The GVP For Arab Countries v3 12 2015
The GVP For Arab Countries v3 12 2015
The GVP For Arab Countries v3 12 2015
Guideline on good
pharmacovigilance
practices (GVP)
For Arab Countries
Version 3
Guideline
On Good Pharmacovigilance Practice
For Arab Countries
Clarification about
Structure of GVP
timelines
Module I
I.B.12. Monitoring the performance &
Addition of minor
effectiveness of PV system and its quality
missing text
system
Module II
Module V
Editorial improvements throughout the Module without impact on its
content
Amended definition of
V.B.1. Terminology Risk minimisation
activity
V.B.1. Terminology Amended definition of
V.B.8.9. RMP module SVIII ―Summary of Missing information &
the safety concerns‖ safety concern
Effective
Version Version
No. date
Change section Changes date for
changes
V.B.12. (& its sub-sections ) RMP part VI
Amended sub-section of
―Summary of activities in the RMP by
the RMP part VI
medicinal product‖
V.C.3. Situations when a risk management
New situations added
plan should be submitted
Regarding the updates of
V.C.4. and V.C.5.
RMP & their submission
Module X
Annex II templates
Module II
Effective
Version Version
No. date
Change section Changes date for
changes
II.B.4.1. PSMF section on QPPV
More details provided
II.C.3.2.1. National PSSF section on LSR
Table II.1 Conditions to submit the More clarification
PSMF and the national PSSF provided
Module XI , Module XII, Module XIV, P II
Change in the expected release date
P.I: Vaccines for prophylaxis against Newly added part
infectious diseases
Clarification provided in
part III & part IV
Annex II.2. Template of the Risk regarding the situations
Management Plan (RMP) for Generics when these parts are
needed
Addition of new
Annex III Other Pharmacovigilance guidance & amendments
guidance of some links
Annex IV ICH guidelines for Amendments of some
Pharmacovigilance links
Preface
Pharmacovigilance has been defined by the World Health Organization as the science and
activities relating to the detection, assessment, understanding and prevention of adverse
effects or any other medicine-related problem. This Guideline of the Arab Countries has
been developed to bring guidance on the requirements, procedures, roles and activities in the
field of human Pharmacovigilance, for Marketing Authorization Holders (MAHs) of
medicinal products for human use in the Arab Countries.
This guidance describes the respective obligations of the MAH to set up a system for
Pharmacovigilance in order to collect, collate and evaluate information about suspected
adverse reactions. All relevant information should be shared between medicines authority in
Arab Countries and the MAH, in order to allow all parties involved in pharmacovigilance
activities to assume their obligations and responsibilities.
The ultimate goal is to ensure that the MAHs are fulfilling their principal role in the safety
monitoring of their medical products for human use, hence enhance efforts in ensuring that
safe, efficacious, and quality medicines are made available for all patients in the Arab
Countries.
With the strategic objectives "to not reinvent the wheel" and "to keep up –moreover-
harmonise with the new development in pharmacovigilance practices & regulations"; this
guideline is greatly adopted from the European Good Pharmacovigilance Practices (EU
GVP) which considered the most compatible ICH pharmacovigilance guideline thus the
most widely applied pharmacovigilance practices in the developed European Countries.
The adoption of the EU GVP as a base for this guideline does NOT undermine the right of a
national medicines authority (NMA) in the Arab Countries to have additional or sometimes
changed requirements. Multinational marketing authorization holders should be
attentive to these national requirements and bring the attention of their headquarters
to them, consequently, take the necessary measure to comply.
This ''Good Pharmacovigilance Practice for Arab Countries'' (GVP- Arab) has been made to
"harmonise the pharmacovigilance practices & regulations in-between the Arab Countries",
though, it is understood that Arab Countries may have different healthcare and regulatory
systems especially with regard to pharmacovigilance. Accordingly, each national medicines
authority in the Arab Countries should consider this guideline as an ''ideal model'' which
they try to adopt as much as they can on their national level whether at the time being or
planned for the near future.
Each national medicines authority in the Arab Countries needs to decide on the following, as
applicable on the national level:
It should be noted that, as with all guidance documents in rapidly evolving technical areas,
that this guidance is intended to be regularly reviewed and updated.
Contributors
In order to cope with these changes and to unify guidelines and performance across the Arab
world, Arab ministers of health came to a common decree (number 7) in their 37th regular
meeting in March 2012. Under the umbrella of the Arab League ‗The Higher Technical
Committee for Medicines‘ was established with representatives from all Arab countries, to
create common Arab guidelines in pharmacovigilance, and in bioequivalence.
This committee elected Dr. Amr Saad, head of the Egyptian centre, to lead the committee
across all its rounds. The committee has finished the final drafts of the two common
guidelines which were submitted to the 38th regular ministers meeting, and which has been
approved by them.
The new guidelines is mainly adapted from the newly-established international Good
Pharmacovigilance Practice, composed of 16 different modules together with some
product/population specific considerations, as well as annexes and templates of submission.
The Guidelines were published in March 2014 and the effective date will be 1st July 2015.
It is expected that these guidelines will significantly influence pharmacovigilance practice
in general in the whole Arab world, and will increase such activities including reporting
rates and signal detection in that part of the world. It will also help some Arab countries to
develop in the area of ‗Regulatory Pharmacovigilance‘.
The contributors from the National Medicines Authorities in the Arab Countries arranged in
alphabetical order:
Country Name
Country Name
Table of Content
Versions change control ............................................................................................ 3
Preface………….. .................................................................................................. …6
Contributors….. ........................................................................................................ 8
Table of Content ...................................................................................................... 10
Introductory note .................................................................................................... 28
Objectives of Pharmacovigilance .................................................................................................... 28
Structure of GVP ............................................................................................................................. 28
Format & layout general requirements ............................................................................................ 29
GVP: Modules.......................................................................................................... 30
Module I– Pharmacovigilance systems and their quality systems ..................... 30
I.A Introduction ............................................................................................................................... 31
I.B. Structures and processes ........................................................................................................... 31
I.B.1. Pharmacovigilance system ...................................................................................................................31
I.B.2. Quality, quality objectives, quality requirements and quality system .................................................31
I.B.3. Quality cycle ........................................................................................................................................32
I.B.4. Overall quality objectives for pharmacovigilance ...............................................................................32
I.B.5. Principles for good pharmacovigilance practices ................................................................................32
I.B.6. Responsibilities for the quality system within an organization ...........................................................33
I.B.7. Training of personnel for pharmacovigilance ......................................................................................34
I.B.8. Facilities and equipment for pharmacovigilance .................................................................................35
I.B.9. Specific quality system procedures and processes ...............................................................................35
I.B.9.1. Compliance management by marketing authorisation holders ...........................................35
I.B.9.2. Compliance management by national medicines authorities..............................................36
I.B.10. Record management ..........................................................................................................................36
I.B.11. Documentation of the quality system ................................................................................................37
I.B.11.1. Additional quality system documentation by marketing authorisation holders ...............38
I.B.11.2. Critical pharmacovigilance processes and business continuity ........................................38
I.B.12. Monitoring of the performance and effectiveness of the pharmacovigilance system and its quality
system ..........................................................................................................................................39
I.B.13. Preparedness planning for pharmacovigilance in public health emergencies ....................................40
I.C. Operation of Pharmacovigilance systems in Arab Countries ................................................... 41
I.C.1. Overall pharmacovigilance responsibilities of the applicant and marketing authorisation holder in the
Arab Countries ............................................................................................................................41
I.C.1.1. Responsibilities of the marketing authorisation holder in relation to the qualified person
responsible for pharmacovigilance in the Arab Country concerned .............................41
I.C.1.2. Qualifications of the qualified person responsible for pharmacovigilance in the Arab
Country concerned ........................................................................................................43
I.C.1.3. Role of the qualified person responsible for pharmacovigilance in the Arab Country
concerned ......................................................................................................................44
I.C.1.4. Specific quality system processes of the marketing authorisation holder in the Arab Country
concerned ......................................................................................................................46
I.C.1.5. Quality system requirements for pharmacovigilance tasks subcontracted by the marketing
authorisation holder ......................................................................................................47
I.C.2. Overall pharmacovigilance responsibilities within each of the Arab Countries ..................................47
I.C.2.1. Role of the national medicines authorities .........................................................................48
I.C.2.2. Role of the national Pharmacovigilance Advisory Committee ..........................................48
I.C.2.3. Specific quality system processes of the quality systems of medicines authorities in Arab
Countries .......................................................................................................................48
I.C.3. Preparedness planning in the Arab Countries for pharmacovigilance in public health emergencies ..49
V.B.8.7.2. RMP module SVII section “Recent study reports with implications for safety concerns”
..............................................................................................................................134
V.B.8.7.3. RMP module SVII section “Details of important identified and potential risks from
clinical development and post-authorisation experience” ...................................134
V.B.8.7.4. RMP module SVII section “Identified and potential interactions including food-drug,
drug-drug interactions and drug-herbal interactions”.........................................136
V.B.8.7.5. RMP module SVII section “Pharmacological class effects” ...................................137
V.B.8.8. RMP module SVII ―Identified and potential risks (ATMP version)‖.............................137
V.B.8.8.1. RMP module SVII section “Newly identified safety concerns (ATMP)” ..................137
V.B.8.8.2. RMP module SVII section “Recent study reports with implications for safety concerns
(ATMP)” ...............................................................................................................137
V.B.8.8.3. RMP module SVII section “Details of important identified and potential risks (ATMP)”
..............................................................................................................................137
V.B.8.9. RMP module SVIII ―Summary of the safety concerns‖ .................................................139
V.B.9. RMP Part III ―Pharmacovigilance plan‖ ..........................................................................................140
V.B.9.1. RMP part III section ―Routine pharmacovigilance activities‖ ........................................141
V.B.9.2. RMP part III section ―Additional pharmacovigilance activities‖ ...................................142
V.B.9.2.1. Particular situations with post authorisation safety studies.....................................144
V.B.9.3. RMP part III section ―Action plans for safety concerns with additional pharmacovigilance
requirements‖ ..............................................................................................................145
V.B.9.4. RMP part III section ―Summary table of additional pharmacovigilance activities‖ .......145
V.B.10. RMP part IV ―Plans for post-authorisation efficacy studies‖ ........................................................147
V.B.10.1. RMP part IV section ―Summary of existing efficacy data‖ ..........................................147
V.B.10.2 Tables of post-authorisation efficacy studies .................................................................148
V.B.11. RMP Part V ―Risk minimisation measures‖ ..................................................................................149
V.B.11.1. RMP part V section ―Routine risk minimisation‖ ........................................................149
V.B.11.2. RMP part V section ―Additional risk minimisation activities‖.....................................152
V.B.11.3. Format of risk minimisation plan(s)..............................................................................153
V.B.11.4. RMP part V section ―Evaluation of the effectiveness of risk minimisation activities‖ 153
V.B.11.5. RMP part V section ―Summary of risk minimisation measures‖ .................................154
V.B.12. RMP part VI ―Summary of activities in the risk management plan by medicinal product‖ ..........154
V.B.12.1. RMP part VI section ―format and content of the summary of the RMP‖ .....................155
V.B.12.2. RMP part VI section ―Summary of safety concerns‖ ...................................................155
V.B.12.3. RMP part VI section ―Summary of risk minimisation measures by safety concern‖ ...156
V.B.12.4. RMP part VI section ―Planned post-authorisation development plan‖ .........................156
V.B.12.5. RMP part VI section ―Summary of changes to the risk management plan over time‖ .157
V.B.13. RMP part VII ―Annexes to the risk management‖ .........................................................................157
V.B.14. The relationship between the risk management plan and the periodic safety update report ..........158
V.B.14.1. Common modules between periodic safety update report and risk management plan .159
V.B.15. Principles for assessment of risk management plans .....................................................................159
X.C.2. Criteria for defining the initial time period of maintenance in the additional monitoring list ..........334
X.C.2.1. Mandatory scope .............................................................................................................334
X.C.2.2. Optional scope.................................................................................................................334
X.C.3. Roles and responsibilities .................................................................................................................334
X.C.3.1. National medicines authority ..........................................................................................334
X.C.3.2. The Marketing authorisation holder ................................................................................335
X.C.4. Creation and maintenance of the list ................................................................................................335
X.C.5. Black symbol and explanatory statements ........................................................................................335
X.C.6. Transparency ....................................................................................................................................336
Introductory note
Objectives of Pharmacovigilance
Pharmacovigilance has been defined by the World Health Organization (WHO) as the science and
activities relating to the detection, assessment, understanding and prevention of adverse effects or
any other medicine-related problem.
In line with this general definition, underlying objectives of the applicable legislation for
pharmacovigilance are:
preventing harm from adverse reactions in humans arising from the use of authorised medicinal
products within or outside the terms of marketing authorisation or from occupational exposure;
and
promoting the safe and effective use of medicinal products, in particular through providing
timely information about the safety of medicinal products to patients, healthcare professionals
and the public.
Pharmacovigilance is therefore an activity contributing to the protection of patients‘ and public
health.
Structure of GVP
Pharmacovigilance activities are organised by distinct but connected processes, and each GVP
Module presents one major pharmacovigilance process. In addition, GVP provides guidance on the
conduct of pharmacovigilance for specific product types or specific populations (P parts) in which
medicines are used. These GVP Considerations apply in conjunction with the process-related
guidance in the Modules.
While the development of GVP is ongoing, some other guidelines developed by the European
Medicines Agency (EMA) under their previous EU regulations remain valid in principle (unless any
aspect is not compatible with this guideline); they are acknowledged –from scientific aspects- in the
Arab Countries. In addition, those other guidelines may be revised at a later point in time for
inclusion in GVP for Arab Countries; they are included under GVP Annex III.
Hence the GVP guideline constitutes mainly of three main building blocks:
GVP Modules
GVP P-parts
GVP Annexes
Section B gives guidance which reflects scientific and regulatory approaches, formats and
standards agreed internationally in various for a; or, where such formal agreements or expert
consensus do not exist, Section B describes approaches which are considered in line with general
current thinking in the field.
Section C focuses on the specifics of applying the approaches, formats and standards in the Arab
Countries and other aspects of operating the respective process in the Arab Countries.
In the context of this document, any stated timeline is in calendar days unless otherwise clearly
stated.
GVP: Modules
I.A Introduction
This Module contains guidance for the establishment and maintenance of quality assured
pharmacovigilance systems for marketing authorisation holders and medicines authorities. How
the systems of these organisations interact while undertaking specific pharmacovigilance
processes is described in each respective Module of GVP.
By following the overall quality objectives in I.B.4. and the guiding principle in I.B.5. to meet the
needs of patients, healthcare professionals and the public in relation to the safety of medicines, the
application of the quality system should be adapted to how crucial each pharmacovigilance task is
for fulfilling the quality objectives for each medicinal product covered by a quality system.
In this Module, all applicable legal requirements are usually identifiable by the modal verb
―shall‖. Guidance for the implementation of legal requirements is provided using the modal verb
―should‖.
A pharmacovigilance system, like any system, is characterised by its structures, processes and
outcomes. For each specific pharmacovigilance process, including its necessary structures, a
dedicated Module is included in GVP.
In general terms, quality is a matter of degree and can be measured. Measuring if the required
degree of quality has been achieved necessitates pre-defined quality requirements. Quality
requirements are those characteristics of a system that are likely to produce the desired outcome,
quality objectives. The overall quality objectives for pharmacovigilance systems are provided
I.B.4.
Specific quality objectives and quality requirements for the specific structures and processes of the
pharmacovigilance systems are provided in each Module of GVP as appropriate.
The quality system is part of the pharmacovigilance system and consists of its own structures and
processes. It shall cover organisational structure, responsibilities, procedures, processes and
resources of the pharmacovigilance system as well as appropriate resource management,
compliance management and record management.
quality planning: establishing structures and planning integrated and consistent processes;
quality adherence: carrying out tasks and responsibilities in accordance with quality
requirements;
quality control and assurance: monitoring and evaluating how effectively the structures and
processes have been established and how effectively the processes are being carried out; and
quality improvements: correcting and improving the structures and processes where necessary.
complying with the legal requirements for pharmacovigilance tasks and responsibilities;
preventing harm from adverse reactions in humans arising from the use of authorised medicinal
products within or outside the terms of marketing authorisation or from occupational exposure;
promoting the safe and effective use of medicinal products, in particular through providing
timely information about the safety of medicinal products to patients, healthcare professionals
and the public; and
contributing to the protection of patients‘ and public health.
The needs of patients, healthcare professionals and the public in relation to the safety of
non-adherence to the requirements of the quality and pharmacovigilance systems and taking
corrective, preventive and escalation action as necessary;
ensuring that audits are performed (see I.B.12.).
In relation to the management responsibilities described above, upper management within an
organisation should provide leadership through:
motivating all staff members, based on shared values, trust and freedom to speak and act with
responsibility and through recognition of staff members‘ contributions within the organisation;
and
assigning roles, responsibilities and authorities to staff members according to their
competencies and communicating and implementing these throughout the organisation.
All personnel involved in the performance of pharmacovigilance activities shall receive initial and
continued training. For marketing authorisation holders, this training shall relate to the roles and
responsibilities of the personnel.
The organisation shall keep training plans and records for documenting, maintaining and
developing the competences of personnel. Training plans should be based on training needs
assessment and should be subject to monitoring.
The training should support continuous improvement of relevant skills, the application of
scientific progress and professional development and ensure that staff members have the
appropriate qualifications, understanding of relevant pharmacovigilance requirements as well as
experience for the assigned tasks and responsibilities. All staff members of the organisation
should receive and be able to seek information about what to do if they become aware of a safety
concern.
There should be a process in place within the organisation to check that training results in the
appropriate levels of understanding and conduct of pharmacovigilance activities for the assigned
tasks and responsibilities, or to identify unmet training needs, in line with professional
development plans agreed for the organisations as well as the individual staff members.
Adequate training should also be considered by the organisation for those staff members to whom
no specific pharmacovigilance tasks and responsibilities have been assigned but whose activities
may have an impact on the pharmacovigilance system or the conduct of pharmacovigilance. Such
activities include but are not limited to those related to clinical trials, technical product
complaints, medical information, terminologies, sales and marketing, regulatory affairs, legal
affairs and audits.
Appropriate instructions on the processes to be used in case of urgency, including business
continuity (see I.B.11.2.), shall be provided by the organisation to their personnel.
the continuous monitoring of pharmacovigilance data, the examination of options for risk
minimisation and prevention and that appropriate measures are taken by the marketing
authorisation holder (see Modules IX and XII );
the scientific evaluation of all information on the risks of medicinal products as regards
patients‘ or public health, in particular as regards adverse reactions in human beings arising
from use of the product within or outside the terms of its marketing authorisation or associated
with occupational exposure (see Modules VI, VII, VIII, IX);
the submission of accurate and verifiable data on serious and non-serious adverse reactions to
the national medicines authorities within the legally required time-limits (see Modules VI and
IX);
the quality, integrity and completeness of the information submitted on the risks of medicinal
products, including processes to avoid duplicate submissions and to validate signals (see
Modules V, VI, VII, VIII and IX);
effective communication by the marketing authorisation holder with national medicines
authorities, including communication on new or changed risks, the pharmacovigilance system
master file (see Module II), risk management systems (see Module V), risk minimisations
measures (see Modules V and XVI), periodic safety update reports (see Module VII), corrective
and preventive actions (see Modules II, III and IV) and post-authorisation safety studies (see
Module VIII);
the update of product information by the marketing authorisation holder in the light of scientific
knowledge (see Module XII);
appropriate communication of relevant safety information to healthcare professionals and
patients (see Module XII and XV).
A record management system shall be put in place for all documents used for pharmacovigilance
activities, ensuring their retrievability as well as traceability of the measures taken to investigate
safety concerns, of the timelines for those investigations and of decisions on safety concerns,
including their date and the decision-making process.
where necessary and only where the parties involved assess this necessity at every stage of the
pharmacovigilance process. As part of a record management system, specific measures should
therefore be taken at each stage in the storage and processing of pharmacovigilance data to ensure
data security and confidentiality. This should involve strict limitation of access to documents and
databases to authorised personnel respecting the medical and administrative confidentiality of the
data.
There should be appropriate structures and processes in place to ensure that pharmacovigilance
data and records are protected from destruction during the applicable record retention period.
A quality plan documents the setting of quality objectives and sets out the processes to be
implemented to achieve them. A procedure is a specified way to carry out a process and may take
the format of a standard operating procedure and other work instruction or quality manual. A
quality manual documents the scope of the quality system, the processes of the quality system and
the interaction between the two. A quality record is a document stating results achieved or
providing evidence of activities performed.
quality objectives specific to their organisations in accordance with the overall quality
objectives provided under I.B.4. and the structure- and process-specific quality objectives in
accordance with each Module of GVP; and
methods for monitoring the effectiveness of the pharmacovigilance system (see I.B.12.).
the methods of monitoring the efficient operation of the quality system and, in particular, its
ability to fulfil the quality objectives;
a record management policy;
records created as a result of pharmacovigilance processes which demonstrate that key steps for
the defined procedures have been taken;
records and reports relating to the facilities and equipment including functionality checks,
qualification and validation activities which demonstrate that all steps required by the
applicable requirements, protocols and procedures have been taken;
records to demonstrate that deficiencies and deviations from the established quality system are
monitored, that corrective and preventive actions have been taken, that solutions have been
applied to deviations or deficiencies and that the effectiveness of the actions taken has been
verified.
their human resource management in the pharmacovigilance system master file (PSMF) (see
Module II)
job descriptions defining the duties of the managerial and supervisory staff.
an organisational chart defining the hierarchical relationships of managerial and supervisory
staff.
instructions on critical processes (see I.B.11.2.) in the pharmacovigilance system master file
(PSMF) (see Module II); and
their record management system in the pharmacovigilance system master file (PSMF) (see
Module II).
It is recommended that the documentation of the quality system additionally includes the
organisational structures and assignments of tasks, responsibilities and authorities to all personnel
directly involved in pharmacovigilance tasks.
For the requirements of documenting the quality system in the pharmacovigilance system master
file (PSMF) or its annexes, see Module II.
establishing, assessing and implementing risk management systems and evaluating the
effectiveness of risk minimisation;
collection, processing, management, quality control, follow-up for missing information, coding,
classification, duplicate detection, evaluation and timely electronic transmission of individual
case safety reports (ICSRs) from any source;
signal management;
scheduling, preparation (including data evaluation and quality control), submission and
assessment of periodic safety update reports;
meeting commitments and responding to requests from national medicines authorities, including
provision of correct and complete information;
interaction between the pharmacovigilance and product quality defect systems;
communication about safety concerns between marketing authorisation holders and national
medicines authorities, in particular notifying changes to the risk-benefit balance of medicinal
products;
communicating information to patients and healthcare professionals about changes to the
risk-benefit balance of products for the aim of safe and effective use of medicinal products;
keeping product information up-to-date with the current scientific knowledge, including the
conclusions of the assessment and recommendations from the applicable medicines authority.
implementation of variations to marketing authorisations for safety reasons according to the
urgency required.
Business continuity plans should be established in a risk-based manner and should include:
provisions for events that could severely impact on the organisation‘s staff and infrastructure in
general or on the structures and processes for pharmacovigilance in particular; and
back-up systems for urgent exchange of information within an organisation, amongst
organisations sharing pharmacovigilance tasks as well as between marketing authorisation
holders and national medicines authorities.
The organisation may use performance indicators to continuously monitor the good performance
pharmacovigilance activities in relation to the quality requirements. The quality requirements for
each pharmacovigilance process are provided in each Module of GVP as appropriate.
The requirements for the quality system itself are laid out in this Module and its effectiveness
should be monitored by managerial staff, who should review the documentation of the quality
system (see I.B.11.) at regular intervals, with the frequency and the extent of the reviews to be
determined in a risk-based manner. Pre-defined programmes for the review of the system should
therefore be in place. Reviews of the quality system should include the review of standard
procedures and work instructions, deviations from the established quality system, audit and
inspections reports as well as the use of the indicators referred to above.
Risk-based audits of the quality system shall be performed at regular intervals to ensure that it
complies with the requirements for the quality system, the human resource management, the
compliance management, the record management and the data retention and to ensure its
effectiveness. Audits of the quality system should include audit of the pharmacovigilance system
which is the subject of the quality system. The methods and processes for the audits are described
in Module IV. In relation to the pharmacovigilance system of a marketing authorisation holder, a
report shall be drawn up on the results for each quality audit and any follow-up audits be sent to
the management responsible for the matters audited. The report should include the results of audits
of organisations or persons the marketing authorisation holder has delegated tasks to, as these are
part of the marketing authorisation holder‗s pharmacovigilance system.
Requirements and methods for evaluating the effectiveness of actions taken upon medicinal
products for the purpose of minimising risks and supporting the safe and effective use of
medicines in patients are described in Module XVI.
There may be circumstances where a marketing authorisation holder may establish more than one
pharmacovigilance system, e.g. specific systems for particular types of products (e.g. vaccines,
products available without medical prescription).
Guidance on the structures and processes on how the marketing authorisation holder should
conduct the pharmacovigilance tasks and responsibilities is provided in the respective GVP
Modules.
I.C.1.1. Responsibilities of the marketing authorisation holder in relation to the
qualified person responsible for pharmacovigilance in the Arab Country
concerned
As part of the pharmacovigilance system, the marketing authorisation holder shall have
permanently and continuously at its disposal an appropriately qualified person responsible for
pharmacovigilance (QPPV) in the Arab Country concerned. For multinational MAHs a Local
Safety Responsible (LSR) may be accepted in some Arab Countries; consult national medicines
authorities for national requirements.
The marketing authorisation holder shall submit the name and contact details of the QPPV/LSR to
the national medicines authorities. Changes to this information should be submitted in accordance
with regulation on the national variations guidelines.
The QPPV/LSR position is a full time job. The duties of the QPPV/LSR shall be defined in a job
description. The appointed person shall be fully dedicated to his job as a QPPV/LSR. The
hierarchical relationship of the QPPV/LSR shall be defined in an organisational chart together
with those of other managerial and supervisory staff.
Information relating to the QPPV shall be included in the pharmacovigilance systems master file
(PSMF) (see Module II).
Each Pharmacovigilance system can have only one QPPV. A QPPV may be employed by more
than one marketing authorisation holder (i.e. only in case of subcontracting to a third party
organisation), for a shared or for separate pharmacovigilance systems or may fulfil the role of
QPPV for more than one pharmacovigilance system of the same marketing authorisation holder,
provided that the QPPV is able to fulfil all obligations.
For multinational MAHs; in addition to the headquarter QPPV, the national medicines authorities
request the nomination of a pharmacovigilance contact person (local safety responsible) in each
concerned Arab Country reporting to the QPPV Reporting in this context relates to
pharmacovigilance tasks and responsibilities and not necessarily to line management. A contact
person at national level may also be nominated as the Local Safety Responsible (LSR)
The marketing authorisation holder shall ensure that the QPPV has sufficient authority to
influence the performance of the quality system and the pharmacovigilance activities of the
marketing authorisation holder. The marketing authorisation holder should therefore ensure that
the QPPV has access to the pharmacovigilance system master file (PSMF) as well as authority
over it and is notified of any changes to it in accordance with Module II (see I.C.1.3). The
authority over the pharmacovigilance system and the PSMF should allow the QPPV to implement
changes to the system and to provide input into risk management plans (see Module V) as well as
into the preparation of regulatory action in response to emerging safety concerns (see Module
XII).
Overall, the marketing authorisation holder should ensure that structures and processes are in
place, so that the QPPV can fulfil the responsibilities listed in I.C.1.3. In order to do this, the
marketing authorisation holder should ensure that mechanisms are in place so that the QPPV
receives all relevant information and that the QPPV can access all information the QPPV
considers relevant, in particular on:
emerging safety concerns and any other information relating to the benefit-risk evaluation of
the medicinal products covered by the pharmacovigilance system;
ongoing or completed clinical trials and other studies the marketing authorisation holder is
aware of and which may be relevant to the safety of the medicinal products;
information from sources other than from the specific marketing authorisation holder, e.g. from
those with whom the marketing authorisation holder has contractual arrangements; and
the procedures relevant to pharmacovigilance which the marketing authorisation holder has in
place at every level in order to ensure consistency and compliance across the organisation.
The outcome of the regular reviews of the quality system referred to in I.B.6. and I.B.12. and the
measures introduced should be communicated by the managerial staff to the QPPV.
Compliance information should be provided to the QPPV on a periodic basis. Such information
also be used to provide assurance to the QPPV that commitments in the framework of risk
management plans and post-authorisation safety systems are being adhered to.
The managerial staff should also inform the QPPV of scheduled pharmacovigilance audits. The
QPPV should be able to trigger an audit where appropriate. The managerial staff should provide
QPPV with a copy of the corrective and preventive action plan following each audit relevant to
pharmacovigilance system the QPPV is responsible for, so that the QPPV can assure that
appropriate corrective actions are implemented.
In particular with regard to its adverse reaction database (or other systems to collate adverse
reaction reports), the marketing authorisation holder should implement a procedure to ensure that
the QPPV is able to obtain information from the database, for example, to respond to urgent
requests for information from the national medicines authorities, at any time. If this procedure
requires the involvement of other personnel, for example database specialists, then this should be
taken into account in the arrangements made by the marketing authorisation holder for supporting
the QPPV outside of normal working hours.
When a marketing authorisation holder intends to expand its product portfolio, for example, by
acquisition of another company or by purchasing individual products from another marketing
authorisation holder, the QPPV should be notified as early as possible in the due diligence process
in order that the potential impact on the pharmacovigilance system can be assessed and the system
be adapted accordingly. The QPPV may also have a role in determining what pharmacovigilance
data should be requested from the other company, either pre- or post-acquisition. In this situation,
the QPPV should be made aware of the sections of the contractual arrangements that relate to
responsibilities for pharmacovigilance activities and safety data exchange and have the authority
to request amendments.
When a marketing authorisation holder intends to establish a partnership with another marketing
authorisation holder, organisation or person that has a direct or indirect impact on the
pharmacovigilance system, the QPPV should be informed early enough and be involved in the
preparation of the corresponding contractual arrangements (see I.C.1.5.) so that all necessary
provisions relevant to the pharmacovigilance system are included.
1
E.g. Pharmacovigilance methods, MedDRA coding, ICSRs processing activities , Evidence based medicine,
How to conduct literature search, Causality assessment , Case Narrative Writing for Reporting Adverse Events,
Pharmacovigilance quality management, Pharmacoepidemiology, Biostatiscis, Signal detection, Medical Aspects
of Adverse Drug Reactions, Risk benefit assessment, National pharmacovigilance regulations ,
Pharmacovigilance Planning and Risk Management Plans, Risk communication
expertise or access to expertise in relevant areas such as medicine, pharmaceutical sciences as well
as epidemiology and biostatistics.
The expectation is that the applicant or marketing authorisation holder will assess the qualification
of the QPPV prior to appointment by, for example, reviewing university qualifications, knowledge
of national pharmacovigilance requirements and experience2 in pharmacovigilance.
The applicant or marketing authorisation holder should provide the QPPV with training in relation
to its pharmacovigilance system, which is appropriate for the role prior to the QPPV taking up the
position and which is appropriately documented. Consideration should be given to additional
training, as needed, of the QPPV in the medicinal products covered by the pharmacovigilance
system.
I.C.1.3. Role of the qualified person responsible for pharmacovigilance in the Arab
Country concerned
The qualified person responsible for pharmacovigilance (QPPV) is a natural3 person.
The QPPV appointed by the marketing authorisation holder shall be appropriately qualified (see
I.C.1.2.) and shall be at the marketing authorisation holder‘s disposal permanently and
continuously Back-up procedures in the case of absence of the QPPV shall be in place and should
be accessible through the QPPV‘s contact details. The QPPV should ensure that the back-up
person has all necessary information to fulfil the role.
The QPPV shall be responsible for the establishment and maintenance of the marketing
authorisation holder‘s pharmacovigilance system and therefore shall have sufficient authority to
influence the performance of the quality system and the pharmacovigilance activities and to
promote, maintain and improve compliance with the legal requirements. Hence, the QPPV should
have access to the pharmacovigilance system master file (PSMF) (see Module II) and be in a
position of authority to ensure and to verify that the information contained in the PSMF is an
accurate and up-to-date reflection of the pharmacovigilance system under the QPPV‘s
responsibility.
In relation to the medicinal products covered by the pharmacovigilance system, specific additional
responsibilities of the QPPV should include:
2
Taking into consideration that pharmacovigilance practice and regulations are relatively new in the Arab
Countries, thus having an experienced QPPV may be challenging. Accordingly, it is accepted by the national
medicines authorities in the Arab Countries that for only a transitional period the QPPV qualifications may be
expressed in terms of his pharmacovigilance training rather than his practical experience in pharmacovigilance.
Under these circumstances, once the QPPV is appointed, the MAH is responsible of providing him the unachieved
trainings in light of the checklist in module II. (Consult with national medicines authority in each Arab Country
for transitional period duration & conditions, if any,).
3
A natural person is a real human being, as distinguished from a corporation which is often treated at law as a
fictitious person.
having an overview of medicinal product safety profiles and any emerging safety concerns;
having awareness of any conditions or obligations adopted as part of the marketing
authorisations and other commitments relating to safety or the safe use of the products;
having awareness of risk minimisation measures;
being aware of and having sufficient authority over the content of risk management plans;
being involved in the review and sign-off of protocols of post-authorisation safety studies
conducted in the Arab Country concerned or pursuant to a risk management plan agreed in the
Arab Country concerned;
having awareness of post-authorisation safety studies requested by the national medicines
authority including the results of such studies;
ensuring conduct of pharmacovigilance and submission of all pharmacovigilance-related
documents in accordance with the national legal requirements and GVP in Arab Countries;
ensuring the necessary quality, including the correctness and completeness, of
pharmacovigilance data submitted to the national medicines authorities;
ensuring a full and prompt response to any request from the national medicines authorities for
the provision of additional information necessary for the benefit-risk evaluation of a medicinal
product;
providing any other information relevant to the benefit-risk evaluation to the national medicines
authorities;
providing input into the preparation of regulatory action in response to emerging safety
concerns (e.g. variations, urgent safety restrictions, and communication to patients and
healthcare professionals);
the QPPV or the LSR shall acting as a single pharmacovigilance contact point for the national
medicines authorities on a 24-hour basis and also as a contact point for pharmacovigilance
inspections.
This responsibility for the pharmacovigilance system means that the QPPV has oversight over the
functioning of the system in all relevant aspects, including its quality system (e.g. standard
operating procedures, contractual arrangements, database operations, compliance data regarding
quality, completeness and timeliness of expedited reporting and submission of periodic update
reports, audit reports and training of personnel in relation to pharmacovigilance). Specifically for
the adverse reaction database, if applicable, the QPPV should be aware of the validation status of
the database, including any failures that occurred during validation and the corrective actions that
have been taken to address the failures. The QPPV should also be informed of significant changes
that are made to the database (e.g. changes that could have an impact on pharmacovigilance
activities).
The QPPV may delegate specific tasks, under supervision, to appropriately qualified and trained
individuals, for example, acting as safety experts for certain products, provided that the QPPV
maintains system oversight and overview of the safety profiles of all products. Such delegation
should be documented.
I.C.1.4. Specific quality system processes of the marketing authorisation holder in the
Arab Country concerned
In applying the requirements set out in I.B.9.1. in the Arab Countries, the marketing authorisation
holder shall put in place the following additional specific quality system processes for ensuring:
During the retention period, retrievability of the documents should be ensured. Documents can be
retained in electronic format, provided that the electronic system has been appropriately validated
and appropriate arrangements exist for system security, access and back-up of data. If documents
in paper format are transferred into an electronic format, the transfer process should ensure that all
of the information present in the original format is retained in a legible manner and that the media
used for storage will remain readable over time. Documents transferred in situations where the
business of the marketing authorisation holder is taken over by another organisation should be
complete.
4
Use of internationally agreed terminology
For the classification, retrieval, presentation, risk-benefit evaluation and assessment, electronic exchange and communication of
pharmacovigilance and medicinal product information, marketing authorisation holders and the health authorities shall apply the
following terminology:
(a) the Medical Dictionary for Regulatory Activities (MedDRA) as developed by the International Conference on Harmonisation
of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), multidisciplinary topic M1;
(b) the terminology set out in EN ISO 11615:2012, Health Informatics, Identification of Medicinal Products (IDMP) standard,
‗Data elements and structures for unique identification and exchange of regulated medicinal product information’
(ISO/FDIS 11615:2012);
(c) the terminology set out in EN ISO 11616:2012 Health Informatics, Identification of Medicinal Products (IDMP) standard,
‗Data elements and structures for unique identification and exchange of regulated pharmaceutical product information’
(ISO/FDIS 11616:2012);
(d) the terminology set out in EN ISO 11238:2012 Health Informatics, Identification of Medicinal Products (IDMP) standard,
‗Data elements and structures for unique identification and exchange of regulated information on substances’ (ISO/FDIS
11238:2012);
(e) the terminology set out in EN ISO 11239:2012 Health Informatics, Identification of Medicinal Products (IDMP) standard,
‗Data elements and structures for unique identification and exchange of regulated information on pharmaceutical dose
forms, units of presentation and routes of administration‘ (ISO/FDIS 11239:2012);
(f) the terminology set out in EN ISO 11240:2012 Health Informatics, Identification of Medicinal Products (IDMP) standard,
‗Data elements and structures for unique identification and exchange of units of measurement‘ (ISO/FDIS 11240:2012).
Where a marketing authorisation holder has subcontracted some tasks of its pharmacovigilance
tasks, it shall retain responsibility for ensuring that an effective quality system is applied in
relation to those tasks. All guidance provided in GVP is also applicable to the other organisation
to which the tasks have been subcontracted.
When subcontracting tasks to another organisation, the marketing authorization holder shall draw
up subcontracts and these should be detailed, up-to-date and clearly document the contractual
arrangements between the marketing authorisation holder and the other organisation, describing
arrangements for delegation and the responsibilities of each party. A description of the
subcontracted activities and/or services shall be included in the PSMF and a list of the
shall be included in an annex to the PSMF, specifying the product(s) concerned (see Module II).
The other organization may be subject to inspection at the discretion of the national medicines
authorities.
Contractual arrangements should be prepared with the aim of enabling compliance with the legal
requirements by each party involved. When preparing contractual arrangements, the marketing
authorisation holder should include sufficiently detailed descriptions of the delegated tasks, the
related interactions and data exchange, together with, for example, agreed definitions, tools,
assignments and timelines. The contractual arrangements should also contain clear information on
the practical management of pharmacovigilance as well as related processes, including those for
the maintenance of pharmacovigilance databases. Further, they should indicate which processes are
in place for checking whether the agreed arrangements are being adhered to on an ongoing basis. In
this respect, regular risk-based audits of the other organisation by the marketing authorisation
holder or introduction of other methods of control and assessment are recommended.
For responsibilities of the MAH towards the QPPV in this context, see I.C.1.1.
For this purpose each national medicines authority shall operate a pharmacovigilance system and
shall establish and use an adequate and effective quality system for performing their
pharmacovigilance activities.
evaluation;
assessing and processing pharmacovigilance data in accordance with the timelines provided by
national regulations;
arranging for the essential documents describing their pharmacovigilance systems to be kept as
long as the system exists and for at least further 5 years after they have been formally
terminated;
ensuring that pharmacovigilance data and documents relating to individual authorised
medicinal products are retained as long as the marketing authorisation exists or for at least
further 10 years after the marketing authorisation has expired.
In this context, documents relating to a medicinal product include documents of a reference
medicinal product where this is applicable.
The retention periods above apply unless the documents shall be retained for a longer period
where national law so requires.
During the retention periods referred to above, retrievability of the documents should be ensured.
Documents can be retained in electronic format, provided that the electronic system has been
appropriately validated and appropriate arrangements exist for system security, access and
back-up of data. If pharmacovigilance documents in paper format are transferred into an electronic
format, the transfer process should ensure that all of the information present in the original format
is retained in a legible manner and that the media used for storage will remain readable over time.
In addition to the above, national medicines authorities shall establish procedures for collecting
and recording all suspected adverse reactions that occur in their territory (see Module VI) .
In addition, the national medicines authorities shall establish procedures for literature monitoring.
In addition to the quality system documentation in accordance with I.B.11., national medicines
authorities shall clearly determine, and to the extent necessary, keep accessible the organisational
structures and the distribution of tasks and responsibilities.
Quality audits of the national medicines authorities ‗pharmacovigilance systems (see I.B.12.) shall
be performed according to a common methodology.
GVP: Modules
II.A. Introduction
There is legal requirement for marketing authorisation holders (MAHs) to maintain and make
available upon request a pharmacovigilance system master file (PSMF) to strengthen the conduct of
pharmacovigilance activities in the Arab Countries.
The pharmacovigilance system master file definition is a detailed description of the
pharmacovigilance system used by the marketing authorisation holder with respect to one or more
authorised medicinal products.
The pharmacovigilance system master file shall be located either at the site where the main
pharmacovigilance activities of the marketing authorisation holder are performed or at the site
where the qualified person responsible for pharmacovigilance operates.
It is a requirement of the marketing authorisation application that summary information about the
pharmacovigilance system is submitted to the national medicines authorities. This summary
includes information on the location of the pharmacovigilance system master file (see II.B.2.1).
There is no requirement for variations for changes in the content of the pharmacovigilance system
master file.
This Module provides detailed guidance regarding the requirements for the pharmacovigilance
system master file, including its maintenance, content and associated submissions to national
medicines authorities.
Special considerations for multinational MAHs/ applicant are provided in II.C.3.
In this Module, all applicable legal requirements are referenced by the modal verb ―shall‖. Guidance
for the implementation of legal requirements is provided using the modal verb ―should‖.
II.B.1. Objectives
The pharmacovigilance system master file shall describe the pharmacovigilance system and
support/document its compliance with the requirements. As well as fulfilling the requirements for a
pharmacovigilance system master file laid down in the national legislation and guidance, it shall
also contribute to the appropriate planning and conduct of audits by the applicant or marketing
authorisations holder(s), the fulfilment of supervisory responsibilities of the QPPV, and of
inspections or other verification of compliance by national medicines authorities. The
pharmacovigilance system master file provides an overview of the pharmacovigilance system,
which may be requested and assessed by national medicines authorities during marketing
authorisation application(s) or post-authorisation.
Through the production and maintenance of the pharmacovigilance system master file, the
marketing authorisation holder and the QPPV should be able to:
gain assurance that a pharmacovigilance system has been implemented in accordance with the
requirements;
confirm aspects of compliance in relation to the system;
obtain information about deficiencies in the system, or non-compliance with the requirements;
obtain information about risks or actual failure in the conduct of specific aspects of
pharmacovigilance.
The use of this information should contribute to the appropriate management of and
improvement(s) to the pharmacovigilance system.
The requirements for submission of a summary of the marketing authorisation holder‘s
pharmacovigilance system, provision of the content of pharmacovigilance system master file and
the history of changes to the relevant authority(ies) should enable the planning and effective
conduct of inspections by national medicines authorities, based on a risk assessment approach.
Responsibilities, in terms of the pharmacovigilance system master file, for marketing authorisation
holders and applicants, national medicines authorities are described in detail in Section C.
For herbal or homeopathic medicinal products, the following requirements apply: to operate a
pharmacovigilance system, to prepare, maintain and make available on request at any time a
pharmacovigilance system master file and to submit a summary of the pharmacovigilance system/
full PSMF as appropriate.
II.B.2.2. Location
The pharmacovigilance system master file shall be located either at the site where the main
pharmacovigilance activities are performed or at the site where the qualified person responsible for
pharmacovigilance operates, irrespective of the format (paper-based or electronic format file).
Based on this rule, the PSMF shall be located in the Arab Country concerned, an exception is in the
situation where the main activities take place outside the Arab Country concerned (e.g.
multinational MAHs/applicants), the location should default to the site where the QPPV operates or
where the main pharmacovigilance activities are performed (e.g. located in the country of
headquarter) provided that:
the PSMF is made available to the national medicines authority in the Arab Country concerned at
any time; and
the local office/ affiliate of the MAH/applicant has detailed description on the pharmacovigilance
system/ activities on the local level
Details about the location of the pharmacovigilance system master file are required to be notified to
the national medicines authority, and any change to the location shall be notified immediately to the
national medicines authority in order to have the information updated. The required location
information for the PSMF is a physical office address of the marketing authorisation holder or a
contracted third party. Where the pharmacovigilance system master file is held in electronic form,
the location stated must be a site where the data stored can be directly accessed, and this is sufficient
in terms of a practical electronic location.
When determining the main site of pharmacovigilance activity, the marketing authorisation holder
should consider the most relevant site for the pharmacovigilance system as a whole, since the
relative importance of particular activities may vary according to products and fluctuate in the short
term. The marketing authorisation holder should have an appropriate rationale for the location
decision.
In the situation where a main site cannot be determined, the location should default to the site where
the QPPV operates.
II.B.2.3. Registration
Each national medicines authority in the Arab Countries should manage a national list/database
which provides a practical mechanism for maintaining up-to-date information about the MAH's
(or contractual partner) pharmacovigilance system master file, its status, its location, the QPPV&/or
LSR contact information and the products relevant to the pharmacovigilance system described in
the pharmacovigilance system master file.
All pharmacovigilance system master files must be registered at the national medicines authority in
the relevant Arab Country in this list/database. The MAH shall submit for such registration. In
addition, the MAH shall notify national medicines authorities to update the database with the
location of the pharmacovigilance system master file for each product, and update the information
immediately upon change.
Any recipient QPPV should explicitly accept the following changes in writing:
Transfer of responsibility for a pharmacovigilance system to a QPPV.
The QPPV should be in a position to ensure and to verify that the information contained in the
pharmacovigilance system master file is an accurate and up to date reflection of the
pharmacovigilance system under his/her responsibility (see Module I).
system shall be described in a separate pharmacovigilance system master file. Those files shall
cumulatively cover all medicinal products of the marketing authorisation holder for which a
marketing authorisation has been granted.
It is anticipated that there will be circumstances where a single marketing authorisation holder
may establish more than one pharmacovigilance system e.g. specific systems for particular types
of products (vaccines, consumer health, etc.), or that the pharmacovigilance system may include
products from more than one marketing authorisation holder. In either case, a single and specific
pharmacovigilance system master file shall be in place to describe each system.
A single QPPV shall be appointed to be responsible for the establishment and maintenance of the
pharmacovigilance system described in the pharmacovigilance system master file.
Where a pharmacovigilance system is shared by several marketing authorisation holders each
marketing authorisation holder is responsible ensuring that a pharmacovigilance system master
file exists to describe the pharmacovigilance system applicable for his products. For a particular
product(s) the marketing authorisation holder may delegate through written agreement (e.g. to a
licensing partner or contractor) part or all of the pharmacovigilance activity for which the
marketing authorisation holder is responsible. In this case the pharmacovigilance system master
file of the marketing authorisation holder may cross refer to all or part of the pharmacovigilance
system master file managed by the system of the party to whom the activity has been delegated
subject to agreement on access to that system‘s information for the marketing authorisation
holder and the authorities. The marketing authorisation holder should be able to assure the
content of the referenced file(s) in relation to the pharmacovigilance system applicable to their
product(s). Activities for maintaining the pharmacovigilance system master file in a current and
accessible state can be delegated.
Where applicable, a list of all pharmacovigilance system master files held by the same marketing
authorisation holder shall be provided in the annex (see II.B.4.8.); this includes their location(s),
details of the responsible QPPV(s) and the relevant product(s).
Submission of summary information to national medicines authorities cannot contain multiple
locations for a single pharmacovigilance system master file. The address of the location of the
pharmacovigilance system master file provided should be an office address which reflects either
the site where the main pharmacovigilance activities of the marketing authorisation holder are
performed or the site where the qualified person responsible for pharmacovigilance operates.
This address may be different to that of the applicant/marketing authorisation holder, for
example, a different office of the marketing authorisation holder or when a third party undertakes
the main activities.
Similarly, the QPPV details aligned to a product may be those of a contract QPPV responsible for
the pharmacovigilance system for a particular medicinal product, and not necessarily a QPPV
directly employed by the marketing authorisation holder.
When delegating any activities concerning the pharmacovigilance system and its master file, the
marketing authorisation holder retains ultimate responsibility for the pharmacovigilance system,
for ensuring submission of information about the pharmacovigilance system master file location,
maintenance of the pharmacovigilance system master file and its provision to national medicines
authorities upon request. Detailed written agreements describing the roles and responsibilities for
checklist on the following required practical experience/ trainings. Taking into consideration
that pharmacovigilance practice and regulations are relatively new in the Arab Countries, thus
having an experienced QPPV may be challenging. Accordingly, it is accepted by the national
medicines authorities in the Arab Countries that for only a transitional period the QPPV
qualifications may be expressed in terms of his pharmacovigilance training rather than his
practical experience in pharmacovigilance. Under these circumstances, once the QPPV is
appointed, the MAH is responsible of providing him the unachieved trainings in light of the
checklist below. (Consult with national medicines authority in each Arab Country for transitional
period duration & conditions, if any,).
Pharmacovigilance methods
MedDRA coding.
Causality assessment
Pharmaco-epidemiology
Biostatiscis
Signal detection
* during the transitional period: add 3rd column to highlight the trainings; the table header will be as
follow (insert √ or X in the respective field):
for multinational MAH/ applicant; information relating to the contact person for
pharmacovigilance (local safety responsible, LSR) nominated at national level, including contact
details.
A list of tasks that have been delegated by the qualified person for pharmacovigilance shall also be
included in the Annexes (see II.B.4.8.). This should outline the activities that are delegated and to
whom, and include the access to a medically qualified person if applicable. This list may be supplied
as a copy of a written procedural document provided the required content is covered.
The details provided in relation to the QPPV should also include the description of the QPPV
qualifications, experience and registrations relevant to pharmacovigilance. The contact details
supplied should include name, postal, telephone, fax and e-mail and represent the usual working
address of the QPPV, which may therefore be different to a marketing authorisation holder address.
If the QPPV is employed by a third party, even if the usual working address is an office of the
marketing authorisation holder, this should be indicated and the name of the company the QPPV
works for provided.
Delegated activities
The pharmacovigilance system master file, where applicable, shall contain a description of the
delegated activities and/or services relating to the fulfillment of pharmacovigilance obligations.
This includes arrangements with other parties in any country, Worldwide and if applicable, to the
pharmacovigilance system applied to products authorised in the Arab Country concerned.
Links with other organisations, such as co-marketing agreements and contracting of
pharmacovigilance activities should be outlined. A description of the location and nature of
contracts and agreements relating to the fulfilment of pharmacovigilance obligations should be
provided. This may be in the form of a list/table to show the parties involved, the roles
undertaken and the concerned product(s) and territories. The list should be organised according
to; service providers (e.g. medical information, auditors, patient support programme providers,
study data management etc.), commercial arrangements (distributors, licensing partners,
co-marketing etc.) and other technical providers (hosting of computer systems etc.). Individual
contractual agreements should be annexed with the PSMF when the later is submitted. Individual
contractual agreements shall be made available at the request of national medicines authorities at
any time or during inspection and audit and the list provided in the Annexes (see II.B.4.8.).
In each area, the marketing authorisation holder should be able to provide evidence of a system that
supports appropriate and timely decision making and action.
The description must be accompanied by the list of the following processes for compliance
management, as well as interfaces with other functions:
1. the continuous monitoring of pharmacovigilance data, the examination of options for risk
minimisation and prevention and appropriate measures are taken by the marketing authorisation
holder;
2. the scientific evaluation by the marketing authorisation holder of all information on the risks of
medicinal products;
3. the submission of accurate and verifiable data on serious and non-serious adverse reactions to
the national medicines authorities within the time limits provided in the national regulations;
4. the quality, integrity and completeness of the information submitted on the risks of medicinal
products, including processes to avoid duplicate submissions and to validate signals;
5. effective communication by the marketing authorisation holder with the national medicines
authorities, including communication on new risks or changed risks, the pharmacovigilance
system master file, risk management systems, risk minimisation measures, periodic safety
update reports, corrective and preventive actions, and post-authorisation studies;
6. the update of product information by the marketing authorisation holder in the light of scientific
knowledge, and on the basis of a continuous monitoring by the marketing authorisation holder
of information released by the national medicines authorities;
7. appropriate communication by the marketing authorisation holder of relevant safety information
to healthcare professionals and patients.
These interfaces with other functions include, but are not limited to, the roles and responsibilities of
the QPPV, responding to national medicines authority requests for information, literature searching,
safety database change control, safety data exchange agreements, safety data archiving,
pharmacovigilance auditing, quality control and training. The list, which may be located in the
Annexes, should comprise in cross matching with each one of the topics highlighted above in this
section the topic name, procedural document reference number, title, effective date and document
type (for all standard operating procedures, work instructions, manuals etc.). Procedures belonging
to service providers and other third parties should be clearly identified. Documents relating to
specific local/country procedures need not be listed, but a list may be requested on a per country
basis. If no or only some countries use specific local procedures, this should be indicated (and the
names of the applicable countries provided).
should be provided to show the timeliness of 15-day and 90-day reporting over the past year;
A description of any metrics used to monitor the quality of submissions and performance of
pharmacovigilance. This should include information provided by national medicines authorities
regarding the quality of ICSR reporting, PSURs or other submissions;
An overview of the timeliness of PSUR reporting to national medicines authorities in the Arab
Country concerned (the annex should reflect the latest figures used by the marketing
authorisation holder to assess compliance);
An overview of the methods used to ensure timeliness of safety variation submissions compared
to internal and national medicines authority deadlines, including the tracking of required safety
variations that have been identified but not yet been submitted;
Where applicable, an overview of adherence to risk management plan commitments, or other
obligations or conditions of marketing authorisation(s) relevant to pharmacovigilance.
Targets for the performance of the pharmacovigilance system shall be described and explained. A
list of performance indicators must be provided in the Annex to the pharmacovigilance system
master file, alongside the results of (actual) performance measurements.
Procedural documents
A general description of the types of documents used in pharmacovigilance (standards, operating
procedures, work instructions etc), the applicability of the various documents at global, regional
or local level within the organisation, and the controls that are applied to their accessibility,
implementation and maintenance.
Information about the documentation systems applied to relevant procedural documents under
the control of third parties.
A list of specific procedures and processes related to the pharmacovigilance activities and interfaces
with other functions, with details of how the procedures can be accessed must be provided, and the
detailed guidance for the inclusion of these is in section II.B.4.5.
Training
Staff should be appropriately trained for performing pharmacovigilance related activities and this
includes not only staff within pharmacovigilance departments but also any individual that may
receive safety reports.
A description of the resource management for the performance of pharmacovigilance activities:
- the organisational chart giving the number of people (full time equivalents) involved in
pharmacovigilance activities, which may be provided in the section describing the
organisational structure (see II.B.4.3)
Information about sites where the personnel are located (this is described under sections II.B.4.2
and II.B.4.3) whereby the sites are provided in the PSMF in relation to the organisation of
specific pharmacovigilance activities and in the Annexes which provide the list of site contacts
for sources of safety data. However, a description should be provided in order to explain the
training organisation in relation to the personnel and site information;
A summary description of the training concept, including a reference to the location training
files, record as well as the trainings materials.
Auditing
Information about quality assurance auditing of the pharmacovigilance system should be included
in the pharmacovigilance system master file. A description of the approach used to plan audits of the
pharmacovigilance system and the reporting mechanism and timelines should be provided, with a
current list of the scheduled and completed audits concerning the pharmacovigilance system
maintained in the annex referred to II.B.4.8.. This list should describe the date(s) (of conduct and of
report), scope and completion status of audits of service providers, specific pharmacovigilance
activities or sites undertaking pharmacovigilance and their operational interfaces relevant to the
fulfilment of the pharmacovigilance obligations, and cover a rolling 5 year period.
The pharmacovigilance system master file shall also contain a note associated with any audit where
significant findings are raised. This means that the presence of findings that fulfil the national
criteria for major or critical findings must be indicated (see Module IV). The audit report must be
documented within the quality system; in the pharmacovigilance system master file it is sufficient to
provide a brief description of the corrective and/or preventative action(s) associated with the
significant finding, the date it was identified and the anticipated resolution date(s), with cross
reference to the audit report and the documented corrective and preventative action plan(s). In the
annex, in the list of audits conducted, those associated with unresolved notes in the
pharmacovigilance system master file, should be identified. The note and associated corrective and
preventative action(s), shall be documented in the pharmacovigilance system master file until the
corrective and/or preventative action(s) have been fully implemented, that is, the note is only
removed once corrective action and/or sufficient improvement can be demonstrated or has been
independently verified. The addition, amendment or removal of the notes must therefore be
recorded in the logbook.
As a means of managing the pharmacovigilance system, and providing a basis for audit or
inspection, the pharmacovigilance system master file should also describe the process for recording,
managing and resolving deviations from the quality system. The master file shall also document
deviations from pharmacovigilance procedures, their impact and management until resolved. This
may be documented in the form of a list referencing a deviation report, and its date and procedure
concerned.
A logbook of any change of the content of the pharmacovigilance system master file made within
the last five years except the changes in annexes and the following QPPV information: CV,
contact details, back-up arrangements and contact person for pharmacovigilance on the national
level. .In addition, other change control documentation should be included as appropriate.
Documented changes shall include at least the date, person responsible for the change and the
nature of the change.
itself or via access to the systems used to generate the Annex content).
Marketing authorisation holders should be able to justify their approach and have document control
procedures in place to govern the maintenance of the pharmacovigilance system master file. As a
basis for audit and inspections, the pharmacovigilance system master file provides a description of
the pharmacovigilance system at the current time, but the functioning and scope of the
pharmacovigilance system in the past may need to be understood.
Changes to the pharmacovigilance system master file should also account for shared
pharmacovigilance systems and delegated activities. A record of the date and nature of notifications
of the changes made available to the national medicines authorities, the QPPV and relevant third
parties should be kept in order to ensure that change control is fully implemented.
The pharmacovigilance system master file should be retained in a manner that ensures its legibility
and accessibility.
The headings used in II.B.4 should be used for the main content of the pharmacovigilance system
master file. The minimum required content of the Annexes is outlined in II.B.4.8, and additional
information may be included in the Annexes, provided that the requirements for the content of the
main sections (II.B.1-7) are also met. The positioning of content in the Annexes is further outlined;
the bulleted points are descriptions of possible content (and not required headings):
Products, Annex H
List(s) of products covered by the pharmacovigilance system
Any notes concerning the MAH per product
II.C.1. Responsibilities
The pharmacovigilance system master file creation, maintenance in a current and accessible state
(permanently available for audit and inspection purposes) and provision to national medicines
authorities can be outsourced to a third party, but the marketing authorisation holder retains ultimate
responsibility for compliance with the legal requirements.
When the QPPV/LSR and related contact details change or when the location of the
pharmacovigilance system master file changes, the marketing authorisation holder is required to
notify/submit the appropriate variation application(s) to the national medicines authorities as
applicable.
MAH/applicant has not previously submit the PSMF in the Arab Country concerned or is in the
process of establishing a new pharmacovigilance system; the first PSMF submission should be
accompanied by the complete version of pharmacovigilance SOPs.
In the situation where the same pharmacovigilance system master file is used by more than one
marketing authorisation holder (where a common pharmacovigilance system is used) the concerned
pharmacovigilance system master file should be accessible to each, as any of the applicable
marketing authorisation holders shall be able to provide the file to the medicines authorities within
14 days, upon request (unless otherwise stated in the request).
The full PSMF (along together with its summary) is requested to be submitted in the marketing
authorisation applications (i.e. pre-authorisation) in the following situations:
the applicant has not previously held a marketing authorisation in the Arab Country concerned,
full PSMF is appropriate to review the description of a pharmacovigilance system;
the applicant has not previously submit the PSMF in the Arab Country concerned or is in the
process of establishing a new pharmacovigilance system;
the applicant had major changes in its organisation, such as mergers and acquisitions or in its
pharmacovigilance system
the applicant has major or critical findings in the previous pharmacovigilance system assessment
by the national medicines authority;
the applicant has a history or culture of pharmacovigilance non-compliance; previous
information (e.g. inspection history and non-compliance notifications or information from other
authorities). In addition to the submission of the full PSMF, if the marketing authorisation holder
has a history of serious and/or persistent pharmacovigilance non-compliance, a pre-authorisation
pharmacovigilance inspection may be one mechanism to confirm that improvements have been
made to the system before a new authorisation is granted (see module III);
where specific concerns about the pharmacovigilance system and/or the product safety profile
exist;
any other situation as seen appropriate by the national medicines authority;
Except in the above situations, the pharmacovigilance system master file should not routinely be
requested during the assessment of new marketing authorisation applications (i.e.
pre-authorisation), but may be requested on an ad hoc basis, particularly if a new
pharmacovigilance system is being implemented, or if product specific safety concerns or issues
with compliance with pharmacovigilance requirements have been identified or in preparation for an
pharmacovigilance inspection.
information for medicinal products authorised for the MAH, with information on the
pharmacovigilance system to the local or regional activities. Despite this fact, pharmacovigilance
activities on the national level as described in the PSMF may not be applied to the same extent by all
the MAH's national offices/ affiliates, furthermore, some additional national requirements and
details may also apply. Accordingly, multinational MAHs/Applicants should provide clear
illustration of the key elements of both global pharmacovigilance system and national
pharmacovigilance sub-system, highlighting the role of LSR, which pharmacovigilance activities
are carried out in the Arab Country concerned, which are carried out in the headquarter/globally and
how they integrate together.
For the Multinational MAH/Applicant the following two documents are required (for submission
requirement see II.C.3.5.):
1. The PSMF (according to European Good Pharmacovigilance Practice which is the base for this
guideline) and,
2. National pharmacovigilance sub-system file (national PSSF) which describes the key
elements of pharmacovigilance activities in the Arab County concerned.
contact details;
details of back-up arrangements to apply in the absence of the LSR;
checklist on the following required practical experience/ trainings:
Taking into consideration that pharmacovigilance practice and regulations are relatively new in
the Arab Countries, thus having an experienced LSR may be challenging. Accordingly, it is
accepted by the national medicines authorities in the Arab Countries that for only a transitional
period the LSR qualifications may be expressed in terms of his pharmacovigilance training
rather than his practical experience in pharmacovigilance. Under these circumstances, once the
LSR is appointed, the MAH is responsible of providing him the unachieved trainings in light of
the check list below. (Consult with national medicines authority in each Arab Country for
transitional period duration & conditions, if any,).
Practical experience*
Topic (insert √ or X in the respective
field)
Pharmacovigilance methods
MedDRA coding.
Causality assessment
Introduction to pharmaco-epidemiology
Biostatiscis
Practical experience*
Topic (insert √ or X in the respective
field)
* during the transitional period: add 3rd column to highlight the trainings the table header will be as
follow: (insert √ or X in the respective field)
If applicable, a list of tasks that have been delegated by the LSR shall also be included in the
Annexes (see II.C.3.2.8.). This should outline the activities that are delegated and to whom.
The details provided in relation to the LSR should also include the description of the LSR
qualifications, experience and registrations relevant to pharmacovigilance. The contact details
supplied should include name, postal, telephone, fax and e-mail and represent the usual working
address of the LSR.
II.C.3.2.2. National PSSF section on the "organisational structure of the MAH's local office"
Remember that the information provided in this section of the national PSSF shall focus on the
national pharmacovigilance sub-system
A description of the organisational structure of the MAH's local office relevant to the national
pharmacovigilance sub-system must be provided. The description should provide a clear
overview of the company(ies) involved, the main pharmacovigilance department and the
relationship(s) between organisations and operational units relevant to the fulfilment of
pharmacovigilance obligations. This should include third parties. Specifically, the national PSSF
shall describe:
- The organisational structure of the MAH's local office, showing the position of the LSR in
the organisation.
- The site(s) where the pharmacovigilance functions on the national level are undertaken
covering individual case safety report collection, evaluation, safety database case entry,
periodic safety update report production (integration with global system), signal detection
and analysis(integration with global system), risk management plan management, pre- and
post-authorisation study management, and management of safety.
Diagrams may be particularly useful; the name of the department or third party should be
indicated.
Delegated activities
The national PSSF, where applicable, shall contain a description of the delegated activities
and/or services relating to the fulfillment of pharmacovigilance obligations.
Links with other organisations, such as co-marketing agreements and contracting of
pharmacovigilance activities on the national level should be outlined. A description of the
location and nature of contracts and agreements relating to the fulfilment of pharmacovigilance
obligations should be provided. This may be in the form of a list/table to show the parties
involved, the roles undertaken and the concerned product(s) and territories. The list should be
organised according to; service providers (e.g. medical information, auditors, patient support
programme providers, study data management etc.), commercial arrangements (distributors,
licensing partners, co-marketing etc.) and other technical providers (hosting of computer systems
etc.). Individual contractual agreements should be annexed with the national PSSF when the later
is submitted. Individual contractual agreements shall be made available at the request of national
medicines authorities at any time or during inspection and audit and the list provided in the
Annexes (see II.C.3.2.8).
Arab Country concerned (at the site where the main pharmacovigilance activities are performed
globally e.g. Headquarter). However, LSR must have online access to national safety cases and all
national pharmacovigilance data of the Arab Country concerned; otherwise at least backup database
of this national data should always be kept in the local office.
The location, functionality and operational responsibility for computerised systems and databases
used (on the national level) to receive, collate, record and report safety information and an
assessment of their fitness for purpose shall be described in the national PSSF.
Where multiple computerised systems/databases are used on national level, the applicability of
these to pharmacovigilance activities should be described in such a way that a clear overview of the
extent of computerisation within the pharmacovigilance system can be understood. The validation
status of key aspects of computer system functionality should also be described; the change control,
nature of testing, back-up procedures and electronic data repositories vital to pharmacovigilance
compliance should be included in summary, and the nature of the documentation available
described. For non-electronic systems (where an electronic system may only be used for expedited
submission of ICSRs), the management of the data, and mechanisms used to assure the integrity and
accessibility of the safety data, and in particular the collation of information about adverse drug
reactions, should be described.
headquarter)
Communication of safety concerns to consumers, healthcare professionals and the national
medicines authorities;
Implementation of safety variations to the summary of product characteristics (SmPC) and
patient information leaflets; procedures should cover both internal (within the MAH) and
external communications.
In each area, the marketing authorisation holder should be able to provide evidence of a sub-system
that supports appropriate and timely decision making and action on the national level (taking into
consideration liaising with the MAH's headquarter).
The description must be accompanied by the list of the following processes for compliance
management, as well as interfaces with other functions (on the national level and as appropriate
in integration with MAH's headquarter):
1. the continuous monitoring of pharmacovigilance data, the examination of options for risk
minimisation and prevention and appropriate measures are taken by the marketing authorisation
holder;
2. the scientific evaluation by the marketing authorisation holder of all information on the risks of
medicinal products;
3. the submission of accurate and verifiable data on serious and non-serious adverse reactions to
the national medicines authorities within the time limits provided in the national regulations;
4. the quality, integrity and completeness of the information submitted on the risks of medicinal
products, including processes to avoid duplicate submissions and to validate signals;
5. effective communication by the marketing authorisation holder with the national medicines
authorities, including communication on new risks or changed risks, the pharmacovigilance
system master file & national PSSF , risk management systems, risk minimisation measures,
periodic safety update reports, corrective and preventive actions, and post-authorisation studies;
6. the update of product information by the marketing authorisation holder in the light of scientific
knowledge, and on the basis of a continuous monitoring by the marketing authorisation holder
of information released by the national medicines authorities;
7. appropriate communication by the marketing authorisation holder of relevant safety information
to healthcare professionals and patients.
These interfaces with other functions include, but are not limited to, the roles and responsibilities of
the LSR, responding to national medicines authority requests for information, literature searching,
safety database change control, safety data exchange agreements, safety data archiving,
pharmacovigilance auditing, quality control and training. The list, which may be located in the
Annexes, should comprise in cross matching with each one of the topics highlighted above in this
section, the topic name, the procedural document reference number, title, effective date and
document type (for all standard operating procedures, work instructions, manuals etc.). Procedures
belonging to service providers and other third parties should be clearly identified. In addition, any
specific local (in the Arab Country concerned) procedures should be also indicated.
Procedural documents
A general description of the types of documents used in pharmacovigilance (standards, operating
procedures, work instructions etc), the applicability of the various documents at local level
within the organisation, and the controls that are applied to their accessibility, implementation
and maintenance.
Information about the documentation systems applied to relevant procedural documents under
the control of third parties.
A list of specific procedures and processes related to the pharmacovigilance activities (on the
national level) and interfaces with other functions, with details of how the procedures can be
accessed must be provided, and the detailed guidance for the inclusion of these is in section
II.C.3.2.5.
Training
Staff should be appropriately trained for performing pharmacovigilance related activities and this
includes not only staff within pharmacovigilance departments but also any individual that may
receive safety reports such as sales personnel or clinical research staff.
A description of the resource management for the performance of pharmacovigilance activities
on the national level:
- the organisational chart giving the number of people (full time equivalents) involved in
pharmacovigilance activities, which may be provided in the section describing the
organisational structure (see II.C.3.2.3.)
Information about sites where the personnel are located (this is described under sections
II.C.3.2.2.) whereby the sites are provided in the national PSSF in relation to the organisation of
specific pharmacovigilance activities. However, a description should be provided in order to
explain the training organisation in relation to the personnel and site information;
A summary description of the training concept, including a reference to the location training
files, record as well as the trainings materials.
Auditing
Information about quality assurance auditing of the national pharmacovigilance sub-system should
be included in the national PSSF. A description of the approach used to plan audits of the national
pharmacovigilance sub-system and the reporting mechanism and timelines should be provided, with
a current list of the scheduled and completed audits concerning the national pharmacovigilance
sub-system maintained in the annex referred to II.C.3.2.8. This list should describe the date(s) (of
conduct and of report), scope and completion status of audits of service providers, specific
pharmacovigilance activities or sites undertaking pharmacovigilance and their operational
interfaces relevant to the fulfilment of the pharmacovigilance obligations, and cover a rolling 5 year
period.
The national PSSF shall also contain a note associated with any audit where significant findings are
raised. This means that the presence of findings that fulfil the national criteria for major or critical
findings must be indicated (see Module IV). The audit report must be documented within the
quality system; in the national PSSF it is sufficient to provide a brief description of the corrective
and/or preventative action(s) associated with the significant finding, the date it was identified and
the anticipated resolution date(s), with cross reference to the audit report and the documented
corrective and preventative action plan(s). In the annex, in the list of audits conducted to the national
pharmacovigilance sub-system, those associated with unresolved notes in national PSSF, should be
identified. The note and associated corrective and preventative action(s), shall be documented in the
national PSSF until the corrective and/or preventative action(s) have been fully implemented, that
is, the note is only removed once corrective action and/or sufficient improvement can be
demonstrated or has been independently verified. The addition, amendment or removal of the notes
must therefore be recorded in the logbook.
As a means of managing the national pharmacovigilance sub-system, and providing a basis for audit
or inspection, the national PSSF should also describe the process for recording, managing and
resolving deviations from the quality system. The national PSSF shall also document deviations
from pharmacovigilance procedures on the national level, their impact and management until
resolved. This may be documented in the form of a list referencing a deviation report, and its date
and procedure concerned.
Where national pharmacovigilance sub-systems are shared, all products that utilise the national
pharmacovigilance sub-system should be included, so that the entire list of products covered by
the file is available. The products lists may be presented separately, organised per MAH.
Alternatively, a single list may be used, which is supplemented with the name of the MAH(s)
for each product, or a separate note can be included to describe the product(s) and the MAH(s)
covered;
A list of written policies and procedures for the compliance management (see II.C.3.2.5.);
A list of contractual agreements covering delegated activities in the Arab Country concerned
including the medicinal products. In addition, a copy of the individual contractual agreements
shall also be included in this annex when the PSMF is submitted to the national medicines
authorities;
A list of tasks that have been delegated by the LSR (if any);
A list of all completed audits on the national level, for a period of five years, and a list of audit
schedules on the national level;
Where applicable, a list of performance indicators (see II.C.3.3.6.);
Where applicable, a list of other national PSSF(s) held by the same marketing authorisation
holder;
This list should include the national PSSF number(s), the name of MAH, the name of the LSR
responsible for the pharmacovigilance sub-system used. If the pharmacovigilance system is
managed by another party that is not a marketing authorisation holder, the name of the service
provider should also be included.
A logbook of any change of the content of the national PSSF made within the last five years
except the changes in annexes and the following LSR information: CV, contact details, back-up
arrangements and contact person for pharmacovigilance on the national level. .In addition, other
change control documentation should be included as appropriate. Documented changes shall
include at least the date, person responsible for the change and the nature of the change.
should be legible, complete, provided in a manner that ensures all documentation is accessible and
allow full traceability of changes. Therefore, it may be appropriate to restrict access to it in order to
ensure appropriate control over the content and to assign specific responsibilities for the national
PSSF in terms of change control and archiving.
The national PSSF should be written in English (unless otherwise is requested by the national
medicines authority in the Arab Country concerned), indexed in a manner consistent with the
headings described in this Module, and allow easy navigation to the contents with. In general,
embedded documents are discouraged. The use of electronic book-marking and searchable text is
recommended. Documents such as copies of signed statements or agreements should be included as
appendices and described in the index.
The documents and particulars of the national PSSF shall be presented with the following headings
and, if hardcopy, in the order outlined:
Cover Page to include:
The unique number assigned by the national medicines authority to national PSSF (if applicable).
The name of the MAH, the MAH of the LSR responsible for the national pharmacovigilance
sub-system described (if different), as well as the relevant QPPV third party company name (if
applicable).
The name of other concerned MAH(s) (sharing the national pharmacovigilance sub-system) (if
applicable)
The list of national PSSF(s) for the MAH (concerning products with a different
pharmacovigilance sub-system) (if applicable)
The date of preparation / last update
The headings used in II.C.3.2. should be used for the main content of the national PSSF. The
minimum required content of the Annexes is outlined in II.C.3.2.8., and additional information may
be included in the Annexes, provided that the requirements for the content of the main sections
(II.C.3.2.1-7) are also met. The positioning of content in the Annexes is further outlined; the
bulleted points are descriptions of possible content (and not required headings):
Products, Annex H
List(s) of products covered by the national pharmacovigilance sub-system described in this
national PSSF
Any notes concerning the MAH per product
fulfil on the national level the pharmacovigilance tasks and responsibilities listed in this GVP
modules;
a reference to the location where the national PSSF for the medicinal product is kept.
The national PPSF should not routinely be submitted during the assessment of new marketing
authorisation applications (i.e. pre-authorisation), but may be requested on an ad hoc basis, (see
II.C.3.5. for submission requirement).
In case that these situations apply to the national PSSF but not the PSMF; then the multinational
MAH can submit the "summary of PSMF" & the "national PSSF", and vice versa.
Except in the above situations, the PSMF and/or the national PSSF (as appropriate) should not
routinely be requested during the assessment of new marketing authorisation applications (i.e.
pre-authorisation), instead the "summary of PSMF" and "summary of national PSSF" should be
submitted. The following table summarises the different scenarios.
Table II.1 Conditions to submit the PSMF and the national PSSF
Situations in II.C.3.5.1 apply to both PSMF and the PSMF & summary PSMF
national PSSF
National PSSF & summary of national PSSF
Situations in II.C.3.5.1 apply to only PSMF PSMF & summary PSMF summary of
national PSSF
The full PSMF and the national PSSF may be requested on an ad hoc basis by the national
medicines authority in the following situations
particularly if a new pharmacovigilance system is being implemented or the MAH has not
previously submit the PSMF and the national PSSF in the Arab Country concerned; or
if product specific safety concerns or issues with compliance with pharmacovigilance
requirements have been identified; or
in preparation for an pharmacovigilance inspection
any time upon request of the national medicines authority
The marketing authorisation holder shall maintain and make available on request a copy of the
PSMF and national PSSF. The marketing authorisation holder must submit the copy within 14 days
after receipt of the request from the national medicines authority in the Arab Countries concerned
(unless otherwise stated in the request). The PSMF and national PSSF should be submitted in a
clearly arranged readable electronic format or clearly arranged printed copy (consult with the
national medicines authority for required format).
GVP: Modules
III.A. Introduction
This Module contains guidance on the planning, conduct, reporting and follow-up of
pharmacovigilance inspections in the Arab Countries and outlines the role of the different parties
involved. General guidance is provided under III.B., while III.C. covers the overall operation of
pharmacovigilance inspections in the Arab Countries.
In order to determine that marketing authorisation holders comply with pharmacovigilance
obligations established within an Arab Country, and to facilitate compliance, the national medicines
authorities concerned shall conduct, pharmacovigilance inspections of marketing authorisation
holders or any firms employed to fulfil marketing authorisation holder‘s pharmacovigilance
obligations. Such inspections shall be carried out by inspectors appointed by the national medicines
authority and empowered to inspect the premises, records, documents and pharmacovigilance
system master file (PSMF) of the marketing authorisation holder or any firms employed by the
marketing authorisation holder to perform the pharmacovigilance activities. In particular, marketing
authorisation holders are required to provide, on request, the pharmacovigilance system master file,
which will be used to inform inspection conduct (see Module II).
The objectives of pharmacovigilance inspections are:
to determine that the marketing authorisation holder has personnel, systems and facilities in place
to meet their pharmacovigilance obligations;
to identify, record and address non-compliance which may pose a risk to public health;
to use the inspection results as a basis for enforcement action, where considered necessary.
For marketing authorisation holders of products in an Arab Country, it is the responsibility of the
national medicines authority of this country to verify, that the marketing authorisation holder for the
medicinal product satisfies the national pharmacovigilance requirements. The pharmacovigilance
system master file shall be located either where the main pharmacovigilance activities of the
marketing authorisation holder are performed or where the qualified person responsible for
pharmacovigilance operates. The national medicines authority may conduct pre-authorisation
inspections to verify the accuracy and successful implementation of the existing or proposed
pharmacovigilance system.
Pharmacovigilance inspection programmes will be implemented, which will include routine
inspections scheduled according to a risk-based approach and will also incorporate ―for cause‖
inspections, which have been triggered to examine suspected non-compliance or potential risks,
usually with impact on a specific product(s).
The results of an inspection will be provided to the inspected entity, who will be given the
opportunity to comment on any non-compliance identified. Any non-compliance should also be
rectified by the marketing authorisation holder in a timely manner through the implementation of a
corrective and preventive action plan.
If the outcome of the inspection is that the marketing authorisation holder does not comply with the
pharmacovigilance obligations, the national medicines authority concerned shall take the necessary
measures to ensure that a marketing authorisation holder is subject to effective, proportionate and
dissuasive penalties.
Sharing of information and communication between pharmacovigilance inspectors and assessors, is
very important to ensure successful prioritisation and targeting of these inspections and for the
proper follow-up of inspections and the provision of recommendations on actions to be taken.
inspected party, to ensure the availability of relevant individuals for the inspection. However, on
occasion, it may be appropriate to conduct unannounced inspections or to announce an inspection at
short notice (e.g. when the announcement could compromise the objectives of the inspection or
when the inspection is conducted in a short timeframe due to urgent safety reasons).
III.B.1.6. Re-inspections
A re-inspection may be conducted on a routine basis as part of a routine inspection programme. Risk
factors will be assessed in order to prioritise re-inspections. Early re-inspection may take place
where significant non-compliance has been identified and where it is necessary to verify actions
taken to address findings and to evaluate ongoing compliance with the obligations, including
evaluation of changes in the pharmacovigilance system. Early re-inspection may also be appropriate
when it is known from a previous inspection that the inspected party had failed to implement
appropriately corrective and preventive actions in response to an earlier inspection.
when establishing pharmacovigilance inspection programmes include, but are not limited to:
inspection related:
- compliance history identified during previous pharmacovigilance inspections or other types
of inspections (GCP, GMP, GLP and GDP);
- re-inspection date recommended by the inspectors or assessors as a result of a previous
inspection;
product related:
- product with additional pharmacovigilance activities or risk-minimisation activities;
- authorisation with conditions associated with safety, e.g. requirement for post-authorisation
safety studies (PASS) or designation for additional monitoring;
- product(s) with large sales volume, i.e. products associated with large patient exposure in the
Arab Country concerned;
- product(s) with limited alternative in the market place;
Marketing authorisation holder related:
- marketing authorisation holder that has never been subject to a pharmacovigilance
inspection;
- marketing authorisation holder with many products on the market in the Arab Country
concerned;
- resources available to the marketing authorisation holder for the pharmacovigilance
activities they undertake;
- marketing authorisation holder with no previous marketing authorisations in the Arab
Country concerned;
- negative information and/or safety concerns raised by the national medicines authority,
other bodies/medicines authorities outside the Arab Country concerned or other areas (i.e.
GCP, GMP, GLP and GDP);
- changes in the marketing authorisation holder organisation, such as mergers and
acquisitions;
pharmacovigilance system related:
- marketing authorisation holder with sub-contracted pharmacovigilance activities (function
of the qualified person responsible for pharmacovigilance (QPPV) in the Arab Country
concerned, reporting of safety data etc.) and/or multiple firms employed to perform
pharmacovigilance activities;
- change of QPPV/local safety responsible (LSR) since the last inspection;
- changes to the pharmacovigilance safety database(s), which could include a change in the
database itself or associated databases, the validation status of the database as well as
information about transferred or migrated data;
- changes in contractual arrangements with pharmacovigilance service providers or the sites
(see ICSRs);
- submission of study results and relevant safety information (e.g. development safety update
reports (DSURs) and information included in PSURs), where applicable, PASS or
post-authorisation efficacy studies (PAES) submissions, particularly when associated with
specific obligations or RMP commitments;
- appropriate selection of reference safety information, maintenance of investigator brochures
and patient information with respect to safety;
- the inclusion of study data in ongoing safety evaluation;
pharmacovigilance system:
- QPPV/LSR roles and responsibilities, e.g. access to the quality system, the
pharmacovigilance system master file, performance metrics, audit and inspection reports,
and their ability to take action to improve compliance;
- the roles and responsibilities of the marketing authorisation holder in relation to the
pharmacovigilance system;
- accuracy, completeness and maintenance of the pharmacovigilance system master file;
- quality and adequacy of training, qualifications and experience of staff;
- coverage and adherence to the quality system in relation to pharmacovigilance, including
quality control and quality assurance processes;
- fitness for purpose of computerised systems;
- contracts and agreements with all relevant parties appropriately reflect responsibilities and
activities in the fulfilment of pharmacovigilance, and are adhered to.
- As a general approach, a marketing authorisation holder should be inspected on the basis of
risk-based considerations, but it is recommended to routinely inspect MAH at least once
every 4 years.
The inspection may include the system for the fulfilment of conditions of a marketing authorisation
and the implementation of risk–minimisation activities, as they relate to any of the above safety
topics.
III.B.4.3. Re-inspections
For the scope of a re-inspection, the following aspects should be considered:
review of the status of the system and/or corrective and preventive action plan(s) resulting from
previous pharmacovigilance inspection(s);
review of significant changes that have been made to the pharmacovigilance system since the last
pharmacovigilance inspection (e.g. change in the pharmacovigilance database, company mergers
or acquisitions, significant changes in contracted activities, change in QPPV/LSR as
appropriate);
review of process and/or product-specific issues identified from the assessment of information
provided by the marketing authorisation holder, or not covered in a prior inspection.
The scope of re-inspection will depend on inspection history. It may be appropriate to conduct a
complete system review, for example if a long time has elapsed since the previous inspection, in
which case the elements listed in III.B.4.1. may be considered for the inspection scope, as
appropriate.
non-compliances, to clarify the legal requirements and the expectations of the regulator, and to
review the marketing authorisation holder‘s proposals for corrective and preventive actions;
provision of information to other medicines authorities (in Arab and non- Arab countries) under
the framework of confidentiality arrangements;
inspection: non-compliant marketing authorisation holders may be inspected to determine the
extent of non-compliance and then re-inspected to ensure compliance is achieved;
warning letter, non-compliance statement or infringement notice; these are instruments which
national medicines authorities may issue stating the legislation and guideline that has been
breached, reminding marketing authorisation holders of their pharmacovigilance obligations or
specifying the steps that the marketing authorisation holder must take and in what timeframe in
order to rectify the non-compliance and in order to prevent a further case of non-compliance;
the national medicines authority may consider making public a list of marketing authorisation
holders found to be seriously or persistently non-compliant;
actions against a marketing authorisation(s) or authorisation application(s) e.g.
- Urgent Safety Restriction;
- variation of the marketing authorisation;
- suspension or revocation of the marketing authorisation;
- delays in approvals of new marketing authorisation applications until corrective and
preventive actions have been implemented or the addition of safety conditions to new
authorisations;
- requests for pre-authorisation inspections;
product recalls e.g. where important safety warnings have been omitted from product
information;
action relating to marketing or advertising information;
amendments or suspension of clinical trials due to product-specific safety issues;
administrative penalties, usually fixed fines or based on company profits or levied on a daily
basis;
referral for criminal prosecution with the possibility of imprisonment (in accordance with
national legislation).
national medicines authority should be officials of, or appointed by, the national medicines
authority in accordance with national regulation and follow the provisions of the national medicines
authority.
It is recommended that inspectors are appointed based upon their experience (especially in
pharmacovigilance) and the minimum requirements defined by the national medicines authority. In
addition, consideration should be given to the recommendations for training and experience
described in the pharmacovigilance inspections procedures.
The inspectors should undergo training to the extent necessary to ensure their competence in the
skills required for preparing, conducting and reporting inspections. They should also be trained in
pharmacovigilance processes and requirements in such way that they are able, if not acquired by
their experience, to comprehend the different aspects of a pharmacovigilance system.
Documented processes should be in place in order to ensure that inspection competencies are
maintained. In particular, inspectors should be kept updated with the current status of
pharmacovigilance legislation and guidance.
Training and experience should be documented individually and evaluated according to the
requirements of the applicable quality system of the concerned medicines authority.
GVP: Modules
IV.A. Introduction
For the purposes of this module reference to pharmacovigilance audit(s) and pharmacovigilance
audit activity(ies) are deemed to include pharmacovigilance system audits and audit(s) of the
quality system for pharmacovigilance activities.
The overall description and objectives of pharmacovigilance systems and quality systems for
pharmacovigilance activities are referred to in Module I, while the specific pharmacovigilance
processes are described in each respective Module of GVP.
In this Module, all applicable legal requirements are referenced by the modal verb ―shall‖. Guidance
for the implementation of legal requirements is provided using the modal verb ―should‖.
This Module provides guidance on planning and conducting the legally required audits, the role,
context and management of pharmacovigilance audit activity. This Module is intended to facilitate
the performance of pharmacovigilance audits, especially to promote harmonisation, and encourage
consistency and simplification of the audit process. The principles in this Module are aligned with
internationally accepted auditing standards, issued by relevant international auditing
standardisation organisations5 and support a risk-based approach to pharmacovigilance audits.
Section IV.B. outlines the general structures and processes that should be followed to identify the
most appropriate pharmacovigilance audit engagements and describes the steps which can be
undertaken by marketing authorisation holders to plan, conduct and report upon an individual
pharmacovigilance audit engagements. This Section also provides an outline of the general quality
system and record management practices for pharmacovigilance audit processes.
Section IV.C. provides an outline of the operation in the Arab Countries in respect of
pharmacovigilance audits.
IV.A.1. Terminology
Audit, Audit findings, Audit plan, Audit programme, Audit recommendations, Upper management:
see in GVP Annex I.
Auditee: [entity] being audited (ISO 19011 (3.7) 6).
Compliance: Conformity and adherence to policies, plans, procedures, laws, regulations, contracts,
or other requirements (IIA International Standards for the Professional Practice of Internal
Auditing 6).
5
For more details regarding The Institute of Internal Auditors (IIA) see www.theiia.org;
the International Organisation for Standardisation (ISO) see www.iso.org ;
Information Systems Audit and Control Association (ISACA) see www.isaca.org ;
The International Auditing and Assurance Standards Board (IAASB) see www.ifac.org ;
The International Organisation of Supreme Audit Institutions (INTOSAI) see www.issai.org .
6
The Institute of Internal Auditors (IIA) see www.theiia.org
Control(s): Any action taken by management and other parties to manage risk and increase the
likelihood that established objectives and goals will be achieved. Management plans, organises, and
directs the performance of sufficient actions to provide reasonable assurance that objectives and
goals will be achieved (IIA International Standards for the Professional Practice of Internal
Auditing 6).
Evaluation (of audit activities): Professional auditing bodies promote compliance with standards,
including in quality assurance of their own activities, and codes of conduct, which can be used to
address adequate fulfilment of the organisation‘s basic expectations of Internal Audit activity and
its conformity to internationally accepted auditing standards.
Finding(s): see Audit findings
Head of the organisation: see Upper management
Auditors‘ independence: The freedom from conditions that threaten objectivity or the appearance of
objectivity. Such threats to objectivity must be managed at the individual auditor, engagement,
functional and organisational levels (IIA International Standards for the Professional Practice of
Internal Auditing 6).
Internal Control: Internal control is an integral process that is effected by an entity‘s management
and personnel and is designed to address risk and provide reasonable assurance that in pursuit of the
entity‘s mission, the following general objectives are being achieved: executing orderly, ethical,
economical, efficient and effective operations, fulfilling accountability obligations, complying with
applicable laws and regulations and safeguarding resources against loss, misuse and damage (for
further information refer to COSO standards).
International Auditing Standards: issued by International Auditing Standardisation Organisations*.
International Auditing Standardisation Organisations: More details regarding:
The Institute of Internal Auditors (IIA) standards can be found at
http://www.theiia.org/guidance/standards-and-guidance/ippf/standards/full-standards;
The International Organisation for Standardisation (ISO) standard 19011 ―Guidelines for
quality and/or environmental management systems auditing. http://www.iso.org/iso/home.html;
Information Systems Audit and Control Association (ISACA) standards can be found at
http://www.isaca.org/Standards;
The International Auditing and Assurance Standards Board (IAASB) standards can be found
at http://www.ifac.org/auditing-assurance/clarity-center/clarified-standards;
The International Organisation of Supreme Audit Institutions (INTOSAI) can be found at
http://www.issai.org/composite-347.htm.
Auditors‘ objectivity: An unbiased mental attitude that allows internal auditors to perform
engagements in such a manner that they have an honest belief in their work product and that no
significant quality compromises are made. Objectivity requires internal auditors not to subordinate
their judgment on audit matters to that of others (IIA International Standards for the Professional
Practice of Internal Auditing 6).
of the marketing authorisation holder, in addition to considering the other factors included in
this list;
outcome of previous audits, e.g. has the area/process ever been audited (if not, then this may need
to be prioritised depending on criticality); if the area/process has previously been audited, the
audit findings* are a factor to consider when deciding when to re-audit the area/process,
including the implementation of agreed actions;
identified procedural gaps relating to specific areas/processes;
other information relating to compliance* with legislation and guidance, for example:
- for national medicines authorities: information from compliance* metrics from complaints,
from external sources, e.g. audits/assessments of the national medicines authority that may
be conducted by external bodies;
- for marketing authorisation holders: information from compliance* metrics from
inspections see Module III, from complaints, from other external sources, e.g. audits;
other organisational changes that could negatively impact on the area/process, e.g. if a change
occurs to a support function (such as information technology support) this could negatively
impact upon pharmacovigilance activities.
IV.B.2.3.2. Reporting
The findings* of the auditors should be documented in an audit report and should be communicated
to management in a timely manner. The audit process should include mechanisms for
communicating the audit findings* to the auditee* and receiving feedback, and reporting the audit
findings* to management and relevant parties, including those responsible for pharmacovigilance
systems, in accordance with legal requirements and guidance on pharmacovigilance audits. Audit
findings should be reported in line with their relative risk level and should be graded in order to
indicate their relative criticality to risks impacting the pharmacovigilance system, processes and
parts of processes. The grading system should be defined in the description of the quality system
for pharmacovigilance, and should take into consideration the thresholds noted below which would
be used in further reporting under the legislation as set out in section IV.C.2:
critical is a fundamental weakness in one or more pharmacovigilance processes or
practices that adversely affects the whole pharmacovigilance system and/or the rights,
safety or well-being of patients, or that poses a potential risk to public health and/or
represents a serious violation of applicable regulatory requirements.
major is a significant weakness in one or more pharmacovigilance processes or
practices, or a fundamental weakness in part of one or more pharmacovigilance
processes or practices that is detrimental to the whole process and/or could potentially
adversely affect the rights, safety or well-being of patients and/or could potentially
pose a risk to public health and/or represents a violation of applicable regulatory
requirements which is however not considered serious.
minor is a weakness in the part of one or more pharmacovigilance processes or
practices that is not expected to adversely affect the whole pharmacovigilance system
or process and/or the rights, safety or well-being of patients.
Issues that need to be urgently addressed should be communicated in an expedited manner to the
auditee*‘s management and the upper management.
IV.C.3. Confidentiality
Documents and information collected by the internal auditor should be treated with appropriate
confidentiality and discretion, and also respect national legislation on the protection of individuals
with regard to the processing of personal data and on the free movement of such data.
GVP: Modules
V.A. Introduction
It is recognised that at the time of authorisation, information on the safety of a medicinal product is
relatively limited. This is due to many factors including the relatively small numbers of subjects in
clinical trials compared with the intended treatment population, restricted population in terms of
age, gender and ethnicity, restricted co-morbidity, restricted co-medication, restricted conditions of
use, relatively short duration of exposure and follow up, and the statistical problems associated with
looking at multiple outcomes.
A medicinal product is authorised on the basis that in the specified indication(s), at the time of
authorisation, the risk-benefit balance is judged to be positive for the target population. A typical
medicinal product will have multiple risks associated with it and individual risks will vary in terms
of severity, effect on individual patients and public health impact. However, not all actual or
potential risks will have been identified at the time when an initial authorisation is sought and many
of the risks associated with the use of a medicinal product will only be discovered and characterised
post-authorisation. Planning of the necessary pharmacovigilance activities to characterise the safety
profile of the medicinal product will be improved if it is more closely based on specific issues
identified from pre- or post-authorisation data and from pharmacological principles.
However, the purpose of risk identification and characterisation is to allow for risk minimisation or
mitigation wherever possible. Therefore, risk management has three stages which are inter-related
and re-iterative:
1. characterisation of the safety profile of the medicinal product including what is known and not
known;
2. planning of pharmacovigilance activities to characterise risks and identify new risks and
increase the knowledge in general about the safety profile of the medicinal product;
3. planning and implementation of risk minimisation and mitigation and assessment of the
effectiveness of these activities.
Historically, risk management systems for medicinal products for human use was based solely on
managing risks. However, when considering how to maximise, or indeed assess, the risk-benefit
balance, risks need to be understood in the context of benefit. In assessing the risk-benefit balance at
the time of authorisation, the assumption is made that these benefits and risks apply to the whole
target population. However, there may be subsets of patients for whom the risk is greater than that
for the target population as a whole, or in whom the benefit may not be as great. In addition, efficacy
in the clinical trial setting may not reflect the true effectiveness of the medicinal product in everyday
medical practice and so the risk-benefit balance of a medicinal product as assessed at the time of
authorisation will inevitably change post-authorisation. Both post-authorisation safety studies and
post-authorisation efficacy studies may be a condition of the marketing authorisation in certain
circumstances and for these studies they shall be included in the risk management plan (RMP).
Risk management is a global activity. However, because of differences in indication and healthcare
systems, target populations may be different across the world and risk minimisation activities will
need to be tailored to the system in place in a particular country or global region. In addition,
differences in disease prevalence and severity, for example, may mean that the benefits of a
medicinal product may also vary between regions. Therefore a product may have different versions
of a RMP for each region although there will be core elements which are common to all. For
example much of the safety specification will be the same regardless of where the medicinal product
is being used but the epidemiology of the disease may vary between e.g. Africa and Europe, and
there may be additional or fewer safety concerns depending upon the target population and
indication.
Risk management, is applicable to medicinal products at any point in their lifecycle. However, this
module concentrates on peri- and post-authorisation risk management.
The risks addressed in this guidance are those related to non-clinical and clinical safety. In addition,
quality issues may be relevant if they impact on the safety and/or efficacy of the product. Where the
disposal of the product might pose a particular risk because of remaining active substance (e.g.
patches) this should also be addressed.
Although this module includes the principles of risk minimisation, and details of routine risk
minimisation measures, more detail on, in particular, additional risk minimisation tools and the
measurement of the effectiveness of risk management can be found in Module XVI.
V.B.1. Terminology
Identified risk
An untoward occurrence for which there is adequate evidence of an association with the medicinal
product of interest. Examples include:
an adverse reaction adequately demonstrated in non-clinical studies and confirmed by clinical
data;
an adverse reaction observed in well-designed clinical trials or epidemiological studies for which
the magnitude of the difference compared with the comparator group, on a parameter of interest
suggests a causal relationship;
an adverse reaction suggested by a number of well-documented spontaneous reports where
causality is strongly supported by temporal relationship and biological plausibility, such as
anaphylactic reactions or application site reactions.
In a clinical trial, the comparator may be placebo, active substance or non-exposure.
Potential risk
An untoward occurrence for which there is some basis for suspicion of an association with the
medicinal product of interest but where this association has not been confirmed. Examples include:
toxicological findings seen in non-clinical safety studies which have not been observed or
resolved in clinical studies;
adverse events observed in clinical trials or epidemiological studies for which the magnitude of
the difference, compared with the comparator group (placebo or active substance, or unexposed
group), on a parameter of interest raises a suspicion of an association, but is not large enough to
suggest a causal relationship;
a signal arising from a spontaneous adverse reaction reporting system;
an event known to be associated with other active substances within the same class or which
could be expected to occur based on the properties of the medicinal product.
Missing information
Gaps in knowledge about a medicinal product, related to safety or use in particular patient
populations, which could be clinically significant..
Examples of missing information include populations not studied (e.g. pregnant women or patients
with severe renal impairment) or where there is a high likelihood of off-label use.
Safety concern
An important identified risk, important potential risk or missing information.
However, the actions and responsibilities within each step of the cycle will vary according to
whether the stakeholder is an applicant/marketing authorisation holder, medicines authority,
healthcare professional or patient. Other players may be involved in risk-benefit management such
as: patient organisations, learned societies, health economists, health authorities, national safety
organisations, environmental advisors, occupational health professionals and pharmaceutical
distributors but their roles will usually be smaller and complementary to that of the main players.
For applicants/marketing authorisation holders and medicines authorities in the Arab Countries,
there should be specific mention of risk management in the national legislation. In the Arab
Countries, the primary document and process for risk management adheres to the principles in the
International Conference for Harmonisation (ICH) Guideline E2E on Pharmacovigilance Planning.
Some other territories may have local legislation enshrining either risk management in general or
adopting the specific ICH E2E guidance or have developed local guidance. For healthcare
professionals, product information, medical treatment guidelines and any materials produced by
marketing authorisation holders, medicines authority will direct prescribing, dispensing, treatment
and management of both benefit and risks. For patients, the majority of medicinal products will be
prescribed by doctors and dispensed by pharmacists so that management of benefits and risks will
primarily involve complying with treatment schedules and recommendations, being aware of
important risks and what actions to take, and reporting to their doctor, pharmacist, and national
medicines authority any untoward effects. However, in some countries patients may buy medicines
directly without guidance from healthcare practitioners so will need to understand the potential
benefits and risks of the product and what measures they need to comply with to use the medicine
safely and effectively. Whatever the setting, patients who understand the potential benefits and
risks of a medicinal product are better equipped to decide whether or not to be treated and to comply
with suggested risk minimisation activities.
expertise in communication and, where appropriate, patients and/or healthcare professionals. Since
a risk management plan is primarily a pharmacovigilance document, ideally the production
of it should be managed by personnel with appropriate pharmacovigilance training in either
the pharmacovigilance or regulatory departments, depending upon company structure.
Regardless of who prepares the RMP, the responsibility for the content and accuracy of the RMP
remains with the marketing authorisation applicant/holder who should ensure oversight by someone
with the appropriate scientific background within the company.
Further guidance on individual risk minimisation activities is provided in Module XVI.
the medicinal product, and the need for post-authorisation safety data. This proportionality can be
achieved in different ways: by reducing the number of modules which need to be submitted for
products meeting certain conditions (such as well-established products/generics see table V.3), and
by ensuring that requirements for post-authorisation studies and risk minimisation activities reflect
the important risks and important uncertainties of the product.
Where a RMP concerns more than one medicinal product, a separate RMP part VI must be
provided for each medicinal product.
Information should be provided in enough detail to enable an assessor to understand the issues being
presented. Unless specifically mentioned in this guidance, cross references to other parts of the
dossier should be avoided since it is intended that the RMP should be a largely stand-alone
document that is a scientific synopsis of the relevant parts of the dossier, emphasising the important
clinically relevant facts. To aid consistency between the information provided in the common
technical document (CTD) and the RMP, the table below indicates the location of information in the
CTD is summarised for the RMP:
RMP CTD
Copies of literature referenced in the RMP should be included in RMP annex 12.
mentioned. The RMP is part of the scientific dossier of a product and as such should be
scientifically based and not be promotional.
In consistency with the European categorization of medicinal products; certain products for human
medicinal use are categorised as advanced therapy medicinal products (ATMPs). . These products
are broadly comprise:
gene therapy medicinal products;
somatic cell therapy medicinal products;
tissue engineered products.
Because of the nature of these products, risks may occur which are not normally a consideration
with other medicinal products including risks to living donors, risks of germ line transformation and
transmission of vectors. For this reason, for ATMPs, RMP module VII Identified and potential risks
(ATMP) should replace RMP module VII Identified and potential risks as this provides greater
flexibility in consideration of the additional risks.
the product is likely to be used i.e. both labelled and off-labelled use), and outstanding safety
questions which warrant further investigation to refine understanding of the risk-benefit balance
during the post-authorisation period. In the RMP, the safety specification will form the basis of the
pharmacovigilance plan, and the risk minimisation plan.
The safety specification consists of eight RMP modules of which RMP modules SI-SV, SVII and
SVIII correspond to safety specification headings in ICH-E2E. RMP module SVI includes
additional elements required to be submitted in the Arab Countries.
Module SI Epidemiology of the indication(s) and target population(s)
Module SII Non-clinical part of the safety specification
Module SIII Clinical trial exposure
Module SIV Populations not studied in clinical trials
Module SV Post-authorisation experience
Module SVI Additional requirements for the safety specification in the Arab country
concerned
Module SVII Identified and potential risks
Module SVIII Summary of the safety concerns
RMP modules SIII–SV form the ―Limitations of the human safety database‖ part of the ICH-E2E
safety specification and these, with the addition of RMP modules SI and SVII form the clinical part
of the safety specification. RMP modules SVI and the ATMP version of SVII are EU specific
although the topics may apply in any territory; therefore; they are adopted in this guideline and
required in the Arab Countries.
The applicants/marketing authorisation holders should follow the structure of elements provided
below when compiling the safety specification. The safety specification can include additional
elements, depending on the nature of the product and its development programme. Elements which
might need to be incorporated include:
quality aspects if relevant in relation to the safety and efficacy of the product;
the disposal of the product where it might pose a particular risk because of remaining
active substance (e.g. patches);
innovative pharmaceutical forms; or
use with a medical device.
indication(s) may vary across regions), but the emphasis should be on the epidemiology in the Arab
County concerned of the proposed indication.
Information should be provided on the important co-morbidities in the target population. For
example: if a medicinal product is intended for treating prostate cancer, the target population is
likely to be men over the age of 50 years. Men over the age of 50 are also at risk of myocardial
infarction. To identify whether a medicinal product might be increasing the risk of myocardial
infarction, it is important to know how many cases would be expected amongst prostate cancer
patients (ideally) or men in the same age group, not taking the medicinal product. Estimation of the
risk in the target population, as compared with the same age/sex group in the general population
may be particularly important if the disease itself increases the risk of a particular adverse event.
The RMP should include a statement of the intended purpose and impact of the product e.g. whether
it is intended to prevent disease, to prevent particular serious outcomes due to a condition or to
reduce progression of a chronic disease.
blinded trial population only and all clinical trial populations.) Stratifications would normally
include:
age and gender;
indication;
dose;
racial origin (see also V.B.8.4).
Duration of exposure should be provided either graphically by plotting numbers of patients against
time or in tabular format.
The exposure of special populations (pregnant women, breast-feeding women, renal impairment,
hepatic impairment, cardiac impairment, sub-populations with relevant genetic polymorphisms,
immuno-compromised) should be provided as appropriate. The degree of renal, hepatic or cardiac
impairment should be specified as well as the genetic polymorphism.
The categories above are only suggestions and tables/graphs should be tailored to the product. For
example, indication may not be a relevant stratification for a medicinal product where only one
indication has been studied, and route of administration, number of courses/immunisations or repeat
administrations may be important categories to be added.
When presenting age data, categories should be chosen which are relevant to the target population.
Broad artificial divisions which are not clinically relevant, such as <65 and >65, should be avoided.
Paediatric data should be divided by categories (e.g. ICH-E11); similarly the data on elderly patients
should be considered for stratification into categories such as 65-74, 75-84 and 85+, although the
age strata should reflect that of the target population. For teratogenic drugs, stratification into age
categories relating to childbearing potential might be appropriate for the female population.
Unless clearly relevant, data should not be presented by individual trial but should be pooled. Totals
should be provided for each table/graph as appropriate. Where patients have been enrolled in more
than one trial (e.g. open label extension study following a trial) they should only be included once in
the age/sex/ethnic origin tables. Where differences in the total numbers of patients arise between
tables, the tables should be annotated to reflect the reasons for discrepancy.
When the RMP is being submitted with an application for a new indication, a new pharmaceutical
form or route, the clinical trial data specific to the application should be presented separately at the
start of the module as well as being included in the summary tables (as described above)
representing pooled data across all indications.
between target populations and those exposed in clinical trials it should be noted that some
differences may arise through trial setting (e.g. hospital or general practice) rather than through
explicit inclusion/exclusion criteria.
The implications, with respect to predicting the safety of the product in the marketplace, of any of
these populations with limited or no research should be explicitly discussed. In addition, the
limitations of the database with regard to the detection of adverse reactions due to:
number of patients studied;
cumulative exposure (e.g. specific organ toxicity);
long term use (e.g. malignancy)
should be discussed. Where the missing information could constitute an important risk to the target
population, it should be included as a safety concern in RMP module SVIII.
Populations to be considered for discussion should include (but might not be limited to):
Paediatric population
Children (from birth to 18 years with consideration given to the different age categories as per
ICH-E11, or, if justified, to other developmentally meaningful groups i.e. taking into account
specific organ maturation). If paediatric development has been limited to certain age categories
then the implications for other paediatric age groups should also be discussed.
Elderly population
Implications for use in patients over the age of 65 should be discussed – with appropriate
consideration given to use in the older end of the age spectrum. The effects of particular
impairments, e.g. renal, hepatic, or of concomitant disease or medication will be discussed
mainly in the appropriate sections below, but discussion in this section should reflect the fact that
in the elderly population many of these factors may co-exist. The cumulative effect of multiple
impairments and multiple medications should be discussed. Consideration of whether particular
laboratory screening should be performed routinely before use of the medicinal product(s) in the
elderly should be discussed. In particular any adverse reactions which might be of special
concern in the elderly e.g. dizziness or central nervous system effects should be explored.
Pregnant or breast-feeding women
If the target population includes women of child-bearing age, the implications for pregnancy
and/or breast-feeding should be discussed. If the medicinal product is not specifically for use
during pregnancy, any pregnancies which have occurred during the developmental programme
and their outcomes should be discussed. For products where pregnancy should be avoided for
safety reasons, , the discussion on pregnancy should also include an analysis of the reasons why
the contraceptive measures in place during the clinical trials failed and the implications for use in
the less controlled conditions of everyday medical practice.
Patients with hepatic impairment
Patients with renal impairment
Patients with other relevant co-morbidity (e.g. cardiovascular or immunucompromised including
organ transplant patients)
Patients with disease severity different from that studied in clinical trials
Any experience of use in patients with different disease severities should be discussed,
particularly if the proposed indication is restricted to those patients with a specific disease
severity.
Sub-populations carrying known and relevant genetic polymorphism
The extent of pharmacogenetic effects and the implications on genetic biomarker use in the
target population should be discussed. Where a proposed drug indication constitutes patients
with or without specific genetic markers, or the clinical development programme has been in
patients with a specific mutation, the marketing authorisation holder should discuss the
implications of this for the target population and explore whether use in patients with an
unknown or different genotype could constitute a safety concern.
If a potentially clinically important genetic polymorphism has been identified but not fully
studied in the clinical development programme, this should be considered as missing information
and/or a potential risk. This should be reflected in the safety specification and pharmacovigilance
plan. Whether it is included as a safety concern for the purposes of risk minimisation will depend
upon the importance of the possible clinical implications.
Patients of different racial and/or ethnic origins
Genetic variants can influence pharmacodynamics and pharmacokinetics, and subsequently
affect the efficacy and/or safety of the administered drug. Inter-ethnic differences in drug
efficacy and safety have been observed in different ethnic groups due to e.g. genetic
polymorphisms.
One example of such inter-ethnic differences is the variation in frequency of the HLA-B*1502
allele. This allele is strongly associated with the occurrence of severe cutaneous adverse
reactions to carbamazepine and has a prevalence of about 10% in some Asian populations, whilst
the prevalence of the allele is negligible in those of European descent. This is why genomic
testing is recommended for patients of some Asian origins when carbamazepine use is planned,
while this testing will not make sense for a patient who is of European descent.
Major inter-ethnic differences in pharmacokinetics of drugs may also occur due to types and/or
frequencies of gene variants coding for drug metabolising enzymes. The consequences of these
inter-ethnic differences could be that the proportion of subjects with particular beneficial effects
or adverse reactions varies, leading to different risk-benefit balances and specific
recommendations in these ethnic populations.
Furthermore, efficacy in patients may be affected by racial origin. One example is that ACE
inhibitors are less potent in black patients of African or Caribbean family origin than in white
patients.
Therefore, information on racial origin may be relevant and valuable for evaluation of efficacy
and safety and for preventing adverse reactions or improving benefits in the target population.
The experience of drug use in patients with different racial and/or ethnic origins should be
discussed including the implications on efficacy and safety, based on pharmacokinetics and
pharmacodynamics, in the target population. If it is likely that efficacy or safety may be affected
by race or ethnicity, consideration should be given to including this either as a safety concern or
as a topic for inclusion in RMP Part IV. Consideration should also be given as to whether
post-authorisation efficacy and/or safety studies are necessary.
V.B.8.5.1. RMP module SV section “Action taken by regulatory authorities and/or marketing
authorisation holders for safety reasons”
List any significant regulatory action (including those initiated by the marketing authorisation
holder), in any market, taken in relation to a safety concern. Significant regulatory action would
include: a restriction to the approved indication, a new contra-indication, a new or strengthened
warning in section 4.4 of the SPC (or equivalent) or any action to suspend or revoke a marketing
authorisation. This list should be cumulative, and specify the country, action taken and the date as
appropriate. Roll-out in multiple countries of a new safety statement initiated by the MAH can be
presented as one action.
When the RMP is updated, a brief description of the reasons leading to any significant actions since
the last submission of the RMP should be provided. It may be appropriate to add comments if the
regulatory action taken is not applicable to certain products/formulations as authorised in the Arab
Country concerned.
If the drug has different routes of administration, e.g. subcutaneous or oral, exposure data should be
presented separately, where possible. Arabian medicines authorities may request additional
stratification of exposure data, e.g. exposure in age groups or within different approved indications.
However, if the drug is used in different indications with different dosing schedules or other
delineating factors suitable for stratification, marketing authorisation holders should consider
routinely providing such data where possible.
A more accurate breakdown of drug exposure based on market research should be provided where
possible.
If a drug utilisation study has been performed, for reimbursement or other reasons, the results, as
they reflect use in the real world setting, should be provided.
V.B.8.5.3. RMP module SV section “Post-authorisation use in populations not studied in clinical
trials”
Where there are data on post-authorisation use in the special populations identified in RMP module
SIV as having no or limited exposure, estimation of the numbers exposed and the method of
calculation should be provided whether or not the usage is on- or off-label. For paediatric use, cross
reference may be made to RMP section ―Specific paediatric issues‖ in RMP module SVI (see
V.B.8.6.6.). Information on the safety profile of the medicinal product in these special populations,
as compared with the rest of the target population, should also be provided. In particular, any
information regarding an increased or decreased benefit in a special population should be provided.
Any special populations found to be at an increased or decreased risk in relation to a particular
safety concern should be discussed under the specific risk in RMP module SVII but reference
should be made in this section as to which risks and populations are affected.
V.B.8.6. RMP module SVI “Additional requirements for the safety specification”
Some safety topics were not included in ICH-E2E but are thought to be of particular interest due to
either legislation or prior experience of a safety issue.
V.B.8.6.1. RMP module SVI section “Potential for harm from overdose”
Special attention should be given to medicinal products where there is an increased risk of harm
from overdose, whether intentional or accidental. Examples include medicinal products where there
is a narrow therapeutic margin or potential for major dose-related toxicity, and/or where there is a
high risk of intentional overdose in the treated population (e.g. in depression). Where harm from
overdose has occurred during clinical trials this should be explicitly mentioned. The potential for
harm from overdose should be discussed in this section and, where appropriate, overdose should be
included as a safety concern in RMP module SVIII and appropriate risk minimisation proposed in
RMP part V.
V.B.8.6.2. RMP module SVI section “Potential for transmission of infectious agents”
The applicant/marketing authorisation holder should discuss the potential for the transmission of an
infectious agent. This may be because of the nature of the manufacturing process or the materials
involved. For vaccines, any potential for transmission of live virus should be discussed. For
advanced therapy medicinal products a cross-reference to RMP module SVII (ATMP) may be
made.
V.B.8.6.3. RMP module SVI section “Potential for misuse for illegal purposes”
The potential for misuse for illegal purposes should be considered. Misuse, as defined in GVP
Module VI, refers to situations where the medicinal product is intentionally and inappropriately
used not in accordance with the authorised product information. Misuse for illegal purposes has the
additional connotation of an intention of misusing the medicinal product to cause an effect in
another person. This includes, amongst others: the sale, to other people, of medicines for
recreational purposes and use of a medicinal product to facilitate assault. If appropriate, the means
of limiting this, e.g. by the use of colorants and/or flavourings in the dosage form, limited pack size
If during the post-marketing period it becomes apparent that adverse reactions are occurring as a
result of medication errors, this topic should be discussed in the updated RMP and ways of limiting
the errors proposed.
If the formulation or strength of a product is being changed, where appropriate, medication error
should be included as a safety concern and the measures that the marketing authorisation holder will
put in place to reduce confusion between old and new ―product‖ should be discussed in the risk
minimisation plan. Similarly, it may be appropriate to discuss risk minimisation activities in relation
to changes to the presentation, pack size, route of administration or release characteristics of the
medicinal product.
If the product is to be administered with a medical device (integrated or not), consideration should
be given to any safety concerns which could represent a risk to the patient (medical device
malfunction).
specific paediatric aspects will be addressed and all paediatric investigation plan recommendations
considered. Cross-reference may be made to RMP modules SIV and SVII and SVII.
Potential for paediatric off-label use
If the disease or disorder which is being treated or prevented is found in the paediatric population,
and the product is not authorised in all paediatric age groups, the potential for off-label paediatric
use in the non-authorised age groups should be discussed. If there are limited treatment options it
should not be assumed that clinicians will adhere to the labelled indication so it is important that
potential paediatric issues are discussed. Any actual use should be discussed in RMP module SV
section ―Non-study post-authorisation exposure‖ (see V.B.8.5.2.) and in RMP module SV section
―Post-authorisation use in populations not studied in clinical trials‖ (see V.B.8.5.3.).
V.B.8.7.2. RMP module SVII section “Recent study reports with implications for safety
concerns”
Study reports (either interim or final, from whichever type of study), since the last RMP, which
contain results which have a significant impact on an existing safety concern should be discussed
here. The conclusions should be incorporated into the other sections of the safety specification as
appropriate (e.g. RMP module SII; section V.B.8.7.3; V.B.8.7.4; V.B.8.7.5; RMP Module SVI and
RMP Module SVIII
V.B.8.7.3. RMP module SVII section “Details of important identified and potential risks from
clinical development and post-authorisation experience”
This RMP section should provide more information on the important identified and potential risks.
This RMP section should be concise and should not be a data dump of tables or lists of adverse
reactions from clinical trials, or the proposed or actual contents of section 4.8 of the summary of
is a risk factor. Where appropriate, the period of major risk should be identified. Identified risk
incidence rates should be presented for the whole population and for relevant population categories.
For important identified risks, the excess (relative incidence compared to a specified comparator
group) should be given. Time to event data should be summarised using survival techniques.
Cumulative hazard functions may also be used to represent the cumulative probability of occurrence
of an adverse reaction in the presence of competing events.
For potential risks, the background incidence/prevalence in the target population(s) should be
provided.
For most RMPs involving single products, risks which relate specifically to an indication or
formulation can usually be handled as individual safety concerns, e.g. accidental intravenous
administration could be a safety concern in a single product with both oral and subcutaneous forms.
For RMPs covering multiple products where there may be significant differences in the identified
and potential risks for different products, it may be appropriate to categorise the risks to make it
clearer which risks relate to which product. Headings which could be considered include:
Risks relating to the active substance
This would include important identified or potential risks which are common to all formulations,
routes of administration and target populations. It is likely that most risks will fall into this
category for the majority of products.
Risks related to a specific formulation or route of administration
Examples might include an RMP with two products: one a depot intramuscular formulation and
the other an oral formulation. Additional concerns relating to accidental intravenous
administration clearly would not be applicable to the oral product.
Risks relating to a specific target population
The paediatric population is an obvious example of a target population where there may be
additional risks relating to physical, mental and sexual development which would not be relevant
to a product intended solely for adult patients.
Risks associated with switch to non-prescription status.
Division of identified and potential risks using headings should only be considered when the risks
clearly do not apply to some products and lack of separation could cause confusion.
V.B.8.7.4. RMP module SVII section “Identified and potential interactions including food-drug,
drug-drug interactions and drug-herbal interactions”
Identified and potential pharmacokinetic and pharmacodynamic interactions should be discussed in
relation to both the treatments for the condition, but also in relation to commonly used medications
in the target population. For each, the evidence supporting the interaction and possible mechanism
should be summarised, and the potential health risks posed for the different indications and in the
different populations should be discussed. Interactions which are important clinically should be
included as a safety concern in RMP module SVIII ―Summary of the safety concerns.‖
V.B.8.8. RMP module SVII “Identified and potential risks (ATMP version)”
Advanced therapy medicinal products (ATMPs) because of their nature may have specific risks that
are usually not applicable to other non-advanced therapy medicinal products (use as a guide: EMA
Guideline on Safety and Efficacy Follow-up – Risk Management of Advanced Therapy Medicinal
Products). For this reason, for ATMPs, this ATMP specific version of RMP module replaces the
standard RMP module SVII.
Although not all of the risks listed in section V.B.8.8.3. are unique to ATMPs or applicable to all
ATMPs, they represent the most relevant ones which need to be considered.
V.B.8.8.1. RMP module SVII section “Newly identified safety concerns (ATMP)”
Safety concerns (important identified and important potential risks) identified since the last
submission of the RMP should be listed here and further discussed in the appropriate section below.
The source of the safety concern should be stated, whether it is an important identified or important
potential risk and whether new studies or risk minimisation activities are proposed (with further
details in the appropriate RMP parts).
V.B.8.8.2. RMP module SVII section “Recent study reports with implications for safety concerns
(ATMP)”
Study reports (either interim or final), since the last RMP, which contain results which have a
significant impact on an existing safety concern should be discussed here. The conclusions should
be incorporated into the other sections of the safety specification as appropriate (e.g. RMP module
SII; section V.B.8.8.3; RMP Module SVI and RMP Module SVII).I.
V.B.8.8.3. RMP module SVII section “Details of important identified and potential risks
(ATMP)”
This section should provide more information on the most important identified and potential risks.
This section should be selective and should not be a data dump of tables or lists of adverse reactions
from clinical trials, or the proposed or actual contents of section 4.8 of the summary of product
characteristics (SmPC).
What constitutes an important risk will depend upon several factors including the impact on the
individual, the seriousness of the risk and the impact on public health. Normally, any risk which is
clinically important and is/is likely to be included in the warnings and precautions section of the
summary of product characteristics should be included here. In addition, risks, which, whilst not
normally serious enough to require specific warnings or precautions but which occur in a significant
proportion of either the patient or donor, affect the quality of life, and which could lead to serious
consequences if untreated should also be considered for inclusion. The additional risks specific to
ATMPs which should be considered for discussion include:
risks to living donors, for instance:
- risks to living donors related to their conditioning prior to procurement (e.g.
immunosuppression, cytotoxic agents, growth factors);
- risks to living donors related to surgical/medical procedures used during or following
procurement, irrespective of whether the tissue was collected or not;
risks to patients related to quality characteristics of the product, in particular:
- species of origin and characteristics of cells (and related body fluids, biomaterials,
biomolecules) that are used during manufacturing, and the safety testing performed;
- characteristics of vectors for gene therapy medicinal products;
- biologically active substances used in manufacturing (e.g. enzymes, antibodies, cytokines,
sera, growth factors, antibiotics);
- quality assurance and characteristics of the finished product in terms of defined
composition, stability, biological activity, and purity with reference to non-physiologic
proteins and fragments thereof;
- risk related to transmissible diseases (e.g. viral, bacterial, parasitical infections and
infestations, but also malignant disease);
risks to patients related to the storage and distribution of the product, for instance:
- risks related to preservation, freezing and thawing;
- risks of breaking the cold chain or other type of controlled temperature conditions;
- risks related to stability of the product;
risks to patients related to administration procedures, for instance:
- biologically active substances used in preparation of the product prior to administration (e.g.
enzymes, antibodies, cytokines, sera, growth factors, antibiotics);
- risks related to conditioning of the patient;
- risks of related medical or surgical procedures (e.g. anaesthesia, infusion, transfusion,
implantation, transplantation or other application method);
concerns.
A safety concern is:
an important identified risk;
an important potential risk; or
missing information (see Annex I).
It is noted that the ICH definition of safety concern is: an important identified risk, important
potential risk or important missing information, i.e. includes the qualifier ―important‖ in relation to
missing information (see Annex IV, ICH-E2C(R2) Guideline). The ICH-E2E Guideline (see Annex
IV) uses the terms safety issue and safety concern interchangeably with the same definition for
safety concern as defined in the ICH-E2C(R2) Guideline.
The change of the term in Arab Countries, to name this concept ―missing information‖ rather than
―important missing information‖, is to be clear that in the Arab Country concerned a marketing
authorisation cannot be granted if there are unacceptable gaps in knowledge, a marketing
authorisation shall be refused if the quality, safety or efficacy are not properly or sufficiently
demonstrated.
For RMPs covering multiple products where there may be significant differences in the important
identified and important potential risks for different products, similar to the presentation of risks in
RMP module SVII, it may be appropriate to subdivide the summary of safety concerns under
specific headings with the relevant identified and potential risks under each heading. Headings
which could be considered include:
safety concerns relating to the active substance;
safety concerns related to a specific formulation or route of administration;
safety concerns relating to the target population;
risks associated with switch to non-prescription status.
Division of safety concerns by headings should only be considered when the risks clearly do not
apply to some products and inclusion as a single list could cause confusion.
SVIII of the safety specification. Early discussions between medicines authority concerned and the
marketing authorisation holder or applicant are recommended to identify whether, and which,
additional pharmacovigilance activities are needed. It is important to note that only a proportion of
risks are likely to be foreseeable and therefore signal detection, which is part of routine
pharmacovigilance, will be an important element in identifying new risks for all products.
Pharmacovigilance activities can be divided into routine pharmacovigilance activities and
additional pharmacovigilance activities. For each safety concern, the applicant/marketing
authorisation holder should list their planned pharmacovigilance activities for that concern.
Pharmacovigilance plans should be proportionate to the risks of the product. If routine
pharmacovigilance is considered sufficient for post-authorisation safety monitoring, without the
need for additional actions (e.g. safety studies) ―routine pharmacovigilance‖ should be entered
against the safety concern.
to achieve its stated scientific purpose, the applicant/marketing authorisation holder will be required
to modify it or remove it from the pharmacovigilance plan and resubmit the RMP.
Pharmacoepidemiology studies included in the pharmacovigilance plan should be designed and
conducted according to the national respective legislation in place and recommendations in the
internationally recognized guidelines [Guidelines for Good Pharmacoepidemiology Practices
(GPP) 7 and the ENCePP Guide on Methodological Standards in Pharmacoepidemiology 8 . For
studies involving children, refer to the EMA Guideline on Conduct of Pharmacovigilance for
Medicines Used by the Paediatric Population9] which is acknowledged in the Arab Countries. It is
highly recommended that expert advice is sought on the design and conduct of any studies –
whether by the scientific advice procedure or by consulting known experts in the appropriate field.
The responsibility for the scientific value of study protocols remains with applicants or marketing
authorisation holders, even if they have been previously discussed with the medicines authorities
concerned.
Further guidance on the conduct of post-authorisation safety studies (PASS) is given in Module
VIII.
For some safety concerns, additional pharmacovigilance activities other than
pharmacoepidemiology studies may be required, e.g. pharmacokinetic studies, clinical trials or
further pre-clinical work. The appropriate guidelines and legislation should be followed in the
conduct of these studies.
Protocols for studies in the pharmacovigilance plan should be provided in RMP annex 6 until
completion of the study and submission to the medicines authorities concerned of the final study
report. Changes to the protocol which do not affect milestones or due dates are not considered to be
updates to the RMP (see also Module VIII).
For studies conducted as an obligation of any of the Arab countries, the marketing authorisation
holder shall submit the study protocol in English (unless other language is requested by the
medicines authority in the Arab Country concerned). For other studies, if the study protocol or the
study report is written in another language, the marketing authorisation should facilitate access to
study information by including an English translation of the title, the abstract of the study protocol
and the abstract of the final study report (see Module VIII.)
Synopses of study reports from additional pharmacovigilance activities should be included in RMP
annex 9. The impact of the new data on the risk-benefit balance of the medicinal product should be
carefully assessed and the safety specification, pharmacovigilance plan and risk minimisation
measures updated accordingly.
7
International Society for Pharmacoepidemiology. Guidelines for good pharmacoepidemiology practices (GPP).
Pharmacoepidemiol Drug Saf. 2005; 14 (8): 589-595; available on the ISPE website
http://www.pharmacoepi.org/resources/guidelines_08027.cfm.
8
ENCePP Guide on Methodological Standards in Pharmacoepidemiology‖; available on http://www.encepp.eu
9
EMEA/CHMP/PhVWP/235910/2005; available on
http://www.ema.europa.eu/ema/index.jsp?curl=pages/regulation/document_listing/document_listing_000087.jsp&mid
=WC0b01ac0580025b90&jsenabled=true
c. Joint studies
If safety concerns apply to more than one medicinal product, the national medicines authority of the
Arab Country concerned shall s encourage the marketing authorisation holders concerned to
conduct a joint PASS. The conduct of a joint study may also be appropriate where there are limited
patients (rare diseases) or the adverse reaction is rare. The national medicines authority should
facilitate the agreement of the concerned marketing authorisation holders in developing a single
protocol for the study and conducting the study. Where the national medicines authority of the Arab
Country concerned decides to impose the same PASS on more than one marketing authorisation
holder and the concerned marketing authorisation holders have failed to agree a common protocol
within a reasonable period of time, as determined by the national medicines authority; the national
medicines authority, with input from the pharmacovigilance or relevant committee, may define
either a common core protocol or key elements within a protocol which the concerned marketing
authorisation holders will have to implement within a timescale laid down within the request.
Hence, the study would become a condition of the marketing authorisation and be reflected in the
RMP. In some circumstances, the encouragement to do joint studies may relate to a single active
substance where there are multiple marketing authorisation holders for the same active substance.
d. Registries
A registry is an organised system that uses observational methods to collect uniform data on
specified outcomes in a population defined by a particular disease, condition, or exposure. A
registry can be used as a data source within which studies can be performed. Entry in a registry is
generally defined either by diagnosis of a disease (disease registry) or prescription of a drug
(exposure registry).
Registries should ideally include a comparator group so a disease registry will usually be more
suitable than a registry confined to a specific product. However, if, an applicant/marketing
authorisation holder institutes a registry as part of an agreed RMP, the protocol for the registry will
allow all patients who are prescribed the active substance or who have the same disease, as
appropriate, to be entered in the registry. Entry to the registry should not be conditional on being
prescribed a product with a particular invented name or marketing authorisation holder unless there
are clear scientific reasons for this. The same applies to similar biological products.
Unless there are over-riding public health or scientific concerns which lead to mandatory inclusion
in a registry, refusal to enter a registry should not normally be a reason for refusing access to a
medicine.
V.B.9.3. RMP part III section “Action plans for safety concerns with additional
pharmacovigilance requirements”
For safety concerns with additional pharmacovigilance activities only, the action plan for each
safety concern should be presented according to the following structure:
safety concern;
proposed action(s);
individual objectives of proposed action(s) (i.e. what aspects of the safety concern they are
intended to characterise); and
for each action:
details of individual action;
- steps; and
- milestones (including expected dates).
As well as listing any additional pharmacovigilance activities under ―proposed actions,‖ protocols
(draft or otherwise) for any formal studies should be provided in RMP annex 6. Marketing
authorisation applicants/holders should also follow the requirements detailed in Module VIII, where
appropriate. It is recommended that the internationally recognized ENCePP Guide on
Methodological Standards in Pharmacoepidemiology10 including the ENCePP Checklist for Study
Protocols11, should be referred to when considering epidemiological protocol design.
10
ENCePP Guide on Methodological Standards in Pharmacoepidemiology‖; available on http://www.encepp.eu
11
ENCePP Checklist for Study Protocols‖; available on http://www.encepp.eu
product, or because they are specific obligations 12 in the context of a MA under exceptional
circumstances 13 . If the obligation is a non-interventional PASS, it will be subject to the
supervision as described in the national regulations of the Arab Country concerned.
The pharmacovigilance plan also includes studies that are conducted or financed by the marketing
authorisation holder to address particular safety concerns. These studies may be on-going or
planned, may have been requested by another regulatory authority or may have been suggested by
the marketing authorisation applicant/holder and agreed with the medicines authority of the Arab
Country concerned as forming part of the pharmacovigilance plan. They may also be conducted to
evaluate the effectiveness of risk minimisation activities.
Finally, the Pharmacovigilance Plan also has a role in providing an overview of studies which,
although not part of the formal agreed plan to identify and characterise specific safety concerns, the
national medicines authority needs to be aware of. These studies are typically requested
post-authorisation by a national medicines authority for reimbursement reasons e.g. drug utilisation
studies.
The summary table of the pharmacovigilance plan should provide clarity to all of these studies
taking into consideration the following:
Clinical interventional studies are subject to the requirements of national regulations in the Arab
Country concerned.
Non-clinical interventional studies are subject to the legal and ethical requirements related to the
protection of laboratory animals, and Good Laboratory Practice as appropriate.
Agreed upon studies in the RMP between MAH and any Arab Medicine authority is a condition
to maintain license of the product in the territory.
12
Specific obligations can only be imposed on marketing authorisations granted under exceptional circumstances (may
be NOT applicable in some Arab Countries, check the national regulations)
13
Exceptional circumstances is a type of marketing authorisation granted to medicines where the applicant is unable to
provide comprehensive data on the efficacy and safety under normal conditions of use, because the condition to be
treated is rare or because collection of full information is not possible or is unethical. (may be NOT applicable in some
Arab Countries, check the national regulations)
relapse, etc.);
a brief statement of the standard against which the medicine was judged;
number of patients in pivotal studies and treatment regimes;
results.
The following areas should be discussed briefly and the need for further studies post authorisation
evaluated:
the robustness of the endpoints on which the efficacy evaluation is based;
applicability of the efficacy data to all patients in the target population;
factors which might affect the efficacy of the product in everyday medical practice;
variability in benefits of treatment for sub populations.
For updates to the RMP, any subsequent data which impacts on efficacy should be mentioned and
its impact on the benefits of the medicinal product discussed.
b. Legal status
Controlling the conditions under which a medicinal product may be made available can reduce the
risks associated with its use or misuse. This can be achieved by controlling the conditions under
which a medicinal product may be prescribed, or the conditions under which a patient may receive a
medicinal product.
When a marketing authorisation is granted, it must include details of any conditions or restrictions
imposed on the supply or the use of the medicinal product, including the conditions under which a
medicinal product may be made available to patients. The conditions under which a medicinal
product is made available is commonly referred to as the ―legal status‖ of a medicinal product.
Typically it includes information on whether or not the medicinal product is subject to medicinal
prescription. It may also restrict where the medicinal product can be administered (e.g. in a hospital,
but see below) or by whom it may be prescribed (e.g. specialist).
For medicinal products only available on prescription, additional conditions may be imposed by
classifying medicinal products into those available only upon either a restricted medical
prescription or a special medical prescription.
Restricted medical prescription
This may be used to control who may initiate treatment, prescribe the medicinal product and the
14
http://ec.europa.eu/health/files/eudralex/vol-2/c/smpc_guideline_rev2_en.pdf
setting in which the medicine can be given or used. When considering classification of a medicinal
product as subject to restricted medical prescription, the following factors shall be taken into
account:
the medicinal product, because of its pharmaceutical characteristics or novelty or in the interests
of public health, is reserved for treatments which can only be followed in a hospital environment;
the medicinal product is used for the treatment of conditions which must be diagnosed in a
hospital environment or in institutions with adequate diagnostic facilities, although
administration and follow up may be carried out elsewhere; or
the medicinal product is intended for outpatients but its use may produce very serious adverse
reactions requiring prescription drawn up as required by a specialist and special supervision
throughout the treatment.
Although the use of legal status is not an activity that can be used directly by a marketing
authorisation applicant for the purposes of risk reduction, the marketing authorisation applicant
could request the medicines authority to consider a particular legal status and this is indicated in the
SmPC.
However, the definition of what constitutes a specialist is not uniform throughout the Arab
Countries so, in practice, the term ―specialist‖ is sometimes phrased in section 4.2 of the summary
of product characteristics (SmPC) as: ―treatment by a physician experienced in the treatment of <the
disease>‖. Although restricting to use in a hospital environment may in practice ensure that the
medicinal product is always prescribed by a specialist, this needs to be balanced against the
inconvenience to patients if they need to attend a hospital for every prescription. Care also needs to
be taken when considering where a medicinal product can be safely administered. For example the
term ―clinic‖ has different connotations depending upon the country. For this reason, the type of
equipment needed should be specified rather than a location: e.g. ―use in a setting where
resuscitation equipment is available.‖
Special medical prescription
For classification as subject to special medical prescription, the following factors shall be taken into
account:
the medicinal product contains, in a non-exempt quantity, a substance classified as a narcotic or a
psychotropic substance within the meaning of the international conventions in force, such as the
United Nations Conventions of 1961 and 1971; or
the medicinal product is likely, if incorrectly used, to present a substantial risk of medicinal
abuse, to lead to addiction or be misused for illegal purposes; or
the medicinal product contains a substance which, by reason of its novelty or properties, could be
considered as belonging to the group envisaged in the previous indent as a precautionary
measure.
Categorisation at Arab Country level
There is the possibility of implementing further sub-categories at Arab Country level which permits
each Arab Country to tailor the broad classifications described above to their national situation. The
definitions and therefore also the implementation varies in those Countries where the sub-categories
exist.
The majority of safety concerns may be adequately addressed by routine risk minimisation
activities. However, for some risks, routine risk minimisation activities will not be sufficient and
additional risk minimisation activities will be necessary.
experts, patients and healthcare professionals are consulted on the design and wording of
educational material and that, where appropriate, it is piloted before releasing for use.
The medicines authority of the Arab County concerned will agree the key elements of what should
be included in the educational material and these key elements will become, once agreed by the
medicines authority, a condition of the marketing authorisation. In addition, the final version of the
educational material will need to be approved by the national medicines authority to check that the
material contains the key elements in an appropriate design and format and is not promotional.
For public health reasons, applicants/marketing authorisation holders for the same active substance
may be required by the medicines authority to have educational material with as similar as possible
layout, content, colour and format to avoid patient confusion. This requirement may also be
extended to other patient material such as patient alert cards and patient monitoring cards. For this
reason, marketing authorisation applicants/holder are strongly recommended to avoid the use of
company logos or other trademarked or patented material in educational material.
Further extensive guidance on additional risk minimisation measures is provided in Module XVI.
minimisation measures and risk minimisation activities are used virtually synonymously in GVP.
The success of risk minimisation activities in delivering these objectives needs to be evaluated
throughout the lifecycle of a product to ensure that the burden of adverse reactions is minimised and
hence the overall risk-benefit balance is optimised.
When the RMP is updated, the risk minimisation plan should include an evaluation of the impact of
routine and/or additional risk minimisation activities as applicable. Such information may be
presented by region, if applicable/relevant. Results of any studies to assess the impact or other
formal assessment(s) of risk minimisation activities should be included when available. As part of
this critical evaluation, the marketing authorisation holder should make observations on factors
contributing to the success or weakness of risk minimisation activities. If a particular risk
minimisation strategy proves ineffective, or to be causing an excessive or undue burden on patients
or the healthcare system then alternative activities need to be put in place. The marketing
authorisation holder should always comment on whether additional or different risk minimisation
activities are needed for each safety concern.
In certain cases it may be judged that risk minimisation cannot control the risks to the extent
possible to ensure a positive risk-benefit balance and that the medicinal product needs to be
withdrawn either from the market or restricted to those patients in whom the benefits outweigh the
risks.
More extensive guidance on monitoring the effectiveness of risk minimisation activities is included
in Module XVI.
post-authorisation efficacy and risk minimisation measures will be included in this section.
There may also be a requirement for additional summaries of the RMP to be provided for inclusion
in these documents; this will be announced by each national medicines authority.
V.B.12.1. RMP part VI section “format and content of the summary of the RMP”
In situations where the RMP covers more than one product, a separate RMP part VI should be
prepared for each product. To present a balanced picture, the risks discussed in the RMP should be
put into context with a very concise and focused description of the benefits of the medicinal product.
The summary of the RMP part VI should contain the following information based on RMP part II
module SVIII and RMP parts III, IV and V:
Summary of safety concerns:
- Important identified risks,
- Important potential risks,
- Missing information,
Summary of risk minimisation activities by safety concern;
Planned post authorisation development plan;
Studies which are a condition of the marketing authorisation (see sections V.B.9.4 and
V.B.10.2)
Major Changes to the Risk Management Plan over time.
The information provided in each section should be brief and focused and in accordance with the
RMP templates.
In addition to the listing of the safety concerns in the above table; each safety concern should be
briefly described using the following tables.
For important identified risks include the frequency and severity of the safety concern for the
important identified risks and their preventability.
For important potential risks the reasons why it is thought to be a potential risk (e.g. toxicology in
animal study, known effect in other members of the pharmaceutical class) should be explained
together with the uncertainties, e.g. ―occurs in other medicinal products in the same class but was
not seen in the clinical trials for this medicinal product which studied 3,761 people‖.
Where there are safety concerns specific to a particular indication or population, or where an ATMP
is involved it may be appropriate to structure the risks by the headings suggested in module SVII
V.B.12.5. RMP part VI section “Summary of changes to the risk management plan
over time”
This table should provide a listing of all significant changes to the RMP in chronological order. This
should include, for example, the date and version number of the RMP when new safety concerns
were added or existing ones removed or changed, dates and version of the RMP when new studies
were added or finished, and a brief summary of changes to risk minimisation activities and the
associated dates these changes were agreed. Since changes to risk minimisation activities may
involve a variation, the date used for changes to risk minimisation activities should be that of the
decision by a national medicines authority. The date for safety concerns and studies should be the
date of the RMP in which they are first added.
trade name(s);
any explanatory comments.
RMP annex 4: Synopsis of on-going and completed clinical trial programme.
RMP annex 5: Synopsis of on-going and completed pharmacoepidemiological study programme.
RMP annex 6: Protocols for proposed and on-going studies in the section ―Summary table of
additional pharmacovigilance activities‖ in RMP part III.
RMP annex 7: Specific adverse event follow-up forms.
RMP annex 8: Protocols for proposed and on-going studies in RMP part IV.
RMP annex 9: Synopsis of newly available study reports for RMP parts III-IV.
RMP annex 10: Details of proposed additional risk minimisation activities (if applicable).
RMP annex 11: Mock up examples in English (unless other language is requested by the medicines
authority of the Arab Country concerned) of the material provided to healthcare
professionals and patients. For those materials directed to patients, in addition to
the English version, Arabic translation of the mock up shall be included as well.
RMP annex 12: Other supporting data (including referenced material).
V.B.14. The relationship between the risk management plan and the periodic
safety update report
The primary post-authorisation pharmacovigilance documents will be the RMP and the periodic
safety update report (PSUR). Although there is some overlap between the documents, the main
objectives of the two are different and the situations when they are required are not always the same.
Regarding objectives, the main purpose of the PSUR is integrated, post-authorisation risk benefit
assessment whilst that of the RMP is pre-and post-authorisation risk-benefit management and
planning. As such the two documents are complementary. Regarding submission, whereas for many
medicinal products, both documents will need to be submitted, for other medicinal products only
one will be required depending upon where the product is in its lifecycle. For this reason both
documents need to be ―stand-alone‖ but it is anticipated that certain modules may be common to
prevent duplication of effort.
The PSUR examines the overall safety profile as part of an integrated benefit-risk evaluation of the
medicinal product at set time periods and as such will consider the overall risk-benefit balance of the
medicinal product (and a much wider range of (suspected) adverse reactions). It is anticipated that
only a small proportion of these would be classified as important identified or important potential
risks and become a safety concern discussed within the RMP. Deciding to add an adverse reaction to
section 4.8 of the summary of product characteristics (SmPC) is not a sufficient cause per se to
include it as a safety concern in the RMP (see V.B.8.7.2.).
When a PSUR and a RMP are to be submitted together, the RMP should reflect the conclusions of
the accompanying PSUR. For example if a new signal is discussed in the PSUR and the PSUR
concludes that this is an important identified or important potential risk, this risk should be included
as a safety concern in the updated RMP submitted with the PSUR. The pharmacovigilance plan and
the risk minimisation plan should be updated to reflect the marketing authorisation holder‘s
proposals to further investigate the safety concern and minimise the risk.
V.B.14.1. Common modules between periodic safety update report and risk
management plan
The proposed PSUR and RMP modular format is intended to minimise duplication by enabling
common (sections of) modules to be utilised interchangeably across both reports. Common
(sections of) modules are identified in the following table.
Table V.3: Common sections between RMP and PSUR (may not be in identical format)
Part II, module SV – ―Post-authorisation experience‖, Section 3 – ―Actions taken in the reporting interval
section ―Regulatory and marketing authorisation for safety reasons‖
holder action for safety reason‖
Part II, module SV – ―Post-authorisation experience‖, Sub-section 5.2 – ―Cumulative and interval patient
section ―Non-study post-authorisation exposure‖ exposure from marketing experience‖
Part II, Module SVII – ―Identified and potential risks‖ Sub-section 16.4 – ―Characterisation of risks‖
Part II, module SVIII – ―Summary of the safety Sub-section 16.1 – ―Summary of safety concerns‖
concerns‖ (as included in the version of the RMP
which was current at the beginning of the PSUR
reporting interval)
a. Safety specification
Have all appropriate parts of the safety specification been included?
Have all appropriate data been reviewed when compiling the safety specification, i.e. are there
important (outstanding) issues from other sections of the dossier which have not been discussed
in the safety specification?
If parts of the target population have not been studied, have appropriate safety concerns in
relation to potential risks and missing information been included?
What are the limitations of the safety database and what reassurance does it provide regarding the
b. Pharmacovigilance plan
Are all safety concerns from the safety specification covered in the pharmacovigilance plan?
Are routine pharmacovigilance activities adequate or are additional pharmacovigilance activities
necessary?
Are the activities in the pharmacovigilance plan clearly defined and described and suitable for
identifying or characterising risks or providing missing information?
Are the safety studies which have been imposed by a medicines authority of the Arab country
concerned as conditions clearly identified?
If medication error is a safety concern, does the RMP include appropriate proposals to monitor
these?
Are the proposed additional studies necessary and/or useful?
When draft protocols are provided, are the proposed studies in the pharmacovigilance plan
adequate to address the scientific questions and are the studies feasible?
Are appropriate timelines and milestones defined for the proposed actions, the submission of
their results and the updating of the pharmacovigilance plan?
Has the marketing authorisation holder considered ways to reduce medication errors?
Has this been translated into appropriate product information (including device design where
appropriate) and pack design?
Are proposed risk minimisation activities appropriate and adequate?
Have additional risk minimisation activities been suggested and if so, are they risk proportionate
and adequately justified?
Are the methodologies for measuring and assessing the effectiveness of risk minimisation
activities well described and appropriate?
Have criteria for evaluating the success of additional risk minimisation activities been defined a
priori?
managing risks. However, when considering how to maximise, or indeed assess, the risk-benefit
balance, risks need to be understood in the context of benefit.
V.C.1. Legal basis for the implementation of risk management within the
Arab Countries
Each Arab Country should have –as appropriate- its national regulations for requirements in relation
to pharmacovigilance and in particular risk management.
product where the new treatment target population differs materially from the one for which
the medicinal product was previously authorised. This includes (but is not limited to): a new
disease area, a new age group (e.g. paediatric indication) or a move from severe disease to a
less severely affected population. It may also include a move from 2nd line or other therapy
or for an oncology product a change to the concomitant medication specified in the
indication.
at the request of the national medicines authority when there is a concern about a risk affecting
the risk-benefit balance;
with a submission of final study results impacting the RMP;
with a PSUR for medicinal product, when the changes to the RMP are a direct result of data
presented in the PSUR.
at the time of the renewal of the marketing authorisation if the product has an existing risk
management plan.
The need for a RMP or an update to the RMP should be discussed with the national medicines
authority, as appropriate, well in advance of the submission of an application involving a significant
change to an existing marketing authorisation.
An updated RMP should always be submitted if there is a significant change to the benefit-risk
balance of one or more medicinal products included in the RMP.
15
‗Generic medicinal product‘: shall mean a medicinal product which has the same qualitative and quantitative
composition in active substances and the same pharmaceutical form as the reference medicinal product, and whose
bioequivalence with the reference medicinal product has been demonstrated by appropriate bioavailability studies. The
different salts, esters, ethers, isomers, mixtures of isomers, complexes or derivatives of an active substance shall be
considered to be the same active substance, unless they differ significantly in properties with regard to safety and/or
efficacy. In such cases, additional information providing proof of the safety and/or efficacy of the various salts, esters or
derivatives of an authorised active substance must be supplied by the applicant. The various immediate-release oral
pharmaceutical forms shall be considered to be one and the same pharmaceutical form.
Part II-Module SV
Part II-Module SII
Part II-Module SI
Part VII
Part IV
Part VI
Part III
Part V
Part I
Similar biological
Generic medicine
Modified requirement
Please note that the naming and numbering of the RMP parts, modules & sections are standardized
thus should NOT be changed or renumbered due to the omission of un-required sections.
For Arab Countries applying eCTD, the RMP is submitted as PDF files within the eCTD
submission.
Marketing authorisation holders submit the RMP annex I in XML format. RMP annex I provides the
key information regarding the RMP in a structured electronic format. Applicable only in some Arab
Countries hence this annex should be submitted only upon request from the medicines authority of
the Arab Countries concerned. Further details will be announced by authorities who require such
annex. In Arab Countries who do not require this annex, it should be omitted (WITHOUT changing
the numbering of the following annexes).
Other submission requirements may also apply in some Arab Countries. Details of submission
requirements will be provided by each national medicines authority.
The initial RMP should be submitted as part of the initial marketing authorisation, or if required, for
those products that do not have an RMP, through the appropriate post-authorisation procedure.
Post-authorisation, submission of a new or updated RMP outside of another regulatory procedure
may constitute a variation. For details, refer to the national variation regulations or consult with the
national medicines authority in the Arab country concerned.
If, when preparing a PSUR, there is a need for consequential changes to the RMP as a result of new
safety concerns, or other data, then an updated RMP should be submitted at the same time. In this
case no stand-alone RMP variation is necessary.
Should only the timing for submission of both documents coincide, but the changes are not related
to each other, the RMP submission should be handled as a stand-alone variation (refer to the local
variation regulations).
When the RMP is updated, the risk minimisation plan should include an evaluation of the impact of
routine and/or additional risk minimisation activities as applicable (see V.B.11.4.).
For MAH/Applicant submitting EU RMP & its National Display; when the referenced EU RMP is
subject to update the National Display of RMP should be updated in accordance.
V.C.6. Procedure for the assessment of the risk management plan within the
national medicines authority
The regulatory oversight of RMPs for authorised products lies with the Pharmacovigilance
Department (and when appropriate) the pharmacovigilance committee of the national medicines
authority.
The national medicines authority may, on a case-by-case basis, consult with healthcare
professionals and patients during the assessment of RMPs to gather their input on proposed risk
minimisation measures.
In these circumstances (submitting the National Display and the EU RMP), the following
conditions apply:
When the referenced EU RMP is subject to update the National Display of RMP should be
updated in accordance.
Minor differences may exist between this guidance and the EU RMP, in this case
MAH/Applicant may be asked by the national medicines authority in the Arab Country
concerned to submit additional information, use different tables, and/or provide
clarification….etc.
The submitted EU RMP shall be the most updated version.
The EU RMP shall be submitted with its annexes and reference materials
Generally, it is required that all the risk management activities applied globally/in the EU to be
applied in the concerned Arab Country as well, especially the risk minimization measures
including the measurement of their effectiveness. Accordingly, all activities, action plans and
details especially the risk minimization ones (including the measurement of their effectiveness)
stated in the submitted EU RMP -although unjustifiably skipped in the ―National Display of the
RMP‖- are expected by default to apply to Arab Country concerned and the MAH is required to
adhere to them, EXCEPT otherwise clearly stated and justified by the MAH/Applicant in the
―National Display of the RMP‖ and agreed by the national medicines authority.
The purpose of the “National Display of the RMP” is:
to highlight to what extent the risk management activities proposed to be implemented nationally
adhere to the globally implemented plan and;
to provide justification for any difference (apart from what implemented in EU) whenever exist
including the needed national tailoring if any.
In addition it should include an assessment whether there are any additional national/
region-specific risks or not, describing the may be added activities to manage those additional
risks.
It provides good evidence that the LSR has clear understanding and commitment about the
activities that will be implemented on the national level and how they will be implemented.
GVP: Modules
VI.A. Introduction
VI.A.1. Scope
This Module addresses the requirements which are applicable to national medicines authorities in
Arab Countries and marketing authorisation holders as regards the collection, data management
and reporting of suspected adverse reactions (serious and non-serious) associated with medicinal
products for human use authorised in the Arab Countries. Recommendations regarding the
reporting of emerging safety issues or of suspected adverse reactions occurring in special situations
are also presented in this Module.
The guidance provided in this Module does not address the collection, management and reporting of
events or patterns of use, which do not result in suspected adverse reactions (e.g. asymptomatic
overdose, abuse, off-label use, misuse or medication error) or which do not require to be reported as
individual case safety report or as Emerging Safety Issues. This information may however need to
be collected and presented in periodic safety update reports for the interpretation of safety data or
for the benefit risk evaluation of medicinal products. In this aspect, guidance provided in Module
VII applies.
All applicable legal requirements detailed in this Module are referenced by the modal verb ―shall‖.
Guidance for the implementation of legal requirements is provided using the modal verb ―should‖.
VI.A.2. Definitions
The definitions provided hereafter shall be applied for the purpose of this Module. Some general
principles presented in the ICH-E2A and ICH-E2D guidelines16 should also be adhered to; they are
included as well in this chapter.
VI.A.2.1.1. Causality
In accordance with the ICH-E2A guideline, the definition of an adverse reaction implies at least a
reasonable possibility of a causal relationship between a suspected medicinal product and an
adverse event. An adverse reaction, in contrast to an adverse event, is characterised by the fact that a
16
http://www.ich.org/products/guidelines/efficacy/article/efficacy-guidelines.html
causal relationship between a medicinal product and an occurrence is suspected. For regulatory
reporting purposes, as detailed in the ICH-E2D guideline, if an event is spontaneously reported,
even if the relationship is unknown or unstated, it meets the definition of an adverse reaction.
Therefore all spontaneous reports notified by healthcare professionals, patients or consumers are
considered suspected adverse reactions, since they convey the suspicions of the primary sources,
unless the reporters specifically state that they believe the events to be unrelated or that a causal
relationship can be excluded.
b. Off-label use
This relates to situations where the medicinal product is intentionally used for a medical purpose
not in accordance with the authorised product information.
c. Misuse
This refers to situations where the medicinal product is intentionally and inappropriately used not
in accordance with the authorised product information.
d. Abuse
This corresponds to the persistent or sporadic, intentional excessive use of a medicinal product,
which is accompanied by harmful physical or psychological effects.
e. Occupational exposure
This refers to the exposure to a medicinal product, as a result of one‘s professional or
non-professional occupation.
medicinal product is authorised with a defined composition, all the adverse reactions suspected to
be related to any of the active substances being part of a medicinal product authorised in the Arab
Country concerned should be managed in accordance with the requirements presented in this
module. This is valid independently of the strengths, pharmaceutical forms, routes of
administration, presentations, authorised indications, or trade names of the medicinal product.
The guidance provided in this Module also applies, subject to amendments where appropriate, to
medicinal products supplied in the context of compassionate use (see VI.C.1.2.2). As the case may
be, this guidance may also apply to named patient use.
VI.A.2.4 Seriousness
As described in the ICH-E2A guideline, a serious adverse reaction corresponds to any untoward
medical occurrence that at any dose results in death, is life-threatening, requires inpatient
hospitalisation or prolongation of existing hospitalisation, results in persistent or significant
disability or incapacity, is a congenital anomaly/birth defect.
The characteristics/consequences should be considered at the time of the reaction to determine the
seriousness of a case. For example, life-threatening refers to a reaction in which the patient was at
risk of death at the time of the reaction; it does not refer to a reaction that hypothetically might have
caused death if more severe.
Medical judgement should be exercised in deciding whether other situations should be considered
as serious reactions. Some medical events may jeopardise the patient or may require an intervention
to prevent one of the above characteristics/consequences. Such important medical events should be
considered as serious 17 . The EudraVigilance Expert Working Group has co-ordinated the
development of an important medical event (IME) terms list based on the Medical Dictionary for
Regulatory Activities (MedDRA). This IME list aims to facilitate the classification of suspected
adverse reactions, the analysis of aggregated data and the assessment of the Individual Case Safety
Reports (ICSRs) in the framework of the day-to-day pharmacovigilance activities. The IME list is
intended for guidance purposes only and is available on the EudraVigilance web site 18 to
stakeholders who wish to use it for their pharmacovigilance activities; accordingly, this list is
acknowledged in the Arab Countries. The list is regularly updated in line with the latest version
of MedDRA.
17
Examples are provided in Section II.B of ICH E2A guideline.
18
(http://eudravigilance.ema.europa.eu/human/textforIME.asp ).
in Arab Countries).
The system should also be structured in a way that allows for reports of suspected adverse reactions
to be validated (see VI.B.2) in a timely manner and exchanged between medicines authorities and
marketing authorisation holders within the legal reporting time frame (see VI.B.7.1).
In accordance with the ICH-E2D guideline, two types of safety reports are distinguished in the
post-authorisation phase; reports originating from unsolicited sources and those reported as
solicited.
19
See VI. Appendix 2. for the detailed guidance on the monitoring of medical and scientific literature.
a marketing authorisation, and to bring them to the attention of the company safety department
as appropriate.
4. Reports of suspected adverse reactions from the scientific and medical literature, including
relevant published abstracts from meetings and draft manuscripts, should be reviewed and
assessed by marketing authorisation holders to identify and record ICSRs originating from
spontaneous reports or non-interventional post-authorisation studies.
If multiple medicinal products are mentioned in the publication, only those which are identified by
the publication's author(s) as having at least a possible causal relationship with the suspected
adverse reaction should be considered by the concerned marketing authorisation holder(s).
Valid ICSRs should be reported according to the modalities detailed in VI.B.7 and VI.B.8.
One case should be created for each single patient identifiable based on characteristics provided in
VI.B.2. Relevant medical information should be provided and the publication author(s) should be
considered as the primary source(s).
VI.B.1.1.4. Information on suspected adverse reactions from the internet or digital media
Marketing authorisation holders should regularly screen internet or digital media 20 under their
management or responsibility, for potential reports of suspected adverse reactions. In this aspect,
digital media is considered to be company sponsored if it is owned, paid for and/or controlled by the
marketing authorisation holder21. The frequency of the screening should allow for potential valid
ICSRs to be reported to the medicines authorities within the appropriate reporting timeframe based
on the date the information was posted on the internet site/digital medium. Marketing authorisation
holders may also consider utilising their websites to facilitate the collection of reports of suspected
adverse reactions (See VI.C.2.2.1)
If a marketing authorisation holder becomes aware of a report of suspected adverse reaction
described in any non-company sponsored digital medium, the report should be assessed to
determine whether it qualifies for reporting.
Unsolicited cases of suspected adverse reactions from the internet or digital media should be
handled as spontaneous reports. The same reporting time frames as for spontaneous reports should
20
Although not exhaustive, the following list should be considered as digital media: web site, web page, blog,
vlog, social network, internet forum, chat room, health portal.
21
A donation (financial or otherwise) to an organisation/site by a marketing authorisation holder does not
constitute ownership, provided that the marketing authorisation holder does not control the final content of the
site.
22
Local data privacy laws regarding patient‘s and reporter‘s identifiability might apply.
23
There is no suspected adverse reaction.
24
For further guidance on reporting of other duplicate ICSRs, refer to ICH-E2B(R2) guideline Section A.1.11
―Other case identifiers in previous transmission‖.
should be included in the ICSR. Guidance on the reporting of the medical confirmation of a case,
provided in ICH-E2B(R2) guideline Section A.1.14 (“Was the case medically confirmed, if not
initially from a healthcare professional?”), should be followed.
For solicited reports of suspected adverse reactions (see VI.B.1.2), where the receiver disagrees
with the reasonable possibility of causal relationship between the suspected medicinal product and
the adverse reaction expressed by the primary source, the case should NOT be downgraded to a
report of non-related adverse event. The opinions of both, the primary source and the receiver,
should be recorded in the ICSR.
The same principle applies to the ICSR seriousness criterion, which should NOT be downgraded
from serious to non-serious if the receiver disagrees with the seriousness reported by the primary
source.
25
For further guidance on reporting this information, refer to ICH-E2B (R2) guideline, Section A.1.14 (―Was the
case medically confirmed, if not initially from a healthcare professional?‖).
Pharmacovigilance for Vaccines for Pre-and Post-exposure Prophylaxis against Infectious Diseases
should also be followed as appropriate.
control procedures, which permit for the validation against the original data or images thereof. In
this aspect, the source data (e.g., letters, emails, records of telephone calls that include details of an
event) or an image of the source data should be easily accessible.
Clear written standard operating procedures should guarantee that the roles and responsibilities and
the required tasks are clear to all parties involved and that there is provision for proper control and,
when needed, change of the system. This is equally applicable to activities that are contracted out to
third parties, whose procedures should be reviewed to verify that they are adequate and compliant
with applicable requirements.
Staff directly performing pharmacovigilance activities, should be appropriately trained in
applicable pharmacovigilance legislation and guidelines in addition to specific training in report
processing activities for which they are responsible and/or undertake. Other personnel who may
receive or process safety reports (e.g. clinical development, sales, medical information, legal,
quality control) should be trained in adverse event collection and reporting in accordance with
internal policies and procedures.
a. Pregnancy
Reports, where the embryo or foetus may have been exposed to medicinal products (either through
maternal exposure or transmission of a medicinal product via semen following paternal exposure),
should be followed-up in order to collect information on the outcome of the pregnancy and
development of the child after birth. The recommendations provided in the Guideline on the
Exposure to Medicinal Products during Pregnancy: Need for Post-Authorisation Data should be
considered as regard the monitoring, collection and reporting of information in these specific
situations in order to facilitate the scientific evaluation. When an active substance (or one of its
metabolites) has a long half-life, this should be taken into account when assessing the possibility of
exposure of the embryo, if the medicinal product was taken before conception.
Not infrequently, pregnant women or healthcare professionals will contact either medicines
authorities or marketing authorisation holders to request information on the teratogenicity of a
medicinal product and/or experience of use during pregnancy. Reasonable attempts should be made
to obtain information on any possible medicinal product exposure to an embryo or foetus and to
follow-up on the outcome of the pregnancy.
Reports of exposure to medicinal products during pregnancy should contain as many detailed
elements as possible in order to assess the causal relationships between any reported adverse events
and the exposure to the suspected medicinal product. In this context, the use of standard structured
questionnaires is recommended.
Individual cases with an abnormal outcome associated with a medicinal product following exposure
during pregnancy are classified as serious reports and should be reported, in accordance with the
requirements outlined in VI.B.7. (See VI.C.6.2.3.1 for electronic reporting recommendations in the
Arab Countries)
This especially refers to:
reports of congenital anomalies or developmental delay, in the foetus or the child;
reports of foetal death and spontaneous abortion; and
reports of suspected adverse reactions in the neonate that are classified as serious.
Other cases, such as reports of induced termination of pregnancy without information on congenital
malformation, reports of pregnancy exposure without outcome data or reports which have a normal
outcome, should not be reported since there is no suspected adverse reaction. These reports should
however be collected and discussed in the periodic safety update reports (See Module VII).
However, in certain circumstances, reports of pregnancy exposure with no suspected reactions may
necessitate to be reported. This may be a condition of the marketing authorisation or stipulated in
the risk management plan; for example pregnancy exposure to medicinal products contraindicated
in pregnancy or medicinal products with a special need for surveillance because of a high
teratogenic potential (e.g. thalidomide, isotretinoin).
A signal of a possible teratogen effect (e.g. through a cluster of similar abnormal outcomes) should
be notified immediately to the medicines authorities in accordance with the recommendations
presented in VI.C.2.2.6.
b. Breastfeeding
Suspected adverse reactions which occur in infants following exposure to a medicinal product from
breast milk should be reported in accordance with the criteria outlined in VI.B.7 (See VI.C.6.2.3.1
for electronic reporting recommendations in the Arab Countries).
on the risk-benefit balance of the medicinal product, they should be notified to the medicines
authorities in accordance with the recommendations provided in VI.C.2.2.6.
Reports associated with suspected adverse reactions should be subject to reporting in accordance
with the criteria outlined in VI.B.7 and with the electronic reporting requirements described in
VI.C.6.2.3.3. They should be routinely followed-up to ensure that the information is as complete as
possible with regards to the symptoms, treatments, outcomes, context of occurrence (e.g., error in
prescription, administration, dispensing, dosage, unauthorised indication or population, etc.).
26
Definition and Application of Terms for vaccine Pharmacovigilance, 2012
organisation, explicit procedures and detailed agreements should exist between the marketing
authorisation holder and the person/organisation to ensure that the marketing authorisation holder
can comply with the reporting obligations. These procedures should in particular specify the
processes for exchange of safety information, including timelines and regulatory reporting
responsibilities and should avoid duplicate reporting to the medicines authorities.
For ICSRs described in the scientific and medical literature (See VI.B.1.1.2), the clock starts (day
zero) with awareness of a publication containing the minimum information for reporting. Where
contractual arrangements are made with a person/organisation to perform literature searches and/or
report valid ICSRs, detailed agreements should exist to ensure that the marketing authorisation
holder can comply with the reporting obligations.
When additional significant information is received for a previously reported case, the reporting
time clock starts again for the submission of a follow-up report from the date of receipt of the
relevant follow-up information. For the purpose of reporting, significant follow-up information
corresponds to new medical or administrative information that could impact on the assessment or
management of a case or could change its seriousness criteria; non-significant information includes
updated comments on the case assessment or corrections of typographical errors in the previous
case version. See also VI.C.6.2.2.7 as regards the distinction between significant and
non-significant follow-up information.
27
http://www.ich.org/
VI.C.1. Interface with safety reporting rules for clinical trials and post
authorisation studies in the Arab Countries
The pharmacovigilance rules laid down in this guideline do not apply to investigational medicinal
products and non-investigational medicinal products 28 used in clinical trials conducted in
accordance with the "National rules governing interventional clinical trials of medicinal products".
Post-authorisation safety or efficacy studies requested by medicines authorities in Arab Countries,
or conducted voluntarily by marketing authorisation holders, can either be clinical trials or
non-interventional studies as shown in Figure VI.1. Both types differ in their safety reporting
requirement as discussed below.
Further guidance on post-authorisation safety studies is provided in Module VIII.
The different types of studies and clinical trials which can be conducted in the Arab Countries are
illustrated in Figure VI.1.
1. The safety reporting for clinical trials corresponding to Section A, B, C and D of Figure VI.1
follows the requirements of "national regulation for pharmacovigilance of clinical trials" and the
"national rules governing interventional clinical trials of medicinal products". (this guideline
28
For guidance on these terms, see "national regulation for pharmacovigilance of clinical trials".
Section A: Clinical trials which are conducted when no marketing authorisation exists in the Arab
County concerned (i.e.pre-authorisation).
Section B: Clinical trials which are conducted in the post-authorisation period, e.g. for new
indication.
Section C: Post-authorisation clinical trials conducted in accordance with the summary of
product characteristics (SmPC) indication and condition of use, but which fall under
the scope of clinical trials regulations due to the nature of the intervention.
Section D: Post-authorisation safety or efficacy clinical trials requested or conducted
voluntarily by marketing authorisation holders, but which fall under the scope of
clinical trials regulations due to the nature of the intervention.
Section E: Non-interventional post-authorisation safety or efficacy studies requested or conducted
voluntarily by the marketing authorisation holders and which follow the requirements
of this guideline.
adverse reaction.
Only valid ICSRs (See VI.B.2) of adverse reactions, which are suspected to be related to the studied
(or supplied) medicinal product by the primary source or the receiver of the case, should be
reported. They should be considered as solicited reports (with the exception of certain reports from
compassionate use or named patient use (See VI.C.1.2.2)) and reported by marketing authorisation
holders in accordance with the requirements provided in VI.C.3, VI.C.4 and VI.C.6. Other reports
of adverse events should only be included in the study report, where applicable.
Electronic reporting recommendations for cases originating in post-authorisation studies are
detailed in VI.C.6.2.3.7.
It may happen that reports of adverse reactions are only suspected to be related to other medicinal
products which are not subject to the scope of the post-authorisation study. If there is no interaction
with the studied (or supplied) medicinal product, these reports should be notified by the primary
source, to the medicines authority in the Arab Country where the reaction occurred or to the
marketing authorisation holder of the suspected medicinal product, but not to both to avoid
duplicate reporting. Where made aware of such case, the concerned medicines authorities or
marketing authorisation holders should apply the reporting requirements described in VI.C.6.2.3.7
Further guidance on post-authorisation studies conducted by marketing authorisation holders is
provided in VI.C.2.2.2.
Academic sponsors should follow local requirements as regards the reporting of cases of suspected
adverse reactions to the medicines authority in the Arab Country where the reaction occurred.
However, where a study is directly financed, or where the design is influenced by a marketing
authorisation holder, the marketing authorisation holder should fulfil the reporting requirements
detailed in this Module.
29
This does not concern donation of a medicinal product for research purpose if the marketing authorisation
holder has no influence on the study.
the view of the primary source as regard the causal role of the studied (or supplied) medicinal
product on the notified adverse event. Where this opinion is missing, the marketing authorisation
holder should exercise its own judgement based on the information available in order to decide
whether the report is a valid ICSR, which should be reported to the national medicines authorities.
This does not apply to study designs based on secondary use of data for which reporting of ICSRs is
not required (See VI.C.1.2.1).
Safety data to be presented in the relevant sections of the periodic safety update report of the
authorised medicinal product are detailed in Module VII.
(see Module VII) and analysed as regards their overall impact on the medicinal product risk-benefit
profile. In addition, any new safety information, which may impact on the risk-benefit profile of a
medicinal product, should be notified immediately to the medicines authorities in the Arab
Countries where the medicinal product is authorised.
A detailed guidance on the monitoring of the scientific and medical literature has been developed; it
is included in VI. Appendix 2.
The electronic reporting recommendations regarding suspected adverse reactions reports published
in the scientific and medical literature are provided in VI.C.6.2.3.2.
VI.C.2.2.4. Suspected adverse reactions related to quality defect or falsified medicinal products
1. When a report of suspected adverse reactions is associated with a suspected or confirmed
falsified medicinal product or quality defect of a medicinal product, a valid ICSR should be
reported. The seriousness of the ICSR is linked to the seriousness of the reported suspected
adverse reactions in accordance with the definitions provided in VI.A.2.4. Electronic reporting
recommendations provided in VI.C.6.2.3.5 should be followed.
2. In addition in order to protect public health, it may become necessary to implement urgent
measures such as the recall of one or more defective batch(es) of a medicinal product from the
market. Therefore, marketing authorisation holders should have a system in place to ensure that
reports of suspected adverse reactions related to falsified medicinal products or to quality
defects of a medicinal products are investigated in a timely fashion and that confirmed quality
defects are notified separately to the manufacturer and to national medicines authorities.
In the context of evaluating a suspected transmission of an infectious agent via a medicinal product,
care should be taken to discriminate, whenever possible, between the cause (e.g., injection/
administration) and the source (e.g., contamination) of the infection and the clinical conditions of
the patient at the time of the infection (immuno-suppressed /vaccinee).
Confirmation of contamination (including inadequate inactivation/attenuation of infectious agents
as active substances) of the concerned medicinal product increases the evidence for transmission of
an infectious agent and may therefore be suggestive of a quality defect for which the procedures
detailed in VI.C.2.2.4 should be applied.
VI.C.2.2.7. Period between the submission of the marketing authorisation application and the
granting of the marketing authorisation
In the period between the submission of the marketing authorisation application and the granting of
the marketing authorisation, information (quality, non-clinical, clinical) that could impact on the
risk-benefit balance of the medicinal product under evaluation may become available to the
applicant. It is the responsibility of the applicant to ensure that this information is immediately
submitted in accordance with the modalities described in VI.C.2.2.6 to the national medicines
authorities in the Arab Countries where the application is under assessment.
VI.C.2.2.10. Reports from patient support programmes and market research programmes
A patient support programme is an organised system where a marketing authorisation holder
receives and collects information relating to the use of its medicinal products. Examples are
post-authorisation patient support and disease management programmes, surveys of patients and
healthcare providers, information gathering on patient compliance, or
compensation/re-imbursement schemes.
A market research programme refers to the systematic collection, recording and analysis by a
marketing authorisation holder of data and findings about its medicinal products, relevant for
marketing and business development.
Safety reports originating from those programmes should be considered as solicited reports.
Marketing authorisation holders should have the same mechanisms in place as for all other solicited
reports (See VI.C.2.2.2) to manage that information and report valid cases of adverse reactions,
which are suspected to be related to the concerned medicinal product.
Valid ICSRs should be reported as solicited in accordance with the electronic reporting
requirements provided in VI.C.6.2.3.7.
serious domestic valid ICSRs shall be reported to medicines authority in the Arab Country
concerned by marketing authorisation holders within 15 days from the date of receipt of the
reports;
non-serious domestic valid ICSRs shall be reported to medicines authority in the Arab Country
concerned by marketing authorisation holders within 90 days from the date of receipt of the
reports.
reporting of serious international valid ICSRs by MAHs may be required in some Arab
Countries; consult with the national medicines authority for national requirements for these
ICSRs.
This should be done in accordance with the reporting modalities detailed in VI.C.4.
a. Serious ICSRs
Marketing authorisation holders shall report all serious ICSRs that occur in the Arab Country
concerned to the national medicines authority of the Arab Country on whose territory the
suspected adverse reactions occurred (i.e. domestic ICSRs).
Only in some Arab Countries; marketing authorisation holders are required to report to
national medicines authority of the Arab Country in which the medicinal product is authorised
the serious ICSRs that occur outside these Arab Countries (i.e. international serious ICSRs),
including those received from medicines authorities. Consult with the national medicines
authority for national requirements about international serious ICSRs.
National medicines authorities in the Arab Countries shall ensure that all serious ICSRs that
occur in their territory and that are reported to them, including those received from marketing
authorisation holders, are made available to the ―National Pharmacovigilance and Safety reports
database‖. National medicines authorities in the Arab Countries should also make available, to
the marketing authorisation holders of the suspected medicinal products, all serious ICSRs
reported directly to them.
b. Non-Serious ICSRs
Marketing authorisation holders shall report all non-serious ICSRs that occur in the Arab
Countries concerned to the national medicines authority of that Arab Country on whose territory
the suspected adverse reactions occurred (i.e. domestic ICSRs).
VI.C.5. Collaboration with the World Health Organization and the National
Pharmacovigilance Centres in the Arab Countries
Arab Countries participating in the WHO Programme for International Drug Monitoring shall
report to the WHO Collaborating Centre for International Drug Monitoring all suspected adverse
reactions reports occurring in their territory. This will take place on a weekly basis after their
transmission to the ―National Pharmacovigilance and Safety reports database‖. Another frequency
may be adopted by the national pharmacovigilance centre as appropriate.
In the situation where it is evident that the sender has not transmitted the complete information
available on the case, the receiver may request the sender to re-transmit the ICSR within 24 hours
with the complete case information in electronic format in accordance with the requirements
applicable for the electronic reporting of ICSRs. This should be seen in the light of the qualitative
signal detection and evaluation activity, where it is important for the receiver to have all the
available information on a case to perform the medical assessment (see VI.C.6.2.4).
Where the suspected adverse reactions reported in a single ICSR impact on the known risk-benefit
balance of a medicinal product, this should be considered as an Emerging Safety Issue (see
VI.C.2.2.6), which should be immediately notified in writing to the national medicines authorities
of the Arab Countries where the medicinal product is authorised. This is in addition to the reporting
requirements detailed in VI.C.4. A summary of the points of concerns and the action proposed
should be recorded in the ICSR in data element ‗Sender‘s comments‘ (ICH-E2B (R2) B.5.4).
the composition with regard the active substance(s) of the proprietary medicinal product should be
provided accordingly.
Where the primary source reports a suspect or interacting branded/proprietary medicinal product
name without indicating the pharmaceutical form/presentation of the product and where the
proprietary/branded medicinal product can be one of two or more possible pharmaceutical
forms/presentations, which have different compositions in a country, the ICSR should be populated
as follows:
data element 'Proprietary medicinal product name' (ICH-E2B(R2) B.4.k.2.1) should be
populated with the medicinal product name as reported by the primary source;
data element 'Active substance name(s)' (ICH-E2B(R2) B.4.k.2.2) should be completed with
those active substances which are in common to all pharmaceutical forms/presentations in the
country of authorisation.
Where medicinal products cannot be described on the basis of the active substances or the invented
names, for example when only the therapeutic class is reported by the primary source, or in case of
other administered therapies that cannot be structured, this information should only be reflected in
the case narrative (data element ICH-E2B(R2) B.5.1). The data elements ‗Proprietary medicinal
product name‘ (ICH-E2B(R2) B.4.k.2.1) and ‗Active substance name(s)‘ (ICH-E2B(R2) B.4.k.2.2)
should not be populated. The same applies if a suspected food interaction is reported (e.g. to
grapefruit juice).
Where a case of adverse reactions is reported to be related only to a therapeutic class, it is
considered incomplete and does not qualify for reporting (see VI.B.2). Efforts should be made to
follow-up the case in order to collect the missing information regarding the suspected medicinal
product (see VI.B.3).
As regards the reporting of drug interactions, which concerns drug/drug (including biological
products), drug/food, drug/device, and drug/alcohol interactions, the coding of the interaction
should be performed in Section ‗Reactions/Events‘ (ICH-E2B(R2) B.2) in line with the latest
version of the ICH-Endorsed Guide for MedDRA Users - MedDRA Term Selection: Points to
Consider Document. In addition, for drug/drug interactions, information on the active
substances/proprietary medicinal product names should be provided in the Section ‗Drug
information‘ (ICH-E2B(R2) B.4), which should be characterised as interacting in the data element
‗Characterisation of drug role‘ (ICH-E2B(R2) B.4.k.1).
If the primary source suspects a possible causal role of one of the ingredients (e.g., excipient or
adjuvant) of the suspected medicinal product, this information should be provided in the Section
‗Drug information‘ (ICH-E2B(R2) B.4) as a separate entry in addition to the information given
regarding the suspected medicinal product. This should also be specified in the case narrative (data
element ICH-E2B(R2) B.5.1). If available, tests results (positive or negative) in relation to the
causal role of the suspected ingredient should be included in the section 'Results of tests and
procedures relevant to the investigation of the patient' (ICH E2B(R2) B.3).
provisional diagnoses with signs and symptoms in the data element 'Reaction/event in MedDRA
terminology (Lowest Level Term)' (ICH-E2B(R2) B.2.i.1) should be performed in line with the
latest version of the ICH-Endorsed Guide for MedDRA Users, MedDRA Term Selection: Points to
Consider.
In practice, if a diagnosis is reported with characteristic signs and symptoms, the preferred option is
to select a term for the diagnosis only and to MedDRA code it in the ICH-E2B(R2) section B.2
'Reaction(s)/event(s)'. If no diagnosis is provided, all reported signs and symptoms should be listed
and MedDRA coded in the ICH-E2B(R2) section B.2 'Reaction(s)/event(s)'. If these signs and
symptoms are typically part of a diagnosis, the diagnosis can be MedDRA coded in addition by
medicines authorities in Arab Countries or marketing authorisation holders in the ICH-E2B(R2)
data element B.5.3 ‗Sender's diagnosis/syndrome and/or reclassification of reaction/event'.
If in the narrative other events have been reported, which are not typically signs or symptoms of the
primary source's diagnosis or provisional diagnosis, and those events are suspected to be adverse
reactions, they should also be listed and MedDRA coded in the ICH-E2B(R2) section B.2
'Reaction(s)/event(s)'.
In case a medicines authority in an Arab Country or a marketing authorisation holder disagrees with
the diagnosis reported by the primary source, an alternative diagnosis can be provided in the
ICH-E2B(R2) data element B.5.3 ‗Sender's diagnosis/syndrome and/or reclassification of
reaction/event‘ in addition to the reported diagnosis provided in the ICH-E2B(R2) section B.2
'Reaction(s)/event(s)'. In this situation, a reasoning should be included in the data element ‗Sender‘s
comments‘ (ICH-E2B(R2) B.5.4) (See VI.C.6.2.2.4).
In the event of death of the patient, the date, cause of death including autopsy-determined causes
shall be provided as available. If the death is unrelated to the reported suspected adverse reaction(s)
and is linked for example to disease progression, the seriousness criterion of the ICSR should not be
considered as fatal.
30
‗Where possible‘ should be interpreted as having received sufficient information from the primary source to
prepare a concise clinical summary of the individual case.
31
Council for International Organizations of Medical Sciences (CIOMS). Current Challenges in Pharmacovigilance:
Pragmatic Approaches (CIOMS V). Geneva: CIOMS; 2001. http://www.cioms.ch/ .
version change of MedDRA, can be considered as a non-significant change as long as this change
has no impact on the medical content of a case. However, an amendment of the MedDRA coding
due to a change in the interpretation of a previously reported suspected adverse reaction may
constitute a significant change and therefore should be reported.
In situations where the case is modified without impacting on its medical evaluation, while no new
follow-up is received (e.g., for correcting a mistake or typographical error), the date of receipt of the
most recent information reported in the data element ‗Date of receipt of the most recent information
for this report‘ (ICH-E2B(R2) A.1.7 ) should not be changed. This data element should however be
updated in any other situations, to the date when new follow-up information is received
(independently whether it is significant or not) or to the date when changes are made which impact
on the interpretation of the case.
Where follow-up information of a case initially reported by a marketing authorisation holder is
received directly by a medicines authority, the ‗Worldwide unique case identification number‘
(ICH-E2B(R2) A.1.10) of the initial report should be maintained, in adherence with the
ICH-E2B(R2) rules. The same principle should be applied if a follow-up is received by a marketing
authorisation holder of a case initially reported by a medicines authority.
indicate that a previously transmitted report should be considered completely void (nullified), for
example when the whole case was found to be erroneous or in case of duplicate reports. It is
essential to use the same case report numbers previously submitted in the data element ‗Sender‘s
(case) safety report unique identifier‘ (ICH-E2B(R2) A.1.0.1) and in the data element ‗Worldwide
unique case identification number‘ (ICH-E2B(R2) A.1.10).
A nullified case is one that should no longer be considered for scientific evaluation. The process of
the nullification of a case is by means of a notification by the sender to the receiver that this is no
longer a valid case. However, the case should be retained in the sender‘s pharmacovigilance
database for auditing purposes.
The principles to be considered when nullifying a case are detailed in VI. Appendix 3.
the father.
VI.C.6.2.3.2. Suspected adverse reaction reports published in the scientific and medical literature
Requirements in relation to the monitoring of suspected drug reactions reported in the scientific and
medical literature are provided in VI.C.2.2.3. With regard to the electronic reporting of ICSRs
published in the scientific and medical literature, the following applies:
The literature references shall be included in the data element ‗Literature reference(s)‘
(ICH-E2B(R2) A.2.2) in the Vancouver Convention (known as ―Vancouver style‖), developed
by the International Committee of Medical Journal Editors . The standard format as well as those
for special situations can be found in the following reference: International Committee of
Medical Journal Editors. Uniform requirements for manuscripts submitted to biomedical
journals. N Engl J Med. 1997; 336: 309-15, which is in the Vancouver style32.
A comprehensive English summary of the article shall be provided in the data element ‗Case
narrative including clinical course, therapeutic measures, outcome and additional relevant
information‘ (ICH-E2B(R2) B.5.1) .
Upon request of the national medicines authority, for specific safety review, a full translation in
English and a copy of the relevant literature article shall be provided by the marketing
authorisation holder that transmitted the initial report, taking into account copyright restrictions.
The recommendations detailed in VI.App2.10, regarding the mailing of the literature article,
should be adhered to.
Recommendations presented in VI.App2.10, for the reporting of several cases when they are
published in the same literature article, should be followed.
VI.C.6.2.3.3. Suspected adverse reactions related to overdose, abuse, off-label use, misuse,
medication error or occupational exposure
General principles are provided in VI.B.6.3.
If a case of overdose, abuse, off-label use, misuse, medication error or occupational exposure is
reported with clinical consequences, the MedDRA Lowest Level Term code, corresponding to the
term closest to the description of the reported overdose, abuse, off-label use, misuse, medication
error or occupational exposure should be added to the observed suspected adverse reaction(s) in the
data element ‗Reaction/event in MedDRA terminology (Lowest Level Term)‘ (ICH-E2B(R2)
B.2.i.1), in line with recommendations included in the latest version of the ICH-Endorsed Guide for
MedDRA Users 'MedDRA Term Selection: Points to Consider'.
32
The Vancouver recommendations are also available on the International Committee of Medical Journal Editors
website http://www.icmje.org .
efficacy, should be provided in the data element ‗Reaction/event in MedDRA terminology (Lowest
Level Term)‘ (ICH-E2B(R2) B.2.i.1), in line with recommendations included in the latest version of
the ICH-Endorsed Guide for MedDRA Users 'MedDRA Term Selection: Points to Consider'.
Unless aggravation of the medical condition occurs, the indication for which the suspected
medicinal product was administered should not be included in the data element ‗Reaction/event in
MedDRA terminology (Lowest Level Term).
The same reporting modalities as for serious ICSRs (See VI.C.4) should be applied for those cases
related to classes of medicinal products where, as described in VI.B.6.4, reports of lack of
therapeutic efficacy should be reported within a 15-day time frame. If no seriousness criterion is
available, it is acceptable to submit the ICSR within 15 days as non-serious.
VI.C.6.2.3.5. Suspected adverse reactions related to quality defect or falsified medicinal products
Requirements are provided in VI.C.2.2.4. In order to be able to clearly identify cases related to
quality defect or falsified medicinal products when they are exchanged between stakeholders, the
following recommendations should be applied:
a) Quality defect
Where a report of suspected adverse reactions is associated with a suspected or confirmed
quality defect of a medicinal product, the MedDRA Lowest Level Term code of the term
corresponding most closely to the product quality issue, should be added to the observed
suspected adverse reaction(s) in the data element ‗Reaction/event in MedDRA terminology
(Lowest Level Term)‘ (ICH-E2B(R2) B.2.i.1).
b) Falsified medicinal products
Where a report of suspected adverse reactions is associated with a suspected or confirmed
falsified ingredient, active substance or medicinal product, the MedDRA Lowest Level Term
code of the term corresponding most closely to the reported information should be added to the
observed suspected adverse reaction(s) in the data element ‗Reaction/event in MedDRA
terminology (Lowest Level Term)‘ (ICH-E2B(R2) B.2.i.1). Information on the suspected
medicinal product, active substance(s) or excipient(s) should be provided in the data elements
‗Proprietary medicinal product name‘ (ICH-E2B(R2) B.4.k.2.1) and/or ‗Active substance
name(s)‘ (ICH-E2B(R2) B.4.k.2.2) as reported by the primary source.
VI.C.6.2.3.7. Reports originating from organised data collection systems and other systems
General safety reporting requirements in the Arab Countries for post-authorisation studies are
provided in VI.C.1 and VI.C.2.2.2. Individual case safety reports originating from those studies
shall contain information on study type, study name and the sponsor‘s study number or study
registration number. This should be provided in ICH E2B(R2) section A.2.3 ‗Study identification‘.
Safety reporting requirements regarding patient support programmes or market research
programmes are provided in VI.C.2.2.10.
The following reporting rules should be applied based on (i) the type of data collection system and
(ii) whether the suspected medicinal product is part of the scope of the data collection system.
1. For all patient support programmes, non-interventional studies with primary data collection
from consumers and healthcare professionals, and for certain compassionate use or named
patient use where adverse events are actively sought:
a) Where the adverse reaction is suspected to be related at least to the studied (or supplied)
medicinal product:
the report should be considered as solicited;
the ICH E2B(R2) data element A.1.4 'Type of report' should be populated with the
value 'Report from study';
the ICH E2B(R2) data element A.2.3.3 'Study type in which the reaction(s)/event(s)
were observed' should be populated with the value ‗Other studies‘ or 'Individual
patient use'.
b) Where the adverse reaction is only suspected to be related to a medicinal product which is not
subject to the scope of the organised data collection system and there is no interaction with the
studied (or supplied) medicinal product:
the report should be considered as spontaneous report; as such it conveys the suspicion
of the primary source;
The ICH E2B(R2) data element A.1.4 'Type of report' should be populated with the
value 'Spontaneous'.
2. For certain compassionate use or named patient use where adverse event reporting is not
solicited:
the report should be considered as spontaneous report; as such it conveys the suspicion
of the primary source;
The ICH E2B(R2) data element A.1.4 'Type of report' should be populated with the
value 'Spontaneous'.
3. For clinical trial and where the adverse reaction is only suspected to be related to a
non-investigational medicinal product (or another medicinal product which is not subject to the
scope of the clinical trial) and there is no interaction with the investigational medicinal product:
the report should be considered as spontaneous report; as such it conveys the suspicion
VI.C.6.2.4. Data quality of individual case safety reports transmitted electronically and duplicate
management
The ―National Pharmacovigilance and Safety reports database‖ should contain all domestic cases of
suspected adverse reactions to support pharmacovigilance activities. This applies to all medicinal
products authorised in the Arab Country concerned.
The ―National Pharmacovigilance and Safety reports database‖ should also be based on the highest
internationally recognised data quality standards.
To achieve these objectives, all medicines authorities in Arab Countries and marketing
authorisation holders should adhere to:
the electronic reporting requirements;
the concepts of data structuring, coding and reporting in line with the guidelines, standards and
principles referred to in VI.C.6.2.2.1.
The national medicines authorities shall, in collaboration with the stakeholder that submitted an
ICSR to the ―National Pharmacovigilance and Safety reports database‖, be responsible for
operating procedures that ensure the highest quality and full integrity of the information collected in
this national database. This includes as well the monitoring of use of the terminologies referred to in
VI.C.6.1.
In this regard, marketing authorisation holders and national medicines authorities in each Arab
Countries should have in place an audit system, which ensures the highest quality of the ICSRs
transmitted electronically to the ―National Pharmacovigilance and Safety reports database‖within
the correct time frames, and which enables the detection and management of duplicate ICSRs in
their system. Those transmitted ICSRs should be complete, entire and undiminished in their
structure, format and content.
High level business process maps and process descriptions in relation to the quality review of ICSRs
and the detection and management of duplicate ICSRs are provided in VI. Appendix 4 and VI.
Appendix 5.
1 Start. Day 0. Receipt of the information for the case that MAH/NCA
Receive report. indicates that one of the suspect drugs is of
biological origin.
2 Does report concern a If Yes, go to step 3
biological medicinal If No, go to step 4
product?
3 Are batch number, brand If Yes, create the case and send it to the correct MAH/NCA
name & active substance all receiver (step 3).
present and identifiable? If there is more than one batch number, structure the
batch number that coincided with the adverse reaction
in the Drug section (ICH-E2B(R2) B.4) and enter the
other batch numbers in the case narrative.
If No, create the case and send it to the correct
receiver (step 3) and follow-up with the reporter (step
3.1).
3.1 Follow-up with reporter. Follow-up with the reporter to attempt to identify the MAH/NCA
missing information.
3.2 Was reporter able to provide If Yes, return to step 1 – the information should be MAH/NCA
the missing information? treated as follow-up and a new version created &
transmitted.
If No, document this (step 3.3).
3.3 Document the required Document in the case that the missing required MAH/NCA
missing information in the information has been sought but the reporter was not
case. able or willing to provide it.
4 Send to receiver, where If the case requires transmission to a receiver, transmit MAH/NCA
applicable. the case [if applicable electronically, in E2B(R2)
format] within the relevant timelines (15 or 90 days),
to the relevant receiver.
5 Receive in DataBase (DB). Receive the case electronically and load it into the Receiver
pharmacovigilance database.
6 Validate products and Validate the products and substances to ensure that Receiver
substances the brand name, active substance & batch number are
all present and identifiable.
This validation should be complementary to the usual
business rules validations.
7 Was validation successful? If Yes, store the case in the pharmacovigilance Receiver
database (step 8).
If No, contact the sender (Step 7.1).
7.1 Contact sender. Contact the sender regarding the missing or not Receiver
identifiable information.
7.2 Is required data in the Upon receipt of communication from the receiver, MAH/NCA
case file? check in the case file to see if the missing or
unidentifiable information is already on file.
If it is on file, correct the case (step 7.3).
If the information is not on file, contact the reporter to
request the missing information (step 3.1).
7.3 Correct case. Correct the case to include the missing information & MAH/NCA
send updated version to receiver (step 4).
VI. App2.1 When to start and stop searching in the scientific and medical
literature
Requirements as regards the monitoring of scientific and medical literature are provided in
VI.C.2.2.3.
In addition to the reporting of serious and non-serious ICSRs or their presentation in periodic safety
update reports, the marketing authorisation holder has an obligation to review the worldwide
experience with medicinal product in the period between the submission of the marketing
authorisation application and the granting of the marketing authorisation. The worldwide
experience includes published scientific and medical literature. For the period between submission
and granting of a marketing authorisation, literature searching should be conducted to identify
published articles that provide information that could impact on the risk-benefit assessment of the
product under evaluation. For the purpose of the preparation of periodic safety update reports (See
Module VII) and the notification of Emerging Safety Issues (See VI.C.2.2.6), the requirement for
literature searching is not dependent on a product being marketed. Literature searches should be
conducted for all products with a marketing authorisation, irrespective of commercial status. It
would therefore be expected that literature searching would start on submission of a marketing
authorisation application and continue while the authorisation is active.
relevant records are not omitted, however, there is no defined acceptable loss of recall when
searching for pharmacovigilance purposes. Term selection should be relevant to the database used
and the subject of the search.
If limits are applied, they should be relevant to the purpose of the search. When searching a
worldwide scientific and medical literature database, titles and abstracts are usually in English
language. The use of limits that reduce the search result to only those published in the English
language is generally not acceptable. Limits applied to patient types, or other aspects of an article,
for example human, would need to be justified in the context of the purpose of a search.
Limits can be applied to produce results for date ranges, for example, weekly searches can be
obtained by specifying the start and end date for the records to be retrieved. Care should be taken to
ensure that the search is inclusive for an entire time period, for example, records that may have been
added later in the day for the day of the search should be covered in the next search period. The
search should also retrieve all records added in that period, and not just those initially entered or
published during the specified period (so that records that have been updated or retrospectively
added are retrieved). This should be checked with the database provider if it is not clear.
Although one of the purposes of searching is to identify ICSRs for reporting, the use of publication
type limits is not robust. ICSRs may be presented within review or study publications, and such
records may not be indexed as "case-reports", resulting in their omission for preparation of periodic
safety update reports from search results limited by publication type.
A common issue in selecting relevant articles from the results of a search is that often this process is
conducted for the purposes of identification of ICSRs only. Whereas the review should also be used
as the basis for collating articles for the periodic safety update report production, therefore relevant
studies with no ICSRs should also be identified, as well as those reports of events that do not qualify
for reporting.
Outputs from searches may contain enough information to be a valid ICSR, in which case the article
should be ordered. All articles for search results that are likely to be relevant to pharmacovigilance
requirements should be obtained, as they may contain valid ICSRs or relevant safety information.
The urgency with which this occurs should be proportionate to the content of the material reviewed
and the resulting requirement for action as applicable for the marketing authorisation holder.
Articles can be excluded from reporting by the marketing authorisation holder if another company's
branded medicinal product is the suspected medicinal product. In the absence of a specified
medicinal product source and/or invented name, ownership of the medicinal product should be
assumed for articles about an active substance. Alternative reasons for the exclusion of a published
article for the reporting of ICSRs are detailed in VI.C.2.2.3.
organisation may share services of a third party to conduct searches for generic active substances. In
this instance, each organisation should satisfy itself that the search and service is appropriate to their
needs and obligations.
Where an organisation is dependent on a particular service provider for literature searching, it is
expected that an assessment of the service(s) is undertaken to determine whether it meets the needs
and obligations of the organisation. In any case, the arrangement should be clearly documented.
The clock start for the reporting of ICSRs begins with awareness of the minimum information by
either the organisation or the contractual partner (whichever is the earliest). This also applies where
a third party provides a review or a collated report from the published scientific and medical
literature, in order to ensure that published literature cases are reported as required within the correct
time frames. That is, day zero is the date the search was run if the minimum criteria are available in
the abstract and not the date the information was supplied to the organisation.
Ex Scenario Action
1 A literature article For Case 1 described in the literature article:
describes suspected ICH-E2B(R2) A.1.10.1 ‗World-Wide Unique Case Identification
adverse reactions that Number‘:
have been
UK-ORGABC-0001
experienced by up to 3
single patients. ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
3 ICSRs should be
UK-ORGABC-0002
created and reported
for each individual ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
identifiable patient linked to this report‘:
described in the UK-ORGABC-0003
literature article.
ICH-E2B(R2) A.2.2 ‗Literature reference(s):
Each ICSR should Literature reference in line with uniform requirements for
contain all the
manuscripts submitted to biomedical journals:
available information
N Engl J Med. 1997;336:309-15.
on the case.
File name for the copy of literature article to be sent via e-mail:
UK-ORGABC-0001.pdf
For Case 2 described in the literature article:
ICH-E2B(R2) A.1.10.1 ‗World-Wide Unique Case Identification
Number‘:
UK-ORGABC-0002
Ex Scenario Action
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
UK-ORGABC-0001
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
UK-ORGABC-0003
ICH-E2B(R2) A.2.2 ‗Literature reference(s):
Literature reference in line with uniform requirements for
manuscripts submitted to biomedical journals:
N Engl J Med. 1997;336:309-15.
No copy of the literature article required since the copy was already
submitted for case 1.
For Case 3 described in the literature article:
ICH-E2B(R2) A.1.10.1 ‗World-Wide Unique Case Identification
Number‘:
UK-ORGABC-0003
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
UK-ORGABC-0001
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
UK-ORGABC-0002
ICH-E2B(R2) A.2.2 ‗Literature reference(s):
Literature reference in line with uniform requirements for
manuscripts submitted to biomedical journals:
N Engl J Med. 1997;336:309-15.
No copy of the literature article required since the copy was already
submitted for case 1.
2 A literature article For the ICSRs which relate to the same literature article, the cross reference in
describes suspected the data element ‗Identification number of the report which is linked to this
adverse reactions that report‘ ICH (E2B(R2) field A.1.12) should be conducted as follows:
have been The first case should be linked to all other cases related to the same
experienced by more article;
than 3 single patients.
All the other cases should be only linked to the first one, as in the
ICSRs should be
example below.
created and reported
for each individual
identifiable patient Example for the reporting of cases originally reported in the scientific and
described in the medical literature referring to a large number of patients:
literature article.
Ex Scenario Action
Each ICSR should For Case 1 described in the literature article:
contain all the ICH E2B(R2) A.1.10.1 ‗Worldwide Unique Case Identification
available information Number‘:
on the case. UK-ORGABC-0001
The cross reference
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
with all the linked
linked to this report‘:
ICSRs from this
UK-ORGABC-0002
literature article
should only be ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
provided in the first linked to this report‘:
case, in the data UK-ORGABC-0003
element
ICH-E2B(R2) A.1.12 ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
‗Identification linked to this report‘:
number of the report UK-ORGABC-0004
which is linked to this ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
report‘. There is no linked to this report‘:
need to repeat all the
UK-ORGABC-000N
cross references in the
other ICSRs. ICH-E2B(R2) A.2.2 ‗Literature reference(s)‘:
N Engl J Med. 1997;336:309-15.
File name for the copy of literature article to be sent via e-mail:
UK-ORGABC-0001.pdf.
For Case 2 described in the literature article:
ICH E2B(R2) A.1.10.1 ‗Worldwide Unique Case Identification
Number‘:
UK-ORGABC-0002
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
UK-ORGABC-0001
ICH-E2B(R2) A.2.2 ‗Literature reference(s)‘:
N Engl J Med. 1997;336:309-15.
No copy of the literature article required since the copy was already
submitted for case 1.
For Case N described in the literature article:
ICH-E2B(R2) A.1.10.1 ‗Worldwide Unique Case Identification
Number‘:
UK-ORGABC-000N
ICH-E2B(R2) A.1.12 ‗Identification number of the report which is
linked to this report‘:
UK-ORGABC-0001
Ex Scenario Action
ICH-E2B(R2) A.2.2 ‗Literature reference(s)‘:
N Engl J Med. 1997;336:309-15.
No copy of the literature article required since the copy was already
submitted for case 1
1 An individual case has been identified as a One of the individual cases should be nullified.
duplicate of another individual case The remaining valid case should be updated
previously submitted. with any additional relevant information from
the nullified case.
2 A wrong ‗Worldwide unique case The case with the wrong ‗Worldwide unique
identification number‘ (ICH-E2B(R2) case identification number‘ (ICH-E2B(R2)
A.1.10) was accidentally used and does not A.1.10) should be nullified.
refer to an existing case. A new case should be created with a correct
‗Worldwide unique case identification number‘.
Individual cases that have been nullified should not be used for scientific evaluation, however,
they should remain in the database for auditing purposes.
In addition, in case of duplicate reports where one report needs to be nullified, the update of the
remaining case should be performed in the form of a follow-up report. Information on the
identification of the nullified case(s) should be provided in the data element ‗Source(s) of the
case identifier (e.g. name of the company, name of regulatory agency)‘ (ICH-E2B(R2) A.1.11.1)
and in the data element ‗Case identifier(s)‘ (ICH-E2B(R2) A.1.11.2).
Table VI.4. Examples of scenarios for which ICSRs should NOT be nullified
7 A wrong ‗Worldwide unique case The report with the wrong ‗Worldwide unique case
identification number‘ (ICH E2B(R2) identification number‘ (ICH-E2B(R2) A.1.10) should
A.1.10) was accidentally used. This wrong not be nullified.
ICH-E2B(R2) A.1.10 ‗Worldwide unique A follow-up report should be submitted to correct the
case identification number‘ referred to an information previously submitted.
existing case. A new ICSR should be created and submitted with the
correct ‗Worldwide unique case identification number‘.
10 Change of the individual case from serious The case should not be nullified.
to non-serious (downgrading).
A follow-up report should be submitted with the data
element ‗Seriousness‘ (ICH-E2B(R2) A.1.5.1)
populated with the value ‗No‘ without selection of a
value for the data element ‗Seriousness criteria‘
(ICH-E2B(R2) A.1.5.2).
The data element ‗Does this case fulfil the local criteria
for an expedited report?‘ (ICH-E2B(R2) field A.1.9)
should remain populated with the value ‗Yes‘.
11 The primary source country has changed, The case should not be nullified.
which has an impact on the ICH-E2B(R2)
The ‗Sender‘s (case) safety report unique identifier‘
convention regarding the creation of the
(ICH-E2B(R2) A.1.0.1) can be updated on the basis of
‗Worldwide unique case identification
the new primary source country code. However, the
number‘ (ICH-E2B(R2) A.1.10).
‗Worldwide unique case identification number‘
(ICH-E2B(R2) A.1.10) should remain unchanged.
12 The suspected medicinal product belongs The case should not be nullified.
to another marketing authorisation holder
It is recommended that the initial sender informs the
(e.g. a product with the same active
other marketing authorisation holder about this case
substance but marketed under a different
(including the ‗Worldwide unique case identification
invented name).
number‘ (ICH-E2B(R2) A.1.10) used). The original
organisation should also submit a follow-up report to
provide this new information.
13 The suspected medicinal product taken The case should not be nullified.
does not belong to the marketing
The marketing authorisation holder should submit a
authorisation holder (same active
follow-up report with this information within the
substance, the invented name is unknown
appropriate time frame.
and the report originates from a country,
where the marketing authorisation holder
has no marketing authorisation for the
medicinal product in question).
14 The case is mistakenly reported by the The case should not be nullified.
marketing authorisation holder A although
An explanation should be sent by the marketing
the marketing authorisation holder B as
authorisation holder A to the co-marketer marketing
co-marketer is responsible for reporting the
authorisation holder B that the case has already been
case.
reported. The marketing authorisation holder B should
provide any additional information on the case as a
follow-up report with the same ‗Worldwide unique case
identification number‘ (ICH-E2B(R2) A.1.10).
Table VI.5. Process description - Data quality monitoring of ICSRs transmitted electronically
The business map and process description describe a system where there is a separation between a
PharmacoVigilance DataBase (PhV DB) holder, the PhV DB holder‘s data Quality Assessors (QA)
and the PhV DB holder‘s auditors; however this is not mandatory and these functions may be
performed by the same people or groups.
2 Sample ICSRs from Take a sample of ICSRs that were transmitted by the QA
Sender. selected sender
3 Check for data quality Check the cases for data quality errors. QA
errors.
The cases should be assessed against appropriate
published standards and similar documents, for example
the MedDRA Term Selection Points to Consider
document.
4 Write report and send The findings from the data quality assessment should be QA
to PhV DB holder. collated into a single report. These can include related
checks, such as 15-day reporting compliance, whether
error reports are corrected and similar statistical
information.
5 Errors found? Were any errors found during the analysis of the cases? PhV DB
holder
If No, go to step 5.1.
If Yes go to steps 5.2, 5.3 & 6.
5.1 End. If there were no errors found, then no further action PhV DB
needs to be taken. The process can end until the next holder
time the sender is assessed.
The pharmacovigilance database (PhV DB) holder
may choose to share this information with the assessed
sender and their auditors who may wish to factor this
in to determinations of which sender to assess.
5.2 Highlight for PhV If the PhV DB holder‘s organisation has an audit PhV DB
audit. department, any significant findings should always be holder
shared with them.
5.2.1 Prioritise for Audit. The audit or inspections department should use the PhV DB
information provided to them to feed into decisions about holder’s
prioritising organisations for audit or inspection. auditors
5.3 INPUT: Findings Any errors found (or even lack thereof) should be PhV DB
from previous incorporated into decisions about which senders to holder
assessments. evaluate & should also inform the performance of the
assessments (e.g. targeting particular types of case) and
the report (documenting whether previously identified
issues have been addressed).
6 Inform sender of Inform the sender of the findings, including requested PhV DB
findings. remedial actions (e.g. retransmitting certain cases) and holder
time frames for those actions
7 Request meeting? The sender should have the option to choose to request a Sender
meeting to discuss the findings and appropriate remedial
action and time frames.
If no meeting is requested, go to step 7.1. If a meeting is
requested go to step 8.
7.1 Address the findings Address all findings, take necessary steps to prevent Sender
& retransmit any recurrence of such findings & retransmit any required
required cases. cases.
7.2 End. Once all findings have been addressed, the necessary Sender
steps taken to prevent recurrence of such findings and
any required cases have been retransmitted, the
process can end until the next time the sender is
assessed.
8 Have meeting. Upon request from one party, a meeting should be held to PhV DB
discuss the findings of quality assessments and holder &
appropriate remedial and preventive actions to ensure that Sender
the cases in the database are correct and shall be so in the
future.
9 End. Unless further action has been specified (e.g. future PhV DB
meetings or assessments), the process can end until the holder
next time the sender is assessed.
2.1 Not a Duplicate: If the cases are assessed as not being duplicates of one DMT
Mark as not a another, then mark both cases as such.
duplicate.
Go to step 3 (End).
2.2 More information There should be some form of tool for tracking when more DMT
needed: Log in information is needed, when correspondence has been
tracking tool. sent, whether an answer was received and, if so, when.
2.2.1 Write to Sender. More information is required in order to be able to make a PhV DB
definite assessment. holder
The sender (who transmitted the case(s) in question to the
PhVDB holder‘s organisation) should be contacted to
request specific information necessary to confirm or deny
duplication.
Personal data protection must remain paramount, so
unsecured communications should not include sufficient
data to identify an individual.
2.2.2 Receive request, Once a request for more information has been received, Sender
draft and send the Sender of the case should respond promptly, either as a
response. follow-up version of the case or by responding to the
requester.
The DMT should then reassess the case based on the new
information (Go back to step 2).
2.3 Duplicates Once cases have been determined to be duplicates of one DMT
Different another and have been transmitted to the PhV DB holder
Senders: Create by different senders or reporters, then they should be
or nominate merged under a master case, following the process
master. described in chapter 2.3 ―Management of duplicate cases‖
of the Guideline on the Detection and Management of
Duplicate Individual Cases and Individual Case Safety
Reports (ICSRs), EMA/13432/2009, (see GVP Annex
III). This is an EMA guideline which is adopted in the
Arab Countries from the scientific point of view.
2.3.1 Deal with If any follow-ups arrive for any of the cases, this DMT
follow-ups. information may require a reassessment of the master
case.
Reassess and, if necessary, amend the master case as with
any received follow-up information.
Go to step 3 (End).
2.4 Duplicates Same Once cases have been determined to be duplicates of one DMT
Sender: Log in another, and have been transmitted to the PhV DB holder
tracking tool. by the same sender, then this decision and the
correspondence referred to in step 2.4.1 should be logged
in the tracking tool referred to in step 2.2.
2.4.1 Write to Sender. The sender organisation, as the source of the duplicates, PhV DB
should be contacted in accordance with chapter 2.3.3 of holder
the Guideline on the Detection and Management of
Duplicate Individual Cases and Individual Case Safety
Reports (ICSRs), EMA/13432/2009 , (see GVP Annex
III). This is an EMA guideline which is adopted in the
Arab Countries from the scientific point of view.
The sender should be asked to confirm or deny duplication
and take appropriate steps in accordance with chapter
2.3.1 of the aforementioned Guideline.
2.4.2 Receive request. Receive and log the communication containing Sender
information on suspected duplicates in the Sender‘s PhV
2.4.3 Is it a duplicate? Assess the potential duplicates. Are the cases duplicates of Sender
one another?
If Yes, go to step 2.4.3.1.
If No, go to step 2.4.3.2.
2.4.3.1 Merge duplicates. Merge the duplicates, taking into account Flowchart 1 of Sender
chapter 2.3.1.3 of the Guideline on the Detection and
Management of Duplicate Individual Cases and Individual
Case Safety Reports (ICSRs), EMA/13432/2009, (see
GVP Annex III). This is an EMA guideline which is
adopted in the Arab Countries from the scientific point of
view.
2.4.3.1.1 Send For the cases that are merged under the master, send a Sender
nullification message to the PhV DB holder.
follow-up
/nullification For the case that is master, send the updated case to the
PhV DB holder as follow-up information. The merging &
transmission should be completed promptly and in any
case within 15 days of the date of receipt of the
information from the PhV DB holder that the cases were
considered to be possible duplicates. This date should be
treated as the date of receipt of most recent information for
regulatory reporting purposes.
2.4.3.1.2 End. The duplicates have now been removed from both the Sender
Sender’s system and that of the PhV DB holder and
only the master should be available for signal detection
and data quality analyses.
Unless follow-up information is received, then no
further steps need be taken.
2.4.3.2 Draft and send a Reply to the PhV DB holder who sent the communication Sender
response. informing that the cases are not duplicates.
2.4.3.2.1 Mark as “Not a Upon receipt of confirmation from the Sender DMT
duplicate”. organisation that the cases are not duplicates, mark the
cases as ―Not a duplicate‖ & go to step 3 (End).
GVP: Modules
VII.A. Introduction
Periodic safety update reports (PSURs) are pharmacovigilance documents intended to provide an
evaluation of the risk-benefit balance of a medicinal product for submission by marketing
authorisation holders at defined time points during the post-authorisation phase.
The legal requirements for submission of PSURs are established in national regulation. All
applicable legal requirements in this Module are referenced by the modal verb ―shall‖. Guidance for
the implementation of legal requirements is provided using the modal verb ―should‖.
This Module provides guidance on the preparation, submission and assessment of PSURs.
The scope, objectives, format and content of the PSUR are described in VII.B. The required format
and content of PSURs in the Arab Countries are based on those for PSUR described in the European
Good Pharmacovigilance Practice as well as for the Periodic Benefit Risk Evaluation Report
(PBRER) described in the ICH-E2C(R2) guideline. The PBRER format replaces the PSUR format
previously described in the ICH-E2C(R1). In line with the national legislation, the report is
described as PSUR in the GVP Modules in the Arab Countries.
The adoption of the "European Good Pharmacovigilance Practice" as a base for this guideline does
not undermine the right of a national medicines authority in the Arab Countries to have additional or
sometimes changed requirements. Multinational marketing authorization holders should be
attentive to these national requirements and take the necessary measure to comply with them.
Further details and guidance for the submission of PSURs in the Arab Countries, including the list
of Union references dates and frequency of submission are provided in VII.C. As this guideline was
based on the European Good Pharmacovigilance Practice; the "list of EU reference dates" is
adopted in this guideline as well. Hence, the PSURs submitted in the Arab Countries shall follow
the dates & frequency stated in the most updated version of this list, despite this, the national
medicines authority in the Arab Countries may request the submission of PSURs at any time or to
change as appropriate the submission frequency on the national level.
The single national assessment of PSURs is covered in VII.C.4. Details related to the quality system
are provided in VII.C.6.
Each marketing authorisation holder shall be responsible for submitting PSURs for its own products
to the national medicines authorities in the Arab Countries according to the following timelines:
within 70 calendar days of the data lock point (day 0) for PSURs covering intervals up to 12
months (including intervals of exactly 12 months); and
within 90 calendar days of the data lock point (day 0) for PSURs covering intervals in excess of
12 months;
the timeline for the submission of ad hoc PSURs requested by national medicines authorities will
normally be specified in the request, otherwise the ad hoc PSURs should be submitted within 90
calendar days of the data lock point.
It should be noted that detailed listings of individual cases shall not be included systematically. The
PSUR should focus on summary information, scientific safety assessment and integrated
benefit-risk evaluation.
The obligations imposed in respect of PSURs should be proportionate to the risks posed by
medicinal products. PSUR reporting should therefore be linked to the risk management systems of a
medicinal product (see Module V). The ―modular approach‖ of the PSUR described in VII.B.5.
aims to minimise duplication and improve efficiency during the preparation and review of PSURs
along with other regulatory documents such as the safety specification in the Risk Management Plan
(RMP), by enabling the common content of particular sections where appropriate to be utilised
interchangeably across different PSURs and RMPs.
Competent National medicines authorities in the Arab Countries shall assess PSURs to determine
whether there are new risks or whether risks have changed or whether there are changes to the
risk-benefit balance of medicinal products.
In order to avoid duplication of efforts and to prioritise the use of limited resources, a single
assessment of PSURs for different authorised medicinal products containing the same active
substance or the same combination of active substances should be performed in each Arab Country.
As part of the assessment, it should be considered whether further investigations need to be carried
out and whether any action concerning the marketing authorisations of products containing the
same active substance or the same combination of active substances, and their product information
is necessary.
PSURs for generic medicinal products, well-established use medicinal products, homeopathic
medicinal products and traditional herbal medicinal products are required to be submitted in the
Arab Countries (unless otherwise is announced by the national medicines authority in each Arab
Country).
intended, in the first instance, for notification of significant new safety or efficacy information or to
provide the means by which new safety issues are detected, (see Module IX and XII). It is
acknowledged that the review of the data in the PSUR may lead to new safety issues being
identified.
33
The ICH-E2C(R2) guideline should not serve to limit the scope of the information to be provided in the
benefit-risk evaluation of a medicinal product. Please refer to the applicable national laws and regulations in the
countries and regions.
For Arab Country specific requirements, see VII.C.5.
cumulative information available since the development international birth date (DIBD), the
date of first authorisation for the conduct of an interventional clinical trial in any country. For
the cases where the DIBD is unknown or the marketing authorisation holder does not have
access to data from the clinical development period, the earliest possible applicable date should
be used as starting point for the inclusion and evaluation of the cumulative information.
4. Summarising any risk minimisation actions that may have been taken or implemented during
the reporting interval, as well as risk minimisation actions that are planned to be implemented.
5. Outlining plans for signal or risk evaluations including timelines and/or proposals for additional
pharmacovigilance activities.
indications‖ components. In order to facilitate the assessment of benefit and risk-benefit balance by
indication in the evaluation sections of the PSUR, the reference product information document
should list all authorised indications in ICH countries or regions. When the PSUR is also submitted
to countries other than the ICH regions (e.g. Arab Countries) in which there are additional locally
authorised indications, these indications may be either added to the reference product information or
handled in the national appendix as considered most appropriate by the marketing authorization
holder and the national medicines authority in the concerned country. The basis for the benefit
evaluation should be the baseline important efficacy and effectiveness information summarised in
the PSUR section 17.1 (―Important baseline efficacy and effectiveness information‖).
Information related to a specific indication, formulation or route of administration should be clearly
identified in the reference product information.
The following possible options can be considered by the marketing authorisation holders when
selecting the most appropriate reference product information for a PSUR:
Company core data sheet (CCDS)
- It is common practice for marketing authorisation holders to prepare their own company
core data sheet which covers data relating to safety, indications, dosing, pharmacology, and
other information concerning the product. The core safety information contained within the
CCDS is referred to as the company core safety information (CCSI). A practical option for
the purpose of the PSUR is for each marketing authorisation holder to use the CCDS in
effect at the end of the reporting interval, as reference product information for both the risk
sections of the PSUR as well as the main authorised indications for which benefit is
evaluated.
- When the CCDS does not contain information on authorised indications, the marketing
authorisation holder should clearly specify which document is used as reference information
for the authorised indications in the PSUR.
Other options for the reference product information
- When no CCDS or CCSI exist for a product (e.g. where the product is authorised in only one
country or region, or for /generics), the marketing authorisation holder should clearly
specify the reference information being used. This may comprise national or regional
product information.
- Where the reference information for the authorised indications is a separate document to the
reference safety information (the core safety information contained within the reference
product information), the version in effect at the end of the reporting interval should be
included as an appendix to the PSUR (see VII.B.5.20.).
The marketing authorisation holder should continuously evaluate whether any revision of the
reference product information/reference safety information is needed whenever new safety
information is obtained during the reporting interval and ensure that significant changes made over
the interval are described in PSUR section 4 (―Changes to the reference safety information‖) and
where relevant, discussed in PSUR section 16 (―Signal and risk evaluation‖). These changes may
include:
changes to contraindications, warnings/precautions sections;
34
ICH-E2D Post-Approval Safety Data Management: Definitions and Standards for Expedited Reporting.
35
For PSURs submission, it is at the discretion of the QPPV to determine the most appropriate person to sign the
document according to the marketing authorisation holder structure and responsibilities. A statement confirming
the designation by the QPPV should be included. No delegation letters should be submitted.
1. Introduction
2. Worldwide marketing authorisation status
3. Actions taken in the reporting interval for safety reasons
a. Actions related to investigational uses (not applicable for generics)
b. Actions related to marketing experience
4. Changes to reference safety information
5. Estimated exposure and use patterns
5.1. Cumulative subject exposure in clinical trials (not applicable for generics)
5.2. Cumulative and interval patient exposure from marketing experience
6. Data in summary tabulations
6.1. Reference information
6.2. Cumulative summary tabulations of serious adverse events from clinical trials (not
applicable for generics)
6.3. Cumulative and interval summary tabulations from post-marketing data sources
7. Summaries of significant findings from clinical trials during the reporting interval (not
applicable for generics)
7.1. Completed clinical trials
7.2. Ongoing clinical trials
7.3. Long-term follow-up
7.4. Other therapeutic use of medicinal product
7.5. New safety data related to fixed combination therapies
8. Findings from non-interventional studies
9. Information from other clinical trials and sources
9.1. Other clinical trials (not applicable for generics)
9.2. Medication errors
10. Non-clinical Data (not applicable for generics)
11. Literature
12. Other periodic reports
13. Lack of efficacy in controlled clinical trials (not applicable for generics)
14. Late-breaking information
15. Overview of signals: new, ongoing or closed
16. Signal and risk evaluation
16.1. Summaries of safety concerns
An abridged PSUR, which is suitable for generic medicinal products in the Arab Countries, can be
used; the cover letter should state ''abridged PSUR''. Sections which are not required from generics
in the abridged PSUR should NOT be omitted instead state that it is not applicable for generics with
referral to this guideline. The not required section from generics in the abridged PSUR are indicated
in the aforeward content.
36
For PSURs covering multiple products, for practical reasons, this information may be provided in the PSUR
Cover Page (See Annex II)
VII.B.5.3. PSUR section “Actions taken in the reporting interval for safety reasons”
This section of the PSUR should include a description of significant actions related to safety that
have been taken worldwide during the reporting interval, related to either investigational uses or
marketing experience by the marketing authorisation holder, sponsors of clinical trial(s), data
monitoring committees, ethics committees or national medicines authorities that had either:
a significant influence on the risk-benefit balance of the authorised medicinal product; and/or
an impact on the conduct of a specific clinical trial(s) or on the overall clinical development
programme.
If known, the reason for each action should be provided and any additional relevant information
should be included as appropriate. Relevant updates to previous actions should also be summarised
in this section.
37
―Partial suspension‖ might include several actions (e.g. suspension of repeat dose studies, but continuation of
single dose studies; suspension of trials in one indication, but continuation in another, and/or suspension of a
particular dosing regimen in a trial but continuation of other doses). ICH-E2C(R2) guideline (see Annex IV).
warnings, precautions, serious adverse drug reactions, interactions, important findings from
ongoing or completed clinical trials and significant non-clinical findings (e.g. carcinogenicity
studies). Specific information relevant to these changes should be provided in the appropriate
sections of the PSUR.
single dose. Such data can be presented separately with an explanation as appropriate;
if the serious adverse events from clinical trials are presented by indication in the summary
tabulations, the patient exposure should also be presented by indication, where available;
for individual trials of particular importance, demographic characteristics should be provided
separately.
Examples of tabular format for the estimated exposure in clinical trials are presented in VII.
Appendix 1, Tables VII.2, VII.3 and VII.4.
VII.B.5.5.2. PSUR sub-section “Cumulative and interval patient exposure from marketing
experience”
Separate estimates should be provided for cumulative exposure (since the IBD), when possible, and
interval exposure (since the data lock point of the previous PSUR). Although it is recognised that it
is often difficult to obtain and validate exposure data, the number of patients exposed should be
provided whenever possible, along with the method(s) used to determine the estimate. Justification
should be provided if it is not possible to estimate the number of patients exposed. In this case,
alternative estimates of exposure, if available, should be presented along with the method(s) used to
derive them. Examples of alternative measures of exposure include patient-days of exposure and
number of prescriptions. Only if such measures are not available, measures of drug sales, such as
tonnage or dosage units, may be used. The concept of a defined daily dose may also be used to arrive
at patient exposure estimates.
The data should be presented according to the following categories:
the adverse event/reaction terms, the preferred term (PT) level and system organ class (SOC) should
be presented in the summary tabulations.
The seriousness of the adverse events/reactions in the summary tabulations should correspond to the
seriousness assigned to events/reactions included in the ICSRs using the criteria established in
ICH-E2A38 (see Annex IV). When serious and non-serious events/reactions are included in the
same ICSR, the individual seriousness per reaction should be reflected in the summary tabulations.
Seriousness should not be changed specifically for the preparation of the PSURs.
VII.B.5.6.2. PSUR sub-section “Cumulative summary tabulations of serious adverse events from
clinical trials”
This PSUR sub-section should provide background for the appendix that provides a cumulative
summary tabulation of serious adverse events reported in the marketing authorisation holder‘s
clinical trials, from the DIBD to the data lock point of the current PSUR. The marketing
authorisation holder should explain any omission of data (e.g. clinical trial data might not be
available for products marketed for many years). The tabulation(s) should be organised by
MedDRA SOC (listed in the internationally agreed order), for the investigational drug, as well as for
the comparator arm(s) (active comparators, placebo) used in the clinical development programme.
Data can be integrated across the programme. Alternatively, when useful and feasible, data can be
presented by trial, indication, route of administration or other variables. In some Arab Countries, it
may be accepted to use the WHO-Art terminology instead of MedDRA, consult with the national
medicines authorities.
This sub-section should not serve to provide analyses or conclusions based on the serious adverse
events.
The following points should be considered:
Causality assessment is generally useful for the evaluation of individual rare adverse drug
reactions. Individual case causality assessment has less value in the analysis of aggregate data,
where group comparisons of rates are possible. Therefore, the summary tabulations should
include all serious adverse events and not just serious adverse reactions for the investigational
drug, comparators and placebo. It may be useful to give rates by dose.
In general, the tabulation(s) of serious adverse events from clinical trials should include only
those terms that were used in defining the case as serious and non-serious events should be
included in the study reports.
The tabulations should include blinded and unblinded clinical trial data. Unblinded serious
adverse events might originate from completed trials and individual cases that have been
unblinded for safety-related reasons (e.g. expedited reporting), if applicable. Sponsors of clinical
38
ICH Topic E2A. Clinical safety data management: Definitions and standards for expedited reporting.
trials and marketing authorisation holders should not unblind data for the specific purpose of
preparing the PSUR.
Certain adverse events can be excluded from the clinical trials summary tabulations, but such
exclusions should be explained in the report. For example, adverse events that have been defined
in the protocol as ―exempt‖ from special collection and entry into the safety database because
they are anticipated in the patient population, and those that represent study endpoints, can be
excluded (e.g. deaths reported in a trial of a drug for congestive heart failure where all-cause
mortality is the primary efficacy endpoint, disease progression in cancer trials).
An example of summary tabulation of serious adverse events from clinical trials can be found in
VII. Appendix 1 Table VII.7.
signals: new, ongoing or closed‖). Evaluation of the signals, whether or not categorised as refuted
signals or either potential or identified risk, that were closed during the reporting interval should be
presented in PSUR section 16.2 (―Signal evaluation‖). New information in relation to any
previously known potential or identified risks and not considered to constitute a newly identified
signal should be evaluated and characterised in PSUR sections 16.3 (―Evaluation of risks and new
information‖) and 16.4 (―Characterisation of risks‖) respectively.
Findings from clinical trials not sponsored by the marketing authorisation holder should be
described in the relevant sections of the PSUR.
When relevant to the benefit-risk evaluation, information on lack of efficacy from clinical trials for
treatments of non-life-threatening diseases in authorised indications should also be summarised in
this section. Information on lack of efficacy from clinical trials with products intended to treat or
prevent serious or life-threatening illness should be summarised in section 13 (―Lack of efficacy in
controlled clinical trials‖) (VII.B.5.13).
Information from other clinical trials/study sources should be included in the PSUR sub-section 9.1
(―other clinical trials‖) (VII.B.5.9.1).
In addition, the marketing authorisation holder should include an appendix listing the sponsored
post-authorisation interventional trials with the primary aim of identifying, characterising, or
quantifying a safety hazard or confirming the safety profile of the medicinal product that were
completed or ongoing during the reporting interval. The listing should include the following
information for each trial:
study ID (e.g. protocol number or other identifier);
study title (abbreviated study title, if applicable);
study type (e.g. randomised clinical trial, cohort study, case-control study);
population studied, including country and other relevant population descriptors (e.g. paediatric
population or trial subjects with impaired renal function);
study start (as defined by the marketing authorisation holder) and projected completion dates;
status: ongoing (clinical trial has begun) or completed (clinical study report is finalised).
39
Examples of synopses can be found in ICH-E3: Structure and Content of Clinical Study Reports and CIOMS VII
(Council for International Organizations of Medical Sciences (CIOMS). Development Safety Update Report (DSUR):
Harmonizing the Format and Content for Periodic Safety Reporting During Clinical Trials - Report of CIOMS Working
Group VII). Geneva: CIOMS; 2006. http://www.cioms.ch/.
VII.B.5.7.5. PSUR sub-section “New safety data related to fixed combination therapies”
Unless otherwise specified by national or regional regulatory requirements, the following options
can be used to present data from combination therapies:
If the active substance that is the subject of the PSURs is also authorised or under development as
a component of a fixed combination product or a multi-drug regimen, this sub-section should
summarise important safety findings from use of the combination therapy.
If the product itself is a fixed combination product, this PSUR sub-section should summarise
important safety information arising from the individual components whether authorised or
under development.
The information specific to the combination can be incorporated into a separate section(s) of the
PSUR for one or all of the individual components of the combination.
during the reporting interval. (see VII.B.5.7. for the information that should be included in the
listing).
Summary information based on aggregate evaluation of data generated from patient support
programs may be included in this section when not presented elsewhere in the PSUR. As for other
information sources, the marketing authorisation holder should present signals or risks identified
from such information in the relevant sections of the PSUR.
VII.B.5.9. PSUR section “Information from other clinical trials and sources”
40
Uniform requirements for manuscripts submitted to biomedical journals. International Committee of Medical
Journal Editors. N Engl J Med. 1997 Jan 23;336(4):309-15. Available online:
http://www.nejm.org/doi/full/10.1056/NEJM199701233360422
41
Uniform Requirements for Manuscripts Submitted to Biomedical Journals: Writing and Editing for Biomedical
Publication [Updated April 2010] Publication Ethics: Sponsorship, Authorship, and Accountability, International
Committee of Medical Journal Editors. http://www.icmje.org/urm_full.pdf
42
―Signal‖ means information arising from one or multiple sources, including observations and experiments,
which suggests a new potentially causal association, or a new aspect of a known association between an
intervention and an event or set of related events, either adverse or beneficial, that is judged to be of sufficient
likelihood to justify verificatory action.
43
For the purpose of the PSUR, the term ―signal‖ in this section corresponds with the term ―validated signal‖
described in GVP Module IX.
A new signal refers to a signal that has been identified during the reporting interval. Where new
clinically significant information on a previously closed signal becomes available during the
reporting interval of the PSUR, this would also be considered a new signal on the basis that a new
aspect of a previously refuted signal or recognised risk warrants further action to verify. New
signals may be classified as closed or ongoing, depending on the status of signal evaluation at the
end of the reporting interval of the PSUR.
Examples of new signals would therefore include new information on a previously:
Close and refuted signal, which would result in the signal being re-opened.
Identified risk where the new information suggests a clinically significant difference in the
severity or frequency of the risk (e.g. transient liver enzyme increases are identified risks and
new information indicative of a more severe outcome such as hepatic failure is received, or
neutropenia is an identified risk and a well documented case report of agranulocytosis with no
presence of possible alternative causes is received).
Identified risk for which a higher frequency or severity of the risk is newly found (e.g. in an
indicated subpopulation).
Potential risk which, if confirmed, would warrant a new warning, precaution, a new
contraindication or restriction in indication(s) or population or other risk minimisation activities.
Within this section, or as an appendix the marketing authorisation holder should provide a
tabulation of all signals ongoing or closed at the end of the reporting interval. This tabulation should
include the following information:
a brief description of the signal;
date when the marketing authorisation holder became aware of the signal;
status of the signal at the end of the reporting interval (close or ongoing);
date when the signal was closed, if applicable;
source of the signal;
a brief summary of the key data;
plans for further evaluation; and
actions taken or planned.
An example of tabulation of signals can be found in VII. Appendix 2.
The detailed signal assessments for closed signals are not to be included in this section but instead
should be presented in sub-section 16.2 (―Signal evaluation‖) of the PSUR.
Evaluation of new information in relation to any previously known identified and potential risks and
not considered to constitute a new signal should be provided in PSUR sub-section 16.3 (―Evaluation
of risks and new information‖).
When a medicines authority (worldwide) has requested that a specific topic (not considered a
signal) be monitored and reported in a PSUR, the marketing authorisation holder should summarise
the result of the analysis in this section if it is negative. If the specific topic becomes a signal, it
should be included in the signal tabulation and discussed in sub-section 16.2 (―Signal evaluation‖).
44
ICH-E2E – Pharmacovigilance planning (see Annex IV).
identified benefit information that support the characterisation of benefit described in sub-section
17.3 (―Characterisation of benefits‖) that in turn supports the benefit-risk evaluation in section 18
(―Integrated benefit-risk analysis for authorised indications‖).
succinct.
This sub-section should provide a concise but critical evaluation of the strengths and limitations of
the evidence on efficacy and effectiveness, considering the following when available:
a brief description of the strength of evidence of benefit, considering comparator(s), effect size,
statistical rigor, methodological strengths and deficiencies, and consistency of findings across
trials/studies;
new information that challenges the validity of a surrogate endpoint, if used;
clinical relevance of the effect size;
generalisability of treatment response across the indicated patient population (e.g. information
that demonstrates lack of treatment effect in a sub-population);
adequacy of characterization of dose-response;
duration of effect;
comparative efficacy; and
a determination of the extent to which efficacy findings from clinical trials are generalisable to
patient populations treated in medical practice.
VII.B.5.18.1. PSUR sub-section “Benefit-risk context - medical need and important alternatives”
This sub-section of the PSUR should provide a brief description of the medical need for the
medicinal product in the authorised indications and summarised alternatives (medical, surgical or
other; including no treatment).
information, not all benefits and risks contribute importantly to the overall benefit-risk
evaluation, therefore, the key benefits and risks considered in the evaluation should be specified.
The key information presented in the previous benefit and risk section/sub-sections should be
carried forward for integration in the benefit-risk evaluation.
Consider the context of use of the medicinal product: the condition to be treated, prevented, or
diagnosed; its severity and seriousness; and the population to be treated (relatively healthy;
chronic illness, rare conditions).
With respect to the key benefit(s), consider its nature, clinical importance, duration, and
generalisability, as well as evidence of efficacy in non-responders to other therapies and
alternative treatments. Consider the effect size. If there are individual elements of benefit,
consider all (e.g. for therapies for rheumatoid arthritis: reduction of symptoms and inhibition of
radiographic progression of joint damage).
With respect to risk, consider its clinical importance, (e.g. nature of toxicity, seriousness,
frequency, predictability, preventability, reversibility, impact on patients), and whether it arose
from clinical trials in unauthorised indications or populations, off-label use, or misuse.
The strengths, weaknesses, and uncertainties of the evidence should be considered when
formulating the benefit-risk evaluation. Describe how uncertainties in the benefits and risks
impact the evaluation. Limitations of the assessment should be discussed.
Provide a clear explanation of the methodology and reasoning used to develop the benefit-risk
evaluation:
The assumptions, considerations, and judgement or weighting that support the conclusions of the
benefit-risk evaluation should be clear.
If a formal quantitative or semi-quantitative assessment of benefit-risk is provided, a summary of
the methods should be included.
Economic considerations (e.g. cost-effectiveness) should not be considered in the benefit-risk
evaluation.
When there is important new information or an ad hoc PSUR has been requested, a detailed
benefit-risk analysis should be presented based on cumulative data. Conversely, where little new
information has become available during the reporting interval, the primary focus of the benefit-risk
evaluation might consist of an evaluation of updated interval safety data.
authority(ies). This may include proposals for additional risk minimisation activities.
For products with a pharmacovigilance or risk management plan, the proposals should also be
considered for incorporation into the pharmacovigilance plan and/or risk minimisation plan, as
appropriate (see Module V).
Based on the evaluation of the cumulative safety data and the risk-benefit analysis, the marketing
authorisation holder shall draw conclusions in the PSUR as to the need for changes and/or actions,
including implications for the approved summary of product characteristics (SmPC) for the
product(s) for which the PSUR is submitted.
Proposed changes to the reference product information should be described in this section of the
PSUR. The national appendix should include proposals for product information (SmPC and
package leaflet) together with information on ongoing changes when applicable.
failure to provide an explicit evaluation of the risk-benefit balance of the medicinal product;
failure to provide adequate proposals for the local authorised product information.
Any significant deviation from the procedures relating to the preparation or submission of PSURs
should be documented and the appropriate corrective and preventive action should be taken. This
documentation should be available at all times.
When marketing authorisation holders are involved in contractual arrangements (e.g.
licensor-licensee), respective responsibilities for preparation and submission of the PSUR to the
medicines authorities should be clearly specified in the written agreement.
When the preparation of the PSUR is delegated to third parties, the marketing authorisation holder
should ensure that they are subject to a quality system compliant with the current legislation.
Explicit procedures and detailed agreements should exist between the marketing authorisation
holder and third parties. The agreements may specifically detail the options to audit the PSUR
preparation process.
information and same format & content) hence, the same PSUR is submitted to several authorities
worldwide.
Therefore for the purpose of not reinventing the wheel and as this guideline was based on the
European Good Pharmacovigilance Practice; the "list of EU reference dates" (EURD) is adopted in
the context of this guideline. Hence the PSURs submitted in the Arab Countries shall follow the
dates & frequency stated in the most updated version of the list; this does not undermine the right of
a national medicines authority in the Arab Countries to request the submission of PSURs at any time
or to change as appropriate the submission frequency on the national level.
For active substances or combination of active substances not included in the EURD list see
VII.C.1.2.2.
The following subsections define the "EU reference dates list" and its process in the EU with the
purpose to provide more understanding about it and how the same list will be adopted and applied in
the Arab Countries.
45
The initial EU reference dates list was adopted by the EMA in September 2012 and was published on 01
October 2012.
health.
Single assessment and reassessment of the risk-benefit balance of an active substance based on
all available safety data:
The list enables the harmonisation of PSUR submissions for medicinal products containing the
same active substance or the same combination of active substances.
A single PSUR assessment provides a mechanism for evaluating the totality of available data on
the benefits and risks of an active substance or combination of active substances. This single
assessment on the national level is important in avoiding duplication of efforts and in prioritising
the use of limited resources.
post-authorisation experiences);
significant changes to the product (e.g. new indication has been authorised, new pharmaceutical
form or route of administration broadening the exposed patient population);
vulnerable patient populations/poorly studied patient populations, missing information (e.g.
children, pregnant women) while these populations are likely to be exposed in the
post-authorisation setting;
signal of/potential for misuse, medication error, risk of overdose or dependency;
the size of the safety database and exposure to the medicinal product;
medicinal products subjected to additional monitoring.
Any change in the criteria listed above for a given active substance or combination of active
substances may lead to an amendment of the list of EU reference dates (e.g. increase of the
frequency for PSUR submission).
Medicinal Product
Is the
Does the marketing No No
substance or Submit request to
authorisation
combination the NMA to define
include a condition
included in frequency
on the frequency of
EURD list?
PSURs submission?
Yes
Yes
Yes
No Is the substance
Consult with the or combination
NMA included in
EURD list?
Yes
If applicable, variation of
the marketing authorisation
to update the frequency as
published in the EURD list
The data lock points included in the "list of EU references dates" enable the synchronisation of
PSURs submission and permit the single assessment on the national level. These data lock points
are fixed on a certain date of the month, and should be used to determine the submission date of the
PSUR.
Unless otherwise specified in the list of EU reference dates and frequency of submission, or agreed
with national medicines authority in the Arab country concerned, as appropriate, a single PSUR
shall be prepared for all medicinal products containing the same active substance and authorised for
one marketing authorisation holder. The PSUR shall cover all indications, routes of administration,
dosage forms and dosing regimens, irrespective of whether authorised under different names.
Where relevant, data relating to a particular indication, dosage form, route of administration or
dosing regimen shall be presented in a separate section of the PSUR and any safety concerns shall
be addressed accordingly.
The adoption of the list of EU reference dates for the submission of PSURs in Arab Countries does
not undermine the right of a national medicines authority in the Arab Countries to request the
submission of PSURs at any time or to change as appropriate the submission frequency on the
national level.
VII.C.1.2.2. Submission of PSURs in case of active substances not included in the EURD list
For medicinal products containing an active substance or a combination of active substances NOT
included in the EU reference dates list, PSURs shall be submitted according to the PSUR frequency
defined in the marketing authorisation or if not specified, the MAH shall submit request to the
national medicines authority in the Arab Country concerned to define the frequency and dates of
submission of PSURs. See figure VII.2.
The national medicines authorities should maintain a track for those active substance and their
defined frequencies & dates as a complementary to the EURD list. In addition, the national
medicines authority should generalise those frequencies & dates for other national medicines
containing the same active substance or a combination of active substances to support the "national
PSUR single assessment" procedure.
The national medicines authority in the Arab Country concerned may use (if no specific concern
about the safety) the following standard submission schedule to define the frequency & date of
PSURs submission for those substances:
at 6 months intervals once the product is authorised, even if it is not marketed;
once a product is marketed, 6 monthly PSUR submission should be continued following initial
placing on the market for 2 years, then once a year for the following 2 years and thereafter at
3-yearly intervals.
VII.C.1.2.3. Medicinal products with conditioned PSURs submission frequency in the marketing
authorisation
Authorised medicinal product for which the frequency and dates of submission of PSURs are laid
down as a condition in its marketing authorisation; see figure VII.2.
if this conditioned marketing authorisation is granted BEFORE the EURD list becomes into
effect in the Arab Country concerned, and, if the active substance or a combination of active
substances of this product is included in the "EU reference dates list", then the MAH should
submit variation -as appropriate- to update the frequency as published in the EURD list.
if this conditioned marketing authorisation is granted AFTER the EURD list becomes into effect
in the Arab Country concerned, then the MAH should follow the frequency laid down in the
marketing authorisation.
Afterward, any changes to the dates and frequencies of submission of PSURs specified in the list
take effect six months after the date of the publication. Where appropriate, marketing
authorisation holders shall submit the relevant variation in order to reflect the changes in their
marketing authorisation, unless the marketing authorisation contains a direct cross reference to
the list of EU references dates adopted in the Arab Countries.
VII.C.1.2.4. Submission of PSURs for generic, well-established use, traditional herbal and
homeopathic medicinal products
As a general rule, PSURs for these kind of medicinal products are required to be submitted in the
Arab Countries (unless otherwise is announced by the national medicines authority in each Arab
Country).
The multinational marketing authorization holders for any of these kinds of medicinal products
which sometimes are exempted from submitting PSURs routinely for these products in the
European Union; should be attentive to the national requirements in the Arab Countries as this
European exemption is NOT applied in the Arab Countries (unless otherwise is announced by the
national medicines authority).
If for any reason the PSURs of some of these products are no longer required by the national
medicines authority in any Arab Country to be submitted routinely, it is expected that marketing
authorisation holders will continue to evaluate the safety of their products on a regular basis and
report any new safety information that impacts on the risk-benefit balance or the product
information (See Module VI and Module IX).
may be accompanied by a request for an analysis of individual case safety reports, (including
information on numbers of cases, details of fatal cases and as necessary, analysis of non-serious
cases), where necessary for the scientific evaluation. Information on the context or rationale for the
request should generally be provided.
Following the assessment of PSURs, the medicines authority in the Arab Country should consider
whether any action concerning the marketing authorisation for the medicinal products containing
the concerned active substance or combination of active substances is necessary (e.g. add a new
contraindication, a restriction of the indication or a reduction of the recommended dose, the need to
conduct a post-authorisation safety study, request an update of the RMP, review of safety issues
and/or close monitoring of events of interest …etc). The national medicines authority should vary,
suspend or revoke the marketing authorization when applicable according to the appropriate
procedure at national level.
Furthermore, marketing authorisation holders are reminded of their obligation to keep their
marketing authorisation up to date.
Amendments to the SmPC, package leaflet and labelling as a result of the PSUR assessment should
be implemented through the appropriate variation.
When the proposals for the product information include new adverse reactions in section 4.8
(―Undesirable effects‖) of the SmPC, or modifications in the description, frequency and severity of
the existing reactions, marketing authorisation holders should provide in the relevant sections of the
PSUR appropriate information to allow the adequate description and classification of the frequency
of the adverse reactions. If other sections of the SmPC (e.g. SmPC section 4.4 ―Special warnings
and precautions for use‖) are considered to be updated, clear proposals should be provided for the
medicines authorities in the Arab Country concerned to consider during the PSUR assessment. The
proposals should be included in the PSUR national appendix (VII.C.5.).
The outcome of the PSUR assessment should incorporate the new safety warnings and key risk
minimisation recommendations, arising from the assessment of the data in the PSUR, to be included
in the relevant sections of the product information.
The assessment results and conclusions of the medicines authority in the Arab Country should be
provided to the marketing authorisation holder.
Section 3 – ―Actions taken in the reporting Part II, module SV – ―Post-authorisation experience‖, section
interval for safety reasons‖ ―Regulatory and marketing authorisation holder action for
safety reason‖
Sub-section 5.2 – ―Cumulative and interval Part II, module SV – ―Post-authorisation experience‖, section
patient exposure from marketing ―Non-study post-authorisation exposure‖
experience‖
Sub-section 16.1 – ―Summary of safety Part II, module SVIII – ―Summary of the safety concerns‖ (as
concerns‖ included in the version of the RMP which was current at the
beginning of the PSUR reporting interval)
Sub-section 16.4 – ―Characterisation of Part II, Module SVII – ―Identified and potential risks‖
risks‖
Sub-section 16.5 – ―Effectiveness of risk Part V – ―Risk minimisation measures‖, section ―Evaluation of
minimisation (if applicable)‖ the effectiveness of risk minimisation activities‖
appendix 1 of the PSUR (see VII.B.5.20.). When a meaningful differences exist between this
reference safety information (e.g. CCDS or CCSI) and the safety information in the
national product information (national SmPC and package leaflet) approved in the Arab
Country concerned, a brief comment should be prepared by the company, describing these
local differences with track change version.
The reference product information document should list all authorised indications in ICH
countries or regions. When there are additional locally authorised indications in the Arab
Country concerned, these indications may be either added to the reference product information or
handled in the national appendix as considered most appropriate by the marketing authorization
holder and the national medicines authority in the concerned country.
Entries should be listed in chronological order of 1st regulatory authorizations. For multiple
authorizations in the same country (e.g., new dosage forms), the IBD for the active substance and
for all PSURs should be the first (initial) authorization date.
This is a cumulative table; accordingly entries must not be removed from the table e.g. if the product
in no more authorized; instead MAH shall changes the relevant information in table. Fictious
examples for different cases are shown on the table below
Current authorisation
Refusal date
explanation
Comments/
Indication
Country
status
Date
any)
2-3-1990 UK <name> Tablet <indicatio authoris 2-3-1995 Marketed
n> ed renewal 7-9-1990
Typically, indications for use, populations treated (e.g. children vs. adults) and dosage forms will be
the same in many or even most countries where the product is authorised. However, when there are
important differences, which would reflect different types of patient exposure, such information
should be noted. This is especially true if there are meaningful differences in the newly reported
safety information that are related to such different exposures.
If more convenient and useful, separate regulatory status tables for different product uses or forms
should be utilized.
VII.C.6.1. Quality systems and record management systems at the level of the
marketing authorisation holder
Specific quality system procedures and processes shall be in place in order to ensure the update of
product information by the marketing authorisation holder in the light of scientific knowledge,
including the assessments and recommendations.
It is the responsibility of the marketing authorisation holder to check regularly the list of EU
reference dates and frequency of submission (adopted also by Arab Countries) published in the
official website of each national medicines authority in Arab countries/ EMA website to ensure
compliance with the PSUR reporting requirements for their medicinal products (see VII.C.1.).
Systems should be in place to schedule the production of PSURs according to:
the list of EU reference dates and frequency of PSURs submission; or
the conditions laid down in the national marketing authorisation; or
as defined by the national medicines authority in the Arab Country concerned as applicable
(without any conditions in their marketing authorisation or not included in the list of EU
references dates and frequency of submission; or
ad hoc requests for PSURs by a medicines authority in an Arab Country.
For those medicinal products where the submission of an RMP is not required, the marketing
authorisation holder should maintain on file a specification of important identified risks, important
potential risks and missing information in order to support the preparation of the PSURs.
The marketing authorisation holder should have procedures in place to follow the requirements
established by the medicines authority(ies) in the Arab Country(s) concerned for the submission of
PSURs.
The QPPV shall be responsible for the establishment and maintenance of the pharmacovigilance
system and therefore should ensure that the pharmacovigilance system in place enables the
compliance with the requirements established for the production and submission of PSURs. In
relation to the medicinal products covered by the pharmacovigilance system, specific additional
responsibilities of the QPPV in relation to PSURs should include:
ensuring the necessary quality, including the correctness and completeness, of the data submitted
in the PSURs;
ensuring full response according to the timelines and within the procedure agreed (e.g. next
PSUR) to any request from the national medicines authorities in Arab Countries concerned
related to PSURs;
awareness of the PSUR and assessment report conclusions and the decisions of the concerned
national medicines authority in order to ensure that appropriate action takes place.
The record retention times for product-related documents in Module I also apply to PSURs and
source documents related to the creation of PSURs, including documents related to actions taken for
safety reasons, clinical trials and post-authorisation studies, relevant benefit information and
documents utilised for the calculation of patient exposure.
VII.C.6.2. Quality systems and record management systems at the level of the
medicines authorities in Arab Countries
Each medicines authority in the Arab Countries shall have in place a pharmacovigilance system for
the surveillance of medicinal products and for receipt and evaluation of all pharmacovigilance data
including PSURs. For the purpose of operating its tasks relating to PSURs in addition to the
pharmacovigilance system the national medicines authorities in Arab Countries should implement a
quality system (see Module I).
National medicines authorities in the Arab Countries should monitor marketing authorisation
holders for compliance with regulatory obligations for PSURs. Additionally, medicines authorities
should take in cases of non-compliance the appropriate regulatory actions as required (e.g.
variation, suspension or revocation…etc). Medicines authorities in Arab Countries may exchange
information in case of MAH non-compliance.
Where MAH's tasks related to PSUR procedures are delegated to third parties, the national
medicines authorities in Arab Countries should ensure that they are subject to a quality system in
compliance with the obligations provided by the national regulation/legislation.
The national medicines authorities should have in place a process to technically validate the
completeness of PSUR submissions.
Data from the ―National Pharmacovigilance and Safety reports database‖ (e.g. line listings and
summary tabulations) should be retrieved and utilised as appropriate to support the PSUR
assessment.
Written procedures should reflect the different steps to follow for the maintenance and publishing of
the list of dates and frequency of submission of PSURs.
The record retention times for product-related documents in Module I also apply to PSUR- system
related documents (e.g. standard operating procedures) and PSUR -related documents (e.g. PSURs,
assessment reports, the data retrieved from the ―National Pharmacovigilance and Safety reports
database‖ or other data used to support the PSUR assessment).
renewal application. The clinical overview should include an addendum containing the relevant
sections for the re-assessment of the risk-benefit balance of the medicinal product. These sections
are identified below.
Addendum to Clinical Overview:
A critical discussion addressing the current benefit/risk balance for the product on the basis of a
consolidated version of safety/efficacy data accumulated since the initial MAA or the last renewal,
taking into account Periodic Safety Update Reports (PSURs) submitted, suspected adverse
reactions reports, additional pharmacovigilance activities and the effectiveness of risk minimisation
measures contained in the RMP, if applicable. In addition, it should make reference to any relevant
new information in the public domain e.g. literature references, clinical trials and clinical
experience, new treatments available, which may change the outcome of the benefit/risk evaluation
at the time of the original authorisation or last renewal.
The information shall include both positive and negative results of clinical trials and other studies in
all indications and populations, whether or not included in the marketing authorisation, as well as
data on the use of the medicinal product where such use is outside the terms of the marketing
authorisation.
This Addendum should be signed and accompanied by the CV of the expert. The clinical expert
should have the necessary technical or professional qualifications and may, but should not
necessarily, be the same qualified person responsible for pharmacovigilance.
In any event, a clear conclusive statement is required from the clinical expert (detailed below).
The Addendum to the Clinical Overview should contain the following information**:
History of pharmacovigilance system inspections (date, inspecting authority, site inspected, type
of inspection and if the inspection is product specific, the list of products concerned) and an
analysis of the impact of the findings overall on the benefit/risk balance of the medicinal product.
Worldwide marketing authorisation status: overview of number of countries where the product
has been approved and marketed worldwide.
Actions taken for safety reasons (worldwide) during the period covered since the initial
marketing authorisation or since the last renewal until 90 days prior to renewal submission:
description of significant actions related to safety that had a potential influence on the
benefit-risk balance of the approved medicinal product (e.g. suspension, withdrawal, temporary
halt or premature ending of clinical trial for safety reasons, issue requiring communication to
healthcare professionals…).
Significant changes made to the Reference Information (RI) during the period covered since the
initial marketing authorisation or since the last renewal. A track changes version of the document
identifying the changes made during the period covered since the initial marketing authorisation
or since the last renewal should also be provided until 90 days prior to renewal submission.
Meaningful differences between the RI and the proposals for the Summary of Product
Characteristics. A proposed SmPC, Package leaflet and Labelling should also be provided
Estimated exposure and used patterns: data on cumulative exposure of subjects in clinical trials
as well as of patients from marketing exposure. If the marketing authorisation holder becomes
aware of a pattern of use of the medicinal product considered relevant for the implementation of
the safety data, a brief description should be provided; such patterns may include in particular
off-label use.
Data in summary tabulations: Summary tabulations of serious adverse events from clinical trials
as well as summary tabulations of adverse reactions from post-marketing data sources reported
during the period covered since the initial marketing authorisation or since the last renewal until
90 days prior to renewal submission.
Summaries of significant safety and efficacy findings from clinical trials and non-interventional
studies: description of any significant safety findings that had an impact on the conduct of
clinical trials or non-interventional studies. It should also address whether milestones from
post-authorisation safety studies, post-authorisation efficacy studies, studies from the RMP
pharmacovigilance plan and studies conducted as condition and obligations of the marketing
authorisation, have been reached in accordance with agreed timeframes.
Literature: review of important literature references published during the period covered since
the initial marketing authorisation or since the last renewal until 90 days prior to renewal
submission that had a potential impact on the benefit/risk of the medicinal product.
Risk evaluation: the MAH should summarise any information related to important safety issues,
evaluation and characterisation of risks as well as effectiveness of risk minimisations for the
period covered since the initial marketing authorisation or since the last renewal until 90 days
prior to renewal submission.
Benefit evaluation: the MAH should summarise important efficacy and effectiveness
information (including information on lack of efficacy) for the period covered since the initial
marketing authorisation or since the last renewal until 90 days prior to renewal submission.
Benefit-risk balance: a discussion on the benefit-risk balance for the approved indication should
be presented, based on the above information.
Late-breaking information: The MAH should summarise the potentially important safety,
efficacy and effectiveness findings that arise after the data lock point but during the period of
preparation of the addendum to the clinical overview.
** Marketing authorisation holders are advised to consider the Good Vigilance Practice Module on
PSURs as guidance for the preparation of the above sections of the clinical overview.
The Clinical Expert Statement should:
Confirm that no new clinical data are available which change or result in a new risk-benefit
evaluation.
Confirm that the product can be safely renewed at the end of a x-year period (check national
regulations) for an unlimited period, or any action recommended or initiated should be specified
and justified.
Confirm that the authorities have been kept informed of any additional data significant for the
assessment of the benefit/risk ration of the product concerned.
Confirm that the product information is up to date with the current scientific knowledge
including the conclusions of the assessments and recommendations made publicly available.
Medicinal product
Comparator
Placebo
Table VII.3. Cumulative subject exposure to investigational drug from completed clinical trials
by age and sex
Number of subjects
Table VII.4. Cumulative subject exposure to investigational drug from completed clinical trials
by racial/ethnic group
Asian
Black
Caucasian
Other
Unknown
Total
Data from completed trials as of <insert date>
Male
Female
2 to ≤16
>16 to 65
>65
Unknown
<40
≥40
Unknown
Intravenous
Oral
concerned
Arab country
EU
Japan
Colombia
US/Canada
Other
Overall
e.g.
Depression
e.g.
Migraine
Table VII.5 includes cumulative data obtained from day/month/year throughout day/month/year,
where available
Female
2 to ≤16
>16 to 65
>65
Unknown
<40
≥40
Unknown
Intravenous
Oral
concerned
Arab country
EU
Japan
Colombia
US/Canada
Other
e.g.
Depression
e.g.
Migraine
Table VII. 6 includes interval data obtained from day/month/year throughout day/month/year
Table VII.7. Cumulative tabulation of serious adverse events from clinical trials
Table VII.8. Numbers of adverse reactions by preferred term from post-authorisation sources*
MedDRA Non-interventional
Spontaneous, including medicines authorities post-marketing study
SOC
(worldwide) and literature and reports from other
PT solicited sources **
Total
Serious Non-serious Serious
Spontaneous
<SOC 1>
<PT>
<PT>
<PT>
<SOC 2>
<PT>
<PT>
<PT>
<PT>
Explanatory notes:
Signal term:
A brief descriptive name of a medical concept for the signal. This may evolve and be refined as
the signal is evaluated. The concept and scope may or may not be limited to specific MedDRA
term(s), depending on the source of signal.
Date detected:
Month and year the marketing authorisation holder became aware of the signal.
Status:
Ongoing: Signal under evaluation at the data lock point of the PSUR. Anticipated completion
date, if known, should be provided.
Closed: Signal for which evaluation was completed before the data lock point of the PSUR.
Note: A new signal of which the marketing authorisation holder became aware during the reporting
interval may be classified as closed or ongoing, depending on the status of the signal evaluation at
the end of the reporting interval of the PSUR.
Date closed:
Month and year when the signal evaluation was completed.
Source of signal:
Data or information source from which a signal arose. Examples include, but may not be limited
to, spontaneous reports, clinical trial data, scientific literature, and non-clinical study results, or
information request or inquiries from a medicines authority (worldwide).
GVP: Modules
VIII.A. Introduction
A post-authorisation safety study (PASS) is defined as any study relating to an authorised medicinal
product conducted with the aim of identifying, characterising or quantifying a safety hazard,
confirming the safety profile of the medicinal product, or of measuring the effectiveness of risk
management measures.
A PASS may be initiated, managed or financed by a marketing authorisation holder voluntarily, or
pursuant to an obligation imposed by a medicines authority.
This Module concerns PASS which are clinical trials or non-interventional studies and does not
address non-clinical safety studies.
A PASS is non-interventional if the following requirements are cumulatively fulfilled:
the medicinal product is prescribed in the usual manner in accordance with the terms of the
marketing authorisation;
the assignment of the patient to a particular therapeutic strategy is not decided in advance by a
trial protocol but falls within current practice and the prescription of the medicine is clearly
separated from the decision to include the patient in the study; and
no additional diagnostic or monitoring procedures are applied to the patients and epidemiological
methods are used for the analysis of collected data.
Non-interventional studies are defined by the methodological approach used and not by its scientific
objectives. Non-interventional studies include database research or review of records where all the
events of interest have already happened (this may include case-control, cross-sectional, cohort or
other study designs making secondary use of data). Non-interventional studies also include those
involving primary data collection (e.g. prospective observational studies and registries in which the
data collected derive from routine clinical care), provided that the conditions set out above are met.
In these studies, interviews, questionnaires and blood samples may be performed as part of normal
clinical practice.
If a PASS is a clinical trial (i.e. interventional study); the national regulation for pharmacovigilance
of clinical trials and the national rules governing interventional clinical trials of medicinal products
in each Arab Country shall be followed.
The purposes of this Module are to:
provide general guidance for the transparency, scientific standards and quality standards of
non-interventional PASS conducted by marketing authorisation holders voluntarily or pursuant
to an obligation imposed by a medicines authority (VIII.B.);
describe procedures whereby medicines authorities may impose to a marketing authorisation
holder an obligation to conduct a clinical trial or a non-interventional study (VIII.C.2.), and the
impact of this obligation on the risk management system (VIII.C.3);
describe procedures that apply to non-interventional PASS imposed as an obligation for the
protocol oversight and reporting of results (VIII.C.4.) and for changes to the marketing
authorisation following results (VIII.C.5.).
In this Module, all applicable legal requirements are usually identifiable by the modal verb ―shall‖.
Guidance for the implementation of legal requirements is provided using the modal verb ―should‖.
Under this module; the role and responsibilities of the national medicines authority include- among
others- the role of the national scientific research ethics committee which is in some Arab Countries
not a part of the national medicines authority, in such cases collaboration between the national
medicines authority and the national scientific research ethics committee should take place.
VIII.B.1. Scope
The guidance in VIII.B. applies to non-interventional PASS which are initiated, managed or
financed by a marketing authorisation holder and conducted in the Arab Country concerned. This
guidance should also be used for studies conducted outside the Arab Country concerned which have
been imposed or required by this medicines authority (studies defined in Module V).
Where applicable, legal requirements which are applicable to studies conducted pursuant to an
obligation are recommended to studies conducted voluntarily in order to support the same level of
transparency, scientific standards and quality standards for all PASS. This applies, for example, to
the format of study protocols, abstracts and final study reports and to the communication of study
information to national medicines authorities. Where relevant, a distinction is made in the text
between situations where the provision of the guidance represents a legal requirement or a
recommendation.
This guidance apply to studies initiated, managed or financed by a marketing authorisation holder as
well as those conducted by a third party on behalf of the marketing authorisation holder.
This guidance applies to studies that involve primary collection of safety data directly from patients
and health care professionals and those that make secondary use of data previously collected from
patients and health care professionals for another purpose.
VIII.B.2. Terminology
Date at which a study commences: date of the start of data collection.
Start of data collection: the date from which information on the first study subject is first recorded in
the study dataset or, in the case of secondary use of data, the date from which data extraction starts.
Simple counts in a database to support the development of the study protocol, for example to inform
the sample size and statistical precision of the study, are not part of this definition.
End of data collection: the date from which the analytical dataset is completely available.
Analytical dataset: the minimum set of data required to perform the statistical analyses leading to
the results for the primary objective(s) of the study.
Substantial amendment to the study protocol: amendment to the protocol likely to have an impact on
the safety, physical or mental well-being of the study participants or that may affect the study results
and their interpretation, such as changes to the primary or secondary objectives of the study, to the
study population, to the sample size, to the definitions of the main exposure, outcome and
confounding variables and to the analytical plan.
VIII.B.3. Principles
A post-authorisation study should be classified as a PASS when the main aim for initiating the study
includes any of the following objectives:
to quantify potential or identified risks, e.g. to characterise the incidence rate, estimate the rate
ratio or rate difference in comparison to a non-exposed population or a population exposed to
another drug or class of drugs, and investigate risk factors and effect modifiers;
to evaluate risks of a medicinal product used in patient populations for which safety information
is limited or missing (e.g. pregnant women, specific age groups, patients with renal or hepatic
impairment);
to evaluate the risks of a medicinal product after long-term use;
to provide evidence about the absence of risks;
to assess patterns of drug utilisation that add knowledge on the safety of the medicinal product
(e.g. indication, dosage, co-medication, medication errors);
to measure the effectiveness of a risk minimisation activity.
Whereas the PASS design should be appropriate to address the study objective(s), the classification
of a post-authorisation study as a PASS is not constrained by the type of design chosen if it fulfils
the criteria as set in definition of the PASS (see VIII.A). For example, a systematic literature review
or a meta-analysis may be considered as PASS depending on their aim.
Relevant scientific guidance should be considered by marketing authorisation holders and
investigators for the development of study protocols, the conduct of studies and the writing of study
reports, and by national medicines authorities for the evaluation of study protocols and study
reports. Relevant scientific guidance includes the ENCePP Guide on Methodological Standards in
Pharmacoepidemiology46, the ENCePP Checklist for Study Protocols 46, the Guideline on Conduct
of Pharmacovigilance for Medicines Used by the Paediatric Population for studies conducted in
children, and the Guidelines for Good Pharmacoepidemiology Practices of the International Society
of Pharmacoepidemiology (ISPE GPP)47.
For studies that are funded by a marketing authorisation holder, including studies developed,
conducted or analysed fully or partially by investigators who are not employees of the marketing
authorisation holder, the marketing authorisation holder should ensure that the investigators are
qualified by education, training and experience to perform their tasks. The research contract
between the marketing authorisation holder and investigators should ensure that the study meets its
regulatory obligations while permitting their scientific expertise to be exercised throughout the
research process. In the research contract, the marketing authorisation holder should consider the
46
http://www.encepp.eu/standards_and_guidances/index.html
47
http://www.pharmacoepi.org/resources/guidelines_08027.cfm
48
provisions of the ENCePP Code of Conduct , and address the following aspects:
rationale, main objectives and brief description of the intended methods of the research to be
carried out by the investigator(s);
rights and obligations of the investigator(s) and marketing authorisation holder;
clear assignment of tasks and responsibilities;
procedure for achieving agreement on the study protocol;
provisions for meeting the marketing authorisation holder‘s pharmacovigilance obligations,
including the reporting of adverse reactions and other safety data by investigators, where
applicable;
intellectual property rights arising from the study and access to study data;
storage and availability of analytical dataset and statistical programmes for audit and inspection;
communication strategy for the scheduled progress and final reports;
publication strategy of interim and final results.
Non-interventional post-authorisation safety studies shall not be performed where the act of
conducting the study promotes the use of a medicinal product. This requirement applies to all
studies and to all activities performed in the study, including for studies conducted by the personnel
of the marketing authorisation holder and by third parties on behalf of the marketing authorisation
holder.
Payments to healthcare professionals for participating shall be restricted to compensation for time
and expenses incurred.
48
http://www.encepp.eu/code_of_conduct/index.html
9.2. Setting: study population defined in terms of persons, place, time period, and selection
criteria, including the rationale for any inclusion and exclusion criteria and their impact
on the number of subjects available for analysis. Where any sampling from a source
population is undertaken, description of the source population and details of sampling
methods should be provided. Where the study design is a systematic review or a
meta-analysis, the criteria for the selection and eligibility of studies should be explained.
9.3. Variables: outcomes, exposures and other variables including measured risk factors
should be addressed separately, including operational definitions; potential confounding
variables and effect modifiers should be specified.
9.4. Data sources: strategies and data sources for determining exposures, outcomes and all
other variables relevant to the study objectives, such as potential confounding variables
and effect modifiers. Where the study will use an existing data source, such as electronic
health records, any information on the validity of the recording and coding of the data
should be reported. If data collection methods or instruments are tested in a pilot study,
plans for the pilot study should be presented. If a pilot study has already been performed,
a summary of the results should be reported. Involvement of any expert committees to
validate diagnoses should be stated. In case of a systematic review or meta-analysis, the
search strategy and processes and any methods for confirming data from investigators
should be described.
9.5. Study size: any projected study size, precision sought for study estimates and any
calculation of the sample size that can minimally detect a pre-specified risk with a
pre-specified statistical precision.
9.6. Data management: data management and statistical programmes to be used in the study,
including procedures for data collection, retrieval and preparation.
9.7. Data analysis: the major steps that lead from raw data to a final result, including methods
used to correct inconsistencies or errors, impute values, modify raw data, categorise,
analyse and present results, and procedures to control sources of bias and their influence
on results; statistical procedures to be applied to the data to obtain point estimates and
confidence intervals of measures of occurrence or association, and sensitivity analyses.
9.8. Quality control: description of any mechanisms and procedures to ensure data quality
and integrity, including accuracy and legibility of collected data and original documents,
extent of source data verification and validation of endpoints, storage of records and
archiving of statistical programmes. As appropriate, certification and/or qualifications of
any supporting laboratory or research groups should be included.
9.9. Limitations of the research methods: any potential limitations of the study design, data
sources, and analytic methods, including issues relating to confounding, bias,
generalisability, and random error. The likely success of efforts taken to reduce errors
should be discussed.
10. Protection of human subjects: safeguards in order to comply with national requirements for
ensuring the well-being and rights of participants in non-interventional post-authorisation safety
studies.
11. Management and reporting of adverse events/adverse reactions: procedures for the
collection, management and reporting of individual cases of adverse reactions and of any new
information that might influence the evaluation of the benefit-risk balance of the product while
the study is being conducted. For studies where reporting is not required (see Module VI), this
should be stated.
12. Plans for disseminating and communicating study results, including any plans for
submission of progress reports and final reports.
13. References.
The format of the study protocol should follow the Guidance for the format and content of the
protocol of non-interventional post-authorisation safety studies (see GVP Annex II).
Feasibility studies that were carried out to support the development of the protocol, for example, the
testing of a questionnaire or simple counts of medical events or prescriptions in a database to
determine the statistical precision of the study, should be reported in the appropriate section of the
study protocol with a summary of their methods and results. The full report should be made
available to the national medicines authorities upon request. Feasibility studies that are part of the
research process should be described in the protocol, for example, a pilot evaluation of the study
questionnaire(s) used for the first set of patients recruited into the study.
An annex should list all separate documents and list or include any additional or complementary
information on specific aspects not previously addressed (e.g. questionnaires, case report forms),
with clear document references.
risk-benefit balance of the medicinal product may include that arising from an analysis of adverse
reactions and aggregated data.
This communication should not affect information on the results of studies which should be
provided by means of periodic safety update reports (PSURs) (see Module VII) and in RMP updates
(see Module V), where applicable.
methods for case ascertainment, as well as number of and reasons for dropouts.
9.4. Variables: all outcomes, exposures, predictors, potential confounders, and effect
modifiers, including operational definitions and diagnostic criteria, if applicable.
9.5. Data sources and measurement: for each variable of interest, sources of data and details
of methods of assessment and measurement. If the study has used an existing data source,
such as electronic health records, any information on the validity of the recording and
coding of the data should be reported. In case of a systematic review or meta-analysis,
description of all information sources, search strategy, methods for selecting studies,
methods of data extraction and any processes for obtaining or confirming data from
investigators.
9.6. Bias: any efforts to assess and address potential sources of bias.
9.7. Study size: study size, rationale for any sample size calculation and any method for
attaining projected study size.
9.8. Data transformation: transformations, calculations or operations on the data, including
how quantitative data were handled in the analyses and which groupings were chosen and
why.
9.9. Statistical methods: description of:
main summary measures
statistical methods applied to the study, including those used to control for
confounding and, for meta-analyses, methods for combining results of studies
any methods used to examine subgroups and interactions
how missing data were addressed
any sensitivity analyses
any amendment to the plan of data analysis included in the study protocol, with a
rationale for the change.
9.10. Quality control: mechanisms to ensure data quality and integrity.
10. Results: presentation of tables, graphs, and illustrations to present the pertinent data and reflect
the analyses performed. Both unadjusted and adjusted results should be presented. Precision of
estimates should be quantified using confidence intervals. This section should include the
following sub-sections:
10.1. Participants: numbers of study subjects at each stage of study, e.g. numbers potentially
eligible, examined for eligibility, confirmed eligible, included in the study, completing
follow-up, and analysed, and reasons for non-participation at any stage. In the case of a
systematic review or meta-analysis, number of studies screened, assessed for eligibility
and included in the review with reasons for exclusion at each stage.
10.2. Descriptive data: characteristics of study participants, information on exposures and
potential confounders and number of participants with missing data for each variable of
interest. In case of a systematic review or meta-analysis, characteristics of each study
format and are available for auditing and inspection. This provision should also be applied to PASS
voluntarily initiated, managed or financed by the marketing authorisation holder.
VIII.C.1. Scope
Provisions of VIII.C. refer specifically to post-authorisation safety studies initiated, managed or
financed by marketing authorisation holders pursuant to obligations imposed by a medicines
authority. Sections VIII.C.2. and VIII.C.3. apply to both interventional and non-interventional
PASS. Sections VIII.C.4. and VIII.C.5. apply to non-interventional PASS.
a. Request for a post-authorisation safety study as part of the initial marketing authorisation
application
A marketing authorisation may be granted by the national medicines authority subject to the
conduct of a PASS.
When the national medicines authority has assessed the final study results, it will produce an
assessment report, including a list of questions as appropriate. If the national medicines authority
addresses a list of questions to the marketing authorisation holder, the conclusion on the study
results including decision will be issued once the marketing authorisation holder has addressed the
questions posed within the timeline specified.
The national medicines authority will inform the marketing authorisation holder in writing and
within the appropriate timelines of its decisions with respect to the assessment of the following:
Study protocol;
Study protocol amendments;
Final study report;
Waiver request for the submission of the final study protocol.
When the marketing authorisation holder submit a request to the national medicines authority for a
pre-submission meeting the later will set up of this meeting as appropriate.
VIII.App1.1.1.3. Registries
A registry is an organised system that uses observational methods to collect uniform data on
specified outcomes in a population defined by a particular disease, condition or exposure. A registry
can be used as a data source within which studies can be performed. Entry in a registry is generally
defined either by diagnosis of a disease (disease registry) or prescription of a drug (exposure
registry).
Disease/outcome registries, such as registries for blood dyscrasias, severe cutaneous reactions, or
congenital malformations may help collect data on drug exposure and other factors associated with
a clinical condition. A disease registry might also be used as a base for a case-control study
comparing the drug exposure of cases identified from the registry and controls selected from either
patients within the registry with another condition or from outside the registry, or for a case-only
design (see VIII.App 1.1.2.4.).
Exposure registries address populations exposed to medicinal products of interest (e.g. registry of
rheumatoid arthritis patients exposed to biological therapies) to determine if a medicinal product
has a special impact on this group of patients. Some exposure registries address exposures to
medicinal products in specific populations, such as pregnant women. Patients may be followed over
time and included in a cohort study to collect data on adverse events using standardised
questionnaires. Simple cohort studies may measure incidence, but, without a comparison group,
cannot evaluate any association between exposures and outcomes. Nonetheless, they may be useful
for signal amplification particularly for rare outcomes. This type of registry may be very valuable
when examining the safety of an orphan drug indicated for a specific condition.
fraction of cases) in the catchment area are captured and the fraction of controls from the source
population is known, an incidence rate can be calculated.
When the source population for the case-control study is a well-defined cohort, it is then possible to
select a random sample from it to form the control series. The name ―nested case-control study‖ has
been coined to designate those studies in which the control sampling is density-based (e.g. the
control series represents the person-time distribution of exposure in the source population). The
case-cohort is also a variant in which the control sampling is performed on those persons who make
up the source population regardless of the duration of time they may have contributed to it.
A case-control approach could also be set up as a permanent scheme to identify and quantify risks
(case-control surveillance). This strategy has been followed for rare diseases with a relevant
aetiology fraction attributed to medicinal products, including blood dyscrasias or serious skin
disorders.
Marketing authorisation holders should select the best data source according to validity (e.g.
completeness of relevant information, possibility of outcome validation) and efficiency criteria (e.g.
time span to provide results). External validity should also be taken into account. As far as feasible
the data source chosen to perform the study should include the population in which the safety
concern has been raised. In case another population is involved, the marketing authorisation holder
should evaluate the differences that may exist in the relevant variables (e.g. age, sex, pattern of use
of the medicinal product) and the potential impact on the results. In the statistical analysis, the
potential effect of modification of such variables should be explored.
With any data source used, the privacy and confidentiality regulations that apply to personal data
should be followed.
GVP: Modules
IX.A. Introduction
The Report of the Council for International Organisations of Medical Sciences Working group VIII
Practical Aspects of Signal Detection in Pharmacovigilance (CIOMS, Geneva 2010) defines a
signal as information that arises from one or multiple sources (including observations and
experiments), which suggests a new potentially causal association, or a new aspect of a known
association, between an intervention and an event or set of related events, either adverse or
beneficial, that is judged to be of sufficient likelihood to justify verificatory action.
For the purpose of this Module, only new information related to adverse effects will be considered.
In order to suggest a new potentially causal association or a new aspect of a known association, any
signal should be validated taking into account other relevant sources of information.
The signal management process can be defined as the set of activities performed to determine
whether, based on an examination of individual case safety reports (ICSRs), aggregated data from
active surveillance systems or studies, literature information or other data sources, there are new
risks associated with an active substance or a medicinal product or whether known risks have
changed. The signal management process shall include all steps from initial signal detection;
through their validation and confirmation; analysis and prioritisation; and signal assessment to
recommending action, as well as the tracking of the steps taken and of any recommendations made.
The signal management process concerns all stakeholders involved in the safety monitoring of
medicinal products including patients, healthcare professionals, marketing authorisation holders
(MAHs), regulatory authorities, scientific committees.
Whereas the ADRs database will be a major source of pharmacovigilance information, the signal
management process covers signals arising from any source, only signals related to an adverse
reaction shall be considered.
In this Module, all applicable legal requirements are referenced as explained in the GVP
Introductory Cover Note and are usually identifiable by the modal verb "shall". Guidance for the
implementation of legal requirements is provided using the modal verb "should".
The objectives of this Module are:
to provide general guidance and requirements on structures and processes involved in signal
management (section IX.B.);
to describe how these structures and processes are applied in the setting of the Arab Countries
pharmacovigilance and regulatory bodies(section IX.C.).
The detection of signals shall be based on a multidisciplinary approach. Signal detection within the
―National Pharmacovigilance and Safety reports database‖ or MAH-specific ADRs database shall
be complemented by statistical analysis where appropriate.
In order to determine the evidentiary value (i.e. the supporting evidence) of a signal a recognised
methodology shall be applied taking into account the clinical relevance, quantitative strength of the
association, the consistency of the data, the exposure-response relationship, the biological
plausibility, experimental findings, possible analogies and the nature and quality of the data.
Different factors may be taken into account for the prioritisation of signals, namely whether the
association or the active substance/medicinal product is new, the strength of the association, the
seriousness of the reaction involved and the documentation of the reports in the ADRs database.
IX.B.3.1. Introduction
The signal management process covers all steps from detecting signals to recommending action(s)
as follows:
signal detection;
signal validation;
signal analysis and prioritisation;
signal assessment;
recommendation for action;
exchange of information.
Although these steps generally follow a logical sequence, the wide range of sources of information
available for signal detection may require some flexibility in the conduct of signal management e.g.:
when signal detection is primarily based on a review of individual case safety reports (ICSRs),
this activity may include validation and preliminary prioritisation of any detected signal;
when a signal is detected from results of a study, it is generally not possible or practical to assess
each individual case, and validation may require collection of additional data;
recommendation for action (followed by decision in accordance with the applicable legislation)
and exchange of information are components to be considered at every step of the process.
For the purpose of this guidance, signals originating from the monitoring of data from spontaneous
reporting systems are considered as the starting point of the signal management process. The same
principles should apply for data originating from other sources.
2010Whichever methods are employed for the detection of signals, the same principles should
apply, namely:
the method used should be appropriate for the data set; for example, the use of complex statistical
tools may not be appropriate for smaller data sets;
data from all appropriate sources should be considered;
systems should be in place to ensure the quality of the signal detection activity;
any outputs from a review of cumulative data should be assessed by an appropriately qualified
person in a timely manner;
the process should be adequately documented, including the rationale for the method and
periodicity of the signal detection activity.
Detection of signals may be performed based on a review of ICSRs, from statistical analyses in
large databases, or from a combination of both.
The periodicity at which statistical reports should be generated and reviewed may vary according to
the active substance/medicinal product, its indication and any known potential or identified risks.
Some active substances/medicinal products may also be subject to an increased frequency of data
monitoring (see IX.C.2.). The duration for this increased frequency of monitoring may also vary and
be flexible with the accumulation of knowledge of the risk profile associated with the use of the
concerned active substance/medicinal product.
IX.B.3.2.3. Combination of statistical methods and review of individual case safety reports
Statistical reports may be designed to provide tools for identifying suspected adverse reactions that
meet pre-defined criteria of frequency, severity, clinical importance, novelty or statistical
association. Such filtering tools may facilitate the selection of ICSRs to be reviewed as a first step.
The thresholds used in this filtering process (for example, at least 3 cases reported) may vary
according to the extent of usage of medicinal products and thus the potential public health impact.
Irrespective of the statistical method used, where statistical reports are used to automate the
screening of a database, signal detection should always involve clinical judgement and the
corresponding ICSRs should be individually reviewed, considering their clinical relevance
(IX.B.3.2.1.)
The statistical method should therefore be a supporting tool in the whole process of signal detection
and subsequent validation.
- strength of evidence for a causal effect (e.g. number of reports, exposure, temporal
association, plausible mechanism, de/re-challenge, alternative explanation/confounders);
- seriousness and severity of the reaction and its outcome;
- novelty of the reaction (e.g. new and serious adverse reactions);
- drug-drug interactions;
- reactions occurring in special populations.
Previous awareness:
- the extent to which information is already included in the summary of product
characteristics (SmPC) or patient leaflet;
- whether the association has already been assessed in a PSUR or RMP, or was discussed at
the level of a scientific committee or has been subject to a regulatory procedure.
In principle only a new signal for which there is no previous awareness should be validated.
However, an already known association may give rise to a new signal if its apparent frequency of
reporting, its duration, its severity or a change in the previously reported outcome (such as new
fatality) suggests new information as compared with the information included in the SmPC or
previously assessed by the competent authority.
Availability of other relevant sources of information providing a richer set of data on the same
association:
the signal in different settings (e.g. general practice and hospital settings), data sources or
countries;
clinical context (e.g. whether the association suggest a clinical syndrome that may include other
reactions);
the public health impact, including the extent of utilisation of the product in the general
population and in special populations (e.g. pregnant women, children or the elderly) and the
patterns of medicinal product utilisation (e.g. off-label use or misuse). The public health impact
may include an estimation of the number of patients that may be affected by an adverse reaction
and this number could be considered in relation to the size of the general population, the
population with the target disease and the treated population;
increased frequency or severity of a known adverse reaction;
novelty of the suspected adverse reaction, e.g. when an unknown suspected adverse reaction
occurs shortly after the marketing of a new medicinal product;
if a marketing authorisation application for a new active substance is still under evaluation.
In some circumstances, priority can also be given to signals identified for medicinal products or
events with potential high media and pharmacovigilance stakeholder interest in order to
communicate the result to the public and healthcare professionals as early as possible.
The outcome of signal prioritisation should include a recommendation of the time frame for the
management of the signal.
The outcome of the signal prioritisation process should be entered in the tracking system, with the
justification for the priority attributed.
49
MedDRA® the Medical Dictionary for Regulatory Activities terminology is the international medical terminology
developed under the auspices of the International Conference on Harmonization of Technical Requirements for
Registration of Pharmaceuticals for Human Use (ICH)
adverse reactions, such as to other terms linked to a complex disease (e.g. optic neuritis as a possible
early sign of multiple sclerosis), to a prior stage of a reaction (e.g. QT prolongation and torsades de
pointes) or to clinical complications of the adverse reaction of interest (e.g. dehydration and acute
renal failure).
Gathering information from various sources may take time. For a new signal of a serious or severe
adverse reaction, measures should be taken at any stage in the management of a signal including
detection, if the information already available supports the conclusion that there is a potential risk
that needs to be prevented or minimised in a timely manner.
The outcomes of signal assessment involving new or changed risks and risks that have an impact on
the benefit-risk balance of the concerned active substance/medicinal products should be
communicated to the public including health care professionals and patients as well as to the
concerned marketing authorisation holders.
IX.B.4.1. Tracking
All validation, prioritisation, assessment, timelines, decisions, actions, plans, reporting as well as all
other key steps should be recorded and tracked systematically. Tracking systems should be used for
documentation and should also include signals, for which the validation process conducted was not
suggestive of a new potentially causal association, or a new aspect of a known association. All
records need to be archived (see Module I).
may maintain, review and publish a list of medical events that have to be taken into account for
the detection of a signal ;
ensure appropriate support for the monitoring of the data in ―National Pharmacovigilance and
Safety reports database‖ by marketing authorisation holders (applicable in only some Arab
Countries);
administer a Pharmacovigilance Issues Tracking Tool (PITT) for validated signals that require
further assessment ;
perform a regular review of the signal management methodology to be used and publish
recommendations as appropriate ;
statistical outputs should contain ADRs in a structured hierarchy (e.g. MedDRA hierarchy) by
active substance(s)/medicinal product(s) and allow filters and thresholds to be applied on several
fields as appropriate.
The baseline frequency for reviewing the statistical outputs from ―National Pharmacovigilance and
Safety reports database‖ should be once- monthly. An increase to the baseline frequency of this data
monitoring may be decided by the the national medicines authority if justified by the identified or
potential risks of the product or by the need for additional information.
For products subject to additional monitoring (see Module X), the frequency for reviewing the
statistical outputs should be every 2 weeks until the end of additional monitoring. A 2-week
frequency for reviewing the statistical outputs may also be applied for any other product taking into
account the following criteria:
any product considered to have an identified or potential risk that could impact significantly on
the benefit-risk balance or have implications for public health. This may include risks associated
with significant misuse, abuse or off-label use. The product may be moved back to baseline
frequency of monitoring if risks are not confirmed;
any product for which the safety information is limited due to low patient exposure during drug
development, including products authorised under conditional approval or under exceptional
circumstances 50 , or for which there are vulnerable or poorly studied patient populations or
important missing information (e.g. children, pregnant women, renal-impaired patients) while
post-marketing exposure is likely to be significant;
any product that contains active substances already authorised in the Arab Country concerned but
is indicated for use in a new patient population or with a new route of administration;
any product for which the existing marketing authorisation has been significantly varied (e.g.
changes to indication, posology, pharmaceutical form or route of administration), thereby
modifying the exposed patient population or the safety profile.
Confirmation of a signal arising from the ―National Pharmacovigilance and Safety reports
database‖ data monitoring activities does not necessarily imply that the product has to be more
frequently monitored and a risk proportionate approach should be applied.
More frequent monitoring than every 2 weeks may be proposed. It should be targeted to a safety
concern of interest especially during public health emergencies (e.g. pandemics) and may be
applied in the context of customised queries.
50
Exceptional circumstances is a type of marketing authorisation granted to medicines where the applicant is unable to
provide a comprehensive data on the efficacy and safety under normal conditions of use, because the condition to be
treated is rare or because collection of full information is not possible or is unethical. (may be NOT applicable in some
Arab Countries, check the national regulations)
GVP: Modules
X.A. Introduction
Pharmacovigilance is a vital public health function with the aim of rapidly detecting and responding
to potential safety hazards associated with the use of medicinal products.
A medicinal product is authorised on the basis that, its benefit-risk balance is considered to be
positive at that time for a specified target population within its approved indication (s). However,
not all risks can be identified at the time of initial authorisation and some of the risks associated with
the use of a medicinal product emerge or are further characterised in the post-authorisation phase of
the product‘s lifecycle. To strengthen the safety monitoring of medicinal products, this guideline
has introduced a framework for enhanced risk proportionate post-authorisation data collection for
medicinal products, including the concept of additional monitoring for certain medicinal products.
National Medicines Authorities, shall set up, maintain and make public a list of medicinal products
that are subject to additional monitoring (hereafter referred to as ―the list‖).
These medicinal products will be readily identifiable by an inverted equilateral black triangle.
That triangle will be followed by an explanatory statement in the summary of product
characteristics (SmPC) as follows:
“This medicinal product is subject to additional monitoring. This will allow quick
identification of new safety information. Healthcare professionals are asked to report any
suspected adverse reactions. See section ....... for how to report adverse reactions.”
A similar statement will also be included in the package leaflet. This explanatory statement should
encourage healthcare professionals and patients to report all suspected adverse reactions.
Post-authorisation spontaneous Adverse Drug Reactions (ADR) reports remain a cornerstone of
pharmacovigilance. Data from ADR reports is a key source of information for signal detection
activities (see Module IX). Increasing the awareness of healthcare professionals and patients of the
need to report suspected adverse drug reactions and encouraging their reporting is therefore an
important means of monitoring the safety profile of a medicinal product.
The concept of additional monitoring originates primarily from the need to enhance the ADR
reporting rates for newly authorised products for which the safety profile might not be fully
characterised or for products with newly emerging safety concerns that also need to be better
characterised. The main goals are to collect additional information as early as possible to further
elucidate the risk profile of products when used in clinical practice and thereby informing the safe
and effective use of medicinal products.
medicinal products that contain a new active substance which, on 1 July 2015, was not contained
in any innovative medicinal product;
any biological medicinal product not covered by the previous category and authorised after 1
July 2015;
products for which a PASS was requested at the time of marketing authorisation
products authorised with specific obligations on the recording or suspected adverse drug
reactions
products for which a PASS was requested following the grant of marketing authorisation
products which were granted a conditional marketing authorisation
products authorised under exceptional circumstances51.
51
Exceptional circumstances is a type of marketing authorisation granted to medicines where the applicant is
unable to provide comprehensive data on the efficacy and safety under normal conditions of use, because the
condition to be treated is rare or because collection of full information is not possible or is unethical. (may be NOT
applicable in some Arab Countries, check the national regulations)
or made subject to conditions before such date, provided they fall within one or more of the above
situations for the optional scope.
Pharmacovigilance rules in general and additional monitoring specifically take into account that the
full safety profile of medicinal products can only be confirmed after products have been placed on
the market. Due consideration should, therefore, be given to the merit of inclusion of a medicinal
product in the list in terms of increasing awareness about the safe and effective use of a medicinal
product and/or providing any additional information for the evaluation of the product. In this regard,
the decision to include a medicinal product subject to conditions in the list should take account of
the nature and scope of the conditions or obligations placed on the marketing authorisation
including their potential public health impact. The decision should also consider the usefulness of
the additional monitoring status in relation to other additional pharmacovigilance activities
proposed in the risk management plan, for example in relation to the objectives of PASS.
X.C.2. Criteria for defining the initial time period of maintenance in the
additional monitoring list
For medicinal products containing new active substances as well as for all biological medicinal
products approved after 1 July 2015, the initial period of time for inclusion is five years after the
marketing authorisation date in the concerned Arab Country.
The national medicines authority: decide which authorised medicinal product should be subject
to additional monitoring (see X.C.1) and therefore included in the list;
is responsible for publishing the list of medicinal products authorised in its territory that are
subject to additional monitoring on its official website where the product information is
publicly available (if applicable);
is responsible for removing medicinal products from the list after a pre-determined time period;
will take into account the list of authorised medicinal products subject to additional monitoring in
determining the frequency and processes of its signal detection activities;
will inform the relevant MAH when an authorised medicinal product has been included to the list
of additional monitored products;
“This medicinal product is subject to additional monitoring. This will allow quick
identification of new safety information. Healthcare professionals are asked to report any
suspected adverse reactions. See section ....... for how to report adverse reactions.”,
preceded by an inverted equilateral black triangle. A similar statement will also be included in the
package leaflet. Once the medicinal product is included or removed from the list, the marketing
authorisation holder shall update the SmPC and the package leaflet to include or remove, as
appropriate, the black symbol, the statement, and the standardised explanatory statement.
If the decision to include or remove a medicinal product from the list is done during the assessment
of a regulatory procedure (e.g. marketing authorisation application, extension of indication,
renewal) the SmPC and the package leaflet should be updated before finalisation of the procedure in
order to include or remove the black triangle symbol and explanatory statement from the product
information.
If the decision to include or remove a medicinal product from the list is done outside a regulatory
procedure, then the marketing authorisation holder is requested to subsequently submit a variation
to update the product information of that product accordingly.
X.C.6. Transparency
Each Arabian Medicines Authority should make publicly available the list of the names and active
substances of all medicinal products approved in their countries subject to additional monitoring
and the general criteria to include medicinal products in the list.
The list will include an electronic link(s) to the relevant webpage where the product information is
publicly available (if applicable).
GVP: Modules
GVP: Modules
GVP: Modules
GVP: Modules
GVP: Modules
XV.A. Introduction
This Module provides guidance to marketing authorisation holders (MAHs), national medicines
authorities on how to communicate and coordinate safety information in the Arab countries.
Communicating safety information to patients and healthcare professionals is a public health
responsibility and is essential for achieving the objectives of pharmacovigilance in terms of
promoting the rational, safe and effective use of medicines, preventing harm from adverse reactions
and contributing to the protection of patients‘ and public health.
Safety communication is a broad term covering different types of information on medicines,
including statutory information as contained in the product information (i.e. the summary of product
characteristics (SmPC), package leaflet (PL) and the labelling of the packaging). Although some
principles in this Module (i.e. Section XV.B.1 and B.2.) apply to all types of safety communication,
the module itself focuses on the communication of ‗new or emerging safety information‘, which
means new information about a previously known or unknown risk of a medicine which has or may
have an impact on a medicine‘s benefit-risk balance and its condition of use. Unless otherwise
stated, the term ‗safety communication‘ in this module should be read as referring to emerging
safety information.
Communication of important new safety information on medicinal products should take into
account the views and expectations of concerned parties, including patients and healthcare
professionals, with due consideration given to relevant legislation. This Module addresses some
aspects of the interaction with concerned parties and supplements the specific guidance will be
given in Module XI on public participation as well as the guidance on communication planning will
be given in Module XII.
Communication is distinct from transparency, which aims to provide public access to information
related to data assessment, decision-making and safety monitoring performed by competent
authorities.
Section XV.B. of this Module describes principles and means of safety communication. Section
XV.C. provides guidance on the coordination and dissemination of safety communications in the
Arab Countries. Both sections give particular consideration to direct healthcare professional
communications (DHPCs), and provide specific guidance for preparing them. This is because of the
central importance of DHPCs in targeting healthcare professionals and because of the level of
coordination required between marketing authorisation holders and national medicines authorities
in their preparation.
Where relevant safety communication should be complemented at a later stage with follow-up
communication e.g. on the resolution of a safety concern or updated recommendations.
The effectiveness of safety communication should be evaluated where appropriate and possible
(see XV.B.7.).
Safety communications should comply with relevant requirements relating to individual data
protection and confidentiality.
where relevant, a reminder of the need to report suspected adverse reactions in accordance with
national spontaneous reporting systems.
The information in the safety communication shall not be misleading and shall be presented
objectively. Safety information should not include any material or statement which might constitute
advertising.
A DHPC should be disseminated in the following situations when there is a need to take immediate
action or change current practice in relation to a medicinal product:
52
For the purpose of this section tools and channels are presented without distinction as they often overlap and
there is no general agreement on their categorisation
XV.B.5.4. Website
A website is a key tool for members of the public (including patients and healthcare professionals)
actively searching the internet for specific information on medicinal products. National medicines
authorities as well as marketing authorisation holders should ensure that important safety
information published on websites under their control is easily accessible and understandable by the
public. Information on websites should be kept up-to-date, with any information that is out-of-date
marked as such or removed. If possible, the official website of the national medicines authority
should contain information on all medicines authorized in its Arab Country.
the dissemination list also known as ―intended recipient list‖: the intended recipients
HCPs groups may be general practitioners, specialists, pharmacists, nurses;
hospitals/ambulatory care/other institutions as appropriate. The list should specify the
intended recipients name, specialty and geographical distribution;
When defining the target groups of recipients, it should be recognized that it is not
only important to communicate with those HCPs who will be able or likely to
prescribe or administer the medicinal product, but also to those who may diagnose
adverse reactions, e.g. emergency units, poison centres, or to appropriate specialists,
e.g. cardiologists. It is also important to consider provision of DHPCs to relevant
pharmacists (hospital and /or community) who serve as information providers within
healthcare systems and provide assistance and information to Patients, HCPs,
including hospital wards and poison centres, as well as the general public.
timetable for disseminating the DHPC: the proposed timetable should be appropriate
according to the urgency of the safety concern (usually maximum of 15 calendar days
is considered appropriate);
dissemination mechanism: how the DHPC is planned to be disseminated, the
proposed mechanism should be selected appropriately to meet the dissemination
timetable.
The last 3 items above are known as the communication plan.
The marketing authorisation holder should submit these documents in the form of one full original
hard copy and one soft copy, after approval by the national medicines authority; the MAH will
receive back the hard copy stamped with ‖approved‖, while the soft copy will be retained at the
authority. These submission requirements may differ in some Arab Countries; consult with the
national medicines authority.
The marketing authorisation holder should allow a minimum of two working days for comments.
However, whenever possible more time should be allowed. The timing may be adapted according to
the urgency of the situation.
The national medicines authority will review the DHPCs (may request advice from its scientific
committees/ pharmacovigilance committee as appropriate.
Once the content of a DHPC and communication plan from the MAH are agreed by national
medicines authorities, the MAH can start dissemination of the agreed DHPC (i.e. the MAH shall
NOT start disseminating the DHPC prior to obtaining the approval from the national medicines
authority).
The MAH should adhere to the Communication Plan agreed with the national medicines authority.
Any significant event or problem occurring during the DHPC dissemination which reveals a need to
change the Communication Plan or a need for further communication to Healthcare Professionals,
this should be notified in a timely manner to national medicines authority to be approved.
After dissemination of a DHPC, a closing review should be performed by the MAH, a progress
report may be submitted upon request of the national medicines authority.
In cases where a medicines authority in other country (Arab or non-Arab) requests the
dissemination of a DHPC in its territory, the marketing authorisation holder should notify the
relevant national medicines authorities of the Arab Country(s) in which this product is also
authorized. This is in the context of the national legal requirement under which the marketing
authorisation holder shall notify the national medicines authorities of any new information which
may impact the benefit-risk balance of a medicinal product. The need for any subsequent
communication, e.g. a DHPC, in the Arab Country(s) concerned should be considered and agreed
on a case-by- case basis.
A flow chart describing the processing of DHPCs is provided in Figure XV.I at the end of the
Module.
Figure VX.1: Flow chart for the processing of Direct Healthcare Professional Communications
(DHPCs) in the Arab Country concerned
NMA: national medicines authority
MAH: Marketing authorization holder
Dissemination of DHPC in
OTHER country is requested (e.g.
in KSA or UK…etc)
No
DHPC and communication
plan agreed by NMA NMA reply the MAH
GVP: Modules
XVI.A. Introduction
Risk minimisation measures are interventions intended to prevent or reduce the occurrence of
adverse reactions associated with the exposure to a medicine, or to reduce their severity or impact
on the patient should adverse reactions occur. Planning and implementing risk minimisation
measures and assessing their effectiveness are key elements of risk management.
The guidance provided in this Module should be considered in the context of the wider GVP
guidance, in particular in conjunction with Module V.
Risk minimisation measures may consist of routine risk minimisation or additional risk
minimisation measures. Routine risk minimisation is applicable to all medicinal products, and
involves the use of the following tools, which are described in detail in Module V:
the summary of product characteristics (SmPC);
the package leaflet;
the labelling;
the pack size and design;
the legal (prescription) status of the product.
Safety concerns of a medicinal product are normally adequately addressed by routine risk
minimisation measures (see Module V). In exceptional cases however, routine risk minimisation
measures will not be sufficient for some risks and additional risk minimisation measures will be
necessary to manage the risk and/or improve the risk-benefit balance of a medicinal product. This
module provides particular guidance on the use of additional risk minimisation measures, including
the selection of tools and the evaluation of their effectiveness. In specific circumstances, however,
the effectiveness evaluation may also apply to routine risk minimisation measures associated with
safety concern(s) which are described in the SmPC/PIL (e.g. the SmPC provides guidance for
clinical actions beyond routine standards of clinical care for either the risk itself or management of
the target population).
On the basis of the safety concerns described in the safety specification (see GVP Module V), the
appropriate risk minimisation measures should be determined. Each safety concern needs to be
individually considered and the selection of the most suitable risk minimisation measure should
take into account the seriousness of the potential adverse reaction(s) and its severity (impact on
patient), its preventability or the clinical actions required to mitigate the risk, the indication, the
route of administration, the target population and the healthcare setting for the use of the product. A
safety concern may be addressed using more than one risk minimisation measure, and a risk
minimisation measure may address more than one safety concern.
The marketing authorisation holder shall include all risk minimisation measures in the risk
management plan and monitor their outcome .
This Module provides guidance on the principles for:
The development and implementation of additional risk minimisation measures, including
introduce undue burden on the healthcare delivery system, the marketing authorisation holders, the
regulators, and, most importantly, on the patients. To this aim, they should have a clearly defined
objective relevant to the minimisation of specific risks and/or optimisation of the risk-benefit
balance. Clear objectives and defined measures of success with milestones need to guide the
development of additional risk minimisation measures and close monitoring of both their
implementation and ultimate effectiveness is necessary. The nature of the safety concern in the
context of the risk-benefit balance of the product, the therapeutic need for the product, the target
population and the required clinical actions for risk minimisation are factors to be considered when
selecting risk minimisation tools and an implementation strategy to accomplish the desired public
health outcome. The evaluation of effectiveness should facilitate early corrective actions if needed
and may require modification over time. It is recognised that this is an evolving area of medical
sciences with no universally agreed standards and approaches. Therefore, it is important to take
advantage of any relevant elements of methodology from pharmacoepidemiology and other
disciplines, such as social/behavioural sciences and qualitative research methods.
The introduction of additional risk minimisation should be considered as a ―programme‖ where
specific tools, together with an implementation scheme and evaluation strategy are developed. The
description of risk minimisation measures, an integral part of the RMP (see Module V), should
therefore give appropriate consideration to the following points:
Rationale: When additional risk minimisation measure(s) are introduced a rationale should be
provided for those additional measures;
Objectives: Each proposed additional risk minimisation measure(s) should include defined
objective(s) and a clear description of how and which safety concern is addressed with the
proposed additional risk minimisation measure(s);
Description: This section of the RMP should describe the selected additional risk minimisation
measures, including tools that will be used and key elements of content;
Implementation: This section of the RMP should provide a detailed proposal for the
implementation of additional risk minimisation measures (e.g. setting and timing or frequency of
intervention, details of the target audience, plan for the distribution of educational tools; how the
action will be coordinated where more than one marketing authorisation holder is involved);
Evaluation: This section of the RMP should provide a detailed plan with milestones for
evaluating the effectiveness of additional risk minimisation measures in process terms and in
terms of overall health outcome measures (e.g. reduction of risk).
complexity. These measures might be used to guide appropriate patient selection with the exclusion
of patients where use is contraindicated, to support on-treatment monitoring relevant to important
risks and/or management of an adverse reaction once detected. Additionally, specific measures may
be developed to minimise the risk of medication error and/or to ensure appropriate administration of
the product where it is not feasible to achieve this through the product information and labelling
alone.
Section XVI.B.2. describes additional risk minimisation measures that may be considered in
addition to the routine measures, including:
Educational programmes;
Controlled access programmes;
Other risk minimisation measures.
example a diary for posology or diagnostic procedures that need to be carried out and recorded by
the patient and eventually discussed with healthcare professionals, to ensure that any steps required
for the effective use of the product are adhered to.
Patient alert card
The aim of this tool should be to ensure that special information regarding the patient‘s current
therapy and its important risks (e.g. potential life-threatening interactions with other therapies) is
held by the patient at all times and reaches the relevant healthcare professional as appropriate. The
information should be kept to the minimum necessary to convey the key minimisation message(s)
and the required mitigating action, in any circumstances, including emergency. Ability to carry with
ease (e.g. can be fitted in a wallet) should be a key feature of this tool.
Periodic review of the effectiveness of one or more specific tools or the overall programme, as
appropriate should be also planned. Time points of particular relevance are as follows:
after initial implementation of a risk minimisation programme (e.g. within 12-18 months), in
order to allow the possibility of amendments, should they be necessary;
in time for the evaluation of the renewal of a marketing authorisation; and
whenever effectiveness is evaluated, careful consideration should be given on the need for
continuing with the additional risk minimisation measure.
Effectiveness evaluation should address different aspects of the risk minimisation, the process itself
(i.e. to what extent the programme has been implemented as planned), its impact on knowledge and
behavioral changes in the target audience (i.e. the measure(s) in affecting behavioural change), and
the outcome (i.e. to what extent the predefined objectives of risk minimisation were met, in the short
and long term). In designing an evaluation strategy, due consideration needs to be made toward
what aspects of process and outcomes can be realistically measured in order to avoid the generation
of inaccurate or misleading data or placing an undue burden on the healthcare system or other
stakeholders. The time of assessing each aspect of the intervention as well as setting of realistic
metrics on which the effectiveness of the tool is judged, should also be carefully considered and
planned prior to initiation.
To evaluate the effectiveness of additional risk minimisation measures two categories of indicators
should be considered:
Process indicators;
Outcome indicators.
Process indicators are necessary to gather evidence that the implementing steps of additional risk
minimisation measures have been successful. These process indicators should provide insight into
what extent the programme has been executed as planned and whether the intended impacts on
behaviour have been observed. Implementation metrics should be identified in advance and tracked
over time. The knowledge gained may be used to support corrective implementation action as
needed. Assessing the implementation process can also improve understanding of the process(es)
and causal mechanism(s) whereby the additional risk minimisation measure(s) did or did not lead,
to the desired control of specified important risks.
Outcome indicators provide an overall measure of the level of risk control that has been achieved
with any risk minimisation measure in place. For example, where the objective of an intervention is
to reduce the frequency and/or severity of an adverse reaction, the ultimate measure of success will
be linked to this objective.
In rare circumstances when it is fully justified that the assessment of outcomes indicators is
unfeasible (e.g. inadequate number of exposed patients, very rare adverse events), the effectiveness
evaluation may be based exclusively on the carefull interpretation of data on process indicators.
The conclusion of the evaluation may be that risk minimisation should remain unchanged or
modifications are to be made to existing activities. Alternatively, the assessment could indicate that
risk minimisation is insufficient and should be strengthened (e.g. through amendment of warnings
or recommendations in the SmPC or package leaflet, improving the clarity of the risk minimisation
advice and/or by adding additional tools or improving existing tools). Another decision may be that
the risk minimisation is disproportionate or lacking a clear focus and could be reduced or simplified
(e.g. by decreasing the number of tools or frequency of intervention, or by eliminating interventions
proved to be non-contributory to risk minimisation). In all circumstances, the burden on the patient
and the healthcare system should be given careful consideration.
In addition to assessing the effectiveness of risk minimisation measures in managing safety
concerns, it is also important to monitor if the risk minimisation intervention may have had
unintended (negative) consequences relevant to the public health question under consideration,
either in the short and/or long term. Examples of unintended consequences may include undue
burden on the healthcare system, or discontinuation of a product even if its risk-benefit balance
remains positive.
This guidance defines ―Any study ….measuring the effectiveness of risk management measures‖ as
a post-authorisation safety study. Therefore, if a study is conducted to assess behavioural or safety
outcome indicators the detailed guidance for conducting a post-authorisation safety study, which is
provided in Module VIII, should be followed. Such guidance does not apply to the measurement of
simple process markers (e.g. distribution of the tools reaching the target population). The ENCePP
Guide on Methodological Standards in Pharmacoepidemiology53 (which is acknowledged in the
Arab Countries) should be considered as appropriate.
53
http://www.encepp.eu
A survey generally includes a core of standard questions administered through telephone contact, in
person interview, or self-administered through postal/electronic communication, which are repeated
over time. Such an approach may be tailored to the monitoring of attitude and knowledge in a
diverse sample, that includes representatives from each segment of interest in the target populations
of healthcare professionals and/or patients. Psychometric measures should be used as appropriate.
Whenever feasible a randomised sample and an adequate sample size should be selected. In
contrast, use of advocacy groups or patient support groups to survey knowledge can be considered
to be inherently biased through self-selection, and should be avoided.
Appropriate attention should be given to the research objectives, study design, sample size and
representativeness, operational definition of dependent and independent variables, and statistical
analysis. Thorough consideration should also be given to the choice of the most appropriate data
collection instruments (e.g. questionnaires).
adverse reaction, obtained for example in the context of post-authorisation safety studies. The use of
appropriate safety-related outcomes of interest should be considered (e.g. a surrogate endpoint such
as an adequate biomarker as a substitute for a clinical endpoint) if such an approach facilitates the
effectiveness evaluation. Under any approach, scientific rigour and recognised principles of
epidemiologic research should always guide the assessment of the final outcome indicator of
interest. Comparisons of frequency before and after the implementation of the risk minimisation
measures (i.e. pre-post design) should be considered. When a pre-post design is unfeasible (e.g. risk
minimisation measures are put in place at the time of initial marketing authorisation), the
comparison of an outcome frequency indicator obtained post-intervention against a predefined
reference value obtained from literature review, historical data, expected frequency in general
population, would be acceptable (i.e. observed versus expected analysis) and should take into
account any stimulated reporting, changes in patient care and/or risk minimisation measures over
time. The selection of any particular reference group should be appropriately justified.
Methods to measure the effectiveness of risk minimisation measure should be proportionate to the
risks being minimised. As such use of spontaneous reporting rates (i.e. number of suspected adverse
reaction reports over a fixed time period) may be acceptable in the context of routine risk
minimisation. Spontaneous reporting should be considered with caution when estimating the
frequency of adverse events in the treated population, but it may be used in very specific
circumstances, for instance when the adverse reaction with the product is rare and there is a
negligible background incidence of the adverse event in the general population and a strong
association between treatment and the adverse event. In those circumstances when a direct measure
on the risk in the treated population is not feasible, spontaneous reporting could offer an
approximation of the frequency of the adverse reaction in the treated population, provided that
reasonably valid data can be obtained to evaluate the reporting rate in the context of product use.
However, the well know biases that affects reporting of suspected adverse reactions may provide
misleading results. For instance, the introduction of a risk minimisation measure in response to a
safety concern detected in the post-authorisation phase of a medicinal product may raise awareness
regarding selected adverse reactions which ultimately may result in an increased reporting rate. In
these circumstances an analysis of spontaneous reporting may lead to the erroneous conclusion that
the intervention was ineffective. Decreasing reporting rates over time may also lead to the erroneous
conclusion that the intervention was effective.
XVI.B.5. Coordination
If several products, referred to as ―generics‖ of the same active substance are available in a market
there should be a consistent approach in the use of additional risk minimisation measures
coordinated and overseen by the national medicines authorities. When a coordinated action for a
class of products is needed a harmonised approach should be agreed if appropriate. Under these
circumstances advanced planning should ensure that the effectiveness of risk minimisation
measures (see XVI.B.4.) can be considered for each individual product as well as for the products
collectively.
implementation of additional risk minimisation measures as agreed with the national medicines
authorities and keep them informed of any changes, challenges or issues encountered in the
implementation of the additional risk minimisation measures. Any relevant changes to the
implementation of the tools should be agreed with the national medicines authorities before
implementation.
In the implementation of web-based tools the applicant or marketing authorisation holder should
apply requirements specific for each Arab country, with particular consideration of potential issues
linked to accessibility, recognisability, responsibility, and privacy and data protection.
For generic products the applicant or marketing authorisation holder should develop risk
minimisation in line with the scope, content, and format of the tools used for the reference medicinal
product. Scheduling and planning of interventions should be carefully coordinated in order to
minimise the burden on the healthcare systems.
For generic products, the effectiveness of risk minimisation measures should be assessed by the
marketing authorisation holders in close co-operation with the national medicines authorities.
Where formal studies are justified, joint studies for all medicinal products involved are strongly
encouraged in order to minimise the burden on the healthcare systems. For instance, if a prospective
cohort study is instituted, study entry should be independent from the prescription of a product with
a specific invented name or marketing authorisation holder. Recording of specific product details
would still be important to enable rapid identification of any new safety hazard with a particular
product.
The marketing authorisation holder shall monitor the outcome of all risk minimisation measures .
General principles for effectiveness evaluation are provided in XVI.B.3..
The applicant or marketing authorisation holder should report the evaluation of the impact of
additional risk minimisation activities when updating the RMP (see V.B.11.4.).
The applicant or marketing authorisation holder should report in the Periodic Safety Update Report
(PSUR) the results of the assessment of the effectiveness of risk minimisation measures which
might have an impact on the safety or risk-benefit assessment (see VII.B.5.16.5.).
The applicant or marketing authorisation holder should ensure timely communication with the
national medicines authorities for relevant regulatory evaluation and actions, as appropriate (see
also XVI.C.2. and Modules V and VII).
In general, the frequency of RMP updates should be proportionate to the risks of the product. The
focus of RMP updates should be on the risk minimisation measures and in providing updates on the
implementation of those measures where applicable. If there is a consequential change to the
summary RMP, this should also be highlighted in the cover letter. Changes to the product
information should not be proposed via a standalone RMP update but rather a variation application
should be submitted. A PSUR can also result directly in an update to product information.
healthcare professionals, sponsor lists, web panels, professional and learned societies may represent
feasible approaches. However, their representativeness for the intended target population of
physicians needs to be carefully reviewed for each study. For patient recruitment the relevant
clinical setting, existing web-panels, and patient advocacy groups should be considered. A
recruitment strategy should be designed while accounting for the chances of achieving accurate and
complete data collection.
Efforts should be made to document the proportion of non-responders and their characteristics to
evaluate potential influences on the representativeness of the sample.
When survey results are weighted, the following key points should be considered:
Differences in selection probabilities (e.g. if certain subgroups were over-sampled);
Differences in response rates;
Post-stratification weighting to the external population;
Clustering.
P.I.A. Introduction
Vaccination is one of the most effective and widely used public health interventions, whose benefits
for individuals and the community has been abundantly demonstrated. Prominent examples are the
global eradication of smallpox and the elimination of poliomyelitis in most countries. As with any
other pharmaceutical product, however, no vaccine is without risks. Robust systems and procedures
must be in place to continuously monitor quality, safety and efficacy. Vaccine pharmacovigilance
has been defined by the CIOMS/WHO Working Group on Vaccine Pharmacovigilance54 as the
science and activities related to the detection, assessment, understanding and communication of
adverse events following immunisation and other vaccine- or immunisation-related issues, and to
the prevention of untoward effects of the vaccine or immunisation.
The objective of this Module is to strengthen the conduct of pharmacovigilance for vaccines. It
should be noted that the overall objectives and processes of pharmacovigilance are similar for
vaccines and other types of medicinal products and this guidance does not replace the information
provided in the other modules of the Good Pharmacovigilance Practices (GVP)- Arab. This Module
focusses on vaccine-specific aspects and unique challenges that should be borne in mind when
designing and implementing pharmacovigilance activities for vaccines.
This Module is relevant to vaccines used for pre- and post-exposure prophylaxis of infectious
diseases and does not cover therapeutic vaccines (e.g. viral-vector based gene therapy, tumour
vaccines, anti-idiotypic vaccines such as monoclonal antibodies used as immunogens). This
guidance is addressed primarily to marketing authorisation holders and competent authorities but
may also be useful to other stakeholders (e.g. sponsors of clinical studies, healthcare professionals,
and public health authorities).
P.I.B. provides guidance specific for vaccines in relation to the main pharmacovigilance processes
described in the Modules of the GVP-Arab. Where applicable, specific recommendations are
provided for situations where vaccines are administered in mass vaccination programmes and where
a large number of reports of suspected adverse reactions is expected in a short period of time.
P.I.C. provides specific guidance related to the operation of the vaccines safety systems in the Arab
Countries.
Other relevant guidance which are acknowledged in the Arab Countries include the EMA Guideline
on Clinical Development of Vaccines55, guidance on design and specific aspects of clinical trials to
be conducted pre and post marketing authorisation, and the EMA Guideline on the Exposure to
Medicinal Products During Pregnancy: Need for Post-Authorisation Data56(see Annex III).
54
Council for International Organizations of Medical Sciences (CIOMS). Definition and application of terms of
vaccine pharmacovigilance (report of CIOMS/WHO Working Group on Vaccine Pharmacovigilance). Genève:
CIOMS; 2012
55
EMEA/CHMP/VWP/164653/2005, available on EMA website http://www.ema.europa.eu.
56
EMEA/CHMP/313666/2005, available on EMA website http://www.ema.europa.eu.
P.I.A.1. Terminology
It is acknowledged that the term Adverse Event Following Immunisation (AEFI) is used at
international level. The term is defined as any untoward medical occurrence which follows
immunisation and which does not necessarily have a causal relationship with the usage of a vaccine.
The adverse event may be any unfavourable or unintended sign, abnormal laboratory finding,
symptom or disease. AEFIs have been further classified by the CIOMS/WHO Working Group on
Vaccine Pharmacovigilance54 (see Annex III) into four categories according to possible causes
(apart from a coincidental event): vaccine product-related, vaccine quality defect-related,
immunisation error-related and immunisation anxiety-related. The term AEFI is not used in this
guidance as the term ―adverse event‖ already designates any untoward medical occurrence in a
patient administered a medicinal product and which does not necessarily have a causal relationship
with this medicinal product (see Annex I).
The terms immunisation (the process of making a person immune) and vaccination (the
administration of a vaccine with the aim to produce immune response) have slightly different
meanings and are not used interchangeably in this guidance. The term vaccination is generally used
unless justified otherwise by the context.
given point in time or under special circumstances, such as in a national emergency, military or
pandemic situation.
Such vaccination programmes are associated with a variety of challenges for pharmacovigilance.
The key ones include:
A large number of suspected adverse reaction reports in a short time period may require
resources for processing, analysing, presenting and communicating data;
It is inevitable that rare or serious incident illnesses will occur in temporal association with
vaccination; new suspected adverse reactions must be very rapidly investigated and
distinguished from coincidental illnesses;
Lack of a comparable unvaccinated concurrent cohort requires alternative statistical and
epidemiological methods to allow appropriate analysis of safety, e.g. case-only designs (see
Appendix 1 of Module VIII and the ENCePP Guide on Methodological Standards in
Pharmacoepidemiology ‖Annex III‖);
Mass vaccination in a short time period may be associated with very unique business continuity
and infrastructure constraints; under such circumstances, specific consideration should be given
to adapting pharmacovigilance plans to meet these challenges and ensure that resource is
prioritised and necessary technical requirements are met (see Module I for public health
emergency planning);
The vaccinated population may include immunocompromised individuals, including those
infected with human immunodeficiency virus (HIV), whose clinical status may not be known at
the time of vaccination and who may be at a higher risk of risk of occurrence of the infectious
disease targeted by the vaccine and of impaired immune response to vaccination, in particular
when vaccinated with live vaccines.
P.I.B.1.2.4. RMP module SVI “Additional EU requirements for the safety specification”
The following aspects should be addressed in this section:
Potential for transmission of infectious agents
For live attenuated vaccines, this section should address aspects such as shedding (including
shedding from vaccinated individuals to unvaccinated close contacts), transmission of the
attenuated agents to close contacts, risk for pregnant women and the foetus, and reversion to
virulence.
As for all biological products, the potential for infections caused by residuals of biological
material used in the manufacturing process as well as contaminations introduced by the
manufacturing process should be evaluated and addressed.
Potential for medication errors
This section should address potential for vaccination errors and mechanisms put in place to
adequately follow-up and investigate the root cause of any errors. Causes of vaccination
errors to be considered include:
Inappropriate handling or breakdown in the cold chain, which may lead to adverse
reactions such as infection due to bacterial contamination of the vaccine, transmission
composition (e.g. adjuvants), immunogenicity and novelty, they represent potential risks that
would need immediate investigation or regulatory action, they could lead to a change in the
benefit-risk balance of the vaccine, or they would require prompt communication to the public by
regulatory or public health authorities; proposal for such adverse events of special interests
(AESIs) may be particularly useful in situations of a mass vaccination programme where it is
expected that a large number of adverse reactions may be reported and their processing may need
to be prioritised.
The information on potential mechanisms for each identified or potential risk should include
available data on association of the risk with the antigen itself, any other ingredient of the vaccine,
including adjuvants, stabilisers, preservatives or residuals of the manufacturing process, the target
population, interactions with other vaccines or medicinal products or the vaccination schedule. If
some of these factors are clearly associated with some identified or potential risks, it may be
appropriate to present these risks in different categories.
Identified and potential interactions with co-administration of other vaccines, including the
increased risk for adverse reactions and clinically relevant immunological interference;
Possible safety concerns reported with combined vaccines such as increased frequency or
severity of known adverse reactions (local or systemic), as small differences of local or systemic
adverse reactions between the combined vaccine and the precursor (combined or individual)
vaccine(s) are usually not detected in pre-authorisation studies;
Any adverse events of special interest (AESIs) identified as an important potential risk in the
safety specification; standard case definitions should be provided (e.g. Brighton Collaboration
case definitions57) and age-stratified data on incidence rates in the population targeted by the
vaccination programme should be compiled; if such data do not exist, they should be included in
the pharmacovigilance plan as data to be collected in the post-authorisation phase (see
P.I.B.1.3.2.);
Inappropriate use of vaccines and patterns of error;
Cases of breakthrough infections, which are expected without necessarily indicating a problem
with the vaccine, as vaccines and vaccination programmes are not 100% effective; although this
issue cannot be fully investigated via spontaneous reporting, reports of vaccine failure can
nonetheless generate signals and risk factors should be analysed (e.g. obesity, age, smoking
status, vaccination schedule, concomitant disease); appropriate case definitions and validated
analytical tests for confirmation of the infective agents should be used whenever possible and the
recommendations of the CIOMS/WHO Working Group on Vaccine Pharmacovigilance54 should
be considered for the definition and classification of cases of vaccination failure;
Adverse reaction reports indicating a possible reversion to virulence, especially for new live
attenuated vaccines; validated and standardised assays, including assays to distinguish between
wild and vaccine strains, should normally be implemented prior to marketing authorisation for
appropriate case assessment.
As part of the routine follow-up of adverse reactions, data should be collected (in addition to data on
the patient, the adverse reaction and the vaccination history) on:
The vaccination schedule and the route of administration;
The vaccine and the diluent (if applicable), including manufacturer(s) and batch number(s);
In case of a suspected quality defect, batch release specifications, expiry date(s) and laboratory
test results about the batch if appropriate, and distribution and administration-related data, such
as storage and handling conditions for vaccines in the healthcare institutions where vaccination
took place;
Relevant comorbidities in the target population (including autoimmune disorders).
Any arrangements established to promptly investigate any emerging issues, such as access to
electronic health records, registries (e.g. pregnancy registries) or other data sources should be
described in this section.
57
Available on Brighton Collaboration website http://www.brightoncollaboration.org.
58
Charlton R and de Vries C, for the European Medicines Agency. Available at
http://www.encepp.eu/structure/documents/Data_sources_for_medicines_in_pregnancy_research.pdf
available to estimate background incidence rates will differ across countries and is likely to impact
diagnostic validity in terms of sensitivity and specificity. The study design should allow
differentiation between prevalent and incident cases. Furthermore, bias could arise from
misclassification of disease type or changes in diagnostic criteria and disease management over the
study period. Whenever possible, data should be stratified by age, sex, geographical region as well
as by other potentially relevant risk factors or confounders. If relevant, seasonal variability should
be taken into account.
In exceptional circumstances (for example in a pandemic with mass vaccination), national
medicines authorities and marketing authorisation holders may agree on an additional system to
rapidly exchange information on emerging safety data whose submission timelines would depend
on the extent of vaccine exposure, epidemiological situation and emerging risk. For example, a
structured worksheet could present the observed and expected numbers of cases and integrate
simple signal detection methods discussed in P.I.B.4., such as observed-to-expected analyses.
Where such an additional system has been agreed, its inclusion as an additional pharmacovigilance
activity in the RMP, along with information on its rationale, format and periodicity, should be
discussed between the marketing authorisation holder and the national medicines authority.
Appropriate national communications to optimise and facilitate reporting may be proposed in
specific situations where mass vaccination takes place and prompt identification and evaluation of
safety concerns are needed. This communication should involve collaboration between marketing
authorisation holders and national regulatory and public health authorities to ensure provision of
information to patients to describe which vaccine they have used, the batch number and how events
can be reported.
in the reporting of adverse events, the MAH should also consider labelling and packaging as risk
minimisation tools.
may however be difficult to implement where they involve populations with high vaccine coverage
rates, an appropriate unvaccinated group is lacking or adequate information on covariates at the
individual level is lacking. The ENCePP Guide on Methodological Standards in
Pharmacoepidemiology describes alternative study designs that can be used in such cases (see
Annex III).
Safety parameters in PASS should be appropriate for the specific vaccine. A pre-requisite is the use
of globally accepted standards for case definitions (e.g. those published by the Brighton
Collaboration57) to compare the frequency of adverse reactions across different studies.
Vaccination registries established in many countries may be used in vaccine safety by creating a
source population for large cohort studies. Using a vaccination registry as a source population for
studies should be made with caution where enrolment may be biased or there is no systematic
collection of exposure in the population. Moreover, a large number of vaccinated individuals is
required for the active surveillance of rare adverse reactions by follow-up of a cohort recruited at the
time of vaccination.
Non-clinical studies and experimental investigations should also be considered to address safety
concerns. This may include virological, bacteriological and/or immunological experiments and
other methods to elucidate the aetiology of an adverse reaction.
59
Council for International Organizations of Medical Sciences (CIOMS). Development and rational use of
Standardised MedDRA Queries (SMQs). Geneva: CIOMS; 2004. Available at http://www.cioms.ch/
objectives of the analysis and the information available in the database. A comparison with all
medicinal products may result in the detection of reactions specifically related to vaccines, but may
also identify a high number of false signals (e.g. SIDS in infants) or already known mild and
expected reactions (e.g. local reactions). On the other hand, using only vaccine-related reports
available in the database may result in signals of age-related reactions (e.g. cardio-vascular
disorders if the vaccine of interest is used in the elderly). In a first step, it may therefore be
appropriate to examine results of statistical methods using both comparator groups, or to use reports
for other vaccines as the comparator group with a stratification made at least by age.
Stratification by geographical region may also be considered and seasonality of vaccine
administration may be relevant for some vaccines and needs consideration. When stratification is
performed, results of both adjusted and non-adjusted analyses should be examined. Results could be
inspected in each stratum as pooled result of a stratified analysis may miss signals.
should be provided, together with information on the date of vaccination, product administered,
manufacturer, batch number, site and route of administration, detailed description and course of the
adverse event/reaction as well as therapeutic intervention. Information on rechallenge, where
applicable, should be recorded. The investigation of clusters of reported adverse events or adverse
reactions is described in the report of the CIOMS/WHO Working Group on Vaccine
Pharmacovigilance54.
Appropriate follow-up of serious suspected adverse reactions is essential, including data on possible
alternative causes. It may be helpful to develop pre-defined check lists or formats for those reactions
which may be anticipated from experience with similar vaccines in order to consistently ascertain
relevant clinical information and support the quality of causality assessment for individual cases
(see also Module VI).
The following aspects need to be considered for signal evaluation:
The incidence of the natural disease in the target population for vaccination and its seasonality,
as this population is usually large and heterogeneous and coincident adverse events are likely to
occur;
Additives and excipients used for the production, inactivation, preservation, and stabilisation of
the vaccine;
Past experience with similar vaccines, adjuvants and types of antigens, in order to identify
adverse reactions which are unexpected and for which a causal relationship remains to be
elucidated;
Distinction between suspected adverse reactions to the vaccine and those reflecting the clinical
picture of the disease for which vaccination has been given (e.g. rash following measles
vaccination);
Public information (public campaign, press) that may favour certain reports in some periods.
vaccines or of alternative vaccines for the vaccination programme should also be considered in this
context.
In situations where a batch-specific quality or safety issue has not been confirmed, measures other
than recall or quarantine may be warranted initially whilst an investigation is on-going, e.g.
providing recommendations on patient surveillance and follow-up post-vaccination. This may be
considered when recall or quarantine may lead to vaccine supply shortages and alternatives are not
widely available.
The following sections present elements that should be taken into account when considering
recalling or quarantining batches.
P.I.B.5.2. Action based on clinical events in the absence of a known quality issue
A batch-specific signal based on an observed clinical event is often based on spontaneous reporting.
In the absence of a known quality issue, decision making on a precautionary recall or quarantine is
difficult, as a causal association with the vaccine can rarely be established at the time when an initial
decision is required.
In the absence of a known quality issue and where there is an apparent increase in frequency or
severity of known adverse reactions without serious clinical risk, consideration should be given to
the geographical distribution of the suspected batch and of the case(s) at the origin of the signal. If it
is established that a suspected batch has been used to a significant extent in many regions/countries
and a signal is apparent in only one geographical area, this could potentially indicate a false signal.
Conversely, an apparent signal in more than one locality may potentially strengthen the signal and
support a recall or quarantine.
For single fatal adverse events, particularly where the cause of death is unknown, the reporting rate
of the event relative to both the usage of the vaccine batch and the ‗expected‘ age-specific all-cause
mortality should be considered before deciding on a recall or quarantine action (see also P.I.B.4.2).
The probability of a chance association should be considered. If a fatal event is initially thought to
be a consequence of a known adverse reaction (e.g. due to anaphylaxis), it would not necessarily
imply a batch-specific issue requiring a recall or a quarantine. On the other hand, where
contamination of a batch is suspected based on individual case details or a localised cluster, due to
possible cold chain and handling deviations, localised action should be considered before escalation
to a national recall or quarantine.
Being transparent and providing explicit information in lay language to the public regarding the use
of (a) vaccine(s) should be fundamental to the communication approach. Incomplete or unclear
messages may lead to confusion of the general public and the decision not to vaccinate or not to be
vaccinated on unsubstantiated grounds. Communication should help preventing anxiety-related
reactions (see Annex I). Any potential risks for specific population groups should be clearly
communicated.
Specific safety communication objectives in relation to vaccines may also aim at avoiding errors in
vaccine handling and administration and at reiterating warnings and precautions for use.
Safety communication about a vaccine should also describe the benefits of vaccines, explain the
risks for individuals and the population of a decrease in vaccination coverage, and explain its impact
on disease control. When drafting communication texts, it should be considered that, as vaccination
programmes mature, incidence rates of the targeted diseases decrease substantially, and so does
personal experience with the disease in a given population. This may result in an increased attention
to concerns related to vaccine safety, and information on the target disease itself may need to be
provided. It should be considered that risk perceptions may differ between stakeholders, especially
when there is uncertainty about a risk. Public confidence in vaccination programmes may only be
maintained by knowledge that systems are in place to ensure complete and rapid assessment and to
take precautionary measures if needed. Therefore, safety communication about vaccines may also
profit from describing key functions of the pharmacovigilance systems.
Communication about vaccines may also include informing vaccinators/healthcare professionals on
the management of vaccine-related anxiety and associated reactions, particularly in individuals with
special conditions (e.g. pregnancy, puberty, immunosensitive conditions, general anxiety or other
mood disorders, epilepsy).
Communication to the public should be a collaborative task undertaken by the industry, regulators
and public health organisations, with input from other stakeholders (see Module XII for collection
of data on information needs and public concerns and Module XI for mechanisms for public
participation).
The processes for planning and implementing safety communication at the level of marketing
authorisation holders and national medicines authorities described in Modules XII and XV apply
and are interlinked with the risk assessment and communication effectiveness evaluation processes
also described in these Modules. Communication interventions may be part of a risk management
plan (RMP) (see Modules V and XVI). During the communication planning and implementing
phases, international collaboration should be facilitated as necessary. Special planning should be
undertaken in case of public health emergencies (see Module I) or pandemics.
Communication planning should include being prepared for frequent public communication needs,
such as those regarding excipients, residues, identified or potential risks for individuals with special
conditions, coincidental events, temporal versus causal association, a single case of an adverse
event rarely identified as a risk, safety monitoring requirements being different to identified risk, or
the mock-up concept not being related to an experimental/not tested/not authorised vaccine. For
the purpose of quantifying safety concerns, relevant background rates, by age group and sex, of
signs and symptoms which are also present in adverse events, whether known to be causally related,
suspected to be causally related or likely to be coincidental, should be kept up-to-date, as well as
exposure data. Communication planning should also include preparing standard texts. Frequently
needed explanations should be ideally tested by representatives of likely target audiences. Concerns
raised by the public should also be addressed by proactively communicating results of benefit-risk
evaluations.
National Medicines authorities should ensure appropriate communication with the public and in
particular the media. Media monitoring should be especially conducted for vaccines. The media can
play an important role in influencing the public perception of vaccine safety, in both a negative and
positive way, and information to the media should be given in timely and meaningful manner (see
Module XII). In this respect, it is essential to maintain a high level of transparency on how
regulatory decisions were reached and on the roles and responsibilities of each stakeholder. In
communication materials, reference should be made to published documents.
of the severe adverse event either from information provided to the vaccinated person or the
patient/carer, or by contacting the medical centre or person that provided the vaccine. Any
suspected adverse reaction should be reported to the national medicines authorities in the Arab
Countries according to national recommendations.
National medicines authorities should have in place a web-based reporting system of suspected
adverse reactions for patients and healthcare professionals, and should encourage these to provide
accurate information on invented names and batch numbers. They should establish channels for an
adequate communication to the public and play an important role in unbiased communication, in
particular in situations where there is a gap between results of scientific analyses made by experts
and public concerns. National medicines authorities should ensure that the public is given important
information on pharmacovigilance concerns relating to the use of the vaccines. Media should
receive timely and relevant information on the benefit-risk balance of vaccines.
National medicines authorities should collaborate with the World Health Organisation in the field of
vaccine safety (see Module XIV).
National medicines authorities should undertake data monitoring of their respective ―National
Pharmacovigilance and Safety reports databases‖, signal detection and signal validation for
respective nationally authorised vaccines (see Module IX).
GVP: Annexes
Annex I: Definitions
Annex II: Templates
Annex II.1. Template of the risk management plan (RMP) in the Arab Countries in
integrated format
Annex II.2. Template of the risk management plan (RMP) in the Arab Countries for generics
Annex II.3. Template of the National Display of the risk management plan (RMP) in the
Arab Countries - for MAH/Applicant having Eu RMP
Annex II.4. Templates: Cover page of periodic safety update report (PSUR)
Annex II.5. Templates: Direct healthcare-professional communication (DHPC)
Annex III: Abbreviations
Annex IV: Other Pharmacovigilance Guidance
Annex V: International Conference on Harmonisation of Technical Requirements for
Registration of Pharmaceuticals for Human Use (ICH) guidelines for
pharmacovigilance
GVP: Annexes
Annex I – Definitions
Adverse reaction; synonyms: Adverse drug reaction (ADR), Suspected adverse (drug)
reaction, Adverse effect, Undesirable effect
A response to a medicinal product which is noxious and unintended60.
Response in this context means that a causal relationship between a medicinal product and an
adverse event is at least a reasonable possibility (see Annex IV, ICH-E2A Guideline).
Adverse reactions may arise from use of the product within or outside the terms of the marketing
authorisation or from occupational exposure. Conditions of use outside the marketing authorisation
include off-label use, overdose, misuse, abuse and medication errors.
See also Adverse event, Serious adverse reaction, Unexpected adverse reaction, Off-label use,
Overdose, Misuse of a medicinal product, Abuse of a medicinal product, Occupational exposure to
a medicinal product
Audit
A systematic, disciplined, independent and documented process for obtaining audit evidence and
evaluating it objectively to determine the extent to which the audit criteria are fulfilled (see ISO
19011 (3.1)61).
60
In the context of clinical trials, an adverse reaction is defined as all untoward and unintended responses to an
investigational medicinal product related to any dose administered.
61
International Organization for Standardization (ISO); www.iso.org
Audit finding(s)
Results of the evaluation of the collected audit evidence against audit criteria (see ISO19011
(3.4)61).
Audit evidence is necessary to support the auditor‘s results of the evaluation, i.e. the auditor‘s
opinion and report. It is cumulative in nature and is primarily obtained from audit procedures
performed during the course of the audit. See also Audit
Audit plan
Description of activities and arrangement for an individual audit (see ISO19011 (3.12)61).
See also Audit
Audit programme
Set of one or more audits planned for a specific timeframe and directed towards a specific purpose
(see ISO 19011 (3.11)61). See also Audit
Audit recommendation
Describes the course of action management might consider to rectify conditions that have gone
awry, and to mitigate weaknesses in systems of management control (see Sawyer LB et al, 200362).
Audit recommendations should be positive and as specific as possible. They should also identify
who is to act on them (Sawyer LB et al, 200362). See also Audit
Clinical trial
Any investigation in human subjects intended to discover or verify the clinical, pharmacological
and/or other pharmacodynamic effects of one or more investigational medicinal product(s), and/or
to identify any adverse reactions to one or more investigational medicinal product(s) and/or to study
absorption, distribution, metabolism and excretion of one or more investigational medicinal
product(s) with the objective of ascertaining its (their) safety and/or efficacy. This includes clinical
trials carried out in either one site or multiple sites, whether in one or more Country.
See also Ongoing clinical trial, Completed clinical trial, Investigational medicinal product
Closed signal
In periodic benefit-risk evaluation reports, a signal for which an evaluation was completed during
the reporting interval (see Annex IV, ICH-E2C(R2) Guideline).
This definition is also applicable to periodic safety update reports. See also Signal
62
Sawyer LB, Dittenhofer MA. Sawyer‘s Internal Auditing. 5th ed. Altamonte Springs, FL: The IIA Research
Foundation; 2003.
Consumer
For the purpose of reporting cases of suspected adverse reactions, a person who is not a healthcare
professional such as a patient, lawyer, friend or relative/parent/child of a patient (see Annex IV,
ICH-E2D Guideline).
Crisis
A situation where, after assessment of the associated risks, urgent and coordinated action within the
country is required to manage and control the situation See also Incident
included in a PSUR.
For a periodic benefit-risk evaluation report (PBRER), the date designated as the cut-off date for
data to be included in a PBRER, based on the international birth date (see Annex IV, ICH-E2C(R2)
Guideline).
For a development safety update report (DSUR), the date designated as the cut-off date for data to
be included in a DSUR, based on the development international birth date (see ICH-E2F Guideline).
Date includes day and month (see ICH-E2F Guideline).
See also Periodic safety update report, Development safety update report, International birth date,
Development international birth date
Failure to vaccinate
63
An indicated vaccine was not administered appropriately for any reason (see CIOMS-WHO ).
For interpreting what is appropriate, consider the explanatory note for Immunisation error-related
reaction.
63
Council for International Organizations of Medical Sciences (CIOMS). Definition and application of terms of
vaccine pharmacovigilance (report of CIOMS/WHO Working Group on Vaccine Pharmacovigilance). Genève:
CIOMS; 2012.
Healthcare professional
For the purposes of reporting suspected adverse reactions, healthcare professionals are defined as
medically qualified persons, such as physicians, dentists, pharmacists, nurses and coroners (see
Annex IV, ICH-E2D Guideline).
Identified risk
An untoward occurrence for which there is adequate evidence of an association with the medicinal
Illegal purposes
See Misuse for illegal purposes
Immunisation
The process of making a person immune.
For the context of Considerations P.I, immunisation refers to the process of making a person
immune to an infection.
See also Vaccination
Incident
A situation where an event occurs or new information arises, irrespective whether this is in the
public domain or not, in relation to (an) authorised medicinal product(s) which could have a serious
impact on public health.
The incident may be related to quality, efficacy or safety concerns, but most likely to safety and/or
quality (and possibly subsequent supply shortages). In addition, situations that do not seem at a first
glance to have a serious impact on public health, but are in the public domain - subject of media
attention or not- and may lead to serious public concerns about the product, may also need to be
considered as incidents. Likewise, other situations which might have a negative impact on the
appropriate use of a medicinal products (e.g. resulting in patients stop taking their medicine) may
fall within the definition of an incident.
Individual case safety report (ICSR); synonym: Adverse (drug) reaction report
Format and content for the reporting of one or several suspected adverse reactions to a medicinal
product that occur in a single patient at a specific point of time.
In the context of a clinical trial, an individual case is the information provided by a primary source
to describe suspected unexpected serious adverse reactions related to the administration of one or
more investigational medicinal products to an individual patient at a particular point of time
See also Minimum criteria for reporting
Investigational drug
Experimental product under study or development. This term is more specific than investigational
medicinal product, which includes comparators and placebos (see ICH-E2F Guideline).
See also Investigational medicinal product
Labelling
Information on the immediate or outer packaging.
Medicinal product
Any substance or combination of substances
presented as having properties for treating or preventing disease in human beings; or
which may be used in or administered to human beings either with a view to restoring, correcting
or modifying physiological functions by exerting a pharmacological, immunological or
metabolic action, or to making a medical diagnosis.
Missing information
Gaps in knowledge, related to safety or particular patient populations, which could be clinically
significant.
The change of the term in Arab Countries, to name this concept ―missing information‖ rather than
―important missing information‖, is to be clear that in the Arab Country concerned a marketing
authorisation cannot be granted if there are unacceptable gaps in knowledge, a marketing
authorisation shall be refused if the quality, safety or efficacy are not properly or sufficiently
demonstrated.
The common name is the international non-proprietary name (INN) recommended by the World
Health Organization, or, if one does not exist, the usual common name.
The complete name of the medicinal product is the name of the medicinal product followed by the
strength and pharmaceutical form.
Off-label use
Situations where a medicinal product is intentionally used for a medical purpose not in accordance
with the authorised product information.
Off-label use includes use in non-authorised paediatric age categories. Unless specifically
requested, it does not include use outside the Arab Country concerned in an indication authorised in
that territory which is not authorised in the this Arab country.
Ongoing signal
In periodic benefit-risk evaluation reports, a signal that remains under evaluation at the data lock
point (see Annex IV, ICH-E2C(R2) Guideline).
This definition is also applicable to periodic safety update reports.
See also Signal, Data lock point
Overdose
Administration of a quantity of a medicinal product given per administration or cumulatively which
is above the maximum recommended dose according to the authorised product information.
Clinical judgement should always be applied.
Package leaflet
A leaflet containing information for the user which accompanies the medicinal product.
Pharmacovigilance
Science and activities relating to the detection, assessment, understanding and prevention of adverse
effects or any other medicine-related problem (see WHO).
In line with this general definition, underlying objectives of pharmacovigilance in accordance with
the applicable legislation for are:
preventing harm from adverse reactions in humans arising from the use of authorised medicinal
products within or outside the terms of marketing authorisation or from occupational exposure;
and
promoting the safe and effective use of medicinal products, in particular through providing
timely information about the safety of medicinal products to patients, healthcare professionals
and the public.
Pharmacovigilance is therefore an activity contributing to the protection of patients‘ and public
health.
Pharmacovigilance system
A system used by the marketing authorisation holder and by national medicines authoritites to fulfil
the pharmacovigilance tasks and responsibilities listed in national regulations and designed to
monitor the safety of authorised medicinal products and detect any change to their risk-benefit
balance.
In general, a pharmacovigilance system is a system used by an organisation to fulfil its legal tasks
and responsibilities in relation to pharmacovigilance and designed to monitor the safety of
authorised medicinal products and detect any change to their risk-benefit balance.
Potential risk
An untoward occurrence for which there is some basis for suspicion of an association with the
medicinal product of interest but where this association has not been confirmed (see ICH-E2F
Guideline.
Examples include:
non-clinical toxicological findings that have not been observed or resolved in clinical studies;
adverse events observed in clinical trials or epidemiological studies for which the magnitude of
the difference, compared with the comparator group (placebo or active substance, or unexposed
group), on the parameter of interest raises a suspicion of, but is not large enough to suggest, a
causal relationship;
a signal arising from a spontaneous adverse reaction reporting system;
an event known to be associated with other active substances within the same class or which
could be expected to occur based on the properties of the medicinal product (see ICH-E2F
Guideline).
See also Adverse event, Signal
Quality adherence
Carrying out tasks and responsibilities in accordance with quality requirements.
See also Quality requirements
Quality assurance
See Quality control and assurance
Quality improvements
Correcting and improving the structures and processes where necessary.
This applies for the purpose of fulfilling quality requirements.
See also Quality requirements
Quality objectives
See Quality requirements
Quality planning
Establishing structures and planning integrated and consistent processes.
This applies for the purpose of fulfilling quality requirements.
See also Quality requirements
Quality requirements
Those characteristics of a system that are likely to produce the desired outcome, or quality
objectives.
See also Pharmacovigilance system, Quality system of a pharmacovigilance system
Registry
An organised system that uses observational methods to collect uniform data on specified outcomes
in a population defined by a particular disease, condition or exposure.
Risk-benefit balance
An evaluation of the positive therapeutic effects of the medicinal product in relation to the risks, i.e.
any risk relating to the quality, safety or efficacy of the medicinal product as regards patients‘ health
or public health.
See also Risks related to use of a medicinal product
Safety concern
An important identified risk, important potential risk or missing information.
It is noted that the ICH definition of safety concern is: an important identified risk, important
potential risk or important missing information, i.e. includes the qualifier ―important‖ in relation to
missing information (see Annex IV, ICH-E2C(R2) Guideline). The ICH-E2E Guideline (see Annex
IV) uses the terms safety issue and safety concern interchangeably with the same definition for
safety concern as defined in the ICH-E2C(R2) Guideline.
See also Important identified risk and Important potential risk, Missing information
Signal
Information arising from one or multiple sources, including observations and experiments, which
suggests a new potentially causal association, or a new aspect of a known association between an
intervention and an event or set of related events, either adverse or beneficial, that is judged to be of
sufficient likelihood to justify verificatory action.
For the purpose of Section 16.2 of the periodic benefit-risk evaluation report, signals relate to
adverse effects (see Annex IV, ICH-E2C(R2) Guideline).
See also Validated signal, Newly identified signal, Closed signal, Ongoing signal, Signal
management process, Adverse reaction
Signal validation
Process of evaluating the data supporting a detected signal in order to verify that the available
documentation contains sufficient evidence demonstrating the existence of a new potentially causal
association, or a new aspect of a known association, and therefore justifies further analysis of the
signal.
See also Validated signal
Stimulated reporting
See Spontaneous report
Substance
Any matter irrespective of origin which may be human (e.g. human blood and human blood
products), animal (e.g. micro-organisms, whole animals, parts of organs, animal secretions, toxins,
extracts, blood products), vegetable (e.g. micro-organisms, plants, part of plants, vegetable
secretions, extracts), chemical (e.g. elements, naturally occurring chemical materials and chemical
products obtained by chemical change or synthesis).
Upper management
Group of persons in charge of the highest executive management of an organisation. Membership of
this group is determined by the governance structure of the organisation. While it is envisaged that
the upper management usually is a group, the head of the organisation is the one person at the top of
the organisation with ultimate responsibility for ensuring that the organisation complies with
relevant legislation.
Vaccination
The administration of a vaccine with the aim to produce immune response.
See also Immunisation
Vaccination failure
Vaccination failure due to actual vaccine failure or failure to vaccinate (see CIOMS-WHO 63).
Vaccination failure may be defined based on clinical endpoints or immunological criteria, where
correlates or surrogate markers for disease protection exist. Primary failure (e.g. lack of
seroconversion or seroprotection) needs to be distinguished from secondary failure (waning
immunity) (see CIOMS-WHO 63).
See also Vaccine failure, Failure to vaccinate
Vaccine
See Immunological medicinal product
Vaccine failure
Confirmed or suspected vaccine failure.
64
For investigational medicinal products, an unexpected adverse reaction is an adverse reaction, the nature or
severity of which is not consistent with the applicable product information (e.g. the investigator‘s brochure for an
unauthorised investigational product or the summary of product characteristics for an authorised product).
Vaccine pharmacovigilance
The science and activities relating to the detection, assessment, understanding and communication
of adverse events following immunisation and other vaccine- or immunisation-related issues, and to
the prevention of untoward effects of the vaccine or immunisation (see CIOMS-WHO 63).
In this definition, immunisation means the usage of a vaccine for the purpose of immunising
individuals (see CIOMS-WHO 63), which in the Arab Countries is preferably referred to as
vaccination (in the report of CIOMS/WHO Working Group on Vaccine Pharmacovigilance the
terms immunisation and vaccination are used interchangeably). Usage includes all processes that
occur after a vaccine product has left the manufacturing/packaging site, i.e. handling, prescribing
and administration of the vaccine (see CIOMS-WHO 63).
An adverse event following immunisation (AEFI) is any untoward medical occurrence which
follows immunisation and which does not necessarily have a causal relationship with the usage of
the vaccine. The adverse event may be any unfavourable or unintended sign, abnormal laboratory
finding, symptom or disease. While this AEFI definition is compatible with the definition of adverse
event applied in the Arab Countries, the AEFI definition is not needed to describe
pharmacovigilance for vaccines in the Arab Countries. However, Arab Countries guidance on
pharmacovigilance for vaccines makes use of the terminology suggested by CIOMS-WHO
regarding possible causes of adverse events, turning them into suspected adverse reactions. A
coincidental event is an AEFI that is caused by something other than the vaccine product,
Validated signal
A signal where the signal validation process of evaluating the data supporting the detected signal
has verified that the available documentation contains sufficient evidence demonstrating the
existence of a new potentially causal association, or a new aspect of a known association, and
therefore justifies further analysis of the signal.
See also Signal
GVP: Annexes
Annex II – Templates
Pharmaco-therapeutic group
(ATC Code):
Data lock point for this RMP <Enter a date> Version number <Enter a version no>
In some circumstances there may be a need to submit a third RMP which is a different version from
both the agreed RMP and a second RMP version currently undergoing evaluation e.g. if new safety
concerns have been recently identified or if a new indication requires different risk minimisation
measures. In this case different versions of a RMP will be simultaneously under evaluation. The
purpose of this section is to provide oversight.
Overview of versions:
Version number of last agreed RMP:
Version number <Enter a version no>
… etc.
Posology and route of Current (if applicable) in the Arab Country concerned
administration in the Arab Country
concerned
Current of the reference medicinal product/ this product in the
EEA
Pharmaceutical form(s) and Current (if applicable) in the Arab Country concerned
strengths
Current of the reference medicinal product/ this product in the
EEA
Country and date of first authorisation worldwide <Enter a country> <Enter a date>
Country and date of first launch worldwide <Enter a country> <Enter a date>
<Enter a date>
65
This is a European system which is adopted by the Arab Countries unless otherwise announced by the
national medicines authority(s). For more information on additional monitoring see GVP in Arab Countries
Module X: additional monitoring.
The list of medicines under additional monitoring includes medicines authorised in the European Union (EU)
that are being monitored particularly closely by regulatory authorities. Medicines under additional
monitoring have a black inverted triangle displayed in their package leaflet and summary of product
characteristics, together with a short sentence explaining what the triangle means.
Indication
Key Safety findings (from non- clinical studies) Relevance to human usage
Toxicity including:
Single and repeat-dose toxicity,
reproductive (must be discussed if medicine
might be used in women of child-bearing
potential)
developmental toxicity
nephrotoxicity
hepatotoxicity
genotoxicity
carcinogenicity
General safety pharmacology:
cardiovascular (including potential for QT
interval prolongation)
nervous system
etc.
Mechanisms for drug interactions
Other toxicity-related information or data
Specify whether there is a need for additional non-clinical data if the medicinal product(s) is/are to
be used in special populations
Indication 1(person time should only be provided for final duration category and total )
Duration of exposure (at least) Persons Person time
1m
3m
6m
12 m etc.
Total person time
Indication 2 (person time should only be provided for final duration category and total )
Duration of exposure (at least) Persons Person time
1m
3m
6m
12 m etc.
Total person time
Total exposed population (person time should only be provided for final duration category and total )
Duration of exposure (at least) Persons Person time
1m
3m
6m
12 m etc.
Total person time
Indication 1
Dose of exposure Persons Person time
Dose level 1
Dose level 2 etc.
Total
Indication 2
Dose of exposure Persons Person time
Dose level 1
Dose level 2 etc.
Total
Total Population
Dose of exposure Persons Person time
Dose level 1
Dose level 2 etc.
Total
When providing data by age group, the age group should be relevant to the target population.
Artificial categories such as <65, >65 should be avoided. Paediatric data should be divided by
categories (e.g. ICH-E11) similarly the data on mature patients should be stratified into categories
such as 65-74, 75-84 and 85+ years. For teratogenic drugs, stratification into age categories
related to childbearing potential might be appropriate for the female population. If the RMP
includes more than one medicinal product, the total population table should be provided for each
product as well as a combined table.
Indication 1
Age group Persons Person time
M F M F
Age group 1
Age group 2 etc.
Total
Indication 2
Age group Persons Person time
M F M F
Age group 1
Age group 2 etc.
Total
Total population
Age group Persons Person time
M F M F
Age group 1
Age group 2 etc.
Total
Indication 1
Ethnic/racial origin Persons Person time
Ethnic origin 1
Ethnic origin 2 etc.
Total
Indication 2
Ethnic/racial origin Persons Person time
Ethnic origin 1
Ethnic origin 2 etc.
Total
Total population
Ethnic/racial origin Persons Person time
Ethnic origin 1
Ethnic origin 2 etc.
Total
Indication 1
Persons Person time
Pregnant women
Lactating women
Renal impairment (specify or categorise)
Hepatic impairment (specify or categorise)
Cardiac impairment (specify or categorise)
Sub populations with genetic polymorphism (specify)
Immuno-compromised
Indication 2
Persons Person time
Pregnant women
Lactating women
Renal impairment (specify or categorise)
Hepatic impairment (specify or categorise)
Cardiac impairment (specify or categorise)
Sub populations with genetic polymorphism (specify)
Immuno-compromised
Total population
Persons Person time
Pregnant women
Lactating women
Renal impairment (specify or categorise)
Hepatic impairment (specify or categorise)
Cardiac impairment (specify or categorise)
Sub populations with genetic polymorphism (specify)
Immuno-compromised
Which are rare (it may be <E.g. 12,600 patients were <E.g. ADRS with a frequency
appropriate to choose other ADR exposed over the whole CT greater than 1 in 4,200 could be
frequencies) programme> detected if there were no
background incidence>
Due to prolonged exposure <E.g. 3000 women were exposed <E.g. There does not appear to be
to X for more than 4 years during an effect on endometrial
which time there were no cases of proliferation during the first 4
endometrial carcinoma. 42 years of treatment. X is thought
women in the treated to ………………etc.>
experienced endometrial
hyperplasia compared with 35 in
the non-exposed group (2000)>
Due to cumulative effects <e.g. specific organ toxicity>
Which have a long latency
Children
Special consideration should be given to the experience in different paediatric age groups – e.g.
ICH-E11 - since these relate to different physiological and anatomical development stages. If
paediatric development has been limited to certain age categories then the implications for other
paediatric age groups should also be discussed.
Pre-term newborns
Neonate (birth to 27 days)
Infants and toddlers (28 days to 23 months)
Children (2 years to e.g. 11 years)
Adolescents (e.g. 12 years to 17 years)
Elderly
Implications on the use in patients of 65 and older should be discussed with appropriate
consideration to the top ranges of the age spectrum. The effect of individual impairment should be
discussed in the sections below but the effects of multiple (minor) co-existing impairments and also
adverse reactions of particular concern in the elderly should be discussed.
Use in different age ranges: e.g. 65-74, 75-84, >85
Need for laboratory screening prior to use
Effect of multiple co-existing impairments
Adrs of special concern – e.g. dizziness, CNS effects
Effect of multiple medications
Patients with a disease severity different from the inclusion criteria in the clinical trial
population
Safety concerns due to limitations of the clinical trial programme Outstanding concern?
Table 1. Detailed description of action taken since last update to this module
Safety issue
Background to issue
Evidence source
Action taken
Countries affected
Date(s) of action
Safety concern 1
SV.2.2 Exposure
Indication
Age Group Persons Exposure (e.g. packs or person years)
M F M F
Age group 1
Age group 2
Etc.
By indication
By route of administration
By dose
Indication
Persons Exposure (e.g. packs or person years)
Dose level 1
Dose level 2
Etc.
By country
Indication
Persons Exposure (e.g. packs or person years)
Arab Country concerned
Other countries
Note the categories provided, are suggestions and other relevant variables can be used e.g. oral
versus i.e., duration of treatment etc.
Paediatric use
Paediatric use
Pre-term new-borns
Neonates (birth to 27 days)
Infants and toddlers (1 month to 23
months)
Children (2 years to e.g. 11 years)
Adolescents (e.g. 12 years to 18 years)
Data source
Method of calculation
Elderly use
65 – 74 years
75 – 84 years
85+ years
Data source
Method of calculation
Pregnant
Breast feeding
Data source
Method of calculation
Hepatic impairment
Mild
Moderate
Severe
Data source
Method of calculation
Renal impairment
Mild
Moderate
Severe
Data source
Method of calculation
Specify category
Specify category
Specify category
Data source
Method of calculation
Part II: Module SVI - Additional requirements for the safety specification
Product name(s)
Product name(s)
Where multiple strengths, posologies or concentrations are available, or where different products
have different formulations, reconstitution differences etc., consideration should be given to
including “medication error” as a safety concern.
SVI.7 Conclusions
Safety concerns from this module (to be carried through to Part II Module SVIII)
SVII.1 Newly identified safety concerns (since this module was last submitted)
Safety concern
Details
Source
New studies proposed in pharmacovigilance plan? Yes/No
New risk minimisation actions proposed? Yes/No
here. The conclusions should be incorporated into the other sections and modules of the safety
specification as appropriate with detailed information on the risk provided in SVII.3.
Details of the above safety concerns should also be provided below.
66
For definitions see Good Vigilance Practices (GVP) Module V, chapter V.B.1 .
Frequency with 95 % CI State clearly which frequency parameter is being used e.g.
incidence rate or incidence risk and the data source e.g. blinded
clinical trial population, epidemiological study. For identified
risks incidence should be presented for the whole population and
relevant subpopulation categories.
(see also section V.B.8.7.3 of GVP Module V)
Where there are clear differences in rates between populations, this
should be discussed
Severity and nature of risk e.g. tabulate grades of severity where available
Risk groups or risk factors Describe patient factors, dose, time or other factors where available
including additive or synergistic factors
Potential public health impact of Describe or enumerate if possible, using e.g. Numbers Needed to
safety concern Harm and/or expected number of patients affected, hospitalisations,
fatalities given the predicted population use.
Evidence source Identify, briefly describe and cross refer to supporting data in CTD
or annex
Effect of interaction
Evidence source
Possible mechanisms
Potential health risk
Discussion
Consider including “interactions” as a safety concern in Part II Module SVIII.
Seriousness/outcomes
Severity and nature of risk e.g. tabulate grades of severity where available
Comment
The Pharmacovigilance plan (PhV Plan) provides details of pharmacovigilance activities/ studies
which are intended to identify and/or characterise safety concerns. What is required will depend
upon the nature of the medicine, the target population, the number of safety concerns and where the
medicine is in its life-cycle. A PhV Plan may also include details of studies to measure the
effectiveness of risk minimisation measures for important measures where a formal study is
required.
Some safety concerns may be well characterised in which case routine PhV will be sufficient.
Depending upon the safety concern, and areas to be investigated, a PhV Plan will often include
epidemiological (non-interventional) studies (such as cohort, case control, registries, drug
utilisation etc.) but may also include interventional studies or more rarely pre-clinical activities
(such as PK/PD, clinical trials, in vivo or in vitro studies). Further information on post
authorisation safety studies is given in GVP Module VIII.
In the PhV Plan, section III.1 reviews each safety concern and what areas need investigation
whereas III.4 gives details of the individual studies and milestones. Section III.2 provides details
of any activities aimed at measuring the effectiveness of risk minimisation activities. The results
of any studies in the PhV Plan should be briefly summarised in section III.3. If the study results
concern the effectiveness of risk minimisation, brief results should be provided in section III.3. If
the results suggest that the risk minimisation measure is failing in its objectives, this should be
discussed with the root cause analysis and proposal for rectification in Part V of the RMP. Section
III.5 summarises the entire PhV plan – both completed, on-going and planned activities.
Study/activity title
Safety concern
Objective(s) of the risk minimisation measures
Routine risk minimisation measures (Proposed) text in SmPC
<E.g. Dose reduction for ……. in section 4.2 of the
SPC………
Warning in section 4.4 to……
Listed in section 4.8>
Comment (e.g. on any differences between SmPCs)
Other routine risk minimisation measures
<E.g. Prescription only medicine
Use restricted to physicians experienced in the
treatment of…….>
Additional risk minimisation measure(s)1 Objective and justification of why needed.
Safety concern
Risk minimisation measure(s)
Component 1 Analysis
Component 2 etc. Analysis
Discussion
Safety concern
Objective(s) of the risk minimisation activities
Routine risk minimisation activities Synopsis of (proposed) text in SmPC
In addition to the listing of the safety concerns in the above table; each safety concern should be
briefly described using the following tables.
Important identified risks
Missing information
Current (or proposed if product is not authorised) local (of the concerned Arab Country) summary
of product characteristics (SmPC) and package leaflet(s) for each product in the RMP.
If multiple versions are included for a product, they should show in which Country(s) they are
applicable. In addition, if available, a core SmPC should be provided with an overview of the
changes applicable to the SmPC in the Arab Country concerned.
*
Enter the date of the most recent change to the licence status: eg date of approval or date of
suspension
RMP Annex 6 - Protocols for proposed and on-going studies in the section “Summary table of
additional pharmacovigilance activities” in RMP part III
*
Draft = not approved
Approved = when agreed by national authority as appropriate
Provide forms
RMP Annex 8 - Protocols for proposed and on-going studies in RMP part IV
*
Draft = not approved
Approved = when agreed by Authority
RMP Annex 9 - Newly available study reports for RMP parts III & IV
Include the study abstract. For non-interventional studies use the abstract format detailed in
Module: VIII Post Authorisation Safety Studies of Good Pharmacovigilance Safety Studies
RMP Annex 10 - Details of proposed additional risk minimisation measures (if applicable)
RMP Annex 11 - Mock-up of proposed additional risk minimisation measures (if applicable)
Mock up examples in English (unless other language is requested by the medicines authority of the
Arab Country concerned) (of the material provided to healthcare professionals and patients. For
those materials directed to patients, in addition to the English version, Arabic translation of the
mock up shall be included as well.
Pharmaco-therapeutic group
(ATC Code):
Data lock point for this RMP <Enter a date> Version number <Enter a version no>
VI.4 Summary of changes to the Risk Management Plan over time .....................................................
Part VII: RMP Annexes ...................................................................................................................
RMP Annex 1 – ―National Pharmacovigilance and Safety reports database‖ /"National Pharmacovigilance
Issues Tracking Tool" Interface ....................................................................................................
RMP Annex 2 - SmPC & Package Leaflet ......................................................................................................
RMP Annex 3 - Worldwide marketing authorisation by country (including Arab Country(s) concerned).....
RMP Annex 4 - Synopsis of on-going and completed clinical trial programme .............................................
RMP Annex 5 - Synopsis of on-going and completed pharmacoepidemiological study programme .............
RMP Annex 6 - Protocols for proposed and on-going studies in the section ―Summary table of additional
pharmacovigilance activities‖ in RMP part III ..............................................................................
RMP Annex 7 - Specific adverse event follow-up forms ................................................................................
RMP Annex 8 - Protocols for proposed and on-going studies in RMP part IV...............................................
RMP Annex 9 - Newly available study reports for RMP parts III & IV .........................................................
RMP Annex 10 - Details of proposed additional risk minimisation measures (if applicable).........................
RMP Annex 11 - Mock-up of proposed additional risk minimisation measures (if applicable) .....................
RMP Annex 12 - Other supporting data (including referenced material)........................................................
This guidance covers the Parts and modules of the abridged RMP which may be required for
applications concerning generics in the Arab Countries. Modules and sections in the RMP which
are ALWAYS NOT required from generics are omitted in this guidance of abridged RMP. Please
note that the naming and numbering of the parts, modules & sections are standardised thus should
NOT be changed due to the omission of unrequired sections.
Other sections of the abridged RMP apply to generics in ONLY certain situations as described
below those have been provided here for completeness. Parts III and IV many not be required and
applicants are encouraged to discuss the need with the competent authority prior to submission of
the RMP.
* A new RMP version number should be assigned each time any Parts/modules are updated
Overview of versions:
Version number of last agreed RMP:
Version number <Enter a version no>
… etc.
Posology and route of Current (if applicable) in the Arab Country concerned
administration in the Arab Country
concerned
Current of the reference medicinal product *
Pharmaceutical form(s) and Current (if applicable) in the Arab Country concerned
strengths
Current of the reference medicinal product *
Country and date of first authorisation worldwide <Enter a country> <Enter a date>
Country and date of first launch worldwide <Enter a country> <Enter a date>
<Enter a date>
Date of first authorisation (if authorised) in the
Arab Country concerned
67
This is a European system which is adopted by the Arab Countries unless otherwise announced by the
national medicines authority(s). For more information on additional monitoring see GVP in Arab Countries
Module X: additional monitoring.
The list of medicines under additional monitoring includes medicines authorised in the European Union (EU)
that are being monitored particularly closely by regulatory authorities. Medicines under additional
monitoring have a black inverted triangle displayed in their package leaflet and summary of product
characteristics, together with a short sentence explaining what the triangle means.
Table 7. Detailed description of action taken since last update to this module
Safety issue
Background to issue
Evidence source
Action taken
Countries affected
Date(s) of action
Safety concern 1
SV.2.2. Exposure
Indication
Age Group Persons Exposure (e.g. packs or person years)
M F M F
Age group 1
Age group 2
Etc.
By indication
By route of administration
By dose
Indication
Persons Exposure (e.g. packs or person years)
Dose level 1
Dose level 2
Etc.
By country
Indication
Persons Exposure (e.g. packs or person years)
Arab Country concerned
Other countries
Note the categories provided, are suggestions and other relevant variables can be used e.g. oral
versus I.V.., duration of treatment etc.
Paediatric use
Pre-term new-borns
Paediatric use
Method of calculation
Elderly use
65 – 74 years
75 – 84 years
85+ years
Data source
Method of calculation
Pregnant
Breast feeding
Data source
Method of calculation
Hepatic impairment
Mild
Moderate
Severe
Data source
Method of calculation
Renal impairment
Mild
Moderate
Severe
Data source
Method of calculation
Specify category
Specify category
Specify category
Data source
Method of calculation
Study/activity title
Study (type and Objectives Efficacy Status (planned, Date for submission
study number) uncertainties started) of interim or final
addressed reports
Safety concern
Objective(s) of the risk minimisation measures
Routine risk minimisation measures (Proposed) text in SmPC
<E.g. Dose reduction for ……. in section 4.2 of the
SPC………
Warning in section 4.4 to……
Listed in section 4.8>
Comment (e.g. on any differences between SmPCs)
Other routine risk minimisation measures
<E.g. Prescription only medicine
Use restricted to physicians experienced in the
treatment of…….>
Additional risk minimisation measure(s)1 Objective and justification of why needed.
Proposed actions/components and rationale
Additional risk minimisation measure(s) 2 Objective and justification of why needed.
(repeat as necessary) Proposed actions/components and rationale
Safety concern
Risk minimisation measure(s)
Component 1 Analysis
Component 2 etc. Analysis
Discussion
Safety concern
Objective(s) of the risk minimisation activities
Routine risk minimisation activities Synopsis of (proposed) text in SmPC
In addition to the listing of the safety concerns in the above table; each safety concern should be
briefly described using the following tables.
Missing information
Current (or proposed if product is not authorised) local(of the concerned Arab Country) summary
of product characteristics (SmPC) and package leaflet(s) for each product in the RMP.
If multiple versions are included for a product, they should show in which Country(s) they are
applicable. In addition, if available, a core SmPC should be provided with an overview of the
changes applicable to the SmPC in the Arab Country concerned.
*
Enter the date of the most recent change to the licence status: eg date of approval or date of
suspension
A3.2 Licensing status in the rest of the world
RMP Annex 6 - Protocols for proposed and on-going studies in the section “Summary table of
additional pharmacovigilance activities” in RMP part III
*
Draft = not approved
Approved = when agreed by national authority as appropriate
Provide forms
RMP Annex 8 - Protocols for proposed and on-going studies in RMP part IV
*
Draft = not approved
Approved = when agreed by Authority
RMP Annex 9 - Newly available study reports for RMP parts III & IV
Include the study abstract. For non-interventional studies use the abstract format detailed in
Module: VIII Post Authorisation Safety Studies of Good Pharmacovigilance Safety Studies
RMP Annex 10 - Details of proposed additional risk minimisation measures (if applicable)
RMP Annex 11 - Mock-up of proposed additional risk minimisation measures (if applicable)
Mock up examples in English (unless other language is requested by the medicines authority of the
Arab Country concerned) (of the material provided to healthcare professionals and patients. For
those materials directed to patients, in addition to the English version, Arabic translation of the
mock up shall be included as well..
For the EU RMP which is the reference of this National Display (referenced EU RMP):
Risk management is a global activity. However, because of differences in indication and healthcare
systems, target populations may be different across the world and risk minimisation activities will
need to be tailored to the system in place in a particular country or global region. In addition,
differences in disease prevalence and severity, for example, may mean that the benefits of a
medicinal product may also vary between regions. Therefore a product may need different or
supplementary activities in the RMP for each region although there will be core elements which
are common to all. For example much of the safety specification will be the same regardless of
where the medicinal product is being used but the epidemiology of the disease may vary between
e.g. Africa and Europe, and there may be additional or fewer safety concerns depending upon the
target population and indication.
Furthermore, individual countries may have different health systems and medical practice may
differ between countries so the conditions and restrictions in the marketing authorisation may be
implemented in different ways depending upon national customs.
MAH/ Applicants are required to submit RMP to the medicines authority of the Arab Country
concerned in the situations described in Module V section V.C.3.
Taking into consideration that the core elements of the product’s RMP are common and as this
guideline was based on the European Good Pharmacovigilance Practice, thus for simplification;
MAH/Applicants having EU RMP in place submit both of the following:
1. the most updated version of the EU RMP (referenced EU RMP including its annexes);
altogether with
2. the National Display of the RMP (including its annexes).
In these circumstances (submitting the National Display and the EU RMP), the following
conditions apply:
When the referenced EU RMP is subject to update the National Display of RMP should be
updated in accordance.
Minor differences may exist between this guidance and the EU RMP, in this case
MAH/Applicant may be asked by the national medicines authority in the Arab Country
concerned to submit additional information, use different tables, and/or provide
clarification….etc.
The submitted EU RMP shall be the most updated version.
The EU RMP shall be submitted with its annexes and reference materials
Generally, it is required that all the risk management activities applied globally/in the EU to be
applied in the concerned Arab Country as well, especially the risk minimization measures
including the measurement of their effectiveness. Accordingly, all activities, action plans and
details especially the risk minimization ones (including the measurement of their effectiveness)
stated in the submitted EU RMP are expected by default to apply to Arab Country concerned
and the MAH is required to adhere to them, EXCEPT otherwise clearly stated and justified by
the MAH/Applicant in the “National Display of the RMP” and agreed by the national medicines
authority. Please pay attention in filling in the National Display of RMP and do not skip any
activity which was in the reference EU RMP without highlighting whether it will be
implemented or not on the national level according to the tables below. Any unjustifiably
skipped activity will be considered as “apply to national level” and the MAH is required to
adhere to.
Contacts
Local Safety Responsible (LSR) name ……………………………………………………………
LSR signature .....……………………………………………..............……....………
Contact person for this RMP …………………………………………………..........…………
E-mail address or telephone number of contact person ………………………….........………
…………........................................................................................................................................
Posology and route of Current (if applicable) in the Arab Country concerned
administration in the Arab Country
concerned
Current of the reference medicinal product in the EEA
Pharmaceutical form(s) and Current (if applicable) in the Arab Country concerned
strengths
Current of the reference medicinal product in the EEA
Activities as Type of the Safety Action plan in the Action plan in the Highlight Justification
stated in the activity Concern/ referenced EU RMP National Display of differences if any
referenced efficacy the RMP
(even minor
EU RMP issue
difference)
a) If the MAH/Applicant proposes not to implement in Arab Country concerned any of the
activities stated in the EU referenced RMP; this should be clearly highlighted in the above table
and comprehensive justification should be supplied, in addition explanation of how the safety
concern intended by this activity will then be managed in Arab Country concerned.
b) If the MAH/Applicant proposes some differences (even minor ones) in the action plan of
specific activity to be followed in the Arab Country concerned other than those described in the
referenced EU RMP; the differences should be clearly highlighted in the table and
comprehensive justification should be supplied as well.
pharmacovigilance activity); fill in the following table, and protocols should be provided in Annex
6 of this National Display of RMP.
Safety concern
Objective(s) of the risk minimisation measures
Routine risk minimisation measures (Proposed) text in SmPC
<E.g. Dose reduction for ……. in section 4.2 of the
SPC………
Warning in section 4.4 to……
Listed in section 4.8>
Comment (e.g. on any differences between SmPCs)
Other routine risk minimisation measures
<E.g. Prescription only medicine
Use restricted to physicians experienced in the
treatment of…….>
Additional risk minimisation measure(s)1 Objective and justification of why needed.
Proposed actions/components and rationale
Additional risk minimisation measure(s) 2 Objective and justification of why needed.
(repeat as necessary) Proposed actions/components and rationale
Current (or proposed if product is not authorised) local (of the concerned Arab Country) summary
of product characteristics (SmPC) and package leaflet(s) for each product in the RMP.
If multiple versions are included for a product, they should show in which Country(s) they are
applicable. In addition, if available, a core SmPC should be provided with an overview of the
changes applicable to the SmPC in the Arab Country concerned.
*
Draft = not approved
Approved = when agreed by national authority as appropriate
Provide forms
Annex 8 - Protocols for proposed and ongoing studies in National Display of RMP section
III.2.b
*
Draft = not approved
Approved = when agreed by Authority
Annex 9 - Synopsis of newly available study reports in National display Section III.2.a. & b.
Include the study abstract. For non-interventional studies use the abstract format detailed in
Module: VIII Post Authorisation Safety Studies of Good Pharmacovigilance Safety Studies
Mock up examples in English (unless other language is requested by the medicines authority of the
Arab Country concerned) (of the material provided to healthcare professionals and patients. For
those materials directed to patients, in addition to the English version, Arabic translation of the
mock up shall be included as well.
OTHER INFORMATION:
<Other identifying or clarifying information if necessary>
<Date>
<Active substance, name of medicinal product and main message (e.g. introduction of a
warning or a contraindication)>
Further information
<Link/reference to other available relevant information, such as information on the website of a
national medicines authority>
<Therapeutic indication of the medicinal product, if not mentioned above>
Annexes
<Relevant sections of the Product Information that have been revised (with changes made visible)>
<Detailed scientific information, if necessary>
<List of literature references, if applicable>
GVP: Annexes
The following are other guidelines developed by the European Medicines Agency (EMA) some of
them are under their previous EU regulations but remain valid in principle (unless any aspect is not
compatible with this guideline). These guidelines are acknowledged –from scientific aspects- in the
Arab Countries, they may be revised at a later point in time for inclusion in GVP for Arab Countries.
Guidance for the format and content of the final study report of
EMA
non-interventional post-authorisation safety studies
GVP: Annexes
First Last
Document(s)
published updated
First Last
Document(s)
published updated
MedDRA users and ICH-Endorsed guide for MedDRA users on data output
* ICH E2B(R2): Maintenance of the ICH guideline on clinical safety-data management: Data
elements for transmission of individual case safety reports. While the implementation of
ICH-E2B(R3) is being prepared for, ICH-E2B(R2) remains the currently applicable format for
transmission of individual case safety reports.
GVP: Annexes
Annex V- Abbreviations
Abbreviation
AE Adverse event
AR Assessment report
DB Database
EU European Union
Abbreviation
IT Information technology
MA Marketing authorisation
Abbreviation
PL Package leaflet
أســس
الممــارست الجيــذة لليقظــت الذوائيــت
بالــذول الـعربيــت