View Article

Abstract

Pharmacovigilance is the science and activities related to detecting, assessing, understanding, and preventing adverse effects or any other drug-related problem. Established as a crucial aspect of healthcare, pharmacovigilance ensures that medicinal products' benefits outweigh their risks, thereby safeguarding patient health. This review provides a comprehensive examination of pharmacovigilance, including its historical background, key components such as adverse drug reaction (ADR) monitoring, methodologies for risk management, and the regulatory frameworks governing drug safety. Key challenges, such as underreporting and data quality issues, are also discussed, alongside recent innovations involving artificial intelligence, real-world evidence, and patient involvement in ADR reporting. Ultimately, a deeper understanding of pharmacovigilance allows healthcare systems to advance in monitoring, detecting, and mitigating risks associated with medicinal products. The review concludes with insights into the future of pharmacovigilance, highlighting the role of global harmonization and technological advancements in drug safety surveillance.

Keywords

Pharmacovigilance, Drug Safety, adverse drug reaction (ADR) monitoring

Introduction

Pharmacovigilance, defined by the World Health Organization (WHO) as “the science and activities related to the detection, assessment, understanding, and prevention of adverse effects or any other drug-related problems,” plays a vital role in ensuring drug safety across healthcare systems worldwide [1]. This discipline was initially established following significant public health crises, most notably the thalidomide tragedy in the 1960s, which underscored the need for systematic drug safety monitoring [2]. The catastrophic effects of thalidomide, primarily used as a sedative in pregnant women, led to thousands of congenital disabilities, ultimately sparking regulatory changes that laid the foundation for modern pharmacovigilance practices [3]. Pharmacovigilance not only focuses on adverse drug reactions (ADRs) but also encompasses broader risk management, which includes tracking, evaluating, and mitigating risks associated with medicinal products. Given the complex landscape of drug safety, pharmacovigilance now involves an interdisciplinary approach, including clinical pharmacy, regulatory science, epidemiology, and data analytics [4]. The goal is to enhance patient safety by continuously assessing the risk-benefit profile of medicinal products as they are developed, tested, approved, and made available in real-world settings [5].

Figure 1: Pharmacovigilance

With the expansion of pharmacovigilance efforts, several methodologies have been adopted, including spontaneous reporting systems, cohort and case-control studies, and automated database systems. These tools have transformed pharmacovigilance into a proactive science, helping detect drug-related risks early and informing safer clinical practices [6]. Moreover, regulatory bodies such as the U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), and the WHO have developed stringent guidelines and frameworks to oversee pharmacovigilance activities, highlighting the importance of harmonized drug safety standards across countries [7]. In recent years, the scope of pharmacovigilance has expanded, particularly with advances in digital health technologies and artificial intelligence (AI), which have increased the capacity to monitor and analyze ADRs from vast amounts of data more effectively. These advancements have enabled a shift toward real-time pharmacovigilance, allowing healthcare providers and regulatory agencies to react promptly to potential safety signals [8]. The ongoing evolution in this field underscores the necessity of integrating modern tools with established practices to create a robust pharmacovigilance system that meets contemporary drug safety challenges.

METHODOLOGY

Pharmacovigilance relies on a range of systematic methodologies designed to identify, assess, and mitigate risks associated with medicinal products. This section describes the primary methodologies used in pharmacovigilance, focusing on both traditional and emerging approaches. These methods include spontaneous reporting systems, cohort and case-control studies, automated database analyses, and the incorporation of artificial intelligence (AI) to manage large volumes of adverse drug reaction (ADR) data.

  1. Spontaneous Reporting Systems (SRS)

Spontaneous reporting is one of the earliest and most widely used methodologies in pharmacovigilance. In this approach, healthcare professionals, and sometimes patients, voluntarily report suspected ADRs to regulatory agencies [9]. National databases like the FDA’s Adverse Event Reporting System (FAERS) and the WHO’s global pharmacovigilance database, VigiBase, have significantly advanced ADR monitoring by providing a centralized repository for ADR data from around the world [10]. While spontaneous reporting is critical for early signal detection, it has inherent limitations, such as underreporting, reporting bias, and lack of causality assessment [11].

  1. Cohort and Case-Control Studies

Epidemiological studies, including cohort and case-control studies, are used in pharmacovigilance to investigate potential ADRs and evaluate causal relationships. In a cohort study, researchers follow groups of patients exposed and unexposed to a drug over time to observe the incidence of ADRs. This design is particularly valuable for identifying ADRs that occur after long-term use [12]. Conversely, case-control studies compare patients who experienced ADRs (cases) with those who did not (controls), offering insights into rare or delayed reactions [13]. Both study designs are essential in validating signals detected through spontaneous reporting systems and enhancing the reliability of pharmacovigilance data.

  1. Automated Database Systems

With advancements in electronic health records (EHRs) and digital data management, automated database systems have become invaluable in pharmacovigilance. Systems such as the FDA’s Sentinel System and the European Union’s EudraVigilance facilitate large-scale data collection and analysis of ADRs, helping to identify patterns that may not be apparent in individual case reports [14]. These systems employ data mining techniques like disproportionality analysis to detect potential safety signals, thereby enhancing the efficiency and accuracy of pharmacovigilance efforts [15]. Automated databases allow for real-time surveillance and are increasingly relied upon in post-marketing surveillance for new drugs.

  1. Signal Detection and Data Mining

Signal detection is a fundamental aspect of pharmacovigilance, involving the identification of new, unexpected ADRs from large datasets. Disproportionality analysis methods, such as the proportional reporting ratio (PRR) and Bayesian data mining, are commonly used to identify signals in pharmacovigilance databases [16]. PRR compares the observed frequency of a specific ADR with the expected frequency, highlighting potential associations that require further investigation [17]. Signal detection enables regulatory authorities to issue safety alerts or mandate further studies when necessary.

  1. Artificial Intelligence and Machine Learning

Artificial intelligence (AI) and machine learning (ML) have emerged as transformative tools in pharmacovigilance, addressing the limitations of traditional methodologies and enhancing signal detection accuracy [18]. Natural language processing (NLP) algorithms, for example, analyze large volumes of unstructured text from sources like social media, patient forums, and EHRs, expanding the scope of ADR monitoring beyond formal reporting systems [19]. AI-driven approaches can also analyze real-world evidence (RWE) from diverse populations, potentially leading to earlier detection of safety signals compared to conventional methods [20].

  1. Real-World Evidence (RWE) and Real-World Data (RWD)

Real-world evidence and real-world data are increasingly utilized in pharmacovigilance, providing insights from sources outside clinical trials, such as patient registries and claims data. RWE complements traditional pharmacovigilance approaches by capturing ADRs in more diverse and routine clinical settings [21]. For example, observational studies using RWD allow researchers to evaluate long-term drug effects, especially in-patient populations that may not have been well- represented in initial clinical trials [22]. RWE has proven particularly useful in evaluating the safety of medications under actual usage conditions, providing a fuller picture of a drug’s risk profile.

Regulatory Frameworks and Guidelines

Pharmacovigilance operates within a complex regulatory landscape shaped by national and international guidelines to ensure the safe and effective use of medicinal products. Regulatory frameworks vary globally but converge in their core principles, emphasizing patient safety, adverse drug reaction (ADR) monitoring, and proactive risk management. This section highlights the key regulatory bodies and guidelines in pharmacovigilance, including those from the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), the World Health Organization (WHO), and the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH).

  1. The U.S. Food and Drug Administration (FDA)

The FDA plays a crucial role in pharmacovigilance in the United States, primarily through its Center for Drug Evaluation and Research (CDER). CDER oversees drug safety across the product lifecycle, from pre-approval clinical trials to post-marketing surveillance. The FDA’s Adverse Event Reporting System (FAERS) is central to its pharmacovigilance efforts, collecting and analyzing ADR data to identify potential safety issues [23]. The FDA also operates the Sentinel System, a national electronic system designed to monitor the safety of FDA-regulated medical products in real-time, improving signal detection and risk assessment processes [24]. The FDA mandates that pharmaceutical companies conduct post-marketing safety studies and report any ADRs to FAERS, providing a structured approach to ongoing drug safety monitoring [25].

  1. The European Medicines Agency (EMA)

In the European Union, the European Medicines Agency (EMA) oversees pharmacovigilance activities through the Pharmacovigilance Risk Assessment Committee (PRAC). The EMA’s primary tool for ADR monitoring is EudraVigilance, a centralized database that collects ADR reports from European Union member states. EudraVigilance serves as a key resource for detecting safety signals and tracking the safety profile of drugs on the market [26]. The EMA also enforces Good Pharmacovigilance Practices (GVP), a set of guidelines covering all aspects of pharmacovigilance activities, including ADR reporting, signal management, and risk minimization strategies [27]. The EMA’s regulatory framework emphasizes transparency and public accessibility, often publishing safety communications and risk management plans (RMPs) to inform healthcare professionals and the public about emerging drug safety issues [28].

  1. World Health Organization (WHO)

The WHO collaborates with national pharmacovigilance centers and operates VigiBase, a global ADR database managed by the Uppsala Monitoring Centre in Sweden. VigiBase collects ADR reports from over 150 countries, enabling the WHO to identify international safety signals and provide guidance on emerging global drug safety concerns [29]. The WHO’s International Drug Monitoring Program promotes pharmacovigilance best practices and provides resources and training for countries developing their pharmacovigilance systems. WHO also publishes guidelines, such as the "WHO Handbook for Reporting ADRs," which outlines standard procedures for reporting and analyzing ADRs across member states [30].

  1. International Council for Harmonisation (ICH) Guidelines

The International Council for Harmonisation (ICH) has established several guidelines to harmonize pharmacovigilance practices worldwide, fostering a unified approach to drug safety. ICH E2E, for example, provides guidance on pharmacovigilance planning, addressing pre- and post-marketing safety data collection and analysis [31]. The ICH E2D guideline outlines requirements for post-approval safety data management, emphasizing the importance of expedited reporting for serious and unexpected ADRs [32]. By standardizing pharmacovigilance terminology and practices, the ICH guidelines help regulatory agencies and pharmaceutical companies align their safety monitoring efforts across different regions, promoting a consistent global approach to pharmacovigilance.

  1. Country-Specific Regulations and Practices

Beyond the frameworks established by major organizations like the FDA, EMA, and WHO, many countries have developed their own pharmacovigilance regulations tailored to local healthcare contexts. For example, Japan’s Pharmaceuticals and Medical Devices Agency (PMDA) has implemented unique guidelines for ADR reporting and risk management, reflecting the specific regulatory needs within its jurisdiction [33]. In India, the Pharmacovigilance Programme of India (PvPI), managed by the Indian Pharmacopoeia Commission, oversees ADR monitoring and reporting for the country, helping to standardize pharmacovigilance practices within a rapidly growing pharmaceutical market [34]. Country-specific regulations highlight the diversity of pharmacovigilance practices worldwide while contributing to a broader, globally harmonized pharmacovigilance network.

Challenges in Pharmacovigilance

Despite the critical role of pharmacovigilance in enhancing patient safety, the field faces significant challenges that can hinder its effectiveness. Key challenges include underreporting of adverse drug reactions (ADRs), data quality and completeness issues, limited resources in low- and middle-income countries, and difficulties in harmonizing pharmacovigilance practices across regions. This section explores these challenges in detail.

  1. Underreporting of Adverse Drug Reactions

Underreporting is a widespread issue in pharmacovigilance, affecting spontaneous reporting systems globally. Studies indicate that up to 95% of ADRs may go unreported, especially for mild or moderate reactions [35]. Several factors contribute to underreporting, including lack of awareness among healthcare providers, fear of legal repercussions, time constraints, and the perception that reporting ADRs has minimal impact [36]. Patient-driven reporting, while beneficial, also faces limitations, as patients may lack the technical knowledge to identify ADRs accurately. Underreporting weakens the effectiveness of pharmacovigilance systems, as critical safety signals may remain undetected or be identified too late.

  1. Data Quality and Completeness

In pharmacovigilance, data quality and completeness are paramount, as these factors influence the accuracy of signal detection and risk assessment. However, pharmacovigilance databases often contain incomplete, inaccurate, or duplicate entries, limiting their utility for meaningful analysis [37]. For example, missing data on dosage, timing, and patient demographics can make it difficult to establish causality between a drug and an ADR [38]. Standardizing data collection practices and improving the reporting infrastructure are essential steps to enhancing data quality. The integration of electronic health records (EHRs) has the potential to improve data completeness, but technical and regulatory barriers still prevent widespread adoption.

  1. Resource Limitations in Low- and Middle-Income Countries

Pharmacovigilance systems in low- and middle-income countries (LMICs) often lack the resources, infrastructure, and trained personnel necessary for effective drug safety monitoring. Limited funding and technology hinder these countries’ ability to establish robust pharmacovigilance programs and participate in international pharmacovigilance networks, such as the WHO Programme for International Drug Monitoring [39]. Resource constraints also affect data collection and analysis, as LMICs may rely on paper-based reporting systems rather than electronic databases, further complicating ADR monitoring efforts [40]. Addressing these disparities is critical for building a global pharmacovigilance system that ensures drug safety across diverse healthcare settings.

  1. Harmonization of Pharmacovigilance Practices

The lack of uniform pharmacovigilance regulations and practices across regions poses challenges for multinational pharmaceutical companies and regulatory bodies. For example, while the International Council for Harmonization (ICH) provides guidelines to harmonize pharmacovigilance activities, varying national regulations and reporting requirements still create complexity [41]. Inconsistencies in regulatory frameworks can delay the global sharing of safety data and impede rapid response to emerging safety signals. Efforts to standardize ADR definitions, reporting formats, and data-sharing protocols are ongoing, but achieving full harmonization remains challenging.

  1. Signal Detection in the Era of Polypharmacy

With the rise of polypharmacy particularly among elderly populations—identifying specific ADRs linked to a single drug has become increasingly complex. Drug-drug interactions complicate pharmacovigilance as they may mask, exacerbate, or create unique ADRs that are difficult to trace to a specific medication [42]. Signal detection algorithms, such as disproportionality analysis, are limited in their ability to disentangle these interactions without more advanced data and analytical methods. Addressing the challenges posed by polypharmacy requires enhanced methodologies in pharmacovigilance, including advanced data mining techniques and patient-level data integration.

  1. Integrating Real-World Evidence (RWE) into Pharmacovigilance

While real-world evidence (RWE) holds potential for improving pharmacovigilance, its integration faces challenges due to the complexity and variability of real-world data (RWD) sources. RWD from electronic health records, insurance claims, and social media lacks standardization, making it challenging to use in pharmacovigilance without significant data cleaning and validation efforts [43]. Moreover, privacy concerns and regulatory hurdles can restrict access to certain RWD sources, further complicating RWE integration [44]. Despite these challenges, RWE remains a valuable resource for monitoring drug safety in diverse populations and real-world settings.

Figure 2: Challenges in Pharmacovigilance

Future Directions in Pharmacovigilance

Pharmacovigilance is poised to undergo substantial transformation as it adapts to technological advances, increased patient involvement, and a more globalized approach to healthcare. Future directions in pharmacovigilance focus on personalized medicine, regulatory innovations and enhanced global collaboration, all aimed at improving drug safety and efficacy on a global scale. This section examines these future directions and the potential impact they may have on the field.

  1. Personalized Pharmacovigilance

Personalized pharmacovigilance is an emerging field that integrates pharmacogenomic data with traditional safety monitoring to create individualized drug safety profiles. By accounting for genetic, environmental, and lifestyle factors, personalized pharmacovigilance can help predict individual risks of adverse drug reactions (ADRs) and enable more tailored treatment options [45]. For instance, patients with genetic markers linked to hypersensitivity to certain drugs could receive alternative therapies or adjusted dosages to reduce the likelihood of ADRs. This approach requires a shift towards more patient-specific data collection and integration, potentially aided by electronic health records (EHRs) and wearable health devices that continuously monitor patient health metrics [46].

  1. Regulatory Adaptations and Agile Pharmacovigilance

To keep pace with rapid drug development and approval processes, regulatory bodies are exploring agile approaches to pharmacovigilance. Agile pharmacovigilance refers to flexible regulatory frameworks that allow for real-time monitoring and adaptive safety assessments [47]. For example, the FDA and EMA have implemented rolling reviews and conditional approvals for certain medications, particularly in response to public health emergencies like the COVID-19 pandemic [48]. Additionally, regulatory bodies are developing frameworks to accommodate novel therapies, such as gene and cell therapies, which may require unique pharmacovigilance protocols given their complex mechanisms of action and potential for long-term effects [49].

  1. Enhanced Global Collaboration

Global collaboration is essential for managing the complexities of pharmacovigilance in a world where medications are manufactured, distributed, and prescribed internationally. Future pharmacovigilance efforts are likely to emphasize shared databases, harmonized regulatorystandards, and collaborative reporting mechanisms. Organizations like the International Council for Harmonisation (ICH) and the World Health Organization (WHO) have been instrumental in standardizing pharmacovigilance practices, and their roles are expected to expand as regulatory bodies work to create a more cohesive global system [50]. Initiatives like the WHO’s VigiBase database and the EMA’s EudraVigilance database enable cross-border sharing of ADR data, improving the ability to detect safety signals globally [51].

  1. Real-World Data (RWD) Integration and Predictive Analytics

The integration of real-world data (RWD) is likely to continue reshaping pharmacovigilance by enabling a more nuanced understanding of drug effects in diverse populations. RWD sources, including EHRs, health insurance claims, and patient-reported outcomes, allow for a more comprehensive view of a drug’s safety profile over its lifecycle [52]. Predictive analytics, fueled by machine learning and artificial intelligence, can then use this data to anticipate ADRs before they occur, facilitating preventive measures [53]. The use of RWD in pharmacovigilance represents a shift towards a proactive, rather than reactive, approach to drug safety.

  1. Ethical and Privacy Considerations

As pharmacovigilance becomes increasingly data-driven, ethical and privacy considerations will play a larger role in shaping its future. Protecting patient privacy while collecting and sharing data across borders remains a challenge, as varying national privacy regulations complicate global data- sharing efforts. The use of blockchain technology is being explored as a solution for creating secure, decentralized databases that can protect patient confidentiality while allowing for transparent data sharing [54]. Additionally, there is a growing focus on the ethical implications of using AI in pharmacovigilance, particularly regarding the potential for bias in algorithms and the need for algorithmic transparency [55].

  1. Expanding Pharmacovigilance to Include Patient-Reported Outcomes and Quality of Life

Traditional pharmacovigilance focuses on detecting ADRs, but future pharmacovigilance efforts may place a greater emphasis on patient-reported outcomes (PROs) and quality of life (QoL) metrics. By tracking how medications impact patients’ daily lives, pharmacovigilance systems can gain a more holistic view of drug safety. Including PROs and QoL measures in safety assessments allows for a better understanding of the trade-offs between drug efficacy and side effects, especially for chronic and long-term therapies [56]. Patient-reported data could also be integrated with wearable technology, providing continuous monitoring and real-time feedback on drug effects.

CONCLUSION

Pharmacovigilance has evolved from a reactive process of detecting and managing adverse drug reactions (ADRs) to a more proactive and data-driven discipline. This transformation is largely fueled by technological advancements, including artificial intelligence (AI), machine learning, real-world data (RWD) integration, and the increasing involvement of patients in reporting ADRs. As pharmacovigilance becomes more personalized, the role of genomics, predictive analytics, and patient-reported outcomes will continue to grow, ensuring that drug safety monitoring evolves in alignment with the complexities of modern medicine. The future of pharmacovigilance hinges on several key developments. Personalized pharmacovigilance, which integrates genetic, environmental, and lifestyle factors, holds the promise of more accurate safety profiles and tailored treatment options. Regulatory bodies are adapting to the pace of innovation, with agile frameworks that allow for real-time monitoring and flexible approval processes for novel therapies. Moreover, enhanced global collaboration through shared databases and harmonized standards will be essential for managing the growing globalized use of medicines. In conclusion, the future of pharmacovigilance is marked by a shift towards a more proactive, data- centric, and patient-centered system. With advancements in technology, regulatory adaptations, and global cooperation, pharmacovigilance will continue to enhance drug safety and improve patient outcomes, ensuring that therapies used worldwide meet the highest standards of safety and efficacy.                                          

REFERENCE

  1. World Health Organization. (2020). The importance of pharmacovigilance: Safety monitoring of medicinal products. Geneva: WHO.
  2. Vargesson, N. (2015). Thalidomide-induced teratogenesis: History and mechanisms. Birth Defects Research Part C: Embryo Today, 105(2), 140–156.
  3. Schott, G., et al. (2017). The history of thalidomide and its current role in disease management. Drug Safety, 40(4), 245–259.
  4. Pirmohamed, M., & Park, B. K. (2019). Adverse drug reactions: back to the future. British Journal of Clinical Pharmacology, 85(1), 4-5.
  5. Edwards, I. R., & Aronson, J. K. (2020). Adverse drug reactions: Definitions, diagnosis, and management. The Lancet, 356(9237), 1255-1259.
  6. Inman, W. H. W. (2019). Spontaneous adverse reaction reporting systems and the future of pharmacovigilance. Pharmacovigilance Journal, 8(3), 234–239.
  7. European Medicines Agency. (2021). Pharmacovigilance guidelines and regulations. Retrieved from https://www.ema.europa.eu/en/documents/regulation.
  8. Liu, R., & Zhang, P. (2022). Artificial intelligence in pharmacovigilance: A new frontier. International Journal of Clinical Pharmacy, 44(5), 1208–1215.
  9. Hazell, L., & Shakir, S. A. (2021). Under-reporting of adverse drug reactions: a systematic review. *Drug Safety*, 38(10), 885-896.
  10. Bate, A., & Evans, S. J. (2020). Quantitative signal detection using spontaneous ADR reporting: Proportional reporting ratios (PRRs) and other thresholding rules. *Drug Safety*, 45(2), 55-67.
  11. Lopez-Gonzalez, E., Herdeiro, M. T., & Figueiras, A. (2019). Determinants of under-reporting of adverse drug reactions: a systematic review. *Drug Safety*, 32(1), 19-31.
  12. Rothman, K. J., Greenland, S., & Lash, T. L. (2018). Cohort studies. *Modern Epidemiology*, 3rd ed. Philadelphia: Lippincott Williams & Wilkins.
  13. Wang, X., & Chen, Y. (2021). Case-control studies in pharmacovigilance: Applications and considerations. *International Journal of Epidemiology*, 48(2), 279-290.
  14. Platt, R., Wilson, M., & Chan, K. A. (2021). The Sentinel System: Early successes and challenges for an innovative national medical product surveillance system. *Pharmacoepidemiology and Drug Safety*, 29(6), 663-671.
  15. Norén, G. N., & Hopstadius, J. (2022). Disproportionality analysis in pharmacovigilance: empirical approaches. *Pharmacovigilance Journal*, 9(3), 189-202.
  16. Hauben, M., & Bate, A. (2021). Data mining in pharmacovigilance. *International Journal of Clinical Pharmacy*, 43(3), 353-361.
  17. Almenoff, J. S., & Tonning, J. M. (2019). Bayesian data mining and its applications to pharmacovigilance. *Drug Safety*, 30(7), 635-645.
  18. Ball, R., & Robb, M. (2020). AI in pharmacovigilance: Enhancing detection and response. *Drug Safety*, 43(1), 103-115.
  19. Xu, X., & Jackson, R. (2022). Natural language processing applications in pharmacovigilance. *Journal of Biomedical Informatics*, 76, 100-109.
  20. Kesselheim, A. S., et al. (2021). Real-world evidence in regulatory decision-making for drugs and devices. *New England Journal of Medicine*, 377(23), 2293-2297.
  21. Schneeweiss, S. (2020). Real-world evidence and the new FDA framework: What it means for the development and safety of new drugs. *Journal of Clinical Epidemiology*, 100(3), 22-27.
  22. Sherman, R. E., & Anderson, S. A. (2022). Real-world evidence: expanding the horizon in drug safety evaluation. *Pharmacovigilance Journal*, 11(4), 412-425.
  23. U.S. Food and Drug Administration. (2021). FDA Adverse Event Reporting System (FAERS). Retrieved from https://www.fda.gov/drugs.
  24. Platt, R., Wilson, M., & Chan, K. A. (2021). The Sentinel System: Early successes and challenges for an innovative national medical product surveillance system. *Pharmacoepidemiology and Drug Safety*, 29(6), 663-671.
  25. Sultana, J., Cutroneo, P., & Trifirò, G. (2022). Clinical and regulatory impact of pharmacovigilance on drug safety: A review. *European Journal of Clinical Pharmacology*, 78(7), 1125-1134.
  26. European Medicines Agency. (2022). EudraVigilance and adverse drug reaction reporting. Retrieved from https://www.ema.europa.eu/en.
  27. European Medicines Agency. (2021). Good Pharmacovigilance Practices (GVP). Retrieved from https://www.ema.europa.eu/en.
  28. Härmark, L., & van Grootheest, A. C. (2019). Pharmacovigilance: Methods, recent developments, and future perspectives. *European Journal of Clinical Pharmacology*, 65(2), 95- 103.
  29. Lindquist, M. (2020). VigiBase, the WHO global ICSR database system: Basic facts. *Drug Information Journal*, 42(5), 409-419.
  30. World Health Organization. (2020). WHO Handbook for Reporting ADRs. Geneva: WHO Press.
  31. International Council for Harmonisation. (2022). ICH E2E: Pharmacovigilance Planning. Retrieved from https://www.ich.org.
  32. International Council for Harmonisation. (2020). ICH E2D: Post-Approval Safety Data Management. Retrieved from https://www.ich.org.
  33. Pharmaceuticals and Medical Devices Agency. (2021). Pharmacovigilance in Japan: PMDA’s Approach to ADRs. Retrieved from https://www.pmda.go.jp.
  34. Kumar, R., & Bhowmik, D. (2021). Pharmacovigilance in India: Current scenario and future perspectives. *Indian Journal of Pharmacology*, 53(1), 5-9.
  35. Hazell, L., & Shakir, S. A. (2021). Under-reporting of adverse drug reactions: A systematic review. *Drug Safety*, 38(10), 885-896.
  36. Lopez-Gonzalez, E., Herdeiro, M. T., & Figueiras, A. (2019). Determinants of under-reporting of adverse drug reactions: A systematic review. *Drug Safety*, 32(1), 19-31.
  37. Härmark, L., & van Grootheest, A. C. (2019). Quality and completeness of adverse drug reaction reports: A review of key challenges. *European Journal of Clinical Pharmacology*, 75(2), 151-160.
  38. Bate, A., & Evans, S. J. (2020). Quantitative signal detection using spontaneous ADR reporting: Proportional reporting ratios (PRRs) and other thresholding rules. *Drug Safety*, 45(2), 55-67.
  39. Olsson, S. (2020). The role of the WHO Programme for International Drug Monitoring in supporting pharmacovigilance in low-income countries. *Drug Safety*, 29(5), 421-426.
  40. Elshafie, S., et al. (2022). Pharmacovigilance challenges in low- and middle-income countries: A narrative review. *BMC Health Services Research*, 22, 87.
  41. International Council for Harmonisation. (2021). ICH E2E: Pharmacovigilance planning and harmonization. Retrieved from https://www.ich.org.
  42. Vande Griend, J. P., et al. (2021). Polypharmacy and drug-drug interactions in pharmacovigilance. *Journal of Clinical Pharmacology*, 50(9), 1170-1182.
  43. Schneeweiss, S., & Avorn, J. (2020). The promise and challenges of real-world evidence in drug safety assessment. *Journal of Clinical Epidemiology*, 100(4), 224-230.
  44. Kesselheim, A. S., et al. (2021). Real-world data in regulatory decision-making for drug safety. *New England Journal of Medicine*, 377(23), 2293-2297
  45. Roden, D. M., et al. (2020). Pharmacogenomics and the future of personalized medicine in pharmacovigilance. *Journal of Clinical Investigation*, 130(3), 1205-1212.
  46. Topol, E. J. (2019). The role of wearable technology in pharmacovigilance and personalized healthcare. *Nature Biotechnology*, 37(1), 1-4.
  47. Eichler, H. G., et al. (2021). Agile pharmacovigilance: Adapting regulatory practices for rapid response. *Regulatory Toxicology and Pharmacology*, 125, 104931.
  48. Alexander, G. C., et al. (2021). The role of real-time monitoring and adaptive safety in COVID- 19 vaccine pharmacovigilance. *JAMA*, 324(6), 599-600.
  49. Wirth, T., et al. (2022). Pharmacovigilance for advanced therapies: Challenges and regulatory adaptations. *Molecular Therapy*, 30(2), 546-552.
  50. Bakker, A., et al. (2020). Harmonization of pharmacovigilance regulations: The role of the ICH and WHO. *Drug Safety*, 43(7), 611-618.
  51. Olsson, S., & Pal, S. N. (2019). The VigiBase initiative: Enhancing global pharmacovigilance through data-sharing. *Drug Safety*, 42(6), 677-685.
  52. Schneeweiss, S., & Avorn, J. (2020). Real-world data in drug safety monitoring: Emerging trends and ethical considerations. *BMJ*, 368, m161.
  53. Laporte, J. R., & Carné, X. (2021). The future of predictive analytics in pharmacovigilance: Toward proactive drug safety. *Drug Safety*, 44(5), 467-473.
  54. Benchoufi, M., & Ravaud, P. (2021). Blockchain technology for improving transparency and privacy in pharmacovigilance. *Blockchain in Healthcare Today*, 3, e205.
  55. Anderson, C. J., & Ratna, S. (2020). AI ethics in pharmacovigilance: Addressing algorithmic bias and transparency. *Pharmacovigilance Review*, 45(3), 215-228.
  56. Basch, E., et al. (2021). Patient-reported outcomes and quality of life in pharmacovigilance: Expanding the scope of drug safety assessment. *Clinical Pharmacology & Therapeutics*, 109(4), 856-862.

Reference

  1. World Health Organization. (2020). The importance of pharmacovigilance: Safety monitoring of medicinal products. Geneva: WHO.
  2. Vargesson, N. (2015). Thalidomide-induced teratogenesis: History and mechanisms. Birth Defects Research Part C: Embryo Today, 105(2), 140–156.
  3. Schott, G., et al. (2017). The history of thalidomide and its current role in disease management. Drug Safety, 40(4), 245–259.
  4. Pirmohamed, M., & Park, B. K. (2019). Adverse drug reactions: back to the future. British Journal of Clinical Pharmacology, 85(1), 4-5.
  5. Edwards, I. R., & Aronson, J. K. (2020). Adverse drug reactions: Definitions, diagnosis, and management. The Lancet, 356(9237), 1255-1259.
  6. Inman, W. H. W. (2019). Spontaneous adverse reaction reporting systems and the future of pharmacovigilance. Pharmacovigilance Journal, 8(3), 234–239.
  7. European Medicines Agency. (2021). Pharmacovigilance guidelines and regulations. Retrieved from https://www.ema.europa.eu/en/documents/regulation.
  8. Liu, R., & Zhang, P. (2022). Artificial intelligence in pharmacovigilance: A new frontier. International Journal of Clinical Pharmacy, 44(5), 1208–1215.
  9. Hazell, L., & Shakir, S. A. (2021). Under-reporting of adverse drug reactions: a systematic review. *Drug Safety*, 38(10), 885-896.
  10. Bate, A., & Evans, S. J. (2020). Quantitative signal detection using spontaneous ADR reporting: Proportional reporting ratios (PRRs) and other thresholding rules. *Drug Safety*, 45(2), 55-67.
  11. Lopez-Gonzalez, E., Herdeiro, M. T., & Figueiras, A. (2019). Determinants of under-reporting of adverse drug reactions: a systematic review. *Drug Safety*, 32(1), 19-31.
  12. Rothman, K. J., Greenland, S., & Lash, T. L. (2018). Cohort studies. *Modern Epidemiology*, 3rd ed. Philadelphia: Lippincott Williams & Wilkins.
  13. Wang, X., & Chen, Y. (2021). Case-control studies in pharmacovigilance: Applications and considerations. *International Journal of Epidemiology*, 48(2), 279-290.
  14. Platt, R., Wilson, M., & Chan, K. A. (2021). The Sentinel System: Early successes and challenges for an innovative national medical product surveillance system. *Pharmacoepidemiology and Drug Safety*, 29(6), 663-671.
  15. Norén, G. N., & Hopstadius, J. (2022). Disproportionality analysis in pharmacovigilance: empirical approaches. *Pharmacovigilance Journal*, 9(3), 189-202.
  16. Hauben, M., & Bate, A. (2021). Data mining in pharmacovigilance. *International Journal of Clinical Pharmacy*, 43(3), 353-361.
  17. Almenoff, J. S., & Tonning, J. M. (2019). Bayesian data mining and its applications to pharmacovigilance. *Drug Safety*, 30(7), 635-645.
  18. Ball, R., & Robb, M. (2020). AI in pharmacovigilance: Enhancing detection and response. *Drug Safety*, 43(1), 103-115.
  19. Xu, X., & Jackson, R. (2022). Natural language processing applications in pharmacovigilance. *Journal of Biomedical Informatics*, 76, 100-109.
  20. Kesselheim, A. S., et al. (2021). Real-world evidence in regulatory decision-making for drugs and devices. *New England Journal of Medicine*, 377(23), 2293-2297.
  21. Schneeweiss, S. (2020). Real-world evidence and the new FDA framework: What it means for the development and safety of new drugs. *Journal of Clinical Epidemiology*, 100(3), 22-27.
  22. Sherman, R. E., & Anderson, S. A. (2022). Real-world evidence: expanding the horizon in drug safety evaluation. *Pharmacovigilance Journal*, 11(4), 412-425.
  23. U.S. Food and Drug Administration. (2021). FDA Adverse Event Reporting System (FAERS). Retrieved from https://www.fda.gov/drugs.
  24. Platt, R., Wilson, M., & Chan, K. A. (2021). The Sentinel System: Early successes and challenges for an innovative national medical product surveillance system. *Pharmacoepidemiology and Drug Safety*, 29(6), 663-671.
  25. Sultana, J., Cutroneo, P., & Trifirò, G. (2022). Clinical and regulatory impact of pharmacovigilance on drug safety: A review. *European Journal of Clinical Pharmacology*, 78(7), 1125-1134.
  26. European Medicines Agency. (2022). EudraVigilance and adverse drug reaction reporting. Retrieved from https://www.ema.europa.eu/en.
  27. European Medicines Agency. (2021). Good Pharmacovigilance Practices (GVP). Retrieved from https://www.ema.europa.eu/en.
  28. Härmark, L., & van Grootheest, A. C. (2019). Pharmacovigilance: Methods, recent developments, and future perspectives. *European Journal of Clinical Pharmacology*, 65(2), 95- 103.
  29. Lindquist, M. (2020). VigiBase, the WHO global ICSR database system: Basic facts. *Drug Information Journal*, 42(5), 409-419.
  30. World Health Organization. (2020). WHO Handbook for Reporting ADRs. Geneva: WHO Press.
  31. International Council for Harmonisation. (2022). ICH E2E: Pharmacovigilance Planning. Retrieved from https://www.ich.org.
  32. International Council for Harmonisation. (2020). ICH E2D: Post-Approval Safety Data Management. Retrieved from https://www.ich.org.
  33. Pharmaceuticals and Medical Devices Agency. (2021). Pharmacovigilance in Japan: PMDA’s Approach to ADRs. Retrieved from https://www.pmda.go.jp.
  34. Kumar, R., & Bhowmik, D. (2021). Pharmacovigilance in India: Current scenario and future perspectives. *Indian Journal of Pharmacology*, 53(1), 5-9.
  35. Hazell, L., & Shakir, S. A. (2021). Under-reporting of adverse drug reactions: A systematic review. *Drug Safety*, 38(10), 885-896.
  36. Lopez-Gonzalez, E., Herdeiro, M. T., & Figueiras, A. (2019). Determinants of under-reporting of adverse drug reactions: A systematic review. *Drug Safety*, 32(1), 19-31.
  37. Härmark, L., & van Grootheest, A. C. (2019). Quality and completeness of adverse drug reaction reports: A review of key challenges. *European Journal of Clinical Pharmacology*, 75(2), 151-160.
  38. Bate, A., & Evans, S. J. (2020). Quantitative signal detection using spontaneous ADR reporting: Proportional reporting ratios (PRRs) and other thresholding rules. *Drug Safety*, 45(2), 55-67.
  39. Olsson, S. (2020). The role of the WHO Programme for International Drug Monitoring in supporting pharmacovigilance in low-income countries. *Drug Safety*, 29(5), 421-426.
  40. Elshafie, S., et al. (2022). Pharmacovigilance challenges in low- and middle-income countries: A narrative review. *BMC Health Services Research*, 22, 87.
  41. International Council for Harmonisation. (2021). ICH E2E: Pharmacovigilance planning and harmonization. Retrieved from https://www.ich.org.
  42. Vande Griend, J. P., et al. (2021). Polypharmacy and drug-drug interactions in pharmacovigilance. *Journal of Clinical Pharmacology*, 50(9), 1170-1182.
  43. Schneeweiss, S., & Avorn, J. (2020). The promise and challenges of real-world evidence in drug safety assessment. *Journal of Clinical Epidemiology*, 100(4), 224-230.
  44. Kesselheim, A. S., et al. (2021). Real-world data in regulatory decision-making for drug safety. *New England Journal of Medicine*, 377(23), 2293-2297
  45. Roden, D. M., et al. (2020). Pharmacogenomics and the future of personalized medicine in pharmacovigilance. *Journal of Clinical Investigation*, 130(3), 1205-1212.
  46. Topol, E. J. (2019). The role of wearable technology in pharmacovigilance and personalized healthcare. *Nature Biotechnology*, 37(1), 1-4.
  47. Eichler, H. G., et al. (2021). Agile pharmacovigilance: Adapting regulatory practices for rapid response. *Regulatory Toxicology and Pharmacology*, 125, 104931.
  48. Alexander, G. C., et al. (2021). The role of real-time monitoring and adaptive safety in COVID- 19 vaccine pharmacovigilance. *JAMA*, 324(6), 599-600.
  49. Wirth, T., et al. (2022). Pharmacovigilance for advanced therapies: Challenges and regulatory adaptations. *Molecular Therapy*, 30(2), 546-552.
  50. Bakker, A., et al. (2020). Harmonization of pharmacovigilance regulations: The role of the ICH and WHO. *Drug Safety*, 43(7), 611-618.
  51. Olsson, S., & Pal, S. N. (2019). The VigiBase initiative: Enhancing global pharmacovigilance through data-sharing. *Drug Safety*, 42(6), 677-685.
  52. Schneeweiss, S., & Avorn, J. (2020). Real-world data in drug safety monitoring: Emerging trends and ethical considerations. *BMJ*, 368, m161.
  53. Laporte, J. R., & Carné, X. (2021). The future of predictive analytics in pharmacovigilance: Toward proactive drug safety. *Drug Safety*, 44(5), 467-473.
  54. Benchoufi, M., & Ravaud, P. (2021). Blockchain technology for improving transparency and privacy in pharmacovigilance. *Blockchain in Healthcare Today*, 3, e205.
  55. Anderson, C. J., & Ratna, S. (2020). AI ethics in pharmacovigilance: Addressing algorithmic bias and transparency. *Pharmacovigilance Review*, 45(3), 215-228.
  56. Basch, E., et al. (2021). Patient-reported outcomes and quality of life in pharmacovigilance: Expanding the scope of drug safety assessment. *Clinical Pharmacology & Therapeutics*, 109(4), 856-862.

Photo
Akanksha Punekar
Corresponding author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Photo
Nikhil Sandhan
Co-author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Photo
Om Pawar
Co-author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Photo
Suraj Pathak
Co-author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Photo
Saurabh Tribhuvan
Co-author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Photo
Dnyaneshwar Mogare
Co-author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Photo
Onkar Shepal
Co-author

Department of Pharmacy, JES’s SND College of Pharmacy, Babhulgaon (Yeola), India

Akanksha Punekar*, Nikhil Sandhan, Om Pawar, Suraj Pathak, Saurabh Tribhuvan, Dnyaneshwar Mogare, Onkar Shepal, The Role of Pharmacovigilance in Drug Safety, Int. J. Sci. R. Tech., 2025, 2 (11), 273-282. https://doi.org/10.5281/zenodo.17563807

More related articles
Radiological Evaluation of Sternal Fusion Pattern ...
Vivek Kumar Yadav, Shivam Kumar, ...
A Comparative Analysis of The Economic Effects of ...
S. Swetha, Sahayaraj Antony Michael, ...
Therapeutic Advance in Transdermal Drug Delivery Systems, Enhancing Efficiency T...
Rushikesh Narode, Vikram Saruk, Manoj Garad, Parvani Wani, ...
Mechanism of Action and Clinical Use of Remdesivir in the Treatment of COVID-19...
Amol Jadhav, Maaz Aquil, Girish Gore, Nishant Gite, Parth Khandelwal, ...
Related Articles
Pharmacovigilance in Methotrexate Therapy: A Review...
Akanksha Punekar, Om Pawar, Nikhil Sandhan, Suraj Pathak, Saurabh Tribhuvan, Onkar Shepal, Prashant ...
The Role of Parijatak and Bael Patra in Polyherbal Cough Formulations...
Vaishnavi Kanade, Maaz Aaquil, Achal Bagore , Sakshi Jadhav, Sakshi Kadam, ...
Effect of Adaptogens on the Central Nervous System and The Molecular Mechanism A...
Pooja Rasal, Saurabh Tribhuvan, Suraj Pathak, Prashant Pawar, Nikhil Sandhan, Om Pawar, ...
AI-Driven Approaches to Enhance Pharmacovigilance...
Khairnar Hrutuja , Lahare Chaitali, Kherud Rohan, Ippar Ashwini, Kotame Vikrant, Pansare Shwetali, ...