Europaudvalget 2023
KOM (2023) 0192
Offentligt
2696964_0001.png
EUROPEAN
COMMISSION
Brussels, 26.4.2023
SWD(2023) 192 final
PART 1/2
COMMISSION STAFF WORKING DOCUMENT
IMPACT ASSESSMENT REPORT
Accompanying the documents
Proposal for a Directive of the European Parliament and of the Council on the Union
code relating to medicinal products for human use, and repealing Directive 2001/83/EC
and Directive 2009/35/EC
Proposal for a Regulation of the European Parliament and of the Council laying down
Union procedures for the authorisation and supervision of medicinal products for
human use and establishing rules governing the European Medicines Agency, amending
Regulation (EC) No 1394/2007 and Regulation (EU) No 536/2014 and repealing
Regulation (EC) No 726/2004, Regulation (EC) No 141/2000 and Regulation (EC) No
1901/2006
{COM(2023) 192 final} - {COM(2023) 193 final} - {SEC(2023) 390 final} -
{SWD(2023) 191 final} - {SWD(2023) 193 final}
EN
EN
kom (2023) 0192 - Ingen titel
GLOSSARY .................................................................................................................................................................... 3
1
INTRODUCTION: POLITICAL AND LEGAL CONTEXT ............................................................................................. 11
1.1
1.2
2
P
OLITICAL CONTEXT
................................................................................................................................................... 12
L
EGAL CONTEXT
........................................................................................................................................................ 14
PROBLEM DEFINITION ....................................................................................................................................... 16
2.1
2.2
2.3
W
HAT ARE THE PROBLEMS
? ........................................................................................................................................ 16
W
HAT ARE THE PROBLEM DRIVERS
? .............................................................................................................................. 20
H
OW LIKELY IS THE PROBLEM TO PERSIST
? ...................................................................................................................... 24
3
WHY SHOULD THE EU ACT? ............................................................................................................................... 25
3.1
3.2
3.3
L
EGAL BASIS
............................................................................................................................................................. 25
S
UBSIDIARITY
: N
ECESSITY OF
EU
ACTION
....................................................................................................................... 25
S
UBSIDIARITY
: A
DDED VALUE OF
EU
ACTION
.................................................................................................................. 26
4
OBJECTIVES: WHAT IS TO BE ACHIEVED? ........................................................................................................... 26
4.1
4.2
4.3
I
NTRODUCTION
......................................................................................................................................................... 26
G
ENERAL OBJECTIVES
................................................................................................................................................. 27
S
PECIFIC OBJECTIVES
.................................................................................................................................................. 27
5
WHAT ARE THE AVAILABLE POLICY OPTIONS? .................................................................................................. 29
5.1 W
HAT IS THE BASELINE FROM WHICH OPTIONS ARE ASSESSED
? .......................................................................................... 29
5.1.1
Projections .................................................................................................................................................. 30
5.2 D
ESCRIPTION OF THE POLICY OPTIONS
........................................................................................................................... 30
5.2.1
Tabular overview of policy options ............................................................................................................. 32
5.2.2
Policy Option A............................................................................................................................................ 33
5.2.3
Policy Option B ............................................................................................................................................ 33
5.2.4
Policy Option C ............................................................................................................................................ 34
5.2.5
Horizontal measures ................................................................................................................................... 36
6
WHAT ARE THE IMPACTS OF THE POLICY OPTIONS? ......................................................................................... 37
6.1 E
CONOMIC IMPACTS
.................................................................................................................................................. 37
6.1.1
Economic impacts of key policy measures .................................................................................................. 40
6.1.2
Option A – combined impact of the measures .......................................................................................... 51
6.1.3
Option B – combined impact of the measures ............................................................................................ 51
6.1.4
Option C – combined impact of the measures ............................................................................................ 53
6.2 S
OCIAL IMPACTS
....................................................................................................................................................... 54
6.3 E
NVIRONMENTAL IMPACT
........................................................................................................................................... 56
7
HOW DO THE OPTIONS COMPARE?................................................................................................................... 57
F
OR EFFICIENCY
,
EFFECTIVENESS AND
EU
ADDED VALUE
,
THE SCORES ARE GIVEN ON THE EXPECTED MAGNITUDE OF IMPACT AS EXPLAINED
ABOVE
: + + +
BEING STRONGLY POSITIVE
, + +
POSITIVE
, +
MODERATELY POSITIVE
, 0
NEUTRAL
, –
MODERATELY NEGATIVE
, – –
NEGATIVE AND
– –
STRONGLY NEGATIVE
. F
OR LEGAL
/
POLITICAL FEASIBILITY AND COHERENCE
, +
MEANS THAT THE ASSESSMENT IS POSITIVE
,
AND
MEANS
THAT IT IS NEGATIVE
............................................................................................................................................................ 58
7.1 E
FFECTIVENESS
......................................................................................................................................................... 58
7.2 E
FFICIENCY ANALYSIS
................................................................................................................................................. 61
7.3 C
OHERENCE
............................................................................................................................................................. 63
7.4 P
ROPORTIONALITY AND SUBSIDIARITY
............................................................................................................................ 64
7.5 L
IMITATIONS OF THE COMPARISON
............................................................................................................................... 64
8
PREFERRED OPTION .......................................................................................................................................... 64
8.1
8.2
8.3
C
OSTS AND BENEFITS OF THE PREFERRED OPTION
............................................................................................................. 65
REFIT (
SIMPLIFICATION AND IMPROVED EFFICIENCY
)........................................................................................................ 69
S
IMPLIFICATION AND BURDEN REDUCTION FOR BUSINESSES
,
SUPPORTING THE ONE IN ONE OUT APPROACH
................................. 70
9
HOW WILL ACTUAL IMPACTS BE MONITORED AND EVALUATED? ..................................................................... 71
1
kom (2023) 0192 - Ingen titel
2
kom (2023) 0192 - Ingen titel
2696964_0004.png
G
LOSSARY
Term or acronym
Accessibility
Meaning or definition
A medicine becomes accessible to patients once it has
been authorised, is being marketed, and can be
reimbursed in a Member State.
Relates to payments to be made by patients (out of
pocket on healthcare or through co-payments) which can
be described as affordability at micro level and to the
sustainability of public funding of the healthcare sector
raised through social security contributions or taxes
(affordability at macro level).
Antimicrobial resistance.
Any substance with a direct action on bacteria that is
used for treatment or prevention of infections or
infectious diseases.
Any substance with a direct action on micro-organisms
used for treatment or prevention of infections or
infectious diseases, including antibiotics, antivirals,
antifungals and anti-protozoals.
The ability of micro-organisms to survive or to grow in
the presence of a concentration of an antimicrobial agent
which is usually sufficient to inhibit or kill micro-
organisms of the same species.
Active Pharmaceutical Ingredient.
Anatomical Therapeutic Chemical code.
Conditional marketing authorisation is the approval to
market a medicine that addresses patients’ unmet
medical needs on the basis of data that is less
comprehensive than that normally required. The
available data must indicate that the medicine’s benefits
outweigh its risks and the applicant should be in a
position to provide comprehensive clinical data in the
future.
Affordability
AMR
Antibacterial/antibiotic
Antimicrobial
Antimicrobial resistance
(AMR)
API
ATC
Conditional marketing
authorisation
3
kom (2023) 0192 - Ingen titel
2696964_0005.png
CMDh
The Coordination Group for Mutual recognition and
Decentralised Procedures – Human is EMA’s committee
responsible for the examination and coordination of
questions relating to the marketing authorisation of
human medicines in two or more Member States in
accordance with the mutual recognition or decentralised
procedure.
European Commission.
The Committee for Orphan Medicinal Products is the
Agency’s committee responsible for recommending
orphan designation of medicines for rare diseases.
The centralised authorisation procedure is the European
Union-wide procedure for the authorisation of
medicines, where there is a single application, a single
evaluation and a single authorisation granted by the
European Commission valid throughout the EU.
Period of protection during which pre-clinical and
clinical data and data from clinical trials handed in to the
authorities by one company cannot be referenced by
another company in their regulatory filings.
The decentralised procedure is the procedure for
authorising medicines in more than one European Union
Member State in parallel. It can be used for medicines
that do not need to be authorised via the centralised
procedure and have not already been authorised in any
Member State. The DCP was introduced by Directive
2004/27/EC, by the 2004 revision.
The European Economic Area includes all EU Member
States and also Iceland, Liechtenstein and Norway.
The European Medicines Agency (‘the Agency’) is an
EU agency founded in 1995 which is responsible for
the scientific evaluation, supervision and safety
monitoring of medicines, both human and veterinary,
across the EU.
Environmental Risk Assessment
European Union.
A centralised European database of suspected adverse
4
COM
COMP
CP
Data protection
DCP
EEA
EMA
ERA
EU
EudraVigilance
kom (2023) 0192 - Ingen titel
2696964_0006.png
reactions to medicines that are authorised or being
studied in clinical trials in the European Economic Area
(EEA).
Evergreening
’Evergreening’ strategies extend the effective patent
period and thus allow drug companies to maintain a
market share after their drug patents expire by
introducing “follow-on drugs” – those with slight
changes made to them after expired patents allow
generic competitors to enter the market.
United States Food and Drug Administration.
Good Distribution Practices.
General Data Protection Regulation.
Good Manufacturing Practices.
Genetically Modified Organism.
A generic medicine contains the same active
substance(s) as the reference medicine, and it is used at
the same dose(s) to treat the same disease(s). The
generic can only be marketed after expiry of the data
and market protection of its reference medicine.
Health Technology Assessment is a multidisciplinary
process that summarises information about the medical,
patient and social aspects and the economic and ethical
issues related to the use of a health technology in a
systematic, transparent, unbiased and robust manner.
High Unmet Medical Need
An impact assessment identifies and describes the
problems to be tackled, establishes objectives,
formulates policy options, assesses the impacts of these
options and describes how the expected results will be
monitored. The Commission's impact assessment
system follows an integrated approach that assesses the
environmental, social and economic impacts of a range
of policy options.
An incremental cost-effectiveness ratio is a summary
measure representing the economic value of an
intervention, compared with an alternative (the
comparator). An ICER is calculated by dividing the
difference in total costs (incremental cost) by the
difference in the chosen measure of health outcome or
effect (incremental effect) to provide a ratio of ‘extra
cost per extra unit of health effect’ for the more
5
FDA
GDP
GDPR
GMP
GMO
Generic medicine
HTA
HUMN
IA
ICER
kom (2023) 0192 - Ingen titel
2696964_0007.png
expensive therapy versus the alternative.
IP
IQVIA
Intellectual property
IQVIA is a contract research and analytical services
organisation that collects data including global
pharmaceutical sales data.
‘Killer acquisitions’ is used as shorthand for:
‘acquisitions’ (in a wide economic sense) of innovative
competitors which have as their object or effect the
discontinuation of overlapping R&D projects to the
detriment of innovation competition and ultimately
consumers. Cunningham, C., Ederer, F. and Ma, S.
(2021), “Killer acquisitions”, Journal of Political
Economy, Vol. 129, No. 3, pp. 649–702. 2
A marketing authorisation is the mandatory approval
process before a medicine enters the market of one,
several or all EU Member States.
Marketing authorisation holder
An application made to a European regulatory authority
for approval to market a medicine within the EU.
A decision granting the marketing authorisation issued
by the relevant authority.
The period after the marketing authorisation of a
medicine for a rare disease when similar medicines for
the same indication cannot be placed on the market and
applications for those medicines cannot be validated.
Under the current legislation, the market exclusivity has
a duration of 10 years.
Period of protection during which generics cannot be
placed on the market.
The United Nations Millennium Development Goals
are 8 goals that UN Member States have agreed to try
to achieve by the year 2015 to reduce extreme poverty.
The MDGs have been superseded by the United
Nations Sustainable Development Goals.
Any deviation(s) from the normal structure or function
of the body, as manifested by a characteristic set of
signs and symptoms (typically a recognised distinct
disease or a syndrome).
Megatrends are long-term driving forces that are
observable now and will most likely have significant
6
Killer acquisitions
MA
MAH
Marketing authorisation
application
Marketing authorisation grant
Market exclusivity
Market protection
MDGs
Medical condition
Megatrend
kom (2023) 0192 - Ingen titel
2696964_0008.png
influence on the future. Megatrends are closely
interlinked between each other and simultaneously
affect many different stakeholders. Thus, a systemic
and global understanding of the issue under study is
necessary to fully picture and illustrate the dynamics at
stake.
See also:
The Megatrends Hub | Knowledge for policy
(europa.eu)
MRP
The mutual recognition procedure (MRP) is a
procedure through which an authorisation of a medicine
in one EU Member State is recognised by another
Member State.
Member States are countries member of the EU.
The national authorisation procedure is a marketing
authorisation procedure where individual Member
States authorise medicines for use in their own
territory. This procedure depends on national
legislation.
New active substances.
National Competent Authority.
New Chemical Entity.
Use of a medicine for an unapproved indication or in an
unapproved age group, dosage, or route of
administration.
A branch of medicine that specialises in the prevention,
diagnosis and treatment of cancer.
A status assigned to a medicine intended for use against
a rare condition. The medicine must fulfil certain
criteria for designation so that it can benefit from
incentives such as market exclusivity.
Parallel import/trade is based on the principle of free
movement of goods in the internal market (TFEU
Articles 34 and 36). This trade is known as "parallel" to
the extent that it takes place outside and – in most cases
– in parallel with the distribution network that the
manufacturers or original suppliers have established for
their products.
An entity responsible for financing or reimbursing
healthcare.
MS
National authorisation
procedure
NAS
NCA
NCE
“Off-label” use
Oncology
Orphan designation
Parallel import
Payer
7
kom (2023) 0192 - Ingen titel
2696964_0009.png
PDCO
The Paediatric Committee is EMA scientific committee
responsible for activities associated with medicines for
children. It supports the development of such medicines
in the EU by providing scientific expertise and defining
paediatric need.
A medical model using characterisation of individuals’
phenotypes and genotypes (e.g. molecular profiling,
medical imaging, lifestyle data) for tailoring the right
therapeutic strategy for the right person at the right
time, and/or to determine the predisposition to disease
and/or to deliver timely and targeted prevention.
The monitoring of the safety of an authorised medicine
and the detection of any change to its benefit-risk
balance.
A paediatric investigation plan is a development plan
designed to ensure that the data required to support the
authorisation of a paediatric medicine are obtained
through studies of its effect on children.
The priority medicine scheme has been launched by the
European Medicines Agency to enhance support for the
development of medicines that target an unmet medical
need. Through this voluntary scheme the Agency offers
early and proactive support to medicine developers to
optimise the generation of robust data on a medicine's
benefits and risks, to optimise development plans and
to enable accelerated assessment of applications.
Quality-adjusted life years refers to a measure of the
state of health of a person or group in which the
benefits, in terms of length of life, are adjusted to
reflect the quality of life. One QALY is equal to one
year of life in perfect health. QALYs are calculated by
estimating the years of life remaining for a patient
following a particular treatment or intervention and
weighting each year with a quality-of-life score (on a 0
to 1 scale). It is often measured in terms of the person’s
ability to carry out the activities of daily life and
freedom from pain and mental disturbance.
Diseases with a particularly low prevalence. The EU
considers diseases to be rare when they affect no more
than 5 per 10,000 people in the EU.
Medicines repurposing identifies new uses for licensed
medicines that are outside of the scope of the originally
intended use for the medicine. This typically involves
taking an existing medicine that already has a
marketing authorisation or licence for human use for a
8
Personalised medicine
Pharmacovigilance
PIP
PRIME
QALYs
Rare disease
Repurposed medicines
kom (2023) 0192 - Ingen titel
2696964_0010.png
particular condition, and then using it to treat another
condition. Alternatively, a repurposed medicine may be
used in a different dose, or form, than its original
licence (for example an inhaled product, rather than a
tablet).
RSB
The Regulatory Scrutiny Board is an independent body
of the Commission that offers advice to the College of
Commissioners. It provides a central quality control
and support function for the Commission’s impact
assessment and evaluation work. The Board examines
and issues opinions and recommendations on all the
Commission's draft impact assessments and its major
evaluations and fitness checks of existing legislation.
Repeat Use Procedure is the use of the Mutual
Recognition Procedure (MRP) after the completion of a
first MRP or Decentralised Procedure (DCP) for the
recognition of a marketing authorisation by other
Member States.
A scientific advice (SA) is the provision of advice by
the Agency on the appropriate tests and studies required
in developing a medicine, or on the quality of a
medicine.
The United Nations Sustainable Development Goals
(UN SDGs) are 17 goals with 169 targets that all UN
Member States have agreed to work towards achieving
by the year 2030. They set out a vision for a world free
from poverty, hunger and disease.
A summary of product characteristics (SmPC)
describes the properties and the officially approved
conditions of use of a medicine.
Micro, small and medium-sized enterprises.
The supplementary protection certificate is an
intellectual property right that serves as an extension to
a patent right. The patent right extension applies to
specific pharmaceutical and plant protection products
that have been authorised by regulatory authorities.
Staff working documents are required to present the
results of all impact assessments and evaluations/fitness
checks.
The proposed indication for the marketing
authorisation. A medical condition that a medicine is
used for. This can include the treatment, prevention and
diagnosis of a disease. The therapeutic indication
9
Repeat use procedure (RUP)
SA
SDGs
SmPC
SMEs
SPC
SWD
Therapeutic indication
kom (2023) 0192 - Ingen titel
2696964_0011.png
granted at the time of marketing authorisation will be
the result of the assessment of quality, safety and
efficacy data submitted with the marketing application.
UMN
Unmet medical need - see Annex 6 for possible criteria
for unmet medical need.
10
kom (2023) 0192 - Ingen titel
2696964_0012.png
1
I
NTRODUCTION
: P
OLITICAL AND LEGAL CONTEXT
This impact assessment covers Directive 2001/83/EC
1
and Regulation (EC) No 726/2004
2
(“general
pharmaceutical legislation”). The EU general pharmaceutical legislation was established in 1965
with the dual objective of safeguarding public health and harmonising the internal market for
medicines. It has developed considerably since then, but these overarching objectives have guided
all revisions. The general pharmaceutical legislation is complemented by the
specialised
legislation
for medicines for rare diseases (‘Orphan Regulation’)
3
, medicines for children (‘Paediatric
Regulation’)
4
, currently under revision, and advanced therapy medicines (‘ATMP Regulation’)
5
. The
general legislation applies to these specialised medicines, while the specialised frameworks provide
additional measures to address their specific characteristics. In particular, they address market
failures by providing specific
incentives
for development of medicines for small number of patients
affected by rare diseases and
rewards
for companies that fulfil the
obligation
to screen adult
medicines under development for use in children
6
. The ATMP regulation adapts the technical
requirements for the authorisation of medicines based on genes, tissues or cells.
The general pharmaceutical legislation governs the granting of marketing authorisations for all
medicines for human use by defining conditions and procedures to enter and remain on the market.
A fundamental principle is that a marketing authorisation is granted only to medicines with a
positive benefit-risk balance after assessment of their quality, safety and efficacy.
The most recent comprehensive revision took place in 2004 while targeted revisions on post-
authorisation monitoring (pharmacovigilance)
7
and on falsified medicines
8
were adopted
subsequently. In the almost 20 years since this revision, the pharmaceutical sector has changed and
has become more globalised, both in terms of development and manufacture. The roles of ‘big
pharma’ and SMEs have changed, with emerging biopharma companies – often SMEs – increasingly
driving innovation and development, with these developments taken over by ‘big pharma’ through
acquisitions or licence agreements.
9
Science and technology have evolved at a rapid pace. However,
there continues to be unmet medical needs
10
, i.e. diseases without or only with suboptimal
treatments. Moreover, some patients may not benefit from innovation because medicines may be
unaffordable or not launched (i.e. placed on the market) in the Member State concerned. There is
1
Directive 2001/83/EC of the European Parliament and of the Council of 6 November 2001 on the Community code
relating to medicinal products for human use,
OJ L 311, 28.11.2001, p.67.
2
Regulation (EC) No 726/2004 of the European Parliament and of the Council of 31 March 2004 laying down Union
procedures for the authorisation and supervision of medicinal products for human use and establishing a European
Medicines Agency,
OJ L136, 30.4.2004, p.1.
3
Regulation (EC) No 141/2000 of the European Parliament and of the Council of 16 December 1999 on orphan
medicinal products
4
Regulation (EC) No 1901/2006 of the European Parliament and of the Council of 12 December 2006 on medicinal
products for paediatric use and amending Regulation (EEC) No 1768/92, Directive 2001/20/EC, Directive 2001/83/EC
and Regulation (EC) No 726/2004
5
Regulation (EC) No 1394/2007 of the European Parliament and of the Council of 13 November 2007 on advanced
therapy medicinal products and amending Directive 2001/83/EC and Regulation (EC) No 726/2004
6
See Annex 6 for further details on the coherence between the two initiatives.
7
Directive 2010/84/EU of the European Parliament and of the Council of 15 December 2010 amending, as regards
pharmacovigilance, Directive 2001/83/EC on the Community code relating to medicinal products for human use, OJ L
348, 31.12.2010, p. 74, and Directive 2012/26/EU of the European Parliament and of the Council of 25 October 2012
amending Directive 2001/83/EC as regards pharmacovigilance, OJ L 299, 27.10.2012, p. 1.
8
Directive 2011/62/EU of the European Parliament and of the Council of 8 June 2011 amending Directive 2001/83/EC
on the Community code relating to medicinal products for human use, as regards the prevention of entry into the legal
supply chain of falsified medicinal products, OJ L 174, 1.7.2011, p. 74.
9
See Annex 9 for further description of the pharmaceutical ecosystem.
10
Possible criteria to define unmet medical need are described in Annex 6.
11
kom (2023) 0192 - Ingen titel
2696964_0013.png
also a greater awareness of the environmental impact of medicines. More recently, the COVID-19
pandemic has stress tested the framework.
This impact assessment (IA) analyses policy options designed to address shortcomings highlighted
in the evaluation
11
of the general pharmaceutical legislation, taking into account the lessons learnt
from the COVID-19 pandemic. It was conducted in parallel with the evaluation (a ‘back-to-back’
exercise).
The revision is part of the implementation of the Pharmaceutical strategy for Europe
12
and aims to:
Promote innovation, in particular for unmet medical needs, while reducing regulatory burden
and the environmental impact of medicines;
Ensure access to innovative and established medicines for patients, with special attention to
enhancing security of supply and addressing risks of shortages, taking into account the
challenges of the smaller markets of the EU;
Create a balanced and competitive system that keeps medicines affordable for health systems
while rewarding innovation.
This revision focuses on provisions relevant to achieve its specific objectives; therefore it covers all
but provisions concerning advertising, falsified medicines, homeopathic and traditional herbal
medicines. The revision of the general pharmaceutical legislation will be presented as a ‘package’
with the revision of the orphan and paediatric legislation. The ATMP regulation is not revised, but
the revision of the general legislation will address some of the issues, e.g. broad application of
hospital exemption, innovative or specific manufacturing methods for these products and
burdensome procedures, identified
13
through the experience accumulated since the entry into force
of the ATMP Regulation and will help translate research into ATMPs available to patients across the
EU while maintaining a high level of public health protection.
1.1
Political context
Since the 2004 revision of the general pharmaceutical legislation, certain aspects such as unequal
patient access, affordability, shortages, or the environmental impact of medicines have become more
prominent and moved up the political agenda. This is evidenced by recent Council conclusions
14
and
resolutions of the European Parliament
15
which called for a balanced system of incentives,
rewarding innovation while improving access. Member States called for revised mechanisms and
incentives for medicines development tailored to the level of unmet medical need, while ensuring
patient access and availability of medicines in all Member States. The COVID-19 pandemic has
spotlighted some critical issues in the European pharmaceutical policy.
The Pharmaceutical strategy for Europe
16
– adopted in November 2020 – is an important building
block of the European Health Union
17
and more than a response to the pandemic. The strategy is a
holistic answer to the current challenges of the pharmaceutical policy with 55 legislative and non-
11
12
Annex 5.
COM(2020) 761 final.
13
COM(2014) 188 final.
14
Council conclusions on strengthening the balance in the pharmaceutical systems in the EU and its Member States, OJ
C, C/269, 23.07.2016, p. 31. Strengthening the European Health Union: improving accessibility to and availability of
medicinal products and medical devices. Council Conclusions on Access to medicines and medical devices for a
Stronger and Resilient EU, (2021/C 269 I/02).
15
European Parliament resolution of 2 March 2017 on EU options for improving access to medicine (2016/2057(INI))
Shortages of medicines,
2020/2071(INI).
16
COM(2020) 761 final https://health.ec.europa.eu/medicinal-products/pharmaceutical-strategy-europe_en
17
COM(2020) 724 final, available at https://ec.europa.eu/info/strategy/priorities-2019-2024/promoting-our-european-
way-life/european-health-union_en.
12
kom (2023) 0192 - Ingen titel
2696964_0014.png
legislative actions interacting together to achieve its overall goal of ensuring Europe's supply of safe
and affordable medicines and supporting the European pharmaceutical industry's innovation
efforts
18
. The revision of the general pharmaceutical legislation and the ongoing revision of the
legislation on medicines for children and rare diseases
19
are flagship initiatives of the strategy.
Although the revision of the general pharmaceutical legislation is a key element in addressing the
objectives of the strategy, its effect needs to be seen with the other actions of the strategy, actions
under EU4Health
20
and other relevant EU and national policies.
The
research and development stage
for medicines is supported by Horizon Europe
21
– a key
funding programme for EU research and innovation – as well as the Innovative Health Initiative
22
,
co-funded by Horizon Europe, to promote innovation of medicines, including planned, specific
partnerships to address unmet medical need
23
and AMR
24
. The Mission on Cancer
25
, together with
Europe’s Beating Cancer Plan
26
will allow to better support development of cancer treatments. The
budget for health research under Horizon Europe amounts to €8.2bn
27
; additional health research is
funded by national programmes. In 2016, Member States from which data are available collectively
budgeted about €11.3bn for health-related R&D; this figure excludes most tax incentives and
funding for higher education and publicly-owned corporations
28
. In the EU, private investment in
R&D in medicines and biotechnology has doubled from around €20bn in 2000 to more than €40bn
in 2018; in the US, starting from a higher level at €40bn it almost doubled to around €75bn in the
same period
29
.
The European Health Data Space
30
- under the European strategy for data
31
– will provide a common
framework across Member States for access to high-quality real world health data. Use of these will
allow progress in research and development of medicines and provide new tools for
pharmacovigilance. The revision of the general pharmaceutical legislation will better accommodate
digital tools and the use of health data
fitting the ambitions of ‘Shaping Europe’s Digital Future’
32
18
19
mission-letter-stella-kyriakides_en.pdf (europa.eu)
Medicines for children & rare diseases – updated rules, available at https://ec.europa.eu/info/law/better-
regulation/have-your-say/initiatives/12767-Medicines-for-children-rare-diseases-updated-rules_en.
20
E.g. a joint action to support the cooperation between competent authorities by organising trainings, improving
scientific assessment capacities and inspections, and an action to contribute to implement the Pharmaceutical Strategy as
it concerns supporting Member States in national pricing and reimbursement policies.
21
Regulation (EU) 2021/695 of the European Parliament and of the Council of 28 April 2021 establishing Horizon
Europe – the Framework Programme for Research and Innovation, laying down its rules for participation and
dissemination, and repealing Regulations (EU) No 1290/2013 and (EU) No 1291/2013, OJ L 170, 12.5.2021, p. 1.
22
Council Regulation (EU) 2021/2085 of 19 November 2021 establishing the Joint Undertakings under Horizon Europe
and repealing Regulations (EC) No 219/2007, (EU) No 557/2014, (EU) No 558/2014, (EU) No 559/2014, (EU) No
560/2014, (EU) No 561/2014 and (EU) No 642/2014, OJ L427, 30.11.2021, p. 17.
23
European Partnership on Rare Diseases will develop a European Clinical Research Network to accelerate clinical trials
for rare diseases; support access to data, information resources to translate research results into safe and effective
medicines; support the scientific work of the International Rare Disease Research Consortium; and integrate basic, pre-
clinical and clinical research. This partnership is planned for the work programme 2023/4.
24
European Partnership: One Health Anti-Microbial Resistance will contribute to achieving the objectives of the
European One Health Action Plan against AMR
24
and the World Health Organization Global Action Plan on AMR
24
, by
reducing the threat of AMR and contribute to achieving the objectives of the Health Emergency Preparedness and
Response Authority (HERA). This partnership is planned for the work programme 2023/4.
25
EU Mission: Cancer, available at
EU Mission: Cancer | European Commission (europa.eu)
26
COM/2021/44 final.
27
European Commission, Directorate-General for Research and Innovation,
Horizon Europe, budget: Horizon Europe -
the most ambitious EU research & innovation programme ever,
2021,
https://data.europa.eu/doi/10.2777/202859.
28
OECD, Pharmaceutical Innovation and Access to Medicines, OECD Health Policy Studies, 2018.
29
Analytical report, indicator RI-8, Annex 10.
30
COM(2022) 197 final.
31
COM(2020) 66 final.
32
COM(2020) 67 final.
13
kom (2023) 0192 - Ingen titel
2696964_0015.png
and the digital transition. By facilitating access to and use of health data the two initiatives together
will support the
competitiveness and innovation capacity
of the EU’s medical industry.
In 2021, the Health Emergency Preparedness and Response (HERA) was created in the aftermath of
the COVID-19 pandemic to prevent, detect and rapidly respond to health emergencies. While HERA
can address medicines shortages related to a health emergency, it will not play a role in addressing
the challenges of
systemic shortages
targeted by the revision of the general pharmaceutical
legislation.
The European One Health Action Plan against
Antimicrobial Resistance
(AMR)
33
aims to reduce
AMR and develop alternative treatments or prevent diseases treated with antimicrobials. The
revision of the general pharmaceutical legislation would contribute to the implementation of this
action plan, together with the planned Council Recommendation on AMR.
The revision will also address
environmental challenges
together with European Green Deal
34
initiatives such as: the EU Action Plan “Towards a Zero Pollution for Air, Water and Soil”
35
, the
revision of the Urban Waste Water Treatment Directive
36
, the revision of the Industrial Emissions
Directive
37
and the revision of the list of surface and groundwater pollutants
38
under the Water
Framework Directive
39
to include some medicines in order to protect the environment and the public
health. Moreover, the EU Strategic Approach to Pharmaceuticals in the Environment
40
lists
measures to address challenges from medicine residues.
Finally, this initiative supports the United Nations’
Sustainable Development Goals
(SDGs)
41
and
in particular SGD 3 (‘ensure healthy lives and promote well-being for all at all ages’), SDG 9 (‘build
resilient infrastructure, promote inclusive and sustainable industrialisation and foster innovation’)
and SDG 10 (‘reduced inequalities’). The objectives and proposed measures relating to unmet
medical need, affordability and unequal access to medicines across the EU are linked to SDG 3 and
SDG 10, while those relating to environmental challenges and addressing inefficiencies of the
regulatory system contribute to SDG 9.
1.2
Legal context
Directive 2001/83/EC and Regulation (EU) No 726/2004 form one policy intervention, the ‘general
pharmaceutical legislation’ that regulates the authorisation, manufacturing, distribution and
monitoring of medicines. It also provides regulatory protection periods to reward innovative
medicines.
42
The legislation is based on cooperation and division of responsibilities between the EU
and Member States. It provides for common standards but different pathways for an authorisation at
EU and at Member State level.
43
Member States are responsible for the authorisation of
manufacturers and wholesale distributors and they conduct inspections of companies.
33
34
A European One Health Action Plan against Antimicrobial Resistance (AMR) (June 2017).
COM (2019) 640 final.
35
COM/2021/400 final
36
Council Directive 91/271/EEC of 21 May 1991 concerning urban waste-water treatment, OJ L 135, 30.5.1991, p. 40.
37
Directive 2010/75/EU of the European Parliament and of the Council of 24 November 2010 on industrial emissions
(integrated pollution prevention and control, OJ L 334, 17.12.2010, p. 17, and COM(2022) 156 final.
38
Integrated water management – revised lists of surface and groundwater pollutants (europa.eu).
39
Directive 2000/60/ EC
of the European Parliament and of the Council of 23 October 2000 establishing a framework
for Community action in the field of water policy,
OJ L 327, 22.12.2000
40
COM(2019) 128 final.
41
Home - United Nations Sustainable Development
42
These regulatory protection periods are described in section 6.1 and in the evaluation SWD, section 3.3, Annex 5.
43
For certain categories of medicines it is a requirement and for others it is an option for companies to apply for a
marketing authorisation granted by the European Commission through the centralised procedure. This authorisation is
valid in all Member States and based on a scientific assessment performed by the EMA. Medicines may also be
authorised through national procedures. The different authorisation procedures are outlined in Annex 7.
14
kom (2023) 0192 - Ingen titel
2696964_0016.png
Pharmacovigilance is a shared responsibility. The legislation does not affect the Member States’
powers regarding the setting of medicine prices or the inclusion of medicines in the scope of national
health insurance schemes.
The general pharmaceutical legislation has touchpoints with other legislation. The ongoing revision
of the legislation on medicines for rare diseases and medicines for children is coherent with this
revision in it aims to address unmet medical needs and improve patient access to medicines; a
description of how the initiatives complement each other can be found in Annex 6.
The Clinical Trials Regulation
44
, applicable since 2022, allows a more efficient approval of clinical
trials in the EU, while the extended EMA mandate, as part of the European Health Union,
strengthens the role of the Agency for a coordinated EU-level response to health crises
45
to ensure
access to medicines in such crisis. The EMA fees legislation
46
is currently under revision. The fees
support EMA and national competent authorities and contribute to the sustainability of the EU
regulatory system.
The revision of the EU legislation on blood, tissues and cells (BTC)
47
is relevant as some substances
of human origin are starting materials for medicines. Coherence between the two revisions is key to
ensure clarity as to which legislation applies to some BTC based therapies.
For
access to medicines,
in addition to the general pharmaceutical legislation, the intellectual
property frameworks (patents and SPCs) as well as the HTA Regulation and the 'Transparency'
Directive
48
play a role. The Intellectual Property Action Plan
49
under the Industrial Strategy
50
includes the modernisation of the system of supplementary protection certificates (SPC) in the form
of a “Unitary SPC” which does not intend to modify the maximum period of a SPC, but may lead to
wider coverage of the SPCs; an impact assessment on these changes is under development.
51
SPCs
extend patent rights to protect
innovation
and compensate for lengthy clinical trials and marketing
authorisation procedures. At the same time, they impact the effect of regulatory protection periods
provided by the pharmaceutical legislation and therefore the entry of generic and biosimilar
medicines and eventually
patient access to medicines and affordability.
Member States’ decisions
on pricing and reimbursement of medicines also influence access. The 'Transparency' Directive
regulates procedural aspects of the Member States’ pricing and reimbursement decisions but do not
44
Regulation (EU) No 536/2014 of the European Parliament and of the Council of 16 April 2014 on clinical trials on
medicinal products for human use, and repealing Directive 2001/20/EC, OJ L 158, 27.5.2014, p. 1.
45
Regulation (EU) 2022/123 of the European Parliament and of the Council of 25 January 2022 on a reinforced role for
the European Medicines Agency in crisis preparedness and management for medicinal products and medical devices,
OJ
L 20, 31.1.2022, p. 1.
46
Council Regulation (EC) No 297/95 of 10 February 1995 on fees payable to the European Agency for the Evaluation
of Medicinal Products,
OJ L 35, 15.2.1995, p. 1, and Regulation (EU) No 658/2014 of the European Parliament and of
the Council on fees payable to the European Medicines Agency for the conduct of pharmacovigilance activities in
respect of medicinal products for human use, OJ L 189, 27.6.2014, p. 112.
These regulations set out fee amounts and
allows for remuneration of the national competent authorities for the contributions to services provided by EMA to
companies, e.g. assessment of application for marketing authorisation.
47
Directive 2002/98/EC of the European Parliament and of the Council of 27 January 2003 setting standards of quality
and safety for the collection, testing, processing, storage and distribution of human blood and blood components and
amending Directive 2001/83/EC, OJ L 33, 8.2.2003, p. 30, and Directive 2004/23/EC of the European Parliament and of
the Council of 31 March 2004 on setting standards of quality and safety for the donation, procurement, testing,
processing, preservation, storage and distribution of human tissues and cells, OJ L 102, 7.4.2004, p. 48.
48
Council Directive 89/105/EEC, of 21 December 1998, relating to the transparency of measures regulating the pricing
of medicinal products for human use and their inclusion in the scope of the national health insurance systems, OJ L 40,
11.2.89, p. 8.
49
COM(2020) 760 final.
50
COM(2021) 350 final.
51
Medicinal & plant protection products – singles procedure for the granting of SPCs
15
kom (2023) 0192 - Ingen titel
2696964_0017.png
impact on the level of price. The Health Technology Assessment (HTA) Regulation
52
will engage
national HTA bodies in joint clinical assessment to provide evidence-based information on the
comparative effectiveness of medicines to help national decisions on pricing and reimbursement.
This contributes to improve affordability and access across the EU. Annex 14 further describes the
multiplicity of factors having an impact and framing access to affordable medicines.
A description of the pharmaceutical ecosystem and legislative landscape can be found in Annex 9
together with a visual overview of the lifecycle of a medicine in Annex 8.
2
2.1
P
ROBLEM DEFINITION
What are the problems?
The evaluation of the general pharmaceutical legislation showed that the legislation continues to be
relevant for the dual overarching objectives of protection of public health and harmonisation of the
internal market for medicines in the EU. The legislation delivered on the objectives of the 2004
revision; albeit not to the same extent for all. The objective to ensure quality, safety and efficacy of
medicines was achieved to the largest extent, while patient access to medicines in all Member States
was achieved only to a limited extent. As to ensuring the competitive functioning of the internal
market and attractiveness in a global context, the legislation has performed to a moderate extent. The
evaluation found that the achievements or shortcomings of the 2004 revision vis-a-vis its objectives
depend on many external factors outside the remit of the legislation, e.g. R&D activities and
international location of R&D clusters, national pricing and reimbursement decisions, business
decisions and market size. The pharmaceutical sector and development of medicines are global;
research and clinical trials conducted on one continent will support development and authorisation in
other continents; likewise the supply chains and manufacturing of medicines are global.
International cooperation to harmonise requirements to support authorisation exists, e.g. the
International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human
Use. The evaluation identified six shortcomings which are not adequately addressed by the
pharmaceutical legislation recognising that they also depend on factors outside its remit:
Medical needs of patients are not sufficiently met
The evaluation showed that the legislation has been less relevant to ensure development of
medicines addressing unmet medical needs, including novel antimicrobials. This related to e.g. lack
of adequate incentives for innovation by SMEs, academic/industry collaborations. Unmet medical
needs with regard to medicines for rare diseases and for children are covered by the parallel revision
of the specialised legislations supported by its own impact assessment.
53
The number of authorised medicines, both innovative and those with well-known active substances
(e.g. generic and biosimilar medicines) is constantly on the rise. Since 2005, between 13 and 43
medicines with new active substances have been authorised in the EU every year, and 4-20 of those
medicines address unmet medical needs
54
. However, there continue to be diseases with no or only
few treatment options, e.g. neurodegenerative or infectious diseases. These unmet medical needs
affect millions of EU citizens
55
. In the public consultation
56
, all stakeholders found that the
52
Regulation (EU) 2021/2282 of the European Parliament and of the Council of 15 December 2021 on health technology
assessment and amending Directive 2011/24/EU,
OJ L 458, 22.12.2021, p. 1.
53
Cf. Ongoing Impact assessment for Medicines for children and rare diseases: https://ec.europa.eu/info/law/better-
regulation/have-your-say/initiatives/12767-Medicines-for-children-rare-diseases-updated-rules_en
54
Analytical report, indicator RI-9, Annex 10.
55
The number of people living with dementia in the EU27 is estimated to be 7,853,705 and Alzheimer's disease is the
most common form of dementia, Other dementias | Alzheimer Europe (alzheimer-europe.org).
56
https://ec.europa.eu/info/law/better-regulation/have-your-say/initiatives/12963-Evaluation-and-revision-of-the-general-
pharmaceutical-legislation/public-consultation_en.
16
kom (2023) 0192 - Ingen titel
2696964_0018.png
legislation moderately promotes the development of medicines for unmet medical needs, with
industry having the most positive view in that regard. While the general pharmaceutical legislation is
not alone responsible for the problem of unmet medical needs
57
, it can be instrumental in addressing
some of the problem drivers within its remit.
AMR - a specific case of unmet medical need
An important area of unmet medical need is drug-resistant infections due to the emergence and
spread of pathogens that have acquired new resistance mechanisms leading to AMR. AMR is
responsible for an estimated 33 000 deaths per year in the EU and amounts to an estimated €1.5bn
every year in healthcare costs and productivity losses
58
. At the same time the pipeline for novel
antimicrobials that can fight resistant pathogens is very weak.
59
There is an apparent market failure
and the lack of market incentives has led to underinvestment by big pharma companies in new
compounds. Annex 15 further describes the market failure in this area.
Unequal access to medicines across the EU
The evaluation showed that the legislation has limited effect and relevance to ensure patient access
to medicines. Access also depends on external factors
60
such as strategic decisions by companies
whether and when to launch a product in a given Member State and national pricing and
reimbursement policies. However, the general pharmaceutical legislation can have an impact on
access through its incentives.
The number of authorised medicines in the EU has increased over time: 1 160 centrally authorised
medicines (CAPs) were authorised in the period 2005-2020 and more than 17 000 medicines,
primarily generic medicines, were authorised through mutual recognition and decentralised
procedures in the same period
61
. However, patient access to medicines varies considerably across the
EU
62
. The number of EU countries in which CAPs are launched has been steadily decreasing
63
.
Substantial differences have been reported in terms of time to entry on the market
64
.
Most medicines are – after authorisation – subject to national pricing and reimbursement decisions
and, in particular for innovative and costly medicines, also HTA. The evidence requirements for
these decisions (on relative/cost effectiveness of new medicines compared to existing treatments) are
different than for the authorisation of those medicines, which is based on a positive benefit-risk
balance for patients. Evidence required for HTA or pricing and reimbursement decisions are (often)
not generated by companies by the time of the authorisation of the medicine and this may delay
access. However, the recently adopted HTA Regulation intends to improve the situation, though its
effects could not yet been taken into account in the evaluation and the consultations.
Evidence
65
shows that, whilst in Germany 133 out of 152 (i.e. 88%) new medicines authorised
between 2016 and 2019 at EU level were accessible to patients, small Member States such as the
Baltic Member States or Member States with comparatively low prices or with low GDP, like
Romania, had fewer than 50 of these available
66
. The time to patient access is also significantly
57
58
External factors (e.g. scientific barriers) are mentioned in the problem drivers for unmet medical need, see section 2.2.
A European One Health Action Plan against Antimicrobial Resistance (AMR) (June 2017).
59
Of 43 antibiotics in development, 15 were in Phase 1 clinical trials, 13 in Phase 2, 13 in Phase 3, and two have had
new drug applications submitted. Historically, about 60% of drugs that enter Phase 3 will be approved.
60
See Annex 14 on the factors influencing access to affordable medicines
61
Analytical report, indicator ACC-1, Annex 10.
62
Technopolis Evaluation study report, figure 10, 2022.
63
Kyle, M.K, (2019). The Single Market in Pharmaceuticals. Review of Industrial Organization, 55(1),111-135.
https://doi.org/10.1007/s11151-019-09694-6
64
Bergmann et al., 2016, Ferrario (2016). Access to innovative oncology medicines in Europe. Annals of Oncology,
27(2), 353-356. https://doi.org/10.1093/ANNONC/MDV547
65
Data from European Federation of Pharmaceutical Industries and Associations (EFPIA) and IQVIA.
66
Newton et al. (2021).
EFPIA Patients W.A.I.T. Indicator 2020 Survey.
17
kom (2023) 0192 - Ingen titel
2696964_0019.png
longer for most of these latter countries, e.g. approximately two years or more after marketing
authorisation in Romania compared to four months in Germany. Similar observations were made
across different subsets of medicines. As a result, patients may not have had access to any
appropriate treatment for their disease.
Although access depends, as explained above, on a multiplicity of factors, most respondents in the
targeted survey, except industry agree that there is still room for improvement of the EU legislation
in terms of access.
Most of the nationally authorised medicines are generic medicines
67
. Generic and biosimilar
medicines can be marketed only after the expiry of regulatory and other intellectual property
protection periods of the original medicine. They normally drive prices down and improve access.
Low volume markets still experience limited access to generics.
Affordability of medicines is a challenge for health systems
Innovative medicines are often costly. Medicine prices vary significantly between Member States
68
.
A study showed that list prices were the highest in Germany and the cheapest in many different EU
countries but never in those with lower GDP like Bulgaria or Romania
69
. The medicines analysed
were unaffordable for many EU health systems. Pharmaceutical budgets also put pressure on health
systems. Medicines in hospitals account for over 20-30% of hospital expenditures and are growing
70
.
In 2013-2019, the average household out-of-pocket (including regulated co-payments) share of non-
hospital medicines is stable, at around 28-30%, but there are big differences between the MS with
countries like Germany and France having shares below 20% and Poland and Bulgaria over
respectively 60 and 70%.
71
Out-of-pocket payment for medicines is outside of the remit of the
pharmaceutical legislation. Other external factors are described in Annex 14.
Against this backdrop, generic and biosimilar entry can be an important factor in terms of
competition, to achieve lower prices, broadening patients’ access and alleviating healthcare costs
72
.
In the EU, the share of generics in total medicines sales revenue modestly increased (from 13% to
16%) between 2002-2020
73
. An analysis shows that the EU is on a similar trend as other comparable
markets (Japan and USA)
74
. Nonetheless, inquiries show that originator companies sometimes use
various practices (such as “evergreening” or “killer acquisitions” early in the pipeline) to delay or
prevent generic/biosimilar entry. These anti-competitive practices can be prosecuted by EU
competition authorities. The evaluation confirms that further efforts can be made to fully exploit the
savings generated by the generic and biosimilar competition; although measures in this regard are
primarily outside the scope of the general pharmaceutical legislation, the revision can improve the
conditions for generic and biosimilar authorisation and competition.
67
Study on the experience acquired as a result of the procedures for authorisation and monitoring of medicinal products
for human use, EY, January 2020, p. 103.
68
The desk research suggests for example an almost 11-fold difference between interferone-beta list prices in Germany
(€1451.17) and Croatia (€132.77); list prices do not include the confidential rebates (if they exist) or ‘price freezes’ and
may therefore not correspond to the actual price.
69
Zaprutko T, Kopciuch D, Kus K, et al. Affordability of medicines in the European Union.
PLoS One.
2017;12(2):e0172753.
70
European Commission, State of health in the EU: companion report 2019 (ISBN 978-92-76-10194-9)
71
OECD, Eurostat and World Health Organization (2017), A System of Health Accounts 2011: Revised edition, OECD
Publishing, Paris.
http://dx.doi.org/10.1787/9789264270985-en).
72
IMS Health (2015) The Role of Generic Medicines in Sustaining Healthcare Systems: A European Perspective
73
Evaluation SWD, section 4.1.1.4, Annex 5.
74
Ibid, footnote 67.
18
kom (2023) 0192 - Ingen titel
2696964_0020.png
The probability of competition is lower for (a) biosimilars than for generics; (b) products with
manufacturing complexity and (c) products with smaller turnover (e.g. for rare diseases).
7576
According to all stakeholder groups, enabling access to affordable medicines is among the areas
where the legislation has been less effective. The rising costs of medicines were key concerns for
academics, healthcare professionals, public authorities and civil society stakeholders.
Shortages of medicines
The evaluation showed that medicine shortages are an increasing problem in the EU; a problem that
was also experienced during the COVID-19 pandemic. Over the last 10 years, there has been a
strong increase in the number of shortages notified in the EU from a few in 2008 to nearly 14 000 in
2019
77
. There are a number of root causes. These include: more complex and diversified global
supply chains, quality and manufacturing challenges and commercial decisions or unexpected
increase in demand. Evidence shows that medicine shortages are placing a significant burden on
health systems, health professionals and are ultimately putting patients at risk of sub-optimal care
and health systems at risk of higher healthcare costs
78
.
Medicine shortages have also a global dimension due to the global supply chain, where external
actions or events impact the supply of medicines in the EU, e.g. the Indian export restriction of
certain active substances during the COVID-19 pandemic. Likewise, problems at a manufacturing
site may cause shortages in several Member States or the whole EU, depending on the supply chain.
The public consultation confirms the importance all stakeholders (in particular civil society
organisations and healthcare professionals) place on medicine shortages. In the targeted survey, civil
society, public authorities and health service stakeholders considered that the legislation is the least
effective in addressing issues related to security of supply and medicine shortages.
The general pharmaceutical legislation can provide harmonised tools to allow Member States to
better handle medicine shortages and thus act as enabler for addressing the problem.
The regulatory system does not sufficiently cater for innovation/unnecessary administrative burden
While the system for authorisation and monitoring of medicines in the EU overall meets the
objectives of the general pharmaceutical legislation, rapid scientific and technological developments
have resulted in new challenges for the system, which has become more complex over time, as
reflected by the expansion of the number of EMA scientific committees and their interactions
79
.
New types of medicines (e.g. personalised medicines), approaches and processes, may raise
questions about whether they fully fit within the scope of the legislation and can find themselves
subject to unintended barriers to innovation, development, production or marketing authorisation.
Products combining medicines with technologies regulated under other frameworks (e.g. medical
devices, artificial intelligence) or products using new platform technologies
80
face uncertainty about
the applicable framework. Likewise, the current framework is not adapted to novel production
technologies or methods (e.g. decentralised manufacturing). Borderline issues for ATMPs with the
BTC framework, which provides starting materials, were also highlighted in the evaluation.
75
76
SWD(2020) 163 final, p. 58.
Understanding Net Pharmaceutical Expenditure Dynamics in Europe, April 2022, IQVIA.
77
Analytical report, indicator SM-1, Annex 10. Data only collected for period 2008-2020, during which many Member
States put in place new systems or requirements for notification of shortages.
78
European Commission, Directorate-General for Health and Food Safety, Jongh, T., Becker, D., Boulestreau, M., et
al., Future-proofing pharmaceutical legislation: study on medicine shortages: final report (revised),
2021, https://data.europa.eu/doi/10.2875/211485.
79
COM(2021) 497 final.
80
When a certain process/method is used to manufacture specific individualised treatments, i.e. adjustments to the
medicine are made based on the characteristics of the patient or the causing pathogen.
19
kom (2023) 0192 - Ingen titel
2696964_0021.png
The consultations showed a consensus between academia/research organisations, patient/consumer
organisations, healthcare professionals and industry that the legislation was not flexible enough to
accommodate scientific advances, such as real-world data in healthcare. Public authorities noted that
medicines regulators need more resources to keep up with the speed of scientific and technological
developments and to assess complex therapies appropriately.
Digital transformation has been changing the health sector. However, there is an overall lack of
transparency and interoperability; digital expertise and infrastructure are not sufficiently available
across the Member States and the EU regulatory network. All stakeholders agreed that EU
telematics systems play an important role in contributing to the efficiency of the system, but also
identified room for improvement (like a very complex governance system for EU telematics).
An assessment of the current authorisation system
81
identified the need for rationalisation and
simplification which the consultations echoed. Stakeholders noted the need for strengthened
coordination between bodies responsible for marketing authorisation procedures, clinical trial
authorisations, HTA and pricing and reimbursement. Several industry respondents stated that
regulatory burden can be costly, duplicative and thus hinder innovation, in particular for innovative
SMEs who may struggle with high fee costs, though fees incentives exist for SMEs
82
.
Medicines in the environment
While the positive effect of medicine for treatment of diseases is undisputed, pollution caused by
medicines is a well-documented risk to the environment and human health, particularly in relation to
antimicrobial resistance. Residues of medicines may enter the environment during their
manufacturing, use by patients and disposal, with the largest source being the use
83
. Residues of
medicines have been found in surface and ground waters, soils and animal tissues across the EU at
concentrations depending on the medicine and the proximity of sources
84
. Traces have also been
found in drinking water. Residues of medicines in the environment is a global problem
85
. The
evaluation confirmed that the current requirement for an environmental risk assessment (ERA)
before marketing authorisation has some weaknesses as regards compliance, content and scope.
In the targeted consultations, the stakeholders (industry, civil society and public authorities) ranked
reducing the environmental impact of medicines among the objectives where the general
pharmaceutical legislation had been the least effective. In the public consultation, the stakeholders
across the board found that the legislation has performed moderately in ensuring that medicines are
manufactured, used and disposed of in an environmentally friendly manner, with citizens, healthcare
professionals and public authorities being the most critical.
2.2
What are the problem drivers?
Figure 1 provides an overview of the problem drivers and their link with the problems identified.
81
82
COM(2021) 497 final.
Commission Regulation (EC) No 2049/2005 provides for specific support for SMEs, including an SME Office in the
EMA and fee reductions and deferrals. Further fee incentives for SMEs are provided in the Rules for implementation of
the two fee regulations (Council Regulation (EC) No 297/95 and Regulation (EU) No 658/2014).
83
COM(2019) 128 final.
84
Analytical report, indicator E-1, Annex 10.
85
Idem.
20
kom (2023) 0192 - Ingen titel
2696964_0022.png
Figure 1
Problem tree diagram for the revision of the general pharmaceutical legislation
The problem drivers that are causing underperformance on the ground are a series of complex,
interlinked factors.
Drivers for unmet medical needs
Despite the fast-paced advances in science and technology, scientific barriers prevent the
development of medicines to treat or cure some diseases such as Alzheimer’s. For unmet needs,
there are a series of different drivers, e.g. market failure, complexity of disease pathologies,
knowledge gaps in molecular and physiological underpinnings of diseases, high risk R&D. While
the EU has a world-leading, research-intensive pharmaceutical industry
86
, evidence suggests that
R&D costs per new medicine have increased over time with estimates ranging from US$944m to
US$2,826m with great variability across therapeutic fields
87
. This is one among the drivers that have
increased the commercial risk of developing new medicines for
unmet medical need.
Big pharma companies tend to disinvest from riskier upstream research and to choose R&D
investments that will maximise their future profits through licensing or acquisitions of products that
are already in later clinical trial stages with good probability for marketing authorisation, sales and
high price.
88
Such business strategies are not always aligned with the public goal of directing efforts
towards the greatest unmet medical needs. Furthermore, the pharmaceutical legislation makes no
distinction in regulatory incentives granted to highly innovative medicines addressing unmet
medical need and those for incremental innovation, such as ‘me-too’ medicines (similar to existing
medicines) without added therapeutic value. This gives less incentive to invest in higher risk
development of the former. There is a concentration of investment in areas where there is less
86
87
The Pharmaceutical Industry in Figures, Key Data 2021 (EFPIA, 2021).
Simoens, S., & Huys, I. (2021). R&D costs of new medicines: a landscape analysis.
Frontiers in medicine, 8,
available
at
https://www.frontiersin.org/articles/10.3389/fmed.2021.760762/full.
88
EPRS_STU(2021)697197_EN.pdf (europa.eu):
European pharmaceutical research and development. Could public
infrastructure overcome market failures?
21
kom (2023) 0192 - Ingen titel
2696964_0023.png
financial risk, e.g. oncology. When companies invest in less risky areas, even incremental innovation
can lead to an economically viable or profitable product.
The
growing resistance of pathogens to antimicrobials (AMR)
combined with the weak global
pipeline of major new classes of antimicrobials are a special driver for unmet medical need. A
growing market failure derives from the fact that the typical cost of surpassing the scientific
challenges involved in developing new antimicrobials is very high and at the same time the typical
income and profit that can be derived from sales of these products are very limited because
healthcare systems want to keep new antimicrobials in reserve or limit their use so as not to fuel the
vicious cycle of AMR, by inappropriate use of already authorised antimicrobials.
Drivers for access to medicines
A key
access problem driver
is that authorised
medicines are not launched in all Member States
or are subsequently withdrawn.
Currently, companies have the choice where and when to launch
centrally authorised medicines, the legislation only requires them to place their product on the
market in at least one Member State within three years of its authorisation (the so-called ‘sunset
clause’). Other than that, companies have a free hand; this creates an unpredictable situation for
patients and Member States. With some Member States companies enter into pricing and
reimbursement negotiations only very long time after marketing authorisation or not at all. The
decision for the company to launch and when depends on different factors for example the size of
the patient population, or national pricing and reimbursement policies, and the organisation of health
systems. These factors influence whether the company can successfully pass a HTA in that Member
State and finally negotiate a price and a reimbursement status for the product.
Access may also differ due to organisational differences in Member States (different medical
protocols, access to specific equipment/infrastructure needed for administration, different
characteristics of the health systems).
The pharmaceutical legislation has no direct influence on HTA and pricing and reimbursement
processes or the organisation of the national health systems. However, the general pharmaceutical
legislation and its system of regulatory incentives can be an enabling factor to improved access by
incentivising market launch by companies, strengthening the position of national pricing and
reimbursement bodies, facilitating collaboration among decision makers along the lifecycle of a
medicine and by increasing competition from generics and biosimilars.
For a more detailed analysis on the factors and dynamics behind the market launch, the access chain,
HTA, pricing/reimbursement process and on pharmaceutical expenditure please refer to Annex 14.
Withdrawals of medicines
disrupt the established access chain (from authorisation to entry into the
health system). An available product abruptly or gradually withdrawn from the market (often for
commercial reasons) can create
shortages
and leave patients without treatments.
Drivers for affordability of medicines
Pharmaceutical expenditure
is largely subsidised by national health systems in order to ensure the
adequate provision of medicines to all their respective citizens. New, highly
innovative medicines
may place pressure on public budgets due to their prices. Therefore, Member States adopt measures
to regulate the prices of medicines and the conditions of their public funding based on their
exclusive competence in this field (Article 168 TFEU). Member States follow different
price and
reimbursement policies
and the pharmaceutical markets remain very fragmented by country (for a
review of pricing policies
89
). The External Reference Pricing (ERP) policy, for which the price set
for the same product in one or several countries is used as a benchmark for setting the product's
price in a given country, is the most frequently used pricing policy in Europe. As a consequence of
89
WHO guideline on country pharmaceutical pricing policies, Geneva: World Health Organization; 2020.
22
kom (2023) 0192 - Ingen titel
2696964_0024.png
differences in prices, the use of ERP and parallel import, and differences in market size, the
availability and entry date of medicines strongly differ among Member States.
The prices by country do not depend only on the government regulation (such as price controls and
reimbursement decisions) but also on several other factors, such as income per capita, the size of the
market, the characteristics of the product (innovative or old, its therapeutic advantages etc.), the
patent status, the presence of competitors and research costs incurred (also for unsuccessful
development of medicine)
90
. However, there is a lack of transparency on R&D costs or public
contributions to these costs. While R&D costs are not relevant for the assessment of a medicine’s
benefit-risk balance, information on such costs are relevant for the downstream actors and may
facilitate their decision-making.
Delay in generic and biosimilar entry is also a driver for expensive innovative medicines.
The general pharmaceutical legislation has only an indirect impact on the affordability of medicines
by facilitating competition and early market entry by generic and biosimilar medicines. In a similar
way, it streamlines procedures and makes the regulatory framework more efficient thereby lowering
costs for authorisation or manufacturing which could have an impact on the price of the medicine.
Drivers for shortages of medicines
Vulnerability in the global supply chains
has arisen from global industry consolidation with
increased complexity in supply chains, in which many different intermediate suppliers may be
connected, and, in particular for generic medicines, from reliance on a few, specialised overseas
suppliers that produce at lower prices. In addition, the notification and obligation to ensure
appropriate and continued supply, varies across Member States with e.g. 4 months in advance
notification of shortages in Italy and at least 6 months in Romania
91
.
While Member States have already introduced a variety of actions at the national level to help
protect their security of supply, the impact of these measures on preventing and mitigating the
impact of shortages is not yet sufficiently understood.
Drivers for medicines into the environment
The
lack of relevant or insufficient regulation and oversight
currently influences the effects
medicines use may cause for the environment, while a lack of environmental expertise influences the
understanding of the effects on the environment from medicines. The largest source of medicines
entering the environment is the use of medicines; due to the chemical and/or metabolic stability of
some medicines, as much as 90% of the active substance is excreted or washed off into the
environment in its original form
92
. Pharmaceuticals mainly reach the environment through:
-
the discharge of effluent from urban waste water (sewage) treatment plants –
containing excreted pharmaceuticals as well as unused pharmaceuticals thrown away
into sinks and toilets, despite the existence of collection schemes;
the spreading of animal manure; and
aquaculture, in which pharmaceuticals are often dispensed with the animal feed.
93
-
-
Another source is the discharge of effluent from manufacturing plants (especially those outside the
Union) with potential impacts that may significantly effect on a local scale when manufacturing
90
Zaprutko T, Kopciuch D, Kus K, et al. Affordability of medicines in the European Union.
PLoS One.
2017;12(2):e0172753.
91
European Commission, Directorate-General for Health and Food Safety,
Future-proofing pharmaceutical legislation:
study on medicine shortages: final report (revised),
2021,
https://data.europa.eu/doi/10.2875/211485.
92
COM(2019) 128 final.
93
Idem as 92.
23
kom (2023) 0192 - Ingen titel
2696964_0025.png
emissions of wastewater are inadequately managed.
94
Environmental legislation, such as the Urban
Waste Water Directive – currently under revision – and other environmental legislation and
initiatives mentioned in section 1.1, is the main instrument for addressing reduction of medicines
residues and hence the environmental impact of the industry; however, not even the best and most
expensive current wastewater treatments are 100% effective. The measures in this revision
complement environmental legislation.
Drivers for lack of innovation and inflexible regulatory framework
The rapid pace of the scientific and technological development is a driver for – and an external
factor to – the problem that the regulatory system does
not sufficiently cater for innovation.
The
general pharmaceutical legislation is often prescriptive, and it takes a long time to amend it. Hence,
the
medicines framework lacks agility
to respond to rapid developments.
Inefficiencies in the regulatory framework
were identified in the evaluation, e.g. redundant
requirements like the 5-year renewal of marketing authorisation, leading to unnecessary
administrative burden. In addition, there is duplication of assessment by the medicines authorities,
for instance when different companies apply for authorisation of the same product with the same
clinical trial in different procedures. There is insufficient pan-European digital infrastructure and
legal basis for optimal use of electronic tools for companies or medicine authorities which
contributes to a loss of competitiveness. Better use of digitalisation in the framework, e.g. through
electronic product information, could help combat shortages, increase access in smaller markets and
also support competition, while improving information on medicines.
2.3
How likely is the problem to persist?
If no EU action is taken, the problems described will persist. While more medicines are expected to
be authorised (for CAPs this might increase to 40-60 medicines containing new active substances
per year
95
), these medicines will not necessarily address unmet medical needs to a greater extent
than today. For example, recently approved antibiotics
96
and the clinical pipeline are insufficient to
tackle the increasing emergence and spread of antimicrobial resistance
97
. The market failures in this
area will not be corrected without interventions on several fronts, including the general
pharmaceutical legislation. The persistence of the problems is also confirmed by some of the
megatrends identified by the EU Joint Research Centre
98
.
The megatrend on shifting health
challenges describes demographic changes and environmental challenges that could create new
unmet medical needs and public health burdens as demonstrated by the COVID-19 pandemic.
Authorised medicines will continue to be inaccessible at affordable prices in some Member States.
The ‘access chain’ mechanism mentioned above and analysed in Annex 14 is affected by
deficiencies that are systemic in nature and some of the ‘links’ lie outside the remit of this
legislation. Nevertheless, the analysis of the policy options in section 6 shows that the revision of the
legislation can act as a key enabler for access and can influence affordability. The policy
interventions in the legislation shall be complemented by other actions of the pharmaceutical
strategy, e.g. best practice exchange between Member States on pricing, payment and procurement.
94
95
Larsson DGJ. 2014 Pollution from drug manufacturing: review and perspectives.
Phil. Trans. R. Soc
369:20130571.
Described in section 5.1.1.
96
Since 2015. 11 antibacterials with new active substance have been granted a Union marketing authorisation, though
none of these products constituted a new class of antibiotic.
97
Antimicrobial products in clinical development for priority pathogens (April, 2021), 68 products are in development
(41 antibiotics and 27 non-traditional antibacterial agents) see https://www.who.int/observatories/global-observatory-on-
health-research-and-development/monitoring/antibacterial-products-in-clinical-development-for-priority-pathogens.
98
The Megatrends Hub | Knowledge for policy (europa.eu)
24
kom (2023) 0192 - Ingen titel
Since new scientific and technological developments will continue, some problems may exacerbate
if the legislation is not future-proof. Current work-arounds which are based on 'creative'
interpretation will become bottlenecks, especially for complex products. Borderlines between
product categories may be more blurred and determination of applicable legal frameworks and their
interaction may become complex, leading to longer development or authorisation processes for
innovative medicines and thus a longer time to reach patients. This impacts negatively innovation
while some innovative products may remain unregulated with negative effect on public health.
If the efficiency of the regulatory system will not be improved and administrative burden not
reduced, e.g. by digitisation, valuable resources might not be available to facilitate the development
and the assessment of innovative medicines. Likewise, resources might not be available to invest in
the expertise needed to cope with new scientific and technological developments. For the industry,
there might be less investment in new medicines and hence fewer new medicines authorised,
reduced innovative capacity and competitiveness. The megatrend on accelerating technological
change and hyperconnectivity is particularly relevant both in terms of development and innovation
of medicines and of digitisation of the regulatory system.
Likewise, the problem of medicine residues in the environment will persist if no EU action is taken
with risks to flora, fauna and habitat due to the pharmacological characteristics of the active
substances. The megatrend on increasing demographic imbalances with the ageing population in the
EU may exacerbate the environmental challenges from medicines as elderly people tend to use more
medicines than young people; this could also put further pressure on national health systems.
3
3.1
W
HY SHOULD THE
EU
ACT
?
Legal basis
The general pharmaceutical legislation is based on Articles 114 and 168 of the Treaty on the
Functioning of the European Union (TFEU). These articles provide the legal basis for the EU to
adopt measures which have as their object the establishment and functioning of the internal market
(Article 114(1)) as well as setting high standards of quality and safety of medicinal products (Article
168(4)(c)). While the internal market and common safety concerns in public health matters fall
within a shared competence of the EU and Member States, once the EU adopts harmonised
legislation in such an area, Member States can no longer exercise their own competence. This is the
case for the general pharmaceutical legislation. Any future legislative proposals, supported by this
impact assessment, will be based on Articles 114(1) and 168(4)(c) TFEU. It will also consider
Article 35 of the EU Charter of Fundamental Rights that provides that the Union is to ensure a high
level of human health protection in the definition and implementation of Union policies.
As per Article 168(7) of the TFEU, Member States are responsible for the definition of their health
policy and for the organisation and delivery of health services. Consequently, coverage and pricing
decisions for medicines are outside the scope of the legislation.
3.2
Subsidiarity: Necessity of EU action
Diseases do not know borders. Common provisions for the authorisation of medicines constitute a
cross-border issue for public health that affects all Member States and thus can effectively be
regulated only at EU level, given that the authorisation of medicines is fully harmonised at EU level.
The objectives this revision intends to achieve benefit all Member States. EU action relies also on
the single market to achieve a stronger impact as regards access to safe, effective and affordable
medicines, as well as the security of supply across the EU. National actions are likely to create
disharmonised solutions resulting in fragmentation, and possibly exacerbate some of the problems to
be solved, distort competition and increase administrative burden for the pharmaceutical companies,
which often operate in more than one Member State. An example of fragmentation is the additional
25
kom (2023) 0192 - Ingen titel
2696964_0027.png
and non-harmonised measures introduced by Member States to prevent and mitigate medicines
shortages
99
. A harmonised approach at EU level also provides greater potential for incentives to
support innovation and for concerted action for development of medicines in areas of unmet needs.
The legislation respects Member States’ exclusive competence in the provision of health services,
including pricing and reimbursement policies and decisions. In this respect, the Pharmaceutical
Strategy provides for supporting non-legislative actions such as cooperation mechanisms, e.g.
through a group of competent authorities, based on mutual learning and best practice exchange on
pricing, payment and procurement policies. These exchanges can be facilitated at EU level.
3.3
Subsidiarity: Added value of EU action
This initiative revises a system with recognised EU added value for the EU patients/citizens,
pharmaceutical industry and medicines authorities through e.g. timely authorisation, patient access
and continuous supply of innovative and established medicines and strong cooperation
100
.
This revision is expected to bring further benefits by addressing unmet medical needs and
contributing to reducing the unequal patient access to medicines across the EU. At the same time,
simplification and streamlining of processes are expected to reduce administrative burden for
companies and authorities and hence improve efficiency and attractiveness of the EU system.
This revision can influence positively the competitive functioning of the market through the review
of the incentives and other measures to facilitate early entry on the market of generic and biosimilar
medicines and hence improve patient access and affordability.
These benefits and cost-savings can best be achieved by EU action, while recognising that external
factors such as national pricing and reimbursement policies and company decisions to launch
medicines have great impact on patients’ access to medicines. Furthermore, science and
technological developments, as well as R&D policies and company investment decisions influence
innovation, especially for unmet medical needs.
The measures to support security of supply under this initiative relate to the responsibilities of
marketing authorisation holders and supply chain actors like wholesalers. Those actors are already
covered by the EU pharmaceutical legislation. However, measures supporting security of supply go
beyond legislative measures; many actions do actually take place already at national level and will
continue to do so. National and EU levels are not alternatives to each other, but complementary.
In a few instances, the evaluation identified problems with a harmonised implementation of the
Directive across Member States
101
. However, these problems relate to vague legal wording of the
respective provision rather than the legal instrument used. Moreover, in 2019, a REFIT Platform
Opinion
102
considered a suggestion to turn the Directive into a Regulation, though that suggestion
did not receive overall support. The opinion showed that many Member States considered the
system sufficiently harmonised and would not see a need for a Regulation.
4
4.1
O
BJECTIVES
: W
HAT IS TO BE ACHIEVED
?
Introduction
This section sets out the general and specific objectives as well as the logic (Figure 2) underpinning
the revision. It addresses the problems identified, and provides a focus for assessing and comparing
99
European Commission, Directorate-General for Health and Food Safety, Future-proofing pharmaceutical legislation :
study on medicine shortages : final report (revised), 2021, https://data.europa.eu/doi/10.2875/211485
100
Evaluation SWD, section 4.2, see Annex 5.
101
E.g. application of the Bolar provision – see page 7 of the evaluation SWD
102
https://wayback.archive-it.org/12090/20200308120955/https:/ec.europa.eu/info/sites/info/files/xi.9.a_medicinal_products_for_human_use.pdf
26
kom (2023) 0192 - Ingen titel
2696964_0028.png
the likely cost-effectiveness of the selected policy options. The two legislations constituting the
general legislation make up a single intervention logic in this policy area.
Figure 2
Intervention logic for the general and specific objectives, problem drivers and problems
4.2
General objectives
The general objectives of the revision remain unchanged in that the general pharmaceutical
legislation aims to ’guarantee a high level of public health by ensuring the quality, safety and
efficacy of medicines for EU patients’ and harmonise the internal market.
4.3
Specific objectives
In response to the problems identified, this revision aims to:
1 . Promote innovation, in particular for unmet medical needs
The objective is to promote innovation with special focus on medical conditions not yet addressed
and which represent a significant EU health burden (unmet medical needs). The revision should
enable major biomedical research advances and ensure a pipeline of innovative new medicines for
use across the EU. It should also support pharmaceutical R&D and strengthen the competitiveness of
the research-based EU pharmaceutical sector.
The objective is also to address the market failure related to the development of novel antimicrobials
through novel incentives that can finance the research required while respecting the need for a as
limited as possible use of antimicrobials to reduce the tendency of pathogens to develop resistance.
2 . Create a balanced system for pharmaceuticals in the EU that promotes affordability for
health systems while rewarding innovation
This objective aims to enable competition, to promote affordability of medicines for health systems
across the EU and ensure healthcare costs are sustainable for Member States. Affordability should
not though be promoted at the expense of innovation, which also benefits patients. Thus, the
underlying ambition is to create a balance where, on the one hand, innovation is rewarded, and on
the other hand, faster market entry of generic and biosimilar medicines is facilitated, as a means to
27
kom (2023) 0192 - Ingen titel
2696964_0029.png
improve competition across the EU. This is expected to drive down costs for medicines with the
additional benefit of strengthening the EU generic and biosimilar industry.
Affordability is a new objective of the revision, which can only indirectly be impacted by the
general pharmaceutical legislation.
3 . Ensure access to innovative and established medicines for patients, with special
attention to enhancing security of the supply across the EU
This objective aims to promote equal access to medicines for all EU citizens, including in smaller
Member States, after a timely authorisation under the EU pharmaceutical system. After a medicine
has been developed and become available after a timely authorisation under the EU pharmaceutical
system, patient access has two dimensions: (i) equal access to/market entry of innovative medicines
across the EU and (ii) continuous supply and limited shortages of all medicines. As regards the first,
the aim is to provide a motivation to companies to rapidly reach an agreement with Member States
and engage Member States in effective negotiations. Facilitating competition from generic and
biosimilars will also serve the same objective. As regards the second dimension (shortages and
keeping products on the market), the aim is to enhance and harmonise notification requirements and
obligations to ensure appropriate and continued supply across Member States.
4 . Reduce the environmental impact of the pharmaceutical product lifecycle
This objective aims to reduce the environmental impact of pharmaceuticals through minimising
medicine residues in the environment from their production, use, and disposal. This would entail an
enhanced assessment of environmental risks of medicines and appropriate risk mitigation measures,
including on their prudent use, especially for AMR.
5 . Reduce the regulatory burden and provide a flexible regulatory framework
This objective aims to create a more flexible regulatory framework, to future-proof innovation and
reduce regulatory burden. Through simplifying and integrating regulatory requirements and
pathways and reducing burden for industry and public authorities alike, this objective aims to
increase the attractiveness of the EU regulatory system. The goal is to provide clarity on the
appropriate regulatory pathway, reduce approval times and costs while maintaining high standards
and robust assessment of the quality, safety, and efficacy of medicines. Leveraging digital
technology and the use of electronic product information could support this objective.
Objectives 1, 2 and 5 work in synergy for promoting innovation as do objectives 2, 3 and 5, with a
range of measures to achieve access to affordable medicines. Trade-offs have to be considered
between objectives 4 and 5 as measures to reduce the medicine residues in the environment are
likely to increase the administrative burden. Trade-offs have also to be carefully considered for
measures under objective 3 to address the risk of shortages while reducing regulatory burden. Trade-
offs between achieving access (objective 3) through possible costs of additional market launches and
affordability (objective 2) may also be necessary. Trade-offs are also inherent in objective 2 between
rewarding innovative medicines and affordability often achieved by generic/biosimilar competition.
The specific objectives are consistent with the European Green Deal and Digital agenda principles
and with the right of access to preventive health care and the right to benefit from medical treatment
set out in the EU Charter of fundamental rights
103
. In particular objectives 1 and 3 on innovation
including for unmet medical needs and on access to medicines will have a positive effect on the
access of patients to the medicines they need which relates to Article 35 of the Charter of
fundamental rights of the EU which establishes the right to benefit from medical treatment under the
conditions established by national laws and practices and a high level of human health protection in
103
https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=celex%3A12012P%2FTXT
28
kom (2023) 0192 - Ingen titel
2696964_0030.png
the definition and implementation of all the Union's policies and activities. Objective 4 which is
expected to reduce medicines’ residues in the environment from their manufacturing, use and
disposal is in line with the objectives set out by Article 37 on environmental protection.
5
5.1
W
HAT ARE THE AVAILABLE POLICY OPTIONS
?
What is the baseline from which options are assessed?
The baseline is represented by the business-as-usual scenario, that is, the situation where no policy
changes are made.
The current system provides 8 years of data protection and 2 years of market protection for all
innovative medicines, to give time to developers to recoup their investment by delaying the entry of
generic or biosimilar medicines. Other incentives also exist in parallel that delay generic/biosimilar
competition (patent, SPC, orphan market exclusivity, paediatric protection extensions), usually
offering a longer than 10-year protection if a medicine is eligible. However, the regulatory data and
market protection is the broadest in terms of eligibility, as it applies to all innovative medicines, and
it is almost impossible to infringe it
104
.
The current legislation also provides an additional 1 year regulatory protection for a new indication
with significant clinical benefit, allowing thus a maximum of 11-year protection. The revision does
not consider changing this incentive. Therefore, this incentive is not presented in the options.
Currently, there are no special incentives or obligations for the development of new antimicrobials
or prudent use of existing ones, nor for conducting comparative clinical trials.
There are no incentives or obligations on MAHs to place their products on the markets that do not
offer a sufficient business case. In essence, even when receiving an EU-wide marketing
authorisation, a company is completely free to choose where and when it will market its product.
There is no predictability for Member States who have no way of obliging the company to initiate
negotiations for pricing and reimbursement. The steps from a medicine’s marketing authorisation to
access and the influencing factors are described in Annex 14. There is no requirement for MAHs to
be transparent about public contribution to R&D costs either.
With regard to shortages, the current system focuses on notifying supply disruptions; it obliges
MAHs to notify competent authorities 2 months in advance if they expect a temporary or permanent
withdrawal of a medicine. Moreover, MAHs and wholesalers have to ensure appropriate and
continued supplies of medicines, however without effective means to enforce the obligations.
The ERA is the main mechanism within the current legislation for addressing environmental impact
of pharmaceuticals. It is required for all new MA applications and covers the environmental risks of
the use, storage and disposal of pharmaceuticals. It does not include environmental effects of
manufacturing. While it provides data to assess the impact of medicine residues released into the
environment, there are gaps in timely enforcement and possible risk minimisation measures.
SMEs have a fundamental role in the development of medicines. According to a recent report from
IQVIA
105
, emerging biopharma companies (defined differently than SMEs in the EU, but essentially
the same category) were responsible for a record 65% of the molecules in the R&D pipeline in 2021,
up from less than 50% in 2016 and 33% in 2001. The trend is that small companies dominate the
earlier development stages, which are not too expensive but very risky. Once the molecule reaches a
104
Before authorising a generic/biosimilar product, national competent authorities check against the data protection or
market exclusivity of the reference medicine and do only authorise the generic if these protections have expired.
105
‘Global Trends in R&D: overview through 2021,’ IQVIA Institute for Human Data Science, February 2022.
29
kom (2023) 0192 - Ingen titel
2696964_0031.png
certain maturity and still looks commercially promising, the SME typically partners
106
with big
pharma companies, which come in at the stage of the expensive late-stage clinical trials, marketing
authorisation and market launch that often require vast capital and global infrastructure.
5.1.1 Projections
The life sciences sectors continue to invest in and advance innovative therapeutics and vaccines, the
total number of products that are in active development globally exceeds 6 000, up 68% over the
2016 level.
107
Rich pipelines translate to more medicine authorisations, and we assume that the
current annual 30-40 authorisations of medicines with new active substances in the EU will expand
to 50-60 in the next 15 years. In our
dynamic baseline,
we will take the middle value at the middle
of the next 15-year period,
45 innovative medicines per year
to analyse the impacts of the various
policy measures proposed.
Against the backdrop of the overall positive outlook for innovation, research efficiency declines and
it costs more money and requires more failures to develop a new medicine
108
. Investments in R&D
are driven by commercial interest rather than public health needs, leaving important unmet medical
needs unaddressed. We expect that
15-20%
of the new innovative medicines, or
7-9 medicines per
year will address a real unmet medical need
without changes to the baseline, based on the current
ratio of accelerated assessments at the EMA
109
.
According to WHO, drug-resistant bacterial diseases already cause at least 700 000 deaths globally a
year, including 230 000 deaths from multidrug-resistant tuberculosis, a figure that could increase to
10 million deaths globally per year by 2050 under the most alarming scenario if no action is taken
and no new antibiotics are developed and authorised.
Regarding access to medicines, a IQVIA survey
110
shows no major improvement over the last year,
with a
90% variance
between Northern and Western European countries and Southern and Eastern
European countries
in terms of patient access to new medicines,
which also largely
corresponds
to the launch patterns according
to market size and purchasing powers
described in section 2.1
and Annex 14 due to pricing and reimbursement policies. The average delay between market
authorisation and patient access can vary by as much as a factor of seven across EU, from as little as
4 months to 29 months. Maintaining the baseline would likely conserve the problem at today’s level.
Available evidence suggests that across the EU the frequency of shortages and their impact on
patients and healthcare providers is increasing
111
.
If no changes are made to current requirements, the effect of the ERA to manage environmental
risks would remain limited. The main effect to reduce medicines in the environment should come
from environmental legislation.
5.2
Description of the policy options
In order to respond to the specific objectives, we considered more than 70 potential policy measures
deriving from the consultation process and initial analysis. These measures were organised around
nine policy blocks reflecting the objectives of the revision and its broad scope
112
.
106
Big pharma may acquire the rights for the product, the whole company, or they develop, authorise and market the
medicine in a joint partnership (e.g. the Pfizer-BioNTech COVID-19 vaccine).
107
‘Global Trends in R&D: overview through 2021,’ IQVIA Institute for Human Data Science, February 2022.
108
idem
109
Annex 5 – Evaluation SWD, p.22
110
EFPIA Patients WAIT Indicator 2021, see:
https://www.efpia.eu/media/636821/efpia-patients-wait-indicator-final.pdf
111
European Commission, Directorate-General for Health and Food Safety, Future-proofing pharmaceutical legislation :
study on medicine shortages : final report (revised), 2021, https://data.europa.eu/doi/10.2875/211485
30
kom (2023) 0192 - Ingen titel
2696964_0032.png
In a second step, taking into account the preliminary evaluation findings, we designed the three
high-level options which represent alternative ways to reach all the objectives of the revision. Each
option is constructed around specific underlying principles behind the grouping:
-
-
-
Option A builds on status quo and achieves the objectives mainly through new incentives;
Option B reaches the objectives through more obligations and oversight;
Option C adopts a ‘quid pro quo’ approach in the sense that positive behaviour is rewarded
and obligations are only used when there are no alternatives.
Each option contains pivotal and non-pivotal measures. Non-pivotal measures are complementary to
the pivotal ones and form an integral part of the policy options. A thorough multi-criteria impact
analysis for each policy measure, based on data, literature review and stakeholder feedback can be
found in Annex 11.
113
Finally, the options are complemented by horizontal measures. Contrary to
the non-pivotal measures, they apply across the board and deliver on simplification and innovation.
The IA report focuses on the ‘pivotal’ measures and the ‘pivotal horizontal measures’. These pivotal
measures were selected on the basis of the magnitude of their impacts and their political importance.
Table 1
shows how the pivotal measures map on to the specific objectives.
112
Directive 2001/83/EC merged 11 prior directives related to medicinal products, and together with the Regulation
(EC) No 726/2004, consists of 220 articles, offering numerous “levers” to adjust the policy.
113
To give an example, a pivotal measure to support market access is making the last 1 or 2 years of regulatory data
protection subject to market launch in all EU countries and this is discussed in the main body of the IA. Access in all
Member States will be supported by other measures, such as facilitating multi-country packs to make launches in smaller
Member States easier, but those measures are rather considered in Annex 11.
31
kom (2023) 0192 - Ingen titel
2696964_0033.png
5.2.1 Tabular overview of policy options
Table 1
Mapping of
pivotal elements
to the
specific objectives
Objective
Promote innovation, in
particular for unmet medical
needs.
Baseline
8 years DP +2 years MP
Option A
8 years DP +2 years MP
Special incentive:
+1 year DP for medicines that address UMN
+6 months DP to include comparative trials
Digitalization, simplification elements from horizontal
measures
Transferable exclusivity vouchers for antimicrobial
products
Baseline + additional rewards for innovation and access.
Comparative trials may lead to public cost savings.
Option B
6 years DP +2 years MP
Special incentive:
+ 2 years DP for originators that address UMN.
Digitalization, simplification elements from horizontal
measures
Option C
6 years DP +2 years MP
Special incentive:
+1 year DP for medicines that address UMN
+ 6 months DP for comparative trials
Digitalization, simplification elements from horizontal
measures
Transferable exclusivity vouchers for antimicrobial products
Incentives to promote the
development of novel
antimicrobials
Create a balanced system for
pharmaceuticals in the EU that
promotes affordability for health
systems while rewarding
innovation
No special incentives for the
development of antimicrobials
Generic and biosimilar entry after
DP/MP periods are over providing a
predictable framework for
competition from generic and
biosimilar medicines.
Pay or play model for antimicrobial products
Earlier entry of generics and biosimilars with 2 years
shorter protection than baseline
+2 years MP for medicines with no return on investment.
Require public transparency on any relevant public
contribution or funding, including of research and
development costs
Obligation to place a centrally authorised medicine on the
market in the majority of MS (small markets included)
within 5 years
If market launch condition not met, earlier entry of generics
and biosimilars
Require transparency on public contribution to R&D costs
in relation to clinical trials included in the MA application
Comparative trials may lead to public cost savings.
+2 years (or 1) DP extension if medicine is placed on all EU
markets within 2 years of authorisation and appropriately
and continuously supplied
Ensure access to innovative and
established medicines for
patients with special attention to
enhancing security of supply
across the EU
Currently no obligation or incentive
to launch in a particular or group of
MS
+6 months additional protection period if centrally
authorised product is placed on market in all MSs within
6 years of the MA (milestone incentive); and allow
generic competition if not launched in majority of MS
within 5 years of MA (disincentive)
Notification requirement same as in baseline
Obligation to notify a withdrawal 2
months before the interruption in
market supply of the product
Notification requirement same as in baseline
Improve data on medicines shortages, through adequate
notification periods for withdrawals and serious shortage
risks; shortage prevention, increased transparency of the
supply chain, mitigation plans for all medicines and
stockpiling of critical medicines Monitoring of shortages is
reinforced with a mechanism of information exchange
between MS.
Same as option B with the inclusion of AMR aspects in
GMP.
Reduce environmental impact of
the pharmaceutical product
lifecycle
An ERA is required for all new MA
applications. Potential risks from
medicines to the environment are
assessed by regulators and
precautionary measures are taken
Not applicable / non legislative
measures
Same as baseline ERA
Strengthen the conditions of use for medicines and ERA
requirements, including the assessment of the environmental
risk of manufacturing and its impact to AMR
Reduce regulatory burden, and
provide a flexible regulatory
framework
Horizontal measures*
*The horizontal measures are applicable to all options,
for details please refer to section 5.2.5.
Horizontal measures*
Horizontal measures*
Notes: AMR=antimicrobial resistance; DP=data protection; EMA/HMA= European Medicines Agency/Heads of Medicines Agencies; ERA= environmental risk assessment; GMP=good manufacturing practice; MA= marketing application; MP=market protection;
MS=member state; R&D=research and development; UMN=unmet medical need
32
kom (2023) 0192 - Ingen titel
2696964_0034.png
5.2.2 Policy Option A
Option A addresses the identified problems through
incentives
rather than setting further obligations
coupled with a stronger enforcement of existing obligations and information requirements.
To stimulate
innovation,
Option A maintains the current system of regulatory incentives (8 years
data + 2 years market protection), supplemented by a targeted incentive, an additional 1 year of
regulatory data protection for products addressing unmet medical need (UMN). Clarifications of the
scope and new definitions should facilitate innovation. It also foresees the introduction of a new
incentive for the conduct of comparative trials,
which bring a more robust evidence base for the
assessment of effectiveness of new treatments and facilitate decision-making downstream in the
lifecycle of medicines.
Option A stimulates the development of novel
antimicrobials
that can fight resistant pathogens
through
transferable exclusivity vouchers.
A transferable regulatory protection voucher
(transferable exclusivity voucher) allows the developer of a novel antimicrobial that reduces AMR
to benefit from an additional year of RP on another product in their portfolio or sell the voucher to
another company. This is a measure supported mostly by industry as a way to underpin the
substantial R&D costs of bringing new classes of antimicrobials to the market
114
. This will be
supported by measures on harmonisation of the summary of product characteristics for nationally
authorised antimicrobials to support good prescription practices.
Option A promotes patient
access
with a 6 month regulatory data protection incentive if a product is
placed on the market in all Member States within 5 years of MA. The rationale behind the measure
is that MAHs can be encouraged to increase the number of markets in which they launch products or
accelerate the timeframe within which they do so, by offering them a reward in exchange.
Measures on
security of supply
retain the current requirement for notifications of withdrawals (at
least two months in advance).
The current
ERA requirements
continue with an additional obligation to include the information on
the environmental impact of supply chain actors in the application dossier. The latter proposal is part
of the package of suggestions to support quality and manufacturing aspects (QMC) for medicines.
Among the
non-pivotal measures
of Option A are a non-binding system for scientific assessment of
evidence for repurposing off-patent medicines to include new indications for allow for innovation,
measures to facilitate multi-country packs to enhance access and inclusion of new manufacturing
methods into the framework to both ensure best quality manufacturing and to cater for innovation.
5.2.3 Policy Option B
Option B uses
more obligations
to address the specific objectives rather than incentives. This option
explores stronger monitoring mechanisms and increased obligations with interventions at different
milestones in the lifecycle of a medicine to foster patient access, affordability and security of supply.
To stimulate
innovation,
especially for unmet medical needs, it introduces a modulated
system of
incentives,
with a reduction in the current standard regulatory protection periods. The new standard
protection
115
for all originator medicines would consist of 6-years data protection and 2-year market
protection. New originator medicines with a demonstrated ability to address UMN would benefit
from an additional 2 years of data protection, thus maintaining the current baseline. Other medicines
will be entitled to strengthened protection only if they can demonstrate no return on investment in
view of investment costs, including for research and development.
114
115
Previously explored in the Joint Action on Antimicrobial Resistance and Healthcare Associated Infections.
Baseline protection is the current regulatory protection of 8 years of data protection and 2 years of market protection
which also applies in Option A; (new) standard protection is the regulatory protections of Options B and C of 6 years of
data protection and 2 years of market protection.
kom (2023) 0192 - Ingen titel
2696964_0035.png
Option B also encourages the development of novel
antimicrobials
that can fight resistant
pathogens through a ‘pay or play’ model. Either a company holds an antimicrobial in its portfolio, or
it pays into a fund for financing the development of novel antimicrobials. It also includes measures
for prudent use of antimicrobials including monitoring consumption, optimising package sizes and
stricter rules for the use and disposal of antimicrobials for human use and tightening of prescription
requirements for example through the mandatory use of diagnostics prior to prescription of
antimicrobials thus target pathogens better.
Access
measures in Option B consist primarily of an obligation to launch centrally authorised
medicines on the market in a majority of Member States (small markets included) within 5 years. If
the obligation is not fulfilled, the medicine loses its protection, and generics can enter the market.
Measures on
security of supply
encourage EU coordination for exchange of information and use
existing guidelines and systems, such as the EU medicines verification system
116
to track supply, and
measures to increase manufacturers’ responsibilities to ensure supply. The notification period for
withdrawals remains identical to the baseline and MAHs are obliged to offer their MA for transfer to
another MAH in case of withdrawals from the market.
The
ERA requirements
and conditions of use for medicines are strengthened. This option also
foresees the assessment of the environmental risk of manufacturing in the ERA as part of the
marketing authorisation. Moreover, it proposes improving oversight of sites through modification of
rules on inspections and a mandatory joint audit scheme for national GMP and GDP inspectorates.
Non-pivotal elements
in Option B include the possibility for regulators to impose a post-
authorisation obligation for comparative studies on the effectiveness of a given medicine compared
with the standard of care. Codification of rolling reviews beyond crisis-related medicines, and
measures to future-proof the regulatory system by reviewing the scope and definition of products
that need to be accommodated under the pharmaceutical legislation and simplifying/clarifying the
regulatory framework for certain categories of medicines (e.g. borderline products) should facilitate
innovation. Anti-competitive practices such as introducing multiple marketing authorisations are
restricted, interchangeability of a biosimilar medicine with its originator medicine will be elaborated
in the product assessment and the Bolar exemption (legal exemptions from patent infringements for
acts relating to the regulatory submission of testing data) will be broadened to facilitate generic
entry. Together with obligation for all MA applicants to publicly disclose any relevant public
funding received (R&D
transparency)
this should address
affordability.
5.2.4 Policy Option C
Option C proposes a ‘quid pro quo approach’ with a modulated system of
incentives combined
with obligations.
The regulatory protection for originator medicines in option C is split into a standard and a
conditional period. The standard is 6 years data protection and 2 years market protection (as in
option B) while the conditional period is 2 years (or 1 year, see box below with a variation of the
option). The conditional year/years are granted only if the product is placed on all EU markets
within 2 years of authorisation and appropriately and continuously supplied thus increasing
access
to
patients. To be pragmatic, the provision has some exemptions (e.g. the possibility for a Member
State to waive
117
the obligation within its territory for the purpose of the incentive). For it to be
predictable for generic and biosimilar companies, a time limit is set (i.e. 2 years before the DP
expires) for a final decision on the prolongation or not. If a company fails to comply with the market
launch requirement, there will be earlier generic competition and increased
affordability for health
116
Directive 2011/62/EU of the European Parliament and of the Council of 8 June 2011 amending Directive 2001/83/EC
on the Community code relating to medicinal products for human use, as regards the prevention of the entry into the
legal supply chain of falsified medicinal products, OJ L 174, 1.7.2011, p. 74.
117
In the case that a MS does not wish to be supplied at that moment.
34
kom (2023) 0192 - Ingen titel
2696964_0036.png
systems
118
. Moreover, originator medicines addressing an UMN would receive an additional 1 year
of data protection to stimulate more innovation in areas of unmet patient need.
The system of special incentives in options A and C are similar but transparency on public
contribution to the costs of clinical trials will be required for all medicines in option C. There is a
special incentive (6 months) to stimulate developers to conduct comparative trials.
Incentives can
be cumulated,
however the total regulatory protection period is
capped at 11 years,
which is a
difference compared to Option A.
Variation to Option C
Option C aims at a balanced mix of obligations and incentives,
which in individual cases may result in a higher level of protection
for companies than the current baseline. To mitigate this result, a
variation
119
to Option C is assessed, where no medicine could reach
a ‘beyond-baseline’ level of protection. The variation consists of a
reduction of the conditional
2 years
protection period to
1 year,
and
a capping of cumulated incentives at 10 years.
The next sections will consider Option C with 2 years conditional
period as default. The differences in impacts between the default
option C and the variation are discussed in section 8.1.
Variation to Option C
6 years DP +
1 years DP if placed
in all EU markets
+2 years MP
Special incentives:
+1 year DP for medicines that
address UMN
+ 6 months DP for comparative trials
Incentives capped at 10 years.
Transferable exclusivity vouchers for
antimicrobial products
With respect to
innovation,
the changes to the scope, definitions and classification advice with
regard to medicines and the codification of rolling reviews and PRIME would be similar to option B.
However, this option also foresees the inclusion of a sandbox environment (i.e. a structured form of
testing before formal regulation) which would more readily accommodate innovation in
breakthrough areas where the current framework does not sufficiently cater for this innovation. A
binding system for scientific assessment of evidence for repurposing off-patent medicines will be
established, and obligations will be simplified to facilitate non-commercial entities (e.g. academic)
to become marketing authorisation holders. To incentivise development of novel
antimicrobials
that
can fight resistant pathogens, a system of transferrable exclusivity vouchers (as in option A) is
explored. The fight against AMR is corroborated with a strong emphasis on prudent use measures
which are similar to those proposed in option B.
With respect to
security of supply,
in addition to an EU definition of shortages, critical shortages
and critical medicines, option C measures include a balance of EU- and Member State-level actions
to mitigate and prevent
shortages
and build on the shortage provisions in the EMA reinforced role
legislation
120
. The approach to reporting shortages is harmonised across the EU, while monitoring of
supply remains with Member States and only critical shortages are escalated to EU-level. As with
option B, support to the management of shortages is increased through earlier, harmonised reporting
on shortages. There is the possibility of information sharing by Member States on critical shortages
and supply chain vulnerabilities.
The
ERA requirements
are similar to option B. It would also strengthen conditions of use of
medicines on a case by case basis to limit the environmental impact without affecting the
118
An alternative consequence could be repealing marketing authorisation of companies not launching in all EU,
however this would deprive patients’ access to the concerned medicine, hence this measure was discarded.
119
During the evaluation several stakeholders from patients’ groups and academia argued that incentives are overly
generous within the EU.
120
Regulation (EU) 2022/123 of the European Parliament and of the Council of 25 January 2022 on a reinforced role for
the European Medicines Agency in crisis preparedness and management for medicinal products and medical devices, OJ
L 20, 31.1.2022, p. 1.
35
kom (2023) 0192 - Ingen titel
2696964_0037.png
appropriate therapeutic use. It will include AMR aspects in GMP to allow a more holistic assessment
of environmental risk along the pharmaceutical lifecycle.
With regard to
non-pivotal elements
121
, this option foresees stronger oversight of manufacturing
supply chains through changes to inspections, reinforced Member State inspection capacity (joint
audits of inspectorates) and increased EMA coordination. The strengthened Bolar provision to
promote competition and hence
affordability
listed in Option B is retained and the transparency
obligation on public funding is limited to clinical trials. Improvements to the current Hospital
Exemption will continue allow for the use of ATMPs without marketing authorisation, but under
stricter conditions to ensure quality, safety and efficacy of these therapies.
Transferable exclusivity vouchers and restrictions
on their granting and use
The transferable exclusivity voucher is a tool to
generate funds for the development of novel
antimicrobials.
The analysis in section 6.1.1.4 points to the conclusion that even though vouchers
can be an expensive solution, they represent a credible measure against AMR if applied under strict
conditions; their benefits and costs need to be weighed against the cost of inaction and the impact of
AMR on health and economy
122
.
By setting strict criteria for antimicrobials that can benefit from the voucher, its value would be
calibrated to benefit the developer of the antimicrobial more than the buyer. The analysis in section
6.1.1.4 explains why vouchers can work only if they are very restricted to a limited number (i.e. max
1 per year). This is also the reason why they score differently in the impact assessment for orphan
and paediatric medicines where such limitation is not possible (see details in Annex 4).
To achieve strict limitations, only those medicines that are ‘game changing’ antimicrobials for
reducing AMR can receive ‘novel
antimicrobial’ status
by the Agency, based on clear criteria set
out in the legislation. The antimicrobial is considered novel, and thus eligible for the voucher if
preclinical and clinical data underpin a significant clinical benefit with respect to antimicrobial
resistance and it either represents a new class of antimicrobials or it has a new mechanism of action
that is distinctly different from the mode of action of any authorised antimicrobial (criteria to be
assessed by qualified experts). Moreover, the active substance should not have been previously
authorised in a medicinal product in the EU that addresses a multi-drug resistant infection or a
serious or life threatening infection. This will also direct investment and research into those game
changing products. Even if found eligible, additional supply requirements, transparency conditions
on funding received and on the sale or transfer of the voucher and other conditions will be set in the
legislation.
There would be moreover a review clause in the legislation to evaluate the application of the
vouchers after some years and decide on the continuation or not of the measure. It may take some
time until an antimicrobial is authorised that is eligible for a voucher, a voucher may not be used
immediately after it has been granted and the effect of the extension of data protection due to a
voucher may also take some time to be seen. Several vouchers have to be granted and been used to
gain sufficient experience for a review of the measure.
5.2.5 Horizontal measures
All options are complemented by a series of horizontal measures. These are necessary to improve
the effectiveness and efficiency of the regulatory system overall and will act on core elements of the
authorisation and lifecycle procedures. They respond to the specific objectives of
innovation,
and
reducing the regulatory burden and providing a flexible regulatory framework.
Generic marketing authorisations will be simplified by enabling a common assessment of
manufacturing data across products, as generic medicines often source active substances from the
121
122
See Annex 11 for details.
AMR-Tackling-the-Burden-in-the-EU-OECD-ECDC-Briefing-Note-2019.Pdf
36
kom (2023) 0192 - Ingen titel
2696964_0038.png
same site. A more efficient repeat use procedure
123
will be provided to reduce administrative and
cost/burden and prevent medicine shortages. Furthermore, the sunset clause and renewal of MAs
after five years will be abolished to simplify procedures. Likewise, the envisaged reduction in the
number of notifiable variations reduces the administrative costs incurred by MAHs and regulators.
Provisions of the legislation will be reviewed with regard to novel combined products (e.g. where
medicines are coupled with medical devices, software, or artificial intelligence). To address
shortcomings highlighted in the evaluation
124
the legislation will ensure complementarity with the
medical devices regulation/in vitro diagnostic regulation in relation to benefit/risk assessment,
responsibilities of the medicine developer, and joint scientific advice.
In addition, delinking the environmental risk assessment of medicines that contain or consist of
GMOs from the GMO legislation and replace it with GMO environmental risk assessment
requirements and procedures adapted to the specificity of medicines under the general
pharmaceutical legislation is considered, but these changes would not constitute a complete
derogation from the GMO legislation.
New concepts will be integrated, such as adaptive clinical trials and full use of health data (real
world evidence), applying the digital by default principle, notably through electronic submissions of
applications, variations to MAs and electronic product information. The provision of authorised
electronic product information for EU medicinal products would enable easier access to data
contained within the product information, taking into account needs of patients, consumers and
healthcare professionals, as well as the risk of digital exclusion.
The working methods of EMA and the European medicines regulatory network will be adapted,
especially with regard to the functioning of the centralised procedure and the decentralised
procedures, the use of expert assessment teams and multi-expert inspections teams to ensure a better
use of the available network resources. The evaluation also identified suboptimal coordination
between the EMA committees that duplicate work, create administrative burden and risking delays
especially in the assessment of medicines for rare diseases and for children
125
and ATMPs. An EU-
wide centrally coordinated process will be foreseen offering early dialogue and more coordination
among clinical trial, marketing authorisation, health technology assessment bodies and pricing and
reimbursement authorities for integrated medicines development and post-authorisation monitoring,
pricing and reimbursement.
6
6.1
W
HAT ARE THE IMPACTS OF THE POLICY OPTIONS
?
Economic impacts
The general pharmaceutical legislation rewards innovators through the
regulatory data and market
protection (RP).
By protecting data on the safety and efficacy of the product, RP guarantees that
during the data protection period no generic/biosimilar medicine can obtain a marketing
authorisation referring to the originator’s data. This effectively protects innovators from generic or
biosimilar competition
126
for 10 or 11
127
years after authorisation. In comparison with other
jurisdictions, the EU ranks high (see
Table 2).
123
124
See glossary.
See Annex 5. The evaluation showed the need for more clarity on roles and responsibilities and for a more integrated
approach in relation to scientific advice on medicines and medical devices.
125
SWD(2020) 163 final.
126
RP does not prevent companies willing to undertake their own clinical testing to seek marketing authorisation for the
same medicinal product if they do not infringe on any patents or SPCs. However, that would be rather costly for entering
a market, where the originator medicine is already present, and hence rarely occurs.
127
An extra year is granted for an additional indication with significant clinical benefit. Historically around 1 in 8
medicines qualify for that.
37
kom (2023) 0192 - Ingen titel
2696964_0039.png
Table 2
Basic regulatory protection periods for medicines globally
128
Country
Canada
EU
Switzerland
USA
USA
Israel
China
Japan
Protection
New Chemical Entity+ Market Protection
New Chemical Entity+ Market Protection
New Chemical Entity
New Chemical Entity (small molecule)
Biosimilar Application Approval Exclusivity (biologic)
Market Protection
New Chemical Entity
New Chemical Entity
Duration
6+2 years
8+2+1 years
10 years
5 years
4+8 years
6 or 6.5 years
6 years
8 years
In addition to RP, medicines are also protected by patents (20 years), SPCs (up to 5 year extension
of primary patent, but maximum 15 years from marketing authorisation), and medicines for rare
diseases also benefit from 10 years market exclusivity (+2 years if paediatric studies were carried
out)
129
. The patent and SPC protection start from the patent filing, and depending on the time until
authorisation they may offer longer or shorter protection than RP. It differs case by case which
instrument provides the longest protection period after entering the market, as demonstrated by
Figure 3
on a representative sample of 200 medicines. Medicines protected by patent or SPC not
only enjoy a longer protection, but on average they generate 2-3 times higher revenues than those
protected only by RP (Table 3).
Table 3
Medicines’ protection period and revenues by their last layer of protection
Last line of protection
Regulatory protection
Market Exclusivity
SPC
Patent
Grand Total
Number
of
products
69
12
95
23
199
Avg. protection
duration
10.1 years
10.7 years
14.3 years
16.7 years
12.9 years
Avg peak
annual sales
1
€ 158.7 m
€ 41.7 m
€ 368.3 m
€ 300.5 m
€ 268.2 m
We expect this ratio among protection
types to remain in the next 15 years,
therefore the changes to the RP would
concern around 1/3 (i.e. 35%) of the
new medicines,
which have a
23%
share among all originator medicine
sales in the EU.
Figure 3
– Ratio of medicines by the length of last layer of protection and type of protection
35%
We provide a conceptual model to explain the economic impacts of the changes in the RP, on the
different stakeholders. The model is based on the commercial lifecycle of a representative innovative
128
129
Data collection by Technopolis Group, 2022.
A diagram with the current regulatory and IP protections in the EU can be found in Annex 9.
38
kom (2023) 0192 - Ingen titel
2696964_0040.png
medicine, an analogue, for which RP is the ultimate protection. To create this analogue, historical
data
130
were examined, and the evolution of sales followed from market authorisation until
protection expiry, and 5 more years from then, along with generic/biosimilar sales,
Figure 4.
The
model uses normalised units to represent prices and volumes across different products, where 100 is
equal to originator’s peak sales, at year -1. It is assumed that the pricing strategy of the
manufacturers remain unchanged. The calculations were done based on the public, list prices (not
the actual, confidential prices).
Figure 4
Normalised sales and volume for products with 8+2 years of RP protection (baseline)
The SPC evaluation
131
highlighted that generic competition is not uniform across medicines. High-
sales medicines, small molecule medicines are more likely to be contested and by more competitors,
leading to quick erosion of the price and the innovator’s premium. On the other hand, biological
medicines, medicines for rare diseases and low revenue products are less likely to be contested,
resulting in slower price erosion, or even maintaining a monopoly position. To account for this
variability, the model considers the average evolution of sales volumes and values across all the RP-
protected medicines in a nine-year cohort, including those medicines that were not contested by
generics after protection expiry. The model represents well real-life at systemic level, even though
some medicines – for example, those that face a high number of competitors – might show a much
steeper erosion, whereas others might see persistently high sales after expiry in the absence of
competitors.
From year 0, the generic medicines enter the market with a lower price, carve out a growing market
share and force the originator to offer discounts
132
. The volume of generic medicines steeply
increases, partly because some users substitute the originator medicine with generics and partly
because the total volume rises with increased affordability. For health systems, the price drop
following generic competition means cost savings. In our analogue, the price drop is 50% on
average at year +5. The lower price extends eligibility and more patients and from more Member
States can have access to the medicine either in its original or generic form. Even with the 32% more
patients served at year +5, health systems pay 34% less than at peak sales in year -1.
To account for the impacts of modifying the RP, we use the above baseline and the 16 years
observation period, which we consider as the commercial lifetime of an RP protected medicine. This
allows to understand how the stakeholders’ positions change under the different scenarios.
Extending the protection allows innovators to seek longer monopoly rents, but it delays cost savings
130
A cohort of medicines approved between 2004 and 2011, where RP is the last defence. Further explanation of the
inputs used for the model is provided in Annex 4.
131
SWD(2020) 292 final.
132
The evaluation (Annex 5) found that originator products can maintain a 30% premium over their generic competitors.
39
kom (2023) 0192 - Ingen titel
2696964_0041.png
and broader access for the public and delays revenues for generic companies. Decreasing protection
has the exact reverse effect.
Profit, sales, cost, volumes – how we measure economic impacts for key stakeholders
For
health payers
we measure the impact of changes by the change in the
cost of medicines,
which can be directly
deducted from total sales of originator and generic medicines in IQVIA data.
For
patients,
we measure the impact of change by the change in the
volume of medicines.
The more the volume,
the more patients could benefit from therapy, either using originator or generic product. We will indicate the
monetary value of the volume difference as “Δ of patients treated (monetised)”.
For
originator
and
generic industry
the key measure of impact is
the profit
that they can realise from their
business operations.
There is no readily available dataset on profits but we have good data on sales (revenues) from the IQVIA database. By
deducting the cost of sales from the revenues, we can calculate the gross profit. The gross profit only includes the
variable costs of manufacturing and distribution, but not the fixed costs, such as R&D and investment in infrastructure.
In our model we distinguish three categories of revenues, each with a different margin of gross profits.
Protected originator sales:
this is the most profitable category during the protected period of new medicines.
Based on a sample of reports from publicly listed companies we apply a 80% gross profit margin on the
revenues (20% cost of sales)
Contested originator sales:
once generics enter the market, originator products are forced into price
competition. Still, originator products can maintain a price premium compared to generics albeit reduced thanks
to brand loyalty and strong sales force. We assume a 50% gross profit margin in this category.
Generic sales:
generic industry operates on a high volume, low margin basis. With low product development
risk, a lower profit margin can be sustainable. We apply a 33% gross profit margin on generic revenues.
6.1.1 Economic impacts of key policy measures
6.1.1.1 Special incentives through increasing regulatory protection (Option A and C)
To understand the economic impact of an increased regulatory protection (either offered for UMN,
comparative trials or market launch) we have added an extra year of protected sales to our model,
and analysed the gains/losses for the different stakeholders during the observed 16 years (Figure
5).
Figure 5 -
Normalised sales and volume for products with 8+2+1 years of RP protection
The longer protection translates into higher profits for the innovator but increases the costs for
patients and payers, and also delays revenues for generic manufacturers. Overall, payers, patients
and the generic industry share the burden of allowing longer streams of monopoly revenues to the
innovator, to compensate for extra costs occurred (comparative trial, market launch), or to reward
and incentivise innovation of high public health benefit (UMN). The exact monetary impact depends
on the length of additional protection, and on the number of medicines expected to benefit from a
certain incentive. Below we assess the special incentives one by one.
Special incentive: 1 year extension of RP for medicines addressing UMN (Option A, C)
This measure affects RP protected medicines as last protection, altogether 35% of all new medicines.
Of these we expect 15-20% to address UMN. Applying these rates on the 45 annual new authorised
40
kom (2023) 0192 - Ingen titel
2696964_0042.png
medicines as per our dynamic baseline,
on average 3 special UMN incentives per year are
expected.
It is worth noting that for orphan medicines too an incentive for high unmet medical needs
is foreseen, extending the market exclusivity period beyond the modulated RP protection for those
orphan medicines.
Table 4 – Impact of change of +1 year regulatory protection for UMN
1 year increase in RP
Originator gross profit
Generic gross profit
Cost to public payer
Patients monetised gains/losses
Patients + payer monetised gain/loss
Product level change
+€94m
-€13m
+€54m
-€28m
-€82m
Systemic change (3 medicines)
+€282m
-€39m
+€162m
-€84m
-€246m
Table 4 summarises the monetary gains and losses of the different stakeholders at a single product
level, and also at systemic level, counting with 3 incentives a year. For affected medicines, the
innovators’
gross profit will increase by €282m a year, and the incentives would increase the cost
for payers by €162m. Taking into account that some patients will not have access to the medicine
due to the sustained higher price, the total
cost will be €246m to the public.
In exchange for this public cost, the UMN incentive would directly reward investment in UMN
R&D and likely would have a spill-over effect: national and EU-level research and innovation
funding could be specifically channelled to UMN, and national pricing and reimbursement systems
could differentiate the UMN addressing medicines, making them even more viable commercially.
We expect that the incentive would attract more investment in UMN and
result in 1-2 additional
UMN medicines per year,
for the benefit of the patients and creating savings for the health systems.
This important and non-monetised
133
benefit has to be seen together with the costs.
The consultations showed that both public authorities and patients support modulating the RP
periods around factors such as UMN. Industry on the other hand said that if incentives were limited
to UMN only, that would disregard the reality of science and incremental innovation and would
introduce uncertainty for businesses as the ultimate duration of the regulatory protection period
would not be fully clear when their investment decision is made
134
.
Special incentive: 6 month RP extension for comparative clinical trials (Option A, C)
Similar to the previous incentive, this measure could benefit medicines for which RP is the last layer
of protection, making around
35% of all new medicines eligible.
Conducting comparative trials
may not be feasible for some medicines, and if the cost of the comparative trial is too high as
opposed to the reward, companies will decide to decline the incentive. Taking these factors into
account, we expect that half of the RP products or
8 medicines annually
could benefit from the
incentive. Table 5 shows the economic impacts on the main stakeholder groups of this incentive both
at individual product level and at systemic level, for the 8 medicines per year.
Table 5 – Impact of change of +6 months year regulatory protection for comparative trials
6-month increase in RP
Originator gross profit
Cost of comparative trial for originator
Generic gross profit
Cost to public payer
Δ of patients treated
(monetised)
Product level change
+€47m
+€35m
-€6.5m
+€27m
-€14m
Systemic change (8 medicines)
+€378m
+€280m
-€52m
+€218m
-€112m
133
Monetising the benefits of an additional new medicine has several challenges: there is a large variation between
medicines’ value, defined by the patient population and severity of disease. Moreover, monetising a medicine’s value
requires putting a monetary value on patients’ life and health, as well as on the physical and emotional burden of their
families and carers. We thus have chosen not to monetise these impacts, but quantify them as much as possible.
134
See Annex 14 for further details on the factors influencing access and affordability.
41
kom (2023) 0192 - Ingen titel
2696964_0043.png
Patients + payer monetised gain/loss
-€41m
-€328m
Comparative clinical trials have a cost. In the absence of publicly available data, we estimate the
cost of a comparative clinical trial at €20-50m (the model uses the middle value of the range),
referring to the paediatric trials as a benchmark
135
. Due to the revenue extending nature of the
incentive, higher sales medicines would have a higher compensation, independent from the cost of
the trial.
For the public, 8 trials a year would cost €328m, but at the same time it would generate important
non-monetised benefits: comparative trial data will enable public authorities making better informed
reimbursement decisions and saving cost down the line. Data from trials would also accelerate
pricing and reimbursement decisions, allowing faster access to patients.
In the consultations, industry stated that comparative data is already provided at authorisation stage
when possible and that some products (e.g. ATMPs, products for ultra-rare diseases) will not benefit
from this incentive. Patients and public authorities on the other hand supported comparative clinical
trials (even as an obligation in the case of the latter).
6.1.1.2 Decreasing standard regulatory protection (Option B)
A key feature to support affordability in Option B
136
is a decreased regulatory protection, from 8+2
years in the baseline to 6+2 years, except for a minority of medicines: UMN addressing medicines
and medicines with no return on investment can maintain 8+2 years RP.
To model for the change, we removed from our analogue the original year -1 and -2, enabling earlier
generic competition. To keep the same 16 years of observation period, we have added year +6 and
+7 in the model, which we assumed to be equal to year +5
137
(Figure
6).
Figure 6
- Normalised sales and volume for products with 6+2 years of RP protection
This measure would only concern medicines that have RP as the last layer of protection, about 1/3 of
the 45 new medicines. Out of this 15 medicines, 20% may be UMN addressing or low revenue thus
exempted from the measure. Some of the RP protected medicines are eligible for SPC protection
between year 8 and 10 from market authorisation, partially offsetting the RP reduction. Overall,
9-12
medicines may be affected
by the reduction annually.
Table 6
summarises the impacts at product
and systemic level for the different stakeholders.
Table 6
– changes between baseline and RP 6+2 per stakeholder
2 year decrease in RP
Product level change
% change
Systemic change (9-12 medicines)
The joint evaluation of the orphan and paediatric regulation estimates the cost of paediatric studies at €22m.
This section discusses Option B solely, the eventual loss of protection in Option C for some medicines not complying
with the access condition is discussed in 6.1.1.3.
137
More on the assumptions in Annex 4.
135
136
42
kom (2023) 0192 - Ingen titel
2696964_0044.png
Originator gross profit
Generic gross profit
Cost to public payer
Δ of patients treated
(monetised)
Patients + payer monetised gain/loss
-€188m
+€25m
-€107m
+€71m
+€178m
-15%
+56%
-6%
+5%
+9%
-€1.97 b
+€266 m
-€1.13 b
+€745 m
+€1.86 b
Compared to the baseline, affected
originators
would lose their two highest-revenue, most-
profitable years. The product would
lose 15% of its lifetime profits.
For the originators this sums
up to €2bn loss annually in gross profits from the EU. More than 75% of originators replying to the
targeted survey expressed a negative stance towards a reduction of protection period for products
that do not address an UMN.
On the other end, the measure would generate €266m additional gross profit for the generic industry,
and €1.13bn direct cost reduction for health payers. Thanks to the lower price, 5% more patients
could benefit from the concerned medicines and accounting for the extra patients served in a
monetised form, the total benefit for the public is €1.86bn, or 0.9% of the total EU pharmaceutical
expenditure. An additional benefit would be a higher proportion of UMN among newly approved
medicines, due to the relative higher reward.
Because of all the other co-existing protections (SPC, patent, market exclusivity), option B
would
leave 75-80% of new medicines unaffected. The saving for payers and patients, would be borne
by a dozen of medicines, which would lose 15% of their profits.
Apart from the imbalanced impact, the measure would have additional costs. With a lower reward,
some developers may decide not to enter the EU market, or delay entry and seek return on other
markets first. An estimated
€670m will be lost for innovation
138
that could benefit patients.
Even though in the consultation civil society organisations in principle supported a reduction of
regulatory protection, patients would pay the highest price for the lost innovation, in that their
medical needs could not be met. Innovation is important for health payers too if new products offer
cost-effective health solutions, and a continuous stream of innovative medicines is needed for the
generics industry for new business opportunities.
Would the RP reduction harm EU competitiveness?
A direct link between EU incentives and EU competitiveness is hard to establish because while the
incentives make the EU markets more attractive, they are agnostic to the medicines’ geographical
origin. Around 20% of new medicines authorised in the EU are from the EU, the others are mainly
from US, UK, Switzerland and Japan that are equally eligible to all EU incentives. Equally EU
based innovative companies can benefit from incentives elsewhere, if they sell their products there.
In June 2016, the Council requested the Commission to conduct an evidence-based analysis of the
impact of incentive mechanisms, notably SPCs. Two studies have been commissioned. One from
Max Planck Institute
139
questions whether the availability of patent or SPC protection affects
companies’ decisions to locate research facilities in one jurisdiction or another, emphasising that
other factors are likely of greater importance. The Copenhagen Economics study
140
argued that SPCs
could play a role in attracting innovation to Europe, pointing out that taxation, education, and other
factors are probably more significant in that respect.
138
139
20% of lost protected sales, the typical R&D rate of revenue for originator companies, calculated in Annex 4.
Max Planck Institute. Study on the legal aspects of supplementary protection certificates in the EU, 2018.
140
Copenhagen Economics. Study of the economic impact of supplementary protection certificates, pharmaceutical
incentives and rewards, 2018.
43
kom (2023) 0192 - Ingen titel
2696964_0045.png
6.1.1.3 Measures to improve market access (Option A, B and C)
All policy options address the challenge of unequal market access to new medicines across the EU
but with different measures. As all options modulate RP, they all would impact those medicines that
have RP as the last layer of protection, 35% of new medicines,
15-16 medicines a year.
Option A
offers a +6 months RP extension incentive for medicines launched in all EU markets within 5 years
of authorisation. Option B instead requires companies to launch their product in the majority of all
EU countries within 5 years, otherwise they lose their protection and generics are allowed to the
market. Option C requires market launch in all EU MS (except those not interested in the product)
within 2 years of authorisation as a conditionality to parts of the protection period. Complying
medicines would gain 2 years of conditional RP (or 1 year in the case of the variation of Option C).
We have also observed a strong correlation between a medicine’s peak sales and its access across
EU countries. The magnitude of the incentive or the loss of protection is commensurate to the peak
sales, meaning that for high sales medicines the motivation is very high to comply. Since high-sales
medicines are launched already in most of the markets, for them the compliance cost is small. The
opposite is true for low sales medicines.
Based on the size of the incentive (or potential loss in option B and C), the compliance is estimated
as the percentage of medicines fulfilling the market launch requirements. From this, the costs or
savings to the public have been calculated (Table
7).
For option A, we used the same model as for
the special incentive for comparative trials, but expecting that only the higher sales medicines would
comply, a higher average peak sales was used in the model (detailed in Annex 4).
In option B and C the concept is reversed. If a medicine complies with the requirements, the
stakeholders’ position do not change. But non-complying medicines would face earlier generic
competition, resulting in losses for originators and in gains for the public and generics. To calculate
public savings stemming from non-complying medicines we used the model of the decreasing
standard regulatory protection (section 6.1.1.2). Again, the average peak-sales value was adjusted,
assuming that the low-sales medicines will be the ones not complying.
Table 7
– Comparative table of measures improving access
Option
Option A
+6 months, if in all
EU
Option B
-5 years, if not in
majority of MS
Option C*
-2 years, if not in
all EU
Expected compliance
50% (6-8 medicines)
75% (11-13 medicines)
Majority of markets
Originator’s reward/loss
+€527 m gross profit
+7.5% gross profit for 7
complying medicines
-€842 m gross profit
-34% gross profit for 4 non-
complying medicines
-€469 m gross profit
-15% gross profit for 5 non-
complying medicines
Cost/benefit for public
+€455 m public cost
€681 m gain from non-
complying medicines
€444 m gain from non-
complying medicines
66% (10-12 medicines)
*
The differences in impacts between the default option C and its variation are discussed in section 8.1
To determine compliance we use assumptions and this inevitably carries uncertainties. Originator
industry is better off with higher compliance and worse off with low compliance, which then results
in profit losses. For the public, high compliance is the desired outcome, resulting in faster and
increased access. However, non-compliance lowers the cost by shortening the protection period and
thus contributes to affordability, also an improved outcome compared to the baseline.
The access measures benefit society, above all patients. These benefits are elaborated in the social
impacts section (6.2). Option B has the disadvantage that it is unpredictable. Until reaching 5 years
on the market, the generic industry will not know for sure whether the originator medicine complies
or not. If generic companies prepare for non-compliance, and start development and production, the
innovator’s compliance would delay their entry by 3 years. And in case of non-compliance without
the generic companies being prepared, there will be no generic competition for quite some time,
neutralising part of the expected impact of the measure.
44
kom (2023) 0192 - Ingen titel
2696964_0046.png
Practical details and impact of modulation of data protection for market launch (option C)
The access conditionality would be a first-of-its-kind policy measure that addresses a problem
specific to the EU, and the primordial goal of it is to increase and accelerate EU patients’ access to
new medicines, regardless of the country they reside. The measure is successful if it is widely used
and a high proportion of new MAHs comply with the requirements and benefit from the incentive. A
low success rate would discourage companies and would not achieve access in all Member States.
Lack of access in a particular Member State can have many reasons. Sometimes companies decide
not to launch or delay launch in a market because of low profitability, small patient populations,
perceived cumbersome procedures, pricing policy, parallel trade. In other cases, Member States deny
access because no therapeutic value is seen, the medicine is not cost-effective according their HTA
assessment, or it would have an unbearable budget impact. There may also be objective roadblocks,
such as the need for highly specialised delivery infrastructure or diagnostic tools for the therapy that
do not exist in the Member State.
The proposed measure in option C targets companies to do their utmost to launch the medicine in all
EU markets within a specific period after authorisation (e.g. 2 years) and ensure a continuous
supply. This includes that companies shall file for pricing and reimbursement in all 27 Member
States, they have to conduct negotiations in good faith, and upon positive decision ensure supply that
covers the Member States’ needs
141
. However, companies could still receive the market launch
incentive if due to reasons beyond their control the market launch is delayed or not happened at all
(e.g. the Member State doesn’t wish to be supplied at that particular moment or doesn’t have the
specialised infrastructure e.g. in case of orphan medicines or ATMPs).
The Commission would grant the extra protection (2 years or 1 year for the variation to option C)
based on a system where Member States will be obliged to confirm within a certain period after
marketing authorisation compliance with the conditions of the incentive, justify a refusal by a
statement of reasons based on objective and verifiable criteria or give a waiver to the company. Non
reaction of a Member State will be considered as tacit confirmation of compliance.
Companies should not find it difficult to comply with the conditions of this incentive, as EFPIA
already made a voluntary commitment
142
: their members would file for pricing and reimbursement
in all EU27 Member States within 2 years from authorisation. This is already a step forward from
the current situation, but it is voluntary, restricted to EFPIA members and there are no controls in the
system. Hence, it does not work to the extent of the incentive, which relates to actual launch and
supply not just filing. The proposed measure adds a significant financial incentive for complying,
and it can also prevent dishonest applications
143
. By making ignoring certain markets or abusive
negotiating practices very expensive, Member States, and especially smaller Member States would
have a more balanced position when dealing with global firms.
The instrument to work adequately would also require Member States to act timely and in good
faith, because if compliance is made unduly difficult and unpredictable, the access goals will not be
met. Considering the common goal of both industry and Member States to ensure wide patient
access in the EU, we expect this change to contribute positively to the negotiations between the two
parties and that blocking the incentive will indeed be reserved to the objectively justified situations.
Ultimately, any alleged abusive behaviour can be subject to judicial control at Member State level
and a revision clause could be built in to take stock of performance after a certain time.
141
The Transparency Directive allows 180 days for Member States to make their pricing and reimbursement decision,
therefore filing at 18 months shall allow a market launch in 2 years.
142
EFPIA
Access to medicines (efpia.eu)
143
We have seen examples in the past that a small member state was offered 4-6 times higher price than Germany.
45
kom (2023) 0192 - Ingen titel
2696964_0047.png
The specific situation of
SMEs and not-for-profit entities
and their capacity to engage in multiple
parallel pricing negotiations will be taken into account by allowing longer period to comply with the
market launch conditions,
3 years from authorisation.
We can expect this measure to spur a
long term behavioural change
of both industry and public
actors to engage more towards increasing access, which is a strong demand from public authorities
and citizens. Ideally, launching new medicines in all 27 Member States in a timely fashion would be
the standard for all medicines, and not only for the 35% of them (with RP as the last layer of
protection) that are directly affected by the incentive.
Such incentive has not yet been tested on the market, however stakeholders were willing to share
their views about it. Public authorities in the targeted survey and a workshop were overall positive to
linking incentives with market launch, while industry was against. For industry, access depends on
factors that are not under their control (e.g. variations in national reimbursement decisions);
however, they agreed that the measure can be a financial incentive to launch in smaller markets. To
address this concern, the design of the measure includes the safeguards explained above. Civil
society organisations, patients, researchers and public authorities considered this measure as very
important. They stressed the need to provide ‘real’ effective access and continuous supplies. Some
public authorities argued that this measure should be an obligation. Member States have highlighted
in a series of Council conclusions
144
that incentives need to be proportionate to the goal of
encouraging innovation while improving patients' access to innovative medicines. They considered
that deferred or missed market launches, and business behaviour, including high priced essential
medicines pose a high burden for patients and health systems. They called the Commission to
evaluate the system and take action.
Would a decreased protection translate into price increase?
Companies may try to increase prices to compensate for a shorter RP if they do not get the incentive,
however, this will result in lower volumes sold, less Member States and fewer patients could afford
the increased price. Rationally behaving companies should not have different pricing policies
because of the length of protection, a higher price does not automatically lead to higher profits
145
.
The Evaluation
146
compared prices of the top-selling almost 200 medicines in the EU, US, Australia,
Canada, Japan and Switzerland.
We could not find any correlation between the prices and data
protection periods,
however in the US prices for the same medicines are often 3-5 times higher
than in other countries despite offering very long effective protection
147
.
6.1.1.4 Measures addressing AMR (Options A, B, C)
Annex 15 describes innovative financing solutions – outside of the general pharmaceutical
legislation – introduced in some EU Member States and some international initiatives to incentivise
development of new antimicrobials.
Pay or play model (Option B)
144
Council conclusions on strengthening the balance in the pharmaceutical systems in the EU and its Member
Stateshttps://www.consilium.europa.eu/en/press/press-releases/2016/06/17/epsco-conclusions-balance-pharmaceutical-
system/;
Council conclusions on innovation for the benefit of patients:
https://eur-lex.europa.eu/legal-
content/EN/TXT/PDF/?uri=CELEX:52014XG1206(03)&from=SK;
Conclusions on strengthening the European Health
Union:
https://data.consilium.europa.eu/doc/document/ST-14029-2021-INIT/en/pdf
145
A recent and extreme example is the case of Zynteglo®, a gene therapy authorised in the EU in 2019. The company
insisted on a high price (more than €1m) that not even the richest markets were willing to pay, and led to zero sales and
zero profits in the EU market.
146
Notably the indicator AFF-1.2 on p100 of Annex 10, Analytical report.
147
On the other hand, more new medicines and much faster than in the EU are made available to US patients, at least for
those who can afford a premium insurance scheme.
46
kom (2023) 0192 - Ingen titel
2696964_0048.png
In this model, a company co-finances the innovation and either holds an antimicrobial in its portfolio
or it pays to a fund to finance the development of novel antimicrobials. A recent analysis
148
found
that a pay or play model would impose additional costs on EU pharmaceutical businesses with the
risk that the costs would be passed on to health systems (insurers and/or patients) through higher
prices and while a minority may look to avoid a levy by developing antimicrobials or acquire
businesses with an antimicrobial in the portfolio, the majority would likely view the surcharge as an
unavoidable cost to be factored into their wider pricing policies. In addition, the fund would generate
only limited amount of money so that only a limited number of rewards can be ensured. The results
of this model could be seen only after several years (when the fund collects enough capital).
The pay or play model would not directly increase the number of novel antimicrobials and may
increase prices of other medicines, creating substantial social costs. The benefits of the incentive
would depend on the use of the collective fund, which is beyond the scope of the general
pharmaceutical legislation.
This measure was supported by patients and other civil society organisations in the public
consultation. Industry was the least supportive, they raised concerns that the model would unfairly
penalise companies (particularly SMEs) with no expertise in AMR product development.
Transferable exclusivity vouchers for novel antimicrobials (Options A and C)
These would benefit in particular SMEs as they would be rewarded as early as regulatory approval
for a new antimicrobial. It would also increase the attractiveness of the field for private financing
mechanisms, such as venture capital. According to EFPIA
149
, the value of such voucher in the EU
should be between €280 m and €440 m per product, based on assumptions around a “fair European
share”, a proportionate contribution of the EU towards the development of a novel antimicrobial
product that would benefit the global population. The voucher could be an important part of the EU
response to AMR for the
development of novel antimicrobials,
i.e. not just products that are
already in the (weak) pipeline. Such response could also include other initiatives, outside the
legislation, such as joint procurement for antimicrobials under HERA to
guarantee revenue
paid to
producers
for ensuring access
to existing or new antimicrobials.
Cost and benefit of transferable exclusivity vouchers
To understand the impacts of a voucher, the model of RP extension has been used, with some
adjustments. The buyers and thus users of the vouchers would be companies that hold the products
with the highest sales among the RP protected medicines. The commercial lifecycle of these
products differs from the average, as their market is more attractive for generics/biosimilars. It
results in a faster erosion of price and sales, therefore an additional year of protection has a higher
value for the originator, and a higher cost for the other stakeholders. We have examined over a 10-
year period the highest selling RP protected medicines, and identified the champions for each year.
We used in our model a €545 m average peak annual sales for these champions (More details on the
model in Annex 4).
Table 8
summarises the effects to the various stakeholders.
Table 8
– Changes to baseline with the voucher and value of voucher
Stakeholder
Originator gross profit
Generic gross profit
Cost to public payer
Patients monetised gain/loss
Patient + payer monetised gain/loss
change
+€387 m
-€54 m
+€283 m
-€158 m
-€441 m
change %
+10.1%
-23%
+4.7%
-3.8%
-7.3%
148
149
(https://academic.oup.com/cid/article/71/8/1994/5736365?login=true).
Representative of innovative industry:
A new EU pull incentive to address Anti-microbial Resistabce (AMR)
Recommendations from EFPIA.
47
kom (2023) 0192 - Ingen titel
2696964_0049.png
The €545m gain of the originator in protected sales is not equal to the value of the voucher for the
originator, because the revenue contains the cost of manufacturing and distribution, as well as the
cost of capital. We assume that the originator can only use the voucher 2 years after buying it, to
ensure that generic competitors can prepare for a delayed entry. Assuming 20% cost of sales and
10% annual cost of capital over 2 years, the
value of the voucher for the originator is € 360m
at a
cost of € 441m for payers and patients
(or €283m in nominal value, disregarding patients’ loss).
Sharing the value of the voucher between buyer and seller
We were able to identify the likely average value of the voucher, however it remains uncertain what
proportion of the value will be transferred to the seller – the actual developer of the rewarded
antimicrobial, often an SME. The negotiating position of the seller will depend on the second
highest selling medicine, the next potential buyer, similar to an auction where the winner has to pay
only a little more than the second highest bidder. The situation is further complicated if there are
more vouchers on the market and the EFPIA paper estimates 1-3 vouchers per year. Each additional
voucher drives down the price for all vouchers in that year, as they generate competition for each
other. For instance, if there are 3 vouchers, the price for all will fall between the value of the voucher
for the 3
rd
and 4
th
best seller medicine. Using historic data on the second, third and fourth best-
selling RP protected medicines in a given year, we can visualise the impact. (Figure
7, Table 9).
Figure 7
Distribution of buyer and seller advantage if 1 or 3 vouchers issued a year
Table 9
– share of value among buyer, seller and the public
1 voucher
Seller rent
Buyer rent
Cost to public in nominal
value
Cost to public incl.
unserved patients
3 vouchers
€205 m
€154 m
€283 m
€441 m
Seller rent
Buyer rent
Cost to public in
nominal value
Cost to public incl.
unserved patients
Voucher
1
€89
m
€270
m
€283
m
€441
m
Voucher
2
€89
m
€97
m
€147
m
€228
m
Voucher
3
€89
m
€50
m
€109
m
€170
m
Total
€267 m
€417 m
€539 m
€839 m
In the model, based on historic sales data,
the buyer captures 43% of the voucher’s value
if there
is one voucher per year, and 61% if there are three vouchers annually. The buyer’s share is sensitive
to the gap in the voucher’s value between one buyer and the next. The smaller the gap, the higher
proportion of the value remains with the developer (seller). Appropriate safeguards and modulation
of the voucher system could potentially improve the buyer/seller value-sharing ratio.
The voucher not only generously rewards the buyer without merits, but the public has to pay a high
price to the developer. We present the cost for the public payer to reward the developer with 1€ in
Table 10
both in nominal value (the net budgetary effect for payers) and with a cost that takes into
account the lost volumes and thus unserved patients.
Table 10 -
cost for the public payer to reward the developer with 1€
Scenario
Cost to public in nominal value
Cost to public incl. unserved patients
1 voucher
1.38 €
2.15 €
2 vouchers
1.40 €
2.18 €
3 vouchers
2.02 €
3.14 €
48
kom (2023) 0192 - Ingen titel
2696964_0050.png
If it were possible to add safeguards, ensuring that 90% of the value of the voucher is captured by
the seller (developer), the ratio of the award and the cost would significantly improve. In this case, it
would cost €87 m to the health payers to give a €100 m reward, but this payer cost does not account
for the unserved patients’ loss
150
.
Regardless of the cost calculation method, the public has to pay more than 1€ for each euro awarded
to the developer. However, it would be a feasible way
to pool sizeable resources and incentivise
antibiotic development,
which so far have proven ineffective with other incentives. These costs
should be put on balance with the current
€1.5bn in health care costs and productivity losses from
AMR
151
and the risk from the high levels of antimicrobial resistance in bacteria from human
infections, a silent pandemic that is not subsiding, and its economic consequences. Benefits are
further detailed in the social impact section (6.2).
In the consultations, some civil society organisations concurred that company profits would rise as a
result of a transferable voucher and thus create an incentive to develop products to address the issue
of AMR. However, they recognised that if this is done the system should be fine-tuned to meet the
needs of patients. Others oppose this incentive as it would delay the entry of generics for other
medicines and could increase substantially the costs for public health systems. Alternative solutions
such as small milestone rewards or longer regulator protection periods should be considered
according to civil society organisations, public authorities, healthcare professionals and citizens. In
the public consultation, innovator industry defended the benefits of transferable vouchers. Public
authorities, civil society and the generics industry expressed opposing views about the voucher
citing arguments linked to overcompensation, high cost to health systems and loss of
competitiveness for generics.
Impact of prudent use measures
The use of smaller packages would enable more sustainable use of antimicrobials and less release of
unused antimicrobials in the environment. On the opposite side, it would increase manufacturing
costs and package waste. Stricter rules on prescription of antimicrobials and mandatory use of
diagnostics would impact prescription behaviour positively, however, it would also result in
switching from broader spectrum antimicrobials to more specific (and expensive) antimicrobials and
costly diagnostic tests. Requirements to adopt AMR lifecycle monitoring plans
152
would help the EU
reduce its overall consumption of antimicrobials and hence AMR. This measure would come with
some cost both to businesses and Member States, however the establishment of appropriate
mechanisms to share information with regulators could mitigate this burden.
6.1.1.5 Horizontal measures
153
The horizontal measures are intended to deliver wide-ranging improvements in terms of efficiency
and effectiveness.
Table 11
presents a qualitative assessment of the benefits of each of the 10
pivotal horizontal measures, rating the likely benefits – against the baseline – on a 3-point scale
(High, Medium, Low) for each stakeholder group. From this perspective, the most promising
horizontal measures – overall, for all stakeholder groups – are the proposals to improve the
governance of the European medicines regulatory network, the development of an integrated, pan-
EU data architecture for the regulatory system and an EU-wide, centrally coordinated process for
early dialogue.
150
151
Unserved patients refer to those patients that were not served due to the delayed entry of generics, i.e. the lost volume
201020_EUJAMRAI_policy-brief_WP7_appropriate-use-of-antibiotics-one-health-perspective.pdf (eu-jamrai.eu)
152
Such AMR lifecycle monitoring plan could cover stewardship, risk mitigation measures to limit AMR, report
resistance to the antimicrobial, educational material to inform more efficient use, monitoring and reporting on the use.
153
Detailed analysis of the measures are in Annex 11.
49
kom (2023) 0192 - Ingen titel
2696964_0051.png
Table 11 -
Qualitative assessment of the benefits of pivotal horizontal measures for key stakeholders
Business
Streamlining and de-duplication
#1 Streamlining of procedures
#2 More efficient RUP
#3 Efficient governance
Regulatory Network
of
the
European
Medicines
H
H
H
M
M
L
H
H
M
H
H
M
H
L
H
M
L
M
M
M
L
L
L
L
EMA
NCAs
SMEs
Health
Systems
Environ-
ment
#4 Facilitate more efficient interaction across regulatory
frameworks
Digitisation
#5 Legal basis to allow network to analyse real world
evidence
#6 Legal basis for setting up electronic product information
for medicines
#7 Electronic submission of applications
Enhanced support and regulatory flexibility
#8 Optimise regulatory support to SMEs and non-commercial
organisations
#9 Adaptation of the regulatory system to support the use of
new concepts
#10 EU-wide centrally coordinated process for early dialogue
M
L
H
M
M
H
H
M
M
H
L
H
H
M
L
M
L
L
L
H
H
M
M
M
L
M
H
H
H
H
H
M
M
L
L
L
Stakeholders’ views are more convergent vis-a-vis horizontal measures. Reducing regulatory burden
(e.g. through efficient governance of EMA committees and authorisation procedures, elimination of
the renewal procedure and digitisation) can be considered as common ground both for industry and
public authorities and improve the competitiveness of the EU as a global destination for businesses.
The introduction of electronic product information is supported by all stakeholder groups. For
healthcare professionals and patients it is important to keep paper package leaflets in certain cases to
ensure access to information for all patients. Member States want that the different national levels of
‘digital readiness’ are respected. The electronic product information will complement the current
paper package leaflet of authorised, statutory information for each medicine, though in certain cases
Member States could allow electronic product information only. It could have positive effects on
shortages and will be more appropriate to the EU’s multi-lingual environment. The electronic
product information will have a limited, positive environmental impact from reducing the number of
paper package leaflets and streamlining the logistics chain.
An EU-wide centrally coordinated process for early dialogue among authorities responsible for
clinical trials, marketing authorisation, health technology assessment, and pricing and
reimbursement will improve business predictability for companies (including SMEs). Such early
dialogues are expected to provide guidance to companies on evidence generation along the medicine
lifecycle. Clearer and more coherent evidence requirements will reduce uncertainty and investment
risks for developers of innovative medicines, in particular in areas of unmet medical need (where
developers often already face significant challenges due to the complexity of the diseases
concerned). Early dialogues can therefore contribute to guiding and steering the investment and
clinical development decisions of companies towards innovations with high added value for health
systems and patients. They will also ultimately contribute to timely patient access to innovative
medicines by providing clarity on evidence requirements of downstream actors for timely generation
of appropriate evidence, facilitating and speeding up their decision-making.
Overall, these measures are expected to generate net benefit of up to €100m a year, shared among
businesses and authorities (Annex 3) in the best case scenario.
50
kom (2023) 0192 - Ingen titel
2696964_0052.png
6.1.2 Option A – combined impact of the measures
Conduct of business: Retention of the current period of RP for all new medicines and special
incentives for UMN, comparative trials and EU-wide product launch would have a positive effect on
businesses that can benefit from the incentives. However, this would negatively impact the generic
and biosimilar industry as it would further delay their access to the market. Measures on security of
supply retain the current requirements hence they would bring no additional burden.
Public authorities: Incentives providing longer data protection periods in general (whether to
promote innovation or EU-wide market launch) would carry a significant cost to national health
systems and payers by delaying generic entry. There would also be additional administrative burden
for the EMA and NCAs involved in the assessment of the additional applications, UMN criteria and
verification of product market launch information to determine whether a MAH has fulfilled all the
conditions to be eligible for longer data protection. On the other hand, a special incentive for
comparative trials would offset an additional period of high prices for payers against a more robust
evidence base for HTAs and payers.
The high cost of a transferable voucher given to developers of novel antimicrobials would be borne
by healthcare payers. This cost needs to be considered in the context of the health costs related to
AMR and possible savings from novel antimicrobials to combat resistant bacteria.
Sectoral competitiveness, trade and investment flows: The special incentives for UMN, including the
transferable voucher and EU-wide market launch are expected to improve competitiveness and
attractiveness of the EU pharmaceutical sector, especially SMEs and support increased investment in
medicine development to address UMN and AMR respectively.
Research and innovation: The special incentives will support increased return on investment for
developers and bring additional investment into R&D for UMN, including AMR. Comparative trials
will contribute to better understanding the clinical benefits of a medicine and its comparators.
Functioning of the internal market: The slight increase in the number of new innovative centrally
authorised medicines owing to incentives and the increase in access to those medicines through the
market launch incentive will improve the functioning of the internal market. On the other hand,
delayed generic entry would hinder competition, and keep prices high for a longer period compared
to the baseline. Overall, option A would make more harm to the functioning of the internal market
than benefit.
Administrative burden on business: Changes to RP for medicines to make them contingent on
market launch should be expected to make the system considerably more complex. It will require
reporting by MAHs on market launches resulting in higher administration costs. The horizontal
measures however would significantly cut red tape.
SMEs: The transferable exclusivity voucher is intended to reward antibiotic developers that are often
SMEs. Thanks to the transferability, they can monetise the value of the voucher by selling it.
Fulfilling the conditions for the market launch incentive is more challenging for SMEs compared to
big companies that may have offices and staff in all Member States. As mentioned in the ‘SME test’
Appendix D of Annex 12, other measures in Option A present no major positive or negative impacts.
6.1.3 Option B – combined impact of the measures
Conduct of business: For originators affected by the reduced RP, the overall income and profitability
from new medicines would be significantly reduced (22% loss in commercial value). It may happen
that developers increase their prices or otherwise rebalance their portfolios towards those market
segments with greater commercial potential. The threat to EU-based originators will be offset to
some degree by giving a boost to EU’s generic industries, broadening their portfolios and potentially
creating a prime-mover advantage in global markets. Similarly, developers of products addressing
UMN will be exempt from the negative impacts of the measure.
51
kom (2023) 0192 - Ingen titel
2696964_0053.png
A pay or play model would impose additional costs on EU pharmaceutical businesses, and while a
minority may look to avoid a levy by developing antimicrobials or acquire businesses with an
antimicrobial in their portfolio, the majority would be likely to view the surcharge as an unavoidable
additional cost to be factored into their wider pricing policies.
Public authorities: Health payers may benefit from lower average lifetime costs for medicines due to
earlier generic entry (because of a reduced data protection period). The extent of these benefits will
depend on originators’ response to the reduced incentives, and it is possible that average prices will
be adjusted upwards to some degree to offset the shortened protection period.
Greater transparency around public support for medicines development may strengthen payers’
position when negotiating with MAHs, helping to place a downward pressure on prices and thereby
helping to maintain or improve access to medicines. Auditing the claim of developers demonstrating
the absence of return on investment can be time consuming for authorities; the global development
and the complex accounting systems raise questions on the overall feasibility of the exercise.
The measures to increase patient access to medicines are expected to improve the situation in
particular in smaller markets, and thus the cost-effectiveness of the health systems.
Creating the infrastructure and monitoring shortages will require a significant investment from
authorities. However, shortages avoided reduce the burden of finding substitutes or new suppliers.
Sectoral competitiveness, trade and investment flows: Reduction in the standard regulatory
protection could weaken the global competitiveness of EU based originators overall, compared with
the current situation. The reduction will affect equally all companies selling their products in the EU,
no matter where their R&D is placed. The proposed pay or play model and access obligation would
raise the cost of doing business in EU. This could affect the competitiveness of pharmaceutical
companies in EU relative to non-EU companies.
Research and innovation: The reduction of the regulatory protection would cause an estimated
annual €670m loss for R&D.
Functioning of the internal market: Earlier generic entry due to lowering of the standard data
protection period for most new medicines (except those addressing an UMN) and increase in access
to medicines through market launch obligations improve access to medicines and the functioning of
the internal market. Reduced number of new innovative medicines would offset parts of the benefit.
Administrative burden on business: For developers that need to demonstrate the absence of a return
on investment (ROI) from their R&D to secure a period of additional regulatory protection, there
would be increased administrative costs associated with the methodology that businesses would need
to follow. The transparency requirements would put an additional burden on companies. The
horizontal measures however (discussed in section 8) would significantly cut red tape.
Obligations on MAHs to place centrally authorised medicines on the market in a majority of
Member States may carry additional costs to the MAH that would have to bear the consequences of
the reduced regulatory protection. The MAH will also have to provide additional information to
regulators to demonstrate their compliance with obligations, raising costs. These obligations will
also increase the costs to MAHs for interacting with HTA bodies in the Member States.
Administrative costs would also be expected for AMR measures in relation to the pay or play model
and prudent use measure, e.g. monitoring of consumption.
SMEs: SME originators may find it more difficult to invest in riskier novel medicines given the
reduction in future returns on investment owing to reduction in the standard data protection period
and their relatively weaker market position when it comes to negotiating prices. On the other hand
SMEs could benefit from the UMN incentive as they are often willing to invest in more risky R&D.
Obligations for market launch in a minimum number of Member States, including smaller markets,
may be more challenging to meet for SMEs that do not yet have market presence.
52
kom (2023) 0192 - Ingen titel
2696964_0054.png
6.1.4 Option C – combined impact of the measures
Conduct of business: Under this option, companies will be able to obtain the same protection period
as in the baseline, but subject to compliance with certain conditions on which the eligibility for those
"conditional" periods depend. Access to additional incentives for market launch and supply in all
Member States, innovation for UMN and AMR as well as comparative trials will grant MAHs a
longer period of exclusive prices compared to the minimum period being introduced, representing
increased revenue and potentially changing behaviour of the sector. For companies not complying
with the criteria for the conditional periods, impacts to conduct of business will be similar to those
for Option B with reduction in overall income and profitability for new medicines. In addition,
generic companies have the opportunity to enter the market earlier when originators have not
fulfilled the RP prolongation conditions.
As regards shortages, submission of shortage prevention plans and additional reporting requirements
to increase transparency of the supply chain would be acceptable to industry stakeholders if the
information remains confidential, as this could be commercially sensitive. In consultations, industry
stakeholders have strongly opposed applying these measures to all authorised medicines rather than
limiting it to critical medicines and those medicines at high risk of shortage.
Public authorities: It is a win-win for public authorities, partly because their role in market launch is
strengthened and no longer depending on companies only. Either after a successful price or
reimbursement negotiation the medicine will become available to patients, or if there is no
compliance, the measure will allow earlier market entry of generics and biosimilars thus reducing
prices through generic and biosimilar competition. The strengthened role of Member States comes
though with increased responsibilities for timely decisions at national level. The special incentive for
comparative trials would lead to increased availability of such data to regulators at time of
authorisation and may provide a better evidence base for HTAs and payers.
There may be additional costs for the public authorities involved in the assessment of UMN criteria
and verification of product market supply to determine whether a MAH is eligible for longer data
protection. Similarly, an increase in notification period for withdrawals and shortages will increase
the complexity and administrative burden of monitoring shortages for Member States’ authorities,
although use of a common template and streamlined reporting for reporting could enable cost
savings in the long term. Monitoring of supply at Member State level is economically advantageous
for NCAs as it builds upon the existing system of national monitoring.
To support market launch of products in Member States, HTA, pricing and reimbursement bodies
would have to conduct a greater number of procedures, in a reduced time period. It is observed that
national pricing and reimbursement decisions for new medicines often take longer than the legally
maximum of 180 days.
154
This can be partly offset by the efficiencies in the new HTA regulation, in
particular better sharing of evidence on the therapeutic benefits of the treatment. Greater
transparency around public support for clinical trials would strengthen pricing and reimbursement
agencies’ negotiating position with MAHs.
Member States would have new burden from supplying marketing authorisation holders with
confirmations, refusals or waivers on the compliance with conditions for market launch extension.
For AMR, public authorities would need extra capacity to assess AMR lifecycle monitoring plans.
The EMA and NCAs may require additional capacity and expertise or incur greater administrative
burden in reviewing and assessing products based on the additional requirements for ERA and GMP
(AMR aspects).
Sectoral competitiveness, trade and investment flows: By providing additional incentives (UMN,
AMR, comparative trial) companies could get the same regulatory protection period as in the
154
The Directive 89/105/CEE sets a maximum period of 180 days. For compliance issues see e.g. SWD(2012) 29 final.
53
kom (2023) 0192 - Ingen titel
2696964_0055.png
baseline (8+2), and the EU pharmaceutical sector would remain attractive. In recent years, global
venture capital investment has seen accelerating growth driven by advances in drug research and
residual unmet need for which it is often easier to demonstrate value to patients/the healthcare
systems
155
. The conditional EU-wide market launch incentive would apply to both EU and non-EU
based companies, therefore the relative competitiveness of EU companies would not be driven
down. The greater obligations and requirements to monitor and prevent shortages (including
reporting and stockpiling requirements) and to address environmental challenges could affect more
the EU pharmaceutical sector, but these measures are proportionate to achieving the objectives of
security of supply of medicines at all times and reducing the environmental impact of
pharmaceuticals. The overall balance of the measures on competitiveness would still be positive.
Research and innovation: Impacts on research and innovation would be similar to Option A.
Functioning of the internal market: The increase in the number of new innovative medicines owing
to incentives and the increase in access across the EU through the market launch incentive will
improve patient coverage and functioning of the internal market. Transferable vouchers would delay
the start date of competition for the product to which the voucher is transferred, but the systemic
impact would be limited due to the low number of vouchers and products benefiting from them.
Administrative burden on business: Additional regulatory data protection period for medicines
contingent on appropriate and continuous supply will require MAHs to seek confirmation of supply
from Member States resulting in higher administration costs. Similarly, an increase in notification
period for withdrawals (12 months) and shortages (6 months) will increase the administrative burden
of reporting shortages for MAHs. Introduction of a common template for reporting withdrawals and
shortages could help reduce the additional administrative burden and promote harmonised data
collection. Keeping monitoring at Member State level will not lead to additional burden for MAHs
as it builds upon existing systems. MAHs will also incur greater costs due to requirements for
stockpiling and shortage prevention and mitigation plans for all medicines. The horizontal measures
however (see section 8) would significantly cut red tape.
Increased transparency around public support for clinical trials is narrower than the proposal under
Option B, where all aspects of public support for medicines development, including various tax
reliefs, have to be considered. Hence, this option would be simpler to implement as information on
support of specific clinical trials through publicly funded R&D grants is more easy to retrieve and
thus will incur less substantial administrative costs.
For AMR, prudent use measures would increase the administrative burden for businesses, e.g. for
AMR lifecycle monitoring plans. Strengthened ERA would also increase the administrative burden
for businesses.
SMEs: There may be additional administrative burden on SMEs to meet the strengthened
requirements for ERA. The greatly expanded obligations and requirements for withdrawal/shortage
reporting and management would also put a relatively larger burden on SMEs compared to their
larger counterparts. On the other hand, SMEs should benefit from the introduction of regulatory
sandboxes to support development of innovative products and scientific support from the Agency, as
well as fee reductions. Incentives for UMN and AMR are also expected to benefit more SMEs,
including biopharmaceutical companies, as they are more active in risky early-stage drug discovery.
6.2
Social impacts
Public health and safety is the key impact assessed
under the social dimension of the legislation and
includes patients’ and health system interests.
155
The financial ecosystem of pharmaceutical R&D: An evidence base to inform further dialogue.
https://www.rijksoverheid.nl/documenten/rapporten/2022/02/28/the-financial-ecosystem-of-pharmaceutical-rd
EU
Figure 8
Avg product accessibility to
54
population
over time, by protection type
kom (2023) 0192 - Ingen titel
2696964_0056.png
Among the specific objectives of this revision, the one on
access
is directly impacting patients.
Analysis of historical data
156
reveals that access to newly authorised medicines in the EU is unequal
and there is a large variation in time to access. Moreover, medicines whose last layer of protection is
SPC are more accessible than RP protected ones (Figure
8).
All policy options seek to address this objective, using either incentives or reducing protection in
case of non-compliance.
Figure 9
shows the likely social impact of the various options. We
compared the options to the baseline in terms of time to access and proportion of EU population
gaining access to a model RP protected medicine.
Figure 9
Proportion of EU population gaining access over time in various options
Based on the assumed compliance rate (Option A – 50%, Option B – 75%
157
, Option C - 66%) and
time limits to comply, we modelled when and what percentage of the EU population can gain access
to the average RP protected medicine (see also section 6.1.1.3).
Option C outperforms all options, by providing access on average to 80% of EU population over the
10 years protected period, 15% higher than in the baseline (65.3%). Also options A and B offer a
higher access than the baseline (67.6% and 70.2% respectively). In other words, in Option A 11
million, in Option B 22 million and in Option C 67 million more EU citizens would have access to a
typical RP protected medicinal product, should they need it
158
compared to the baseline.
The special incentives under Options A and C should support increased R&D investment, especially
in areas of UMN and this should flow through to an increase in treatment options and benefit more
patients. Comparative trials will provide a better evidence base for reimbursement decisions,
potentially leading to cost-effective medicines becoming more readily available to those that need
them. Such trials also tend to assess patient relevant parameters, such as their quality of life and
provide better information to healthcare providers for evidence based treatment decisions.
The reduced regulatory protection in Option B would allow earlier generic/biosimilar entry, lower
prices and eventually increase the number of patients treated with the concerned medicines. The
positive impacts would be somewhat offset by reduced innovation, and the delayed or no entry of
some innovative products to the EU market.
156
157
See Annex 4 (analytical methods and methodology) and Annex 5 (evaluation SWD).
Not all, but for majority of markets.
158
The medicines that were modelled with the average medicine, can be manifold in fact. They may address a small or
big patient population, can offer higher or lower therapeutic value, therefore we refrained from converting the coverage
rate into QALYs or other similar indicator that could thus compromise the integrity of the analysis.
55
kom (2023) 0192 - Ingen titel
2696964_0057.png
The transferable exclusivity voucher in Option A and C would help develop novel antibiotics. While
the scheme would apply to a limited number of novel antibiotics which need to be used selectively,
i.e. as a last-line therapeutic option (to avoid bacteria developing resistance against them), they serve
as an 'insurance' scheme for the EU and global population. The growing threat of antimicrobial
resistance means that routine hospital procedures such as a hip replacement or a caesarean section
can turn fatal. So far, these events are sporadic within the EU, but can develop into a dangerous
public health emergency in the future. Novel antibiotics on the shelf can protect citizens from such a
crisis and the health and economic cost of AMR in case of inaction may be much higher. Moreover,
strict conditions for defining a 'novel' antibiotics will help to ensure that this incentive is not just a
windfall profit for products already in the (weak) pipeline, but encourage additional investment in
research.
In the public consultation, stakeholders rate access to medicines in the EU as ‘moderate’ or ‘poor’
(64.1%). The favoured policy responses differ between respondents; industry placing the root causes
as factors outside the control of the legislation, and public authorities and patients advocating for
obligations or conditions as incentives for access or stronger notification requirements (e.g. for
shortages and withdrawals). For AMR, the highest ranking measure to address AMR was
introduction of a ‘pay or play’ model (Option B) mostly supported by civil society organisations and
opposed by the industry which supported additional market protection period for novel
antimicrobials and the transferable exclusivity voucher.
6.3
Environmental impact
To address the issue of pharmaceutical residues in the environment, and in drinking and natural
waters, different measures have been considered under the policy options. The general
pharmaceutical legislation addresses the impact of pharmaceuticals in the environment through
requirements for an environmental risk assessment (ERA) and related conditions of use and
mitigation measures along the lifecycle of medicines. These measures complement those under the
environmental policy and legislation to reduce the environmental impact of medicines; several
specific environmental legal acts are under review, see section 1.1.
A common measure across all policy options is the more prudent prescription rules for
antimicrobials, which should result in fewer antibiotics entering the environment.
For Option A, the current
ERA requirements
continue with an additional obligation to include the
information on the environmental impact of the supply chain in the application dossier. The impact
of Option A would not be very different to the baseline, though a greater environmental awareness
of the supply chain actors could be envisaged.
Option B increases the requirements for ERA, by including the assessment of the environmental risk
of manufacturing as part of the marketing authorisation process. Option C would in addition
strengthen the conditions of use of medicines and include AMR aspects in GMP to allow a more
holistic assessment of environmental risk along the pharmaceutical lifecycle.
159
The overall impact of options B and C should be less residues (e.g. genotoxic substances,
antimicrobials) in the environment and less disruptions to the ecosystem and human health.
Strengthening the ERA in the general pharmaceutical legislation is expected to have a positive effect
by increasing environmental awareness and responsibilities in the pharmaceutical sector.
Furthermore, a strengthened ERA will also provide an improved basis for taking environmental risk
minimisation measures, enhanced obligations for ERA updates, monitoring of medicines use and
conditions for prudent use. Enforcement should be strengthened as well. The inclusion of assessment
of environmental risk of manufacturing in the ERA would allow tracking the environmental risks of
manufacturing across the supply chain providing a more comprehensive assessment of the potential
159
Annex 11 describes the assessment of the proposed measures (tables 47 and 64) in qualitative terms.
56
kom (2023) 0192 - Ingen titel
2696964_0058.png
environmental impact of a new medicine, but the measure could result in high costs and
administrative burden and pharmaceutical inspectors may not have expertise to check compliance.
For option C, inclusion of AMR aspects into GMP would help minimise amounts of antibiotics
entering the environment via manufacturing and thus prevent or reduce emergence of AMR from
manufacturing of medicines. Companies would have additional costs to comply with AMR
requirements in GMP and public authorities would have additional enforcement costs.
Some limited positive environmental impacts are expected from digitalisation such as electronic
package leaflet and electronic submission of applications in terms of reduced use of paper and
streamlining of the logistics chain.
In the consultations, stakeholders have pointed out that the introduction of new rules at an EU level
has been known to be a trigger for other regions, leveraging on EU actions. There is variable
stakeholder support on strengthening the ERA which ranges from support for it to cover all stages of
pharmaceutical lifecycle, from raw materials to end-product (public authorities and citizens) to
views considering existing measures (controls, benchmarking on the manufacturing and disposal of
products in the environment) stringent enough (industry). According to the targeted survey (Annex
2), the inclusion of assessment of environmental risk of manufacturing in the ERA was mostly
negatively rated by industry while all other stakeholder groups viewed this option as bringing a
positive impact. A workshop conducted for this IA confirmed the general view that there is a tension
between reducing regulatory burden while expanding environmental obligations.
The policy options are aligned with the EU climate-neutrality objective and consistent with ensuring
progress on adaption to climate change. The policy options aim at reducing medicine residues in the
environment and thereby reducing the environmental footprint.
7
H
OW DO THE OPTIONS COMPARE
?
This section compares the expected impacts of the options in relation to the baseline in terms of their
effectiveness, efficiency, coherence, EU-added value, proportionality and subsidiarity.
The comparison focusses on the pivotal elements as these have the most significant impacts and will
allow clear differentiation between the options. The horizontal measures together with the pivotal
elements respond to the objective of
innovation and
will impact on the objective of reducing
regulatory burden and providing a flexible regulatory framework. The other objectives are mainly
impacted by the pivotal elements alone. The overall comparison of the options against the relevant
criteria is presented in
Table 12.
The complete analysis of all the elements is provided in Annex 11.
Table 12
Criteria
Effectiveness: contributing to achieving the policy objectives
Promote innovation,
in particular for unmet medical needs
Create a balanced system for pharmaceuticals in the EU that promotes
affordability for health systems while rewarding innovation
Ensure access to innovative and established medicines for patients with
special attention to enhancing security of supply across the EU
Reduce environmental impact of the pharmaceutical product lifecycle
Reduce regulatory burden and provide a flexible regulatory framework
Effectiveness: other impacts
Competitiveness, SME, single markets
Social impacts (patients, public health and safety)
Environmental impacts
0
0
0
+
++
+
+
+
++
++
+++
+++
0
0
0
0
0
0
++
+++
--
+
+
+++
-
0
++
++
++
++
+
+++
+
+++
+++
++
Overall comparison of policy options
Baseline
Policy
Option A
Policy
Option B
Policy
Option C
57
kom (2023) 0192 - Ingen titel
2696964_0059.png
Criteria
Efficiency
Administrative and compliance costs
Savings and benefits
Coherence
EU added value
Proportionality and subsidiarity
Overall
Baseline
Policy
Option A
Policy
Option B
Policy
Option C
0
0
0
0
0
0
++
+
+
++
+
+
++
++
++
++
+
+
+
+++
++
+++
++
+++
For efficiency, effectiveness, coherence and EU added value, the scores are given on the expected magnitude of impact as explained above: + + +
being strongly positive, + + positive, + moderately positive, 0 neutral, – moderately negative, – – negative and – – strongly negative.
7.1
Effectiveness
Innovation
Option A offers the same default incentives for innovation as the baseline with some additional ones
in particular for UMN and AMR. Overall, Option A is slightly more generous towards innovators, as
in this option incentives can be freely cumulated. Option C on the other hand offers lower default
incentives for innovation than Option A, however under Option C companies can still get the
baseline protection period if they comply with certain conditions (market launch, UMN, comparative
trials etc.). In Option C, the maximum period of RP is capped. Option B keeps the baseline
protection period for UMN medicines, whereas for other RP protected originator medicines there
will be a 15% loss in profits. We estimate that this translates into €670m loss to innovation funding
annually. The pay or play model in Option B is considered less effective than the transferable
exclusivity voucher of Option A and C in stimulating AMR related innovation. It is important to
note that the revision does not affect the incentives pertaining to intellectual property rights (patents
and SPCs). These offer IP protection to the invention(s) associated with the medicine and can extend
the effective protection period beyond RP. As Figure 3 illustrates, for about half of the medicines on
the market, SPC is the protection that expires last. This important incentive for innovation would
still be available for most of the products on the market despite a modulation of regulatory
incentives. The revised SPC regime will not change the duration, but streamlines the way an SPC
can be obtained through a single granting mechanism or a unitary SPC and ensuring legal certainty
for innovative companies.
Horizontal measures will facilitate the secondary use of health data, including real-world evidence,
for innovators (including SMEs and academia), and for regulatory decision-making. Wider and more
systematic access to real world evidence will be integrated in the lifecycle of a medicine, from early
stage of development (complementarity with clinical trials data), to authorisation and post-marketing
supervision. In this context, the European Health Data Space infrastructure will provide a significant
positive economic impact of at least €5.4bn over the next 10 years, stemming from efficiency gains
as a results of a less costly access to health data by reusers (€3.4bn), greater information
transparency for policy-makers and regulators (€0.8bn), and increased value for patients, healthcare
providers and innovators thanks to further reuse of health data
160
. The complementarity of this
initiative with the European Health Data Space, via the facilitation of the secondary use of health
data, will have a direct benefit for all pharmaceutical companies, including SMEs.
Option C combined with horizontal elements, especially simplification, regulatory flexibilities and
digitalisation is more beneficial to innovation compared to the baseline.
160
COM SWD(2022) 131 final
https://data.consilium.europa.eu/doc/document/ST-8751-2022-ADD-3/en/pdf
58
kom (2023) 0192 - Ingen titel
2696964_0060.png
Affordability
In terms of affordability, the general pharmaceutical legislation has a limited role to play, as pricing
and reimbursement of medicines is a Member State prerogative. Nevertheless, the regulatory
protection has an impact on affordability, as it delays generic competition and keeps prices higher.
As demonstrated in section 6.1, two-thirds of the medicines are protected from generic competition
thanks to their SPC or patent protection, therefore any change to the RP would have no effect on
them. According to the draft impact assessment on the revision of the SPC legislation, the unitary
SPC system would not significantly affect the entry of generics and biosimilars on less attractive
(smaller or peripheral) markets in the EU; the larger and more central EU markets usually remain
unaffected as SPCs are sought there anyway. This is possibly so as other factors play a far more
important role in a decision to enter a market, such as: pricing and reimbursement rules, legal
uncertainty connected to the country, quality and readiness of healthcare systems, differences in the
value assessment process, overall levels of pharmaceutical spending and size of the market. The
additional annual expenditure on medicines that might be a result of wider territorial SPC coverage
due to the unitary SPC is estimated at €37m.
161
With these limitations, Option B offers the most effective measure in terms of affordability, offering
€1.13bn direct cost reduction for health payers with the reduced RP period (6+2 years). This
reduction of 0.5%-0.6% of the EU pharmaceutical expenditure would heavily impact 20-25%
162
of
the new medicines (they would lose 15% of their gross profits) while other, often more profitable,
medicines would be unaffected. Option A keeps the baseline protection period. The R&D
transparency requirements in option B and C are supposed to indirectly contribute to affordability
too, better equipping with additional evidence national bodies for price negotiations.
The market launch in option B is an obligation with no additional period of protection whereas in
option C market launch is linked to an incentive. In both cases, if the market launch does not take
place, it would at least result in cost savings to the public as non-complying medicines would lose a
part of their protection period resulting in an earlier entry of generics or biosimilars. In option A, the
market launch incentive would come with an extra €455m cost to the public. Options A and C offer
additional incentives for UMN, and for the transferable exclusivity voucher, which come with
additional costs. This is a
trade-off between innovation and affordability.
Options A and C also
offer an incentive for comparative trials, however the cost of that incentive is counterbalanced by
savings to the health systems by more informed pricing and reimbursement decisions, with an
expected overall neutral/positive impact on affordability. However, this could not be quantified.
Options B and C include an expansion of the so-called Bolar provision to facilitate market entry of
generic and biosimilar medicines immediately after the expiry of regulatory or intellectual property
right protection periods. Market entry of these medicines lower generally the price of the innovator
product and are themselves cheaper
163
and thus make savings for the healthcare systems, e.g. in
2020, the list price savings (excluding confidential rebates and discounts) accounted for €5.7bn in
savings from biosimilar medicine versus the pre-biosimilar cost of the originator
164
.
Option C is the most advantageous by far from a patient/public health perspective, and it represents
a fair balance between originator and generic industry, along with public authorities and payers.
161
Based on historic data (2010-2021) the country with the most significant estimated impact was Latvia in 2019, where
additional spending could reach up to 0.48% of pharmaceutical expenditure, cf. section 6.6.2 of the draft SPC IA.
162
Those having SPC or patent protection, having an orphan market exclusivity, or having an UMN or no return on
investment status in option B would be exempt from the impacts of the decreased RP.
163
Analytical report, indicator AFF-6, Annex 10.
164
The Impact of Biosimilar Competition in Europe, December 2021, IQVIA.
59
kom (2023) 0192 - Ingen titel
Access and shortages
All options result in more and quicker market access of new medicines, compared to the baseline.
The least increase is with Option A and that is the costliest measure for the public. Options B and C
are not only more effective, but they are synergistic with affordability. In these options, the public
wins in either case: more timely access across the EU if companies comply with market launch
conditions, or earlier generic competition and affordability if they do not. The gain in access is
highest with option C, thanks to the shorter deadline to compliance (2 years) and to the all-EU
launch requirement (vs majority of Member States in option B).
Option A does not represent a significant change to the baseline in terms of shortages management,
whereas Option B proposes a more coordinated reporting system, and option C even goes beyond
that, and also requires earlier notification in case of shortages and withdrawals. As such, Option C
has the highest positive impact on shortages, followed by B and A. There is a trade-off among
shortages and administrative burden, better and more reporting is needed to address shortages but
that comes with a certain administrative cost.
Environment
Option A does not impose additional requirements for the ERA, whereas Option B obliges
companies to report about the environmental risks of manufacturing too as part of their MA
application. Option C goes further than B, demanding more stringent conditions of use for medicines
than the baseline. As with the shortages, there is a trade-off among environment protecting measures
and administrative burden.
Regarding the impact on AMR, Option C offers the highest safeguards against the impacts of the
release of antimicrobials into the environment, followed by option B, and with no impact for option
A. All options feature prudent antibiotic use measures, to reduce antibiotics in the environment, and
lower the risk of AMR. Options A and C are also the most effective in financing Europe’s ‘fair
share’ of the cost for novel antimicrobial development through a transferable exclusivity voucher
while in Option B the ‘pay or play’ model would not directly increase the number of novel
antimicrobials and may risk increasing prices in a broad range of medicines without resulting
necessarily in the development of novel antimicrobials (while for the voucher this would concern
only the product on which the voucher would be applied).
Regulatory burden and providing a flexible regulatory framework
Horizontal measures feature uniformly across the options, and they will represent a very significant
burden reduction for companies and public authorities, through streamlining of procedures,
digitisation, enhanced support and regulatory flexibility. In terms of regulatory burden, the
difference among the options is restricted to the increased requirements due to more stringent
shortages and environmental measures, where options C and B score worse than option A. However,
this difference compared to the positive impacts from the horizontal measures is minor.
Impacts on competitiveness and SMEs
In terms of effect on
competitiveness,
the proposed incentives do not make a geographic distinction,
they equally offer regulatory protection for products developed in the EU, or anywhere in the world
which ensures a level playing field between EU-based and third country-based companies. While the
EU regulatory framework is attractive for developers, competitiveness also depends on many other
factors e.g. tax system and incentives; available grants, loans and other funding (e.g. the European
Innovation Council Accelerator); pool of talents; proximity of top academia; clinical trials
infrastructures; market size; security of supply chains; favourable reimbursement decisions.
The horizontal measures described in section 5.2.5 (e.g. simplification, digitalisation, elimination of
duplications) and those pertaining to innovation and the futureproofing of the legislation (e.g.
flexibility of the framework, clarification of scope, sandboxes, codification of rolling reviews and
PRIME) are applicable to all options. They are set to enhance the attractiveness of the EU
framework globally. In this context, other policies and initiatives working in synergy with this
60
kom (2023) 0192 - Ingen titel
2696964_0062.png
revision, like the R&I policy, industrial strategy, the EU system of intellectual property rights
(patents and supplementary protection periods), the creation of the European Health Data Space, are
key factors to promote innovation and EU competitiveness.
In terms of
effects on SMEs,
Option A emphasises support for innovation, but otherwise presents no
major positive or negative impacts for SMEs specifically. Option B includes several measures that
are expected to negatively impact SMEs disproportionately. In terms of innovation, SME originators
may find it more difficult to invest in riskier novel medicines given the reduction in the standard
data protection period and their relatively weaker market position when it comes to negotiating
prices. In terms of obligations for market placement in a minimum number of MSs, including
smaller markets, may be more challenging to meet for SMEs that do not yet have market presence or
distribution channels in such markets. The proposed measures in Option C would be the same for
big pharma and SMEs, however some of the measures may have greater impact on SMEs, e.g. due
to their limited ability to absorb such a reduction in market protection. Mitigating measures such as
longer timeframes to comply with requirements for market launch for example would eliminate any
disproportionate burden on SMEs. Regulatory sandboxes and the transferable exclusivity voucher
for novel antibiotics could be especially beneficial to SMEs because they are more active in
innovative fields than big pharma. Similarly, incentives for UMN would benefit SMEs, which are
generally willing to make early-stage investments in areas of high risk, by giving more value to their
assets even if they are acquired by big pharma in late-stage development. SMEs already enjoy fee
exemptions and reductions for regulatory procedures and through the new horizontal measures
SMEs will benefit from optimised scientific support with a greater likelihood of success for
authorisation. Overall, with the increasing investment in biopharmaceutical R&D and the increasing
share of SMEs among developers, biopharma SMEs in the EU and elsewhere would have excellent
prospects for the future.
Overall, Option C scores the highest in the multi-criteria analysis, this option addresses the most
effectively the specific objectives of the revision, and has the most positive economic, social and
environmental impacts.
7.2
Efficiency analysis
This section compares the cost-effectiveness of the policy measures in the different options, based
on the models and calculations in section 6. The data in tables are always compared to the baseline.
The measures tackling access and affordability (changes in the regulatory protection period) and the
incentives for UNM and AMR are the ones expected to have the most substantive economic impacts
on the various stakeholders. Tables 13a, 13b and 13c compare the options with all relevant measures
(the cost-benefit analysis of the variation to option C is presented in section 8.1).
Table 13a
Cost-benefit table of key measures in
Option A
Option A
+1 year extension of RP for
medicines addressing
UMN
+6 months extension of RP for
conducting
comparative clinical
trials
+6 months extension of RP for all EU
market launch
Transferable exclusivity voucher
Cost/benefit for public payer and
patients
+ €246m cost
+ 1-2 new UMN addressing
medicines
+ €328m cost
+ faster access and cost saving
thanks to improved reimbursement
decisions
+€455 m public cost
+3% access
+€441m cost
+ 1 novel antibiotic
+ €1.470m cost
+ 1-2 new UMN medicines
+comparative data
+3% access
+1 novel antibiotic
Cost/benefit for originators
+ €282 gross profit
(3 incentives)
+ €378m gross profit
+€280m cost
(8 medicines)
+€527 m gross profit
(7 complying medicines)
+€387m gross profit
(1 voucher)
+€1.294m gross profit
Cost/benefit for generic
industry
- €39m gross profit
- €52m gross profit
- €71m gross profit
- €54m gross profit
- €216m gross profit
Total balance
61
kom (2023) 0192 - Ingen titel
2696964_0063.png
Table 13b
Cost-benefit table of key measures in
Option B
Option B
2 year
reduction of RP
(except for
UMN)
Loss of RP, if no
market launch
in
majority of EU within 5 years
Total balance
Cost/benefit for public payer and
patients
+€1860m gain
innovation loss
+€681m gain
+5% access
+ €2.541m gain
+5% access
innovation loss
Cost/benefit for originators
-€1.970m gross profit
(9-12 medicines)
-€842m gross profit
(4 non-complying medicines)
- €2.812m gross profit
Cost/benefit for generic
industry
+€266m gross profit
+€101m gross profit
+€367m gross profit
Table 13c
Cost-benefit table of key measures in
Option C
Option C
2 year conditional protection for
all
EU launch in 2 years
+1 year extension of RP for
medicines addressing
UMN
+6 months extension of RP for
conducting
comparative clinical
trials
Transferable exclusivity voucher
Cost/benefit for public payer and
patients
€444 m gain
+15% access
+ €246m cost
+ 1-2 new UMN addressing
medicines
+ €328m cost
+ faster access and cost saving
thanks to improved reimbursement
decisions
+€441m cost
+ 1 novel antibiotic
+ €571m cost
+ 1-2 new UMN medicines
+comparative clinical data
+15% access
+1 novel antibiotic
Cost/benefit for originators
-€469m gross profit
(5 non-complying MP)
+ €282m gross profit
(3 incentives)
+ €378m gross profit
+€280m cost
(8 medicines)
+€387m gross profit
(1 voucher)
+€298m gross profit
Cost/benefit for generic
industry
+€63m gross profit
- €39m gross profit
- €52m gross profit
- €54m gross profit
- €82m gross profit
Total balance
The tables provide an overview of the costs and benefits of the different options and on different
stakeholder groups. Whenever it was possible, we presented the cost/benefits in a monetised form,
however for certain social benefits putting a monetary value was either not possible or not
appropriate. Therefore the societal benefits of new UMN addressing medicines, of improved access,
of new innovative antibiotics and of comparative clinical data of new medicines are only mentioned
in the table, without a monetary value.
In terms of efficiency, option A delivers quite well on all targets and creates the desired societal
benefits, however at a significant cost for the public, missing the affordability target. Option B on
the other hand is very cost-efficient for patients and public payers, offering altogether €2.5bn
savings to the public, around 1% of the annual pharma expenditure. Option B does improve patient
access and UMN medicines would receive a relatively higher support (though unchanged compared
to baseline). The savings to the public would be borne mostly by the originator industry.
Option C distributes the cost of the additional societal benefits more evenly among the stakeholders,
and also effectively delivers on all objectives. In terms of efficiency, option C offers the most cost-
effective mix of policy measures. The variation to option C (presented in section 8.1) equally
delivers on all objectives in a cost-efficient manner, with a slightly different distribution of cost to
offer more gains for public payers and patients.
Horizontal and other measures
In Annex 3, the analysis concluded that the horizontal measures are – in the best case scenario –
expected to generate up to around
€300m savings annually regardless of the selected option,
shared among businesses (one-third) and authorities (two-thirds). Additional administrative costs
resulting from measures on shortages and environment would offset as a minimum 10% of these
savings (min. €30m additional cost) for businesses; likewise for administrations.
Option C offers the most cost-effective solution to achieve the specific objectives, followed by
Options B and A.
62
kom (2023) 0192 - Ingen titel
2696964_0064.png
7.3
Coherence
Options B and C are consistent with the EU Strategic approach to pharmaceuticals in the
environment and complementary to the ongoing revisions of the environmental legislation
mentioned in section 1.1. All policy options are coherent with the EU Action Plan on Antimicrobial
Resistance
165
.
All three options contribute to SDG 3 (“health and well-being), SDG 9 (“innovation
and infrastructure”) and SDG 10 (“reduced inequalities”)
166
(section 1).
The objective of patient access to affordable medicines is coherent with the objective of the HTA
Regulation on timely patient access. Option C with its incentives for both EU-wide access and
comparative clinical trials provides the best alignment followed by Option A.
Through the horizontal measures all options will ensure coherence with the sectorial legislations
medicines for rare diseases and for children, EMA fees legislation and with EU legal frameworks on
medical devices/in vitro diagnostic and on BTC through efficient interaction and synergies between
these regulatory frameworks. In addition, options B and C will create more clarity on the interplay
between these legal frameworks through the proposed changes in definitions and classification
advice. More details available in Annex 6 with regard to medicines for rare diseased and children
and in Annex 9 for BTC.
The access related measures in Option C such as the modulation of incentives or the additional
obligations of supply will not only have a positive effect on access but also a systemic effect on
public and private actors’ behaviour, as explained in section 6. At the same time, the European
Health Data Space will provide actors access to harmonised EU health data which unlocks
possibilities and efficiencies along the pharmaceutical lifecycle in the development of medicines
promoting innovation, in the monitoring of medicines for both regulators and marketing
authorisation holders and in evidence generation for downstream decisions after marketing
authorisation.
The revision of the general pharmaceutical legislation and the revision of the SPC regime with a
unitary SPC are coherent in the objectives to promote innovation and reduce regulatory burden.
However, the unitary SPC may have a small negative effect on affordability, as mentioned in section
7.1, and a hypothetical risk
167
of delaying generic or biosimilar entry in markets, where the
originator has never been present which would have a negative effect on patient access. On the other
hand, the predictability for generic/biosimilar companies will increase in the new SPC regime,
through a central SPC database, effectively streamlining decision, less risk of litigation and, if
litigation occurs, the avoidance of multiple litigation. Together with the measures undertaken under
the pharmaceutical revision to support day 1 entry of generics and biosimilars this will facilitate
patient access to those products.
HERA would support solutions from the public procurement side to the market failures in the area of
antimicrobials. This unprecedented, combination of policy changes is a result of a combined set of
actions in related areas (data, procurement, pharmaceuticals) that complement each other and should
not be seen in isolation from each other. Together with the futureproofing and simplification
elements of this revision they constitute a holistic response which can be expected to radically
upgrade the EU’s position globally as a place for medicine innovation.
165
A European One Health Action Plan against Antimicrobial Resistance (AMR) (June, 2017), available at:
https://ec.europa.eu/health/system/files/2020-01/amr_2017_action-plan_0.pdf
166
Sustainable development in the European Union, overview of progress towards the SDGs in an EU context, 2022
edition, Eurostat (2022)
167
The risk is considered hypothetical because it is only in very limited cases that generic or biosimilar medicines enter a
market where a SPC has not been requested or granted, i.e. a market where the originator has never been present.
63
kom (2023) 0192 - Ingen titel
7.4
Proportionality and subsidiarity
All three options are consistent with the EU’s right to act under the Treaty of the Functioning of the
EU (covering public health protection, the single market and the free movement of products within
the EU). Moreover, all three options propose actions that will allow the objectives of the revision to
be addressed to a greater extent than if Member States were acting alone.
The principle of proportionality is strongly reflected in the discussion of certain trade-offs to be
made between the different objectives (section 4). To give an example, trade-offs are inherent
between the objective of innovation and affordability often achieved by generic/biosimilar
competition. The incentives will remain a key element for innovation but they have to be adapted to
better take into account that medicines are not sufficiently accessible by patients in all Member
States. This is reflected in Option C which modulates incentives to reward innovation, especially for
UMN, but also make the regulatory protection period conditioned to market launch in all Member
States. If this condition is not fulfilled generic competition will start earlier, resulting in increased
affordability.
With regards to subsidiarity, all options pursue the objectives of the revision and provide a clear
demarcation between EU level and Member State level actions. They do not propose any change to
the national health care systems which are in the exclusive power of Member States (Article 168
TFEU), but certain measure (e.g. transparency requirements, better evidence base, early dialogue
between regulators, HTA bodies and payers) will facilitate decisions of Member States in these areas
e.g. pricing and reimbursement.
7.5
Limitations of the comparison
There is a level of potential uncertainty in the findings described in section 7 owing to the influence
of other contextual factors such as developments in the pharmaceutical sector, other relevant
legislations (e.g. HTA Regulation, Urban Waste Water Directive and SPC Regulation) and policies
at Member State level (see for details of factors influencing access to affordable medicines – annex
14). While the influence of external factors has been considered in the design of the options and their
analysis there is an unavoidable risk that they may impact or delay some of the expected benefits.
Their effects and anticipated unintended consequences (e.g. the effect of some measures on prices of
medicines, or the effect of conditionality of certain incentives on innovation) are analysed to the
extent possible in section 6. There is also a level of uncertainty owing to the limitations and
assumptions involved in assessing and quantifying the likely impacts of the options provided.
All methods applied to our research encountered a varying degree of difficulty in relation to lack of
quantitative data available in the databases and sources examined. We did not find enough data to
quantify all relevant impacts of every policy measure discussed in the policy options for the future of
the legislation. Whenever possible, we have made reasonable assumptions to assess the impacts, but
this lack of quantitative data is a key limitation to our analysis.
8
P
REFERRED OPTION
The impact assessment indicates that policy option C is most effectively addressing all the objectives
of the revision of the general pharmaceutical legislation in an efficient and consistent manner. The
measures of option C address in a proportionate manner the underlying problem drives; a mapping
of measures against problem drivers can be found in Annex 16.
This option proposes a modulated trade-off between incentivising innovation (for both unmet
medical need and antimicrobial resistance) and improving access, R&D transparency, and security
of supply of medicines as well as reducing the environmental impact of medicines. The costs and
benefits of Option C for different stakeholder types are described below. The below section
considers the pivotal measures but also
takes into account the other measures assessed in Annex
11,
along with the impacts of the horizontal measures.
64
kom (2023) 0192 - Ingen titel
2696964_0066.png
The preferred option conforms to the principles of subsidiarity and proportionality. It respects the
national competence on the organisation of the Member States’ healthcare systems and provides
clear demarcations between EU level and Member State level actions. Given the objectives the
revision aims to achieve, the trade-offs and new burdens on companies and authorities are
acceptable and proportionate.
It is expected that the revision will not change the current legal instruments, i.e. a Directive and a
Regulation, for the general pharmaceutical legislation.
8.1
Costs and benefits of the preferred option
Table 14
reviews the most significant costs and benefits stemming from the pivotal measures, and
also includes the variation to Option C described in section 5.2.4. The variation would decrease the 2
year conditional protection to 1 year. As a result, the overall protection level moves down by 1 year
for all RP protected medicines, and only 1 year protection remains dependent on the launch
condition. The 1 conditional year is a lower “prize” for compliance, thus we assumed that fewer
medicines would meet the requirement (50% vs. 66% in the default). The variation allows the
legislator to consider the impacts on the various stakeholder groups by “moving the cursor”.
Table 14a
Cost-benefit table of incentives in Option C (6+2+2) compared to baseline (8+2)
Option C
2 year conditional protection for
all
EU launch in 2 years
+1 year extension of RP for
medicines addressing
UMN
+6 months extension of RP for
conducting
comparative clinical
trials
Transferable exclusivity voucher
Total balance
Cost/benefit for public payer and
patients
€444 m gain
+15% access
+ €246m cost
+ 1-2 new UMN addressing
medicines
+ €328m cost
+ faster access and cost saving
thanks to improved
reimbursement decisions
+€441m cost
+ 1 novel antibiotic
+ €571m cost
+ 1-2 new UMN medicines
+comparative clinical data
+15% access
+1 novel antibiotic
Cost/benefit for originators
-€469m gross profit
(5 non-complying MP)
+ €282m gross profit
(3 incentives)
+ €378m gross profit
+€280m cost
(8 medicines)
+€387m gross profit
(1 voucher)
+€298m gross profit
Cost/benefit for generic
industry
+€63m gross profit
- €39m gross profit
- €52m gross profit
- €54m gross profit
- €82m gross profit
Table 14b
Cost-benefit table of incentives in Option C Variation (6+2+1) compared to baseline (8+2)
Variation to Option C
1 year general reduction of the RP
1 year conditional protection for
all
EU launch in 2 years
+1 year extension of RP for
medicines addressing
UMN
+6 months extension of RP for
conducting
comparative clinical
trials
Transferable exclusivity voucher
Cost/benefit for public payer and
patients
+€1,008m
+€384 m gain
+8% access
+ €246m cost
+ 1-2 new UMN addressing
medicines
+ €328m cost
+ faster access and cost saving
thanks to improved reimbursement
decisions
+€441m cost
+ 1 novel antibiotic
+ €377m gain
+ 1-2 new UMN medicines
+comparative clinical data
+8% access
+1 novel antibiotic
Cost/benefit for originators
-€991m gross profit
-€378m gross profit
(8 non-complying MP)
+ €282m gross profit
(3 incentives)
+ €378m gross profit
+€280m cost
(8 medicines)
+€387m gross profit
(1 voucher)
-€602m gross profit
Cost/benefit for generic
industry
+€133m gross profit
+€51m gross profit
- €39m gross profit
- €52m gross profit
- €54m gross profit
+€39m gross profit
Total balance
In the default Option C, the higher market access is achieved without extra cost to the public, even
some gains could be expected in case of non-complying medicines. The other incentives would
mean an extra cost to the public and to generics, nonetheless it is expected that the indirect benefits
from the medicines addressing UMN and faster and better reimbursement decisions, would offset
65
kom (2023) 0192 - Ingen titel
2696964_0067.png
these costs. The originator companies would have additional costs and benefits from the incentives
and the market launch conditionality, and overall they would see an increase in their sales.
In the variation of option C the public would gain significantly compared to the baseline in monetary
terms and also enjoy the benefits of the measures. The gains would even allow financing the
transferable voucher to support development of novel antimicrobials, without turning the public
monetary balance into negative. In the variation, all the costs of the positive social impacts would be
translated into reduced revenues for innovator companies, though a significant proportion of the
costs would come from non-compliance (e.g. not launching in all EU markets, not carrying out
comparative trials), which companies should avoid by complying.
In the variation the cost is put only on a subset of innovator companies, e.g. high-sales, SPC
protected medicines would be unaffected. The shorter conditional period for market launch (1
instead of 2 years) means a smaller loss of revenue if companies do not launch in all EU markets,
therefore a lower compliance rate (50%) is assumed, resulting in smaller positive effect on patient
access. The loss to innovators may translate into slightly less innovation.
Option C and its variant are both cost-effective alternatives to reach all the objectives, the slight
difference between the two being the different focus on more access or more affordability (+15%
access and €571m more cost vs. +8% access and €377m gains) for the public payer and patients.
Patients, Citizens and Healthcare services
Option C will bring
benefits to patients and citizens
by facilitating the work of healthcare
professionals, pharmacies, hospitals and strengthening health. The new measures to promote access
across all Member States, by incentivising companies to launch their products on all EU markets,
coupled with lower revenues for companies in case of non-compliance will be the first EU-level
legislative measure to address the long-standing inequalities
and will increase patient access to
innovative medicines.
Facilitating the entry of generics and biosimilars will increase affordability
and consequently increase the number of patients treated. The additional incentive for addressing
UMN will incentivise the development of more medicines with high public health benefit.
Transferable vouchers would lead to development of novel antimicrobials, reduce EU deaths and
health system costs due to AMR, and ensure a better preparedness against the increasing threat of
resistant bacteria.
Security of supply
measures will improve continuous availability of both critical
and non-critical medicines, which will significantly reduce shortages of medicines and benefit
patients and healthcare services. Citizens will also benefit from measures taken to reduce the impact
of pharmaceuticals on the environment and on public health via the environment through a
strengthened environmental risk assessment of medicines along their lifecycle and imposition of
appropriate measures to mitigate these risks.
Several other measures discussed in Annex 11 will corroborate the impacts of the pivotal measures:
Option C would give a push to repurposing of medicines, as a cost-efficient way to expand
therapeutic uses of medicines instead of a rather selective and even risky off-label use (C.1.2.,
C.1.3.)
168
. Along with the measures facilitating generic entry right after protection expiry (C.1.4.,
C.5.1., C.5.2., C.5.4., C.5.5.), these will further expand patients’ access to medicines. Prudent use
measures for
antimicrobials
will help decrease the risk of AMR (C.2.3, C.2.4, C.2.5).
A harmonised system for authorisation of medicines in the EU – through the general pharmaceutical
legislation – offers clear EU-added value for public health to enable access to and innovation of
medicines. In addition, EU-level action is the most efficient mechanism – in the scope of this
revision – to address the concerns Member States have raised about unequal access and
affordability, in particular for the centrally authorised medicines.
168
The codes in brackets refer to the codes of the measures in Annex 11for easier identification
66
kom (2023) 0192 - Ingen titel
2696964_0068.png
Future proofing
measures of Option C will ensure patient safety in areas of rapid technological
change, including personalised medicine. Currently, Directive 2001/83/EC covers all ‘medicinal
products’ that are “either prepared industrially or that are manufactured by a method involving an
industrial process”. “Delinking” the legislation’s scope from the way medicines are manufactured
will address potential regulatory gaps (without changing the overall scope) due to scientific and
technological developments e.g. low-volume products, bedside-manufactured or single batch
personalised medicines that do not involve an industrial manufacturing process
169
(C.3.3.). Adapted
regulatory pathways, e.g. for less complex cell-based medicinal products, and regulatory sandboxes
will also increase the chance of faster patient access to cutting edge medicinal products (C.3.5.,
C.3.6.). Lastly, allowing electronic product information will bring advances to readability for
patients and opportunities for healthcare professionals to communicate information more effectively
(Horizontal 6).
Industry
For the originator industry, the modulation of the regulatory protection means a lower standard
duration of regulatory protection,
but companies can achieve a similar/same (depending on the
variant in this option) protection as of today if they comply with the
condition to launch in all EU-
markets.
The extra condition would entail some additional administrative cost, but that would be
somewhat compensated by burden reduction, such as allowing multi-country packs for certain types
of medicines (C.4.2.). The special incentive for addressing UMN would offer a longer period of
protected sales and thus a higher return on investment, a €282m additional gross profit at industry
level. The special incentive for comparative trials will recompense the additional costs from carrying
out the trials, and the data will help faster pricing and reimbursement decisions, and earlier market
entry. It comes with €378m extra gross profit, but also with €280m cost. The trial data would allow
better negotiating position for payers, which may limit company’s profits. The transferable
exclusivity voucher would reward developers of novel antibiotics, and also the buyers of the
vouchers would have gains.
The incentives involving extension of data protection would delay generic entry and keep generic
companies out of the market for longer. In the case of UMN incentive of an additional 1 year to
originators, it represents a loss of €39m in gross profit per year for generic companies, and €52m for
comparative trials. They would also have increased costs from the obligation to include smaller
markets in their own mutual recognition procedure (or decentralised procedure) applications (C.1.5,
C.1.6.). On the other hand, there should be an increase in R&D activity for generic/biosimilar
medicines with a streamlined and clearer regulatory pathway (C.5.1.) and by measures facilitating
generic entry right after protection expiry.
Option C also brings greater certainty for businesses by adding clarity and predictability to the
regulatory system and the legal pathway (see references to "delinking" in the previous section, as
well as adaptation of definitions), streamline the GMO assessment in the authorisation of clinical
trials that involve investigational medicines with a GMO component (C.3.2.). These measures
should promote
innovation
and attract investment to the EU. SMEs should also benefit from the
introduction of regulatory sandboxes to support development of innovative products (C.3.6.) and
enhanced support in addition to the current fee reductions.
The preferred option continues to provide a favourable incentive structure for innovation in the EU
which remains competitive against what other regions offer. The incentives apply equally to all
products, regardless of where they are developed – in the EU or elsewhere; in this regard, the EU
competitiveness is not negatively impacted by this option.
169
Organised in close coordination with other EU legal frameworks (medical devices, substances of human origin) to
avoid shifts of therapies that are already regulated
67
kom (2023) 0192 - Ingen titel
2696964_0069.png
Greater use of multi-country packs is also expected to facilitate the movement of medicines within
the EU internal market, which will help all businesses. In terms of
security of supply,
option C
introduces several obligations and requirements on MAHs and wholesalers that likely will carry
additional costs to these parties including costs associated with warehousing (for stockpiling),
operations and capital (C.6.1. to C.6.9.). Stakeholder consultations estimated that increasing
warehouse capacity to accommodate 10% additional stock will have a cost of €500k – 1m per
warehouse. This policy option will also require more
transparency
and at the same time obligations
regarding supply chain actors and environmental risk assessments, which will result in additional
costs for businesses for inspections, compliance and other additional responsibilities. This will likely
represent a substantial burden on SMEs in particular.
The horizontal measures on the other hand simplify the regulatory system and reduce burden on
industry, reducing compliance costs and administrative burden in the range of €80-160m per year.
For industry, a harmonised and predictable medicines regulatory framework – through the general
pharmaceutical legislation – offers clear EU-added value by reducing duplication, simplifying
requirements and making the system easier to navigate. The preferred option aims at harmonising
requirement concerning shortages.
Despite the new obligations for companies, the preferred option is proportionate when balanced with
the efficiency gains, including those from secondary use of health data via the European Health Data
Space (see section 7.1), and simplifications introduced and the recognition that other objectives such
as patient access and the wider policy ambitions on strategic autonomy and green deal have to be
factored in.
Competitiveness and future of innovation under reduced regulatory data protection
Industry stakeholders frequently claim that the reduction of regulatory data protection period would
harm future innovation and EU competitiveness. In section 6.1.1.2 we demonstrated that the
incentives are agnostic to the geographic origin of the medicines, therefore the reduction would not
harm EU companies more than non-EU companies coming to the European market (non-EU
companies develop 80% of new medicines introduced to the EU market).
However, lower profits may transform into less innovation at a global scale. Option C results in a
slight gain in gross profits but the variation of option C estimates a total loss of €602m in gross
profits. Industry re-invests on average 25% of their gross profit into R&D, consequently €150m may
be lost for innovation. In 2021 the global pharmaceutical industry has invested €230b in R&D
170
,
hence the potential loss amounts to 0.07% of global R&D investment. If we wanted to translate this
to medicines, only 1 in the next 1500 new medicines would not be developed because of the
reduction, a likely invisible loss over the next 15 years.
Public authorities, agencies and payers
Incentives involving additional data protection periods will lengthen the period in which health
systems can be charged higher prices for medicines. For example, transferable vouchers would have
indirect healthcare costs for the healthcare payer.
Public authorities will require additional budget and expertise for reviewing MA applications (larger
number of applications, change in ERA requirements, etc.), enforcement of obligations (e.g. for
market launch, lifecycle management of antimicrobials), inspections of manufacturing sites,
increased commitments to provide advice (e.g. on interchangeability of biosimilar medicines, ERA,
green manufacturing, classification of borderline products etc.) as well as setting up of new
centralised infrastructure for information exchange (e.g. for shortage monitoring; one-off costs).
Additional costs for EMA in assessing the application for new antimicrobials and the associated
170
$238b -
EvaluatePharma - World Preview 2022, Outlook to 2028, page 20
68
kom (2023) 0192 - Ingen titel
2696964_0070.png
voucher are estimated at €2m per year. The workload of pricing and reimbursement agencies would
also increase with incentives for market launch driving up the number of applications, while their
workload should decrease from better evidence provided from more comparative trials.
Health payers would also benefit from measures to promote post-authorisation studies and
comparative trials, which would enable access to evidence that supports pricing and reimbursement
decisions for HTA bodies. Rejecting immature marketing authorisation applications at time of
validation would reduce workload of medicine regulators (C.9.1.) with estimated savings for the
EMA and NCAs at 3% of annual costs.
Measures to improve security of supply will facilitate information exchange between Member State
authorities and improve strategies to tackle shortages. Both aspects should reduce long-term costs to
authorities. However, public authorities will also need to increase capacity to assess shortage
prevention plans provided by MAHs, and, depending on the cost and risk-sharing agreements for
reserve stock, authorities may also incur direct costs for storage. While measures to improve quality,
manufacturing and environmental impact of pharmaceuticals will increase workload for EMA and
NCAs, increased coordination, joint audits and data sharing could also result in efficiencies.
Academic/research institutions
Option C will bring benefits for clinical researchers and academics in the form of opportunities to be
more involved in the development work and trials, as a binding system for scientific assessment of
evidence for repurposing off-patent medicines will be established (C.1.2), and obligations will be
simplified to facilitate non-commercial entities (e.g. academic) to become MAHs (C.1.2). This
option also brings increased requirements of efficacy and safety for use of hospital exemption (e.g.
trial data and good manufacturing practices capability), dedicated pathways for less-complex cell
based medicinal products and a regulatory sandbox (C.3.5. and C.3.6.), which may impact the
activities of academic researchers and research institutions under this exemption, but should support
data collection, safe and efficacious use and ATMP development. Academics and research
institutions will also benefit from streamlining ‘horizontal’ measures such as fee reduction and more
scientific support to help non-commercial entities to bring innovative medicines to the market.
8.2
REFIT (simplification and improved efficiency)
The review aims at
simplifying the regulatory framework and improving its effectiveness and
efficiency thereby reducing the administrative costs borne by companies and administrations
171
. The
horizontal measures are envisaged in that regard and most of them will act on the core elements of
the authorisation and life-cycle procedures, which are at the centre of this legislation. These
measures can be grouped as follows:
Streamlining and acceleration of processes and coordination of the network
The proposed abolishment of the sunset clause and renewal of MAs after five years would avoid
unnecessary duplication and a burden on MAHs and regulators
172
. The envisaged reduction in the
number of notifiable variations could potentially reduce the administrative costs incurred by MAHs
and regulators. For generic applications, in order to avoid duplicative assessments of the same data
for medicines containing the same active substance, to reduce administrative costs for both
administrations and companies, worksharing procedures and a more efficient repeat use procedure
are proposed.
A quantification of these costs is presented in Annex 3.
The latter not adding value regarding safety, given the availability of Periodic Safety Update Reports that accumulate safety data
and any impacts on the known benefit-risk balance.
172
171
69
kom (2023) 0192 - Ingen titel
2696964_0071.png
The revision will also look to streamline efficient interaction (early dialogue) between different
regulatory authorities (EMA, NCAs, HTA, etc.) as well as synergies between different but related
regulatory frameworks, e.g. interplay with BTC framework, medical devices (for certain types of
products) and health technology assessments. This, together with a structural simplification of EMA
(e.g. as regards the committees) should further reduce the administrative costs for both the
administration and the business.
Digitalisation
The envisaged revision aims at an enhanced digitisation of different applications to EMA and NCAs,
which should result, overall, in cost reductions. This would induce initial, one-off, costs for the
administrations but should bring efficiencies and therefore cost reductions with time. Finally, the
envisaged use of the electronic product information, i.e. the electronic leaflet as opposed to paper
leaflets, should also, in the long term, adduce additional administrative cost reductions.
Adaptations to accommodate new concepts and support SMEs and non-commercial organisation
The revision foresees adaptations to accommodate new concepts and regulatory processes such as
adaptive clinical trials, use of real world evidence, and new uses of health data within the regulatory
framework. This should result in cost reductions for businesses and administrations. It also
envisages optimising the regulatory support to SMEs and non-commercial organisations. This
should in turn result in additional reductions of administrative costs for these parties.
8.3
Simplification and burden reduction for businesses, supporting the one in one out
approach
This section evaluates the administrative costs induced by the implementation of the
preferred
option for
businesses and citizens/patients, in comparison to the baseline. Moreover, all options include some
administrative costs related to horizontal elements, which are also evaluated in comparison to the
baseline
173
.
As regards companies, there are a number of cost reductions resulting from
the implementation of the
preferred
option.
The reduction is done for reasons of good policy but also in part to create the
financial headroom to introduce new legislative actions and procedures that will inevitably bring
additional costs in pursuit of additional social benefits. As a case in point, the strengthening of the
environmental risk assessment within the overall assessment process (e.g. in consideration of
manufacturing and supply chain issues) will add costs, compared with the current situation, as will
the inclusion of environmental issues within post-market authorisation monitoring and the measures
on security of supply.
As regards companies, there are also costs reductions resulting from
the implementation of
horizontal
measures which apply to all the options. The
revision aims at
simplifying the regulatory framework
and improving its effectiveness and efficiency thereby reducing the administrative costs. Annex 3
presents the cost for the horizontal measures that relate most directly to streamlining of processes
and coordination of network as well as digitisation measures. The table summarises the balance of
costs and benefits, and suggests that the measures as proposed may deliver a reduction in
compliance costs and administrative burden in the range of €524.5-1,050m for the industry
174
.
More specifically:
The proposed streamlining procedures, including enhanced support, will yield useful cost
savings for European pharmaceutical businesses, with estimated cost savings falling in the range
of €412.5-825m over the next 15-years.
A quantification of these costs and savings is presented in Annex 3.
Methodological details underpinning the calculations are described in Annex 4.
173
174
70
kom (2023) 0192 - Ingen titel
2696964_0072.png
The proposed digitalisation measures will provide relatively modest financial savings to
industry, given the primary focus is on the integration of regulatory systems and platforms across
the EU and support for the re-use of data. Electronic submission will however deliver industry
cost savings. These are estimated at €112m-€225m over 15 years.
For citizens/patients, there are many improvements foreseen in all areas of importance
175
but there
are no obligations and therefore costs induced by the legislation.
9
H
OW WILL ACTUAL IMPACTS BE MONITORED AND EVALUATED
?
Indicators for the preferred option, in relation to the core objectives, with suggested data sources and
proposed frequency of data collection are presented in table 15. The Commission will review the
indicators periodically.
Much of the data collected by EMA are already collected today and published in its annual reports;
the new data collected by EMA would result in only a minor additional burden. The burden on the
Member States to provide data on the number of shortages, variations and authorised antimicrobials
would also be minor, and even further reduced by digitisation. The Commission has access to the
IQVIA data and data from the other sources are already being collected.
The development of medicines is a long process and the completion of clinical development plans
can take up to 10-15 years. Regulatory protection periods of the preferred option exert their effect up
to 11 years after marketing authorisation. For certain measures concerning incentives for innovation,
affordability and access, a meaningful evaluation of the revised legislation can take place only 15
years from its application. The Commission will monitor though the indicators and assess the need
for an earlier revision.
Table 15
Proposed list of monitoring and evaluation indicators
Specific objective
Promote
innovation,
particular for UMN
in
Monitoring indicators
Number of authorised medicines with new active
substance
Number of authorised medicines addressing UMN
Number of authorised antimicrobials
Number of authorised novel antibiotics/transferable
vouchers granted
Number of incentives granted for comparative trials
Use of pre-marketing regulatory support (scientific
advice, PRIME)
Number of sandboxes used
Data source/frequency
EMA data/annual
EMA/annual
EMA and NCAs/annual
EMA/annual
EMA/annual
EMA/annual
EMA/annual
IQVIA data/biannual
Euripid database, IQVIA data,
OECD data/biannual
Eurostat, OECD data/biannual
IQVIA data/biannual
Create a balanced system for
pharmaceuticals in the EU that
promotes affordability for
health systems while rewarding
innovation
Ensure access to innovative and
established
medicines
for
patients, with special attention
to enhancing the security of
supply across the EU
Market share of generic and biosimilar medicines
Development of prices of medicines
Member States’ pharmaceutical spending
Time from authorisation to market launch
Number of Member States where basket of medicines
(both innovative and established medicines) are
launched
Number of market access incentives granted
Number of withdrawal of medicines reported </> 1
year in advance
Number of withdrawals for which, as a result of the
IQVIA data/biannual
EMA and NCAs/annual
175
The legislation aims at improving the flow of cutting-edge treatments for conditions for which there are no effective
treatments currently (UMN), reversing the decline in investment in antimicrobial research and encircling the issues
driving AMR, incentivising access in all Member States, a broader repurposing, and the generic and biosimilar entry. A
more robust ERA will support environmental goals. Measures on security of supply will improve access to medicines.
71
kom (2023) 0192 - Ingen titel
2696964_0073.png
notification, measures could be identified to mitigate,
prevent or alleviate a critical impact on the health
system or on patients of the withdrawal
Total number of shortages
Number of shortages reported </> 6 months in
advance, specifying number of critical shortages
Number, root cause and duration of critical shortages
and identification of measures that mitigated,
prevented or alleviated impact on the shortage
Number of NCAs automatically sharing information
with the EMA platform and number of NCAs
manually submitting information with the EMA
platform
Presence of medicines residues in the environment
Consumption of antimicrobials
GHG emissions of EU-based pharmaceutical
manufacturers
EMA and NCAs/annual
EMA and NCAs/annual
EMA and NCAs/annual
EMA and NCAs/annual
Reduce the environmental
impact of the pharmaceutical
product lifecycle
Reduce the regulatory burden
and
provide
a
flexible
regulatory framework
Number of variations
Number of meeting of EMA scientific committees and
their working parties
Number of early dialogues/ scientific advice including
other public authorities than medicine authorities
Number of scientific advice given to SMEs and
academia
EMA
Information Platform for
Chemical Monitoring that
includes data on occurrence of
pharmaceuticals in the
environment
ECDC annual report on
antimicrobial consumption
Eurostat/annually
EMA, CMDh and
NCAs/annually
EMA/annually
EMA/annually
EMA/annually
72