UNIT IV
History and purpose of GCP development of ICH GCP
History
of GCP
Early 1960s : Widespread concern
about the safety and control of investigational drugs and the clinical research
process developed among member of the medical profession, the scientific
community, regulatory authorities,
and general public.
WHO WHO’s s “Guidelines for Good
Clinical Practice (GCP) for trials on pharmaceutical products products”1995.
• The GCP guideline is Topic E6 ICH,
1997
• International Standard Organization
(ISO), “Clinical investigation of medical devices for human subjects, Part I
(General requirements) and Part 2 (Clinical investigation plans) (2001)
• Pan American Health Organization
(PAHO). Pan American Network on Drug Regulatory Harmonization (PANDRH). “GCP:
Document of the Americas Americas” (2005)
History
of ICH--GCP
•Need for safe and efficient
products for various health problemshealth problems
•Drug development is expensive and
time consumingDrug consuming
•Need for efficient quality systemsNeed
systems••Global drug marketGlobal market
•Existence of national laws and
regulations for drug developmentdevelopment.
Goal
• To provide a unified standard to
facilitate the mutual acceptance of clinical data by the
regulatory authorities
• Remove redundancy /duplication in development
and review process.
Process
• Developed guidelines applicable
for
• Drugs
• Biologics
• Medical devices
• Approved by ICH members
• Adopted by National Regulatory
Authorities
Good Clinical Practice (GCP)
An international ethical and
scientific quality standard for designing, conducting, recording, and
reporting trials that involve the
participation of human subjects Public assurance that the rights, safety, and
well well- being of trial subjects are protected
• Consistent with the Declaration of
Helsinki
• Results in credible data
Who are responsible for GCP
The responsibility for GCP is shared
by all of the parties involved, including including:
– sponsors
– investigators and site staff
– contract research organizations (
CROs CROs)
– ethics committees
– regulatory authorities
– research subjects.
WHO Principles of GCP
Principle 1 : Ethical Conduct
Research involving humans should be scientifically
sound and conducted in accordance with basic ethical principles, which have
their origin in the Declaration of Helsinki. Three basic ethical principles of equal
importance, namely respect for persons, beneficence, and justice justice, permeate
all other GCP principles.
Principle 2 Research Described in a
Protocol Research involving humans should be scientifically justified and
described in a clear, detailed protocol.
Principle 3: Risk Identification :
Before research involving humans is
initiated,
foreseeable risks and discomforts
and any
anticipated benefit (s) for the
individual
research subject and society should
be
identified. Research of
investigational products
or procedures should be supported by
adequate non non-clinical and, when
applicable,
clinical information.
Principle 4: Benefit Benefit-Risk
Assessment
Research involving humans should be
initiated only if the anticipated benefit(s s) for the ) individual research
subject and society clearly outweigh the risks. Although the benefit of the results
of the trial to science and society should be taken into account, the most
important considerations are those related to the rights, safety, and well
well-being of the research subjects.
Principle 5 : Review by IEC/IRB
Research involving humans should
receive independent ethics committee/institutional review board (IEC/IRB)
approval/ favourable opinion prior to initiation
Principle 6 : Protocol Compliance
Research involving humans should be conducted
in compliance with the approved
protocol.
Principle 7 : Informed Consent
Freely given informed consent should
be obtained from every subject prior to research participation in accordance
with national culture (s) and requirements. When a subject is not capable of
giving informed consent, the permission of a legally authorized representative
should be obtained in accordance with applicable law.
Principle 8 : Continuing
Review/Ongoing
Benefit Benefit-Risk Assessment Research
involving humans should be continued only if the benefit benefit-risk profile remains
favorable.
Principle 9 : Investigator
Qualifications
Qualified and duly licensed medical
personnel (i.e. physician or, when appropriate, dentist) should be responsible
for the medical care of research subjects, and for any medical decision (s)
made on their behalf.
Principle
10: Staff Qualifications
Each
individual involved in conducting a trial should be qualified by education,
training,
and
experience to perform his or her respective task (s) and currently licensed to
do so,
where required.
Principle
11 : Records
All
clinical trial information should be recorded, handled, and stored in a way
that
allows its
accurate reporting, interpretation, and verification.
Principle 12 :
Confidentiality/Privacy
The confidentiality of records that
could identify subjects should be protected, respecting the privacy and
confidentiality rules in accordance with the applicable regulatory requirement
(s)
Principle 13 : Good Manufacturing
Practice
Investigational products should be manufactured,
handled, and stored in accordance with applicable Good Manufacturing Practice
(GMP) and should be used in accordance with the approved protocol.
Principle 14: Quality Systems
Systems with procedures that assure
the quality of every aspect of the trial should be implemented.
GCP Design Standards
• Written protocol
• Investigator brochure
• Scientific soundness
• Feasibility
• Adequate resources
• Randomization / blinding
GCP Conduct Standards
• IRB & Regulatory approval
• Compliance with protocol
• Informed consent
• Confidentiality of data
• Medical management of adverse
events
• Product accountability
• Qualification & training
GCP Recording Standards
• CRF completion
• Data handling
• Security maintenance
• Audit requirements
• Product accountability
• Management of study
files/essential
Documents
GCP Reporting Standards
• Adverse events
• Interim reviews
• Progress reports
• Final reports
• Monitoring / audit reports
To
••SponsorsSponsors••IRB/IECIRB/IEC••Regulatory authoritiesRegulatory
authorities
• Other investigators
(ref : http://www.jirb.org.tw/DB/File/Download/970127
03_History%20and %20 Principles% 20of%20 Good%20Clinical%20PracticeTaipei_Christine%20Maure.pdf)
Roles
and responsibilities in clinical research according to ICH GCP
Roles
and Responsibilities of Clinical Research Coordinator (CRC)
Categories: Clinical Research,
Ethics in Clinical Research | Tags: CRC job profile, Role of Clinical
Research Coordinator (CRC), Role of CRC, Roles and Responsibilities of a CRC
A clinical research coordinator
(CRC) is responsible for conducting clinical trials at clinical trial sites
according to the protocol, ICH-GCP and other regulatory requirements. The role
of a study coordinator in clinical trials is very important. Every clinical trial
site may have one or more study coordinators depending on the workload at the
trial site. Clinical trials at site level can be roughly divided into 3 stages.
Let us discuss about the role of the study coordinator in these three stages.
The three stages are:
1) Before the start of the clinical
trial
2) During the conduct of the
clinical trial
3) After close out of the clinical
trial
1) Before the start of the Clinical
Trial:
During this stage the study
coordinator has to collect and complete feasibility questionnaires received
form different CRO’s and Sponsor’s. CRC’s also have to collect required
information from the principal investigator, and send it back to the respective
people who contacted the site regarding the study. Sponsors select sites based on
the responses filled in the feasibility questionnaire and conduct Pre-site
selection visits to confirm the sites participating in the study.
The sponsors or CRO’s after
selecting clinical trial sites conduct Investigator meetings, which have to be
attended by the study coordinator along with the principal or co-investigator.
Investigator meetings are conducted either at national level or international
level. Before start of the trial CRC’s are usually busy with submitting all
study documents to the ethics committee.
Documents to be submitted to the
ethics committee normally include the study protocol, investigator brochure and
informed consent forms (vernacular languages at site) with translation
certificates. If any amendments are available those also need to be submitted
to the ethics committee. In addition to these documents, investigator’s CV with
MRC, FDA-1572, Financial disclosure forms, Insurance statement, subject diaries
if applicable, clinical trial agreement, indemnification letter, protocol signature
page and different study logs, eCRF or CRF entry guidelines, blank CRF’s etc
also need to be submitted.
In all these processes a clinical
study coordinator plays a vital role. After obtaining approval from the ethics
committee the clinical trial can be initiated at the site level.
2) During the conduct of clinical
trial:
By the time a clinical trial is
initiated at a site the CRC must have a good understanding of the study
protocol and must know well about inclusion and exclusion criteria. During the
screening time, the study coordinator has to obtain informed consent from the
subject in if delegated to do so by the Principal Investigator. Study
coordinators have to collect subjects previous medical records and according to
study protocol he/she has to conduct subjects scheduled visits. Before
randomization visit of the subject the CRC must check inclusion and exclusion
criteria thoroughly, and only then eligible subjects must be enrolled into the
study.
After completion of all visit
procedures coordinators have to enter data in case report forms (CRF). CRF’s
are two types, one is paper CRF and another one is eCRF. eCRF’s are usually of
5 types namely , Phase forward – Inform, oracle clinical remote data capture,
medi data rave, novartis EDC (or) Pasco and Data track. All the visit details
like demographics, vital signs, subject past medical history, any SAE or AE
details have to be entered in these online data collection tools based on
source documentation available. During monitoring, sponsor representatives
(CRA’s) cross verify both the source documents & the data filled by the CRC
in the case report forms. During the monitoring visit the CRA’s also verify EC
notifications and Investigator Site Files (ISF). To keep all these documents up
to date is the responsibility of the clinical study coordinator. Study
coordinator also has to maintain site SOP’s, EC sops and EC members list.
Whenever study subjects come for
next schedule visit, study drug accountability has to be calculated by the CRC.
Along with that subject diaries have to be reviewed if applicable. IVRS
(Interactive voice response system) and IWRS (Interactive web response system)
to record the subject visit have to be performed as per the study requirement.
IVRS reports have to be maintained in the ISF’s. IP (Investigational product)
is the major part in clinical trial and study coordinators have to store the
same in proper condition and maintain required temperature logs.
In case of any adverse events (AE’s)
or serious adverse events (SAE’s) that occurred at the trial site, coordinators
have to collect all applicable details such as start date, stop date, severity
(Mild, Moderate or Severe), concomitant medications consumed by the subject and
the route of administration. Also information related to any dechallenge and
rechallenge, therapy provided to the subject during the SAE, recovery details
and whether it is related to study drug or not have to be collected. Study
Coordinators have to notify SAE’s to the Ethics committee within 7 working days
and details of SAE have to sent to the sponsor within 24 hours.
Throughout the clinical trial the
study coordinators have to check all Central lab reports as well as local lab
reports and take PI signatures on them to document that the PI reviewed the lab
reports. CRC is also responsible for telephonic reminders to the subjects regarding
the visits. If the CRC is involved in data entry then it is the study
coordinators responsibility to resolve all queries within 48 hours or as per
timeline specified by the sponsor. Queries are of three types i.e. 1) system
generated queries 2) data management generated queries and 3) CRA generated
queries.
3) After Close out of clinical
trial:
Before a clinical trial is closed at
the site, study coordinators have to check all documents and all the documents
have to be updated. On the day of close out CRA will verify all documents.
After verification of all documents by the CRA, CRC will assist in archiving
the documents at site. Site has to maintain all study related records for 15 to
20 years.
As you can see a CRC plays a vital
role in managing a clinical trial at the site level and acts as a link between
the sponsor, EC and the Investigator Site.
Sponser
5.
SPONSOR
5.1
Quality Assurance and Quality Control
5.1.1 The sponsor is responsible for implementing and
maintaining quality assurance and
quality control systems with written SOPs to ensure that trials are conducted and data are generated,
documented (recorded), and reported in compliance
with the protocol, GCP, and the applicable regulatory requirement(s).
5.1.2 The sponsor is responsible for securing
agreement from all involved parties to ensure
direct access (see 1.21) to all trial related sites, source data/documents
, and reports for the purpose of
monitoring and auditing by the sponsor, and
inspection by domestic and foreign regulatory authorities.
5.1.3 Quality control should be applied to each
stage of data handling to ensure that all
data are reliable and have been processed correctly.
5.1.4 Agreements, made by the sponsor with the investigator/institution
and any other parties involved with the
clinical trial, should be in writing, as part of the protocol or in a separate agreement.
5.2 Contract Research Organization (CRO)
5.2.1 A sponsor may transfer any or all of the sponsor's
trial-related duties and functions to a
CRO, but the ultimate responsibility for the quality and integrity of the trial data always resides with
the sponsor. The CRO should implement
quality assurance and quality control.
5.2.2 Any trial-related duty and function that is transferred
to and assumed by a CRO should be
specified in writing.
5.2.3 Any trial-related duties and functions not specifically
transferred to and assumed by a CRO are
retained by the sponsor.
5.2.4 All references to a sponsor in this guideline
also apply to a CRO to the extent that a
CRO has assumed the trial related duties and functions of a sponsor.
5.3 Medical Expertise
The sponsor should designate appropriately qualified
medical personnel who will be readily
available to advise on trial related medical questions or problems. If necessary, outside consultant(s) may be
appointed for this purpose.
5.4 Trial Design
5.4.1 The sponsor should utilize qualified
individuals (e.g. biostatisticians, clinical
pharmacologists, and physicians) as appropriate, throughout all stages
of the trial process, from designing the
protocol and CRFs and planning the analyses to analyzing and preparing interim
and final clinical trial reports.
5.4.2 For further guidance: Clinical Trial Protocol and
Protocol Amendment(s) (see 6.), the ICH Guideline for Structure and Content of
Clinical Study Reports, and other
appropriate ICH guidance on trial design, protocol and conduct.
5.5 Trial Management, Data Handling, and Record
Keeping
5.5.1 The sponsor should utilize appropriately
qualified individuals to supervise the overall
conduct of the trial, to handle the data, to verify the data, to conduct the statistical analyses, and to prepare the
trial reports.
5.5.2 The sponsor may consider establishing an
independent data-monitoring committee
(IDMC) to assess the progress of a clinical trial, including the safety data and the critical efficacy endpoints at
intervals, and to recommend to the sponsor
whether to continue, modify, or stop a trial. The IDMC should have written operating procedures and maintain
written records of all its meetings.
5.5.3 When using electronic trial data handling
and/or remote electronic trial data systems,
the sponsor should:
(a) Ensure and document that the electronic data
processing system(s) conforms to the
sponsor’s established requirements for completeness, accuracy, reliability, and consistent
intended performance (i.e. validation).
(b) Maintains SOPs for using these systems.
(c) Ensure that the systems are designed to permit
data changes in such a way that the data
changes are documented and that there is no deletion of entered data (i.e. maintain an audit trail,
data trail, edit trail).
(d) Maintain a security system that prevents
unauthorized access to the data.
(e) Maintain a list of the individuals who are
authorized to make data changes
(see 4.1.5 and 4.9.3).
(f) Maintain adequate backup of the data.
(g) Safeguard the blinding, if any (e.g. maintain
the blinding during data entry and
processing).
5.5.4 If data are transformed during processing, it
should always be possible to
compare the original data and observations with the
processed data.
5.5.5 The sponsor should use an unambiguous subject identification
code (see 1.58) that allows
identification of all the data reported for each subject.
5.5.6 The sponsor, or other owners of the data,
should retain all of the sponsorspecific essential documents pertaining to the
trial (see 8. Essential Documents for the Conduct of a Clinical Trial).
5.5.7 The sponsor should retain all sponsor-specific
essential documents in conformance with
the applicable regulatory requirement(s) of the country(ies) where the product is approved, and/or where
the sponsor intends to apply for approval(s).
5.5.8 If the sponsor discontinues the clinical
development of an investigational product
(i.e. for any or all indications, routes of administration, or dosage forms), the sponsor should maintain all
sponsor-specific essential documents for
at least 2 years after formal discontinuation or in conformance with the applicable regulatory requirement(s).
5.5.9 If the sponsor discontinues the clinical
development of an investigational product,
the sponsor should notify all the trial investigators/institutions and all the regulatory authorities.
5.5.10 Any transfer of ownership of the data should be
reported to the appropriate authority(ies),
as required by the applicable regulatory requirement(s).
5.5.11 The sponsor specific essential documents
should be retained until at least 2 years
after the last approval of a marketing application in an ICH region and until there are no pending or contemplated
marketing applications in an ICH region
or at least 2 years have elapsed since the formal discontinuation of clinical development of the investigational product.
These documents should be retained for a
longer period however if required by the applicable regulatory requirement(s) or if needed by the sponsor.
5.5.12 The sponsor should inform the
investigator(s)/institution(s) in writing of the need for record retention and should notify
the investigator(s)/institution(s) in writing
when the trial related records are no longer needed.
5.6 Investigator Selection
5.6.1 The sponsor is responsible for selecting the
investigator(s)/institution(s). Each investigator
should be qualified by training and experience and should have adequate resources (see 4.1, 4.2) to properly
conduct the trial for which the investigator
is selected. If organization of a coordinating committee and/or selection of coordinating investigator(s) are
to be utilized in multicentre trials, their
organization and/or selection are the sponsor's responsibility.
5.6.2 Before entering an agreement with an
investigator/institution to conduct a trial,
the sponsor should provide the investigator(s)/institution(s) with the protocol and an up-to-date Investigator's
Brochure, and should provide sufficient
time for the investigator/institution to review the protocol and the information provided.
5.6.3 The sponsor should obtain the
investigator's/institution's agreement:
(a) to conduct the trial in compliance with GCP,
with the applicable regulatory requirement(s)
(see 4.1.3), and with the protocol agreed to by the sponsor and given approval/favourable opinion by the
IRB/IEC (see 4.5.1);
(b) to comply with procedures for data
recording/reporting;
(c) to permit monitoring, auditing and inspection
(see 4.1.4) and
(d) to retain the trial related essential documents
until the sponsor informs the investigator/institution
these documents are no longer needed (see
4.9.4 and 5.5.12).
The sponsor and the investigator/institution should
sign the protocol, or an alternative
document, to confirm this agreement.
5.7 Allocation of Responsibilities
Prior to initiating a trial, the sponsor should
define, establish, and allocate all trialrelated duties and functions.
5.8 Compensation to Subjects and Investigators
5.8.1 If required by the applicable regulatory requirement(s),
the sponsor should provide insurance or
should indemnify (legal and financial coverage) the investigator/the institution against claims
arising from the trial, except for claims
that arise from malpractice and/or negligence.
5.8.2 The sponsor's policies and procedures should
address the costs of treatment of trial
subjects in the event of trial-related injuries in accordance with the applicable regulatory requirement(s).
5.8.3 When trial subjects receive compensation, the
method and manner of compensation should
comply with applicable regulatory requirement(s).
5.9 Financing
The financial aspects of the trial should be
documented in an agreement between the sponsor
and the investigator/institution.
5.10 Notification/Submission to Regulatory
Authority(ies)
Before initiating the clinical trial(s), the sponsor
(or the sponsor and the investigator, if
required by the applicable regulatory requirement(s)) should submit any required application(s) to the appropriate
authority(ies) for review, acceptance, and/or
permission (as required by the applicable regulatory requirement(s)) to
begin the trial(s). Any
notification/submission should be dated and contain sufficient information to identify the protocol.
5.11 Confirmation of Review by IRB/IEC
5.11.1 The sponsor should obtain from the
investigator/institution:
(a) The name and address of the investigator's/institution’s
IRB/IEC.
(b) A statement obtained from the IRB/IEC that it is
organized and operates according to GCP
and the applicable laws and regulations.
(c) Documented IRB/IEC approval/favourable opinion
and, if requested by the sponsor, a
current copy of protocol, written informed consent form(s) and any other written information to be provided
to subjects, subject recruiting procedures,
and documents related to payments and compensation available to the subjects, and any other
documents that the IRB/IEC may
have requested.
5.11.2 If the IRB/IEC conditions its approval/favourable
opinion upon change(s) in any aspect of
the trial, such as modification(s) of the protocol, written informed consent form and any other written
information to be provided to subjects, and/or
other procedures, the sponsor should obtain from the investigator/institution a copy of the modification(s)
made and the date approval/favourable
opinion was given by the IRB/IEC.
5.11.3 The sponsor should obtain from the investigator/institution
documentation and dates of any IRB/IEC
reapprovals/re-evaluations with favourable opinion, and of any withdrawals or suspensions of
approval/favourable opinion.
5.12 Information on Investigational Product(s)
5.12.1 When planning trials, the sponsor should ensure
that sufficient safety and efficacy data
from nonclinical studies and/or clinical trials are available to support human exposure by the route, at the
dosages, for the duration, and in the
trial population to be studied.
5.12.2 The sponsor should update the Investigator's
Brochure as significant new information
becomes available (see 7. Investigator's Brochure).
5.13 Manufacturing, Packaging, Labelling, and Coding
Investigational Product(s)
5.13.1 The sponsor should ensure that the
investigational product(s) (including active comparator(s) and placebo, if applicable) is
characterized as appropriate to the stage
of development of the product(s), is manufactured in accordance with any applicable GMP, and is coded and labelled in
a manner that protects the blinding, if
applicable. In addition, the labelling should comply with applicable regulatory requirement(s).
5.13.2 The sponsor should determine, for the
investigational product(s), acceptable storage
temperatures, storage conditions (e.g. protection from light), storage times, reconstitution fluids and procedures,
and devices for product infusion, if any.
The sponsor should inform all involved parties (e.g. monitors, investigators, pharmacists, storage managers)
of these determinations.
5.13.3 The investigational product(s) should be packaged
to prevent contamination and
unacceptable deterioration during transport and storage.
5.13.4 In blinded trials, the coding system for the
investigational product(s) should include
a mechanism that permits rapid identification of the product(s) in case of a medical emergency, but does not permit
undetectable breaks of the blinding.
5.13.5 If significant formulation changes are made in
the investigational or comparator
product(s) during the course of clinical development, the results of any additional studies of the formulated
product(s) (e.g. stability, dissolution rate,
bioavailability) needed to assess whether these changes would significantly alter the pharmacokinetic profile
of the product should be available prior
to the use of the new formulation in clinical trials.
5.14 Supplying and Handling Investigational
Product(s)
5.14.1 The sponsor is responsible for supplying the
investigator(s)/institution(s) with the
investigational product(s).
5.14.2 The sponsor should not supply an
investigator/institution with the investigational
product(s) until the sponsor obtains all required documentation (e.g. approval/favourable opinion from
IRB/IEC and regulatory authority(ies)).
5.14.3 The sponsor should ensure that written procedures
include instructions that the
investigator/institution should follow for the handling and storage of investigational product(s) for the trial and
documentation thereof. The procedures
should address adequate and safe receipt, handling, storage, dispensing, retrieval of unused product from subjects,
and return of unused investigational
product(s) to the sponsor (or alternative disposition if authorized by the sponsor and in compliance with
the applicable regulatory requirement(s)).
5.14.4 The sponsor should:
(a) Ensure timely delivery of investigational
product(s) to the investigator(s).
(b) Maintain records that document shipment, receipt,
disposition, return, and destruction of
the investigational product(s) (see 8. Essential Documents for the Conduct of a Clinical Trial).
(c) Maintain a system for retrieving investigational
products and documenting this retrieval
(e.g. for deficient product recall, reclaim after trial completion, expired product reclaim).
(d) Maintain a system for the disposition of unused
investigational product(s) and for the
documentation of this disposition.
5.14.5 The sponsor should:
(a) Take steps to ensure that the investigational product(s)
are stable over the period of use.
(b) Maintain sufficient quantities of the
investigational product(s) used in the trials
to reconfirm specifications, should this become necessary, and maintain records of batch sample analyses and
characteristics. To the extent stability
permits, samples should be retained either until the analyses of the trial data are complete or as
required by the applicable regulatory
requirement(s), whichever represents the longer retention period.
5.15 Record Access
5.15.1 The sponsor should ensure that it is
specified in the protocol or other written agreement that the
investigator(s)/institution(s) provide direct access to source data/documents for trial-related monitoring,
audits, IRB/IEC review, and regulatory
inspection.
5.15.2 The sponsor should verify that each subject
has consented, in writing, to direct access
to his/her original medical records for trial-related monitoring, audit, IRB/IEC review, and regulatory inspection.
5.16 Safety Information
5.16.1 The sponsor is responsible for the ongoing
safety evaluation of the investigational
product(s).
5.16.2 The sponsor should promptly notify all
concerned investigator(s)/institution(s) and the regulatory authority(ies) of findings
that could affect adversely the safety of subjects, impact the conduct of the
trial, or alter the IRB/IEC's approval/favourable
opinion to continue the trial.
5.17 Adverse Drug Reaction Reporting
5.17.1 The sponsor should expedite the reporting to
all concerned investigator(s)/institutions(s),
to the IRB(s)/IEC(s), where required, and to the regulatory authority(ies) of all adverse drug
reactions (ADRs) that are both serious and
unexpected.
5.17.2 Such expedited reports should comply with the
applicable regulatory requirement(s) and
with the ICH Guideline for Clinical Safety Data
Management: Definitions and Standards for Expedited Reporting.
5.17.3 The sponsor should submit to the regulatory
authority(ies) all safety updates and
periodic reports, as required by applicable regulatory requirement(s).
5.18
Monitoring
5.18.1 Purpose
The purposes
of trial monitoring are to verify that:
(a) The rights and well-being of human subjects are
protected.
(b) The reported trial data are accurate, complete,
and verifiable from source documents.
(c) The conduct of the trial is in compliance with
the currently approved protocol/amendment(s),
with GCP, and with the applicable regulatory
requirement(s).
5.18.2 Selection and Qualifications of Monitors
(a) Monitors should be appointed by the sponsor.
(b) Monitors should be appropriately trained, and
should have the scientific and/or
clinical knowledge needed to monitor the trial adequately. A monitor’s qualifications should be
documented.
(c) Monitors should be thoroughly familiar with the
investigational product(s), the
protocol, written informed consent form and any other written information to be provided to subjects, the
sponsor’s SOPs, GCP, and the applicable
regulatory requirement(s).
5.18.3 Extent and Nature of Monitoring
The sponsor
should ensure that the trials are adequately monitored. The sponsor should determine the appropriate extent
and nature of monitoring. The
determination of the extent and nature of monitoring should be based on considerations such as the objective,
purpose, design, complexity, blinding, size,
and endpoints of the trial. In general there is a need for on-site monitoring, before, during, and after the
trial; however in exceptional circumstances
the sponsor may determine that central monitoring in conjunction with procedures such as
investigators’ training and meetings, and
extensive written guidance can assure appropriate conduct of the trial
in accordance with GCP. Statistically
controlled sampling may be an acceptable
method for selecting the data to be verified.
5.18.4 Monitor's Responsibilities
The monitor(s) in accordance with the sponsor’s
requirements should ensure that the
trial is conducted and documented properly by carrying out the following activities when relevant and
necessary to the trial and the trial site:
(a) Acting as the main line of communication between
the sponsor and the investigator.
(b) Verifying that the investigator has adequate
qualifications and resources (see 4.1,
4.2, 5.6) and remain adequate throughout the trial period, that facilities, including laboratories, equipment,
and staff, are adequate to safely and
properly conduct the trial and remain adequate throughout the trial period.
(c) Verifying, for the investigational product(s):
(i) That storage times and conditions are
acceptable, and that supplies are sufficient
throughout the trial.
(ii) That the investigational product(s) are
supplied only to subjects who are
eligible to receive it and at the protocol specified dose(s).
(iii) That subjects are provided with necessary
instruction on properly using, handling,
storing, and returning the investigational
product(s).
(iv) That the receipt, use, and return of the
investigational product(s) at the trial
sites are controlled and documented adequately.
(v) That the disposition of unused investigational product(s)
at the trial sites complies with
applicable regulatory requirement(s) and is in
accordance with the sponsor.
(d) Verifying that the investigator follows the
approved protocol and all approved
amendment(s), if any.
(e) Verifying that written informed consent was obtained
before each subject's participation in
the trial.
(f) Ensuring that the investigator receives the
current Investigator's Brochure, all
documents, and all trial supplies needed to conduct the trial properly and to comply with the applicable
regulatory requirement(s).
(g) Ensuring that the investigator and the investigator's
trial staff are adequately informed
about the trial.
(h) Verifying that the investigator and the
investigator's trial staff are performing
the specified trial functions, in accordance with the protocol and any other written agreement between the
sponsor and the investigator/institution,
and have not delegated these functions to
unauthorized individuals.
(i) Verifying that the investigator is enroling only
eligible subjects.
(j) Reporting the subject recruitment rate.
(k) Verifying that source documents and other trial
records are accurate, complete, kept
up-to-date and maintained.
(l) Verifying that the investigator provides all the
required reports, notifications,
applications, and submissions, and that these documents are accurate, complete, timely, legible, dated,
and identify the trial.
(m) Checking the accuracy and completeness of the
CRF entries, source documents and other
trial-related records against each other. The monitor specifically should verify that:
(i) The data required by the protocol are reported
accurately on the CRFs and are
consistent with the source documents.
(ii) Any dose and/or therapy modifications are well
documented for each of the trial
subjects.
(iii) Adverse events, concomitant medications and
intercurrent illnesses are reported in
accordance with the protocol on the CRFs.
(iv) Visits that the subjects fail to make, tests
that are not conducted, and examinations
that are not performed are clearly reported as such on the CRFs.
(v) All withdrawals and dropouts of enrolled
subjects from the trial are reported and
explained on the CRFs.
(n) Informing the investigator of any CRF entry error,
omission, or illegibility. The monitor
should ensure that appropriate corrections, additions, or deletions are made, dated, explained (if necessary),
and initialled by the investigator or by
a member of the investigator's trial staff who is authorized to initial CRF changes for the investigator.
This authorization should be documented.
(o) Determining whether all adverse events (AEs) are
appropriately reported within the time
periods required by GCP, the protocol, the IRB/IEC, the sponsor, and the applicable regulatory
requirement(s).
(p) Determining whether the investigator is
maintaining the essential
documents (see 8. Essential Documents for the
Conduct of a Clinical Trial).
(q) Communicating deviations from the protocol,
SOPs, GCP, and the applicable regulatory
requirements to the investigator and taking
appropriate action designed to prevent recurrence of the detected deviations.
5.18.5 Monitoring Procedures
The
monitor(s) should follow the sponsor’s established written SOPs as well as those procedures that are specified by the
sponsor for monitoring a specific trial.
5.18.6 Monitoring Report
(a) The monitor should submit a written report to
the sponsor after each trialsite visit or trial related communication.
(b) Reports should include the date, site, name of
the monitor, and name of the
investigator or other individual(s) contacted.
(c) Reports should include a summary of what the
monitor reviewed and the monitor's
statements concerning the significant findings/facts, deviations and deficiencies, conclusions, actions taken
or to be taken and/or actions recommended
to secure compliance.
(d) The review and follow-up of the monitoring report
with the sponsor should be documented by
the sponsor’s designated representative.
5.19 Audit
If or when sponsors perform audits, as part of
implementing quality assurance, they should
consider:
5.19.1 Purpose
The purpose
of a sponsor's audit, which is independent of and separate from routine monitoring or quality control
functions, should be to evaluate trial conduct
and compliance with the protocol, SOPs, GCP, and the applicable regulatory requirements.
5.19.2 Selection and Qualification of Auditors
(a) The sponsor should appoint individuals, who are
independent of the clinical
trials/systems, to conduct audits.
(b) The sponsor should ensure that the auditors are
qualified by training and experience to
conduct audits properly. An auditor’s qualifications should be documented.
5.19.3 Auditing Procedures
(a) The sponsor should ensure that the auditing of
clinical trials/systems is conducted in
accordance with the sponsor's written procedures on what to audit, how to audit, the frequency of audits,
and the form and content of audit
reports.
(b) The sponsor's audit plan and procedures for a
trial audit should be guided by the
importance of the trial to submissions to regulatory authorities, the number of subjects in the trial, the type and
complexity of the trial, the level of
risks to the trial subjects, and any identified problem(s).
(c) The observations and findings of the auditor(s)
should be documented.
(d) To preserve the independence and value of the
audit function, the regulatory
authority(ies) should not routinely request the audit reports. Regulatory authority(ies) may seek access to
an audit report on a case by case basis
when evidence of serious GCP non-compliance exists, or in the course of legal proceedings.
(e) When required by applicable law or regulation,
the sponsor should provide an audit
certificate.
5.20 Noncompliance
5.20.1 Noncompliance with the protocol, SOPs, GCP,
and/or applicable regulatory requirement(s)
by an investigator/institution, or by member(s) of the sponsor's staff should lead to prompt action by the
sponsor to secure compliance.
5.20.2 If the monitoring and/or auditing identifies
serious and/or persistent noncompliance
on the part of an investigator/institution, the sponsor should terminate the investigator's/institution’s
participation in the trial. When an investigator's/institution’s
participation is terminated because of noncompliance,
the sponsor should notify promptly the regulatory authority(ies).
5.21 Premature Termination or Suspension of a Trial
If a trial is prematurely terminated or suspended,
the sponsor should promptly inform the
investigators/institutions, and the regulatory authority(ies) of the
termination or suspension and the
reason(s) for the termination or suspension. The IRB/IEC should also be informed promptly and provided the reason(s)
for the termination or suspension by the
sponsor or by the investigator/institution, as specified by the applicable regulatory requirement(s).
5.22 Clinical Trial/Study Reports
Whether the trial is completed or prematurely
terminated, the sponsor should ensure that
the clinical trial reports are prepared and provided to the regulatory
agency(ies) as required by the
applicable regulatory requirement(s). The sponsor should also ensure that the clinical trial reports in
marketing applications meet the standards of
the ICH Guideline for Structure and Content of Clinical Study Reports.
(NOTE: The ICH Guideline for Structure
and Content of Clinical Study Reports specifies that abbreviated study reports may be acceptable
in certain cases.)
5.23 Multicentre Trials
For multicentre trials, the sponsor should ensure
that:
5.23.1 All investigators conduct the trial in strict
compliance with the protocol agreed to
by the sponsor and, if required, by the regulatory authority(ies), and
given approval/favourable opinion by the
IRB/IEC.
5.23.2 The CRFs are designed to capture the required
data at all multicentre trial
sites. For those investigators who are collecting
additional data, supplemental CRFs
should also be provided that are designed to capture the additional data.
5.23.3 The responsibilities of coordinating
investigator(s) and the other participating
investigators are documented prior to the start of the trial.
5.23.4 All investigators are given instructions on
following the protocol, on complying with
a uniform set of standards for the assessment of clinical and laboratory findings, and on completing the CRFs.
5.23.5 Communication between investigators is
facilitated.
4.
INVESTIGATOR
4.1
Investigator’s Qualifications and Agreements
4.1.1 The investigator(s) should be qualified by
education, training, and experience to assume responsibility for the proper
conduct of the trial, should meet all the qualifications specified by the
applicable regulatory requirement(s), and should provide evidence of such
qualifications through up-to-date curriculum vitae and/or other relevant
documentation requested by the sponsor, the IRB/IEC, and/or the regulatory
authority(ies).
4.1.2 The investigator should be thoroughly familiar
with the appropriate use of the investigational product(s), as described in the
protocol, in the current Investigator’s Brochure, in the product information
and in other information sources provided by the sponsor.
4.1.3 The investigator should be aware of, and
should comply with, GCP and the applicable regulatory requirements.
4.1.4 The investigator/institution should permit
monitoring and auditing by the sponsor, and inspection by the appropriate
regulatory authority(ies).
4.1.5 The investigator should maintain a list of
appropriately qualified persons to whom the investigator has delegated
significant trial-related duties.
4.2 Adequate Resources
4.2.1 The investigator should be able to demonstrate
(e.g., based on retrospective data) a potential for recruiting the required
number of suitable subjects within the agreed recruitment period.
4.2.2 The investigator should have sufficient time
to properly conduct and complete the trial within the agreed trial period.
4.2.3 The investigator should have available an
adequate number of qualified staff and adequate facilities for the foreseen
duration of the trial to conduct the trial properly and safely.
4.2.4 The investigator should ensure that all
persons assisting with the trial are adequately informed about the protocol,
the investigational product(s), and their trial-related duties and functions.
4.3 Medical Care of Trial Subjects
4.3.1 A qualified physician (or dentist, when
appropriate), who is an investigator or a sub-investigator for the trial,
should be responsible for all trial-related medical (or dental) decisions.
4.3.2 During and following a subject’s participation
in a trial, the investigator/institution should ensure that adequate medical
care is provided to a subject for any adverse events, including clinically
significant laboratory values, related to the trial. The
investigator/institution should inform a subject when medical care is needed
for intercurrent illness(es) of which the investigator becomes aware.
4.3.3 It is recommended that the investigator inform
the subject’s primary physician about the subject’s participation in the trial
if the subject has a primary physician and if the subject agrees to the primary
physician being informed.
4.3.4 Although a subject is not obliged to give
his/her reason(s) for withdrawing prematurely from a trial, the investigator
should make a reasonable effort to ascertain the reason(s), while fully
respecting the subject’s rights.
4.4 Communication with IRB/IEC
4.4.1 Before initiating a trial, the
investigator/institution should have written and dated approval/favourable opinion
from the IRB/IEC for the trial protocol, written informed consent form, consent
form updates, subject recruitment procedures (e.g., advertisements), and any
other written information to be provided to subjects.
4.4.2 As part of the investigator’s/institution’s
written application to the IRB/IEC, the investigator/institution should provide
the IRB/IEC with a current copy of the Investigator’s Brochure. If the
Investigator’s Brochure is updated during the trial, the
investigator/institution should supply a copy of the updated Investigator’s
Brochure to the IRB/IEC.
4.4.3 During the trial the investigator/institution
should provide to the IRB/IEC all documents subject to review.
4.5 Compliance with Protocol
4.5.1 The investigator/institution should conduct
the trial in compliance with the protocol agreed to by the sponsor and, if
required, by the regulatory authority(ies) and which was given
approval/favourable opinion by theIRB/IEC. The investigator/institution and the
sponsor should sign the protocol, or an alternative contract, to confirm
agreement.
4.5.2 The investigator should not implement any
deviation from, or changes of the protocol without agreement by the sponsor and
prior review and documented approval/favourable opinion from the IRB/IEC of an
amendment, except where necessary to eliminate an immediate hazard(s) to trial
subjects, or when the change(s) involves only logistical or administrative
aspects of the trial (e.g., change in monitor(s), change of telephone
number(s)).
4.5.3 The investigator, or person designated by the
investigator, should document and explain any deviation from the approved
protocol.
4.5.4 The investigator may implement a deviation
from, or a change of, the protocol to eliminate an immediate hazard(s) to trial
subjects without prior IRB/IEC approval/favourable opinion. As soon as
possible, the implemented deviation or change, the reasons for it, and, if
appropriate, the proposed protocol amendment(s) should be submitted: (a) to the
IRB/IEC for review and approval/favourable opinion, (b) to the sponsor for
agreement and, if required, (c) to the regulatory authority(ies).
4.6 Investigational Product(s)
4.6.1 Responsibility for investigational product(s)
accountability at the trial site(s) rests with the investigator/institution.
4.6.2 Where allowed/required, the
investigator/institution may/should assign some or all of the
investigator’s/institution’s duties for investigational product(s)
accountability at the trial site(s) to an appropriate pharmacist or another
appropriate individual who is under the supervision of the
investigator/institution..
4.6.3 The investigator/institution and/or a
pharmacist or other appropriate individual, who is designated by the
investigator/institution, should maintain records of the product’s delivery to
the trial site, the inventory at the site, the use by each subject, and the
return to the sponsor or alternative disposition of unused product(s). These
records should include dates, quantities, batch/serial numbers, expiration
dates (if applicable), and the unique code numbers assigned to the
investigational product(s) and trial subjects. Investigators should maintain
records that document adequately that the subjects were provided the doses specified
by the protocol and reconcile all investigational product(s) received from the
sponsor.
4.6.4 The investigational product(s) should be
stored as specified by the sponsor (see 5.13.2 and 5.14.3) and in accordance
with applicable regulatory requirement(s).
4.6.5 The investigator should ensure that the
investigational product(s) are used only in accordance with the approved
protocol.
4.6.6 The investigator, or a person designated by
the investigator/institution, should explain the correct use of the investigational
product(s) to each subject and should check, at intervals appropriate for the
trial, that each subject is following the instructions properly.
4.7 Randomization Procedures and Unblinding
The investigator should follow the trial’s
randomization procedures, if any, and should ensure that the code is broken
only in accordance with the protocol. If the trial is blinded, the investigator
should promptly document and explain to the sponsor any premature unblinding
(e.g., accidental unblinding, unblinding due to a serious adverse event) of the
investigational product(s).
4.8 Informed Consent of Trial Subjects
4.8.1 In obtaining and documenting informed consent,
the investigator should comply with the applicable regulatory requirement(s),
and should adhere to GCP and to the ethical principles that have their origin
in the Declaration of Helsinki. Prior to the beginning of the trial, the
investigator should have the IRB/IEC’s written approval/favourable opinion of
the written informed consent form and any other written information to be
provided to subjects.
4.8.2 The written informed consent form and any
other written information to be provided to subjects should be revised whenever
important new information becomes available that may be relevant to the
subject’s consent. Any revised written informed consent form, and written
information should receive the IRB/IEC’s approval/favourable opinion in advance
of use. The subject or the subject’s legally acceptable representative should
be informed in a timely manner if new information becomes available that may be
relevant to the subject’s willingness to continue participation in the trial.
The communication of this information should be documented.
4.8.3 Neither the investigator, nor the trial staff,
should coerce or unduly influence a subject to participate or to continue to
participate in a trial.
4.8.4 None of the oral and written information
concerning the trial, including the written informed consent form, should
contain any language that causes the subject or the subject’s legally
acceptable representative to waive or to appear to waive any legal rights, or
that releases or appears to release the investigator, the institution, the
sponsor, or their agents from liability for negligence.
4.8.5 The investigator, or a person designated by
the investigator, should fully inform the subject or, if the subject is unable
to provide informed consent, the subject’s legally acceptable representative,
of all pertinent aspects of the trial including the written information and the
approval/ favourable opinion by the IRB/IEC.
4.8.6 The language used in the oral and written
information about the trial, including the written informed consent form,
should be as non-technical as practical and should be understandable to the
subject or the subject’s legally acceptable representative and the impartial
witness, where applicable.
4.8.7 Before informed consent may be obtained, the
investigator, or a person designated by the investigator, should provide the
subject or the subject’s legally acceptable representative ample time and
opportunity to inquire about details of the trial and to decide whether or not
to participate in the trial. All questions about the trial should be answered
to the satisfaction of the subject or the subject’s legally acceptable
representative.
4.8.8 Prior to a subject’s participation in the
trial, the written informed consent form should be signed and personally dated
by the subject or by the subject’s legallyacceptable representative, and by the
person who conducted the informed consent discussion.
4.8.9 If a subject is unable to read or if a legally
acceptable representative is unable to read, an impartial witness should be
present during the entire informed consent discussion. After the written
informed consent form and any other written information to be provided to
subjects, is read and explained to the subject or the subject’s legally
acceptable representative, and after the subject or the subject’s legally
acceptable representative has orally consented to the subject’s participation
in the trial and, if capable of doing so, has signed and personally dated the
informed consent form, the witness should sign and personally date the consent
form. By signing the consent form, the witness attests that the information in
the consent form and any other written information was accurately explained to,
and apparently understood by, the subject or the subject’s legally acceptable
representative, and that informed consent was freely given by the subject or
the subject’s legally acceptable representative.
4.8.10 Both the informed consent discussion and the
written informed consent form and any other written information to be provided
to subjects should include explanations of the following:
(a) That the trial involves research.
(b) The purpose of the trial.
(c) The trial treatment(s) and the probability for
random assignment to each treatment.
(d) The trial procedures to be followed, including
all invasive procedures.
(e) The subject’s responsibilities.
(f) Those aspects of the trial that are
experimental.
(g) The reasonably foreseeable risks or
inconveniences to the subject and, when applicable, to an embryo, fetus, or
nursing infant.
(h) The reasonably expected benefits. When there is
no intended clinical benefit to the subject, the subject should be made aware
of this.
(i) The alternative procedure(s) or course(s) of
treatment that may be available to the subject, and their important potential
benefits and risks.
(j) The compensation and/or treatment available to
the subject in the event of trial-related injury.
(k) The anticipated prorated payment, if any, to the
subject for participating in the trial.
(l) The anticipated expenses, if any, to the subject
for participating in the trial.
(m) That the subject’s participation in the trial is
voluntary and that the subject may refuse to participate or withdraw from the
trial, at any time, without penalty or loss of benefits to which the subject is
otherwise entitled.
(n) That the monitor(s), the auditor(s), the
IRB/IEC, and the regulatory authority(ies) will be granted direct access to the
subject’s original medical records for verification of clinical trial
procedures and/or data, without violating the confidentiality of the subject,
to the extent permitted by theapplicable laws and regulations and that, by
signing a written informed consent form, the subject or the subject’s legally
acceptable representative is authorizing such access.
(o) That records identifying the subject will be
kept confidential and, to the extent permitted by the applicable laws and/or
regulations, will not be made publicly available. If the results of the trial
are published, the subject’s identity will remain confidential.
(p) That the subject or the subject’s legally
acceptable representative will be informed in a timely manner if information
becomes available that may be relevant to the subject’s willingness to continue
participation in the trial.
(q) The person(s) to contact for further information
regarding the trial and the rights of trial subjects, and whom to contact in
the event of trial-related injury.
(r) The foreseeable circumstances and/or reasons
under which the subject’s participation in the trial may be terminated.
(s) The expected duration of the subject’s
participation in the trial.
(t) The approximate number of subjects involved in
the trial.
4.8.11 Prior to participation in the trial, the
subject or the subject’s legally acceptable representative should receive a
copy of the signed and dated written informed consent form and any other
written information provided to the subjects. During a subject’s participation
in the trial, the subject or the subject’s legally acceptable representative
should receive a copy of the signed and dated consent form updates and a copy
of any amendments to the written information provided to subjects.
4.8.12 When a clinical trial (therapeutic or
non-therapeutic) includes subjects who can only be enrolled in the trial with
the consent of the subject’s legally acceptable representative (e.g., minors,
or patients with severe dementia), the subject should be informed about the
trial to the extent compatible with the subject’s understanding and, if
capable, the subject should sign and personally date the written informed
consent.
4.8.13 Except as described in 4.8.14, a
non-therapeutic trial (i.e. a trial in which there is no anticipated direct
clinical benefit to the subject), should be conducted in subjects who
personally give consent and who sign and date the written informed consent
form.
4.8.14 Non-therapeutic trials may be conducted in
subjects with consent of a legally acceptable representative provided the
following conditions are fulfilled:
(a) The objectives of the trial can not be met by
means of a trial in subjects who can give informed consent personally.
(b) The foreseeable risks to the subjects are low.
(c) The negative impact on the subject’s well-being
is minimized and low.
(d) The trial is not prohibited by law.
(e) The approval/favourable opinion of the IRB/IEC
is expressly sought on the inclusion of such subjects, and the written
approval/ favourable opinion covers this aspect.
Such trials, unless an exception is justified,
should be conducted in patients having a disease or condition for which the
investigational product is intended. Subjects in these trials should be
particularly closely monitored and should be withdrawn if they appear to be
unduly distressed.
4.8.15 In emergency situations, when prior consent
of the subject is not possible, the consent of the subject’s legally acceptable
representative, if present, should be requested. When prior consent of the
subject is not possible, and the subject’s legally acceptable representative is
not available, enrolment of the subject should require measures described in
the protocol and/or elsewhere, with documented approval/favourable opinion by
the IRB/IEC, to protect the rights, safety and well-being of the subject and to
ensure compliance with applicable regulatory requirements. The subject or the
subject’s legally acceptable representative should be informed about the trial
as soon as possible and consent to continue and other consent as appropriate
(see 4.8.10) should be requested.
4.9 Records and Reports
4.9.1 The investigator should ensure the accuracy,
completeness, legibility, and timeliness of the data reported to the sponsor in
the CRFs and in all required reports.
4.9.2 Data reported on the CRF, that are derived
from source documents, should be consistent with the source documents or the
discrepancies should be explained.
4.9.3 Any change or correction to a CRF should be
dated, initialed, and explained (if necessary) and should not obscure the
original entry (i.e. an audit trail should be maintained); this applies to both
written and electronic changes or corrections (see 5.18.4 (n)). Sponsors should
provide guidance to investigators and/or the investigators’ designated
representatives on making such corrections. Sponsors should have written
procedures to assure that changes or corrections in CRFs made by sponsor’s
designated representatives are documented, are necessary, and are endorsed by
the investigator. The investigator should retain records of the changes and
corrections.
4.9.4 The investigator/institution should maintain
the trial documents as specified in Essential Documents for the Conduct of a
Clinical Trial (see 8.) and as required by the applicable regulatory
requirement(s). The investigator/institution should take measures to prevent
accidental or premature destruction of these documents.
4.9.5 Essential documents should be retained until
at least 2 years after the last approval of a marketing application in an ICH
region and until there are no pending or contemplated marketing applications in
an ICH region or at least 2 years have elapsed since the formal discontinuation
of clinical development of the investigational product. These documents should
be retained for a longer period however if required by the applicable
regulatory requirements or by an agreement with the sponsor. It is the
responsibility of the sponsor to inform the investigator/institution as to when
these documents no longer need to be retained.
4.9.6 The financial aspects of the trial should be
documented in an agreement between the sponsor and the
investigator/institution.
4.9.7 Upon request of the monitor, auditor, IRB/IEC,
or regulatory authority, the investigator/institution should make available for
direct access all requested trial-related records.
4.10 Progress Reports
4.10.1 The investigator should submit written
summaries of the trial status to the IRB/IEC annually, or more frequently, if
requested by the IRB/IEC.
4.10.2 The investigator should promptly provide
written reports to the sponsor, the IRB/IEC (see 3.3.8) and, where applicable,
the institution on any changes significantly affecting the conduct of the
trial, and/or increasing the risk to subjects.
4.11 Safety Reporting
4.11.1 All serious adverse events (SAEs) should be
reported immediately to the sponsor except for those SAEs that the protocol or
other document (e.g., Investigator’s Brochure) identifies as not needing
immediate reporting. The immediate reports should be followed promptly by
detailed, written reports. The immediate and follow-up reports should identify
subjects by unique code numbers assigned to the trial subjects rather than by
the subjects’ names, personal identification numbers, and/or addresses. The
investigator should also comply with the applicable regulatory requirement(s)
related to the reporting of unexpected serious adverse drug reactions to the
regulatory authority(ies) and the IRB/IEC.
4.11.2 Adverse events and/or laboratory
abnormalities identified in the protocol as critical to safety evaluations
should be reported to the sponsor according to the reporting requirements and
within the time periods specified by the sponsor in the protocol.
4.11.3 For reported deaths, the investigator should
supply the sponsor and the IRB/IEC with any additional requested information
(e.g., autopsy reports and terminal medical reports).
4.12 Premature Termination or Suspension of a Trial
If the trial is prematurely terminated or suspended
for any reason, the investigator/institution should promptly inform the trial
subjects, should assure appropriate therapy and follow-up for the subjects,
and, where required by the applicable regulatory requirement(s), should inform
the regulatory authority(ies). In addition:
4.12.1 If the investigator terminates or suspends a
trial without prior agreement of the sponsor, the investigator should inform
the institution where applicable, and the investigator/institution should
promptly inform the sponsor and the IRB/IEC, and should provide the sponsor and
the IRB/IEC a detailed written explanation of the termination or suspension.
4.12.2 If the sponsor terminates or suspends a trial
(see 5.21), the investigator should promptly inform the institution where
applicable and the investigator/institution should promptly inform the IRB/IEC
and provide the IRB/IEC a detailed written explanation of the termination or
suspension.
4.12.3 If the IRB/IEC terminates or suspends its approval/favourable
opinion of a trial (see 3.1.2 and 3.3.9), the investigator should inform the
institution where applicable and the investigator/institution should promptly
notify the sponsor and provide the sponsor with a detailed written explanation
of the termination or suspension.
4.13 Final Report(s) by Investigator
Upon completion of the trial, the investigator,
where applicable, should inform the institution; the investigator/institution
should provide the IRB/IEC with a summary of the trial’s outcome, and the
regulatory authority(ies) with any reports required.
IRB / IEC Essential Documentation
3. INSTITUTIONAL REVIEW
BOARD/INDEPENDENT ETHICS COMMITTEE (IRB/IEC)
3.1
Responsibilities
3.1.1 An IRB/IEC should safeguard the rights,
safety, and well-being of all trial subjects. Special attention should be paid
to trials that may include vulnerable subjects.
3.1.2 The IRB/IEC should obtain the following
documents:
trial
protocol(s)/amendment(s), written informed consent form(s) and consent form
updates that the investigator proposes for use in the trial, subject
recruitment procedures (e.g. advertisements), written information to be
provided to subjects, Investigator's Brochure (IB), available safety
information, information about payments and compensation available to subjects,
the investigator’s current curriculum vitae and/or other documentation
evidencing qualifications, and any other documents that the IRB/IEC may need to
fulfil its responsibilities.
The
IRB/IEC should review a proposed clinical trial within a reasonable time and
document its views in writing, clearly identifying the trial, the documents
reviewed and the dates for the following:
- approval/favourable opinion;
- modifications required prior to
its approval/favourable opinion;
- disapproval / negative opinion;
and
- termination/suspension of any
prior approval/favourable opinion.
3.1.3 The IRB/IEC should consider the qualifications
of the investigator for the proposed trial, as documented by a current
curriculum vitae and/or by any other relevant documentation the IRB/IEC
requests.
3.1.4 The IRB/IEC should conduct continuing review
of each ongoing trial at intervals appropriate to the degree of risk to human
subjects, but at least once per year.
3.1.5 The IRB/IEC may request more information than
is outlined in paragraph 4.8.10 be given to subjects when, in the judgement of
the IRB/IEC, the additional information would add meaningfully to the
protection of the rights, safety and/or well-being of the subjects.
3.1.6 When a non-therapeutic trial is to be carried
out with the consent of the subject’s legally acceptable representative (see
4.8.12, 4.8.14), the IRB/IEC should determine that the proposed protocol and/or
other document(s) adequately addresses relevant ethical concerns and meets
applicable regulatory requirements for such trials.
3.1.7 Where the protocol indicates that prior
consent of the trial subject or the subject’s legally acceptable representative
is not possible (see 4.8.15), the IRB/IEC should determine that the proposed
protocol and/or other document(s) adequately addresses relevant ethical
concerns and meets applicable regulatory requirements for such trials (i.e. in
emergency situations).
3.1.8 The IRB/IEC should review both the amount and
method of payment to subjects to assure that neither presents problems of
coercion or undue influence on the trial subjects. Payments to a subject should
be prorated and not wholly contingent on completion of the trial by the
subject.
3.1.9 The IRB/IEC should ensure that information
regarding payment to subjects, including the methods, amounts, and schedule of
payment to trial subjects, is set forth in the written informed consent form
and any other written information to be provided to subjects. The way payment
will be prorated should be specified.
3.2
Composition, Functions and Operations
3.2.1 The IRB/IEC should consist of a reasonable
number of members, who collectively have the qualifications and experience to
review and evaluate the science, medical aspects, and ethics of the proposed
trial. It is recommended that the IRB/IEC should include:
(a) At least five members.
(b) At least one member whose
primary area of interest is in a nonscientific area.
(c) At least one member who is
independent of the institution/trial site.
Only
those IRB/IEC members who are independent of the investigator and the sponsor
of the trial should vote/provide opinion on a trial-related matter.
A
list of IRB/IEC members and their qualifications should be maintained.
3.2.2 The IRB/IEC should perform its functions
according to written operating procedures, should maintain written records of
its activities and minutes of its meetings, and should comply with GCP and with
the applicable regulatory requirement(s).
3.2.3 An IRB/IEC should make its decisions at
announced meetings at which at least a quorum, as stipulated in its written
operating procedures, is present.
3.2.4 Only members who participate in the IRB/IEC
review and discussion should vote/provide their opinion and/or advise.
3.2.5 The investigator may provide information on
any aspect of the trial, but should not participate in the deliberations of the
IRB/IEC or in the vote/opinion of the IRB/IEC.
3.2.6 An IRB/IEC may invite nonmembers with
expertise in special areas for assistance.
3.3
Procedures
The IRB/IEC should establish,
document in writing, and follow its procedures, which should include:
3.3.1 Determining its composition (names and
qualifications of the members) and the authority under which it is established.
3.3.2 Scheduling, notifying its members of, and
conducting its meetings.
3.3.3 Conducting initial and continuing review of
trials.
3.3.4 Determining the frequency of continuing
review, as appropriate.
3.3.5 Providing, according to the applicable
regulatory requirements, expedited review and approval/favourable opinion of
minor change(s) in ongoing trials that have the approval/favourable opinion of
the IRB/IEC.
3.3.6 Specifying that no subject should be admitted
to a trial before the IRB/IEC issues its written approval/favourable opinion of
the trial.
3.3.7 Specifying that no deviations from, or changes
of, the protocol should be initiated without prior written IRB/IEC
approval/favourable opinion of an appropriate amendment, except when necessary
to eliminate immediate hazards to the subjects or when the change(s) involves
only logistical or number(s)) (see 4.5.2).
3.3.8 Specifying that the investigator should
promptly report to the IRB/IEC:
(a) Deviations from, or changes
of, the protocol to eliminate immediate hazards to the trial subjects (see
3.3.7, 4.5.2, 4.5.4).
(b) Changes increasing the risk
to subjects and/or affecting significantly the conduct of the trial (see
4.10.2).
(c) All adverse drug reactions
(ADRs) that are both serious and unexpected.
(d) New information that may
affect adversely the safety of the subjects or the conduct of the trial.
3.3.9 Ensuring that the IRB/IEC promptly notify in
writing the investigator/institution concerning:
(a) Its trial-related
decisions/opinions.
(b) The reasons for its
decisions/opinions.
(c) Procedures for appeal of its
decisions/opinions.
3.4
Records
The IRB/IEC should retain all
relevant records (e.g., written procedures, membership lists, lists of
occupations/affiliations of members, submitted documents, minutes of meetings,
and correspondence) for a period of at least 3 years after completion of the
trial and make them available upon request from the regulatory authority(ies).
The IRB/IEC may be asked by investigators, sponsors
or regulatory authorities to provide its written procedures and membership
lists.
The
INDIAN / USA / EU Directives on GCP in Clinical Trials
The
INDIAN Directives on GCP in Clinical Trials
INTRODUCTION
The history of Good Clinical Practice (GCP) statute
traces back to one of the oldest enduring traditions in the history of
medicine: The Hippocratic Oath. As the
guiding ethical code it is primarily known for its edict to do no harm to the
patient. However, the complexities of
modern medicine research necessitate a more elaborate set of guidelines that
address a Physician’s ethical and scientific responsibilities such as obtaining
informed consent or disclosing risk while involved in biomedical research.
Good Clinical Practice is a set of guidelines for
biomedical studies which encompasses the design, conduct, termination, audit,
analysis, reporting and documentation of the studies involving human
subjects. The fundamental tenet of GCP
is that in research on man, the interest of science and society should never
take precedence over considerations related to the well being of the study
subject. It aims to ensure that the
studies are scientifically and ethically sound and that the clinical properties
of the pharmaceutical substances under investigation are properly
documented. The guidelines seek to
establish two cardinal principles: protection of the rights of human subjects
and authenticity of biomedical data generated.
These
guidelines have been evolved with consideration of WHO, ICH, USFDA and European
GCP guidelines as well as the Ethical Guidelines for Biomedical research on
Human Subjects issued by the Indian Council of Medical Research. They should be followed for carrying out all
biomedical research in India at all stages of drug development, whether prior
or subsequent to product registration in India.
DEFINITIONS
Act
Wherever relevant, the Act means Drugs &
Cosmetics Act 1940 (23 of 1940) and the Rules made thereunder.
Adverse Event (AE)
Any untoward medical occurrence (including a symptom
/ disease or an abnormal laboratory finding) during treatment with a
pharmaceutical product in a patient or a human volunteer that does not
necessarily have a relationship with the treatment being given. Also see
Serious Adverse Event
Adverse Drug Reaction (ADR)
(a) In case of approved pharmaceutical
products: A noxious and unintended
response at doses normally used or tested in humans
(b) In case
of new unregistered pharmaceutical products (or those products which are not
yet approved for the medical condition where they are being tested): A noxious
and unintended response at any dose(s)
The phrase ADR differs from AE, in case of an ADR
there appears to be a reasonable possibility that the adverse event is related
with the medicinal product being studied.
In clinical trials, an untoward medical occurrence
seemingly caused by overdosing, abuse / dependence and interactions with other
medicinal products is also considered as an ADR.
Adverse drug reactions are type A (pharmacological)
or type B (idiosyncratic). Type A reactions represent an augmentation of the
pharmacological actions of a drug. They are dose-dependent and are, therefore,
readily reversible on reducing the dose or withdrawing the drug. In contrast,
type B adverse reactions are bizarre and cannot be predicted from the known
pharmacology of the drug.
Audit of a Trial
A systematic verification of the study, carried out
by persons not directly involved, such as:
(a) Study
related activities to determine consistency with the Protocol
(b) Study
data to ensure that there are no contradictions on Source Documents. The audit should also compare data on the
Source Documents with the interim or final report. It should also aim to find out if practices
were employed in the development of data that would impair their validity.
(c)
Compliance with the adopted Standard Operating Procedures (SOPs)
Blinding / Masking
A method of “control experimentation” in which one
or more parties involved are not informed of the treatment being given. Single blind refers to the study subject(s)
being unaware, while Double blind refers to the study subject(s) and/or
investigator(s), monitor, data analyst(s) are being unaware of the treatment
assigned.
Case Record Form (CRF)
A document designed in consonance with the Protocol,
to record data and other information on each trial subject. The Case Record Form should be in such a form
and format that allows accurate input, presentation, verification, audit and
inspection of the recorded data. A CRF
may be in printed or electronic format.
Clinical
Trial (Clinical Study)
A systematic study of pharmaceutical products on
human subjects – (whether patients or non-patient volunteers) – in order to
discover or verify the clinical, pharmacological (including pharmacodynamics /
pharmacokinetics), and / or adverse effects, with the object of determining
their safety and / or efficacy.
Human/Clinical
Pharmacology trials (Phase I)
The objective of phase I of trials is to determine
the maximum tolerated dose in humans; pharmacodynamic effect, adverse
reactions, if any, with their nature and intensity; and pharmacokinetic
behaviour of the drug as far as possible. These studies are often carried out
in healthy adult volunteers using clinical, physiological and biochemical
observations. At least 2 subjects should be used on each dose.
Phase I trials are usually carried out by
investigators trained in clinical pharmacology and having the necessary
facilities to closely observe and monitor the subjects. These may be carried
out at one or two centres.
Exploratory
trials (Phase II)
In phase II trials a limited number of patients are
studied carefully to determine possible therapeutic uses, effective dose range
and further evaluation of safety and pharmacokinetics. Normally 10-12 patients
should be studied at each dose level. These studies are usually limited to 3-4
centres and carried out by clinicians specialized on the concerned therapeutic
areas and having adequate facilities to perform the necessary investigations
for efficacy and safety.
Confirmatory
trials (Phase III)
The purpose of these trials is to obtain sufficient
evidence about the efficacy and safety of the drug in a larger number of
patients, generally in comparison with a standard drug and/or a placebo as
appropriate. These trials may be carried out by clinicians in the concerned
therapeutic areas, having facilities appropriate to the protocol. If the drug
is already approved/marketed in other countries, phase III data should
generally be obtained on at least 100 patients distributed over 3-4 centres
primarily to confirm the efficacy and safety of the drug, in Indian patients
when used as recommended in the product monograph for the claims made.
Data on ADRs observed during clinical use of the
drug should be reported along with a report on its efficacy in the prescribed
format. The selection of clinicians for such monitoring and supply of drug to
them will need approval of the licensing authority under Rule 21 of the Act.
Phase
IV
Studies performed after marketing of the
pharmaceutical product. Trials in phase IV are carried out on the basis of the
product characteristics on which the marketing authorization was granted and
are normally in the form of post-marketing surveillance, assessment of
therapeutic value, treatment strategies used and safety profile. Phase IV
studies should use the same scientific and ethical standards as applied in
pre-marketing studies.
After a product has been placed on the market,
clinical trials designed to explore new indications, new methods of
administration or new combinations, etc. are normally considered as trials for
new pharmaceutical products.
Comparator Product
A pharmaceutical product (including placebo) used as
a reference in a clinical trial.
Confidentiality
Maintenance of privacy of study subjects including
their personal identity and all medical information, from individuals other
than those prescribed in the Protocol.
Confidentiality also covers the prevention of disclosure of sponsor’s
proprietary information to unauthorised persons.
Co-Investigator
A person legally qualified to be an investigator, to
whom the Investigator delegates a part of his responsibilities.
Co-ordinating Investigator
See Principal Investigator
Clinical Research Organisation (CRO)
An organisation to which the sponsor may transfer or
delegate some or all of the tasks, duties and / or obligations regarding a
Clinical Study. All such contractual
transfers of obligations should be defined in writing. A CRO is a scientific body – commercial,
academic or other.
Contract
A written, dated and signed document describing the
agreement between two or more parties involved in a biomedical study, namely
Investigator, Sponsor, Institution. Typically, a contract sets out delegation /
distribution of responsibilities, financial arrangements and other pertinent
terms. The “Protocol” may form the basis of “Contract”.
Documentation
All records (including written documents,
electronic, magnetic or optical records, scans, x-rays etc.) that describe or
record the methods, conduct and results of the study, and the actions
taken. The Documents include Protocol,
copies of submissions and approvals from the office of the Drugs Controller
General of India, ethics committee, investigator(s)’ particulars, consent
forms, monitor reports, audit certificates, relevant letters, reference ranges,
raw data, completed CRFs and the final report. Also see: Essential Documents
Escape
Treatment
A supplementary treatment, usually given to
alleviate pain in placebo-controlled trials, to relieve the trial subject of
the symptoms caused by the investigated disease in a study.
Essential
Documents
The Documents that permit evaluation of the conduct
of a study and the quality of the data generated. See Appendix V.
Ethics
Committee
An independent review board or committee comprising
of medical / scientific and non-medical / non-scientific members, whose
responsibility is to verify the protection of the rights, safety and well-being
of human subjects involved in a study.
The independent review provides public reassurance by objectively,
independently and impartially reviewing and approving the “Protocol”, the
suitability of the investigator(s), facilities, methods and material to be used
for obtaining and documenting “Informed Consent” of the study subjects and
adequacy of confidentiality safeguards.
Final
Report
A complete and comprehensive description of the
study after its completion. It includes
description of experimental and statistical methods and materials, presentation
and evaluation of the results, statistical analyses and a critical ethical,
statistical and clinical appraisal. The Investigator’s declaration closing the
study is a part of the Final Report.
Good
Clinical Practice (GCP)
It is a standard for clinical studies or trials that
encompasses the design, conduct, monitoring, termination, audit, analyses,
reporting and documentation of the studies.
It ensures that the studies are implemented and reported in such a
manner that there is public assurance that the data are credible, accurate and
that the rights, integrity and confidentiality of the subjects are
protected. GCP aims to ensure that the
studies are scientifically authentic and that the clinical properties of the
“Investigational Product” are properly documented.
Impartial
Witness
An impartial independent witness who will not be
influenced in any way by those who are involved in the Clinical Trial, who
assists at the informed consent process and documents the freely given oral
consent by signing and dating the written confirmation of this consent.
Informed
Consent
Voluntary written assent of a subject’s willingness
to participate in a particular study and in its documentation. The confirmation is sought only after
information about the trial including an explanation of its status as research,
its objectives, potential benefits, risks and inconveniences, alternative treatment
that may be available and of the subject’s rights and responsibilities has been
provided to the potential subject.
Inspection
An official review/ examination conducted by
regulatory authority(ies) of the documents, facilities, records and any other
resources that are deemed by the authority(ies) to be related to the
study. The inspection may be carried out
at the site of the trial, at the sponsor’s / or CRO’s facilities in order to
verify adherence to GCP as set out in these documents.
Institution
Any public or private medical facility where a
clinical study is conducted.
Investigator
A person responsible for the conduct of the study at
the trial site. Investigator is
responsible for the rights, health and welfare of the study subjects. In case the study is conducted by a team of
investigators at the study site then the designated leader of the team should
be the Principal Investigator. Also see Principal Investigator,
Sub-investigator.
Investigational Labelling
Labelling developed specifically for products
involved in the study.
Investigational Product
A pharmaceutical product (including the Comparator
Product) being tested or used as reference in a clinical study. An
Investigational Product may be an active chemical entity or a formulated dosage
form.
Investigator’s Brochure
A collection of data (including justification for
the proposed study) for the Investigator consisting of all the clinical as well
as non-clinical information available on the Investigational Product(s) known
prior to the onset of the trial. There should be adequate data to justify the
nature, scale and duration of the proposed trial and to evaluate the potential
safety and need for special precautions. If new substantially relevant data is
generated during the trial, the information in the Investigator’s Brochure must
be updated. See Appendix IV.
Monitor
A person appointed by the Sponsor or Contract
Research Organisation (CRO) for monitoring and reporting the progress of the
trial and for verification of data. The monitor ensures that the trial is
conducted, recorded and reported in accordance with the Protocol, Standard
Operating Procedures (SOPs), Good Clinical Practice (GCP) and the applicable
regulatory requirements.
Multi-Centric Study
A clinical trial conducted according to one single
protocol in which the trial is taking place at different investigational sites,
therefore carried out by more than one investigator.
Non-Clinical Study
Biomedical studies that are not performed on human
subjects.
Non-Therapeutic Study
A study in which there is no anticipated direct
clinical benefit to the Subject(s). Such
studies, unless an exception is justified, should be conducted in patient(s)
having a disease or condition for which the Investigational Product is
intended. Subject(s) in these studies
should be particularly closely monitored and should be withdrawn if they appear
to be unduly distressed.
Pharmaceutical Product(s)
Any substance or combination of substances which has
a therapeutic, prophylactic or diagnostic purpose or is intended to modify
physiological functions, and presented in a dosage form suitable for
administration to humans.
Principal Investigator
The investigator who has the responsibility to
co-ordinate between the different Investigators involved in a study at one site
or different sites in case of a multi-center study.
Protocol
A document that states the background, objectives,
rationale, design, methodology (including the methods for dealing with AEs,
withdrawals etc.) and statistical considerations of the study. It also states
the conditions under which the study shall be performed and managed.
A list of
items to be included in the Protocol is compiled in a subsequent chapter.
The content and format of the protocol should take
into consideration the adopted SOPs, the regulatory requirements and the
guiding principles of GCP.
The term Protocol, unless otherwise specified,
relates to the latest amended version of the document, read in conjunction with
all its appendices and enclosures.
Protocol Amendment(s)
Any changes or formal clarifications appended to the
protocol. All Protocol Amendments should
be agreed upon and signed by the persons who were the signatories to the
Protocol.
Quality Assurance (QA)
Systems and processes established to ensure that the
trial is performed and the data are generated in compliance with GCP. QA is validated through in-process Quality
Control and in and post-process auditing of clinical trial process as well as
data.
Quality Control (QC)
The operational techniques and activities undertaken
within the system of QA to verify that the requirements for quality of the
trial related activities have been fulfilled.
QC activities concern everybody involved with planning, conducting,
monitoring, evaluating, data handling and reporting. The objective of QC is to
avoid exposure of study subjects to unnecessary risks and to avoid false
conclusions being drawn from unreliable data.
Randomisation
The process of assigning study subjects to either
the treatment or the control group.
Randomisation gives all subjects the same chance of being in either
group in order to reduce bias.
Regulatory Authority
The Drugs Controller General of India or an office
nominated by him is the regulatory authority for the purpose of carrying out
Clinical Trials in India. The Regulatory Authority approves the study Protocol,
reviews the submitted data and conducts inspections.
Raw Data
It refers to all records or certified copies of the
original clinical and laboratory findings or other activities in a clinical
study necessary for the reconstruction and evaluation of the trial. Also see
Source Data.
Serious
Adverse Event (SAE) or Serious Adverse Drug Reaction (SADR)
An AE or ADR that is associated with death,
inpatient hospitalisation (in case the study was being conducted on
out-patients), prolongation of hospitalisation (in case the study was being
conducted on in-patients), persistent or significant disability or incapacity,
a congenital anomaly or birth defect, or is otherwise life threatening.
Schedule
Unless repugnant to the context, the Schedule means
Schedule Y to the Drugs & Cosmetics
Rules. (Reproduced here at Appendix II)
Source
Data
Original documents (or their verified and certified
copies) necessary for evaluation of the Clinical Trial. These documents may include Study Subjects’
files, recordings from automated instruments, tracings, X-Ray and other films,
laboratory notes, photographic negatives, magnetic media, hospital records,
clinical and office charts, Subjects’ diaries, evaluation check-lists, and
pharmacy dispensing records.
Sponsor
An individual or a company or an institution that
takes the responsibility for the initiation, management and / or financing of a
Clinical Study. An Investigator who
independently initiates and takes full responsibility for a trial automatically
assumes the role of a Sponsor.
Study
Product
Any Pharmaceutical Product or Comparator Product
used in a clinical study.
Sub-Investigator
See
Co-Investigator
Subject
Files / Patient Files
A file containing demographic and medical
information about a study subject. It
includes hospital files, consultation records or special subject files allowing
the authenticity of the information presented in CRF to be verified and where
necessary allowing it to be completed or corrected. The conditions regulating the use and
consultation of such documents must be honoured as prescribed under Confidentiality.
Study Subject (Subject)
An individual participating in a clinical trial as a
recipient of the Investigational Product.
A Study Subject may be a healthy person volunteering
in a trial or a person with a medical condition that is unrelated to the use of
the Investigational Product or a person whose medical condition is relevant to
the use of the Investigational Product.
Standard Operating Procedures (SOP)
Standard elaborate written instructions to achieve
uniformity of performance in the management of clinical studies. SOPs provide a general framework for the
efficient implementation and performance of all the functions and activities
related to a particular study.
Subject Identification Code
A unique identification number / code assigned by
the Investigator to each Study Subject to protect the Subject’s identity. Subject Identification Code is used in lieu
of the Subject’s name for all matters related to the study.
Study Management
Steering, supervising, data management and
verification, statistical processing and preparation of the study report.
Validation
Validation of Study: The process of proving, in
accordance with the principles of Good Clinical Practice, that any procedure,
process equipment, material, activity or system actually leads to the expected
results
Validation of Data: The procedures carried out to
ensure and prove that the data contained in the final report match the original
observations. The procedure is applied
to Raw Data, CRFs, computer software, printouts, statistical analyses and
consumption of Study Product / Comparator Product.
The
USA Directives on GCP
Refer
the following link
The
EU Directives on GCP
Good clinical practice (GCP) is an international
ethical and scientific quality standard for designing, recording and reporting
trials that involve the participation of human subjects. Compliance with this
standard provides public assurance that the rights, safety and wellbeing of
trial subjects are protected and that clinical-trial data are credible.
The protection of clinical trial subjects is
consistent with the principles set out in the Declaration of Helsinki. This is
a statement of ethical principles developed by the World Medical Association.
Requirements for the conduct of clinical trials in
the European Union (EU), including GCP and good manufacturing practice (GMP)
and GCP or GMP inspections, are implemented in:
the 'Clinical Trial Directive' (Directive
2001/20/EC);
the 'GCP Directive' (Directive 2005/28/EC).
Information concerning the activities in EU Member
States can be found via the Heads of Medicines Agencies.
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.
The development of this paper followed on from the
publication of the strategy paper on acceptance of clinical trials conducted in
third countries and an international workshop in September 2010.
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
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 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.
How will the introduction affect
clinical research
-----------------------------------------------------------------
Extracts
from the guidance documents Possible sanctions for non- compliance
Guidance
documents accessible from this page represent the Agency's current thinking on
good clinical practice (GCP) and the conduct of clinical trials. As with all
guidance documents, they do not create or confer any rights for or on any
person and do not operate to bind FDA or the public. An alternative approach
may be used if such approach satisfies the requirements of the applicable
statute and regulations. However, in many places throughout these documents,
specific regulations are cited and the requirements of the regulations are
reiterated. The regulations are enforceable.
Guidance
Documents Grouped by Topic:
·
General
General
This
guidance is intended to assist clinical investigators, Institutional Review
Boards (IRBs), sponsors, and other interested parties in understanding the
FDA's process for handling clinical investigations that include children as
subjects and that have been referred to FDA for review under 21 CFR 50.54.
This
guidance is intended to assist sponsors of clinical trials in determining when
a data monitoring committee (DMC) is needed for study monitoring, and how such
committees should operate.
This
guidance describes FDA’s longstanding policy that already-accrued data,
relating to individual who cease participating in a study, are to be maintained
as part of the study data. This pertains to data from individuals who
decide to discontinue participation in a study, who are withdrawn by their
legally authorized representative, as applicable, or who are discontinued from
participation by the clinical investigator.
This
guidance is intended to assist Institutional Review Boards (IRBs), clinical
investigators and sponsors in the development, conduct, and oversight of
investigations to determine the safety and effectiveness of FDA regulated
products (e.g., drugs, including biological drug products, devices) in
emergency settings when an exception from the informed consent requirements is
requested under Title 21, Code of Federal Regulations, Section 50.24 (21 CFR
50.24).
This
guidance is intended to assist clinical investigators, industry, and
FDA staff in interpreting and complying with the regulations
governing financial disclosure by clinical investigators, 21 CFR
part 54.
This
guidance document, developed at the department (DHHS) level, applies to all
human subject research conducted or supported by HHS agencies or regulated by
the Food and Drug Administration.
This
guidance provides an overview of the responsibilities of a person who conducts
a clinical investigation of a drug, biological product, or medical device (an
investigator as defined in 21 CFR 312.3(b) and 21 CFR 812.3(i)). It is
intended to clarify for investigators and sponsors FDA’s expectations
concerning the investigator’s responsibility (1) to supervise a clinical study
in which some study tasks are delegated to employees or colleagues of the
investigator or other third parties and (2) to protect the rights, safety, and
welfare of study subjects.
Monitoring
of Clinical Investigators, Guideline for the, Guidance for Industry (Document Removed)
FDA is
currently revising this guidance. If assistance is needed concerning
monitoring of clinical investigators, please contact the appropriate Center or,
for general monitoring questions, the Office of Good Clinical Practice (OGCP)
via our GCP questions box at gcp.questions@fda.hhs.gov.
This
guidance describes how FDA reviews and evaluates existing, modified, or newly
created patient-reported outcome (PRO) instruments used to support claims in
approved medical product labeling. A PRO instrument (i.e., a
questionnaire plus the information and documentation that support its use) is a
means to capture PRO data used to measure treatment benefit or risk in medical
product clinical trials. This guidance does not address the use of PRO
instruments for purposes beyond evaluation of claims made about a medical
product in labeling nor disease-specific issues.
The
guidance provides recommendations to sponsors holding investigational new drug
applications (INDs), new drug applications (NDAs), and biologics license
applications (BLAs) on what pharmacogenomic data to submit to the agency during
the drug development process, the format of submissions, and how the data will
be used in regulatory decision making. The guidance is intended to facilitate
scientific progress in the area of pharmacogenomics.
This
guidance recommends using a standardized approach for collecting and reporting
race and ethnicity information in clinical trials conducted in the United
States and abroad for certain FDA regulated products. The recommended
standardized approach was developed by the Office of Management and Budget
(OMB). The guidance lists the OMB categories for race and ethnicity and
describes FDA's reasons for recommending the use of these categories. In
addition, this guidance recommends a format for race and ethnicity data within
study data that are submitted in standardized data sets such as the Study Data
Tabulation Model or in the electronic Common Technical Document (eCTD).
This
guidance is intended to provide assistance to the research community in
interpreting requirements for submitting reports of unanticipated problems,
including certain adverse events reports, to the IRB.
This
guidance is intended to assist sponsors, institutions, institutional review
boards (IRBs), and clinical investigators involved in multicenter clinical
research in meeting the requirements of 21 CFR part 56 by facilitating the use
of a centralized IRB review process (use of a single central IRB), especially
in situations where centralized review could improve efficiency of IRB review.
This
guidance provides clarification for IRBs of their responsibilities for
reviewing and approving stand-alone authorizations under the HIPAA Privacy
Rule.
This
guidance is intended to help sponsors, investigators and Institutional Review
Boards better understand the new informed consent requirement set forth in 21
CFR 50.25(c). The guidance will assist those involved in applicable
FDA-regulated clinical trials better understand the new informed consent
requirement, including small businesses.
IRB Continuing Review after Clinical Investigation
Approval, Guidance for IRBs, Clinical Investigators, and Sponsors (PDF -
320 KB)
This guidance is intended to assist institutional review boards (IRBs) in carrying out their continuing review responsibility under 21 CFR 56.108(a) and 56.109(f) by providing recommendations regarding the criteria, process, and frequency of continuing review to assure the protection of the rights and welfare of human subjects enrolled in clinical investigations. This guidance should also help clinical investigators and sponsors better understand their responsibilities related to continuing review.
This guidance is intended to assist institutional review boards (IRBs) in carrying out their continuing review responsibility under 21 CFR 56.108(a) and 56.109(f) by providing recommendations regarding the criteria, process, and frequency of continuing review to assure the protection of the rights and welfare of human subjects enrolled in clinical investigations. This guidance should also help clinical investigators and sponsors better understand their responsibilities related to continuing review.
This
guidance is intended to assist IRBs in complying with the requirement for IRB
registration under amended 21 CFR 56.106, effective July 14, 2009.
Registration is accomplished through a modified version of the Internet-based
registration system used by OHRP for registration of IRBs that are designated
by institutions under FWAs. This guidance document addresses basic
information, such as why FDA issued a new rule requiring registration, which
IRBs are subject to the regulation, the type of information to be provided when
registering, and implications of non-compliance.
This
guidance is intended to provide information for those using radioactive drugs
for certain research purposes to help determine whether research studies can be
conducted under 21 CFR 361.1, Prescription Drugs for Human Use Generally
Recognized as Safe and Effective and Not Misbranded: Drugs Used in Research, or
whether research studies must be conducted under 21 CFR part 312,
Investigational New Drug Application (IND).
This
guidance is intended to provide information for those using radioactive drugs
for certain research purposes to help determine whether research studies can be
conducted under 21 CFR 361.1, Prescription Drugs for Human Use Generally
Recognized as Safe and Effective and Not Misbranded: Drugs Used in Research, or
whether research studies must be conducted under 21 CFR part 312,
Investigational New Drug Application (IND).
This
guidance is intended to provide guidance to industry on the meaning of the term
"available therapy" as currently used by the Center for Drug
Evaluation and Research (CDER) and the Center for Biologics Evaluation and
Research (CBER) in the FDA in the specific circumstances described in the
guidance.
Inspection
of clinical and analytical sites that perform bioavailability (BA) and
bioequivalence (BE) studies frequently reveals the absence of reserve samples
at the testing facilities where the studies are conducted. The guidance is
intended to clarify how to distribute test articles and reference standards to
testing facilities, how to randomly select reserve samples, and how to retain
reserve samples.
This
guidance for industry and clinical investigators provides information on one
use by FDA of its authority to impose a clinical hold on a study or study site
if FDA finds that human subjects are or would be exposed to unreasonable and
significant risk of illness or injury. Specifically, this guidance
describes circumstances in which FDA may impose a clinical hold based on
credible evidence that a clinical investigator conducting the study has
committed serious violations of FDA regulations on clinical trials of human
drugs and biologics.
Enforcement
of Safety Reporting Requirements for INDs and BA/BE Studies, Guidance for
Industry and Investigators (PDF - 41KB)
This document provides guidance to sponsors and investigators on enforcement of FDA’s final rule, "Investigational New Drug Safety Reporting Requirements for Human Drug and Biological Products and Safety Reporting Requirements for Bioavailability and Bioequivalence Studies in Humans" (75 FR 59935, September 29, 2010). This guidance contains information regarding the Agency’s intent to exercise enforcement discretion regarding the reporting requirements in the final rule until September 28, 2011.
This document provides guidance to sponsors and investigators on enforcement of FDA’s final rule, "Investigational New Drug Safety Reporting Requirements for Human Drug and Biological Products and Safety Reporting Requirements for Bioavailability and Bioequivalence Studies in Humans" (75 FR 59935, September 29, 2010). This guidance contains information regarding the Agency’s intent to exercise enforcement discretion regarding the reporting requirements in the final rule until September 28, 2011.
This
guidance describes the preclinical and clinical issues as well as chemistry,
manufacturing and controls information that should be considered when planning
exploratory studies including studies of related drugs or biologics under an
investigational new drug (IND) application.
FDA Acceptance of Foreign Clinical Studies Not
Conducted Under an IND, Frequently Asked Questions, Guidance for Industry and
FDA Staff (PDF - 108KB)
This guidance document is intended to clarify for sponsors and applicants how they can demonstrate compliance with the requirements of 21 CFR 312.120. It provides recommendations for the submission of information, whether in an IND or application for marketing approval for a drug or biological drug product, to demonstrate that a non-IND foreign clinical study was conducted in accordance with GCP.
This guidance document is intended to clarify for sponsors and applicants how they can demonstrate compliance with the requirements of 21 CFR 312.120. It provides recommendations for the submission of information, whether in an IND or application for marketing approval for a drug or biological drug product, to demonstrate that a non-IND foreign clinical study was conducted in accordance with GCP.
This
guidance provides recommendations to sponsors and/or applicants planning to
conduct food-effect bioavailability (BA) and fed bioequivalence (BE) studies
for orally administered drug products as part of investigational new drug
applications (INDs), new drug applications (NDAs) and abbreviated new drug
applications (ANDAs), and supplemental applications.
This
guideline presents guidance on FDA's expectations regarding inclusion of both
genders in drug development.
This
guidance is intended to assist industry with risk management activities for
drug products, including biological drug products, in the Center for Drug
Evaluation and Research (CDER) and the Center for Biologics Evaluation and
Research (CBER).
This
guidance is intended to assist sponsors in deciding whether a study of marketed
drugs or biological products for treating cancer falls within the exemption
under § 312.2(b)(1) (21 CFR 312.2(b)(1)) from the general requirement to submit
an investigational new drug application (IND).
This
guidance is intended to assist industry with risk management activities for
drug products, including biological drug products, in the Center for Drug
Evaluation and Research (CDER) and the Center for Biologics Evaluation and
Research (CBER).
This
guidance discusses issues related to the electronic submission of new drug
applications (NDAs), abbreviated new drug applications (ANDAs), biologics
license applications (BLAs), investigational new drug applications (INDs),
master files, advertising material, and promotional labeling using the
electronic common technical document (eCTD) specifications.
This
guidance is intended to help sponsors and investigators comply with the
requirements for investigational new drug (IND) safety reporting and safety
reporting for bioavailability (BA) and bioequivalence (BE) studies under 21 CFR
312.32, 312.64(b), and 320.31(d)(3). This document provides guidance to
sponsors and investigators on expedited safety reporting requirements for human
drug and biological products that are being investigated under an IND and for
drugs that are the subjects of BA and BE studies that are exempt from the IND
requirements. This guidance defines terms used for safety reporting, makes
recommendations on when and how to submit a safety report, and provides advice
on other safety reporting issues that have arisen from sponsors and
investigators.
This
guidance is intended to help small businesses understand and comply with FDA’s
safety reporting regulations for human drug and biological products that are
being investigated under an investigational new drug application (IND) and for
drugs that are the subjects of bioavailability (BA) and bioequivalence (BE)
studies that are exempt from the IND requirements. The FDA has prepared this
guidance in accordance with section 212 of the Small Business Regulatory
Enforcement Fairness Act (Public Law 104-121).
This
guidance is intended to assist sponsors who will be submitting information to
the Clinical Trials Data Bank. It addresses statutory and procedural issues for
submitting information to the data bank.
This
guidance document is intended to clarify the regulations regarding commercially
distributed analyte specific reagents (ASRs) (21 CFR 809.10(e), 809.30, and
864.4020), and the role and responsibilities of ASR manufacturers. This
document is not intended to provide guidance on the role of clinical
laboratories in the development of laboratory developed tests (LDTs). The
guidance follows the substance, spirit, and meaning of the ASR regulations
already in place.
This
guidance document answers commonly asked questions about Humanitarian Use
Devices (HUDs) and applications for Humanitarian Device Exemption (HDE)
authorized by section 510(m)(2) of the Federal Food, Drug, and Cosmetic Act
(the Act). It also reflects the additional requirements set forth in the
Pediatric Medical Device Safety and Improvement Act of 2007.
This
guidance document is intended to assist applicants in the preparation and
submission of Humanitarian Use Device (HUD) designation requests to the U.S.
Food and Drug Administration’s (FDA or Agency) Office of Orphan Products
Development (OOPD). It is also designed to assist FDA reviewers in their
evaluation and analysis of HUD designation requests ("HUD requests"
or "requests").
This
guidance informs sponsors, institutional review boards (IRBs), clinical investigators,
and agency staff that the FDA intends to exercise enforcement discretion, under
certain circumstances, with respect to its current regulations governing the
requirement for informed consent when human specimens are used for FDA
regulated in vitro diagnostic device investigations.
This
guidance document outlines FDA regulations applicable to studies for
investigational IVD devices, including those regulations related to human
subject protection. The guidance also explains data considerations that
ultimately will affect the quality of the premarket submission. It includes a
glossary, a reference list with related web addresses, and a quick-reference
table.
The
primary purpose of this guidance is to outline general validation principles
that the FDA considers applicable to the validation of medical device software
or the validation of software used to design, develop, or manufacture medical
devices. In addition, it contains useful validation principles applicable
to software used in the conduct of clinical trials.
This
guidance is intended to assist in applying current good manufacturing practice
(CGMP) required under section 501(a)(2)(B) of the Federal Food, Drug, and
Cosmetic Act (FD&C Act) in the manufacture of most investigational new
drugs (IND) used in phase 1 clinical trials. These drugs, which include
biological drugs, are exempt from complying with 21 CFR part 211 under 21 CFR
210.2(c) (referred to as phase 1 investigational drugs).
This
guidance is intended to assist manufacturers in understanding quality system
requirements concerning design controls. Assistance is provided by interpreting
the language of the quality systems requirements and explaining the underlying
concepts in practical terms. As discussed under Device Advice, devices approved under an
investigational device exemption (IDE) are exempt from the Quality System (QS)
regulation, except for the design control requirements under §820.30. However,
the sponsor may state an intention to comply with other parts of the QS
regulation. The extent to which the Quality System regulation will be followed
in manufacturing the device must be documented in the sponsor’s IDE records
[§812.140(b)(4)(v)].
This
guidance provides recommendations to sponsors of investigational new drug
applications (INDs) on the chemistry, manufacturing, and controls (CMC)
information that would be submitted for phase 2 and phase 3 studies conducted
under INDs. This document applies to human drugs (as defined in the Federal
Food, Drug, and Cosmetic Act). It does not apply to botanical drug products,
protein drug products derived from natural sources or produced by the use of
biotechnology, or other biologics. The goals of this document are to (1) ensure
that sufficient data will be submitted to the Agency to assess the safety, as
well as the quality of the proposed clinical studies from the CMC perspective,
(2) expedite the entry of new drug products into the marketplace by clarifying
the type, extent, and reporting of CMC information for phase 2 and phase 3
studies, and (3) facilitate drug discovery and development.
This
guidance provides recommendations to sponsors, contract research organizations,
data management centers, clinical investigators and institutional review boards
regarding the use of computerized systems in clinical investigations.
This
guidance is intended to describe the FDA's current thinking regarding the scope
and application of part 11 of Title 21 of the Code of Federal Regulations;
Electronic Records; Electronic Signatures (21 CFR Part 11).