Download as pdf or txt
Download as pdf or txt
You are on page 1of 78

Essential

Good Clinical
Practice
Professor David Hutchinson
© Professor David Hutchinson, 2017
Published by Canary Ltd, 5 Studley Court Mews, Chobham, Surrey, GU24 8EB, UK
Website: www.canarybooks.com
ISBN 978-1-912055-18-0
EAN 9781912055180
First edition, January 2009; Second edition, November 2017
All rights reserved. No part of this book may be reproduced, stored in a retrieval
system, or transmitted in any form or by any means, electronic, electrostatic,
magnetic tape, mechanical, photocopying, recording or otherwise, without prior
written permission of the publisher.
Artwork by: LIMA Graphics, Fleet, Hampshire, UK
Printed by: Surrey Litho, Great Bookham, Surrey, UK
Essential
Good Clinical
Practice
By Professor David Hutchinson
Contents
Page

Preface 1
Chapter 1: Introduction to good clinical practice 2
Factors affecting clinical trials 2
GCP development 5
Principles of GCP 10
Compliance with GCP 12
Documenting GCP compliance 13

Chapter 2: General GCP responsibilities and subject protection 14


The investigator and study site 14
Addressing the risks of trials 17
Management of investigational medicinal product 20
Product accountability 21
The study protocol 22
Recruiting trial subjects 24
Subject protection 25

Chapter 3: Study conduct and GCP compliance 30


Data collection in clinical trials 30
Safety monitoring, recording and reporting 34
Analysis and reporting of trial data 42
End of study procedures 43
GCP compliance and inspections 45
Due diligence is the key 46

Chapter 4: Essential study documentation 47


Computerized systems 48
Checklists of trial documents 50

Chapter 5: The sponsors’ quality management system 55


Risk-based approach to trial monitoring 57

Quiz 62

Answers to quiz 70
Essential Good Clinical Practice

Preface
Good clinical practice (GCP) is an international quality standard. In order for
clinical trials on medicinal products in humans to achieve international recognition,
they should be conducted in accordance with GCP. In many countries this is a
legal requirement, as GCP principles have been incorporated into national laws.
Adherence to GCP is also a requirement for publication in many international
medical journals. Any reputable clinical trial should therefore be compliant with GCP.
This book is based on the GCP guidelines developed by The International Council
for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use
(ICH GCP E6(R2)). The guidelines (ICH GCP) have become a proven international
standard that is globally recognized and is the guidance of choice for both
commercial and non-commercial trials. The ICH GCP requirements mainly affect the
development of new medicinal products. The principles can, however, be applied
to studies involving marketed products, other healthcare products such as herbal
and consumer medicines, and trials on nutritional products. Different standards
apply to studies involving medical devices.
The book is a revision of the 2009 edition and takes into account the latest GCP
changes. It aims to provide practical guidance and information about some of the
most important GCP responsibilities of sponsors and investigators. It looks at the
development of GCP; provides an overview of the principles of GCP; and examines
in more detail the GCP requirements relating to ethics, subject consent, protocol
compliance, data collection, safety reporting, managing the investigational product,
documentation, trial oversight and quality management.
This book is an ideal companion for those setting up trials (the sponsors) and those
performing trials (the investigators). It is also suitable for those who regulate trials,
such as members of ethics committees.
I hope that you enjoy it and find it useful.

Professor David Hutchinson


1
CHAPTER 1 Essential Good Clinical Practice

Introduction to good
clinical practice
Factors affecting clinical trials
Everyone involved in clinical trials in design and undertake trials as they
humans has a responsibility to ensure wished. A few voluntary codes existed.
that the trials are conducted to the
Today, human clinical research is
highest possible standards.
heavily regulated. A quality standard
Around 35 years ago there were called good clinical practice
few rules and regulations (GCP) should be followed in
affecting clinical trial all trials, wherever they are
conduct. Most sponsors and conducted.
investigators were free to

2
Introduction to good clinical practice

Declaration of Helsinki of participation; requires informed


The most notable code of practice that consent to be obtained; prohibits
underpins medical research in humans coercion by investigators; and allows
is the Declaration of Helsinki. It is a set subjects to withdraw from the trial at
of ethical principles established by the any time without detriment to their
World Medical Association (WMA). future care.
It has been modified on many
There have been many
occasions since it was first
controversial aspects to the
published in 1964, following
Declaration of Helsinki, in
a General Assembly of the
particular the use of placebo
WMA in Helsinki (hence its
(inactive substance) in trials
name).
and the requirement to provide
The Declaration of Helsinki the best proven treatment at
provides protection for subjects the end of a trial. The latest version
who take part in trials. It ensures of the Declaration of Helsinki can be
that subjects are provided with obtained from the WMA website at
information on the risks and benefits <www.wma.net>.

3
Essential Good Clinical Practice

Other regulations and guidelines countries, there are many similar


A process called ICH (International procedures, making pan-European
Council for Harmonisation) has trials easier to perform.
generated guidance documents
The regulations in the USA – published
for virtually every stage of drug
in the Federal Register and referred
development, from non-clinical
to as the Code of Federal Regulations
to human studies.
(CFR) – affect most commercial
Within the European Union (EU) research because, more often than not,
framework – expanded to the trials performed globally will be used
European Economic Area (EEA), to support marketing applications in
with the agreed involvement of the USA.
Norway, Iceland and Liechtenstein –
There are also strict data protection
directives and regulations have been
laws (affecting the transfer and
developed to harmonize procedures
processing of trial subjects’ data).
throughout Europe. These have
been incorporated into the national Inspectors from the regulatory
laws of EEA Member States. Whilst authorities visit sponsors and
there has not been complete investigators to check that the laws
harmonization between European and regulations are being followed.

REMEMBER:
In Europe, directives must be passed into Member States’ laws
within a specified time, but may be ‘gold plated’ to add additional
requirements within that country. Regulations are binding for all
EU Member States.
Europe accounts for about one-third of patients recruited into clinical
trials submitted as part of marketing authorizations. Another third
of the data are generated in the USA, leaving a large proportion
generated outside Europe and North America. For maximum
credibility, these trials should be performed in accordance with
internationally recognized GCP.

4
Introduction to good clinical practice

GCP development
What is GCP? Who does GCP affect?
GCP is a quality standard. It applies to Everyone involved in a clinical trial
every step of a clinical trial: from its has a duty to ensure that the trial is
planning and design through to the performed properly and to the highest
reporting of results. possible standards. Therefore, GCP
affects everyone who is involved in
GCP can be defined as follows: a clinical trial.
“A standard for the design, conduct,
performance, monitoring, auditing, Responsibilities are defined in the GCP
recording, analyses, and reporting of guidelines for
clinical trials that provides assurance • trial sponsors – those that organize,
that the data and reported results are manage and fund clinical trials, such
credible and accurate, and that the as pharmaceutical companies (and
rights, integrity and confidentiality of other commercial organizations) and
the subjects are protected” (ICH GCP non-commercial organizations
guidelines, Section 1.24).
• investigators – physicians or
From this we can summarize that dentists and members of their study
GCP requires teams, such as other doctors, nurses
• procedures to be implemented to and administrators
help ensure that the participants • ethics committees.
of studies – the trial subjects – are
fully protected; the subjects can be
healthy volunteers or patients
• procedures to be carried out so
that the data collected are accurate
and reliable.

5
Essential Good Clinical Practice

Where did GCP start? European GCP


Specific requirements for doctors/ A period of time then ensued where
investigators performing human individual countries developed
clinical trials were first introduced in their own GCP guidelines. This was
the USA in the late 1970s. This set followed in 1990 by the adoption of
the standard for research. The US a harmonized set of European GCP
regulatory authorities then deemed guidelines. These guidelines then had
research conducted outside the USA to to be implemented into the national
be less acceptable. This often meant laws of the EU Member States. Some
that trials that had not been performed Member States achieved this by 1992.
within the USA had to be repeated By 1996, GCP had finally become a
to US standards, before they would legal requirement in all EU Member
be accepted as part of a marketing States. Despite this, studies performed
authorization application. This costly in one country were not always
and time-consuming duplication of acceptable in another country.
research quickly led to other countries
introducing guidelines to improve the Harmonization of procedures
quality and acceptability of their own The EU prompted a meeting to discuss
clinical trials. the wider harmonization of drug
development procedures, so that
one method would be acceptable
to all. The first ICH conference took
place in Brussels in April 1990. The
unique meeting brought together
representatives from the world’s
most active pharmaceutical research
and development regions. It involved
the regulatory authorities and
pharmaceutical industry associations
from Europe, Japan and the USA.

6
Introduction to good clinical practice

The conference has since developed The ICH ‘family’ is now truly global and
into an effective and highly successful representatives of many organizations
process to provide guidance on all have membership of ICH. A list of the
aspects of drug development, known current members is available on the
simply as ICH. The ICH website ICH website.
contains a great deal of useful
The ICH process has produced a great
information about the process,
deal of useful guidance covering all
including its products and plans for
aspects of drug development. This
the future (see <www.ich.org>).
includes guidance relating to the
The ICH Management Committee is quality of medicines (the ‘Q’ guidelines),
made up of the six original members guidance on non-clinical safety studies
(regulators and pharmaceutical (the ‘S’ guidelines), guidance on studies
industry representatives) as well in humans (the ‘E’ guidelines) and
as Health Canada, Swissmedic, the guidance on miscellaneous topics
World Health Organization (WHO) (the ‘M’ guidelines). The E guidelines
and the International Federation of are the most important products for
Pharmaceutical Manufacturers and human clinical trials. A summary of
Associations (IFPMA). The original the ICH products in the E section is
pioneers of the ICH process were: provided overleaf.
• regulators: European Commission
(EU); Japanese Ministry of
Health, Labour and Welfare
(MHLW/PDMA); and the US Food
and Drug Administration (FDA)
• pharmaceutical industry
representatives: European
Federation of Pharmaceutical
Industries and Associations
(EFPIA); Japan Pharmaceutical
Manufacturers Association (JPMA);
and Pharmaceutical Research and
Manufacturers of America (PhRMA).

7
Essential Good Clinical Practice

ICH GCP guidelines


One of the most important
SUMMARY OF ICH products is the ICH GCP
ICH PRODUCTS: guidelines. The guidelines have
E1: Extent of population exposure to assess become the internationally
safety accepted way in which to
E2: Safety reporting (in parts E2A, B, C, D, undertake clinical trials and their
E and F) principles have been incorporated
E3: Clinical study reports into the national laws of many
E4: Dose response information to support countries. By following the ICH
registration GCP guidelines, clinical trials will
E5: Ethnic considerations be accepted by the regulatory
E6: GCP guidelines authorities of all European
E7: Clinical trials in special populations – countries, and in the USA, Japan,
geriatrics Canada, Switzerland and many
E8: General considerations other countries.
E9: Statistical principles The ICH GCP guidelines are
E10: Choice of control group divided into eight sections:
E11: Studies in children 1. Glossary of terms (definitions)
E12: Therapeutic area guidelines 2. Principles of GCP
E14: Clinical evaluation for anti-arrhythmic
drugs 3. Institutional review board/
independent ethics committee
E15, E16, E18: Genomics topics
4. Investigator
E17: Multi-regional clinical trials
5. Sponsor
E19: Safety data collection
6. Clinical trial protocol and
The list of ICH products is always increasing
amendments
and existing guidelines are regularly being
revised. For the most up-to-date information, 7. Investigator’s Brochure
the ICH website <www.ich.org> should be
8. Essential documents.
regularly checked.

8
Introduction to good clinical practice

The ICH GCP requirements broadly Data quality


focus on three main areas: subject Requirements include the careful and
protection, the reliability and integrity diligent collection of data, accurate
of data, and documentation. record form completion, frequent
monitoring of the trial and a series
Subject protection of quality checks to ensure that all
All trials have to be performed in recorded data are reliable.
accordance with a set of ethical
principles laid down in the Declaration Good documentation
of Helsinki. The risks of participation GCP encourages good documentation
in a trial should be carefully evaluated practices. Every step of the trial has
and documented in a formal risk to be documented to prove GCP
assessment. Risks should be compliance. These factors all help
outweighed by the benefits to the to improve the quality of trials and
subject. The informed consent of this helps regulators to make reliable
trial subjects should be obtained. decisions when considering marketing
A favourable ethics committee applications.
opinion is required before the study
commences. Any decisions about
the subjects’ medical care should be
made by a doctor.

9
Essential Good Clinical Practice

Principles of GCP
Following the principles of GCP is a • Before a trial is initiated, foreseeable
legal requirement in most countries. risks and inconveniences should be
In some countries, such as the UK, the weighed against the anticipated
wording in the ICH GCP guidelines has benefit for the individual trial
been slightly amended but the spirit of subject and society. A trial should be
the principles remains the same. The initiated and continued only if the
thirteen principles of ICH GCP are listed anticipated benefits justify the risks.
below (reproduced from Section 2 of
• The rights, safety, and well-being
the ICH GCP guidelines).
of the trial subjects are the most
• Clinical trials should be conducted important considerations and should
in accordance with the ethical prevail over interests of science and
principles that have their origin in society.
the Declaration of Helsinki, and that
• The available nonclinical and clinical
are consistent with GCP and the
information on an investigational
applicable regulatory requirement(s).
product should be adequate to
support the proposed clinical trial.
• Clinical trials should be scientifically
sound, and described in a clear,
detailed protocol.
• A trial should be conducted in
compliance with the protocol that
has received prior institutional
review board (IRB)/independent
ethics committee (IEC) approval/
favourable opinion.

10
Introduction to good clinical practice

• The medical care given to, and


medical decisions made on behalf
of, subjects should always be the
responsibility of a qualified physician
or, when appropriate, of a qualified
dentist.
• Each individual involved in
conducting a trial should be
qualified by education, training,
and experience to perform his or
her respective task(s).
• Freely given informed consent
should be obtained from every • The confidentiality of records that
subject prior to clinical trial could identify subjects should be
participation. protected, respecting the privacy and
confidentiality rules in accordance
• All clinical trial information should with the applicable regulatory
be recorded, handled, and stored requirement(s).
in a way that allows its accurate
reporting, interpretation and • Investigational products should be
verification. This principle applies manufactured, handled, and stored
to all records referenced in this in accordance with applicable good
guideline, irrespective of the type manufacturing practice (GMP). They
of media used. should be used in accordance with
the approved protocol.
• Systems with procedures that
assure the quality of every aspect
of the trial should be implemented.
Aspects of the trial that are essential
to ensure human subject protection
and reliability of trial results should
be the focus of such systems.

11
Essential Good Clinical Practice

Compliance with GCP


For a trial to be globally acceptable Organisations are able to ensure
to regulatory authorities, it is essential that their staff comply with GCP
for sponsors and investigators requirements by establishing a set
performing both commercial and of Standard Operating Procedure
non-commercial trials to (SOPs). These should describe
comply with the principles every procedure undertaken
of GCP. The national laws in a clinical trial. SOPs are
of many countries require relevant for sponsors,
this. In addition, the investigators and ethics
major medical journals committees. They should
will not accept studies for form the working manual for
publication if they have not been those who have to undertake
performed in accordance with GCP. the procedures, and should provide
Inspections are undertaken by the details of exactly how a procedure
regulatory authorities to check GCP must be performed and documented.
compliance. Failure to comply may SOPs should be regularly reviewed and
lead to sanctions being imposed on the updated.
sponsor and/or investigator.
GCP protects subjects and facilitates
Furthermore, for the consideration of the collection of reliable data – it is an
a study for publication in many of the important quality standard.
major medical journals, the editors
require a declaration by the authors
that the study has been performed to
GCP standards.

12
Introduction to good clinical practice

Documenting GCP compliance


Both sponsors and investigators carefully defined and a log maintained
need to demonstrate that they have of the documents kept within it, and
complied with GCP, their SOPs and all their location. All documents stored in
legal requirements. Documenting the TMF must be quickly retrievable
every step of the trial is one way and readily available for audit
to demonstrate this. and inspection.
Documents are generated Many of the documents are
before, during and after common to both study files.
a trial. They are called However, some are exclusive
essential documents. Essential to either the sponsor or the
documents may be paper- investigator site. For example,
based or may be electronic records. documents that identify trial subjects
As the trial progresses, the essential are unique to the investigator site. The
documents are collected together in original copies of documents that do
a study file. There will be a study file not identify subjects are usually kept
at both the investigator and sponsor in the sponsor’s TMF.
sites. At the sponsor site, the study file
Someone should be appointed to
is called the Trial Master File (TMF). The
manage the study files. To ensure that
TMF may be totally paper, completely
documents are not lost or damaged,
electronic or it may be a mixture of both
access to the study files must be
paper and electronic records. Whatever
restricted.
the system used, the TMF must be

REMEMBER:
If it is not documented, it didn’t happen!
The minimum content of the study file at both the sponsor
and investigator sites is listed in Section 8 of the ICH
GCP guidelines. This is not a definitive list and there are
many other documents that make up the TMF. The TMF
documents list is trial-specific.
13
CHAPTER 2 Essential Good Clinical Practice

General GCP responsibilities


and subject protection
The investigator and study site
The success of any clinical trial is It is usual for the principal investigator
down to the selection of a suitable – ie. the person who is responsible for
investigator and study site personnel. the overall conduct of the trial – to
They must be appropriately qualified, delegate duties to other members
trained and have the experience and of the study team. However, it is
expertise to undertake the trial. The important that members of the study
sponsor must carefully consider the team only perform the duties delegated
personal and professional qualities to them. They must be legally allowed
of the investigator. GCP requires the to undertake those duties, which
sponsor to undertake a thorough site must be within their experience and
assessment to ensure that both the capabilities. The sponsor must check
investigator and the proposed trial site compliance with this requirement.
are suitable.

REMEMBER:
The investigator must have time to undertake the study,
sufficient potential trial subjects, and suitable facilities and
supporting staff.
14
General GCP responsibilities and subject protection

Before they take part in a study, all In order to demonstrate that a


staff should be properly trained and person involved in a trial is suitably
briefed. They should be aware of and experienced and has the qualifications
should follow the study protocol, and required, an up-to-date signed and
should know about the medicines dated curriculum vitae (CV) should be
being used in the trial. provided by that person. This should
be kept on file and updated when
The investigator plays a central part in
appropriate. CVs are particularly
a hub of trial activities and processes.
important for team members who
Log of study personnel have significant trial-related duties,
It is important for investigators to keep such as obtaining the consent of
a log of the site personnel involved in subjects, undertaking assessments and
the study. This log should document completing record forms. CVs might not
who was involved, their qualifications, be required for personnel with minor,
when they were involved (start and end supervised roles; however, if in doubt
dates/times, depending on the nature a CV should be kept on file.
of the study) and what their trial-
related duties were.

15
Essential Good Clinical Practice

Oversight of the trial


The investigator is responsible for
supervising any individual or party
who is delegated trial-related
duties and functions. For example,
the delegation of the consent
process to a subinvestigator, the
involvement of a trial pharmacist
to dispense trial medication or the
collection of data by a study nurse
all require appropriate oversight. The
investigator should
• ensure that the person or party is
qualified and experienced enough
to do the task
• make sure the person is named,
and their qualifications and
experience are documented
Equipment requirements
• check that the person is
The study site must have the relevant
performing the duty or function
equipment and facilities in order to
correctly – and document how
perform the trial. Equipment used
they have done this.
in a trial must be suitable, properly
maintained and calibrated; records should
be available to confirm this. For example,
a refrigerator used to store trial samples
should have a temperature alarm in case
of failure; its temperature and condition
should be regularly checked by study
staff (signing a log of activity); and the
refrigerator should ideally be dedicated
to the trial to avoid cross-contamination.
The equipment and facilities must be
available to the trial when required.

16
General GCP responsibilities and subject protection

Addressing the risks of trials


Clinical trials in humans can carry a Disasters in trials are rare. However, all
significant risk for those who take members of the study team should be
part. This was plainly obvious when aware of what to do in an emergency
a first-in-man (Phase I) clinical trial in situation (eg. following accidental
eight healthy volunteers went wrong overdose or the emergence of sudden
in March 2006. Six of the subjects and serious side-effects).
who were taking part were given a
single dose of a biological product (a Indemnity
monoclonal antibody), and suffered European legislation implemented
immediate and unexpected life- into national laws makes it a legal
threatening side-effects. The other two requirement that sponsors of trials
subjects taking placebo witnessed the have insurance that indemnifies the
dreadful events but were unharmed investigator, as well as providing
themselves. Events such as this lead compensation to trial subjects, in the
to the review of trial regulations. event of trial-related injuries.
Another ‘first-in-human’ The investigator would not usually
study in France in 2016 be indemnified in the event of
also ended in tragedy, his/her own negligence (eg.
and this too led to new normal medical procedures
guidelines being issued on going wrong, and perhaps
the conduct of clinical trials failure to comply with the
in humans being given a new protocol requirements). However,
medicine for the first time. indemnity would be provided to cover
The risks of all clinical trials, in any investigators in the event of trial-
phase of a product’s development, and product-related damages.
should be carefully evaluated by the It is important that an adequate
sponsor and investigator, and plans amount of insurance cover is provided
put into place to identify, evaluate by the sponsor to cover damages,
and control risks to both trial subjects particularly for a number of subjects
and the data being collected. Plans suffering problems in the same trial;
should be in place to deal with any this amount should be checked by
unforeseen event. the investigator.

17
Essential Good Clinical Practice

Compensation Both non-commercial research


Procedures should be in place to institutions and pharmaceutical
compensate subjects in the event of companies usually take out insurance
trial-related injuries. Subjects should policies to cover claims arising from
be informed that compensation is clinical trials. The limit of indemnity
available and details provided on covered by the insurance is variable,
how to obtain it. The discussion of and many policies have conditions
compensation with potential subjects attached.
is clearly a sensitive issue and
Numerous pharmaceutical
needs to be managed openly
companies and their
but carefully.
investigator sites agree to
Before embarking on a trial, use standard clinical trial
the investigator should compensation guidelines,
ensure that these protections such as those published
are in place and that by the Association of the
agreements with the sponsor British Pharmaceutical Industry
have been signed. (ABPI). The ABPI guidelines provide
a simple and expeditious procedure
Both indemnity and compensation
for the provision of compensation
arrangements are likely to be reviewed
for injury caused by participation in a
by the ethics committee when an
clinical trial. The guidelines form part
application for an opinion is made.
of commonly used model clinical trial
agreements. Some basic principles for
compensation (based on some of the
ABPI principles) are given below.

18
General GCP responsibilities and subject protection

Compensation should be paid


• regardless of whether the patient is able to prove that the company has been
negligent in relation to research or development of the medicinal product
under trial
• when, on the balance of probabilities, the injury was attributable to the
administration of a medicinal product under trial, or any clinical intervention
or procedure provided for by the protocol that would not have occurred but
for the inclusion of the patient in the trial
• for more serious injury of an enduring and disabling character, and not for
temporary pain or discomfort or less serious or curable complaints.

Even if an adverse reaction causing the part in the trial – this should not
injury was foreseeable or predictable exclude a patient from consideration
– and regardless of the fact that the for compensation.
patient has freely consented to take

19
Essential Good Clinical Practice

Management of investigational medicinal


product
Information and knowledge updated by the sponsor at least once
When a clinical trial involves the every year.
administration of a medicinal product,
The Investigator’s Brochure forms
it is important that all members of
part of the documentation that allows
staff are familiar with the correct and
the investigator to evaluate the risks
appropriate use of that medicine.
and benefits of the trial, when
Information on the trial’s deciding whether or not to
investigational medicinal participate. The Investigator’s
products (IMPs) will be Brochure is also useful
provided by the sponsor in when determining whether
the form of an Investigator’s observed untoward effects
Brochure. This concise, ‘state- are related to the IMP.
of-the-art’ document should be
If the product is already marketed,
read by the investigator and made
information about the medicine can
available to those who need to refer
be found in its Summary of Product
to it. The Investigator’s Brochure is a
Characteristics. This is an approved
compilation of non-clinical and clinical
data sheet.
knowledge on the IMP. It should be

REMEMBER:
The information in the Investigator’s Brochure should be
presented in a concise, simple, objective, balanced and non-
promotional form.
20
General GCP responsibilities and subject protection

Product accountability
An important GCP responsibility at IMPs should be stored securely in
the investigator site is to ensure that suitable conditions, with access by
IMPs are only used to treat subjects authorized persons only. Ideally, trial
in the trial. Every single dose of study medications should be stored in a
medication has to be accounted for in pharmacy, under the supervision of
written documentation. This is a nominated trial pharmacist.
called product accountability. The trial materials should be
Records include kept in a separate area of the
pharmacy (quarantined).
• how much product has
been received from the Sponsors should have
sponsor, and when detailed records of the IMP
manufactured, compliance
• what has been dispensed to
with Good Manufacturing Practice,
the trial subjects, and when
expiry dates, and what has been
• what is returned unused by the supplied to and retrieved from the
subject (and this should reconcile investigator site.
with what is in stock)
Product accountability records aid
• what is returned to the sponsor, compliance evaluations to determine if
and when. the trial subjects have taken their trial
Unused medicines are usually returned medication as required.
to the sponsor for reconciliation and
destruction.

21
Essential Good Clinical Practice

The study protocol


One of the most important documents Compliance with the study protocol
relating to the trial is the study is essential. Violations of any aspect
protocol. This is usually prepared by of the protocol are unwelcome.
the trial sponsor. It describes all the The quality of the trial – and the
procedures that have to be followed performance of the site – will be
by the investigator. measured against the number and
nature of any protocol violations.
The content of the protocol includes
Everyone involved in the trial
• the aims of the study
should be familiar with the
• which subjects are to be protocol and should follow it.
recruited, using inclusion They should ensure that they
and exclusion criteria are working with the current
• how and when the version that includes any
medicines are to be amendments.
administered Appropriate study site staff must
• how and when evaluations are to be know where the protocol is kept and
performed must have access to it. They must refer
to it at all times.
• what to do if adverse events occur
In the undesirable event that a protocol
• how all study data will be collected violation occurs (eg. if an assessment
and analysed. is not undertaken at the right time), it
Protocols may vary in structure and must be documented and brought to
format between different sponsors, the attention of the sponsor. Mistakes
but their content will essentially be do happen in trials, but they must not
the same. They should be prepared in be concealed.
accordance with SOPs and Section 6
of the ICH GCP guidelines (Section 6
contains more information on clinical
trial protocols).

22
General GCP responsibilities and subject protection

Finally, it is not acceptable for In Europe, procedures for the approval


a sponsor of a trial to allow an of substantial amendments or
investigator to enter subjects who do modifications are documented in
not strictly fulfil the entry criteria, or national law. These involve notification
to permit any other deviation from the of both the competent (regulatory)
protocol using waivers. Waivers breach authority and the ethics committee.
ethics and regulatory approval, and The sponsor will help investigators to
may have serious implications for the comply with this process.
statistical integrity of the study.
In the event of an emergency involving
Protocol amendments an immediate hazard, it is possible
It is possible to amend a protocol to amend the protocol to protect trial
during a trial. However, there are strict subjects at risk. However, the correct
procedures for doing this and ad hoc process of informing the sponsor
changes must not be implemented and ethics committee must then
without the proper approvals. be followed.
A protocol amendment must be
agreed by the sponsor and investigator.
Unless it is of a very administrative
nature, it should then be submitted
to the appropriate ethics committee
for evaluation. The amendment
may not be implemented until the
favourable opinion/approval of the
ethics committee has been obtained
in writing.

23
Essential Good Clinical Practice

Recruiting trial subjects


The investigator needs to evaluate the Subject screening or enrolment log
potential recruitment of trial subjects. Information about subjects who have
This estimate should be based on the been screened and entered into the
throughput of subjects who are likely to trial should be recorded in the subject
meet the study entry criteria described screening log or the subject enrolment
in the study protocol. A common error is log. This records subjects’ names
for investigators to base their estimates and their unique study numbers. It
on the total number of subjects with may also include their dates of birth,
the particular condition being studied, health service numbers and dates
forgetting that as many as 50–60% of participation in the trial. Only the
may have to be excluded from the study. investigational site keeps a record of
the subjects’ identities.
The investigator site needs to keep a
record of who is screened for entry to The subject enrolment log is an
the study – this demonstrates that important document, as it is unique to
proper selection is being undertaken the study site and is the main way in
based on the protocol – and who is which a subject can be identified later.
actually entered into the study.

24
General GCP responsibilities and subject protection

Subject protection
Ethics committee opinion Some of the submitted documents
The favourable opinion (‘approval’) of need to be approved by the
an independent ethics committee (IEC) IEC/IRB, whilst others are submitted
(or institutional review board (IRB) as to help the IEC/IRB make its decision.
they are called in some countries) The documents needing specific
is required before the trial can approval are
begin. IECs/IRBs are made up • the final version of the
of medical and non-medical study protocol and any
members. It is their job to subsequent amendments
ensure that the rights, safety
and well-being of the trial • the documentation used
subjects are protected. to provide information to and
obtain the consent of the subject
To obtain an opinion, a comprehensive (and any updates), eg. subject
package of documents and information information sheets and consent
has to be submitted to the IEC/IRB. In forms
some regions, a standard application
• the materials used to recruit study
form is used. The IEC/IRB will then
subjects, such as advertisements
review the application and provide its
and posters.
opinion. In Europe, strict timelines are
placed on the ethical review of clinical The IEC/IRB should review its opinion/
trials. A new Regulation in Europe to approval annually. Sponsors and
be implemented in 2019 will require investigators should ensure that
an electronic application to obtain a this occurs.
single combined regulatory and ethics
approval. In other regions there may be
no timeline established and obtaining
an ethics committee opinion may be a
significant rate-limiting step.

25
Essential Good Clinical Practice

Consent procedures document, but should be presented


Freely given written informed consent in a simple, plain-language format.
is required before any subject
Subjects should be given ample time to
enters the trial and any trial-related
decide about the trial. They must not
assessments are undertaken (including
be coerced into participation. They are
those that would usually be conducted
always free to withdraw from the
as part of normal clinical practice:
study at any time, even though
if the data are recorded for
they initially gave consent to
the trial, they are considered
participate. It is good practice
trial-related!).
to check that the subject
Each prospective subject understands the information
must be provided with before he/she signs the
information verbally and in consent form.
writing using an approved subject
When subjects cannot consent for
information sheet. In some countries
themselves (eg. children), are unable
(eg. the USA), the document may need
to read or write, or are mentally
to bear the stamp of the approving
impaired, special consent procedures
ethics committee.
exist. These are laid down in national
ICH GCP Section 4.8.10 requires the law. In these cases a witness or a legal
information sheet to contain 20 items representative may be required.
of information. The subject information
sheet tends to be quite a long

26
General GCP responsibilities and subject protection

Information to be provided (h) The reasonably expected benefits.


The items of information to be When there is no intended clinical
presented to trial subjects are stated benefit to the subject, the subject
in the ICH GCP guidelines (Section should be made aware of this.
4.8.10), and reproduced below:
(i) The alternative procedure(s) or
(a) That the trial involves research. course(s) of treatment that may
be available to the subject, and
(b) The purpose of the trial.
their important potential benefits
(c) The trial treatment(s) and the and risks.
probability for random assignment
(j) The compensation and/or
to each treatment.
treatment available to the subject
(d) The trial procedures to be followed, in the event of trial-related injury.
including all invasive procedures.
(k) The anticipated prorated
(e) The subject’s responsibilities. payment, if any, to the subject
(f) Those aspects of the trial that for participating in the trial.
are experimental. (l) The anticipated expenses, if any,
(g) The reasonably foreseeable risks or to the subject for participating
inconveniences to the subject and, in the trial.
when applicable, to an embryo, (m) That the subject’s participation in
fetus, or nursing infant. 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.

27
Essential Good Clinical Practice

(n) That the monitor(s), the auditor(s), (o) That records identifying the subject
the IRB/IEC, and the regulatory will be kept confidential and, to the
authority(ies) will be granted direct extent permitted by the applicable
access to the subject’s original laws and/or regulations, will not
medical records for verification be made publicly available. If the
of clinical trial procedures and/ results of the trial are published,
or data, without violating the subject’s identity will remain
the confidentiality of confidential.
the subject, to the
(p) That the subject or the
extent permitted by the
subject’s legally acceptable
applicable laws and
representative will be
regulations and that, by
informed in a timely manner if
signing a written informed
information becomes available
consent form, the subject or
that may be relevant to the
the subject’s legally acceptable
subject’s willingness to continue
representative is authorizing such
participation in the trial.
access.
(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.

28
General GCP responsibilities and subject protection

Other parts of the GCP guidelines In some cases, the investigator may
have consent implications, such as delegate the consent process to an
Section 4.3.3 which recommends that appropriate member of his/her study
a subject’s primary care physician team. However, as with all delegated
is informed about the subject’s trial obligations, the investigator
participation in the trial. retains overall responsibility for the
consent process and should
The process of obtaining
ensure that it is undertaken
properly informed consent
in accordance with GCP and
is a serious matter. It is
any relevant national laws.
not easy to ensure that all
the information is provided Updates to consent forms
verbally. Investigators may If information becomes
choose to use the subject available during the course of
information sheet as a prompt. a clinical trial that might affect the
A key item to note from the list is the subject’s willingness to participate
provision of information in a balanced – for example if a large number of
way about the risks, inconveniences serious adverse reactions become
and benefits of participation. Data evident – the consent form should
protection laws also affect the content be updated and the subject informed
of consent forms, and subjects should of the new information. It might be
consent to their personal data being necessary in these cases to suspend
processed and transferred. subject recruitment until the ethics
committee has reviewed and approved
Signing consent forms the use of the updated information
When consent is obtained, the subject sheet and consent form. The sponsor
signs and dates the consent form. should advise the investigator on how
In addition, the person obtaining best to proceed in these circumstances.
consent must also sign and date
the form, together with any witness
when required. It is not acceptable
for the investigator to date the
subject’s consent.

29
CHAPTER 3 Essential Good Clinical Practice

Study conduct and GCP


compliance
Data collection in clinical trials
Data collected in a clinical trial • attributable – showing who
are recorded in a subject-specific undertook the observation and
document called a case report form recorded it
(CRF). The CRF identifies the subject
• legible – clear and readable
using a unique study number. The
subject’s initials and date of birth • contemporaneous – recorded at the
may be collected, but to preserve same time the observation is made
confidentiality subjects’ names should • original – be careful where data are
never be recorded in the CRF or in any recorded for the first time; copies of
other document that will leave the data are not acceptable unless they
study site (eg. laboratory reports). have been checked and certified
There are some basic rules for the • accurate – diligent observations
collection of data in clinical trials. should be made and recorded
These are referred to in ICH GCP as correctly
the ALCOA-C principles, whereby all
data should be attributable, legible, • complete – missing data should
contemporaneous, original, accurate be avoided, and source documents
and complete: should be complete and record all
data generated.

REMEMBER:
Investigators should make sure that they clearly record
a subject’s participation in a trial in their medical records
(patient files).
30
Study conduct and GCP compliance

This means that:


REMEMBER:
• CRFs must be completed accurately,
Source documents are
fully and legibly the original documents,
• data should be collected and entered
data and records –
in the CRF within the right timescale effectively the first
• the data should be attributable to
practicable place where
a particular subject data are recorded.
• missing data must be avoided
• Source data. All information in
• it should be possible to identify
original records and certified copies
who collected and then entered
of original records of clinical findings,
the data in a CRF.
observations, or other activities
The original pages from CRFs are in a clinical trial necessary for the
usually collected by the sponsor as reconstruction and evaluation of the
the trial progresses. This enables the trial. Source data are contained in
data to be checked and entered into source documents (original records
the trial database in a timely manner. or certified copies).
The investigator should retain a copy of
• Source documents. Original
the data (eg. a copy of the CRF pages).
documents, data and records (eg.
Source documents hospital records, clinical and
Maintaining source documents office charts, laboratory notes,
to support the data recorded memoranda, subjects’ diaries
in trial-specific CRFs is a key or evaluation checklists,
GCP requirement. Data in pharmacy dispensing
documents should be detailed records, recorded data from
enough to allow the trial to be automated instruments, copies
reconstructed from investigator- or transcriptions certified after
site documents. The location of verification as being accurate copies,
the source documents should be microfiches, photographic negatives,
documented. microfilm or magnetic media, X-rays,
subject files, and records kept at the
Source data and source documents are pharmacy, at the laboratories and
defined by ICH GCP (Sections 1.51 and at medico-technical departments
1.52, respectively) as follows. involved in the clinical trial).
31
Essential Good Clinical Practice

The subject’s medical record (or patient When recording data, investigators
file) is often the source document. should think carefully before they
In addition, laboratory result sheets, write the result down. The place they
recordings from an automated write it becomes the source document,
instrument like an ECG and a subject’s and it is this document that study
bedside chart – and even making an monitors, auditors and inspectors
informal note of a blood pressure will wish to see.
on a scrap of paper – are also
If investigators need to change
source documents.
source data in their own
Sometimes the CRF is used records, the original entry
to record data directly. It should not be obscured; if
then becomes the source it is unclear why the change
document. The protocol should has been made, this should
identify any source data that be explained.
will be recorded directly into the CRF.
However, auditors and inspectors much Monitoring and source data
prefer that the source document is verification
something other than the CRF, so that It is a GCP requirement that sponsor’s
data can be recreated if the CRF pages monitors visit the investigator site
are lost or damaged. before, during and after the trial.
At one of the pre-study visits
the monitor will undertake a site
assessment. Once a site is selected, the
monitor will then brief the study team
about the trial. This will be extensive
as every aspect of the protocol, subject
REMEMBER: recruitment, data collection and IMP
management will be discussed.
To minimize further effort,
The monitor will visit the study site
investigators should regularly during the trial. He/she
ensure that any data will check on trial progress and will
recorded in CRFs match identify any areas of concern that the
those in original source investigator team may have. Another
documents. important role of the monitor is to look
at the data recorded in the CRFs.
32
Study conduct and GCP compliance

The monitor will also compare the Regulatory Agency) in writing, within
source documents with the data in 7 days of discovery.
the CRFs. This process is called source
When significant non-compliance
data verification. The monitor will need
with the protocol or GCP is identified,
a period of ‘quiet time’ during his/her
the sponsor should perform a root
visit to undertake this process. The
cause analysis and implement, and
investigator will be asked to explain
document, appropriate corrective and
and resolve any discrepancies between
preventative actions.
the source data and the data recorded
in the CRF. Correcting data in CRFs
If a change is required to data
Breaches of protocol and GCP
already recorded in a CRF, there is a
If a member of the sponsor or
very precise way in which this must
investigator team becomes aware
be undertaken. The sponsor’s trial
of a breach of GCP or a violation
monitors will ensure that this process
of the study protocol, they should
has been properly followed and will
immediately report this to their
require remedies to be made if it
supervisor or the principal investigator.
has not.
The violation should usually be
In order to change data already
documented in the trial file, stating
recorded in a CRF, the investigator
what the violation was, how
simply crosses through the
it was corrected and what
incorrect data with a single
steps were put into place to
line, writes the new data
prevent its recurrence. If an
alongside, and initials
inspection is undertaken by
and dates the change.
the regulatory authorities, the
For example:
inspectors may ask for a list of
protocol violations.
A special situation exists in the UK,
where it is a legal requirement for
sponsors to report serious breaches
4
DRH 15/9/17
of GCP and serious protocol violations
to the UK regulatory authority (the
Medicines and Healthcare products

33
Essential Good Clinical Practice

Where the reason for the change is This process should also be followed by
not obvious it will need to be added, investigators when there is a need to
for example if data are changed a long change critical efficacy or safety data in
time after they were initially recorded. their source documents.
Correcting fluids should not be used to
hide a previous piece of data.

WHY MONITOR?
The purpose of trial monitoring is 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).
(ICH GCP guidelines, Section 5.18.1)

Safety monitoring, recording and reporting


The process of safety monitoring, • E2A – Clinical Safety Data
recording and reporting is called Management: Definitions and
pharmacovigilance. There are strict Standards for Expedited Reporting
regulations that state how safety
• E2B – Clinical Safety Data
information should be reported in
Management: Data Elements for
clinical trials.
Transmission of Individual Case
The ICH E2 guidelines (see Safety Reports (also E2B(M))
<www.ich.org>) provide more detail
• E2C – Clinical Safety Data
on the requirements for safety,
Management: Periodic Safety
for example:
Update Reports for Marketed Drugs

34
Study conduct and GCP compliance

• Addendum to E2C: Periodic Safety


Update Reports for Marketed Drugs Commonly used safety
abbreviations
• E2D – Post-Approval Safety Data
PV pharmacovigilance
Management: Definitions and
Standards for Expedited Reporting AE adverse event
• E2E – Pharmacovigilance Planning ADR adverse drug reaction
• E2F – Development Safety Update AR adverse reaction
Report. SAE serious adverse event
ICH guideline E19 provides information SAR serious adverse reaction
on safety data collection.
SUSAR suspected unexpected
As part of the essential safety serious adverse reaction
monitoring process, investigators
should identify and record any PSUR periodic safety update
untoward medical events that occur report
during the trial. QPPV Pharmacovigilance
Qualified Person
Adverse events
Investigators are asked to record all
adverse events in the CRF, even those subject administered a pharmaceutical
that are expected and unrelated to product and which does not necessarily
treatment. The ICH GCP guidelines have a causal relationship with
(Section 1.2) define adverse events this treatment”.
as “Any untoward medical occurrence An adverse event can therefore be
in a patient or clinical investigation any unfavourable and unintended
sign (including an abnormal laboratory
finding), symptom or disease
associated with the use of an IMP.
The italicized part of the above
REMEMBER: definition is important: it should be
noted that an adverse event does not
The safety of subjects necessarily have to be related to the
in trials is the most investigational product.
important consideration.
35
Essential Good Clinical Practice

When recording an adverse event, • the investigator could ask the


investigators are usually asked to subject if they have had any
determine and record problems, using a ‘non-leading’
question such as, “How have
• details of the nature and timescale
you been since your last visit?”
of the event
or “Has the treatment upset you
• if it has a causal relationship with in any way?”
study medication
• the subject could be asked leading
• the severity of any adverse event questions using an adverse events
• the outcome. checklist.

Adverse events should be followed In addition to general observation, the


up by the investigator until their most commonly used method to identify
resolution, updating the CRF as adverse events is to ask the subject a
required. non-leading question. As different ways
of obtaining information about adverse
There are a number of ways in which events give rise to different frequencies
investigators may obtain information in reporting, it is important that the
from subjects about adverse events: same method is used for each subject
• a trial subject could spontaneously in each trial. This aids the comparison of
volunteer information during a visit adverse event profiles between studies.
The method to be used should be
• the investigator could observe a sign
described in the study protocol.
during the examination of a patient

Examples of adverse events:


• an elderly patient falls in the street and fractures her wrist
• a child who is a known asthma sufferer has an attack soon after taking trial
medicine and is hospitalized for several days
• a patient suffers from a severe headache after taking study medication
• a patient suffers from mild stomach ache for several days and cannot take
study medication
• a patient dies due to an illness unrelated to the trial
• a patient suffers a serious eye injury playing squash, leading to blindness.

36
Study conduct and GCP compliance

Serious adverse events The term “life-threatening” in the


Some adverse events are classified as definition of serious adverse event
serious adverse events (SAEs). These refers to an event in which the patient
have to be reported by the investigator was at risk of death at the time of
immediately to the sponsor – usually the event; it does not refer to an
by phone, e-mail or fax. The protocol event which hypothetically might
describes the procedures to be have caused death if it were
followed. The sponsor should more severe.
have a 24-hour service to
ICH guideline E2A also
deal with SAE reports.
states that, “Medical and
The ICH GCP guidelines and scientific judgement should
the ICH E2A guideline define be exercised in deciding
a serious adverse event or whether expedited reporting
reaction as any untoward medical is appropriate in other situations,
occurrence that at any dose such as important medical events
that may not be immediately life-
• results in death
threatening or result in death or
• is life-threatening hospitalisation but may jeopardise the
• requires in-patient hospitalization patient or may require intervention
or prolongation of existing to prevent one of the other outcomes
hospitalization listed in the definition above. These
should also usually be considered
• results in persistent or significant serious”. These are sometimes
disability/incapacity, or classified as “other significant
• is a congenital anomaly/birth defect. medical events”.
It should be noted that the severity of
an adverse event is not the same as
its seriousness. An event can be severe
but not serious, as it may not fall into
one of the above categories.

37
Essential Good Clinical Practice

Adverse reactions Expedited reporting of serious


When an adverse event is considered adverse reactions
to have some causal relationship – The regulatory authorities are
possibly, probably or definitely – with particularly interested in reports of
the medicinal product, it is then called serious adverse reactions caused by
an adverse reaction. Adverse reactions IMPs. The sponsor has to follow strict
(ARs) are called adverse drug reactions rules in reporting these. When an
(ADRs) in some countries. investigator reports a serious adverse
event to the sponsor, the following
The definition of an adverse reaction
procedure is followed.
in the ICH E2A guidelines, relating to
pre-marketed products, is as follows: • The sponsor will check that the
“all noxious and unintended responses reported event is serious according
to a medicinal product related to any to the definition.
dose should be considered adverse
• The causality of the event (ie.
drug reactions”.
whether it is an adverse reaction)
The phrase “responses to a medicinal will be determined, based on the
product” means that a causal investigator’s assessment and
relationship between a medicinal a further assessment by the
product and an adverse event sponsor’s pharmacovigilance
is at least a reasonable experts. The investigator’s
possibility, ie. the relationship opinion will not be
cannot be ruled out. downgraded; however,
an adverse event might
For marketed medicinal
be upgraded by the sponsor
products, a well-accepted
to an adverse reaction,
definition of an adverse drug
based on previous experiences
reaction in the post-marketing setting
with the product.
is: “A response to a drug which is
noxious and unintended and which
occurs at doses normally used in man
for prophylaxis, diagnosis, or therapy
of disease or for modification of
physiological function”.

38
Study conduct and GCP compliance

• If it is an adverse reaction, the This should be followed by as


sponsor will then assess whether complete a report as possible within
or not the adverse event was 8 additional calendar days. This
expected or unexpected. An report must include an assessment
unexpected reaction is one in which of the importance and implication
the severity or frequency of the of the findings, including relevant
adverse reaction is not consistent previous experience with the same
with the current product or similar medicinal products.
information (eg. as described in
• Serious unexpected
the Investigator’s Brochure).
adverse reactions that
Adverse reactions that are are not fatal or life-
serious and unexpected – threatening must be
in Europe these are called reported as soon as
suspected unexpected serious possible, but no later than
adverse reactions (SUSARs) – must 15 calendar days after first
be expeditiously reported to the knowledge by the sponsor.
appropriate regulatory authority
according to a certain timescale.
• Fatal or life-threatening unexpected
adverse reactions occurring in
clinical investigations qualify for very
rapid reporting. Regulatory agencies
should be notified (eg. by telephone,
fax or in writing) as soon as possible,
but no later than 7 calendar days
after first knowledge by the sponsor.

39
Essential Good Clinical Practice

Managing other safety reports All adverse events are recorded in


Sponsors require an effective the CRFs. All collected safety data
pharmacovigilance system to are evaluated carefully during and
manage safety reports. In Europe, after the trial. At the end of the trial,
the person in charge of the all adverse events – serious or not,
pharmacovigilance system is related to treatment or not –
called a Pharmacovigilance will be summarized, analysed
Qualified Person (QPPV). and reported in the final
There is also a requirement study report.
to have a local safety contact
It is important that everyone
working in conjunction with
in an organization who might
the QPPV.
encounter product safety
In addition to expedited reporting, information – from receptionist
sponsors have to make annual to Chief Executive – recognizes it
safety reports that include a listing and knows how to report it to their
of the serious adverse reactions safety contact. This is an important
and individual case safety reports. requirement of the pharmacovigilance
The QPPV will co-ordinate this activity. system.

40
Study conduct and GCP compliance

Example of a pharmacovigilance decision-making process by the sponsor


A patient with hay fever is taking part in a clinical trial of a new antihistamine
(the IMP). At the Week 3 visit the investigator discovers an abnormality
on an electrocardiogram (ECG) undertaken as part of the trial. The patient
is referred to a cardiologist, who immediately hospitalizes the patient for
3 days. The IMP is stopped and the patient recovers fully (with no further or
permanent ECG abnormalities). The event is not listed in the Investigator’s
Brochure.
• Is the event serious? Yes, because the subject is hospitalized.
• Is it an adverse reaction? Yes, because causality cannot be ruled out
(clue: the subject recovered when the IMP was stopped).
• Is it unexpected? Yes, because it is not listed in the Investigator’s Brochure.
The adverse event can therefore be reclassified as a suspected unexpected
serious adverse reaction (SUSAR). The event has to be reported expeditiously.
• Is the SUSAR immediately life-threatening or fatal? No.
Therefore the SUSAR has to be reported within 15 calendar days.

41
Essential Good Clinical Practice

Analysis and reporting of trial data


Data collected in CRFs are checked by protocol. Analysis and interpretation
the sponsor and any discrepancies or are usually undertaken by a study
missing data are accounted for. biostatistician.
The data are then entered into a A final study report is written – this
validated database prior to analysis. may be a team effort involving the
The database is usually constructed by biostatistician, medical experts, the
the sponsor. It has to be tested prior to investigator and the medical writer
use to ensure that it is fit for purpose. who is preparing the report.
There are important guidelines that
The final report describes the methods
have to be followed relating to the
used, summarizes and tabulates
validation of computerized
the results obtained, and
systems used in clinical trials.
provides interpretations of
These are discussed further
the findings. Listings of all
in Chapter 4.
data collected in the trial
Checks are performed on form part of the final report.
the database before it is
It is very important that no
‘locked’. At this point, no further
unplanned data analyses are
alterations may be made or more
undertaken whilst the study is in
data entered.
progress. These may introduce a
Analyses are then performed. These bias and thus affect the scientific
should be in keeping with the statistical integrity of the study. Even tabulating
analysis plan documented in the study data to look for trends constitutes

REMEMBER:
In a blinded study, the study code is broken when the final
report is written. The investigator should not break any
study blinding unless in an emergency.
42
Study conduct and GCP compliance

an unplanned interim analysis. If an study team – will be used to examine


interim analysis of the data is data as the trial progresses,
planned before the study starts, to determine whether a
this will be taken into account determined outcome has
when estimating the sample been achieved. ICH guideline
size and it will be documented E9 (Statistical Principles
in the statistical analysis for Clinical Trials) is a useful
plan. Often, a Data Monitoring source of further information
Committee (DMC) – consisting of on this subject.
a group of experts independent of the

End of study procedures


The regulatory authority that The sponsor’s documents are collected
authorized the study and the relevant together in the Trial Master File (TMF),
ethics committee(s) should be informed which should be located in a safe and
when the trial has ended. In some secure environment. Access to the
regions (eg. Europe) this has to be done TMF should be restricted to authorized
within specified timescales at the end personnel. It is vital that the TMF is
of a trial (often defined as last subject, well defined as documents will be
last visit). located in many different places.
Documentation is the key to proving
that a study is in compliance with
GCP. Meticulous record keeping is
therefore vital. Essential documents –
see Chapter 4 – should not be stored
where they can be lost or damaged,
and files should always be kept
up-to-date.

43
Essential Good Clinical Practice

Clear indexing and signposting is It is important that trial documents


required so that it is possible to locate are archived in a safe place at the end
trial documents quickly and efficiently of the trial. An index of the documents
when required. All documents held in an archive is essential.
have to be readily available
Although there are guidelines
on request by auditors and
on the length of time for
inspectors.
which trial documents need
After a trial has ended, the to be kept (eg. ICH GCP
sponsor is responsible for guidelines, Sections 5.5.7 –
archiving the TMF. 5.5.12), these vary from region
to region. In practice, a sponsor is
Whilst many documents are
unlikely to destroy trial documentation.
common to both the sponsor and
Sponsors will advise investigators
investigator sites, some are unique to
on the length of time to keep study
the study site (eg. the enrolment log,
documentation. Investigators should
signed consent forms and medical
expect this to be for a considerable
records such as patient files). It is
period, maybe 15 years or longer.
vital to preserve the integrity of these
documents. As storage space at the
investigator site is usually limited, the
sponsor may help the investigator
to ensure that these documents
are safely archived once the study
has ended.

REMEMBER:
A log of all TMF documents should be kept. Know where
each document is located. Clear signposting is required
so that all documents can be readily retrieved and made
available when needed.
44
Study conduct and GCP compliance

GCP compliance and inspections


Audits may be undertaken by sponsors. Inspections are often conducted on
This is to ensure that the sponsor’s a routine cyclical basis. The selection
procedures are being correctly followed of sponsors/ investigators for an
and that the investigator site is inspection may be based on risk
compliant with national laws, GCP assessment (eg. those performing
and the protocol. more trials involving a greater
number of subjects are at
Regulatory authority
greater risk of inspection).
inspections are undertaken to
Unannounced inspections
check compliance.
may also be triggered by
Inspections are frequently complaints or adverse
carried out by the authorities publicity.
in both their own countries and
There may be severe penalties for
in other territories. Inspections are
non-compliance with the regulations.
regularly undertaken by the US Food
For example, inventing trial data will
and Drug Administration, the national
certainly lead to criminal prosecution
competent authorities in Europe which
and possibly a jail sentence. Non-
also undertake inspections on behalf
compliance with GCP will risk the study
of the European Medicines Agency,
data being rejected, as its reliability
and Japan’s regulatory body (the
cannot be ascertained. Investigators
Japanese Ministry of Health, Labour
in the USA can be banned from taking
and Welfare).
part in clinical trials. Entire research
programmes in academic institutions
can be (and have been) closed down
temporarily or permanently.

45
Essential Good Clinical Practice

Due diligence is the key


Due diligence is required when GCP should not be considered a burden.
performing clinical trials. A good rule to Remember, it is in place to protect the
remember is to document every aspect subjects who enter clinical trials and to
of the trial. If it is not written down make sure that the data collected are
and filed away, it didn’t exist! Mistakes useful and reliable.
are allowed, but these must be
documented and procedures should be
put in place to prevent their recurrence.
Everyone involved in a clinical trial has
a duty to be aware of their SOPs and
GCP responsibilities. Compliance will
usually be achieved if the protocol and
the organization’s SOPs are adhered to.

46
CHAPTER 4 Essential GCP

Essential study
documentation
Many of the documents that should • A log of all documents collected,
be part of the Trial Master File have and where they are stored, should
already been discussed. However, be kept.
numerous other documents that are
• Original documents are precious.
not listed in Section 8 of the ICH GCP
Once collected, they should not be
guidelines will also be generated and
removed from the files. Copies can
will form part of the overall study file.
be taken and these can be used as
There are some basic rules for working documents.
managing trial documents.
• Someone must be in charge of the
• A proper system of filing and storage trial files. The nominated person
is required, which might include the should ensure that there is no
use of filing cabinets. If this is the unauthorized access to documents.
case, specific drawers should be All trial files should be kept under
dedicated to the study. Avoid mixing lock and key.
up documentation in the same
filing area.
• Trial documents should not
be left around offices, waiting
rooms, clinics or other working
areas. Vital documents can get
lost and damaged, as well as
read by those who should not
be looking at them!

47
Essential Good Clinical Practice

• The location of the filing area is are kept away from the office, even
important. The documents must be in a different country. Validated
stored securely in an adequate and copies will be acceptable in these
suitable space, free from risk of loss, circumstances.
premature destruction and damage.
• If documents are scanned
For example, thought must be given
or filmed, the copy must be
to how to protect the documents
validated to ensure that it is a true
from fire, floods, pests and other
reproduction of the original. This is
problems of nature. Inspectors
called a ‘certified copy’ and has all
will check that the file location
of the same content, context and
is suitable.
structure as the original document.
• When inspections occur, it is Documents should remain complete
important that the requested and legible throughout the required
documents can be retrieved retention period (ie. they are
efficiently. Sometimes documents enduring).

Computerized systems
The increasing use of computers in Guidelines on the use of computerized
clinical trials means that many of systems in clinical trials have been
the essential documents are in an published by the European Medicines
electronic format. For example, Agency1 and the US Food
data may be entered directly and Drug Administration2.
onto a computer by the Compliance with the current
investigator. The data are requirements helps to ensure
then transferred electronically that any data collected using
to the sponsor’s computerized computerized systems are
system. Laboratory data may validated3.
be sent directly to the sponsor’s
system.

1.
Inspectors in Europe check the compliance of a computerized system against the PIC/S guidance (Good Practices
for Computerised Systems in Regulated GxP Environments (PIC PI 011-3, September 2007)).
2.
The FDA has published guidance entitled ‘Computerized Systems Used in Clinical Investigations’, dated May 2007.
3.
 ll guidance is regularly updated to reflect current thinking, and requirements (such as those referred to above)
A
are changed.

48
Essential study documentation

There is an extensive list of • the system should be able to create


requirements for a computerized a complete record of all entries and
system, including the following: amendments (an ‘audit trail’)
• computerized systems used in trials • for quality auditing purposes, it
should be identified and listed should be possible to obtain clear
printed copies of electronically
• standard operating procedures
stored data
should be in place to cover system
set up, installation and use • stored data should be checked for
accessibility, durability and accuracy
• all critical systems should be
validated and tested. This should • data should be backed-up at regular
be based on a risk assessment with intervals
the key focus on human subject
• back-up data should be stored for as
protection and the reliability of the
long as necessary at a separate and
trial results
secure location.
• clear user manuals and suitable
training should be provided
• equipment should be located in
suitable places, where extraneous
factors cannot interfere with
the system
• data should only be entered or
amended by persons authorized
to do so
• there should be suitable methods for
deterring the unauthorized entry of
data, including the use of keys, pass
cards, personal codes and restricted
access to computer terminals

49
Essential Good Clinical Practice

Checklists of trial documents


Some of the many documents that only. There is no standard document
make up the Trial Master File, as well checklist – every trial is different and
as other essential documents, are will generate its own documents.
listed below. These lists demonstrate Furthermore, as new regulations are
the vast quantity of documentation implemented, new documentation will
that can make up a typical clinical trial. be generated, and new standard forms
are being developed all the time.
The lists below from Section 8 of
ICH GCP are intended as guidance

Pre-study documents
Sponsor Investigator
4 4 Investigator’s Brochure
4 4 Signed protocol and amendments
4 4 Subject information sheet and consent form
4 4 Written instructions for subject
4 4 Advertisement for recruitment
4 4 Financial agreement with investigator
4 Contract research organization (CRO) agreement (if any)
4 4 Insurance statement
4 4 Investigator agreement
4 4 Independent ethics committee (IEC)/institutional review
board (IRB) opinion on final protocol, information sheets,
consent form, recruitment advertisements, etc
4 4 IEC/IRB composition
4 4 Agreement with pharmacy
4 4 Regulatory opinion notification
4 4 CVs of relevant study site staff
4 4 CV of principal investigator
4 4 Reference ranges for laboratory evaluations
4 4 Reference ranges for study tests
4 4 Laboratory procedures and quality assurance (QA)
records

50
Essential study documentation

Sponsor Investigator
4 4 Laboratory certification/accreditation
4 Sample of label (investigational medicinal product; IMP)
4 4 Handling instructions (IMP)
4 4 Shipping records (IMP)
4 (4) Certificate of analysis (IMP)
4 4 Signing off IMP by Qualified Person (QP)
4 4 Decoding procedures
4 Master randomization list (blinded)
4 Pre-trial monitoring report (of site)
4 4 Study initiation report
4 4 Investigator’s study personnel log
4 4 Manufacturing/import authorization for IMPs (if
required)
4 4 Documentation relating to any Data Monitoring
Committee
4 4 Final case report form (CRF)
4 4 Documents relevant to investigational new drug (IND)
applications (eg. Form FDA 1572), if any
4 Statistical analysis plan
4 Blank CRF

During study documents


Sponsor Investigator
4 4 Investigator’s Brochure updates
4 4 Amendments to protocol, subject information sheet,
consent form, CRF updates, advertisements
4 4 IEC/IRB opinion for updated documents
4 4 Documents relating to applications for amendment to
protocol
4 4 Regulatory opinion of amendments
4 4 IEC/IRB notification of continuing review
4 4 CVs of new staff continued

51
Essential Good Clinical Practice

During study documents continued

Sponsor Investigator
4 4 Updates to reference ranges
4 4 Updates to laboratory procedures
4 4 Records of product shipment to investigator
4 (4) Certificates of analysis for new IMPs
4 4 Signing off new IMP by QP
4 Monitoring visit reports
4 4 Log of monitoring visits made to site
4 4 Letters and other records of communication with study
site
4 4 Completed CRF pages (sponsor – original; investigator
– copy)
4 4 Documented changes to CRFs
4 4 Signature sheet (those making CRF entries/corrections)
4 4 Notifications of serious adverse events (investigator to
sponsor)
4 (4) Notifications of serious adverse reactions (sponsor to
authorities)
4 4 Safety information provided to investigators
4 4 Safety updates to competent authorities
4 4 Safety information provided to IEC/IRB
4 4 Annual report to IEC/IRB
4 4 Product accountability at study site
4 4 Record of retained samples and body fluids/tissues
4 4 Updates to investigator’s study personnel log
4 4 Documentation and explanation by investigator of any
protocol deviation
4 4 Communication from monitor/sponsor to investigator on
protocol deviations
4 Code-break envelopes

52
Essential study documentation

Post-study documents

Sponsor Investigator
4 4 Final product accountability at study site
4 4 Documentation relating to product destruction
4 Audit certificate(s)
4 Final study close-out report
4 Treatment allocation and master decoding information
4 4 Final clinical study report
4 4 Notification to authorities of end of trial
4 4 Notifications to IEC/IRB about study end
4 4 List of archived documents and location (archive index/
log)
4 4 List of Standard Operating Procedures and versions used
during trial
4 4 Completed list of sponsor/CRO personnel involved in
study
4 4 Details of any transfer of ownership of study-related
documentation
4 4 Identification of archivist(s)
4 4 Written information to investigator regarding record
retention

Investigator site only


Sponsor Investigator
4 Signed consent forms
4 Source documents, eg. patient files
4 Subject identification list
4 Subject screening log
4 Subject enrolment log
4 Copies of completed CRF pages
4 Documented changes to CRFs (copy)
4 Subject consent to allow viewing by third parties
of patient files continued

53
Essential Good Clinical Practice

Investigator site only continued


Sponsor Investigator
4 Authority of subject to contact primary care physician
4 Final report by investigator to IEC/IRB/regulatory
authority (where required/applicable)
4 Consent of subject to process and transfer personal data

Sponsor general files

Sponsor Investigator
4 List of SOPs and versions used
4 Internal meeting minutes
4 CVs of sponsor trial staff
4 Training records of sponsor trial staff
4 Study audit reports (in QA files not general Trial Master
File)
4 Correspondence with regulatory authorities
4 Meetings with regulatory authorities
4 Scientific advice
4 Documentation relating to inspections
4 Out-of-date/draft versions of trial documentation
(eg. protocol)
4 Master randomization list (unblinded)
4 Trial registration documentation, eg. EudraCT, Clinical
Trial Authorization/IND application
4 Manufacturing and/or import authorization
4 Sponsor’s quality control records

The location of some documents may vary (eg. a certificate of IMP analysis might
not always be copied to the investigator’s file). A sponsor’s SOPs will determine
which documents are kept in sponsor files only. Original CRF pages will be kept at
the study site until collection by the sponsor. Thereafter, the investigator will retain
a copy of the CRF pages.

54
CHAPTER 5 Essential GCP

The sponsors’ quality


management system
A major change to the ICH GCP • identify critical processes to
guidelines in 2016, brought about ensure subject protection and
by the E6(R2) addendum, was the data reliability
introduction of guidance on quality
• identify risks at both the system
management.
and trial level
Sponsors should implement
• evaluate the identified risks
a system to manage quality
throughout all stages of the trial • decide which risks to control, accept
process. The system should focus or reduce and establish predefined
activities on the protection of trial tolerance limits
subjects and the reliability of data. • communicate quality management
This includes the design of efficient activities to those who are affected
protocols, CRFs and other trial by such activities
documents, and trial processes
should avoid unnecessary complexity. • periodically review risk control
A risk-based approach should measures
• describe the quality management
approach and summarize important
deviations from the predefined
tolerance limits, plus any remedial
actions, in the clinical study report.

55
Essential Good Clinical Practice

A documented risk assessment is a When considering the risks in a trial,


fundamental requirement for all trials. many factors have to be considered,
It should identify both the higher including
risk and lower risk areas of
• the personnel
the trial. For the higher risk
areas, thought must be • the vendors
given to how risks can be • the medicinal product
mitigated. These actions
should be documented and • blinding
their effectiveness assessed • safety and adverse events
once the trial is in progress. For lower
• data collection
risk areas, the risks may be deemed
acceptable without the need for • the investigators and sites
further action.
• the consent process
• the size and complexity of the trial
and assessments.

REMEMBER:
Risk assessments are now a vital part of every clinical trial.
These should focus on human subject protection and data
reliability. Risks should be constantly monitored by both the
investigator and sponsor as the trial progresses.
56
The sponsors’ quality management system

Risk-based approach to trial monitoring


A trial must be monitored. Those According to ICH GCP, sponsors should
undertaking trial monitoring must be develop a systematic, prioritized,
competent and qualified by experience risk-based approach to the monitoring
and training; a good monitor will have of clinical trials. This is a flexible
a wide range of skills. The purpose of and varied approach to monitoring
monitoring is to verify that the rights to improve its effectiveness and
and well-being of the subjects who efficiency. The major regulators
participate in a trial are protected. in Europe and the USA encourage
Furthermore, monitoring confirms this modern approach, although
that an enrolled subject exists. regulators in other regions have not
Monitoring also checks the integrity been so accepting of it. Sponsors
of trial data and ensures that the data have therefore been cautious when
are accurate, complete and verifiable adopting risk-based approaches to
from source documents. trial management.
Finally, monitoring checks are made Risk-based monitoring may involve
to determine if the trial has been more targeted monitoring visits to
conducted in accordance with the the investigator site. Greater
regulatory requirements, the emphasis may be placed by
study protocol and trial plan sponsors (or contract research
and, of course, GCP. Effective organizations acting on
monitoring can detect behalf of a sponsor) on
problems early and prevent remote monitoring, such
issues with human subject as via telephone calls and
protection and the integrity of monitoring data centrally using
the data being collected. validated computerized systems.
Co-monitoring is often undertaken The sponsor may choose on-site
as part of the quality management monitoring, a combination of on-site
process relating to trial monitoring and centralized monitoring, or –
oversight. Co-monitoring ensures when it can be justified – centralized
that monitors are performing their monitoring only.
duties correctly and in accordance
with the organization’s SOPs and
monitoring plan.

57
Essential Good Clinical Practice

Data management has a big role Centralized monitoring


to play in risk-based approaches to Centralized monitoring involves
trial monitoring (RBM) and quality the review of accumulating data
management. Together with the and can be used to identify missing
biostatistician, the data manager and inconsistent data and protocol
should therefore have input into the deviations, and in trend and metrics
monitoring plan and the selection of analyses. It has to be performed
appropriate data monitoring tools. in a timely manner by suitably
They should also have input into the qualified and trained persons. Medical
risk assessment, particularly reviewers, trial managers, data
in relation to data integrity managers and biostatisticians
issues. The identification have a significant part to play
of critical data and plans in this.
to protect data integrity
This method of remote
are crucial. Data managers
monitoring can be used to
should also have appropriate
identify the need for site visits
data integrity input into risk
and maximize the effectiveness of
identification, risk evaluation, risk
site management.
control, communication, review and
reporting. Data managers may also The FDA guidance supports risk-based
have a significant impact on blinding approaches to monitoring clinical trials.
methods and the impact of breaches. It argues that the types of monitoring
activities and the extent to which
centralized monitoring practices can be
employed depend on various factors.

58
The sponsors’ quality management system

These include the sponsor’s use of • examine data trends such as the
electronic systems, the sponsor’s range, consistency and variability of
access to subjects’ electronic records, data within and across sites
the timeliness of data entry
• check for systematic or
from paper CRFs, and the
significant errors in data
communication tools
collection and reporting at
available to the sponsor
a site or across sites, or
and study site. Sponsors
potential data manipulation
who plan to use centralized
or data integrity problems
monitoring processes should
ensure that the processes and • analyse site characteristics
expectations for site record keeping, and performance metrics
data entry and reporting are well- • select sites and/or processes for
defined and ensure timely access targeted on-site monitoring.
to clinical trial data and supporting
documentation.
According to ICH GCP E6(R2) Section
5.18.3, centralized monitoring
processes can be used to:
• identify missing data, inconsistent
data, data outliers, unexpected lack
of variability and protocol deviations

Three key monitoring activities:


On site Remote Centralized
• Visit to site • Telephone calls • Data review
• Meetings with • Internet meetings • Statistical review
study team
• E-mail, etc • Variability, trend
• Data verification analysis

59
Essential Good Clinical Practice

Some of the metrics that might be When centralized monitoring is


measured include serious adverse undertaken the activities must be
event reporting, time to CRF documented formally. The reporting
completion, numbers of deviations of activities should be regular
from predefined tolerance limits, – these reports form an
rates of withdrawals and important part of the Trial
rates of eligibility criteria Master File and subsequent
breaches. document archive. The
reports should be detailed
Central monitoring activities
enough to demonstrate
should be undertaken early
that the activities defined in
and well before the end of a
the monitoring plan have been
trial. Anything detected that falls
undertaken. Any deviations from the
outside the predetermined tolerance
plan must be explained and justified.
limits should then be used to trigger a
monitoring visit and perhaps prevent
further protocol violations or data
integrity issues.

REMEMBER:
As risk-based monitoring may involve fewer visits by the
sponsor’s monitor to the investigator site, the investigator
must take on greater responsibility for ensuring that the
study is conducted correctly and must ensure proper
oversight of the trial at all times.
60
The sponsors’ quality management system

Monitoring plan Dealing with identified non-


Whichever methods the sponsor uses compliance
to monitor the trial, the strategy, If non-compliance with the protocol,
methods, responsibilities and SOPs, GCP or the regulatory
requirements for trial monitoring requirements is detected that does
must be documented in a monitoring or may affect subject protection
plan. This plan must be adhered to or the reliability of the trial results,
– compliance with it will be checked the sponsor should perform a root
during a regulatory inspection. In some cause analysis and implement – and
regions (eg. the USA) the monitoring document – appropriate corrective and
plan can be submitted to the regulator preventative actions.
for an opinion on its acceptability.
These activities must be documented
Review of monitoring activities and will form part of the sponsor’s
Reports of both on-site and centralized Trial Master File. All deviations should
monitoring should be maintained by be evaluated and the decision to
the sponsor. These should be reviewed consider them as significant or non-
and followed up on a timely basis to significant should be a transparent,
ensure that activities are in compliance documented process.
with the monitoring plan. A periodic
review of the monitoring plan is
required so that any emerging issues
can be considered and adaptations can
be incorporated, as appropriate.

61
QUIZ Essential Good Clinical Practice

Test your knowledge


1. What is good clinical practice? 4. Which accepted code of practice
describes the investigator’s
A) A quality standard applied
ethical principles applicable in
to trials in order to improve
global clinical trials?
subject protection and the
quality of data collected in A) Declaration of Helsinki
the trial
B) Code of Federal Regulations
B) Best practices in medicine
C) European Directive
and surgery
2003/94/EC
C) An international standard
devised by pharmaceutical 5. What are Standard Operating
companies to ensure that their Procedures (SOPs)?
trials are performed correctly
A) Documented procedures
and systems that describe
2. Who is affected by GCP?
all trial processes to ensure
A) Sponsors, investigators, ethics compliance with regulations
committees and the organization’s policies
B) Only the study sponsor B) The procedures to be followed
by trial monitors when
C) Only the study investigator
assessing the suitability of an
and ethics committee
investigational site
3. What is meant by the term ICH? C) Guidelines to be followed
by a study nurse at the
A) International Clinical Hospital
investigational site
group
B) International Clinical
Harmonisation of trials
C) International Council for
Harmonisation

62
Quiz

6. What impact does GCP 9. True or false?


implementation have on clinical All clinical trial information
trials? Choose 3 answers. should be recorded, handled and
stored in a way that allows its
A) Facilitates better subject
accurate reporting, interpretation
protection
and verification irrespective of
B) Facilitates collection of the media used.
credible data
C) Facilitates better study 10. In addition to following
documentation GCP guidelines, what other
requirements should a study
D) Requires annual inspections sponsor always adhere to?
and audits Choose 2 options.
E) Facilitates better standards in A) N
 ational legislation of the
non-clinical studies country where the trial is being
carried out
7. True or false?
Available non-clinical and clinical B) Sponsor’s own Standard
information should be adequate Operating Procedures (SOPs)
to support the proposed clinical C) Only EU Directives and
trial. Regulations relating to
clinical trials
8. True or false?
D) Only FDA guidance documents
The rights, safety and well-being
of the trial subjects are the most
important considerations but
need not prevail over interests
of science and society.

63 Answers on page 70.


Essential Good Clinical Practice

11. According to GCP, how should 14. How does GCP require the
the investigator show that informed consent of a trial
he/she has the appropriate subject to be obtained?
qualifications and experience
A) Obtain written informed
to do the study?
consent from each subject
A) Providing an up-to-date, before they undertake any
signed and dated CV study-related procedures
B) Providing a copy of the B) Obtain verbal consent as long
certificate of medical as this is undertaken before the
registration study starts
C) Providing the CV used to obtain C) O
 btain consent once it has
the current position been determined that the
subject is suitable for the study
12. True or false? after undertaking baseline
Trial sponsors are required to assessments
have insurance to cover the
compensation of trial subjects 15. Which of the following are
in the event of product-related required to ensure consent is
damage and also to indemnify correctly documented?
investigators. Select 2 answers.
A) Consent form is signed and
13. How should a researcher become dated by the person obtaining
familiar with the investigational consent
product to be used in a clinical
trial? B) Consent form is signed by
the subject but dated by the
A) U
 ndertaking a self-designed investigator
pilot study in a few patients
prior to the study C) Consent form is signed and
dated by the subject
B) O
 btaining and reading the up-
to-date Investigator’s Brochure D) A witness is always required to
from the sponsor of the trial sign the consent form
C) R
 eading literature relevant to
that therapeutic area

64
Quiz

16. True or false? 18. According to ICH GCP, which


A log identifying subjects items require the ‘favourable
recruited to the study should opinion’ (‘approval’) of an ethics
be kept by the investigator and committee? Select 3 answers.
a photocopy of this should be
A) Protocol and any subsequent
handed to the sponsor/monitor
amendments
at the end of the trial?
B) Consent form and subject
17. A patient is considered by the information sheet
investigator to be suitable for C) Investigator’s Brochure
entry into the study. However
the patient is 6 months too old, D) Case report form
according to the exclusion criteria E) Recruitment methods
in the protocol. What should the (eg. advertisements, posters)
investigator do in order to fully
comply with GCP and regulatory
19. Select 3 options that apply
requirements?
to the use of equipment in
A) T he investigator may obtain a clinical trial.
permission from the sponsor to
A) It should be calibrated and
enter the patient
the results documented
B) T he investigator should not
B) It should be purchased
enter the patient as it would be
specifically for the trial
a protocol violation
C) The equipment should
C) T he investigator can amend the
be regularly maintained
protocol and then inform the
and documented in a
sponsor
maintenance log
D) The equipment should be
available for use when it is
required for trial purposes
E) N
 ormal health service
equipment may be used
without calibration or
maintenance

65 Answers on page 70.


Essential Good Clinical Practice

20.  Whilst collecting samples as part 22. True or false?


of a clinical trial, a subject faints. The filing cabinet containing the
The investigator is about to take essential documents for the trial
the timed 1-hour blood sample should be left unlocked, and in an
from the subject. The event area that allows general access.
means that the sample will now
be taken 20 minutes late. What 23. True or false?
should happen? The names of subjects should not
A) T ake the sample and label it as be recorded in case report forms
a 1-hour sample, making no to preserve confidentiality.
file notes
24. When completing case report
B) T ake the sample and make a forms, investigators should
file note that it was collected ensure that .... Select 3 answers.
late, giving the reason why
A) There are no missing data
C) M
 iss the sample completely
and take the next scheduled B) Data are recorded accurately
sample, making no file notes C) Data are legible
D) Incorrect data are hidden using
21. Which essential documents are
correcting fluids
only kept by the study site with
no copy retained by the sponsor? E) A
 ll data are entered at the end
Select 3 answers. of the study
A) Signed consent forms
25. What best describes the term
B) Subject identification log “source document”?
C) F inal signed version of protocol A) The place where data are
D) Patient or subject files recorded in the first practicably
retainable place
E) Product accountability records
B) Always the case report form
C) Always the patient file

66
Quiz

26. How should data be changed in a 28. Which of the following would be
case report form? classified as a serious adverse
event?
A) C
 ross out the original entry
with a single line, enter the A) Severe headache experienced
new data alongside, date and in the no treatment run-in to
initial the change the study, painkillers prescribed
B) E
 nsure that the original data B) Dizziness thought by the
are completely covered, enter investigator to be possibly
the new data alongside, date related to study medication;
and initial the change subject managed at home
C) W
 ait for the sponsor’s monitor C) S ickness in a patient taking
to review the case report forms placebo, hospitalized for 2 days
and then ask the monitor to for rehydration therapy
make the necessary change
D) Chest pain considered by the
investigator to be related to
27. What best describes an adverse trial medication, treated in an
event? emergency room but released
A) A
 n unwanted medical from hospital after 2 hours
occurrence in a trial subject
that may or may not be caused
by the study medication
B) A
 n unwanted medical
occurrence in a trial subject
that is caused by the study
medication
C) A
 ny event that leads to the
subject being hospitalized,
probably caused by the
study medication

67 Answers on page 70.


Essential Good Clinical Practice

29. Which of the following 31. What is product accountability?


relationships to trial medication
A) The process of accounting
would mean that an adverse
for every dose of medication
event is upgraded to an adverse
in a clinical trial from its
reaction? More than one answer!
manufacture to destruction
A) Definitely related to treatment
B) Tracking the amount of
B) Causal relationship cannot be
investigational product
ruled out
delivered to the investigator
C) Considered unrelated to site
treatment by both the
investigator and sponsor C) Records
 that indicate that
the investigational product
D) Considered unrelated to
has been manufactured
treatment by the sponsor but
in accordance with Good
possibly related to treatment
Manufacturing Practice
by the investigator
32. True or false?
30. Which of the following would be
Investigational medicinal product
classified as adverse reactions?
can still be used to treat the
Select 2 answers.
investigator’s patients who do
A) S evere headache experienced not fulfil trial entry criteria as
in the no treatment run-in to long as a case report form is
the study, painkillers prescribed completed.
B) D izziness thought by the
investigator to be possibly
related to study medication;
subject managed at home
C) S ickness in a patient taking
placebo, hospitalized for 2 days
for rehydration therapy
D) Chest pain considered by the
investigator to be related to
trial medication, treated in an
emergency room but released
from hospital after 2 hours
68
Study conduct and GCP compliance
Quiz

33. Where should all specific trial- 34. What are the potential
related documentation be kept? consequences of failing to
adhere to good clinical practice
A) F or the required period of time
guidelines when performing a
in a Trial Master File that is
clinical trial on an investigational
secure with limited access
product? Select 2 answers.
B) F or the required period of
A) The trial results could be
time in a Trial Master File
rejected and those involved in
with unrestricted access to
the trial may face sanctions
investigators and monitors
B) Nothing at all; GCP is optional
C) In a secure third-party storage
with no legal requirement to
facility with access controlled
comply
by the investigator
C) T his may trigger an inspection
by the regulatory authorities
D) In all cases the investigator
and sponsor will face criminal
prosecution with no chance
of remedy

69 Answers on page 70.


ANSWERS Essential Good Clinical Practice

Answers to quiz
1. A 13. B 25. A
2. A 14.  A 26.  A
3. C 15. A, C 27.  A
4.  A 16. False 28. C
5. A 17.  B 29. A, B, D
6.  , B, C
A 18. A, B, E 30. B, D
7. True 19. A, C, D 31.  A
8. False 20.  B 32. False
9. True 21. A, B, D 33.  A
10. A, B 22. False 34. A, C
11. A 23. True
12. True 24. A, B, C

70
Good Clinical Practice (GCP) is a global quality process that is applied to the
conduct of all clinical trials on new medicinal products. This book aims to
explain the key areas of GCP to help both the non-technical and technical
undertake their roles more effectively. It is based on the internationally
accepted GCP guidelines of the International Council for Harmonisation
(ICH) and encompasses the changes made in 2016 in version E6(R2).

Professor David Hutchinson is an internationally renowned teacher and writer


specializing in clinical research and GCP. He spent many years as an organizer
and manager of clinical trials on new drugs before, during and after the ‘birth’ of
GCP. His books include the best-selling 12 Golden GCP Rules for Investigators,
as well as 15 Golden IND/GCP Rules for Investigators and10 Golden Rules for
Pharmacists. He has taught GCP to personnel from more than 50 countries
worldwide, including investigators, research nurses, physicians and research
associates in the pharmaceutical industry, university students, regulatory
authorities and members of national ethics committees. He is currently Editor
and Publisher of Clinical Research and Quality Assurance Advisor.

www.canarybooks.com

You might also like