Pharmacovigilance Medical Writing in Risk Evaluation and Management

4.1 Introduction


The pharmacovigilance documents associated with risk evaluation and management are often required components of Marketing Authorization Application (MAA) and New Drug Application (NDA) submissions, and thus could have been included in Chapter 3 (Pharmacovigilance Medical Writing for Marketing Authorization). However, they have been accorded a stand-alone chapter as they are ‘living’ documents, which are continually updated and amended throughout the medicinal product’s post-marketing life, as and when warranted by the emergence of new safety information.


In the EU, the Risk Management Plan (RMP) is a mandated component of the MAA for most medicinal products. The EU RMP outlines all safety information yet to be established for the medicinal product and describes the processes that will be utilized by the company to acquire this information. Furthermore, the RMP outlines the measures that will be applied by the company to minimize the product’s determined risks and how the effectiveness and success of these endeavors will be determined.


In effect, the EU RMP, when submitted at the time of application for marketing authorization, affords both the sponsor and regulatory authority the opportunity to proactively plan all required pharmacovigilance activities before the medicinal product is actually licensed for use by the public. As new safety data is gathered during the medicinal product’s early post-marketing life, this document is updated, either upon the initiative of the Marketing Authorization Holder (MAH) or at the behest of the regulators, to further fine-tune understanding of the product’s safety profile and resulting impact on the benefit-risk balance. As such, the EU RMP prepared at the time of application for marketing authorization remains a live document that will continue to be updated in the course of the medicinal product’s post-marketing life.


In the US, the Risk Evaluation and Mitigation Strategies (REMS) Report can be requested by the Food and Drug Administration (FDA) as part of the registration or authorization process for any medicinal product considered as requiring additional risk management processes. In effect, the REMS Report serves the same purpose in the US as the EU RMP in the EU. Although the REMS Report came into effect as a pharmacovigilance document in 2007, the FDA can also request a REMS Report for medicinal products authorized prior to 2007, if considered necessary due to new safety data. As an alternative, it is also possible for the sponsor or MAH to proactively prepare and submit a REMS Report without a specific request from the FDA.


Preparation of the EU RMP and REMS Report involves a significant degree of pre-submission discussion and correspondence between the applicant (i.e. MAH or sponsor) and the regulators, which serve to guide the applicant on the regulators’ views regarding acceptable risk minimization measures for the medicinal product.


The Benefit-Risk Evaluation Report, which presents a cumulative assessment of the medicinal products’ benefits (in specific indications) weighed against the associated risks, is developed during the pre-authorization period. As with other pharmacovigilance documents utilized in risk evaluation and management, the Benefit-Risk Evaluation Report continues to be updated and amended throughout the medicinal product’s life cycle, and is required for product authorization, with subsequent updating and amendment usually required at other critical decision-making points, such as registration of new indications, license renewal, and, on occasions, of product withdrawal.


A summary of the key pharmacovigilance documents required for risk evaluation and management is presented below (see Figure 1.1):



  • EU RMP (EU only);
  • REMS Report (US only);
  • Benefit-Risk Evaluation reports.

4.2 The EU Risk Management Plan


4.2.1 Regulatory Guidelines and General Principles


Volume 9A, the Rules Governing Medicinal Products in the European Union [1], provides the legal underpinning for the EU RMP, with guidance, including a template published by the ICH (Harmonised Tripartite Guideline Pharmacovigilance Planning E2E) [2] and the European Medicines Agency [3, 4].


The general principles of the EU RMP are summarized in Table 4.1.


Table 4.1 General principles of the EU Risk Management Plan.





















Principle Description
Scope of Data The EU RMP encompasses numerous data, including clinical, non-clinical, epidemiological, and literature data.
Risk Assessment The initial component of the EU RMP; comprises the Safety Specification (describing important identified risks, potential risks, and missing information) and Pharmacovigilance Plan (describing standard pharmacovigilance processes and measures to further examine safety concerns described in the Safety Specification).
Risk Minimization The second component of the EU RMP; comprises assessment of the need for further pharmacovigilance activities and methods for monitoring the effectiveness of the implemented measures.
Related Safety Documents The safety data presented in the EU RMP should be consistent with that presented in the other related safety documents (e.g. PSUR, DSUR, Investigator’s Brochure, and product labeling).
DSUR = Development Safety Update Report; EU = European Union; PSUR = Periodic Safety Update Report; RMP = Risk Management Plan

4.2.2 When are EU Risk Management Plans Prepared?


EU RMPs can be prepared and submitted at any time during the medicinal product’s pre- and post-marketing life cycle. EU RMPs are prepared or updated on the following occasions:



  • at the time of the initial MAA (i.e. at the time of product registration/authorization);
  • application for registration of new indications or changes to approved use (e.g. new dose, new route of administration, or a modified manufacturing process for biotechnology products);
  • authorization of treatment for a special treatment population (e.g. paediatrics and the elderly);
  • identification of a new safety concern;
  • upon request from the relevant competent authority;
  • within 60 days of important pharmacovigilance or risk minimization activity milestones being achieved or availability of new data from studies.

4.2.3 Data Sources for the EU Risk Management Plan


A summary of the data required for preparation of EU RMPs and the company departments usually responsible for the provision of these data are presented in Table 4.2. As noted with other documents, the precise designations of the departments/functions may vary from company to company.


Table 4.2 Source data for the EU Risk Management Plan.



























EU RMP Data Data Source
Non-clinical Data Non-clinical R&D; data relating to all pertinent safety issues not yet resolved in the clinical setting (e.g. toxicity, drug interactions, and pharmacology related complications).
Clinical Data
   – Clinical trials
   – Marketplace
Clinical Operations; patient exposure data in studies
Drug Safety (Pharmacovigilance) or Sales and Marketing; post-marketing exposure data.
Regulatory Actions Taken Regulatory Affairs
Adverse Events/Adverse Experience Drug Safety (Pharmacovigilance)In addition:
   – Non-clinical R&D; identified and potential drug interactions.
   – Clinical and Non-clinical R&D; epidemiology of indications and important adverse events.
Pharmacovigilance Plan Drug Safety (Pharmacovigilance)
Literature Medical Information/Scientific Information Services
EU = European Union; R&D = Research and Development

4.2.4 Review of the EU Risk Management Plan


As with all other pharmacovigilance documents, the preparation of the EU RMP involves contributions from a multidisciplinary team, which is tasked with participation in document planning, provision of data from their respective departments, and review and approval of the RMP. A summary of the team that should be involved in preparation of the EU RMP is presented in Table 4.3.


Table 4.3 The EU Risk Management Plan review team.



























Reviewer Key Areas of Responsibility/Sections to Review
Drug Safety Physician The whole EU RMP
EU Qualified Person/Director of Safety The whole EU RMP
Medical Affairs The whole EU RMP
Clinical and Non-clinical R&D Clinical: the whole EU RMP
Non-clinical: Section 1: Safety Specification
Regulatory Affairs Section 1.4.2: Regulatory Actions Taken
Quality Control The whole EU RMP
EU = European Union; R&D = Research and Development; RMP = Risk Management Plan

4.2.5 A Timeline – Planning for the EU Risk Management Plan


As a general rule, preparation of an EU RMP can be divided into the following four key activities:



  • RMP planning and collation of source data;
  • writing of the draft EU RMP;
  • review of the EU RMP;
  • QC, finalization, and approval of the EU RMP.

An example timeline appropriate for preparation of the EU RMP is presented in Figure 4.1.



Figure 4.1 Example timeline for EU Risk Management Plan preparation

img

4.2.6 Generic Model of the EU Risk Management Plan


A generic model EU RMP template, based on guidelines and format recommended by the European Medicines Agency [3] is presented in this section.







Risk Management Plan

[Generic Product Name]

Report Date:

[MAH’s Name and Address]

[MAH’s confidentiality statement]

Table of Contents












































































Abbreviations
Product Information
1 Safety Specification
1.1 Non-clinical
1.2 Clinical
1.2.1 Limitations of the Human Safety Database
1.3 Populations not Studied in the Pre-authorization Phase
1.4 Post-authorization Experience
1.4.1 Post-authorization Usage Data
1.4.2 Regulatory Actions Taken
1.5 Adverse Events/Adverse Reactions
1.5.1 Newly Identified Safety Concerns since the Last Submitted EU RMP
1.5.2 Important Identified and Potential Risks (including Newly Identified Risks)
1.6 Identified and Potential Interactions with other Medicinal Products, Food, and other Substances
1.7 Epidemiology of the Indications and Important Adverse Events
1.8 Pharmacological Class Effects
1.9 Additional EU Requirements
1.9.1 Potential for Overdose
1.9.2 Potential for Transmission of Infectious Agents
1.9.3 Potential for Misuse for Illegal Purposes
1.9.4 Potential for Off-Label Use
1.9.5 Potential for Off-Label Paediatric Use
1.10 Summary – Ongoing Safety Concerns
2 Pharmacovigilance Plan
2.1 Routine Pharmacovigilance Practices
2.2 Summary of Safety Concern and Planned Pharmacovigilance Actions
2.3 Detailed Action Plan for Specific Safety Concerns
2.4 Overview of Study Protocols for the Pharmacovigilance Plan
2.5 For Updates to the EU RMP
2.6 Summary of Outstanding Actions, including Milestone
3 Evaluation of the Need for Risk Minimization Activities
3.1 Summary Table of Planned Actions
3.2 Potential for Medication Errors
4 Risk Minimization Plan
5 Summary of EU Risk Management Plan
6 Contact Person Details
Annexes

Abbreviations

Insert standard abbreviations and definitions table as follows.



























































Abbreviation Definition
ATC Anatomical Therapeutic Chemical
CCSI Company Core Safety Information
EEA European Economic Area
EU European Union
EUQPPV EU Qualified Person for Pharmacoviglance
Professional
INN International non-proprietary name
MAA Marketing Authorization Application
MAH Marketing Authorization Holder
MedDRA Medical Dictionary for Regulatory Activities
PD Pharmacodynamic
PK Pharmacokinetic
PSUR Periodic Safety Update Report
PT Preferred term
RMP Risk Management Plan
SmPC Summary of Product Characteristics




Note: The table needs to be expanded and completed as required.

Product Information

This section functions as an introduction to the report and presents a tabulated summary of information relating to the medicinal product, as illustrated in the example below.
































Invented name of the medicinal product (product short name) Add details of product brand name.
Active substance(s) (INN or common name): Add details of the product generic name.
Pharmaco-therapeutic group (ATC Code): Add products ATC code.
Medicinal product code (From Eudra Vigilance) Add details.
Authorization procedure(s) (central, mutual recognition, decentralized, national) Add details of the product’s registration procedure in the EU.
Name of Marketing Authorization Holder or Applicant Add company details.
Date and country of first authorization worldwide Complete as appropriate.
Date and country of first launch worldwide Complete as appropriate.
Date and country of first authorization in the EEA Complete as appropriate – if different from above.
Date and country of first launch in the EEA Complete as appropriate – if different from above.








Data lock point for EU RMP Add date of data cut-off.
Version Add version number (i.e. version 1.0 for the first final version, and versions 2.0, 3.0, etc. on subsequent updates).
















Brief description of product (chemical class, mode of action, etc.) This section should include a brief description of the product, including the active ingredient, therapeutic class, and mechanism of action/pharmacology.
Indication(s) Add details of the proposed indications if this is the first RMP prepared as part of the MAA dossier, or, details of registered indications if this is an update of an existing RMP for marketed products.
Dosage This section should present details of the proposed (or registered) doses for the medicinal product. If the product is proposed (or approved) for more than one indication, this section should be separated into subsections according to indication and dosage details presented for each indication.
Pharmaceutical form(s) and strength(s) The physical form in which the medicinal product is presented for use, and the amount of active substance in each unit is described in this section.
ATC = Anatomical Therapeutic Chemical; EEA = European Economic Area; EU = European Union; INN = International non-proprietary name; MAA = Marketing Authorization Application; RMP = Risk Management Plan

1 Safety Specification

The Safety Specification functions as a summary of the medicinal product’s safety profile, and describes all significant:


  • identified risks;
  • potential risks;
  • unknown or missing information.

Review of the medicinal product’s safety profile in the Safety Specification is undertaken in the context of the intended treatment population(s), and highlights any unresolved safety issues that require additional research and analysis during the post-marketing phase. This allows for the gradual fine tuning of the product’s benefit-risk profile.

The information set out in the Safety Specification is of critical importance in that it determines (for both the sponsor or MAH and the regulators) additional safety data requirements and monitoring activities, and therefore feeds into the sponsor/MAH obligations that should be outlined in the Pharmacovigilance Plan section of the EU RMP.

Furthermore, the information presented in the Safety Specification also links into subsequent sections of the EU RMP by serving as the premise for determining the need for risk minimization activities and the risk minimization plan (if applicable).

The Safety Specification is structured according to the subsections presented below. However, the PV Medical Writer should note that European regulators regard these subsections as guidance, and encourage modifications to the format to allow for the inclusion of additional subsections, as necessitated by the given medicinal product and clinical program. Still further, the regulators also note that marketed safety products with newly identified safety concerns should not require the exhaustive list of subsections, and the sponsor can use their discretion to select redundant sections for exclusion from the report.

1.1 Non-clinical

The non-clinical section of the Safety Specification presents a summary of all non-clinical safety issues that have not been fully elucidated and resolved by data from the clinical program. Examples of such issues include:


  • toxicity (e.g. repeat-dose toxicity, reproductive/development toxicity, nephrotoxicity, hepatotoxicity, genotoxicity, and carcinogenicity);
  • general pharmacology (e.g. cardiovascular, nervous system);
  • drug interactions.

The PV Medical Writer should endeavor to explain how the identified non-clinical issues relate to use of the medicinal product in the clinical setting; the EU RMP template suggests use of tabulated format, as illustrated in the table below.











Safety Concern from Non-clinical Studies Relevance to Human Usage
Repeat-Dose Toxicity Complete as appropriate.
Reproductive Toxicity Complete as appropriate.

If the medicinal product is proposed for use in special patient populations (e.g. paediatrics), the non-clinical section should be presented as two subsections, the first outlining non-clinical safety concerns yet to be resolved with clinical data, and the second dedicated to showing how the identified non-clinical findings relate to this patient population, and if there is non-clinical data pertinent to this group.

1.2 Clinical

1.2.1 Limitations of the Human Safety Database

This section of the Safety Specification presents a summary of patient exposure to the medicinal product and can be separated into three subsections to show:


  • clinical trial exposure;
  • epidemiological study exposure;
  • post-marketing exposure.

Presentation of clinical trial exposure should be presented according to the treatment indication and tabulated to show exposure according to type of study, patient category, and other study variables:


  • type of study (e.g. blinded randomized studies, open studies, observational studies, overall exposure);
  • age;
  • gender;
  • doses administered;
  • duration of treatment.

Presentation of patient exposure to the medicinal product is followed by an assessment of the limitations of the safety database, which could be due to the size of the database or the conditions under which the clinical studies were performed (e.g. exclusion of particular patient groups), and how these limitations impact on the assessment of the product’s genuine safety profile and safe use in the post-marketing setting.

If the size of the human safety database is limited, the resulting impact on detecting adverse reactions reported at low frequencies should be discussed, and the duration of exposure should be explored with respect to assessing the safety profile of medicinal products intended for long-term use.

The PV Medical Writer is referred to Section 5.2.6 of Chapter 4 (Pharmacovigilance Medical Writing for Marketed Products) for guidance on the presentation of post-marketing data.

1.3 Populations not Studied in the Pre-authorization Phase

This part of the Safety Specification presents a summary of all patient populations excluded from the clinical program, or those that were only assessed to a limited level during the pre-marketing phase. These patient populations may include:


  • children;
  • elderly patients (usually defined as aged ≥65 years);
  • pregnant or breast-feeding women;
  • patients with organ dysfunction (e.g. hepatic or renal impairment);
  • patient with disease severity different from the clinical study population;
  • patient populations with relevant genetic polymorphism;
  • patient populations from varying ethnic/racial origins.

The ramifications of and limitations in assessing the safe use of the product in any excluded patient population should be discussed. For medicinal products, where paediatric development was restricted to particular age groups, the deliberations should include ramifications for other paediatric groups.

1.4 Post-authorization Experience

1.4.1 Post-authorization Usage Data

For subsequent updates of the EU RMP, review of post-marketing data should be used to update the Safety Specification anddescribe how the actual use of the marketed product, including use for unapproved indications, correlates with the information in the Summary of Product Characteristics (SmPC; including indications and contraindications for use).

Subsequent updates should also detail any recently identified safety concerns, particularly those that may be associated with use of the product in patient populations not assessed during the clinical development program.

1.4.2 Regulatory Actions Taken

This section of the Safety Specification presents a summary of any actions taken by regulatory authorities for reasons of safety.

1.5 Adverse Events/Adverse Experience

This section of the Safety Specification presents a summary of all significant identified and potential risks that need further elucidation, and is divided into two subsections, one showing safety concerns identified since the last EU RMP (for updated RMPs) and another summarizing all significant identified and potential safety issues.

1.5.1 Newly Identified Safety Concerns since the Last Submitted EU RMP

The template for the EU RMP recommends a tabulated summary of all newly identified safety issues since the last RMP, as illustrated in Table 1.

Table 1 Summary of newly identified safety concerns for Product X since the last RMP


























Safety Concern #1
Details: Add summary of safety concern.
Source: Add summary of data used to identify the safety issue.
Implications for product literature Add a summary of modifications proposed for product literature (e.g. SmPC, CCSI, and PI) as a result of the newly identified safety issue.
New studies proposed in the Pharmacovigilance Plan Yes/No
New risk minimization actions proposed Yes/No
Safety Concern #2
Expand table as required
CCSI = Company Core Safety Information; PI = Package Insert; SmPC = Summary of Product Characteristics

1.5.2 Important Identified and Potential Risks (including Newly Identified Risks)

The template for the EU RMP recommends a tabulated summary of all identified safety issues, as illustrated in the Table 2.

Table 2 Summary of Important Identified and Potential Risks for Product X






































Identified Risk of <x> Add MedDRA PT term
Seriousness/outcome Complete as appropriate (e.g., the proportion of patients with outcomes recorded as: fatal, recovered with or without sequelae, not recovered, hospitalized).
Severity and nature of risk Include a tabulated summary of severities (e.g., % of mild, moderate or severe).
Frequency with 95% CI Present the relative incidence as well as incidence compared to placebo/comparator treatments.
Background incidence/prevalence Present the background incidence and the general prevalence in the target population(s).
Risk groups or risk factors Describe use, including doses and patient susceptibility or other risk factors.
Potential mechanisms Describe mechanisms.
Preventability Present available data on the potential to predict and prevent the risk(s).
Potential public health impact of safety concern If feasible include the number of patients expected to be affected, hospitalizations, fatal outcomes etc.
Evidence source Present data sources by cross-referencing to CTD modules or specific studies and other reports (e.g., PSURs).
Regulatory action taken Specify the country and type of action taken.
CTD = Common Technical Documentation; MedDRA = Medical Dictionary for Regulatory Activities; PSUR = Periodic Safety Update Report; PT = preferred term,

1.6 Identified and Potential Interactions with other Medicinal Products, Food, and other Substances
< div class='tao-gold-member'>

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 23, 2016 | Posted by in PHARMACY | Comments Off on Pharmacovigilance Medical Writing in Risk Evaluation and Management

Full access? Get Clinical Tree

Get Clinical Tree app for offline access