Ansh File Final
Ansh File Final
Ansh File Final
Submitted by Submitted to
This is to certify that the work presented in the project entitled “ICH Guidelines for
Pharmacovigilance” has been carried out by me for the partial fulfillment of degree of
‘Bachelor of Pharmacy’ under the supervision of Mrs. Pinki Assistant Professor at Lotus
Institute of Pharmacy, Bareilly.
The Project embodies results of the original work and studies carried out by me and the contents
of the project have not formed the basis for the award of any degree/ diploma by any other
University or Institution.
I also confirm that all the material which I have borrowed from other sources and incorporated in
this report is duly acknowledged.
Place: Bareilly
Napees Ahmad
Date: / / 20095705000060
CERTIFICATE
I hereby, declare that this project work “ICH Guidelines for Pharmacovigilance” embodies the
results of the original project work carried out by me. The extent source of information is derived
from the various case studies, existing literature and has indicated throughout the project at
appropriate places. This Project is carried out under the supervision of Mrs. Pinki, Assistant
Professor at Lotus Institute of Pharmacy, Bareilly.
titled “ICH Guidelines for Pharmacovigilance." The conducive academic atmosphere at Lotus
Institute of Pharmacy, Bareilly, greatly facilitated the execution of this project. I extend my
sincere thanks to Dr. Abhishek Agarwal, Principal, for his unwavering support and guidance
throughout the project duration, offering invaluable insights into various aspects and solutions
related to the topic. Special appreciation goes to my mentor, Mrs. Pinki, whose dedicated
assistance and expert advice played a pivotal role in shaping this project. Her profound
Napees Ahmad
2009570500060
Tables of Contents
3. History of ICH 4
4. Mission 6
5. ICH Guidelines 8
6. ICH Guidelines for Pharmacovigilance 8
Introduction
Pharmacovigilance is a branch of healthcare that focuses on the detection, assessment,
understanding, and prevention of adverse effects or any other drug-related problems associated
with pharmaceutical products. It plays a crucial role in ensuring drug safety and protecting public
health.
Objectives of Pharmacovigilance :
3.Risk Assessment and Evaluation: Pharmacovigilance experts assess drug risks and evaluate
the benefits versus the potential harms. This information is crucial for healthcare professionals
and regulatory authorities in making informed decisions about the use of specific drugs.
The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for
Human Use (ICH) is unique in bringing together the regulatory authorities and pharmaceutical
industry to discuss scientific and technical aspects of pharmaceuticals and develop ICH
guidelines
The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for
Human Use (ICH) was established in 1990 as a collaborative initiative between regulatory
authorities and the pharmaceutical industry from Europe, Japan, and the United States. The
primary objective of ICH is to facilitate the development and registration of new pharmaceutical
products by harmonizing technical requirements for drug approval across regions, thereby
reducing duplication of efforts and minimizing barriers to global drug development.
The history and evolution of ICH guidelines can be traced back to the recognition of the need for
international harmonization in response to divergent regulatory requirements and practices
among different regulatory authorities. The initial focus of ICH was on developing guidelines
related to quality, safety, and efficacy of pharmaceutical products. Over the years, ICH has
expanded its scope to cover various aspects of drug development, including pharmacovigilance,
clinical trials, pharmaceutical quality systems, and regulatory communication.
The International Council for Harmonisation (ICH), formerly the International Conference on
Harmonisation (ICH) held the inaugural Assembly meetings on 23 October 2015 establishing
ICH as an international association, a legal entity under Swiss law.
This step built upon a 25-year track record of successful delivery of harmonised guidelines for
global pharmaceutical development as well as their regulation, and a longer standing recognition
of the need to harmonise.
The realisation that it was important to have an independent evaluation of medicinal products
before they are allowed on the market was reached at different times in different regions.
However in many cases the realisation was driven by tragedies, such as that with thalidomide in
Europe in the 1960s.
For most countries, whether or not they had initiated product registration controls earlier, the
1960s and 1970s saw a rapid increase in laws, regulations and guidelines for reporting and
evaluating the data on safety, quality and efficacy of new medicinal products. The industry, at
the time, was becoming more international and seeking new global markets; however the
divergence in technical requirements from country to country was such that industry found it
necessary to duplicate many time-consuming and expensive test procedures, in order to market
new products, internationally.
The urgent need to rationalise and harmonise regulation was impelled by concerns over rising
costs of health care, escalation of the cost of R&D and the need to meet the public expectation
that there should be a minimum of delay in making safe and efficacious new treatments available
to patients in need.
Initiation of ICH
Harmonisation of regulatory requirements was pioneered by the EC, Europe, in the 1980s, as the
EC, Europe moved towards the development of a single market for pharmaceuticals. The success
achieved in Europe demonstrated that harmonisation was feasible. At the same time there were
The birth of ICH took place at a meeting in April 1990, hosted by EFPIA in Brussels.
Representatives of the regulatory agencies and industry associations of Europe, Japan and the US
met, primarily, to plan an International Conference but the meeting also discussed the wider
implications and terms of reference of ICH.
At the first ICH Steering Committee meeting of ICH the Terms of Reference were agreed and it
was decided that the Topics selected for harmonisation would be divided into Safety, Quality and
Efficacy to reflect the three criteria which are the basis for approving and authorising new
medicinal products.
Since ICH's inception in 1990, the ICH process has gradually evolved. ICH's first decade saw
significant progress in the development of ICH Guidelines on Safety, Quality and Efficacy
topics. Work was also undertaken on a number of important multidisciplinary topics, which
included MedDRA (Medical Dictionary for Regulatory Activities) and the CTD (Common
Technical Document). As ICH started into a new millennium, the need to expand communication
and dissemination of information on ICH Guidelines with non-ICH regions became a key focus.
Attention was also directed throughout the second decade towards facilitating the
implementation of ICH Guidelines in ICH's own regions and maintaining already existing ICH
Guidelines as science and technology continued to evolve.
Now in its fourth decade of activity, ICH's attention is directed towards extending the benefits o f
harmonisation beyond the founding ICH regions. A significant step was taken in 2015 to
facilitate this which saw ICH undergoing a series of organisational changes. These changes
constituted a number of reforms including: increasing international outreach; changing ICH’s
governance structure; disseminating more information on ICH processes to a wider number of
The resulting ICH association establishes an Assembly as the over-arching governing body with
the aim of focusing global pharmaceutical regulatory harmonisation work in one venue that
allows pharmaceutical regulatory authorities and notably concerned industry organisations to be
more actively involved in ICH’s harmonisation work.
Mission
The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for
Human Use (ICH) is unique in bringing together the regulatory authorities and pharmaceutical
industry to discuss scientific and technical aspects of drug registration. Since its inception in
1990, ICH has gradually evolved, to respond to the increasingly global face of drug
development. ICH's mission is to achieve greater harmonisation worldwide to ensure that safe,
effective, and high quality medicines are developed and registered in the most resource-efficient
manner. Harmonisation is achieved through the development of ICH Guidelines via a process of
scientific consensus with regulatory and industry experts working side-by-side. Key to the
success of this process is the commitment of the ICH regulators to implement the final
Guidelines.
With ICH’s establishment as an international non-profit Association under Swiss law on October
23, 2015, ICH’s mission has been embodied in its Articles of Association as follows:
Adverse Drug Reaction (ADR): Any harmful or unintended response to a medicinal product
that occurs at doses normally used in humans for the prophylaxis, diagnosis, or treatment of
disease, or for modifying physiological function.
Signal Detection: The process of identifying new or changing safety concerns associated with
pharmaceutical products based on analysis of pharmacovigilance data.
Risk Management: The systematic process of identifying, assessing, and mitigating risks
associated with pharmaceutical products to optimize their benefit-risk profile.
Periodic Safety Update Report (PSUR): A comprehensive document that summarizes the
safety profile of a pharmaceutical product based on aggregated safety data from various sources
and provides an evaluation of its benefit-risk balance.
- Collection, Recording, and Processing of Adve rse Drug Reactions (ADRs): Pharmaceutica l
companies are required to establish systems for the collection, recording, and processing of ADR
reports from healthcare professionals, patients, and other sources. This includes data entry,
coding, and assessment of ADR reports to determine causality and seriousness.
- Periodic Safety Update Reports (PSURs): Pharmaceutical companies are required to submit
PSURs to regulatory authorities at predefined intervals (e.g., every six months or annually) to
provide a comprehensive overview of the safety profile of their products. PSURs include an
evaluation of safety data, analysis of cumulative safety information, and an assessment of the
benefit-risk balance, along with proposed risk management strategies.
ICH E2B - Clinical Safety Data Management: Definitions and Standards for
Expedited Reporting
Expedited reporting of serious and unexpected adverse drug reactions (ADRs) during clinical
trials is crucial for ensuring the safety of study participants and maintaining the integrity of
clinical trial data. Serious and unexpected ADRs are those that are life-threatening, result in
death, require hospitalization or prolongation of existing hospitalization, result in persistent or
significant disability/incapacity, or constitute a congenital anomaly/birth defect.
Timely reporting of such ADRs allows regulatory authorities, sponsors, and investigators to
take appropriate actions to protect the safety and well-being of trial participants, including
implementing risk mitigation measures, modifying study protocols, or terminating the trial if
necessary. Expedited reporting also facilitates the accumulation of safety data for ongoing safety
monitoring and assessment throughout the clinical trial process.
ICH E2B provides standardized guidelines for the format and content of expedited safety
reports submitted during clinical trials. These reports are typically submitted electronically to
regulatory authorities using the International Conference on Harmonisation (ICH) Electronic
Common Technical Document (eCTD) format. Key elements of expedited safety reports include:
- Adverse Event Information: Description of the adverse event, including onset date, severity,
outcome, and relationship to the investigational product.
- Investigator Assessment: Assessment of the causality and seriousness of the adverse event by
the investigator.
- Action Taken: Actions taken in response to the adverse event, such as treatment modifications
or study protocol amendments.
- Follow-Up Information: Follow-up data on the adverse event, including resolution status and
any subsequent outcomes.
Standardized coding systems, such as the Medical Dictionary for Regulatory Activities
(MedDRA), are used to classify adverse events and ensure consistency in reporting across
different studies and regions.
ICH E2B defines specific timelines and procedures for the submission of expedited safety
reports to regulatory authorities during clinical trials. The reporting requirements vary depending
on the seriousness and expectedness of the adverse event:
- Immediate Reporting: Serious and unexpected adverse events that result in death or are life-
threatening should be reported immediately, typically within 7 calendar days of the sponsor's
awareness of the event.
- Expedited Reporting: Other serious and unexpected adverse events should be reported
expeditedly, usually within 15 calendar days of the sponsor's awareness of the event.
Sponsors are responsible for ensuring compliance with these reporting timelines and
procedures, coordinating with investigators and regulatory authorities, and maintaining accurate
and complete documentation of all expedited safety reports throughout the duration of the
clinical trial.
Overall, expedited reporting of serious and unexpected ADRs during clinical trials is essential
for protecting the safety of trial participants, maintaining data integrity, and facilitating
regulatory oversight. Compliance with standardized format and content requirements, as outlined
in ICH E2B, ensures consistency and transparency in the reporting process, enabling timely
assessment and action to address safety concerns.
1. Concept of a Pharmacovigilance Plan and its Role in Proactive Drug Safety Monitoring:
a. Safety Specification:
- Risk Identification and Prioritization: The plan outlines methodologies and criteria for
identifying and prioritizing potential safety signals, including adverse drug reactions (ADRs),
product quality issues, medication errors, and other drug-related problems. Risk prioritization
ensures that resources are allocated effectively to address the most significant safety concerns.
- Risk Assessment and Evaluation: The pharmacovigilance plan describes procedures for
assessing and evaluating the significance of identified risks, including their potential impact on
patient safety, public health, and regulatory compliance. This involves analyzing quantitative and
qualitative data, conducting benefit-risk assessments, and considering factors such as the
severity, frequency, and preventability of adverse events.
- Risk Minimization Strategies: The plan outlines risk minimization strategies and
interventions aimed at reducing the likelihood or severity of identified risks while preserving the
therapeutic benefits of the medicinal product. This may include modifications to product
labeling, implementation of risk communication initiatives, development of risk management
plans (RMPs), and post-marketing studies to further characterize risks.
- Data Collection and Analysis: The pharmacovigilance plan describes methodologies and
procedures for the collection, recording, and analysis of safety data from various sources,
including spontaneous reports, clinical trials, literature reviews, and post-marketing surveillance
- Signal Detection and Management: The plan outlines methodologies for detecting,
evaluating, and managing safety signals, including statistical methods, data mining techniques,
and signal validation processes. This involves establishing signal detection thresholds,
conducting signal validation assessments, and implementing risk management measures in
response to confirmed or emerging safety signals.
- Communication and Reporting: The plan defines procedures for communicating safety
information to regulatory authorities, healthcare professionals, patients, and other stakeholders,
including requirements for expedited reporting of serious and unexpected ADRs. This involves
preparing periodic safety update reports (PSURs), safety communications, and regulatory
submissions in compliance with applicable regulatory requirements.
The pharmacovigilance plan is a dynamic document that is regularly updated and revised
throughout the lifecycle of the medicinal product to reflect evolving safety data, regulatory
requirements, and risk management strategies. By implementing a robust pharmacovigilance
plan, pharmaceutical companies can proactively monitor the safety of their products, mitigate
potential risks, and ensure the ongoing protection of patient health and public safety.
The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for
Human Use (ICH) plays a pivotal role in harmonizing regulatory standards and guidelines for
drug development and registration globally. Pharmaceutical companies adhere to ICH guidelines
to ensure consistency and quality in the development, registration, and post-marketing
surveillance of medicinal products. Implementing ICH guidelines presents various challenges
and considerations, particularly in different regions with diverse regulatory frameworks, cultural
norms, and healthcare infrastructures. This paper explores the challenges and considerations for
implementing ICH guidelines, strategies for developing effective pharmacovigilance systems,
practical examples of ICH guideline application in drug development, and the role of technology
in facilitating compliance with ICH requirements.
Implementing ICH guidelines in different regions poses several challenges and considerations,
including:
2.Cultural and Linguistic Differences: Cultural and linguistic differences may influence
pharmacovigilance practices, reporting behaviors, and communication strategies. Effective
implementation of ICH guidelines requires sensitivity to cultural norms, language barriers, and
healthcare practices to promote accurate and timely reporting of adverse events and ensure
effective communication among stakeholders.
Pharmaceutical companies can develop and maintain effective pharmacovigilance systems that
comply with ICH requirements by:
2.Training and Education: Providing ongoing training and education to staff involved in
pharmacovigilance ensures awareness of ICH requirements, regulatory obligations, and best
practices. Training programs should cover topics such as ADR reporting, data collection, signal
detection methods, and regulatory submissions.
4.Conducting Regular Audits and Quality Assurance Checks: Performing regular audits and
quality assurance checks of pharmacovigilance processes and systems ensures adherence to ICH
guidelines, identifies areas for improvement, and mitigates compliance risks. Audits should
assess the completeness, accuracy, and timeliness of adverse event reporting, data management
practices, and regulatory submissions.
ICH guidelines are applied throughout the drug development process to ensure the safety,
efficacy, and quality of medicinal products. Practical examples of ICH guideline application
include:
1.Clinical Trial Design and Conduct: ICH E6 (Good Clinical Practice) provides standards for
the design, conduct, monitoring, and reporting of clinical trials. Pharmaceutical companies
follow ICH E6 guidelines to ensure the ethical treatment of human subjects, maintain data
integrity, and generate reliable clinical trial results.
3.Risk Management: ICH E2E (Pharmacovigilance Planning) and ICH E2D (Post-Approval
Safety Data Management) provide guidance on risk management planning and implementation.
Pharmaceutical companies develop risk management plans (RMPs) in accordance with ICH
guidelines to identify, characterize, and mitigate risks associated with their products, including
the implementation of risk minimization measures and post-marketing surveillance activities.
Technology plays a critical role in facilitating the efficient implementation of ICH guidelines by
enabling:
1. Data Capture and Management: Advanced pharmacovigilance databases and EDC systems
streamline data capture, management, and analysis processes, improving the accuracy,
completeness, and timeliness of pharmacovigilance data.
The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for
Human Use (ICH) plays a crucial role in harmonizing regulatory standards and guidelines for
drug development and registration worldwide. Since its inception, ICH has made significant
contributions to enhancing drug safety and promoting public health through the development and
implementation of consensus-based guidelines. This paper analyzes the positive impact of ICH
guidelines on improving drug safety worldwide, with a focus on increased reporting of adverse
drug reactions (ADRs), early detection of potential safety risks, improved communication and
collaboration between stakeholders, and more informed regulatory decision-making.
One of the key benefits of ICH guidelines is the promotion of increased reporting of ADRs,
leading to enhanced pharmacovigilance and patient safety. By standardizing criteria for
identifying, assessing, and reporting ADRs, ICH guidelines provide clear guidance to healthcare
professionals, pharmaceutical companies, and regulatory authorities on their roles and
responsibilities in detecting and reporting adverse events associated with medicinal products. As
a result, there has been a significant improvement in the quantity and quality of ADR reports
submitted to regulatory agencies worldwide.
Another significant impact of ICH guidelines is the early detection of potential safety risks
associated with medicinal products. By establishing standardized methods and criteria for signal
detection, risk assessment, and risk management, ICH guidelines enable pharmaceutical
companies and regulatory authorities to proactively monitor the safety of drugs throughout their
lifecycle. Through regular safety data reviews, signal detection activities, and risk assessment
exercises, stakeholders can identify and evaluate potential safety signals promptly, even before
widespread use of the drug in the market.
ICH guidelines have also contributed to improved communication and collaboration between
stakeholders involved in drug development, pharmacovigilance, and regulatory oversight. By
establishing common standards and terminology for safety reporting, data exchange, and
regulatory submissions, ICH guidelines facilitate seamless communication and information
Finally, ICH guidelines have contributed to more informed regulatory decision- making by
providing evidence-based recommendations and criteria for evaluating the safety, efficacy, and
quality of medicinal products. Regulatory authorities rely on ICH guidelines to assess the
benefit-risk profile of drugs, make regulatory decisions, and communicate safety information to
healthcare professionals and patients. By standardizing regulatory requirements and assessment
criteria, ICH guidelines promote consistency and predictability in regulatory processes, enabling
faster and more transparent regulatory reviews and approvals.
ICH guidelines facilitate regulatory decision- making by providing guidance on safety data
requirements, study designs, statistical analyses, and risk assessment methodologies. Regulatory
authorities use this guidance to evaluate the safety profile of drugs based on clinical trial data,
post-marketing surveillance data, and other relevant information. In cases where safety concerns
arise, regulatory authorities can take appropriate regulatory actions, such as labeling changes,
product recalls, or market withdrawals, to protect public health and ensure patient safety.
Throughout our discussion, we have highlighted the importance of ICH guidelines in several key
areas. Firstly, these guidelines enhance the reporting of adverse drug reactions (ADRs) by
providing clear criteria and procedures for identifying, assessing, and reporting safety concerns
associated with medicinal products. By standardizing reporting practices, ICH guidelines
facilitate the timely collection and analysis of safety data, enabling regulatory authorities to
detect and evaluate potential risks early on.
Secondly, ICH guidelines promote proactive risk management by outlining strategies for
identifying, assessing, and mitigating risks associated with medicinal products. By incorporating
risk management plans (RMPs) into pharmacovigilance activities, pharmaceutical companies can
systematically monitor the safety profile of their products and implement appropriate risk
mitigation measures to protect patient health.
Looking ahead, future directions for pharmacovigilance will likely involve further advancements
in technology, data analytics, and regulatory science. The emergence of big data analytics,
artificial intelligence, and real- world evidence is poised to revolutionize pharmacovigilance
practices, enabling more efficient and proactive identification of safety signals and trends.
Additionally, ongoing efforts to enhance international collaboration and harmonization of
In terms of potential revisions to existing ICH guidelines, continuous review and adaptation are
necessary to keep pace with evolving scientific knowledge, technological advancements, and
regulatory requirements. As new safety challenges emerge and pharmacovigilance practices
evolve, ICH guidelines may need to be revised or expanded to address emerging issues,
incorporate new methodologies, and promote innovation in drug safety monitoring.
In conclusion, ICH guidelines play a pivotal role in shaping pharmacovigilance practices and
ensuring the safety and efficacy of medicinal products worldwide. By providing a standardized
framework for pharmacovigilance activities, promoting collaboration among stakeholders, and
facilitating regulatory oversight, ICH guidelines contribute to the advancement of public health
and the well-being of patients around the globe. As we continue to navigate the complexities of
drug safety in an ever-changing landscape, adherence to ICH guidelines will remain essential for
promoting patient safety and optimizing medication outcomes.
definitions/
19. European Medicines Agency. (2020). Guideline on good pharmacovigilance practices
(GVP) Module VI - Management and reporting of adverse reactions to medicinal
products. Retrieved from https://www.ema.europa.eu/en/documents/scientific-
guideline/guideline-good-pharmacovigilance-practices-gvp- module-vi- management-
reporting-adverse-reactions_en.pdf