View Article

Abstract

Pharmacovigilance is the science dedicated to detecting, assessing, understanding, and preventing adverse drug reactions (ADRs) and other drug-related problems, which has become an essential part of ensuring drug safety and public health. Over the decades, pharmacovigilance has evolved from a predominantly reactive system focused on post-marketing surveillance to a more comprehensive, proactive approach that engages multiple stakeholders, including regulatory authorities, healthcare professionals, pharmaceutical companies, and patients. This review explores the historical evolution of pharmacovigilance, highlighting key milestones such as the establishment of pivotal regulatory bodies like the U.S. FDA, European Medicines Agency (EMA), and the UK's MHRA, which laid the foundation for modern drug safety practices. Programs like MedWatch, EudraVigilance, and the Yellow Card Scheme were developed to support the surveillance of drug safety after market approval, enabling early detection of ADRs and the implementation of corrective actions. The review further examines the systems that have shaped pharmacovigilance today, emphasizing the shift from spontaneous reporting systems to more advanced methods like signal detection, risk management, and global data sharing. Moreover, the involvement of multiple stakeholders in pharmacovigilance processes is explored, with an emphasis on the role of pharmaceutical companies in maintaining safety profiles, healthcare providers in identifying and reporting ADRs, and patients in directly contributing to the safety reporting process. The integration of advanced technologies such as artificial intelligence and big data analytics is also discussed as key drivers in the evolution of pharmacovigilance, facilitating more efficient safety monitoring. This review underscores the significance of an integrated, collaborative, and transparent pharmacovigilance system that continues to evolve in response to the growing complexity of drug therapies and healthcare systems globally.

Keywords

Pharmacovigilance, Adverse Drug Reactions (ADRs), Drug Safety, PostMarketing Surveillance, Regulatory Authorities.

Introduction

Pharmacovigilance plays a pivotal role in ensuring drug safety and improving public health by monitoring, detecting, assessing, and preventing adverse drug reactions (ADRs) and other drugrelated issues. Since its inception, the system of pharmacovigilance has undergone a remarkable evolution, adapting to the growing complexity of drug development, global regulatory landscapes, and public health challenges. The thalidomide tragedy in the 1960s catalyzed the establishment of organized drug safety systems, shaping the foundation for modern pharmacovigilance practices.

This review aims to provide a comprehensive overview of the evolution and current system of pharmacovigilance, exploring its historical milestones, advancements in regulatory frameworks, integration of technology, and emerging trends. The article will also discuss the role of international collaborations, such as the World Health Organization's (WHO) Program for International Drug Monitoring, in harmonizing pharmacovigilance practices globally. [1] Pharmacovigilance, often regarded as the cornerstone of drug safety, plays a critical role in protecting public health by monitoring and assessing the risks and benefits of medicines. It is defined as the science and activities related to the detection, assessment, understanding, and prevention of adverse effects or any other drug-related problems. The importance of pharmacovigilance cannot be overstated, as it serves as a safeguard in ensuring that medicines approved for public use remain safe and effective throughout their lifecycle, from pre-clinical trials to post-marketing surveillance.
       
            fig 1.jpg
       


       
            fig 2.png
       

The evolution of pharmacovigilance has been shaped by major drug safety crises, starting with the infamous thalidomide tragedy in the early 1960s, which led to widespread birth defects. This incident revealed significant gaps in drug safety monitoring and became the catalyst for establishing robust pharmacovigilance systems globally. Over the years, these systems have evolved from rudimentary mechanisms focused on spontaneous adverse event reporting to sophisticated frameworks that now encompass advanced technologies such as artificial intelligence (AI), big data analytics, and real-world evidence (RWE) generation. These advances have enabled health authorities, pharmaceutical companies, and healthcare professionals to proactively identify and mitigate risks associated with medicinal products. [2]

The regulatory landscape surrounding pharmacovigilance has also undergone significant transformation, with international collaboration efforts such as the International Council for Harmonisation (ICH) harmonizing the regulatory requirements for pharmacovigilance across different regions. Key guidelines like ICH E2E on pharmacovigilance planning and ICH E2F on periodic benefit-risk evaluation reports (PBRER) have brought consistency and a unified approach to drug safety monitoring. The role of regulatory bodies such as the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), and the World Health Organization (WHO) has expanded to include rigorous post-marketing surveillance requirements, risk management strategies, and proactive pharmacovigilance systems. [3]

Pharmacovigilance has become even more critical in the current era of rapid technological and therapeutic innovation. The development of complex biological therapies, including monoclonal antibodies, gene therapies, and personalized medicine, presents new challenges for pharmacovigilance systems that were initially designed to monitor traditional small-molecule drugs. As the complexity of treatments increases, the need for specialized approaches to safety monitoring has become apparent. Additionally, the emergence of global health crises such as the COVID-19 pandemic has placed unprecedented pressure on pharmacovigilance systems, particularly in the context of the accelerated development and emergency use of vaccines and treatments. [4]  The current pharmacovigilance landscape is multifaceted, involving a wide array of stakeholders, including regulatory authorities, pharmaceutical companies, healthcare professionals, and patients. Each of these groups plays a crucial role in the effective functioning of pharmacovigilance systems. Healthcare professionals and patients contribute through adverse event reporting, while pharmaceutical companies are responsible for continuous safety monitoring and regulatory compliance. Regulatory authorities, in turn, oversee the entire process, ensuring that drug safety data is rigorously evaluated and that necessary actions, such as labeling changes, risk mitigation strategies, or product withdrawals, are implemented to protect public health. [5]  Despite significant advancements, challenges remain in the field of pharmacovigilance. Underreporting of adverse drug reactions (ADRs), particularly in resource-limited settings, continues to undermine the effectiveness of pharmacovigilance systems. Additionally, the sheer volume of data generated from multiple sources, including spontaneous reports, clinical trials, and real-world evidence, presents logistical and analytical challenges. However, innovations in data science, machine learning, and signal detection are being leveraged to enhance the efficiency and accuracy of pharmacovigilance activities. [6]

This review article aims to provide a comprehensive overview of the system and evolution of pharmacovigilance, exploring its historical development, regulatory frameworks, current challenges, and emerging trends. By examining key case studies, technological innovations, and global efforts to harmonize pharmacovigilance practices, this article highlights the critical role pharmacovigilance plays in ensuring the safety of medicinal products worldwide. The ongoing evolution of pharmacovigilance reflects the dynamic nature of drug development and public health needs, emphasizing the importance of continuous vigilance and adaptation to safeguard patients and improve therapeutic outcomes. [7]  Pharmacovigilance is a vital component of the healthcare system, designed to ensure that medicines remain safe for patients, not just during clinical trials but throughout their entire life cycle. As a specialized branch of medical science, pharmacovigilance (PV) involves the continuous monitoring, detection, assessment, and prevention of adverse drug reactions (ADRs) and other drug-related problems. The overarching goal is to minimize risks while maximizing the therapeutic benefits of medications. This is especially crucial in today’s complex therapeutic environment, where new, more advanced drugs such as biologics, gene therapies, and personalized medicines are regularly introduced to the market. [8]

1.1. Historical Background:

The evolution of pharmacovigilance is deeply rooted in history, particularly in response to public health tragedies. The most notable of these was the thalidomide disaster of the 1960s, which resulted in thousands of children being born with severe birth defects due to the drug’s unanticipated teratogenic effects. Thalidomide was initially marketed as a safe sedative for pregnant women, but its catastrophic side effects revealed significant gaps in the drug safety systems of the time. This event marked a turning point in global drug regulation and underscored the necessity for robust pharmacovigilance mechanisms to prevent such incidents from happening again.

In response to this crisis, the World Health Organization (WHO) established the Programme for International Drug Monitoring in 1968, which later gave rise to the Uppsala Monitoring Centre (UMC) in 1978. The UMC became the central hub for collecting and analyzing reports of suspected ADRs from member countries worldwide, creating a global network of pharmacovigilance. This marked the formalization of a global effort toward drug safety monitoring, laying the groundwork for modern pharmacovigilance systems. [9]

1.2. The Current Pharmacovigilance Landscape: A Global Network:

Today, pharmacovigilance operates as a highly structured system that involves multiple stakeholders, including regulatory bodies (such as the FDA in the United States and the EMA in Europe), pharmaceutical companies, healthcare professionals, and patients. These stakeholders contribute to an interconnected network designed to detect and manage the risks associated with the use of medicines, especially after they have been approved for public use.

The development of pharmacovigilance systems has also been shaped by international collaboration and the creation of harmonized guidelines. The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) plays a critical role in this process. ICH guidelines, such as E2E (Pharmacovigilance Planning) and E2F (Periodic Benefit-Risk Evaluation Reports), provide a framework that standardizes pharmacovigilance practices across different regions, ensuring a consistent approach to drug safety. [10]

1.3. Technological Advances: Shaping the Future of Pharmacovigilance:

As pharmacovigilance has evolved, so have the tools and methods used to manage drug safety. The advent of artificial intelligence (AI) and machine learning (ML) is transforming the way drug safety data is analyzed. These technologies allow for the processing of vast amounts of data from multiple sources, including electronic health records (EHRs), social media, and patient registries. By using AI and ML, pharmacovigilance systems can identify patterns and signals faster and more accurately than ever before. This enhances the ability to detect ADRs that may not have been apparent during clinical trials, ultimately leading to earlier interventions and increased patient safety.  Big data analytics is another area revolutionizing pharmacovigilance. With the ability to handle extensive datasets from diverse sources, big data allows for a more comprehensive analysis of drug safety across different populations. This approach is particularly useful for identifying rare adverse events that may occur only in specific subgroups or after long-term use. Additionally, the use of real-world evidence (RWE) is becoming more prevalent in pharmacovigilance, as it provides insights into how drugs perform in everyday clinical settings, outside of the controlled environment of clinical trials. [11]

EVOLUTION OF PHARMACOVIGILANCE:

Pre-1960s: The Pre-Pharmacovigilance Era:

Lack of Structured Systems:

•     Before the 1960s, formal systems for drug safety monitoring were largely absent. Regulatory authorities focused primarily on ensuring the quality, efficacy, and purity of drugs during manufacturing but paid little attention to post-marketing surveillance.

•     Safety concerns were often identified only after a drug was already widely used, relying on anecdotal evidence from healthcare professionals or patient reports. [12]

2.1.2.                                     The Limitations of Early Drug Monitoring:

•     Drug safety was mostly assessed through limited pre-marketing clinical trials that had small sample sizes and often lacked long-term data on side effects.

•     This reactive approach to drug safety frequently led to delayed responses to harmful side effects, as adverse drug reactions (ADRs) were identified only after they had caused widespread harm. [13]

2.2. 1960s to 1980s: Birth of Modern Pharmacovigilance:

2.2.1.                                  Thalidomide Tragedy: A Turning Point (1961):

•     The thalidomide tragedy in the early 1960s marked the beginning of structured pharmacovigilance systems. Thalidomide, introduced in the late 1950s as a sedative, was widely prescribed to pregnant women to alleviate morning sickness. However, it was later discovered to cause severe birth defects (phocomelia).

•     Thousands of children were born with deformities, particularly in Europe, due to a lack of safety monitoring for drugs during pregnancy. This tragedy revealed the deficiencies in pre-approval clinical trials and post-marketing drug surveillance. [14]

2.2.2.              Creation of the WHO Programme for International Drug Monitoring:

•     In response to the thalidomide disaster, the World Health Organization (WHO) launched the Programme for International Drug Monitoring in 1968. The program was designed to coordinate and centralize the global collection of adverse drug reaction (ADR) reports from member states.

•     The program encouraged countries to establish their own national pharmacovigilance systems, leading to the creation of formal reporting structures for ADRs. [15]

2.2.3.                             Establishment of Uppsala Monitoring Centre (1978):

•     The WHO established the Uppsala Monitoring Centre (UMC) in Sweden in 1978 to serve as the global hub for ADR reporting and data analysis. The UMC has played a crucial role in developing standardized reporting practices and facilitating international collaboration on drug safety.

•     The centre oversees VigiBase, a global database for individual case safety reports (ICSRs) submitted by national pharmacovigilance centers from WHO member countries. This marked the first global effort to systematically monitor drug safety. [16]

2.3. 1990s to Early 2000s: Global Harmonization and Expansion:

2.3.1.                                           Growth of Regulatory Frameworks:

•     In the 1990s, as the pharmaceutical industry expanded and drugs became more complex, there was a growing recognition of the need for standardized global pharmacovigilance practices.

•     This led to the formation of the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) in 1990, which aimed to harmonize regulatory standards across Europe, the United States, and Japan. [17]

2.3.2.                           Introduction of ICH Guidelines for Pharmacovigilance:

•     The ICH E2 series guidelines, developed in the 1990s, were pivotal in standardizing pharmacovigilance practices globally. Key guidelines include: o ICH E2E (Pharmacovigilance Planning): A guideline for pre- and postmarketing pharmacovigilance planning. o ICH E2F (Development Safety Update Report - DSUR): Guidance for ongoing safety evaluations during clinical development.

•     These guidelines ensured that countries and pharmaceutical companies followed similar protocols for ADR reporting, signal detection, and risk-benefit assessments. [18]

3.3 Regulatory Expansion and Formalization:

•     By the late 1990s, many countries developed formal regulatory agencies with specific departments for pharmacovigilance.

o     The U.S. Food and Drug Administration (FDA) implemented new regulations for post-marketing surveillance through programs like MedWatch.

o     The European Medicines Agency (EMA) established EudraVigilance for collecting and managing information on suspected ADRs for medicines authorized in the

European Economic Area. [19]

2.3.4 Integration of Risk Management Strategies:

•     Pharmacovigilance systems began to incorporate Risk Management Plans (RMPs) and Risk Evaluation and Mitigation Strategies (REMS), ensuring that identified risks were managed proactively after drug approval. This approach included measures like restricted distribution systems, special labeling, and ongoing safety studies. [20]

3.4. Mid-2000s to Present: Technological Integration and Advanced Pharmacovigilance:

3.4.1 Shift Toward Proactive Pharmacovigilance:

•     In recent years, pharmacovigilance has evolved from a primarily reactive system—focused on identifying ADRs after they occur—to a more proactive approach that involves risk prediction, early signal detection, and benefit-risk management throughout a drug’s lifecycle.

•     The increasing complexity of therapies (biologics, gene therapies) and the rise of personalized medicine have prompted more sophisticated pharmacovigilance tools and strategies. [21]

3.4.2 Artificial Intelligence and Machine Learning:

•     The advent of artificial intelligence (AI) and machine learning (ML) has transformed the field of pharmacovigilance. These technologies allow for the rapid processing and analysis of vast amounts of ADR data, enabling faster detection of signals and safety trends.

•     AI-driven systems are capable of real-time monitoring of multiple data sources (clinical trials, electronic health records, social media, patient registries), improving the timeliness and accuracy of ADR detection. [22]

Case Example: Advanced AI Integration:

Example: FDA’s Sentinel Initiative: The Sentinel System, initiated by the U.S. Food and Drug Administration (FDA), integrates AI/ML to monitor the safety of approved drugs. Sentinel processes massive datasets from healthcare systems to detect ADRs in near real-time. Advanced ML models analyze these data to predict risk factors associated with drug use, enabling proactive regulatory action.

Impact:

•     Real-time risk detection has shortened the time from signal detection to intervention.

•     Enhanced identification of drug-drug interactions in complex patient populations. [23]

3.4.3 Big Data and Real-World Evidence:

•     Big data analytics has emerged as a key tool in pharmacovigilance, allowing for the analysis of large datasets from various sources, including real-world evidence (RWE) gathered from electronic health records (EHRs), patient wearables, and health insurance claims. These insights provide a clearer understanding of a drug’s safety profile in broader, real-world populations beyond clinical trial settings.

•     Real-world data (RWD) allows for continuous monitoring and assessment of drugs postapproval, especially in rare populations or long-term use cases. [23,24]

3.4.4 Social Media and Digital Pharmacovigilance:

•     The use of social media monitoring and digital health platforms has become an emerging source of ADR reports. Patients often share their drug experiences online, providing pharmacovigilance systems with additional data to identify potential safety concerns that may not be captured through traditional channels. [24]

3.4.5 Pharmacovigilance in the COVID-19 Era: 

•     The COVID-19 pandemic significantly accelerated the evolution of pharmacovigilance. The rapid development, approval, and distribution of vaccines and treatments created a need for real-time pharmacovigilance systems that could assess and address emerging safety signals.

•     Health authorities worldwide adapted their systems to handle the vast influx of data on vaccine-related ADRs. Technologies like AI and cloud-based platforms were instrumental in monitoring the large-scale vaccination programs globally, ensuring the safety of millions of people. [25]

4.    SYSTEM OF PHARMACOVIGILANCE:

4.1. Key Components of Pharmacovigilance Systems:

4.1.1 Regulatory Authorities:

•     Role: Regulatory authorities are responsible for overseeing and enforcing pharmacovigilance activities. They ensure that drug manufacturers comply with safety monitoring requirements and that adverse drug reactions (ADRs) are reported and managed appropriately.

•     Examples:

o FDA (U.S.): Oversees the MedWatch program for ADR reporting and conducts inspections of pharmaceutical manufacturers. o EMA (EU): Manages EudraVigilance, the system for monitoring ADRs in the European Economic Area (EEA). o MHRA (UK): Oversees the Yellow Card Scheme for ADR reporting in the UK. [26]

4.1.2 Pharmaceutical Companies:

•     Role: Drug manufacturers have an obligation to continuously monitor the safety of their products once they reach the market. They are required to submit reports on ADRs, conduct

risk assessments, and ensure that any safety issues are communicated to regulatory authorities and healthcare providers.

•     Post-Marketing Surveillance: Manufacturers must monitor drugs for adverse events through post-marketing studies, patient registries, and spontaneous reporting.

•     Risk Management Plans (RMPs): Companies must develop and implement RMPs that outline measures to minimize risks associated with their products. [27]

4.1.3 Healthcare Professionals:

•     Role: Healthcare professionals (e.g., doctors, pharmacists, nurses) are essential for identifying and reporting ADRs. They play a critical role in detecting safety signals, especially through spontaneous reporting.

•     Spontaneous Reporting Systems: Healthcare providers report suspected ADRs to regulatory bodies through established systems (e.g., FDA's MedWatch or the WHO Programme for International Drug Monitoring).

•     Education and Training: Healthcare professionals need to be educated on pharmacovigilance principles and the importance of ADR reporting. [28]

4.1.4 Patients and Consumers:

•     Role: Patients themselves can contribute to pharmacovigilance by reporting adverse events they experience, either directly or through their healthcare providers.

•     Patient Reporting Systems: Many countries allow patients to report ADRs directly to regulatory authorities (e.g., MedWatch in the U.S. allows patient self-reporting). In some systems, patients can also report ADRs through mobile apps or online platforms.

4.2. Pharmacovigilance Processes:

4.2.1 Adverse Event Reporting:

•     Spontaneous Reporting: Healthcare professionals, patients, and pharmaceutical companies submit reports about suspected ADRs voluntarily. This is one of the most common and essential methods for collecting data on drug safety.

•     Mandatory Reporting: In some cases, regulatory authorities require pharmaceutical companies to report ADRs to ensure safety monitoring throughout the lifecycle of a drug.

•     Electronic Reporting: With advances in technology, ADR reporting has increasingly become electronic, allowing for faster data submission and processing. [29]

4.2.2 Signal Detection and Analysis:

•     Signal Detection: The process of identifying potential safety issues based on patterns in ADR reports. Signals could arise from an analysis of multiple ADR reports indicating a recurring pattern or unusual incidence.

•     Methods: Signal detection relies on statistical methods, such as disproportionality analysis (e.g., Bayesian data mining) and data mining techniques, to analyze large volumes of ADR data.

•     Tools: Automated tools like VigiBase (managed by UMC) and FDA Adverse Event Reporting System (FAERS) help to identify safety signals by analyzing ADR reports from multiple sources. 

4.2.3 Risk Assessment:

•     Risk-Benefit Assessment: Regulatory authorities and pharmaceutical companies assess the risk-benefit balance of a drug based on available data. This involves determining whether the benefits of a drug outweigh its potential risks, considering both preclinical and post-marketing data.

•     Risk Communication: Regulatory authorities may issue warnings, safety alerts, or updated labeling to communicate new safety information about a drug. These include black box warnings or contraindications based on the identified risks. [30]

4.2.4 Risk Minimization Strategies:

•     Risk Management Plans (RMPs): Pharmaceutical companies develop and implement RMPs for drugs with known risks, outlining strategies to minimize those risks. This can include additional monitoring, restricted access, or patient education.

•     Risk Evaluation and Mitigation Strategies (REMS): In the U.S., drugs with serious risks may be subject to REMS programs, which include requirements like restricted distribution, patient monitoring, and special labeling.

4.3. Data Management and Reporting Systems:

4,3.1 International Reporting Networks:

•     WHO Programme for International Drug Monitoring: Since its creation in 1968, the WHO has played a central role in coordinating ADR reporting globally through the Uppsala Monitoring Centre (UMC). VigiBase is the world’s largest repository of ADR reports.

•     EudraVigilance: The EMA’s database, used for managing ADR data across Europe, allows for the detection of safety signals and ensures that regulatory action can be taken swiftly in case of adverse events.

•     FDA’s FAERS: The FDA Adverse Event Reporting System is a database that collects and analyzes ADR reports from the U.S. healthcare system. [31]

4.3.2 Data Mining and Analytics:

•     Signal Detection Tools: Tools like VigiLyze, FDA's REMS, and EudraVigilance leverage data mining to identify patterns in ADRs, helping pharmacovigilance teams identify new safety signals and trends from large datasets.

•     Big Data and AI: With the growing use of big data, pharmacovigilance systems can incorporate real-time data from various sources, including electronic health records

(EHRs), social media, and health apps, to identify ADRs and emerging risks. [36]

4.3.3. National Regulations:

•     FDA: In the U.S., the FDA mandates post-marketing safety surveillance through the FD&C Act and the MedWatch program, which requires manufacturers to report ADRs and conduct risk assessments.

•     EMA: The European Medicines Agency oversees pharmacovigilance in the European Union through EudraVigilance and the Pharmacovigilance Legislation (Directive 2010/84/EU), ensuring drugs meet safety standards through constant monitoring and reporting. [32]

•     National Pharmacovigilance Centers: Many countries (e.g., India’s Pharmacovigilance Programme of India (PvPI)) have established their own national pharmacovigilance centers to facilitate ADR reporting and drug safety monitoring at a national level.

4.4.4. Post-Marketing Surveillance Regulations:

•     Post-Approval Studies: Post-marketing surveillance is an essential part of pharmacovigilance. Pharmaceutical companies are often required to conduct Phase IV clinical trials or post-marketing surveillance studies to monitor the safety of a drug once it has been approved.

•     Post-Approval Safety Alerts: If an ADR is detected post-marketing, regulatory bodies may issue safety alerts, restrict drug use, or withdraw the product from the market. [33]

5.    Challenges In The Pharmacovigilance System: 

5.1. Underreporting of Adverse Events:

•     Healthcare professionals and patients often fail to report ADRs due to lack of awareness, time constraints, or fear of legal repercussions.

•     Solutions: Increasing education and awareness among healthcare providers and patients about the importance of ADR reporting is critical to improving the pharmacovigilance system. 

5.2. Data Quality and Completeness:

•     Incomplete or inaccurate data can hinder the effectiveness of pharmacovigilance systems. This issue arises from inconsistent reporting formats, inadequate follow-up, or missing patient information.

•     Solutions: Standardization of reporting formats and better training for healthcare professionals can address data quality issues. 

5.3. Regulatory Disparities:

•     Different countries may have varying pharmacovigilance standards and reporting requirements, creating challenges for global drug safety monitoring.

•     Solutions: Harmonization of pharmacovigilance guidelines through international bodies like ICH and WHO can improve global collaboration and consistency. [34]

6.    Future Aspects Of Pharmacovigilance:

6.1. Technological Advancements in Pharmacovigilance:

6.1.1                                    Artificial Intelligence and Machine Learning:

•     AI in Signal Detection: Machine learning (ML) and artificial intelligence (AI) are expected to revolutionize pharmacovigilance by enhancing signal detection and predictive analytics. AI algorithms can mine large datasets of ADR reports, EHRs. These systems can flag potential adverse drug reactions earlier and with greater accuracy than traditional methods.

•     Natural Language Processing (NLP): NLP technologies will enable the extraction of useful information from unstructured data, such as clinical notes, social media discussions, and patient reports, significantly improving the speed and quality of ADR detection.

•     Predictive Analytics: AI-powered predictive models will help foresee safety risks, enabling proactive intervention even before ADRs manifest in large populations. These models will integrate patient demographics, genetic information, and drug interactions to predict potential adverse effects. [35]

6.1.2                                            Big Data and Real-World Evidence:

•     Real-World Data (RWD): The future of pharmacovigilance will increasingly rely on RWD from sources such as electronic health records (EHRs), insurance claims, and patient registries. This data will provide insights into the long-term safety of drugs in diverse populations, including rare and underrepresented groups. It will help improve the understanding of drug safety in everyday clinical settings.

•     Epidemiological Studies: Large-scale epidemiological studies using big data will enable more accurate assessment of drug safety and the identification of rare ADRs. These studies will help in real-time monitoring of public health trends related to drug use.

6.1.3                                       Blockchain Technology for Data Integrity:

•     Decentralized Data Management: Blockchain technology could be used to ensure the integrity and transparency of pharmacovigilance data. By creating an immutable, decentralized ledger of ADR reports, blockchain could improve data accuracy and reduce the potential for data manipulation or fraud.

•     Secure Data Sharing: Blockchain could also facilitate the secure sharing of ADR data between healthcare providers, pharmaceutical companies, and regulatory agencies while maintaining patient privacy and complying with data protection laws (e.g., GDPR,

HIPAA). [36]

6.2. Advancements in Reporting Systems:

6.2.1                                        Mobile and Digital Reporting Platforms:

•     Mobile Applications for ADR Reporting: The future of pharmacovigilance will see the widespread use of mobile apps for ADR reporting. Patients and healthcare professionals will have easy access to platforms where they can directly report adverse events. These apps will improve ADR collection by making the process more user-friendly and accessible.

•     Wearables and Smart Devices: Devices like wearables (smartwatches, fitness trackers) that monitor health in real-time will play a role in pharmacovigilance. These devices could automatically detect health changes and alert users about possible ADRs, thus providing a continuous stream of data to pharmacovigilance systems. [37]

6.2.2 Patient-Centered Pharmacovigilance:

•     Enhanced Patient Reporting: As patients become more engaged in their healthcare, they will have a greater role in reporting ADRs. Direct reporting by patients through apps, online platforms, or social media will become more commonplace. This patient-centered approach will enhance the timeliness and comprehensiveness of ADR reporting.

•     Personalized Drug Safety Monitoring: With the rise of personalized medicine, pharmacovigilance systems will need to adapt to individual patient profiles (e.g., genetic makeup, comorbidities, lifestyle) to identify ADRs more accurately. Personalized pharmacovigilance will become integral in monitoring the safety of tailored treatments, such as gene therapies or biologics.

6.3. Integration of Global Pharmacovigilance Systems:

6.3.1                                         Global Data Sharing and Collaboration:

•     Cross-Border Collaboration: As medicines are marketed globally, the pharmacovigilance systems will require enhanced international cooperation. Regulatory bodies such as the WHO, EMA, FDA, and others will need to establish seamless data-sharing frameworks to facilitate global monitoring of ADRs.

•     International Standardization: As pharmacovigilance evolves, there will be greater efforts to standardize the reporting and evaluation of ADRs across countries. This could include harmonizing databases, reporting formats, and safety guidelines to improve the efficiency of global drug safety monitoring.

6.3.2 Centralized Global Databases:

•     Unified ADR Database: The future may see the creation of a global, centralized ADR database that integrates reports from multiple countries and regions. This centralized system would allow regulators, pharmaceutical companies, and researchers to access a comprehensive and unified dataset for drug safety evaluation.

•     Enhanced Signal Detection Across Borders: A centralized system would improve signal detection by aggregating data from various healthcare systems worldwide, increasing the likelihood of identifying rare or previously unknown ADRs that might not be visible in isolated national databases. [38]

6.4. Personalized and Precision Pharmacovigilance:

6.4.1                               Pharmacogenomics and Genomic Data Integration:

•     Genetic Screening for ADR Risk: As pharmacogenomics becomes more prominent, pharmacovigilance systems will increasingly integrate genetic data to predict and monitor

ADRs. This will allow for personalized ADR risk assessments based on an individual's genetic predisposition to certain side effects.

•     Tailored Drug Safety Monitoring: For patients undergoing treatments such as gene therapy or targeted biologics, pharmacovigilance systems will need to account for specific genetic or molecular markers. This will involve personalized monitoring to track how different patients respond to treatments and identify potential safety concerns early.

6.4.2                                 Real-Time Monitoring of Personalized Medicine:

•     Smart Drugs and Biologics: The growing use of biologics and gene therapies will necessitate real-time pharmacovigilance systems to track patient responses and long-term safety. These advanced therapies often have complex safety profiles that require continuous monitoring.

•     Individualized Risk Profiles: Personalized risk profiles, which consider genetic, environmental, and lifestyle factors, will become part of routine pharmacovigilance processes. This could enable more proactive management of drug safety by predicting potential ADRs before they occur. [39] 6.5. Regulatory Changes and Future Guidelines:

6.5.1 Strengthening Post-Market Surveillance:

•     Enhanced Post-Approval Studies: Regulatory authorities will likely mandate more extensive post-approval surveillance and long-term monitoring for newly approved drugs, particularly biologics and novel therapies. Ongoing safety studies will help to ensure that ADRs are identified early and appropriately managed after market launch.

•     Continuous Monitoring for High-Risk Drugs: Drugs that are deemed to pose a higher risk to patients (e.g., immunotherapies, new vaccines, or targeted therapies) will undergo more intensive and continuous monitoring throughout their market life, with strict regulatory requirements for periodic safety updates.

6.5.2 Adaptive Regulatory Frameworks:

•     Dynamic Safety Regulations: The future of pharmacovigilance will see the adoption of more adaptive regulatory frameworks that can quickly respond to new safety concerns and emerging data. This may involve more agile regulatory approvals and post-market conditions that can be updated in real time based on evolving safety data.

•     Global Regulatory Harmonization: The ICH and other global organizations will likely continue to push for the harmonization of pharmacovigilance standards, facilitating smoother collaboration between global regulatory agencies, pharmaceutical companies, and healthcare providers. [40]

6.6. Ethical and Social Considerations

6.6.1 Patient Privacy and Data Security

•     Ensuring Data Privacy: With the growing reliance on big data, AI, and personal health data, ensuring the privacy and security of patient information will be a major challenge. Regulatory authorities will need to balance the need for comprehensive data collection with robust data protection measures, adhering to regulations like GDPR and HIPAA.

•     Informed Consent in Digital Pharmacovigilance: As digital tools and mobile apps become more integrated into pharmacovigilance systems, patients will need to provide informed consent for the collection and use of their health data. This may require revisiting ethical standards and consent processes in the digital age.

6.6.2 Enhancing Public Awareness and Engagement

•     Patient-Centric Pharmacovigilance: The future of pharmacovigilance will focus on empowering patients to take an active role in drug safety. Public education campaigns will be critical to raising awareness about ADR reporting and the role of patients in pharmacovigilance.

•     Transparency in Safety Communication: As pharmacovigilance systems evolve, there will be an increasing demand for transparency in how drug safety information is communicated to the public. This could include real-time safety data dashboards or more frequent safety updates from regulatory authorities. [40]

7.    CHALLENGES AND OPPORTUNITIES

7.1. Underreporting and Incomplete Data

•     Solutions: To address underreporting, especially among healthcare professionals, patient engagement, and education will be crucial. Innovations like mobile apps and digital platforms can make reporting easier and more accessible.

•     Opportunities: By embracing digital tools and AI, pharmacovigilance systems can gather data more efficiently, reducing the burden of underreporting.

7.2. Evolving Risk Assessment Methods

•     Solutions: As new therapies are developed, regulatory bodies will need to adopt more dynamic and flexible risk assessment methodologies to address complex safety profiles, particularly in personalized medicine.

•     Opportunities: Incorporating AI, real-world data, and genomic information into risk assessments will provide a more accurate and comprehensive understanding of drug safety. [41]

SUMMARY: 

In conclusion, pharmacovigilance has undergone significant evolution from its early days of reactive post-marketing surveillance to a more comprehensive, proactive, and global approach to drug safety monitoring. The establishment of regulatory authorities such as the FDA, EMA, and MHRA, along with the creation of reporting systems like MedWatch, EudraVigilance, and the Yellow Card Scheme, has laid the groundwork for robust drug safety frameworks. Over time, the role of multiple stakeholders including pharmaceutical companies, healthcare professionals, and patients has become integral to the success of pharmacovigilance systems, ensuring more accurate and timely detection of adverse drug reactions (ADRs). The increasing use of advanced technologies such as data mining, artificial intelligence, and real-world evidence is expected to further enhance pharmacovigilance efforts by improving signal detection, data analysis, and risk management. As pharmacovigilance continues to evolve, global collaboration, harmonization of regulations, and the integration of personalized medicine will be key in ensuring the safety and well-being of patients. Ultimately, a more proactive, integrated, and technologically advanced pharmacovigilance system will play a crucial role in safeguarding public health and improving the therapeutic outcomes of drug treatments.

REFERENCES

  1. Waller, P. (2018). Pharmacovigilance: A definition. In An Introduction to Pharmacovigilance (pp. 1-7). Wiley.
  2. World Health Organization (WHO). (2002). "Pharmacovigilance: A guide for its implementation."
  3. Lazarou, J., Pomeranz, B. H., & Corey, P. N. (1998). "Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies." JAMA, 279(15), 1200-1205.
  4. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). (2003). "E2E Pharmacovigilance."
  5. Sánchez, S. R., & Prado, A. (2019). "Challenges in pharmacovigilance: The future of drug safety monitoring." Pharmacological Research, 142, 251-261.
  6. Ezzeldin, E., & Gomaa, A. (2021). "Pharmacovigilance: The role of stakeholders in drug safety." Journal of Clinical Pharmacy and Therapeutics, 46(5), 1207-1216.
  7. Moulds, R. W., & Smith, D. J. (2019). "Advances in pharmacovigilance: The role of big data and machine learning." Drug Safety, 42(10), 1233-1244.
  8. Boulanger, L., & Green, D. (2020). "The future of pharmacovigilance in the age of precision medicine." Journal of Pharmaceutical Sciences, 109(1), 5-11.
  9. Simoens, S., & Laekeman, G. (2005). "Pharmacovigilance: An integral part of drug safety." Acta Clinica Belgica, 60(1), 10-17.
  10. Kemp, T. (2003). "The Thalidomide Tragedy." Pharmacology & Therapeutics, 99(2), 177188.
  11. Lazarou, J., Pomeranz, B. H., & Corey, P. N. (1998). "Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies." JAMA, 279(15), 1200-1205.
  12. Dinev, T., & Sitar, S. (2021). "Artificial Intelligence in Pharmacovigilance: Applications and Future Directions." Journal of Clinical Pharmacology, 61(7), 830-838.
  13. Edwards, I. R., & Aronson, J. K. (2000). Adverse drug reactions: definitions, diagnosis, and management. Lancet, 356(9237), 1255-1259. https://doi.org/10.1016/S0140-6736(00)02759-9
  14. Hennessy, S., & Cohen, J. T. (2013). The shortcomings of early drug safety monitoring. Drug Safety, 36(5), 295-301. https://doi.org/10.1007/s40264-013-0034-3
  15. Pomeranz, B., & Schurr, T. (2011). The Thalidomide Tragedy and its Legacy: The Birth of Modern Pharmacovigilance. Therapeutic Advances in Drug Safety, 2(2), 63-69. https://doi.org/10.1177/2042098610390090
  16. The World Health Organization. (2008). The WHO Programme for International Drug Monitoring: Progress and Future Directions. Pharmacovigilance, 14(1), 1-9. https://doi.org/10.1002/phar.0431
  17. Ågren, T., & Edwards, I. R. (2013). Uppsala Monitoring Centre: A World Leader in Pharmacovigilance. Drug Safety, 36(6), 413-420. https://doi.org/10.1007/s40264-013-0069-3
  18. Mazzoni, G., & Grandi, M. (2005). Regulatory Frameworks in Pharmacovigilance: Harmonization Efforts. Journal of Regulatory Science, 9(3), 112-118.
  19. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). (1996). ICH E2E Pharmacovigilance: Guidelines for Reporting Adverse Drug Reactions. Pharmaceutical Medicine, 10(1), 42-48. https://doi.org/10.1007/BF03292232
  20. Ross, S. R., & Pillinger, M. (2000). Post-Marketing Drug Surveillance and Regulatory Agencies. Journal of Clinical Pharmacology, 40(5), 525-533. https://doi.org/10.1002/j.1552-4604.2000.tb00119.x
  21. European Medicines Agency (EMA). (2014). Risk Management Plans: Enhancing Pharmacovigilance. Pharmaceutical Policy and Law, 16(2), 65-77. https://doi.org/10.3233/PPL-140019
  22. "FDA's Sentinel Initiative" The Sentinel System integrates AI/ML to process large datasets from healthcare systems, identifying adverse drug reactions in real-time and enhancing the detection of drug-drug interactions. For further details, visit the FDA Sentinel Initiative official page
  23. van Puijenbroek, E., & Bate, A. (2015). Proactive Pharmacovigilance: From Reactive Surveillance to Predictive Risk Assessment. Drug Safety, 38(4), 351-360. https://doi.org/10.1007/s40264-015-0271-4
  24. Muenster, J., & Bosch, T. (2020). The Role of Artificial Intelligence in Pharmacovigilance. Journal of Pharmaceutical Sciences, 109(7), 2199-2207.https://doi.org/10.1016/j.xphs.2020.03.025
  25. Baker, L., & Wilson, C. (2017). Big Data and Real-World Evidence in Pharmacovigilance. Journal of Clinical Pharmacology, 57(9), 1073-1079. https://doi.org/10.1002/jcph.1028
  26. Patel, P. K., & Ghosh, S. (2019). Social Media as a Tool for Pharmacovigilance: New Insights. Pharmaceutical Medicine, 33(4), 183-190. https://doi.org/10.1007/s40290-019-00313-9
  27. Aymé, S., & Smith, G. (2021). Pharmacovigilance in the COVID-19 Era: Real-Time Safety Monitoring of Vaccines. The Lancet Infectious Diseases, 21(8), e161-e169. https://doi.org/10.1016/S1473-3099(21)00258-0
  28. World Health Organization. (2021). Global Individual Case Safety Reports (ICSRs) for Pharmacovigilance." https://www.who.int
  29. Bourne, D. E., & Smith, S. (2018). Risk Management and Drug Safety: Understanding Post-Marketing Surveillance. Pharmaceutical Journal, 301(7922), 222–226."
  30. Hernandez, A. V., & Shaikh, A. (2020). The Role of Healthcare Professionals in Pharmacovigilance. Journal of Clinical Pharmacology, 60(2), 242-249."
  31. "Lazarou, J., Pomeranz, B. H., & Corey, P. N. (1998). Incidence of Adverse Drug Reactions in Hospitalized Patients: A Meta-Analysis of Prospective Studies. JAMA, 279(15), 1200-1205."
  32. "Liu, T., & Wang, J. (2019). Advances in Adverse Drug Reaction Reporting Systems: A Global Overview. Pharmacoepidemiology and Drug Safety, 28(9), 1205-1213."
  33. "Patel, V., & Mott, A. (2021). Signal Detection in Pharmacovigilance: Challenges and Tools. Regulatory Toxicology and Pharmacology, 120, 1045-1052."
  34. Goldstein, R., & Morgan, M. (2022). Risk-Benefit Assessment in Pharmacovigilance: A Critical Review. International Journal of Risk Assessment and Management, 23(3), 193-201."
  35. "Hughes, R. L., & Johnson, T. (2020). Risk Management Plans and Their Role in Minimizing Drug Safety Risks. Journal of Pharmaceutical Health Services Research, 11(4), 250-255."
  36. World Health Organization. (2019). The Uppsala Monitoring Centre (UMC) and the WHO Programme for International Drug Monitoring. WHO Bulletin, 97(1), 56-60."
  37. "Smith, M. J., & Baker, J. (2019). Data Privacy and Security in Pharmacovigilance. Pharmaceutical Technology, 43(8), 123-127."
  38. Zhao, C., & Wang, T. (2022). Big Data and AI in Pharmacovigilance: Tools and Techniques. Pharmacoepidemiology and Drug Safety, 31(2), 197-205."
  39. "U.S. Food and Drug Administration. (2020). MedWatch: The FDA Safety Information and Adverse Event Reporting Program. FDA.gov."
  40. Kesselheim, A. S., & Avorn, J. (2019). Underreporting of Adverse Drug Events: Causes, Solutions, and Implications. New England Journal of Medicine, 378(22), 2124-2132."
  41. Mousavi, S. R., & Mostafavi, F. (2021). Improving the Quality and Completeness of Pharmacovigilance Data: Challenges and Strategies. Journal of Clinical Pharmacy and Therapeutics, 46(4), 1123-1130."
  42. "Bai, S., & Zhang, W. (2021). Artificial Intelligence in Pharmacovigilance: Signal Detection and Risk Management. Journal of Pharmaceutical Innovation, 16(1), 43-50."
  43. "Lee, J., & Ryu, D. (2020). Real-World Evidence in Pharmacovigilance: Leveraging Big Data for Better Drug Safety Monitoring. Journal of Pharmacovigilance, 27(2), 101-109."
  44. "Miller, D., & Lee, S. (2020). The Future of ADR Reporting: The Role of Mobile Apps and Digital Platforms. Drug Safety, 43(6), 677-683."

Reference

  1. Waller, P. (2018). Pharmacovigilance: A definition. In An Introduction to Pharmacovigilance (pp. 1-7). Wiley.
  2. World Health Organization (WHO). (2002). "Pharmacovigilance: A guide for its implementation."
  3. Lazarou, J., Pomeranz, B. H., & Corey, P. N. (1998). "Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies." JAMA, 279(15), 1200-1205.
  4. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). (2003). "E2E Pharmacovigilance."
  5. Sánchez, S. R., & Prado, A. (2019). "Challenges in pharmacovigilance: The future of drug safety monitoring." Pharmacological Research, 142, 251-261.
  6. Ezzeldin, E., & Gomaa, A. (2021). "Pharmacovigilance: The role of stakeholders in drug safety." Journal of Clinical Pharmacy and Therapeutics, 46(5), 1207-1216.
  7. Moulds, R. W., & Smith, D. J. (2019). "Advances in pharmacovigilance: The role of big data and machine learning." Drug Safety, 42(10), 1233-1244.
  8. Boulanger, L., & Green, D. (2020). "The future of pharmacovigilance in the age of precision medicine." Journal of Pharmaceutical Sciences, 109(1), 5-11.
  9. Simoens, S., & Laekeman, G. (2005). "Pharmacovigilance: An integral part of drug safety." Acta Clinica Belgica, 60(1), 10-17.
  10. Kemp, T. (2003). "The Thalidomide Tragedy." Pharmacology & Therapeutics, 99(2), 177188.
  11. Lazarou, J., Pomeranz, B. H., & Corey, P. N. (1998). "Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies." JAMA, 279(15), 1200-1205.
  12. Dinev, T., & Sitar, S. (2021). "Artificial Intelligence in Pharmacovigilance: Applications and Future Directions." Journal of Clinical Pharmacology, 61(7), 830-838.
  13. Edwards, I. R., & Aronson, J. K. (2000). Adverse drug reactions: definitions, diagnosis, and management. Lancet, 356(9237), 1255-1259. https://doi.org/10.1016/S0140-6736(00)02759-9
  14. Hennessy, S., & Cohen, J. T. (2013). The shortcomings of early drug safety monitoring. Drug Safety, 36(5), 295-301. https://doi.org/10.1007/s40264-013-0034-3
  15. Pomeranz, B., & Schurr, T. (2011). The Thalidomide Tragedy and its Legacy: The Birth of Modern Pharmacovigilance. Therapeutic Advances in Drug Safety, 2(2), 63-69. https://doi.org/10.1177/2042098610390090
  16. The World Health Organization. (2008). The WHO Programme for International Drug Monitoring: Progress and Future Directions. Pharmacovigilance, 14(1), 1-9. https://doi.org/10.1002/phar.0431
  17. Ågren, T., & Edwards, I. R. (2013). Uppsala Monitoring Centre: A World Leader in Pharmacovigilance. Drug Safety, 36(6), 413-420. https://doi.org/10.1007/s40264-013-0069-3
  18. Mazzoni, G., & Grandi, M. (2005). Regulatory Frameworks in Pharmacovigilance: Harmonization Efforts. Journal of Regulatory Science, 9(3), 112-118.
  19. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). (1996). ICH E2E Pharmacovigilance: Guidelines for Reporting Adverse Drug Reactions. Pharmaceutical Medicine, 10(1), 42-48. https://doi.org/10.1007/BF03292232
  20. Ross, S. R., & Pillinger, M. (2000). Post-Marketing Drug Surveillance and Regulatory Agencies. Journal of Clinical Pharmacology, 40(5), 525-533. https://doi.org/10.1002/j.1552-4604.2000.tb00119.x
  21. European Medicines Agency (EMA). (2014). Risk Management Plans: Enhancing Pharmacovigilance. Pharmaceutical Policy and Law, 16(2), 65-77. https://doi.org/10.3233/PPL-140019
  22. "FDA's Sentinel Initiative" The Sentinel System integrates AI/ML to process large datasets from healthcare systems, identifying adverse drug reactions in real-time and enhancing the detection of drug-drug interactions. For further details, visit the FDA Sentinel Initiative official page
  23. van Puijenbroek, E., & Bate, A. (2015). Proactive Pharmacovigilance: From Reactive Surveillance to Predictive Risk Assessment. Drug Safety, 38(4), 351-360. https://doi.org/10.1007/s40264-015-0271-4
  24. Muenster, J., & Bosch, T. (2020). The Role of Artificial Intelligence in Pharmacovigilance. Journal of Pharmaceutical Sciences, 109(7), 2199-2207.https://doi.org/10.1016/j.xphs.2020.03.025
  25. Baker, L., & Wilson, C. (2017). Big Data and Real-World Evidence in Pharmacovigilance. Journal of Clinical Pharmacology, 57(9), 1073-1079. https://doi.org/10.1002/jcph.1028
  26. Patel, P. K., & Ghosh, S. (2019). Social Media as a Tool for Pharmacovigilance: New Insights. Pharmaceutical Medicine, 33(4), 183-190. https://doi.org/10.1007/s40290-019-00313-9
  27. Aymé, S., & Smith, G. (2021). Pharmacovigilance in the COVID-19 Era: Real-Time Safety Monitoring of Vaccines. The Lancet Infectious Diseases, 21(8), e161-e169. https://doi.org/10.1016/S1473-3099(21)00258-0
  28. World Health Organization. (2021). Global Individual Case Safety Reports (ICSRs) for Pharmacovigilance." https://www.who.int
  29. Bourne, D. E., & Smith, S. (2018). Risk Management and Drug Safety: Understanding Post-Marketing Surveillance. Pharmaceutical Journal, 301(7922), 222–226."
  30. Hernandez, A. V., & Shaikh, A. (2020). The Role of Healthcare Professionals in Pharmacovigilance. Journal of Clinical Pharmacology, 60(2), 242-249."
  31. "Lazarou, J., Pomeranz, B. H., & Corey, P. N. (1998). Incidence of Adverse Drug Reactions in Hospitalized Patients: A Meta-Analysis of Prospective Studies. JAMA, 279(15), 1200-1205."
  32. "Liu, T., & Wang, J. (2019). Advances in Adverse Drug Reaction Reporting Systems: A Global Overview. Pharmacoepidemiology and Drug Safety, 28(9), 1205-1213."
  33. "Patel, V., & Mott, A. (2021). Signal Detection in Pharmacovigilance: Challenges and Tools. Regulatory Toxicology and Pharmacology, 120, 1045-1052."
  34. Goldstein, R., & Morgan, M. (2022). Risk-Benefit Assessment in Pharmacovigilance: A Critical Review. International Journal of Risk Assessment and Management, 23(3), 193-201."
  35. "Hughes, R. L., & Johnson, T. (2020). Risk Management Plans and Their Role in Minimizing Drug Safety Risks. Journal of Pharmaceutical Health Services Research, 11(4), 250-255."
  36. World Health Organization. (2019). The Uppsala Monitoring Centre (UMC) and the WHO Programme for International Drug Monitoring. WHO Bulletin, 97(1), 56-60."
  37. "Smith, M. J., & Baker, J. (2019). Data Privacy and Security in Pharmacovigilance. Pharmaceutical Technology, 43(8), 123-127."
  38. Zhao, C., & Wang, T. (2022). Big Data and AI in Pharmacovigilance: Tools and Techniques. Pharmacoepidemiology and Drug Safety, 31(2), 197-205."
  39. "U.S. Food and Drug Administration. (2020). MedWatch: The FDA Safety Information and Adverse Event Reporting Program. FDA.gov."
  40. Kesselheim, A. S., & Avorn, J. (2019). Underreporting of Adverse Drug Events: Causes, Solutions, and Implications. New England Journal of Medicine, 378(22), 2124-2132."
  41. Mousavi, S. R., & Mostafavi, F. (2021). Improving the Quality and Completeness of Pharmacovigilance Data: Challenges and Strategies. Journal of Clinical Pharmacy and Therapeutics, 46(4), 1123-1130."
  42. "Bai, S., & Zhang, W. (2021). Artificial Intelligence in Pharmacovigilance: Signal Detection and Risk Management. Journal of Pharmaceutical Innovation, 16(1), 43-50."
  43. "Lee, J., & Ryu, D. (2020). Real-World Evidence in Pharmacovigilance: Leveraging Big Data for Better Drug Safety Monitoring. Journal of Pharmacovigilance, 27(2), 101-109."
  44. "Miller, D., & Lee, S. (2020). The Future of ADR Reporting: The Role of Mobile Apps and Digital Platforms. Drug Safety, 43(6), 677-683."

Photo
Sakshi Kodilkar
Corresponding author

Ashokrao Mane institute of pharmacy, Ambap

Photo
Sonal Kumbhar
Co-author

Ashokrao Mane institute of pharmacy, Ambap

Photo
Dr. N. B. Chougule
Co-author

Ashokrao Mane institute of pharmacy, Ambap

Photo
Sakshi Raghunath Patil
Co-author

Ashokrao Mane institute of pharmacy, Ambap

Photo
Aditi Maruti Mahanwar
Co-author

Ashokrao Mane institute of pharmacy, Ambap

Sakshi Kodilkar*, Sonal Kumbhar, Dr. N. B. Chougule, Sakshi Patil, Aditi Mahanwar, A Review on: Evolution and System of Pharmacovigilances, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 12, 223-237. https://doi.org/10.5281/zenodo.14264405

More related articles
Navigating India's Expanding OTC Healthcare Market...
Ashutosh Chand Kaushal, Arjun Jaiswal, Sushil Chaurasiya, Shiva, ...
Innovations In Oral Drug Administration: Exploring...
VIGNESH . R, Kamaleshwari B., Kaviya G., Kovarthanan M., Mohan Ra...
Anti-Cancer Activity Of Cisplatin And Its Toxicity...
Pinki Chandra , Gagan Panda, Sudhir Kathane, Shruti Rathore , ...
Morphology and Phytochemical analysis of Plant Girardinia diversifolia also know...
Anita kumari , Dev Prakash Dahiya, Chinu Kumari , Munish Choudhary , Pratibha Rana , Dinesh Kumar, ...
A Comparative Study of Diabetic Treatment – Allopathy vs. Ayurveda...
Angad Bhaskar Newre, Abhay S. Jejurkar, Akash S. Pagar, Akshay S. Gavali, Dr. Rajendra M. Kawde, ...
An Interventional Study On Hemodialysis Patient And Impact Of Patient Counseling...
Basheera. V, Rahna Rasheed, Remeeza Jabbar, Rinsa. A, ...
Related Articles
A Review On Analytical Method Development And Validation For Broccoli...
G. M. Srimyvizhiy, K. B. Ilango, T. Geetha, S. Jayashree, S. Kaviyanchally, M. Mutharasi, A. Naveenk...
Review Article On Organogels Methodology And Types Of Organogelators...
Rushikesh Khemnar, Sonia Singh, Nikhil Sutar, Pallavi Kaple, Avadhut Kulkarni, vitthal Dherange, Pan...
Pharmacovigilance In The Digital Age...
Shruti Aher, Manoj Mahajan, Aman Upaganlawar, Chandrashekhar Upasani, ...
Navigating India's Expanding OTC Healthcare Market: Trends, Challenges, And Stra...
Ashutosh Chand Kaushal, Arjun Jaiswal, Sushil Chaurasiya, Shiva, Ajeet Sharma, Ramayan Yadav, Neha C...
More related articles
Navigating India's Expanding OTC Healthcare Market: Trends, Challenges, And Stra...
Ashutosh Chand Kaushal, Arjun Jaiswal, Sushil Chaurasiya, Shiva, Ajeet Sharma, Ramayan Yadav, Neha C...
Innovations In Oral Drug Administration: Exploring Gastro Retentive Raft Forming...
VIGNESH . R, Kamaleshwari B., Kaviya G., Kovarthanan M., Mohan Raj U., ...
Anti-Cancer Activity Of Cisplatin And Its Toxicity...
Pinki Chandra , Gagan Panda, Sudhir Kathane, Shruti Rathore , ...
Navigating India's Expanding OTC Healthcare Market: Trends, Challenges, And Stra...
Ashutosh Chand Kaushal, Arjun Jaiswal, Sushil Chaurasiya, Shiva, Ajeet Sharma, Ramayan Yadav, Neha C...
Innovations In Oral Drug Administration: Exploring Gastro Retentive Raft Forming...
VIGNESH . R, Kamaleshwari B., Kaviya G., Kovarthanan M., Mohan Raj U., ...
Anti-Cancer Activity Of Cisplatin And Its Toxicity...
Pinki Chandra , Gagan Panda, Sudhir Kathane, Shruti Rathore , ...