|
|
International Clinical Trials
|
Pharmaceutical safety is a continuum that begins with preclinical
animal testing, continues throughout human clinical trials and up to
market approval. However, rather than ending with its emergence on the
market, performing ongoing surveillance and assessment of new safety
information is critical to mitigating and managing the potential of a
pharmaceutical (drug or biologic) for adverse effects in patients, and
overall public health. Thus, planning for how pharmacovigilance is to
be performed upon approval, as well as constant re-evaluation of a
product’s benefit/risk profile throughout its life cycle, are of great
necessity.
Towards this goal, multinational initiatives conducted under the
International Conference on Harmonization (ICH) of Technical
Requirements for Registration of Pharmaceuticals for Human Use
auspices, along with the Council for International Organizations of
Medical Sciences (CIOMS) and other regulatory-related projects, have
attempted to foster a consistency of approach and accepted practices
industry-wide. At the same time, exciting advances in clinical
pharmacology, pharmacogenomics and pharmacogenetics have added to the
knowledge of how individual patients may respond to pharmaceutical
agents, and valuable insight into the natural history of varied
diseases has been gained through expanded use of pharmacoepidemiology
techniques.
Postmarketing pharmacovigilance and risk management are now truly
global; therefore, understanding national and international approaches
to signal detection, case evaluation and risk mitigation is of critical
importance. Just as significant is understanding the methods used to
communicate risk information to healthcare professionals and consumers,
and how the effectiveness of risk management programmes is to be
measured.
This article will examine current perspectives in two of the main ICH
regions (US and EU) and approaches to pharmacovigilance planning and
risk management throughout the medical product life cycle, from
clinical trials to product approval to marketing. The US Food and Drug
Administration (FDA) and European Medicines Agency (EMA) regulatory
requirements and standards, in combination with ICH guidelines, will be
discussed along with examples of risk assessment and minimisation
methods and the challenges that the industry faces in performing
high-quality risk management in a global environment.Harmonising Safety
Reporting Consultant Stephen A Goldman examines current perspectives
and approaches to pharmacovigilance planning and risk management
throughout the medical product life cycle, from clinical trials through
product approval to marketing.
FDA TASK FORCE
In 1998, an FDA task force was established to assess the system for
managing risks associated with the use of FDAapproved medical products.
The task force was asked to concentrate on the FDA’s role in three
basic areas: quality of its premarketing review and risk assessment;
strengths and weaknesses of the agency’s postmarketing surveillance and
risk assessment; and other the FDA risk assessment activities. The
resulting May 1999 report to the FDA Commissioner was a landmark
document that established FDA’s philosophy in this regard (1,2). In
combination with international safetyrelated initiatives under ICH (for
example, the E2A Guideline) and CIOMS (for example, CIOMS IV), the task
force report provided a framework for the FDA’s ongoing programmes and
initiatives for safe use of medical products with both national and
global applicability (3,4).
The task force examined all FDA risk management activities within the
context of the entire healthcare delivery system. Further, by assessing
the FDA’s role in both pre- and postmarketing phases, the task force
fostered the critical concept of risk management being a continuum
spanning all phases of a medical product’s life cycle.
In its report, the task force emphasised that responsibility for risk
management of medical products used in the US does not rest solely with
the FDA, but is shared by the FDA with industry, healthcare
professionals, patients, other federal groups, healthcare delivery
systems and professional organisations.
SAFETY REPORTING: NEW RULE
The report’s influence on the FDA’s risk management programmes is
apparent in regulatory actions taken in relation to the unsafe use of
marketed pharmaceuticals, such as coprescription of known
contraindicated drugs, and its emphasis on monitoring pharmaceutical
benefit/risk profiles in the 2003 safety reporting Proposed Rule (5,6).
Its effect outside the US is unclear – while barely mentioned in the
2001 Summary Report of the Heads of Agencies Ad Hoc Working Group, both
reports have common concerns and provided recommendations for
enhancement of public health in relation to risks associated with
medication use (7).
Under the 2003 Proposed Rule, currently mandated postmarketing
expedited submission of serious, unexpected adverse events and
associated follow-up information within 15 calendar days would also
require expedited reporting of information sufficient to consider
changes in administration of a marketed pharmaceutical based on
appropriate medical judgment, encompassing significant unexpected in
vitro, animal or human (clinical; epidemiological) study safety
findings or aggregate data from studies suggesting significant risk to
humans (for example, mutagenicity, teratogenicity or carcinogenicity)
(6). While this paradigm is considered start-of-the-art, and is
consistent with Volume 9A of the rules governing medicinal products in
the EU, it remains a proposed rule in the US (8).
This is in contrast with clinical trial safety reporting, as the FDA
released investigational new drug safety reporting requirements for
human drug and biological products in September 2010 – this rule became
effective on 28 March 2011 (9).
The new rule requires reporting of serious, unexpected, adverse drug
reactions for which “there is evidence to suggest a causal relationship
between the drug and the adverse event”. In an effort to enhance safety
signal detection, examples are given as to when a single event should
be reported (for example, uncommon events known to be strongly
associated with drug exposure), when there is need to wait for more
than one occurrence, and aggregate analysis of specific events (9,10).
In addition, other safety information now to be reported to the FDA
within 15 days of becoming aware of an occurrence includes findings
from clinical or epidemiological studies suggesting significant risk to
study participants; serious suspected adverse reactions occurring at
higher than expected rate; and serious adverse events from
bioavailability studies which determine what percentage and at what
rate a drug is absorbed by bloodstream, and bioequivalence studies
which determine whether a generic drug has the same bioavailability as
a brand name drug.
Under the new rule, definitions and reporting standards have been
revised for greater consistency with ICH and CIOMS so as to help ensure
harmonised reporting of globally conducted clinical trials.
RISK MINIMISATION
This finalised IND reporting rule is the latest component of the FDA’s
risk management activities. In 2005, the FDA published three risk
minimisation guidances: ‘Premarketing Risk Assessment’, ‘Development
and Use of Risk Minimization Action Plans [RiskMAPs]’ and ‘Good
Pharmacovigilance Practices and Pharmacoepidemiologic Assessment’ (11).
Regarding RiskMAPs, routine risk minimisation measures – such as
periodic updating of FDA-approved professional labelling to incorporate
information from postmarketing surveillance or studies revealing new
benefits or risk concerns – along with good reporting practices, were
thought by the FDA to be sufficient for most products, without the need
to consider a RiskMAP (12). A RiskMAP is a strategic safety programme
designed to meet specific goals and objectives in terms of minimising a
product’s known risks, while preserving its benefits; one or more
safety-related health outcomes or goals is targeted, and one or more
tools is used to achieve the goals set out. Tools that can be used to
achieve RiskMAP goals and objectives were classed into three
categories: targeted education and outreach; reminder systems; and
performance-linked access systems.
Targeted Education and Outreach
These tools include healthcare practitioner letters, training
programmes for healthcare practitioners or patients, continuing
education for healthcare practitioners, prominent professional or
public notifications, and patient labelling such as medication guides
and patient package inserts.
With respect to tool effectiveness, major categories of perceived risk
(drug-drug interactions, off-label use, recommended blood test
monitoring and teratogenicity) must also be part of the evaluative
process – as behaviours associated with each category of risk may
differ, so may communication methods utilised optimally (5). Multiple
modes of risk communication and maximal publicity can heighten overall
effectiveness; further, in assessing effectiveness of risk
communication, desired results must be clearly stated, as a fair degree
of achieved success may not be seen as effective enough to prevent
market withdrawal of the pharmaceutical product.
Reminder Systems
When targeted education and outreach tools are insufficient to minimise
risks, reminder systems tools are recommended for additional use.
Examples include healthcare provider training programmes; testing or
other documentation of physician knowledge and understanding; enrolment
of physicians, pharmacies and/or patients in special data collection
that reinforces appropriate use; and specialised systems or records
attesting to safety measures having been satisfied (for example,
prescription stickers and physician attestation of capabilities).
Performance-Linked Access Systems
When routine risk minimisation measures, targeted education and
outreach tools, and reminder systems are insufficient to minimise
risks, and products have significant or otherwise unique benefits in a
particular patient group or condition, but unusual risk – such as
irreversible disability or death – also exists, then performance-linked
access systems are recommended for use. These include systems that link
product access to laboratory testing results or other documentation,
such as a sponsor’s use of compulsory reminder systems, prescription
only by specially certified healthcare practitioners, dispensing only
by pharmacies or practitioners that elect special certification, and
dispensing only to patients with evidence or other documentation of
safe-use conditions (for example, lab test results). A successful
example of the latter is the programme for clozapine and
agranulocytosis (13,14).
The RiskMAP and the other two minimisation guidances are high quality
and of great utility in the US and abroad, but RiskMAPs remain under a
guidance, having not been incorporated into rules or regulations.
PHARMACOVIGILANCE PLANNING
The same situation exists in the US regarding the influential ICH
guideline, ‘E2E: Pharmacovigilance Planning’, crafted to foster
international harmonisation and consistency, especially in the early
postmarketing period of a new drug or biologic (15). Its underlying
principles are: planning pharmacovigilance activities throughout a
product’s life cycle; a science-based approach to risk documentation;
effective collaboration between regulators and industry; and
applicability of the pharmacovigilance plan across ICH regions.
The guideline focuses on a safety specification and pharmacovigilance
plan that might be submitted at the time of application for marketing
authorisation, with an annex describing pharmacovigilance methods. The
safety specification is a method for summarising important identified
and potential risks and missing information, including potentially
at-risk populations and situations where the medical product is likely
to be used that were not studied pre-approval. Thus, by using the
premarketing safety database and available worldwide experience and
scientific literature, factors that might affect the product’s
benefit/risk balance are identified for further evaluation. The
guideline also provides a proposed pharmacovigilance plan structure and
principles of good practice for design and conduct of observational
studies, but does not describe other methods to reduce risks from
products, such as risk communication. While not covering the entire
scope of pharmacovigilance, E2E encompasses the use of
pharmacoepidemiological studies, and is most useful for: new chemical
entities, biotechnology-derived products and vaccines; significant
changes in established products; and established products to be
introduced to new populations, with significant new indications, or
when a new major safety concern has arisen.
The pharmacovigilance plan should be based on the safety specification
and can be written as two parts of the same document. Normally
developed by the sponsor, it can be discussed with regulators during
product development, prior to approval (when marketing application
submitted), or when a safety concern arises postmarketing.
Continuous monitoring of the product’s safety profile, through signal
detection, issue evaluation, label updating and liaison with regulatory
authorities is considered standard as part of ‘routine
pharmacovigilance’ – this is in keeping with the 1999 FDA Task Force
Report, 2001 EU Summary Report of the Heads of Agencies, and the FDA’s
2003 Proposed Rule.
E2E was operationalised in all three ICH regions in 2005 – however, the
approach taken in Europe differs from that in the US. The then EMEA’s
‘Guideline on Risk Management Systems for Medicinal Products for Human
Use’ went into effect in November 2005 with an EU Risk Management Plan
(EU-RMP) that utilised E2E concepts and language – the guideline was
later incorporated into Volume 9A (16).
Circumstances when an EU-RMP is required are consistent with E2E, and consists of two elements:
- Part I: Safety Specification and Pharmacovigilance Plan (as in
E2E), with additional EU requirements as to potentials for overdose,
transmission of infectious agents, and misuse for illegal purposes in
the safety specification
- Part II: Evaluation of need for risk minimisation activities –
if there is need for additional (non-routine) risk minimisation
activities, RMP also includes risk minimisation plan that includes both
routine and additional risk minimisation activities
The guideline’s ‘Annex A: Epidemiological methods for PASS
[post-authorisation safety studies]’ is in line with E2E, with ‘Annex
B: Methods for Risk Minimization’ having elements in common with FDA’s
RiskMAP guidance. Annex C, the EURMP Template, was released in
September 2006 as a separate document. Under the guideline, an EU-RMP
is to be submitted with an application for a new marketing
authorisation, or an application entailing a significant change in a
prior marketing authorisation.
RISK EVALUATION & MITIGATION
The situation is different in the US – there are no corresponding FDA
regulatory requirements for submission of a safety specification and
pharmacovigilance plan with every new application for pharmaceutical
marketing authorisation, and RiskMAPs remain under guidance. However,
in September 2007, the FDA Amendments Act (FDAAA) was signed into law,
with a new category of mandatory risk management plan enacted: risk
evaluation and mitigation strategies (REMS) (17).
REMS, under FDAAA, are quite similar to RiskMAPs, as both are needed
only when judged necessary by the FDA in order to ensure that a drug’s
benefits outweigh its risks, and the same criteria are to be used to
make that determination looking at aspects such as, the seriousness of
the disease or condition to be treated and the expected benefits of the
drug. If a REMS is not required when an application is approved, it may
subsequently be required (including the FDA acting on supplemental
application for new indication) if there is awareness of new safety
information, and determination made that a REMS is necessary to ensure
the drug’s benefits outweigh risks.
Of the original list of drugs and biologics approved before FDAAA
provisions took effect on 25 March 2008 deemed to have REMS, all had
RiskMAPs in place prior to that determination (18). Drugs and biologics
approved before this date that only had a medication guide and no
elements to assure safe use (ETASUs) as defined in FDAAA, were not
deemed to have REMS under FDAAA. With respect to enforcement under
FDAAA, there are civil monetary penalties for violations of REMS
provisions, or a drug or biologic can be deemed misbranded and FDA
could obtain injunctive relief.
In September 2009, the FDA released a draft guidance stating that many
of the principles in the RiskMAP guidance were embodied in FDAAA REMS
provisions as implemented by the FDA (19). The RiskMAP guidance
continues to apply to products with existing RiskMAPs (for example,
those not deemed to have an approved REMS in effect) and those with new
RiskMAPs (such as ANDAs for which reference listed drug has RiskMAP).
Products previously approved with a medication guide or patient package
insert that meets statutory requirements for REMS will now be required
to have REMS, and the draft guidance reiterates that REMS are subject
to inspection and enforceable. The content of a proposed REMS includes
the product and contact information, goals, additional potential REMS
elements, ETASUs, an implementation system, and a timetable for
submission of assessment of the REMS. The content of a REMS supporting
document includes background, goals section, supporting information
about proposed REMS elements, REMS Assessment Plan, and other relevant
information.
As for new developments in the EU, there is an ongoing EMA benefit-risk
methodology project, which started in early 2009 and is scheduled to
run until the end of 2011. It has five steps and is being conducted in
collaboration with academia such as experts in decision theory from the
London School of Economics and Political Science, and with the
University of Groningen. Through this project, the EMA aims to make its
decision-making on the benefits and risks of medicines more
transparent, more consistent and easier to audit (20).
To date, two work package reports have been released. The first (in
March 2010) provided a description of the current practice of
benefit-risk assessment for centralised procedure products in the EU
(21). In the second (from August 2010), approaches for balancing
benefits and risks in decisionmaking about medicinal products were
reviewed, with perceived usefulness to regulators in making pre- and
postapproval decisions completing each review (22).
NEW AMENDMENTS: EU
With respect to regulation, on 22 September 2010 the European
Parliament adopted a proposal for a regulation and directive amending,
as regards pharmacovigilance, regulation (EC) 726/2004 and 1394/2007
Directive 2001/83/EC of the Community code relating to medicinal
products for human use which was published in the Official Journal of
the EU on 31 December 2010 (23). This Directive enters into force on
the 20th day following its publication in the Official Journal of the
European Union (that is, Sunday 20 January 2011), but member states
have until 21 July 2012
to adopt its provisions. The regulation is not due to come into force
until 2 July 2012. Among the amendments which reinforces existing
requirements are that:
- The marketing authorisation holder (MAH) should establish a
pharmacovigilance system to ensure monitoring and supervision of one or
more of its authorised medicinal products, recorded in a
pharmacovigilance system master file permanently accessible for
inspection. A summary of the pharmacovigilance system should be
submitted with the marketing authorisation application and include a
reference to the site where the pharmacovigilance system master file
for the medicinal product concerned is maintained and accessible for
inspection by the competent authorities.
- Planning of pharmacovigilance for each individual medicinal
product by the MAH should take place in the context of a risk
management system and should be proportionate to identified risks,
potential risks and the need for additional information on the
medicinal product. It should also be ensured that any key measures
contained in a risk management system are included in the marketing
authorisation as conditions.
- National competent authorities are to make publicly available
the marketing authorisation, package leaflet, summary of product
characteristics (SmPC) and any deadlines for fulfilment of conditions
where necessary for each medicinal product which they have authorised –
the public assessment report shall include a summary written in a
manner understandable to the public.
- MAHs can be required to conduct post-authorisation studies on
safety and efficacy at time of marketing authorisation or later, and it
should be part of marketing authorisation.
- All medicinal products with a new active substance and
biological medicinal products including biosimilars for which
pharmacovigilance activities are prioritised are authorised subject to
additional monitoring. This may also apply at the request of competent
authorities to specific products, subject to requirement to conduct a
post-authorisation safety study or where there are conditions or
restrictions with regard to safe and effective use of the medicinal
product.
- Products subject to additional monitoring should be identified
by a black symbol and corresponding explanatory sentence on SmPC and
patient information leaflet.
- In order to increase transparency on pharmacovigilance
processes, member states should create and maintain medicines
web-portals. To the same end, MAHs should provide authorities with
prior or simultaneous warnings about safety announcements and
authorities should provide each other with such warnings.
References
- US Department of Health and Human Services, Food and Drug
Administration, Task Force on Risk Management, Report to the FDA
Commissioner: Managing the Risks from Medical Product Use: Creating a
Risk Management Framework, Rockville, Maryland, 1999
- Goldman SA, US Postmarketing Pharmacovigilance Compliance in the
Midst of Regulatory Uncertainty, Food Drug Law J 62(3): pp513-528, 2007
- International Conference on Harmonization of Technical
Requirements for Registration of Pharmaceuticals for Human Use,
Guideline E2A: Clinical Safety Data Management: Definitions and
Standards for Expedited Reporting, Step 4 version, 1994
- Council for International Organizations of Medical Sciences,
CIOMS IV Working Group, Benefit-Risk Balance for Marketed Drugs:
Evaluating Safety Signals, Geneva, Switzerland, 1998
- Goldman SA, Communication of Medical Product Risk: How Effective Is Effective Enough?, Drug Safety 27: pp519-534, 2004
- FDA, Safety Reporting Requirements for Human Drug and Biological
Products: Proposed Rule, Federal Register 68: pp12,406-12,497, 2003
- Summary Report of the Heads of Agencies Ad Hoc Working Group, Establishing a European Risk Management Strategy, 2003
- European Commission, Volume 9A of the Rules Governing Medicinal
Products in the European Union: Guidelines on Pharmacovigilance for
Medicinal Products for Human Use, 2008
- FDA, Investigational New Drug Safety Reporting Requirements for
Human Drug and Biological Products and Safety Reporting Requirements
for Bioavailability and Bioequivalence Studies in Humans: Final Rule,
Federal Register 75: pp59,935-59,963, 2010
- FDA, Center for Drug Evaluation and Research & Center for
Biologics Evaluation and Research, Draft Guidance for Industry and
Investigators: Safety Reporting Requirements for INDs and BA/BE
Studies, 2010
- FDA, www.fda.gov/NewsEvents/Newsroom/ PressAnnouncements/2005/ucm108425.htm
- FDA, Center for Drug Evaluation and Research & Center for
Biologics Evaluation and Research, Guidance for Industry: Development
and Use of Risk Minimization Action Plans, 2005
- Clozaril Tablets Label (Novartis Pharmaceuticals Corporation),
January 2010, www.accessdata.fda.gov/
drugsatfda_docs/label/2010/019758s062lbl.pdf
- Novartis Pharmaceuticals Corporation, Drug Warning and
Information [Clozaril], 2005 www.fda.gov/downloads/
Safety/MedWatch/SafetyInformation/SafetyAlertsforHuman
MedicalProducts/UCM153074.pdf
- FDA, Center for Drug Evaluation and Research & Center for
Biologics Evaluation and Research, Guidance for Industry: E2E
Pharmacovigilance Planning, 2005
- European Medicines Agency, Committee for Medicinal Products for
Human Use, Guideline on Risk Management Systems for Medicinal Products
for Human Use, 2005
- FDA Amendments Act of 2007,
http://frwebgate.access.gpo.gov/cgi-bin/getdoc.cgi?
dbname=110_cong_public_laws&docid=f:publ085.110
- FDA, Identification of Drug and Biological Products Deemed to
Have Risk Evaluation and Mitigation Strategies for Purposes of the Food
and Drug Administration Amendments Act of 2007: Notice, Federal
Register 73: pp16,313-16,314, 2008
- FDA, Center for Drug Evaluation and Research & Center for
Biologics Evaluation and Research, Draft Guidance for Industry: Format
and Content of Proposed Risk Evaluation and Mitigation Strategies
(REMS), REMS Assessments, and Proposed REMS Modifications, 2009
- www.ema.europa.eu/ema/index.jsp?curl=pages/
news_and_events/news/2010/10/news_detail_001129.jsp&m
url=menus/news_and_events/news_and_events.jsp&mid=WC0
b01ac058004d5c1&jsenabled=true
- European Medicines Agency, Benefit-Risk Methodology Project:
Description of the Current Practice of Benefit-Risk Assessment for
Centralised Procedure Products in the EU Regulatory Network, 2010
www.ema.europa.eu/docs/en_GB/
document_library/Report/2010/04/WC500089603.pdf
- European Medicines Agency, Work Package 2 Report: Applicability
of Current Tools and Processes for Regulatory Benefit-Risk Assessment,
2010
- The Pharmacovigilance Regulation (EU) 1235/2010 amending
Regulations (EC) 726/2004 and (EC) 1394/2007 Official Journal of the
European Union on 31 December 2010, http://eurlex.europa.eu/LexUriServ/
LexUriServ.do?uri=OJ:L:2010:348:0001:0016:EN:PDF. The corresponding
Pharmacovigilance Directive 2010/84/EU amending Directive 2001/83/EC
was also published in the same edition of the Official Journal of the
European Union, http://eur-lex.europa.eu/LexUriServ/
LexUriServ.do?uri=OJ:L:2010:348:0074:0099:EN:PDF
Acknowledgement
The author wishes to thank Brian Edwards, MB, MD, MRCP for his editorial contributions
|
Read full article from PDF >>
|
 |
 |
 |
Rate this article |
You must be a member of the site to make a vote. |
|
Average rating: |
0 |
| | | | |
|
|
Stephen A Goldman is an independent consultant who is Managing Member of Stephen A Goldman Consulting Services, LLC (Morris Plains, NJ, US) and former Medical Director of MedWatch, US FDA. Internationally, he provides pharmaceutical and medical device companies with safety-related services (consulting, auditing and training) involving clinical trials, postmarketing vigilance, signal detection and evaluation, regulatory compliance as well as risk management. He previously served as Director, Pharmacoepidemiology at Knoll Pharmaceutical Company (Abbott Laboratories) after an extended FDA service – during which he completed a three-year fellowship in Clinical Pharmacology and Regulatory Drug Evaluation Sciences prior to joining MedWatch. A board-certified psychiatrist, he is a Fellow of the Academy of Psychosomatic Medicine (FAPM) and Distinguished Fellow of the American Psychiatric Association (DFAPA). Email: sagcs@aol.com
|
|
 |
 |
 |
|

 |
White Papers |
 |
Securing the Global Pharmaceutical Supply Chain against the Threat of Counterfeit Drugs
World Courier
With a fully integrated GxP-compliant transport, storage and
distribution system in place and over 140 wholly owned ISO 9001 -
certified offices in more than 50 countries, WORLD COURIER, an
AmerisourceBergen® company, is the world’s largest and most experienced
provider of specialty courier services. It is uniquely positioned to
meet the most demanding industry requirements for managing the global
distribution of time - and temperaturesensitive pharmaceutical products
and IMPs used commercially or in clinical trials.
More info >> |
|
|