Latest
developments in ICH
EMEA: Enhancement of International activities
• EMEA Roadmap: Enhancement of International
activity to better protect public
health, facilitate access to medicines and stimulate innovation
• EC-EMEA Bilateral relationships: Confidentiality
arrangements with USA (2003), Japan (2007), Canada (2007)
• Mutlinational relatioships: ICH remains the key
forum for harmonisation of human
pharmaceuticals. ICH is an important technical
basis for our international collaboration success
Over 50 ICH Guidelines finalised, new, under revision
• Efficacy
- 14
topics/17 guidelines
• Safety
- 8 topics/16
guidelines
• Quality
- 9 topics/23
guidelines
• Medical Dictionary
- MedDRA
• Electronic Standards
- eCTD,
ESTRI, E2B,
• Common Technical Document- CTD
Guidelines extend over entire product life cycle
(ref:http://www.emea.europa.eu/docs/en_GB/document_library/Presentation/2009/11/WC500010482.pdf)
Purpose
The purpose of ICH is to reduce or obviate the need
to duplicate the testing carried out during the research and development of new
medicines by recommending ways to achieve greater harmonisation in the
interpretation and application of technical guidelines and requirements for
product registration. Harmonisation would lead to a more economical use of
human, animal and material resources, and the elimination of unnecessary delay
in the global development and availability of new medicines while maintaining
safeguards on quality, safety, and efficacy, and regulatory obligations to
protect public health.
ICH guidelines have been adopted as law in several
countries, but are only used as guidance for the U.S. Food and Drug
Administration.
Structure
The ICH has four major parts:
ICH Steering Committee,
ICH Coordinators,
ICH Secretariat and
ICH Working Groups.
The Steering Committee, made of six ICH Parties,
governs the ICH, determining the policies and procedures, selecting topics for
harmonisation and monitoring progress of harmonisation initiatives. The ICH
consists of:
European Commission,
European Federation of Pharmaceutical Industries and
Associations (EFPIA)
Ministry of Health, Labour and Welfare (Japan)
Japan Pharmaceutical Manufacturers Association
(JPMA)
Food and Drug Administration (FDA)
Pharmaceutical Research and Manufacturers of America
(PhRMA)
The ICH Coordinators represents each ICH Party to
the ICH Secretariat on a day-to-day basis.
The ICH Secretariat is primarily concerned with
preparations for, and documentation of, meetings of the Steering Committee as
well as coordination of preparations for Working Group (EWG, IWG, Informal WG)
and Discussion Group meetings.
The ICH Working Groups are created by the Steering
Committee when a new topic is accepted for harmonisation, and is charged with
developing a harmonised guideline that meets the objectives outlined in the
Concept Paper and Business Plan.
(Ref: http://www.ich.org/about/faqs/article/how-is-ich-structured.html)
Implications
In the field of drug efficacy and safety ICH has
produced 26 guidelines describing technical requirements related to the process
of registration of new chemical entities and products obtained by
biotechnology. As already mentioned, the scientific level of each guideline is
high and reflects state-of-the-art technology.
Although intended primarily for ICH countries,
guidelines related to drug safety and efficacy may also be of use to non-ICH
countries to create awareness of the subject, and for reference and guidance
purposes for national drug regulatory authorities when producing national
legislation. They may also be used for education and training purposes. This
applies particularly to ICH guidelines in the safety area (guidelines S1A to
ICH S7, especially those on carcinogenicity and genotoxicity). It should be
stressed that the majority of ICH guidelines in the areas of safety and
efficacy deal with topics for which no documents of a regulatory nature
previously existed, which considerably to their usefulness.
It should be recognized that WHO has produced only a
limited number of materials on topics concerned with safety and efficacy. They
relate to good clinical practice (GCP), for which WHO has produced guidelines,
and to reporting of adverse drug reactions, for which WHO maintains its Centre
for International Drug Monitoring in Uppsala, Sweden. As the WHO GCP guidelines
are now in the process of revision, it is intended that the revised version
should indicate the material contained in ICH's guideline.
Attempts to apply some ICH guidelines related to
drug safety and efficacy in practice in non-ICH countries can give rise to
considerable difficulties, especially when full implementation is attempted in
countries that lack adequate resources. Specific examples of such difficulties
are presented below.
ICH guideline E5, "Ethnic factors in the
acceptability of foreign clinical data", recommends a framework for
evaluating the impact of ethnic factors on the effect of the medical product.
Some commentators claim that the guideline may limit the scope of decision of
national regulatory authorities as to the need for clinical studies to be
conducted in the country. This may create problems with the verification of the
actual efficacy of drugs in local situations, and may also create obstacles for
local pharmaceutical development. At the same time, the need for local clinical
trials must be scientifically justified.
ICH guideline E2C "Clinical safety data
management: periodic safety update reports (PSUR) for marketed drugs",
which lays down a format and content for comprehensive periodic safety updates
and recommends a six-monthly periodicity for such update reports, is highly
resource-intensive. The resources required to maintain the system are beyond
the means of most non-ICH countries. It may be considered that non-ICH countries
would benefit more from a digest of work done by the ICH drug regulatory
authorities on each individual product, provided such digests are produced and
suitably disseminated.
The ICH GCP guideline (E6, "Good Clinical
Practice: consolidated guideline") deals with the planning, conduct,
monitoring and reporting of clinical trials. Its object is to facilitate the
mutual acceptance of clinical trial data in ICH countries. However, WHO's
constituency includes all the world's cultures. The purpose of the WHO GCP
guidelines is to provide globally applicable guidance for the conduct of
clinical trials, thereby assuring the ethical and scientific integrity of
research.
The ICH GCP guidelines are used by non-ICH countries
to develop their own GCP guidelines. They are more detailed and user-friendly
than current WHO GCP guidelines, but they do not address country-specific
issues. The conduct of clinical trials should be linked to scientific
justification. There is a need for WHO to revise its current guidelines taking
these factors into account. The WHO GCP guidelines are currently being revised
to adopt a modular approach that will enhance their educational value.
ICH guideline M3, "The timing of non-clinical
safety studies for the conduct of human clinical trials for
pharmaceuticals", is important for regulatory authorities that control and
assess product development prior to drug product approval. It also provides the
pharmaceutical industry with guidance on the approach that should be taken to
perform various tests prior to submission of evidence to regulatory
authorities. It would be useful to non-ICH countries for better awareness and
understanding of requirements so that they could establish national approval
requirements appropriate to their situation.
ICH guideline M1, "Medical terminology"
(including Medical Dictionary for Regulatory Activities Terminology, MedDRA
Terminology), was developed as a medical terminology covering all aspects of
drug regulation ranging from authorization to post-marketing surveillance. It
contains terms for adverse drug reactions (ADRs) and also for diseases,
investigations and patient history. It includes terms from the WHO Adverse
Reaction Terminology (WHO-ART) and ICD-9, but uses a different structure and
many more terms. The advantages of using MedDRA have been stated as specificity
for data entry, flexibility for data retrieval, and standardization for
international communication. However, there are significant problems in the use
of MedDRA by non-ICH countries, particularly difficulties in installing and
using the terminology and its cost. Recommendations by WHO to ICH to retain
WHO-ART as a pharmacovigilance subset of MedDRA and to involve WHO in the
review of MedDRA were not considered by ICH to be workable. ICH's position has
forced WHO to continue with the further development of WHO-ART. The
relationship of WHO-ART with ICD-9 and 10 also has important public health and
epidemiology aspects. There is no satisfactory proposal from ICH to address
these points.
A new ICH initiative to develop activities in the
area of post-marketing surveillance was proposed at the Fifth International
Conference on Harmonization (November 2000). Three topics were suggested in the
first instance:
(1) periodic safety update information;
(2) safe roll-out of new drug products;
(3) case reporting.
(Ref : http://apps.who.int/medicinedocs/en/d/Jh2993e/7.html)
Guidance
notes
The ICH topics are divided into four categories and
ICH topic codes are assigned according to these categories.
1.
Quality Guidelines
Harmonisation achievements in the Quality area
include pivotal milestones such as the conduct of stability studies, defining
relevant thresholds for impurities testing and a more flexible approach to
pharmaceutical quality based on Good Manufacturing Practice (GMP) risk
management.
Stability Q1A - Q1F
Analytical Validation Q2
Impurities Q3A - Q3D
Pharmacopoeias Q4 - Q4B
Quality of Biotechnological Products Q5A - Q5E
Specifications Q6A- Q6B
Good Manufacturing Practice Q7
Pharmaceutical Development Q8
Quality Risk Management Q9
Pharmaceutical Quality System Q10
Development and Manufacture of Drug Substances Q11
Cross-cutting Topics
2.
Safety Guidelines
ICH has produced a comprehensive
set of safety Guidelines to uncover potential risks like . testing strategy for
assessing the QT interval prolongation liability: the single most important
cause of drug withdrawals in recent years.
Zip file with all ICH Safety Guidelines in Word
format
Carcinogenicity Studies S1A - S1C
Genotoxicity Studies S2
Toxicokinetics and Pharmacokinetics S3A - S3B
Toxicity Testing S4
Reproductive Toxicology S5
Biotechnological Products S6
Pharmacology Studies S7A - S7B
Immunotoxicology Studies S8
Nonclinical Evaluation for Anticancer
Pharmaceuticals S9
Photosafety Evaluation S10
Cross-cutting Topics
3.
Efficacy
Guidelines
The work carried out by ICH under the Efficacy
heading is concerned with the design, conduct, safety and reporting of clinical
trials. It also covers novel types of
medicines derived from biotechnological processes and the use of
pharmacogenetics/ pharmacogenomics techniques to produce better targeted
medicines.
Zip file with all Efficacy Guidelines in Word format
Clinical Safety E1 - E2F
Clinical Study Reports E3
Dose-Response Studies E4
Ethnic Factors E5
Good Clinical Practice E6
Clinical Trials E7 - E11
Clinical Evaluation by Therapeutic Category E12
Clinical Evaluation E14
Pharmacogenomics E15 - E16
Cross-cutting Topics
4.
Multidisciplinary
Guidelines
Those
are the cross-cutting topics which do not fit uniquely into one of the Quality,
Safety and Efficacy categories. It
includes the ICH medical terminology (MedDRA), the Common Technical Document
(CTD) and the development of Electronic Standards for the Transfer of Regulatory
Information (ESTRI).
Zip file with all Multidisciplinary Guidelines in
Word format
MedDRA Terminology M1
Electronic Standards M2
Nonclinical Safety Studies M3
Common Technical Document M4
Data Elements and Standards for Drug Dictionaries M5
Gene Therapy M6
Genotoxic Impurities M7
Electronic Common Technical Document (eCTD) M8
(Ref: http://www.ich.org/products)
Inspections
INDIAN / USA / EU Ethics approval system
INDIAN
Regulatory inspections are
important to evaluate the integrity of the data submitted to health authorities
(HAs), protect patient safety, and assess adequacy of site/sponsor quality
systems to achieve the same. Inspections generally occur after submission of
data for marketing approval of an investigational drug. In recent years, there
has been a significant increase in number of inspections by different HAs,
including in India. The assessors/inspectors generally do a thorough review of
site data before inspections. All aspects of ICH-GCP, site infrastructure, and
quality control systems are assessed during the inspection. Findings are
discussed during the close out meeting and a detailed inspection report issued
afterward, which has to be responded to within 15–30 days with effective
Corrective and Preventive Action Plan (CAPA). Protocol noncompliance,
inadequate/inaccurate records, inadequate drug accountability, informed consent
issues, and adverse event reporting were some of the most common findings
observed during recent Food and Drug Administration (FDA) inspections. Drug
development is being increasingly globalized and an increased number of
patients enrolled in studies submitted as part of applications come from all
over the world including India. Because of the steep increase in research
activity in the country, inexperienced sites, and more stakeholders, increased
efforts will be required to ensure continuous quality and compliance. HAs have
also made clear that enforcement will be increased and be swift, aggressive,
and effective.
Regulatory inspections are an
important and essential part of clinical research to evaluate the integrity of
the data submitted to health authorities (HAs), presence of infrastructure to
conduct clinical research, measures implemented to protect patient’s interest
and safety, adequacy of site/sponsor quality systems, and verification of
compliance with the principles of ICH-GCP as well as local regulations.
Inspections
generally occur after submission of data for marketing approval of an
investigational drug; however, inspections may happen at any time during the
conduct of the trial like FDA’s Early Intervention Program.
All HAs like
US FDA, EMA (European Medicines Agency) and others to whom data have been
submitted from Indian site(s) may conduct inspections at the respective study
sites.
Drug
Controller General India (DCGI) has jurisdiction to inspect all clinical trials.
DCGI has conducted inspections to investigate issues, where required. A routine
inspection program will also start in near future.
A closer
look at recent inspection frequency figures suggests a significant number of
inspections occurring outside the US including in the Asia Pacific region. In
fiscal year 2009, FDA Center for Drug Evaluation Research (CDER) clinical
investigator inspections totaled 458 and 119 of those (26%) were outside the
US. The FDA Clinical Investigator Inspection List (CLIL) website, an online
posting of inspections with final classifications, shows 78 investigator site
inspections in the Asia-Pac region, out of which 14 (6 in 2009) have been in
India.
The results
have been positive with either “No Action Indicated” or “Voluntary action
Indicated.” The findings were in categories of Drug Accountability, Protocol
Compliance, Inaccurate Records, and Failure to follow Investigational Plan.
Because of
the steep increase in research activity, inexperienced sites, and more
stakeholders, a lot of effort will be required to ensure continuous quality and
compliance to maintain this positive trend in inspection outcomes.
EMA has also
increased inspections in the region with India being one of the most inspected
countries (eight inspections between 1997 and 2008) outside the EU (European
Union). This is in keeping with the increased number of subjects recruited from
the region (11.6% in pivotal studies submitted in 2007 versus 4% for
submissions between 2005 and 2007) for the pivotal studies submitted to EMA for
approval
(Ref : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3043366/)
USA
This guidance is intended to provide information
about FDA inspections of clinical investigators
conducted under FDA’s Bioresearch Monitoring (BIMO)
Program. This document supersedes
FDA’s Information Sheet Guidance, "FDA Inspections
of Clinical Investigators," dated January
2006. This document has been revised to provide
updated information and is being issued in
accordance with the agency’s regulations on Good
Guidance Practices (21 CFR 10.115).
FDA's guidance documents, including this guidance,
do not establish legally enforceable
responsibilities. Instead, guidances describe the
agency's current thinking on a topic and should
be viewed only as recommendations, unless specific
regulatory or statutory requirements are
cited. The use of the word should in agency
guidances means that something is suggested or
recommended, but not required.
FDA developed its BIMO Program to help ensure the
protection of the rights, safety, and welfare
of human research subjects involved in FDA-regulated
clinical trials, to verify the accuracy and
reliability of clinical trial data submitted to FDA
in support of research or marketing applications, and to assess compliance with
statutory requirements and FDA's regulations governing the conduct of clinical trials.
Among other activities, the FDA BIMO Program
involves site visits to clinical investigators,
sponsors, monitors, contract research organizations,
Institutional Review Boards (IRBs), nonclinical
(animal) laboratories, and bioequivalence
analytical laboratories. This document addresses
site visits to clinical investigators who conduct
clinical investigations that are regulated by FDA
under 21 USC 355(i) or 21 USC 360j(g).
WHEN
ARE CLINICAL INVESTIGATOR INSPECTIONS CONDUCTED?
FDA conducts clinical investigator inspections to
determine if the clinical investigators are
conducting clinical studies in compliance with
applicable statutory and regulatory requirements.
Clinical investigators who conduct FDA-regulated
clinical investigations are required to permit
FDA investigators to access, copy, and verify any
records or reports made by the clinical
investigator with regard to, among other records,
the disposition of the investigational product
and subjects’ case histories. See 21 CFR 312.68 and
812.145. The FDA investigator typically
performs this oversight function through on-site
inspections designed to document how the study
was actually conducted at the clinical
investigator’s site. For investigational drug studies,
clinical investigators must retain study records for
a period of two years following the date a
marketing application is approved for the drug for
the indication for which it is being
investigated; or, if no application is to be filed
or if the application is not approved for such
indication, until two years after the investigation
is discontinued and FDA is notified. See 21
CFR 312.62(c). For investigational device studies,
clinical investigators must retain study
records for a period of two years after the latter
of the following two dates: The date on which
the investigation is terminated or completed, or the
date that the records are no longer required
for purposes of supporting a premarket approval
application or a notice of completion of a
product development protocol. See 21 CFR 812.140(d).
FDA conducts both announced and unannounced
inspections of clinical investigator sites,
typically under the following circumstances:
• to verify the accuracy and reliability of data
that has been submitted to the agency;
• as a result of a complaint to the agency about the
conduct of the study at a particular
investigational site;
• in response to sponsor concerns;
• upon termination of the clinical site;
• during ongoing clinical trials to provide
real-time assessment of the investigator’s
conduct of the trial and protection of human
subjects;
• at the request of an FDA review division; and
• related to certain classes of investigational products
that FDA has identified as products
of special interest in its current work plan (i.e.,
targeted inspections based on current
public health concerns).
HOW
ARE CLINICAL INVESTIGATOR INSPECTIONS CONDUCTED?
Upon arrival at the clinical investigator study
site, the FDA investigator will display his/her FDA
credentials and issue a completed Form FDA 482
(Notice of Inspection) to the clinical
investigator or appropriate study staff. FDA is
authorized at reasonable times to access, inspect,
and copy any required records related to the
clinical investigation. See section 704 of the
Federal Food, Drug, and Cosmetic Act (21 USC 374),
21 CFR 312.68, and 21 CFR 812.145.
During an inspection at the site of a clinical
investigator, the FDA investigator typically verifies
compliance with the regulations governing the use of
investigational products and human subject
protections at 21 CFR parts 50, 56, 312, and/or 812,
by inspecting records and talking to
individuals involved in the conduct of the study to
ascertain:
• who performed various aspects of the protocol for
the study (e.g., who verified inclusion
and exclusion criteria, who obtained informed
consent, who collected adverse event
data);
• whether the IRB approved the protocol, informed
consent form, and any amendments to
the protocol prior to implementation;
• whether the clinical investigator and study staff
adhered to the sponsor’s protocol and
investigational plan and whether protocol deviations
were documented and reported
appropriately;
• whether informed consent documents were signed by
the subject or the subjects’ legallyauthorized representative prior to entry in
the study (i.e., performance of any studyrelated procedures);
• whether authority to conduct aspects of the study
was delegated, and if so, how the
conduct of the study was supervised by the clinical
investigator2;
• where specific aspects of the investigation were
performed;
• how the study data were obtained and where the
study data were recorded;
• accountability for the investigational product,
including shipping records and disposition
of unused investigational product;
• whether the clinical investigator disclosed
information regarding his financial interests to
the sponsor and/or interests of any
subinvestigator(s), spouse(s) and dependent children3;
• the monitor’s communications with the clinical
investigator;
• the monitor’s evaluations of the progress of the
investigation; and
• corrective actions in response to previous FDA
inspections, if any, and regulatory
correspondence or sponsor and/or monitor
correspondence.
The FDA investigator also may audit the study data
by comparing the data filed with the agency
or the sponsor, if available, with records related
to the clinical investigation. Such records may
include the case report forms and supporting source
documentation including signed and dated
consent forms and medical records including, for
example, progress notes of the physician, the
subject’s hospital chart(s), and the nurses’ notes.
These records may be in hard copy and/or an
electronic format. For electronic records and/or
electronic signatures, the FDA investigator may
gather information to determine whether 21 CFR part
11 requirements have been met. FDA may also examine subjects’
medical records that are part of the clinical investigation and
predate the study to verify whether the condition
under study was in fact diagnosed, the study
eligibility criteria were met, and whether the
subject received any potentially interfering
medication prohibited by the protocol.
The FDA investigator may also review subjects’
records covering a period after
completion of study-related activities to determine if there was proper follow-up as outlined in the protocol, and if
the clinical investigator submitted all reportable adverse events (including all clinical signs
and symptoms). See 21 CFR 312.64(b) and 812.150(a).
For more information about the procedures FDA uses during inspections of
clinical investigator sites, see FDA’s
Compliance Program Guidance Manual (CPGM), Bioresearch Monitoring, Clinical
Investigators and Sponsor-Investigators, Program 7348.811.5.
INTERNATIONAL
INSPECTIONS
FDA’s inspection of clinical investigators is not
limited to the United States (U.S.). International
inspections are generally conducted when the studies
are part of a marketing application
submitted to FDA and provide data critical to
decision-making on product approval. See FDA’s
CPGM, Program 7348.811, “Clinical Investigators and
Sponsor-Investigators.”6
Such assignments include studies that are
conducted under an FDA investigational new drug
application (IND), as well as studies at non-U.S.
sites that are not conducted under an IND or
under an investigational device exemption (IDE).
(Ref: http://www.fda.gov/downloads/RegulatoryInformation/Guidances/UCM126553.pdf)
EU
EU
harmonisation
The European Medicines Agency plays an important
role in the harmonisation and co-ordination of GCP-related activity at an EU
level. It is involved in:
co-ordinating GCP inspections for the centralised
procedure;
preparing guidance on GCP topics through the work of
the GCP Inspectors Working Group;
co-ordinating advice on the interpretation of EU GCP
requirements and related technical issues;
developing of EU-wide guidelines on GCP inspections
and related procedures for the centralised procedure.
For more information on clinical trial
authorisation, safety monitoring, GCP inspections, and GCP and GMP requirements
for clinical trials in the European Economic Area (EEA), see volume 10:
clinical-trial guidelines of the rules governing medicinal products in the EU.
International
clinical trials
Regardless of where they are conducted, all clinical
trials included in applications for marketing authorisation in the EEA must be
in accordance with:the GCP Directive (Directive 2001/83/EC Annex I, as amended
by Directive 2003/63/EC);the ethical standards of the Clinical Trials Directive
(Directive 2001/20/EC).
In July 1996, the EU adopted the guideline for good
clinical practice, which lays out unified GCP standards for Europe, the United
States of America and Japan.The Council for International Organizations of
Medical Science (CIOMS);the World Medical Association.
Clinical
trials conducted in countries outside the EU
The number of clinical trials and clinical-trial
subjects outside Western Europe and North America has been increasing for a
number of years. The Agency has been tracking the geographic origins of
patients included in pivotal trials submitted in marketing authorisations to
the centralised procedure.
In April 2012, the Agency published the final
version of the reflection paper on ethical and GCP aspects of clinical trials
of medicinal products for human use conducted outside of the EU / EEA and submitted
in marketing authorisation applications to the EU regulatory authorities. The
aim of the paper is to strengthen existing processes to provide assurance to
regulators and stakeholders that clinical trials meet the required ethical and
GCP standards, no matter where in the world they have been conducted.
The reflection paper is part of the Agency’s
strategy developed to address the challenges arising from the increasing
globalisation of clinical research. These challenges are addressed in a two-fold
manner, by: putting forward concrete steps for international cooperation
between regulatory authorities in the regulation of clinical trials, with a
specific emphasis on capacity-building initiatives for a common approach to
oversight of trials and to ensure a robust framework for the oversight and
conduct of clinical trials;clarifying and determining the practical steps by
which EU regulators will gain assurance that ethical and GCP standards are
applied to clinical trials for human medicines, both during the development and
during the marketing-authorisation-application phase.
Working
Group on Clinical Trials conducted outside of the EU / EEA
The Agency's Working Group on Clinical Trials
Conducted Outside of the EU / EEA was established in 2009 to develop practical
proposals for tasks and procedures or guidance
in the four areas identified in the strategy paper on the acceptance of
clinical trials conducted outside of the EU/EEA:clarify the practical
application of ethical standards for clinical trials, in the context of the
Agency's activities;determine the practical steps to be undertaken during the
provision of guidance and advice in the drug-development phase;determine the
practical steps to be undertaken during the marketing-authorisation phase;international
cooperation in the regulation of clinical trials, their review and inspection
and capacity building in this area and to develop a reflection paper on ethical
and GCP aspects of clinical trials conducted outside the EU / EEA.
The Working Group includes representatives of the
Committee for Medicinal Products for Human Use (CHMP), Paediatric Committee
(PDCO), Committee for Orphan Medicinal Products (COMP), Coordination Group for
Mutual Recognition and Decentralised Procedures (CMD), Clinical Trials
Facilitation Group, GCP inspectors, Patients' and Consumers' Working Party,
Healthcare Professionals' Working Group, Agency secretariat and European
Commission.
The
Working Group meets on a regular basis at the Agency. Collaboration
with the Food and Drug Administration The Agency and the
United States Food and Drug Administration (FDA) agreed to launch a joint
initiative to collaborate on international GCP inspection activities in July
2009.
The initiative forms an important contribution to
ensuring the protection of clinical-trial subjects in the context of the
increasing globalisation of clinical research. The initiative comes under the
scope of the confidentiality arrangements between the European Commission, the
European Medicines Agency (EMA) and the FDA. For more information on these arrangements,
see United States.
EMA-FDA GCP initiative
GCP initiative: frequently asked questions and
answers
The initiative's main objectives are to:
conduct periodic information exchanges on
GCP-related information, to streamline sharing of GCP inspection planning
information, and to communicate on inspection outcomes effectively and in a
timely manner;
conduct collaborative GCP inspections by sharing
information, experience and inspection procedures, co-operating in the conduct
of inspections and sharing knowledge of best practice;
share information on the interpretation of GCP, by
keeping each other informed of GCP-related legislation, regulatory guidance and
related documents, and to identify and act together to benefit the clinical
research process.
The initiative began with a pilot phase that ran
between September 2009 and March 2011. During the pilot, the EMA and the FDA
exchanged more than 250 documents relating to 54 different medicines. They also
organised 13 joint inspections of clinical trials in conjunction with the GCP
inspectors of the EU Member States.
A report and question-and-answer document on the
outcomes of the pilot are available, which detail the success of the
information-sharing and collaboration on inspections relating to clinical trials:
Report
of the EMA-FDA pilot GCP initiativeQuestions
and answers on the EMA-FDA GCP initiative
The pilot has laid the foundation for a more
efficient use of limited resources, improved inspection coverage and better
understanding of each agency’s inspection procedures. It demonstrates how the
agencies can work together to improve human subject protection and better
ensure the integrity of data submitted as the basis for drug approvals.
Based on the positive experience, the EMA and the
FDA intend to continue with the initiative, incorporating lessons learned
during the pilot. The agencies are updating the terms of engagement and
procedures for participating authorities to reflect this.Applicants interested
in volunteering to take part in a collaborative inspection during the pilot
phase should contact the EMA or the FDA.
(ref:http://www.emea.europa.eu/ema/index.jsp?curl=pages/regulation/general/general_content_000072.jsp&mid=WC0b01ac05800268ad)
Overview
Recent developments
The Food and Drug Administration (FDA) is
responsible for deciding whether new human drugs and therapeutic biologics can
be marketed in the United States. Given the substantial investment of time and
resources required to bring a drug or biologic to market, clinical trial
sponsors closely scrutinize application reviews, seeking to understand the
factors that influence FDA’s decisions to approve or deny a New Drug
Application (NDA) or Biologics License Application (BLA). In particular,
sponsors’ perceptions of what inspectional findings bring data integrity into
question and lead to refusal to approve an application may influence how they
plan, conduct and oversee their trials.
Within the Center for Drug Evaluation and Research
(CDER), the Division of Scientific Investigations (DSI) in the Office of
Compliance has specific responsibility for overseeing the inspections that
verify the integrity of application data and determine whether clinical trials
supporting applications are conducted in compliance with current FDA
regulations and statutory requirements. To provide insight into how DSI
verifies whether data are of sufficient quality to support conclusions drawn in
an NDA or BLA, we completed an analysis of our application review documentation
from first quarter (1Q) fiscal year (FY) 2010 through 1Q FY 2011, covering the
most recent time period for which we had complete data. We also evaluated
documentation related to our decisions to classify clinical investigator inspections
as “official action indicated” (OAI) during the same time period, focusing on
factors that might lead to a recommendation to reject data in this subset of
inspections
DSI’s
Role in Drug Approval
Data supporting each NDA or BLA that CDER receives
undergoes multi-disciplinary review. During this review, DSI collaborates with
reviewers in CDER’s Office of New Drugs (OND) to identify inspections important
to determining that data supporting efficacy and safety conclusions are
accurate and complete. Ultimately, DSI reviews a subset of submitted original and
supplemental NDAs and BLAs. Over the past five years, DSI has overseen
approximately 480 on-site inspections of clinical investigators, clinical trial
sponsors and contract research organizations (CRO) annually. Of these,
approximately 70 percent were conducted as part of NDA or BLA review, with the
remainder arising from complaints concerning the various parties engaged in
FDA-regulated research.
Selection of sites for NDA and BLA-related
inspections is generally based upon a site’s importance to the data supporting
an application, such as the number of subjects enrolled or the site’s relative
contribution to study efficacy conclusions. Where an application relates to a
new molecular entity, FDA generally inspects the sponsor in addition to the
clinical investigator sites. More recently, CDER has been piloting a risk-based
clinical investigator site selection tool that permits quantitative evaluation
of an array of factors that may indicate a higher risk of data integrity
concerns at a site. FDA may also conduct sponsor and CRO inspections in
response to site-level inspectional findings that indicate possible deficits in
sponsor or CRO oversight.
Once sites are selected, DSI issues inspection
assignments to field investigators within FDA’s Office of Regulatory Affairs
who carry out the on-site inspections. On occasion, DSI reviewers may
participate in the inspection as clinical and/or scientific expert consultants.
However, a DSI reviewer’s primary role during NDA and BLA review is to
critically evaluate inspectional findings and their overall impact on site and
study data integrity and human subject protections.
When a field investigator observes conditions during
an inspection that he/she believes constitute a regulatory violation, the field
investigator will recommend an action, taking into consideration the nature of
the inspectional findings, any actions that occurred following the findings and
agency policy. DSI reviews the recommended action, determines whether is the
action is appropriate and supported by adequate evidence and makes a final
decision about the inspection classification. Depending on the nature of the
violations, FDA may ultimately find that corrective action may be left to the
establishment to take voluntarily (voluntary action indicated, or VAI).
Alternatively, FDA may determine that regulatory and/or administrative
sanctions are necessary to more immediately address significant objectionable
conditions or practices and classify the inspection as OAI. OAI actions may
range from issuance of a warning letter to pursuit of disqualification of a
clinical investigator.
In addition to reviewing the outcomes of individual
inspections to determine appropriate action for the inspected entity, DSI
reviewers also consider whether and how inspectional findings inform FDA’s
conclusions about the reliability of data in a marketing application. For this
analysis, reviewers generally focus on the data most critical to important
conclusions about a product’s safety and efficacy.1 A Clinical Inspection
Summary (CIS) memo documents DSI’s formal recommendations to the OND review
division at two levels: (1) whether DSI considers data from an individual
clinical investigator or another inspected party reliable, taking into account
the nature, extent, clinical significance and possible root causes of any
observed noncompliance and (2) whether DSI, after careful review of findings
across inspections, considers data from the clinical investigation as a whole
reliable in support of a proposed indication. The analysis below is intended to
shed light on this review process.
(ref: http://www.fdli.org/resources/resources-order-box-detail-view/current-trends-in-fda-inspections-assessing-clinical-trial-quality-an-analysis-of-cder-s-experience)
Current
issues in Clinical research
Clinical Trial Billing Issues
- Interpretation
of Medicare clinical trial policy
- Sponsors as
secondary payers to insurance
- Process
issues
- Medicare :
July
2007 National Coverage Decision (NCD)
- Qualifying
Trials: Trial must “qualify” for coverage:
Four requirements to
qualify:
The trial must be
“deemed” to have desirable characteristics (based on funding source)
The investigational
item or service must fall within a Medicare benefit category
The study must enroll
patients with diagnosed disease
The study must have
therapeutic intent
Regional Medicare contractor’s
Medical Director determines which trials/what factors constitute therapeutic
intent
Phase I trials probably
will not qualify
You need to speak with
your Medicare contractor’s Medical Director
Routine Costs: Medicare
pays for “routine costs”
Conventional Care/SOC
Items/services required
for provision of the investigational item or drug
e.g., administration of
chemotherapeutic agent
Items/services for the
detection and prevention of complications
What about standard of
care services for a trial that does not qualify?
Ask your Medical
Director if billable
- Sponsors as
Secondary Payers: Cannot bill Medicare if a sponsor
agrees to pay for a service if an insurer (including Medicare) first
denies the claim
- If you bill
insurance and claim denied, you must pursue payment from patient
- Exceptions
for indigent patients
- See CMS
SE0822 (released Sep 2008 and modified Jan 09)
Institutional
Stance/Policies: Must have an institutional policy Some
issues open to interpretation –institution needs to take a stance Research
personnel must be trained on the policy
Process Issues: The
complicated part!
Form a cross-functional
team:
revenue cycle
research teams
Sponsors programs
Billing system IT
experts
Executive support
necessary for success
Process Team: The
team will:
Examine current
processes and information flows
Develop processes to
ensure compliant billing
Implement processes
Monitor progress – are
the new processes working?
Don’t just disband and
assume everything will work swell!
Clinical Research Coordinators: Researchers and Study Financial
Administrators
Current Issues in Clinical Trials Contracting
(ref: www.ncura.edu)
Confidentiality
issues
Researchers in the biomedical as well as social and
behavioral sciences are expected
to be proactive in designing and performing research
to ensure that the dignity, welfare,
and privacy of individual research subjects are
protected and that information about an
individual remains confidential. This expectation is
expressed in the ethical codes of
conduct of professional societies. Protecting the
confidentiality of information collected
about individuals is also vital to fulfilling the
ethical responsibilities described in the
Belmont Report.
Research in the biomedical and social sciences
encompasses a broad array of topical
areas, designs, and degree of risk. Many studies
pose minimal risk to research
subjects. Some studies, however, are inaccurately
perceived as conveying minimal
risk. In such studies, disclosure of identifiable
data may present a significant risk to the
subject as a result of the sensitive nature of the
topic, the variety of social interactions,
or possible financial or legal implications of the
activity being studied. In such research,
especially in the social and behavioral sciences,
protecting the confidentiality of data
collected from or about private individuals is often
the key element in minimizing risk.
In addition to protecting research subjects from
harm that might result from their
participation in research, applying appropriate
confidentiality protections provides other
important benefits. Confidentiality protections
minimize subjects’ concerns over the use
(or misuse) of the data. Subjects consequently
provide more accurate information to
investigators, thereby improving the data used in
the analysis and thus the overall
quality of the research. Confidentiality protections
allow researchers to continue to
conduct difficult research on important societal
problems (e.g., drug abuse, the spread
of HIV, genetic predispositions, high risk sexual
behaviors, violence). Such research
provides a scientifically-informed basis for making
important public policy decisions and
fosters advances in medicine and in all fields of
science. The benefits of these results
accrue not only to the research subjects, but to
society at large.
Confidentiality issues need to be recognized and
considered at every stage of the
research process. These stages include the initial
study design; identification,
recruitment, and consent processes for the study
population; security, analysis, and
final disposition of data; and publication or
dissemination of data and results.
Intentional or inadvertent breaches of
confidentiality by investigators or their staff may
occur. In addition, there may be attempts (usually
in a legal context) to force or compel
disclosure of confidential information for
non-research purposes. The likelihood of such
an attempt cannot be anticipated by virtue of the
subject matter or setting of the
research. [An informative overview of this issue can
be found in Joe S. Cecil and
Gerald T. Wetherington, Special Editors,
Court-Ordered Disclosure of Academic
Research: A Clash of Values of Science and Law. 59
LAW AND CONTEMPORARY
PROBLEMS. Number 3, Summer 1996.] The purpose of
this paper is not to address all
dimensions of this issue, but to focus on those
aspects that are especially important in
protecting against breaches of confidentiality.
Reducing
Risk Through Confidentiality Protections
Confidentiality issues do not inhere in all human
subjects research. For example,
observation of behavior in public places where there
is no interaction between the
observer and the observed and where data are
recorded in anonymous form involves
no issue of confidentiality for subjects,
investigators or IRBs. In some studies, the
consent agreement establishes that research subjects
neither seek nor want
confidentiality (e.g., a political science study of
legislative changes where directors of
interest groups agree to participate knowing that
what they report will be presented as
part of the analysis of factors leading to change).
In circumstances where a promise of
confidentiality is not a part of an informed consent
agreement, the protocol makes clear
to IRBs the nature of the consent agreement and why
biographical anonymity and
confidentiality are not sought.
Issues of data confidentiality typically come into
play when biomedical, social or
behavioral science research involves data collection
on identifiable individuals.
Confidentiality protections should be developed
consistent with the study design and the
potential risk of harm from breaches of
confidentiality. As the risk of harm incurred by
disclosure increases, so should the level of
protection from such harm. In some cases,
the collected data may not require as high a level
of security as in other cases (e.g.,
laboratory studies on the level of boredom
associated with repetitive tasks does not
involve the same risk of data disclosure as surveys
of personal sexual orientation and
experience; clinical laboratory data generally do
not involve the same risk of disclosure
as data from genetic testing or screening). In all
cases where a promise of confidentiality is included in the
consent agreement, it must be granted and
secured—regardless of the level of risk.
Much of the risk in social and behavioral science
research is related to inadvertent or
unintended disclosure. An adequate data protection
plan can and should reduce the
risk of such occurrences. The OHRP has clarified
that the Common Rule allows
institutions and IRBs the flexibility to review and
approve appropriately designed
confidentiality protections.
Protocols should be designed to minimize the need to
collect identifiable data by
determining whether there is a legitimate reason to
collect or maintain identifiers. Data
can often be collected anonymously, or the
identifiers can be removed and destroyed
after various data have been merged. When it is
necessary to collect and maintain
identifiable data, a data protection plan should
describe the appropriate level of
confidentiality protections based on the potential
magnitude of the risk of harm from
disclosure. All members of the research team and
staff should receive appropriate
training about securing and maintaining
confidentiality and safeguarding data. Data
should be physically secure, and all identifiable,
confidential data not intended for
secure archiving should be destroyed.
Confidentiality
Protections
Efforts can and should be made to buffer or insulate
research data from encroachment.
When a determination is reached that the sensitive
nature of the data and the potential
risk of harm to individual subjects occasion legally
supported confidentiality protections,
the investigator (with the support of the
institution) should pursue appropriate
protections.
One such mechanism involves securing a certificate
of confidentiality from the
Department of Health and Human Services for
applicable categories of research
(biomedical, behavioral, clinical, mental health,
drug or alcohol abuse)..
Another involves
investigator and institutional compliance with mandatory confidentiality
protections such as those provided through statutes
covering the DOJ and DOEd.. It is
important to note that each of these confidentiality
provisions has important limitations.
It may apply only to certain categories of research
or to research sponsored by a
specific agency. It may protect the identity of the
research subject, but not the data.
Or, it may provide protection against compelled
disclosure of data, but not voluntary
disclosure . OHRP should lead efforts to strengthen
the current system of confidentiality
protections.
Given the limits of these statutory protections,
both investigators (and their research
teams and staff) and their institutions are morally
obligated to resist attempts to breach
confidentiality through compelled or forced
disclosures (e.g., subpoenas). This not only
fulfills ethical obligations to the research
subject, but also serves to prevent important
breaches of confidentiality. It is important to note
that courts may subpoena either data
or investigators who have had conversations with
participants.
(Ref: http://www.hhs.gov/ohrp/archive/nhrpac/documents/nhrpac14.pdf)
Medicines
for human use ( clinical trials ) regulations 2004
PART 1 INTRODUCTORY PROVISIONS
1.Citation and commencement
2.Interpretation
3.Sponsor of a clinical trial
4.Responsibility for functions under the Directive
Collapse -PART 2 ETHICS COMMITTEES
5.United Kingdom Ethics Committees Authority
6.Establishment of ethics committees
7.Recognition of ethics committees
8.Revocation of recognition
9.Constitution and operation of ethics committees
10.Other functions of the Authority
Collapse -PART 3 AUTHORISATION FOR CLINICAL TRIALS
AND ETHICS COMMITTEE OPINION
11.Interpretation of Part 3
12.Requirement for authorisation and ethics
committee opinion
13.Supply of investigational medicinal products for
the purpose of clinical trials
14.Application for ethics committee opinion
15.Ethics committee opinion
16.Review and appeal relating to ethics committee
opinion
17.Request for authorisation to conduct a clinical
trial
18.Authorisation procedure for clinical trials
involving general medicinal products
19.Authorisation procedure for clinical trials
involving medicinal products for gene therapy etc.
20.Authorisation procedure for clinical trials
involving medicinal products with special characteristics
21.Clinical trials conducted in third countries
22.Amendments to clinical trial authorisation
23.Amendments by the licensing authority
24.Amendments by the sponsor
25.Modifying or adapting rejected proposals for
amendment
26.Reference to the appropriate committee or the
Medicines Commission
27.Conclusion of clinical trial
Collapse -PART 4 GOOD CLINICAL PRACTICE AND THE
CONDUCT OF CLINICAL TRIALS
28.Good clinical practice and protection of clinical
trial subjects
29.Conduct of trial in accordance with clinical trial
authorisation etc.
30.Urgent safety measures
31.Suspension or termination of clinical trial
Collapse -PART 5 PHARMACOVIGILANCE
32.Notification of adverse events
33.Notification of suspected unexpected serious
adverse reactions
34.Clinical trials conducted in third countries
35.Annual list of suspected serious adverse
reactions and safety report
Collapse -PART 6 MANUFACTURE AND IMPORTATION OF
INVESTIGATIONAL MEDICINAL PRODUCTS
36.Requirement for authorisation to manufacture or
import investigational medicinal products
37.Exemption for hospitals and health centres
38.Application for manufacturing authorisation
39.Consideration of application for manufacturing
authorisation
40.Grant or refusal of manufacturing authorisation
41.Application and effect of manufacturing
authorisation
42.Obligations of manufacturing authorisation holder
43.Qualified persons
44.Variation of manufacturing authorisation
45.Suspension and revocation of manufacturing
authorisation
Collapse -PART 7 LABELLING OF INVESTIGATIONAL
MEDICINAL PRODUCTS
46.Labelling
Collapse -PART 8 ENFORCEMENT AND RELATED PROVISIONS
47.Application of enforcement provisions of the Act
48.Infringement notices
49.Offences
50.False or misleading information
51.Defence of due diligence
52.Penalties
Collapse -PART 9 MISCELLANEOUS PROVISIONS
53.Construction of references to specified
publications
54.Consequential and other amendments to enactments
55.Revocations
56.Transitional provisions
Signature
SCHEDULES
1.Conditions and principles of good clinical
practice and the protection of clinical trial subjects
2.Additional provisions relating to ethics
committees
3.Particulars and documents that must accompany an
application for an ethics committee opinion, a request for authorisation, a
notice of amendment and a notification of the conclusion of a trial
4.Appeal against unfavourable ethics committee
opinion
5.Procedural provisions relating to the refusal or
amendment of, or imposition of conditions relating to, clinical trial
authorisations and the suspension or termination of clinical trials
6.Particulars that must accompany an application for
a manufacturing authorisation
7.Standard provisions for manufacturing
authorisations
8.Procedural provisions relating to proposals to
grant, refuse to grant, vary, suspend or revoke manufacturing authorisations
9.Modification of the enforcement provisions of the
Act subject to which those provisions are applied for the purposes of these
Regulations
10.Consequential and other amendments of enactments
11.Revocations
12.Transitional provisions
Other
relevant issues
Challenges in Clinical Research
Cooperation among a diverse group of
stakeholders—including research sponsors (industry, academia, government,
nonprofit organizations, and patient advocates), clinical investigators,
patients, payers, physicians, and regulators—is necessary in conducting a
clinical trial today. Each stakeholder offers a different set of tools to
support the essential components of a clinical trial. These resources form the
infrastructure that currently supports clinical research in the United States.
Time, money, personnel, materials (e.g., medical supplies), support systems
(informatics as well as manpower), and a clear plan for completing the
necessary steps in a trial are all part of the clinical research
infrastructure. A number of workshop participants lamented that most clinical
trials are conducted in a “one-off” manner.The following popper user interface
control may not be accessible. Tab to the next button to revert the control to
an accessible version.Destroy user interface control1 Significant time, energy,
and money are spent on bringing the disparate resources for each trial
together. Some workshop attendees suggested that efficiencies could be gained
by streamlining the clinical trials infrastructure so that those investigating
new research questions could quickly draw on resources already in place instead
of reinventing the wheel for each trial.
Challenges
Facing Investigators in Academic Health Centers
Woodcock discussed a number of important obstacles
facing investigators conducting research using the current infrastructure.
Clinical investigators, those who lead a research idea through the clinical
trial process, face multiple small obstacles that together can appear
insurmountable. These obstacles include locating funding, responding to multiple
review cycles, obtaining Institutional Review Board (IRB) approvals,
establishing clinical trial and material transfer agreements with sponsors and
medical centers, recruiting patients, administering complicated informed
consent agreements, securing protected research time from medical school
departments, and completing large amounts of associated paperwork. As a result
of these challenges, many who try their hand at clinical investigation drop out
after their first trial. Especially in the case of investigator-initiated
trials, where an individual’s idea and desire to explore a research question
are the primary force behind the trial, the complex task of seeing a clinical
trial through from beginning to end is making the clinical research career path
unattractive for many young scientists and clinicians. Woodcock noted that in
her experience, successful clinical investigators represent a select subset of
clinicians—highly tenacious and persistent individuals with exceptional
motivation to complete the clinical trial process.
According to Robert Califf, Vice Chancellor for
Clinical Research and Director of the Duke Translational Medicine Institute,
some of the challenges to participating in clinical research mentioned by
clinical cardiovascular investigators include:
- the time and
financial demands of clinical practice;
- the overall
shortage of cardiovascular specialists;
- the
increasing complexity of regulations;
- the
increasing complexity of contracts;
- the lack of
local supportive infrastructure;
- inadequate
research training;
- less
enjoyment from participation (e.g., increasing business aspects, contract
research organization pressures); and
- data
collection challenges (medical records, reimbursement, quality control,
pay for performance).
Challenges
Confronting Community Physicians
Practitioners face a number of challenges to their
involvement in clinical research. Busy patient practices and the associated
billing and reporting requirements leave them with limited time for research. A
further barrier is the lack of a supportive clinical research infrastructure,
especially in the form of administrative and financial support. For
practitioners who become engaged in running a clinical trial and recruiting
patients, their financial reimbursement per patient can, in some cases, be less
than they would receive from regular practice. In addition, there is a
financial disincentive for physicians to refer their patients to clinical trials.
Physicians who do so must often refer those patients away from their care; thus
each patient referred represents a lost revenue stream.
Challenges
Facing Patients
Patients also face challenges to participating in
clinical research. Many workshop participants noted that patients often are
unaware of the possibility of enrolling in a clinical trial. If they are aware
of this opportunity, it is often difficult for them to locate a trial. Patients
may reside far from study centers; even the largest multicenter trials can pose
geographic challenges for those wishing to participate. Moreover, depending on
the number of clinic visits required by the study protocol, significant travel
and time costs may be associated with participation. In addition, trials designed
with narrow eligibility criteria for participation purposely eliminate many
patients who might have the disease being studied but are ineligible because of
other characteristics (e.g., age, level of disease progression, exposure to
certain medicines).
Informed
Consent
Informed consent refers to the process and documents
associated with educating individuals on the details of a clinical trial and
potentially gaining their consent to participate in the study.The following
popper user interface control may not be accessible. Tab to the next button to
revert the control to an accessible version.Destroy user interface control5
Obtaining informed consent from each subject in a clinical trial requires a
significant amount of time. The informed consent process includes developing
appropriately worded consent documents, discussing the documents and the
clinical trial process with individual patients, obtaining the required patient
signatures on the documents, and keeping track of the paperwork generated
throughout the enrollment process.
Patient
Education
Mayer presented the results of a Harris Interactive
Survey of 6,000 cancer patients that found that 85 percent were unaware that
participation in clinical trials was even an option. Of the patients surveyed,
75 percent said that if participation in a clinical trial had been offered,
they would have been receptive to the idea. Of those aware of clinical trials
and offered the possibility of participation, 71 percent chose not to
participate. However, almost all who participated were satisfied with the
experience. Thus, according to these survey results, patients’ preconceived
notions about trial participation could pose a barrier to clinical trial
enrollment.
Patient
Recruitment
According to Woodcock, sites for clinical trials are
frequently selected on the basis of where the investigators are located, as
opposed to where the patients are, creating difficulties in patient
recruitment. When patient recruitment is impeded, the trial is delayed,
sometimes by years, until the number of patients required by the study protocol
can be enrolled. Once a trial protocol has been activated, the recruitment of
patients requires a significant amount of time and money. Canetta reported that
the ability to recruit patients into a trial successfully is similar for the
pharmaceutical industry and NCI. Regardless of the trial sponsor, recruitment
of patients who meet the requirements of the protocol is difficult: in one
study of 14 cancer centers approximately 50 percent of study sites failed to
recruit a single patient (Durivage et al., 2009). Thus, patient enrollment can
directly affect the number of trials that are completed.
(ref: http://www.ncbi.nlm.nih.gov/books/NBK50888/)