Download Chapter 8.3 - WHO archives

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Declaration of Helsinki wikipedia , lookup

Medical research wikipedia , lookup

Patient safety wikipedia , lookup

Clinical trial wikipedia , lookup

Theralizumab wikipedia , lookup

Multiple sclerosis research wikipedia , lookup

Quackery wikipedia , lookup

Transcript
8.3 Barriers to innovation in pharmaceutical research and
development
The impetus for this Project arose from the various reports issued between 2000 and
2003 (Appendix 2). Coincidentally, three papers were published in March 2004 by the
EMEA and FDA (Appendix 8.3) and by Michael Rawlins in Nature (Appendix 8.3.4). In
addition, a paper prepared by individuals working in the pharmaceutical industry was
prepared for this Report (Annex 8.3). These papers all maintained that the present costs
and time spent in developing new medicines were unsustainable. The authors
suggested similar but slightly different solutions to the main barriers with the EMEA,
FDA and Rawlins papers focusing on regulatory obstacles, while the industry paper
was broader.
In recent years, the cost1 and duration of medicines development have increased and
this has been coupled with low output of innovative medicines from the
pharmaceutical pipeline.2 Recent estimates suggest that it costs nearly US$ 1 billion for
each new chemical entity and takes 10 to 15 years on average from discovery to market
authorization. While there may be many projects in early development stages, the fact
remains that little of this potential innovation is being translated into market approvals.
For some diseases, there remains an inadequate understanding of basic science, and
potential targets for medicines development have not been identified.
Regulatory barriers
All authors agreed that every aspect of the regulatory process should be re-examined
and that the evidence base for regulatory practices should be critically analysed using
modern methodologies. In particular, this includes preclinical regulatory "rituals." For
clinical research, there is a suggestion from Rawlins that alternatives to randomized
controlled trials should be investigated. Under some circumstances, he suggests,
historical controls could be utilized and alternative analytical statistical techniques
using Bayesian statistics could be used to analyse data. A key recommendation of all
the authors is the need to improve communication between industry, physicians and
regulators in the regulatory process.
What is particularly striking about the EMEA, Rawlins and FDA papers are two
significant omissions. Apart from the industry paper, none of the three regulatory
papers mention any role for patients in the regulatory process. They are referred to as
beneficiaries of the process but never as contributors to the decision-making. This is
surprising as patients have been very influential in the rapid authorization of AIDS
medicines and in the orphan drug movement. It is not clear how patients could be
most effectively involved in promoting innovation and removing barriers but this is
clearly an area for research. The second striking omission is the absence of any
discussion of post-marketing surveillance as a critical component of the overall process.
The FDA diagram of the stages of the medicine development process omits Phase IV
from its description of all of the steps in medicine development (see Figure 8.3.1 in
Background Chapter 8.3).
Medicines development process
Preclinical studies: Most of the tools used for toxicology and human safety testing are
decades old and may fail to predict the specific safety problem that ultimately halts
development or that requires post authorization withdrawal. Each aspect of preclinical
safety studies (pharmacological ‘screening’ for unintended effects; pharmacokinetic
investigations in species used for toxicology testing; single- and repeat-dose toxicity
testing; and special toxicology testing (such as mutagenicity) has not been rigorously
tested by a robust analysis of its predictive power. The FDA has suggested that
scientists working within regulatory agencies have a wealth of application data
available to undertake such a critical review.
More generally, there are too few analytic tools (e.g., analytical devices, assay systems,
surrogate markers and cell culture methods) to assist in providing medicine safety and
effectiveness studies more quickly, with more certainty, and at lower cost. Key
enabling technologies involving the use of animals and the use of human tissue in
biomedical research are subject to complex regulation. Any increase in complexity of
regulation or indeed blocking of access to these technologies by public opinion
pressures has the potential to seriously disrupt basic and applied biomedical research.
Clinical development: Regulatory authorities are becoming more risk-averse. This lack
of flexibility only entrenches the existing regulatory requirements and perceptions, and
often results in the need for expanded studies to quantify potential adverse events.
Many requirements of medicines regulatory authorities are based on the opinion of
experts, rather than on a robust body of evidence to support the continuing inclusion
of each measure as a regulatory requirement. Randomized, controlled, blinded,
parallel-group clinical trials are not the only possible approach to investigating the
safety and efficacy of a new medicine. It may be that new methodological research to
critically evaluate alternative approaches, including actual experiments comparing
novel and traditional (RCT) designs, will find that medicines development can be
made more efficient without sacrificing safety and efficacy. Such alternative
approaches have been successfully used for high risk diseases such as cancer or AIDS
where accepting results from limited size studies combined with post-authorization
monitoring have allowed products to come to market far more quickly than by
conventional approaches. The role of patient groups in the AIDS example cannot be
underestimated. Indeed, patients and patients’ organizations could be equal
stakeholders participating in decision-making processes that occur in all aspects of
pharmaceutical innovation including: clinical trials design; regulatory processes;
treatment guidelines; marketing and pricing of medications.
There is often poor communication between the industry, physicians, and regulators
during medicines development. This results in requests for additional data and
regulatory questions following submission, and in turn these requests lead to
increasing unpredictability of outcomes and delays in the marketing authorization
process. Payers do not interact with the industry at an early enough stage in the
development process.
Post-marketing studies: In the four papers mentioned above, none deals with the
important role of post-marketing surveillance. In practice, there is relatively scant
attention paid to Phase IV post-marketing studies or systems. Computer-based
registries of patients and their clinical outcomes do not presently have sufficient data
on both incidence of exposure to the medicine and of adverse events in long-term,
post-marketing studies of the risks and benefits of medicines. European countries with
their national reimbursement systems are in a strong position to develop such
integrated systems while preserving patient confidentiality.
Reimbursement mechanisms: A lack of predictability about the timing and level of
reimbursement decisions leads to uncertainty among stakeholders. In Europe, each
national reimbursement system is primarily responsible for cost containment. This is
achieved by setting prices at a level that may not fully reward innovation and
sometimes by delaying decisions about reimbursement. These problems lead to
companies choosing to launch their products in the USA which, at present, has no such
single national system or government-based price control system.
Within its Sixth Framework Programme, the European Commission has recently called
for research proposals that include new approaches for accelerated development of
new, safe and more effective medicines. Bottlenecks and barriers in the current
medicines development process are to be identified and solutions elaborated to
overcome them. The project is expected to involve a range of stakeholders, such as
academia, clinicians, patient organizations, large and small industry, regulatory and
ethics specialists (Appendix 8.3.3).
As part of this call for proposals, it is recommended that the EU should create and
support a broad research agenda, so that every requirement within the medicines
development process, whether clinical or preclinical, is questioned for its regulatory
relevance, costing and predictive value. The involvement of the EMEA and the
various European national regulatory agencies and their scientists appears to be
critical to the success of this key initiative.
1
DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of drug
development costs. J Health Econ 2003;22(2):151-185.
2
Aspden P, editor. Medical innovation in the changing healthcare marketplace:
Conference summary. Washington, D.C.: National Academy Press; 2002. Available
from: http://www.nap.edu/catalog/10358.html (accessed 2 August 2004).