Fact-checked by Grok 2 weeks ago

Adverse event

An adverse event (AE) is defined as any untoward medical occurrence associated with the use of a or medical product in humans, whether or not considered drug-related, encompassing any unfavorable sign, symptom, or temporally linked to the intervention without requiring proof of . This broad definition, established by regulatory bodies such as the FDA and , distinguishes AEs from adverse reactions, which imply a reasonable causal association with the product. In clinical trials and , AEs are systematically collected to monitor safety, with serious adverse events (SAEs)—those resulting in death, life-threatening conditions, hospitalization, or significant —demanding expedited reporting to regulatory authorities. AE reporting forms the backbone of post-marketing surveillance systems like the FDA's FAERS database, which aggregates voluntary reports to detect potential safety signals amid background noise of coincidental events, though limitations such as underreporting and lack of denominator data necessitate cautious interpretation grounded in epidemiological analysis rather than raw counts. In clinical trials, mandatory AE documentation ensures ongoing risk-benefit assessments, enabling protocol amendments or trial halts if harms outweigh efficacy, as evidenced by historical precedents where AE signals prompted regulatory actions. Despite standardized guidelines from bodies like ICH, challenges persist in harmonizing global reporting, verifying causality via methods such as disproportionality analysis, and addressing biases in data submission that may underrepresent rare events or inflate temporal associations. This framework underscores AE monitoring's role in causal realism, prioritizing empirical patterns over presumed intent while safeguarding through transparent, data-driven scrutiny.

Definition and Conceptual Foundations

Core Definition and Scope

An adverse event (AE) is defined as any untoward medical occurrence in a or subject administered a pharmaceutical product, which does not necessarily have a causal relationship with the treatment. This encompasses any unfavorable and unintended sign—such as an abnormal laboratory finding—symptom, or disease temporally associated with the use of a medicinal product, regardless of suspected . The U.S. (FDA) similarly describes it as any undesirable experience associated with the use of a medical product, emphasizing its occurrence in a without requiring proof of causation. This definition originates from international harmonized guidelines, such as those from the International Council for Harmonisation (ICH), to standardize reporting across regulatory frameworks. The scope of adverse events extends beyond pharmaceuticals to include vaccines, medical devices, and biological products, capturing events during clinical trials, post-marketing surveillance, and routine healthcare delivery. In clinical investigations, AEs are monitored prospectively to assess safety profiles, with mandatory reporting thresholds established by regulators like the FDA under 21 CFR 312 for investigational new drugs. Post-approval, systems such as the FDA's Adverse Event Reporting System (FAERS) aggregate voluntary and mandatory reports to detect signals of potential harm, though underreporting remains a known limitation due to reliance on healthcare providers and patients. Pharmacovigilance programs, as defined by the (WHO), integrate AE detection into broader activities for preventing adverse effects or any other drug-related problems, applying globally to ensure ongoing risk-benefit evaluation. Key to the concept is its temporal association criterion, which broadens scope to include coincidental events, distinguishing AEs from adverse reactions that imply . This non-causal threshold facilitates comprehensive but necessitates subsequent to differentiate true risks, as over 90% of reported AEs in some databases lack confirmed relatedness upon review. Regulatory bodies mandate AE reporting within specified timelines—e.g., 15 days for serious events to the FDA—to enable signal detection, though the broad definition can inflate volumes and challenge in . An adverse event is defined as any unfavorable and unintended sign, symptom, disease, or abnormal laboratory finding temporally associated with the use of a medicinal product, without necessitating a causal relationship to the treatment. This broad scope contrasts with an , which specifically denotes a noxious and unintended response to a at normal doses where causality is reasonably suspected or established, as per pharmacovigilance criteria. Adverse events thus serve as a surveillance net capturing potential signals for further investigation, while adverse drug reactions imply a direct pharmacological link requiring attribution assessment. The term , often conflated in lay usage, typically describes predictable secondary pharmacological actions of a —such as drowsiness from antihistamines—that are known, dose-related, and may not qualify as harmful, distinguishing them from the potentially serious or unexpected outcomes encompassed by adverse events. Adverse drug events further broaden this by including not only reactions but also injuries from dosing errors, misuse, or overdoses, emphasizing harm from any aspect of drug administration rather than mere temporal coincidence. In procedural contexts, adverse events differ from complications, which denote secondary conditions or injuries arising directly from a medical intervention or progression, often with implied and potential preventability through technique refinement. Complications may be anticipated risks inherent to the procedure, whereas adverse events maintain neutrality regarding origin, capturing both treatment-related and coincidental occurrences. errors, by contrast, focus on process failures—such as incorrect dosing or misdiagnosis—that can precipitate adverse events but represent lapses in execution rather than the resultant harm itself. This distinction underscores that not all adverse events stem from errors, nor do all errors yield reportable events.

Classification Schemes

Types by Nature and Predictability

Adverse events are classified by nature into those stemming from predictable pharmacological or physiological extensions of a treatment's intended effects and those arising from idiosyncratic or mechanisms unrelated to dose or primary action. This distinction aligns with the predominant Type A and Type B categorization, originally developed for adverse drug reactions but extended to broader adverse events in clinical settings. Type A events, comprising approximately 80% of reported cases, are augmented responses directly tied to a drug's or intervention's known properties, such as exaggerated therapeutic effects leading to toxicity. Type A events are highly predictable based on dose, patient factors like age or renal function, and drug , allowing for risk mitigation through dose adjustments or . For instance, opioid-induced respiratory exemplifies a Type A event, foreseeable from the drug's depressant action and proportional to dosage. These events often occur in susceptible populations, such as the elderly, where altered metabolism amplifies effects, and their predictability facilitates preventive strategies like . In contrast, Type B events exhibit a non-dose-dependent nature, often involving immune-mediated or genetic predispositions, rendering them unpredictable from standard pharmacological profiles. These account for 10-15% of adverse events and include from penicillin in sensitized individuals or from , which cannot be anticipated from preclinical data or dosing alone. Their rarity and host-specific triggers, such as HLA variants, underscore challenges in pre-market detection, though post-marketing surveillance has identified patterns in certain cohorts. Extended classifications incorporate additional types for (Type C), delayed (Type D, e.g., teratogenicity), withdrawal-related (Type E), or therapeutic (Type F) events, which blend elements of predictability based on duration of exposure or cessation. However, these remain secondary to the core A/B , which emphasizes causal by linking to mechanistic predictability rather than assuming uniform risk across interventions. Empirical data from databases, such as those analyzed in large-scale reviews, confirm Type A's dominance in frequency and preventability, while Type B's unpredictability drives ongoing genetic screening efforts to enhance foresight where possible.

Severity and Seriousness Criteria

Seriousness of an adverse event is determined by its potential impact on patient health and function, independent of intensity, and serves primarily as a for expedited regulatory reporting. According to the International Council for Harmonisation (ICH) E2A guideline, a serious adverse event is defined as any untoward medical occurrence that at any dose results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant /incapacity; or is a congenital anomaly/. The U.S. (FDA) aligns with this, adding "other serious important medical events" that may jeopardize the patient or require medical intervention to prevent one of the listed outcomes, such as allergic requiring emergency treatment. Seriousness is assessed as a —serious or non-serious—focusing on clinical outcomes rather than subjective intensity, with regulatory agencies emphasizing that events like attempts or qualify even if not immediately fatal. In contrast, severity evaluates the degree or intensity of the adverse event's symptoms or effects, often using standardized grading scales to quantify clinical impact for monitoring and analysis. The National Cancer Institute's Common Terminology Criteria for Adverse Events (CTCAE) provides a widely adopted five-grade system: Grade 1 (mild; or mild symptoms, no intervention needed); Grade 2 (moderate; minimal intervention required, limiting age-appropriate instrumental ); Grade 3 (severe or medically significant but not immediately life-threatening; limiting , often requiring hospitalization); Grade 4 (life-threatening consequences; urgent intervention indicated); and Grade 5 (death related to the event). Other systems, such as the Division of AIDS (DAIDS) Toxicity Grading Tables, similarly grade events across organ systems (e.g., hematologic, neurologic) based on objective measures like laboratory values or symptom duration, with severity escalating from mild (Grade 1) to life-threatening (Grade 4). These scales facilitate consistent in clinical trials but require clinician judgment for subjective symptoms. The distinction between seriousness and severity is critical to avoid , as a severe (high-intensity symptoms) may not meet seriousness criteria if it resolves without major sequelae, such as a severe but transient , whereas a non-severe like a hospitalization for a potential allergic qualifies as serious. ICH guidelines explicitly note that "serious" and "severe" are not synonymous, with guiding timelines (e.g., 15-day expedited reports for serious s) while severity informs risk-benefit evaluations and dose adjustments. In practice, both assessments are performed concurrently during , with prioritized for immediate action and severity for longitudinal tracking of patterns across populations.

Preventability and Attribution Categories

Adverse events are classified by preventability to differentiate those stemming from modifiable factors in care delivery from inherent treatment risks, enabling targeted interventions to reduce harm. The primary categories include preventable, ameliorable, and non-preventable events. Preventable adverse events result from errors or failures to follow evidence-based practices, such as prescribing contraindicated medications, incorrect dosing, or inadequate monitoring, accounting for approximately 28% of inpatient adverse drug events in systematic reviews of pediatric populations. Ameliorable events occur without initial prevention but could have been substantially lessened through vigilant response, like prompt recognition of symptoms leading to dose adjustment. Non-preventable events reflect unavoidable pharmacological or physiological responses despite adherence to optimal protocols, often comprising the majority of adverse drug reactions in controlled settings. Assessing preventability typically employs structured criteria, such as the Schumock-Thornton scale adapted for adverse events, which evaluates factors like inappropriate selection or lack of documentation to deem events "definitely preventable," "probably preventable," or "not preventable." Empirical data from studies show preventability rates varying by method, with chart reviews identifying up to 50% preventable events compared to lower estimates from spontaneous reports, highlighting methodological inconsistencies that may underestimate system failures. In hospitalized older adults, preventable events frequently involved disturbances or from anticoagulants and analgesics, underscoring classes prone to error-related harm. Attribution categories for adverse events focus on linking harm to specific causal pathways, distinct from overall assessments, to inform prevention strategies. These often delineate types: prescribing errors (e.g., wrong or dose), dispensing errors (e.g., incorrect labeling), administration errors (e.g., improper route), and monitoring failures (e.g., overlooked lab abnormalities). Patient-related attributions include non-adherence or comorbidities exacerbating risks, while system-level factors like inadequate staffing contribute indirectly. In analyses, attribution to errors versus intrinsic adverse reactions separates preventable iatrogenic harm—estimated at 50% of adverse events—from non-modifiable outcomes, with antibiotics and cardiovascular agents commonly implicated in attributable cases. Such categorizations reveal that physician-related decisions account for over 40% of preventable attributions in some cohorts, emphasizing gaps over patient blame.
Preventability CategoryDescriptionExample Attribution FactorsEstimated Prevalence in Inpatient Settings
PreventableDue to deviation from standardsPrescribing error, lack of 20-50% of adverse events
AmeliorableHarm reducible post-onsetDelayed response10-20% overlapping with preventable
Non-preventableInherent to despite optimal carePredictable at therapeutic dose50-80% of events
These classifications, while aiding quality improvement, face challenges from subjective interpretation, with varying by 20-30% across evaluators, necessitating standardized tools for consistent application.

Causality Assessment

Methodologies and Scales

Causality assessment methodologies for adverse events, particularly adverse drug reactions (ADRs), employ structured tools to evaluate the plausibility of a causal link between a suspect agent and the observed outcome, incorporating factors such as temporal , dechallenge ( upon discontinuation), rechallenge (recurrence upon re-exposure), explanations, and biological plausibility. These approaches aim to standardize judgments that are inherently subjective due to variables like comorbidities and , though no method achieves perfect objectivity or inter-rater consistency. Probabilistic scales assign numerical scores, while categorical systems rely on qualitative descriptors, with the choice often depending on such as clinical trials or post-marketing . The Adverse Drug Reaction Probability Scale, developed in 1981, is one of the most widely adopted tools, featuring a 10-item that scores responses from -1 to +2 across criteria including prior reports of the reaction, temporal fit, and response. Total scores categorize as definite (≥9 points), probable (5-8 points), possible (1-4 points), or doubtful (≤0 points), facilitating quantitative estimation in . Despite its simplicity and broad application, the scale exhibits moderate (kappa values around 0.69-0.86) and limitations in handling complex cases involving multiple drugs or rare events. The World Health Organization-Uppsala Monitoring Centre (WHO-UMC) system provides a non-numerical, expert-driven framework with six categories: certain (undeniable link via rechallenge and exclusion of alternatives), probable/likely (reasonable temporal fit and incompatibility with underlying disease), possible (plausible but with alternatives), unlikely (temporal mismatch or stronger alternatives), conditional/unclassified (pending further data), and unassessable (insufficient information). This emphasizes clinical-pharmacological context and documentation quality, making it suitable for global reporting databases, though it requires trained assessors and can yield variable results across evaluators. The Liverpool Causality Assessment Tool (LCAT), introduced in the early 2010s, uses a decision-tree flowchart with binary yes/no questions on elements like previous similar reports, dose-response relationships, and confounding factors, yielding categories of definite, probable, possible, or unlikely. Designed for improved reproducibility, LCAT demonstrates higher inter-rater agreement compared to Naranjo in validation studies (e.g., kappa >0.8 in some cohorts), particularly for pediatric and observational data, but remains less disseminated than older scales. Comparative analyses reveal only fair to moderate concordance between LCAT, Naranjo, and WHO-UMC (kappa 0.2-0.6), underscoring the absence of a universal gold standard and the need for context-specific selection.

Empirical Challenges and Limitations

Assessing for adverse events empirically is hindered by the nonspecific manifestations of most reactions, which overlap with symptoms of underlying conditions, infections, or unrelated comorbidities, making of - or intervention-specific effects challenging without controlled experimentation. Diagnostic tests to confirm are typically unavailable, and deliberate rechallenge—the gold standard for in controlled settings—is ethically prohibited in clinical practice due to potential harm. These constraints shift reliance to observational data from case reports, where temporal proximity to exposure serves as a but fails to disentangle correlation from causation amid variables like , patient heterogeneity, and background incidence rates of similar events. Structured tools such as the and WHO-UMC causality scale aim to standardize evaluation through probabilistic scoring, yet they demonstrate empirical limitations including subjective interpretation of criteria like "previous conclusive reports" or "alternative causes," leading to inconsistent outcomes. Inter-rater agreement across these methods is moderate at best, with coefficients frequently ranging from 0.29 to 0.51 in comparative studies, reflecting fair reliability but substantial variability among assessors. The scale, in particular, shows low sensitivity for detecting probable or possible cases while prioritizing specificity, potentially underestimating weaker signals in rare events. Validation of these scales lacks a robust empirical foundation, as no universal gold standard exists against which to benchmark accuracy, resulting in assessments that often devolve to expert judgment rather than reproducible, data-driven inference. Pharmacovigilance databases compound these issues with incomplete reporting, missing confounders, and selection biases, where only suspected events are captured, skewing toward positive associations without denominator data on non-events. Statistical challenges, such as low event rates necessitating large populations for signal detection, further limit causal inference, as disproportionality analyses (e.g., reporting odds ratios) cannot fully adjust for exposure variability or latency periods. Overall, these empirical barriers underscore the tentative nature of causality attributions, emphasizing the need for complementary approaches like large-scale cohort studies or Bayesian methods to enhance evidentiary rigor.

Grading and Quantification

Standard Grading Systems

Standard grading systems for adverse events provide standardized criteria to assess severity, facilitating consistent reporting, comparison across studies, and clinical decision-making in and clinical trials. The most widely adopted system is the Common Terminology Criteria for Adverse Events (CTCAE), developed by the U.S. (NCI), which categorizes events into five grades based on clinical impact, required interventions, and outcomes. Originally focused on trials, CTCAE has been extended for broader use in interventional studies, offering detailed descriptors for over 800 adverse event terms across organ systems. Version 5.0, released in 2017, remains the reference standard, with updates like version 6.0 in 2025 incorporating refinements for emerging toxicities. CTCAE grades are defined as follows:
GradeDescription
1Mild; asymptomatic or mild symptoms; clinical or diagnostic observations only; not indicated.
2Moderate; minimal, local, or noninvasive indicated; limiting age-appropriate instrumental (ADL).
3Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self-care ADL.
4Life-threatening consequences; urgent indicated.
5Death related to the adverse event.
This scale emphasizes objective criteria, such as laboratory thresholds (e.g., 3 as <8.0 g/dL) and functional , to minimize subjectivity. In non-oncology contexts, adaptations exist, such as the Division of AIDS (DAIDS) Toxicity Grading Table, which aligns with CTCAE but tailors parameters for infectious disease trials, using similar 1-5 scales for parameters like or liver enzymes. For healthy volunteer studies, the FDA employs a 1-4 scale in its Toxicity Grading Scale, excluding grade 5 to reflect lower expected risks, with grade 1 as transient mild discomfort and grade 4 as life-threatening. These systems enhance data comparability but require training for reliable application, as inter-rater variability can occur for subjective symptoms like . Regulatory bodies like the FDA and mandate their use in trial protocols to standardize safety assessments.

Application in Clinical and Research Contexts

In clinical practice, standardized grading systems such as the Common Terminology Criteria for Adverse Events (CTCAE), developed by the , enable clinicians to quantify the severity of adverse events through objective criteria encompassing symptoms, laboratory values, and . This facilitates real-time patient monitoring, where grade 1 (mild) events may require only observation, while grade 3 (severe, limiting self-care) or grade 4 (life-threatening) events necessitate interventions like dose reductions, treatment holds, or supportive care to prevent progression. For example, in settings, CTCAE grading informs algorithms for managing immunotherapy-related toxicities, such as grading or to guide administration or discontinuation. These systems also support interdisciplinary communication and documentation in electronic health records, standardizing severity assessments across providers to reduce variability in care decisions. In non-oncology contexts, such as trials or general therapeutics, the FDA-recommended grading scales for healthy volunteers categorize events by clinical impact, aiding in the identification of signals warranting amendments or enhanced during routine follow-up. In research trials, grading scales are mandated for adverse event reporting to institutional review boards (IRBs) and regulators, ensuring consistent evaluation of safety profiles across study arms. The , in its version 5.0 released in 2017 and updated periodically, is required in most cancer clinical trials for defining dose-limiting toxicities and calculating rates of grade 3+ events, which influence efficacy-safety balances in regulatory submissions. FDA guidelines emphasize using such scales to grade events by predefined criteria, enabling meta-analyses and post-marketing surveillance by providing comparable data on event frequency and intensity. Patient-reported outcome versions like integrate subjective symptomatic grading, complementing assessments in trials to capture discrepancies—such as underreporting of mild events by providers—and improve holistic endpoints. In I-III studies, aggregated grading determine stopping rules; for instance, exceeding predefined thresholds for serious adverse events (grade 3 or higher) can halt recruitment, as seen in protocols adhering to NCI and FDA standards.
Grading AspectClinical Application ExampleResearch Application Example
Mild (Grade 1)Asymptomatic lab abnormality; monitor without interventionLow toxicity rate in lead-in cohorts; supports dose escalation
Moderate (Grade 2)Minimal interference with daily activities; symptomatic managementContributes to overall tolerability profile; tracked for cumulative incidence
Severe (Grade 3)Hospitalization or intervention required; dose adjustmentDefines dose-limiting toxicity; triggers data monitoring committee review
Life-Threatening (Grade 4)Urgent care needed; potential permanent damageHalts trial arm; reported as serious adverse event to FDA within 15 days
Death (Grade 5)Fatal outcome; immediate Primary ; analyzed for causality in final reports

Detection, Reporting, and Surveillance

Mechanisms of Identification

Adverse events are primarily identified through passive surveillance mechanisms, such as spontaneous reporting systems where healthcare professionals, patients, or manufacturers submit unsolicited reports of suspected events to national pharmacovigilance centers. These reports form the backbone of post-marketing monitoring, with systems like the U.S. Food and Drug Administration's (FDA) Adverse Event Reporting System (FAERS) receiving millions of submissions annually to detect potential safety signals. In , the EudraVigilance database similarly aggregates voluntary reports to facilitate early identification of risks not evident in pre-approval studies. In clinical trials and registries involving direct patient contact, identification occurs via prospective monitoring protocols, including scheduled clinical assessments, laboratory testing, vital sign tracking, and patient diaries to capture both solicited and unsolicited events. Serious adverse events, defined by criteria such as life-threatening conditions or hospitalization, trigger immediate expedited reporting to ethics committees and regulators, often within 15 days for fatal or life-threatening cases. Medical record reviews and standardized questionnaires further enhance detection in these structured settings, allowing for systematic documentation of temporal associations between interventions and outcomes. Post-marketing active surveillance employs observational methods like studies, case-control analyses, and prescription-event to identify events in real-world populations, contrasting with passive systems by proactively querying databases such as electronic health records or claims data. algorithms, including disproportionality metrics like the reporting , scan aggregated reports for statistical anomalies indicating potential causality, as applied by the FDA's Center for Biologics Evaluation and Research to flag excess events relative to comparators. These techniques prioritize rare or delayed events missed in trials, with case-control designs particularly suited for investigating associations with specific outcomes in large populations. Emerging computational approaches, such as of clinical narratives and models trained on historical data, augment traditional methods by automating signal detection in unstructured sources like electronic medical records, though their implementation remains limited by validation needs and data quality issues. Overall, identification relies on integrating multiple data streams to overcome the limitations of any single mechanism, ensuring comprehensive coverage across pre- and post-approval phases.

Regulatory and Voluntary Reporting Protocols

Regulatory protocols for adverse event reporting mandate submissions from pharmaceutical manufacturers, clinical trial sponsors, and certain healthcare facilities to national regulatory authorities, typically focusing on serious or unexpected events to ensure timely . In the United States, under the Federal Food, Drug, and Cosmetic Act, manufacturers must report serious adverse drug experiences to the FDA's Adverse Event Reporting System (FAERS) within 15 calendar days of initial receipt, including follow-up information as it becomes available. For medical devices, user facilities and manufacturers are required to report deaths or serious injuries to the FDA's Manufacturer and User Facility Device Experience (MAUDE) database, with timelines of 10 days for serious events or 30 days for certain malfunctions. In the , marketing authorisation holders submit individual case safety reports (ICSRs) electronically to EudraVigilance, adhering to Good Practices (GVP) Module VI, which specifies expedited reporting for serious reactions within 15 days and non-serious within 90 days. The (WHO) supports global harmonization through its programme, requiring member states' national centers to forward aggregated data to VigiBase, though specific timelines vary by country protocol. Voluntary reporting protocols complement regulatory mandates by enabling healthcare professionals, patients, and consumers to submit suspected adverse events without legal obligation, facilitating early signal detection despite limitations in verification. The FDA's MedWatch program accepts voluntary reports via Form 3500 (for consumers) or online portals for prescription drugs, biologics, and devices, emphasizing serious events like hospitalizations or deaths to aid post-marketing surveillance. For vaccines, the Vaccine Adverse Event Reporting System (VAERS), jointly operated by the FDA and CDC, permits anonymous submissions from any individual using an online form or PDF, serving as a passive surveillance tool established under the 1986 National Childhood Vaccine Injury Act. In Europe, national competent authorities integrate voluntary reports into EudraVigilance, where healthcare providers report via portals linked to the European database of adverse drug reaction reports. WHO's VigiFlow system enables voluntary contributions from low-resource settings, promoting standardized forms that include patient demographics, event descriptions, and causality assessments for global aggregation. These protocols often require standardized data elements, such as the ICH E2B format for electronic transmissions, to ensure across systems, though regulatory enforcement focuses on compliance verification rather than causal confirmation at the reporting stage. Non-compliance with mandatory timelines can result in regulatory actions, including fines or product withdrawals, underscoring the protocols' role in balancing rapid signal detection with resource constraints in .

Underreporting Phenomena and Contributing Factors

Underreporting of adverse events represents a pervasive challenge in and surveillance, where spontaneous reporting systems capture only a of occurrences. Systematic reviews estimate that just 6–10% of adverse drug reactions (ADRs) are reported to authorities, severely limiting the ability to quantify incidence rates and detect safety signals promptly. This discrepancy arises because many events go undocumented in formal channels, despite being noted in clinical records or patient experiences, leading to potential underestimation of risks in drugs, devices, and procedures. Contributing factors to underreporting are multifaceted, often categorized by Ronald Inman's framework of the "seven deadly sins" influencing healthcare professionals' (HCPs) behavior, derived from empirical surveys and qualitative analyses. , the belief that only severe or novel ADRs warrant reporting, affects up to 95% of non-reporters among physicians. Diffidence, or fear of criticism for incomplete causality confirmation, impacts 72% of cases. , encompassing procrastination due to workload and lack of time, is cited in 77% of non-reporting instances. Complacency, assuming marketed products are sufficiently safe, contributes in 47% of scenarios. regarding definitive drug-event causality and indifference (viewing single reports as insignificant) each deter around 67% of potential submissions. Practical and systemic barriers exacerbate these attitudinal issues. Time constraints and workflow disruptions, such as non-integrated systems requiring redundant (e.g., 35+ fields separate from electronic health records), discourage clinicians amid high loads and interruptions. Organizational factors include inadequate training, absence of feedback on reports' impact, and resource shortages, with studies in resource-limited settings like showing zero reports from many peripheral hospitals due to unawareness of programs. Among , underreporting stems from similar knowledge gaps, though research is scarcer; and complacency predominate, with attitudes toward reporting tied to perceived severity. Fear of legal repercussions, such as or blame, further inhibits across HCPs and institutions. These factors interact causally: poor system design amplifies lethargy, while lack of perpetuates , collectively yielding reporting rates as low as 0.003–0.006% for specific classes in targeted studies. Addressing underreporting demands targeted interventions like streamlined digital tools, mandatory training, and cultural shifts to emphasize reporting's value, though persistent biases in institutional priorities may hinder progress.

Data Repositories and Analysis

Pharmacological and Drug-Focused Databases

The FDA Adverse Event Reporting System (FAERS) is a database that collects and analyzes post-marketing reports of adverse events, medication errors, and product quality complaints associated with drugs and therapeutic biologic products in the United States. Reports are submitted mandatorily by manufacturers under FDA regulations and voluntarily by healthcare professionals and consumers, with data used for signal detection to identify potential safety issues warranting further investigation. Since August 2025, FAERS has provided real-time public access through an interactive dashboard, though entries may include duplicates, incomplete information, and unverified causality, as reports represent suspected rather than confirmed associations. In the , EudraVigilance functions as the centralized system for managing suspected s to authorized s, including those under centralized, decentralized, and mutual recognition procedures. It receives electronic reports from national competent authorities, marketing authorization holders, and healthcare professionals, aggregating over millions of individual case safety reports for analysis, such as disproportionality assessments to flag safety signals. Public access is available via the European database of suspected reports, which allows searching by active substance or name, but data limitations include variability in reporting quality across member states and reliance on unconfirmed suspicions. VigiBase, maintained by the Uppsala Monitoring Centre under the Programme for International Drug Monitoring, represents the largest global repository of adverse event reports for medicines and vaccines, exceeding 40 million entries as of October 2025 from over 150 participating countries. It integrates national data in a structured format, enabling quantitative signal detection methods like the vigiReact tool for Bayesian confidence propagation neural networks to prioritize potential risks. Public querying is facilitated through VigiAccess, which lists reported side effects by medicinal product, though interpretations must account for underreporting biases, differential national participation, and the absence of denominator data on drug exposure. These databases primarily support passive surveillance via spontaneous reporting, contrasting with active methods, and are analyzed using metrics such as reporting odds ratios to detect disproportionate signals amid known confounders like Weber effect (early post-approval reporting surges) and notoriety bias (increased reports following publicity). Access for researchers often requires agreements to ensure appropriate use, given the raw nature of data unsuitable for direct causality inference without supplementary evidence from clinical studies or controlled analyses.

Medical Device and Procedural Databases

The Manufacturer and User Facility Device Experience (MAUDE) database, maintained by the U.S. (FDA), functions as the central U.S. repository for adverse event reports associated with medical s. It aggregates mandatory reports under the Medical Device Reporting (MDR) regulation from manufacturers, importers, and user facilities—required within 30 days for events involving or serious , or malfunctions that could lead to such outcomes—as well as voluntary submissions from healthcare providers and s. Launched in June 1993, MAUDE contains over 6 million records as of 2023, covering identifiers, malfunction descriptions, outcomes (e.g., injuries, hospitalizations, s), and reporter narratives, with monthly updates and public searchability to support post-market and signal detection for interventions like recalls or labeling changes. MAUDE reports emphasize device-related problems such as failures, labeling errors, or use issues, but lack standardized assessments, often including unverified or narrative-driven data prone to duplicates, underreporting, or incomplete follow-up, which necessitates supplementary for regulatory decisions. For instance, between 2000 and 2013, MAUDE documented over 10,000 adverse events tied to robotic surgical systems, highlighting procedural risks like system errors during operations. Procedural adverse events, frequently linked to devices used in surgeries or interventions (e.g., stents, pacemakers, or endoscopic tools), are primarily captured in MAUDE when device involvement is reported, but broader surgical databases provide complementary procedural context. The National Surgical Quality Improvement Program (ACS NSQIP), a prospective registry involving over 850 U.S. and international hospitals, tracks 30-day postoperative outcomes for more than 1 million cases annually across 25+ procedures, including adverse events like infections, reoperations, or thromboses that may arise from device-assisted interventions. NSQIP employs trained surgical clinical reviewers for data abstraction and risk adjustment, enabling hospital benchmarking, but it aggregates procedural complications without mandating device-specific isolation, limiting granularity for pure device attribution. Globally, device-specific registries augment general databases by monitoring long-term procedural outcomes, such as the Australian Orthopaedic Association National Joint Replacement Registry, which since 2001 has recorded over 1.5 million procedures and adverse events like prosthetic revisions due to loosening or infection, informing device performance trends. In the European Union, the European Database on Medical Devices (EUDAMED) under Regulation (EU) 2017/745 incorporates a planned vigilance module for adverse event reporting, including procedural incidents, to enable cross-border analysis; however, as of October 2025, this module awaits full functionality (projected for Q1 2026 or later), with manufacturers currently relying on national systems for mandatory submissions within 15 days of serious incidents. EUDAMED's eventual integration of unique device identification (UDI) data aims to enhance traceability for procedural devices, though delays have prolonged fragmentation in EU surveillance.

Global and Integrated Surveillance Systems

The World Health Organization's Programme for International Drug Monitoring (PIDM), established in , serves as the primary global framework for coordinating efforts to detect, assess, and prevent adverse drug reactions and vaccine-related events. It involves national centers from member states submitting data, enabling pooled analysis that identifies rare safety signals not detectable in isolated national systems. As of March 2025, PIDM includes 160 full members and 22 associate members, covering nearly 99% of the global population. At the core of PIDM is VigiBase, the WHO global database of Individual Case Safety Reports (ICSRs), managed by the Uppsala Monitoring Centre (UMC) in . VigiBase contains over 40 million reports as of February 2025, aggregated from more than 180 PIDM members, including integrations with major regional databases such as the U.S. FAERS and VAERS, and the European Union's EudraVigilance. Reports are standardized using terminologies like for adverse events and WHODrug Global for medications, with quality controls ensuring completeness, accuracy, and causality assessments via UMC methods. This integration facilitates disproportionality analyses and signal detection for emerging risks, such as those from new medicines or vaccines, supporting regulatory actions worldwide. Data submission to VigiBase occurs through structured electronic formats from national centers, with UMC performing routine analyses for trends and novel signals. for members is provided via secure tools and training, while public interfaces like VigiAccess offer anonymized summaries to promote transparency without compromising confidentiality. In March 2025, WHO and UMC updated VigiBase access conditions to clarify data-sharing protocols, enhancing trust and utility in global . These systems emphasize empirical aggregation over isolated reporting, though limitations persist in data completeness due to voluntary national contributions and varying reporting standards across regions. Beyond PIDM, efforts toward integration include regional initiatives bridging local databases to VigiBase, such as those in the for improved ICSR transmission quality, but no comparable standalone global systems exist for comprehensive adverse event . VigiBase remains the largest and most integrated repository, underscoring PIDM's role in causal risk identification through scale.

Historical Evolution

Early Conceptualization and Milestones

The concept of adverse events in medicine traces its roots to systematic observations of unintended harms from therapeutic interventions, with early documentation emerging in the amid growing pharmaceutical practices. In 1848, the British Medical Journal published reports on adverse reactions to mercury and compounds, marking one of the first structured collections of such data, driven by clinicians' recognition that drugs could produce effects independent of their intended therapeutic actions. This period saw pharmacologists like Oswald Schmiedeberg emphasize experimental to identify toxic potentials, laying groundwork for distinguishing beneficial from harmful outcomes without formal regulatory frameworks. A pivotal milestone occurred in 1937 with the disaster in the United States, where over 100 deaths resulted from a solvent in the untested formulation, exposing gaps in pre-market safety assessments and prompting the 1938 Federal Food, , and Cosmetic Act to mandate safety demonstrations before marketing, though it did not yet require post-approval adverse event reporting. The tragedy of 1961–1962, involving severe birth defects in thousands of infants exposed in utero, catalyzed global recognition of latent risks in approved , leading to the 1962 Kefauver-Harris Amendments in the U.S., which enshrined requirements for efficacy proof and manufacturer reporting of serious adverse reactions to regulators. Concurrently, the launched the Yellow Card Scheme in 1964 for voluntary adverse reaction reporting, while the initiated its international program on adverse drug reactions in 1968, formalizing as a discipline focused on post-marketing surveillance. By 1969, the U.S. established its Reporting System (later evolving into FAERS), enabling aggregation of spontaneous reports to detect signals of harm, with approximately 2.3 million cases documented through 2002 as a foundation for ongoing monitoring. These developments shifted conceptualization from anecdotal clinician notes to institutionalized systems, emphasizing causality assessment via criteria like those later formalized by WHO (e.g., precursors), though early systems grappled with underreporting due to voluntary participation and lack of standardized definitions distinguishing adverse events from reactions.

Regulatory Developments and Key Reforms

The 1938 Federal Food, Drug, and Cosmetic Act, enacted in response to the 1937 disaster that killed over 100 people due to a toxic solvent, marked an early regulatory milestone by mandating pre-market safety testing for drugs but lacked requirements for ongoing post-marketing surveillance of adverse events. This was followed by the 1962 Kefauver-Harris Amendments, prompted by the thalidomide tragedy exposing thousands of birth defects in Europe, which revolutionized U.S. regulation by requiring pharmaceutical manufacturers to submit substantial evidence of both safety and efficacy for new drug approvals and to report all serious adverse drug reactions to the FDA within specified timelines, shifting from voluntary to mandatory post-approval monitoring. In the international arena, the established its Programme for International Drug Monitoring in 1968, followed by the creation of the Monitoring Centre in 1972 as a collaborating center to coordinate global efforts and analyze spontaneous adverse event reports from member states. Concurrently, the FDA formalized its Reporting System (later evolving into the FDA Adverse Event Reporting System, or FAERS) in the late 1960s, initially relying on voluntary healthcare professional reports but expanding to include mandatory manufacturer submissions under the 1962 amendments, with significant revisions in 1997 to enhance data capture and analysis capabilities. Key reforms in the included the 1995 establishment of the (), which harmonized across member states, and subsequent directives like 2001/83/EC that strengthened requirements for risk management plans and expedited reporting of serious adverse events. In the U.S., the 2012 Safety and Innovation Act further reformed surveillance by mandating enhanced post-marketing studies for certain drugs and improving FAERS electronic submission processes to address underreporting and signal detection delays. These developments emphasized proactive risk minimization over reactive responses, though challenges persist in standardizing global data and incentivizing complete reporting.

Controversies, Criticisms, and Debates

Issues of Underreporting Versus Signal Inflation

Underreporting of adverse events in spontaneous reporting systems, such as the U.S. (VAERS) and similar databases, is a well-documented limitation that leads to underestimation of true incidence rates. Studies indicate that fewer than 10% of adverse drug reactions (ADRs) are typically reported, with systematic reviews across multiple countries estimating underreporting rates exceeding 90%. For VAERS specifically, sensitivity for capturing serious events like or Guillain-Barré ranges from 13% to 76%, depending on the and context, reflecting factors such as reluctance due to time constraints, legal fears, lack of awareness, or doubt about causality. This underreporting is exacerbated in voluntary systems reliant on healthcare providers and consumers, where only a fraction of events—often estimated at less than 1% for —reach authorities. Conversely, signal inflation arises from the same passive mechanisms, where unverified reports, duplicates, and stimulated reporting can generate disproportionate associations that do not reflect causal risks. In spontaneous systems, false-positive signals emerge from temporal coincidences, media-driven reporting biases (e.g., the Weber effect, an initial surge post-approval), or confounding co-prescriptions, potentially distorting analyses like disproportionality metrics. For instance, duplicate ADRs in databases can artificially elevate event frequencies, compromising signal validity and leading regulators to investigate spurious alerts. Algorithms for signal detection, while enhancing hypothesis generation, must account for these artifacts; otherwise, they risk overemphasizing noise over true hazards, as seen in evaluations of FDA's FAERS where unadjusted data yield non-causal patterns. The tension between underreporting and signal inflation challenges regulatory decision-making, as minimizing one exacerbates the other: aggressive promotion of reporting amplifies unconfirmed signals, while conservative thresholds miss emerging risks. experts emphasize that spontaneous reports serve primarily for signal detection, not incidence estimation, requiring with active or observational data to validate findings and mitigate biases. This duality underscores the need for methodological refinements, such as automated de-duplication and bias-adjusted analyses, to balance sensitivity against specificity in protecting without undue alarmism.

Influence on Policy, Approvals, and Litigation

Reports of adverse events have significantly shaped pharmaceutical approvals by triggering post-market reviews that result in safety label updates, black box warnings, or outright withdrawals. The FDA's Adverse Event Reporting System (FAERS) has contributed to over 50% of postmarket safety-related label changes by identifying signals of serious risks, such as or cardiovascular events, which prompt further epidemiological investigations and regulatory actions. For instance, between 2001 and 2010, more than 70 FDA-approved drugs encountered post-approval safety issues, including 26 withdrawals linked to adverse event data revealing risks like heart attacks or deaths not fully evident in pre-approval trials. Similarly, from 1998 to 2013, 41% of drugs approved on limited efficacy data received subsequent safety warnings or were withdrawn due to adverse event signals. These signals have influenced broader reforms emphasizing enhanced . Historical withdrawals, totaling 462 medicinal products between 1953 and 2013 primarily due to adverse drug reactions like immune-mediated disorders, underscored the need for robust , leading to policies such as mandatory and strategies (REMS) for high-risk drugs. In the context, adverse event reports of respiratory depression and overdose deaths highlighted FDA approval shortcomings, contributing to shifts like the 2016 CDC prescribing guidelines and expanded monitoring requirements. FAERS data, analyzed for disproportionality in event reporting, has directly informed quarterly FDA signal assessments, resulting in actions like restricted distribution programs. However, reliance on voluntary reporting systems like FAERS introduces challenges, as underreporting or factors can delay responses until corroborated by controlled studies. In litigation, adverse event databases serve as key evidence in claims alleging manufacturers failed to adequately warn of risks or concealed safety data. Plaintiffs often cite FAERS or similar reports to establish causation or notice of defects, as seen in the Vioxx cases where cardiovascular adverse events prompted Merck's 2004 withdrawal and a $4.85 billion settlement for thousands of heart attack and stroke claims. multidistrict litigation, fueled by adverse event documentation of addiction and overdose risks, has yielded settlements exceeding $50 billion across manufacturers like , with courts referencing post-approval surveillance failures. Other examples include Depakote off-label promotion suits, settled for $1.5 billion, where adverse event signals of agitation control risks were central to fraud and injury claims. Such cases highlight how underreported or disputed events can inflate litigation signals, yet verified clusters have driven corporate accountability and influenced approval standards by increasing scrutiny on pre-market risk-benefit analyses.

Balanced Perspectives on Systemic Biases

Systemic biases in adverse event reporting encompass methodological, institutional, and sociocultural factors that can distort signal detection and interpretation in systems like VAERS and FAERS. Passive inherently suffers from underreporting, with VAERS capturing only a fraction of actual events—estimated lower for mild cases but higher for severe ones—due to reliance on voluntary submissions from healthcare providers and the public. Conversely, stimulated reporting can inflate signals for high-profile products, such as vaccines, where intense media scrutiny and public awareness prompted disproportionate submissions unverified for causality. These countervailing biases necessitate cautious analysis, as raw disproportionality metrics like reporting odds ratios may amplify differences unrelated to true risk. Institutional influences exacerbate these issues, with pharmaceutical companies mandated to report adverse events yet facing incentives to minimize perceived risks through selective emphasis in trial data or post-marketing summaries. Regulatory bodies like the FDA exhibit potential conflicts from their approval roles overlapping with surveillance, leading critics to argue for "regulatory capture" where industry funding—evident in advisory committees—may temper aggressive signal pursuit. Peer-reviewed analyses highlight inconsistent definitions and reporting thresholds in trials, often understating harms in industry-sponsored studies, though raw data biases appear limited when scrutinized independently. Balanced assessment requires cross-verifying with , acknowledging that while pharma-driven underemphasis occurs, overreporting biases from novelty or litigation fears can equally mislead. Sociocultural and media dynamics introduce further skews, with coverage of new drugs frequently downplaying harms to highlight benefits, influenced by from sponsors. A study found pharmaceutical ads correlate with reduced emphasis on potential risks, fostering that prioritize efficacy over safety signals. In crises like , ideological polarization compounds this: trust in institutions predicts higher intent and potentially lower scrutiny of adverse reports, while hesitancy correlates with amplified signal interpretation among skeptics. Mainstream outlets and academia, often aligned with progressive paradigms, exhibit systemic reluctance to elevate dissenting safety concerns, as seen in delayed acknowledgments of rare risks despite data; this contrasts with contrarian sources prone to overinflation via unverified VAERS misuse. Truth-seeking demands prioritizing empirical over narrative fidelity, discounting sources with evident institutional entanglements that undervalue patient-reported events from marginalized or ideologically opposed groups.

References

  1. [1]
    IND Application Reporting: Safety Reports - FDA
    Oct 19, 2021 · Adverse event means any untoward medical occurrence associated with the use of a drug in humans, whether or not considered drug related.
  2. [2]
    Adverse event | European Medicines Agency (EMA)
    An untoward medical occurrence after exposure to a medicine, which is not necessarily caused by that medicine.
  3. [3]
    2. Adverse Event (AE) versus Adverse Reaction (adverse drug ...
    An adverse reaction, in contrast to an adverse event, is characterised by the fact that a causal relationship between a medicinal product and an occurrence is ...
  4. [4]
    What is a Serious Adverse Event? - FDA
    May 18, 2023 · An adverse event is any undesirable experience associated with the use of a medical product in a patient. The event is serious and should be reported to FDA ...
  5. [5]
    FDA's Adverse Event Reporting System (FAERS)
    Nov 8, 2024 · The FDA Adverse Event Reporting System (FAERS) database contains adverse event reports, medication error reports and product quality complaintsFeedback form · FAQ · International Regulatory...
  6. [6]
    Adverse Event Detection, Processing, and Reporting - NCBI - NIH
    Medical device reporting is required for incidents in which the device may have caused or contributed to a death or serious injury, or may have malfunctioned ...Introduction · Identifying and Reporting... · Coding · Adverse Event Required...
  7. [7]
    Safety Reporting - CTTI - Clinical Trials Transformation Initiative
    During a clinical trial of an investigational new drug, it is important to detect whether the drug is causing unexpected health problems in study participants ...
  8. [8]
    Strengthening the reporting of harms of all interventions in clinical ...
    Nov 12, 2022 · Harms reporting in clinical trials is critical to understanding the nature and likelihood of experiencing unwanted effects resulting from or following ...
  9. [9]
    [PDF] E2D_Guideline.pdf - ICH
    Nov 12, 2003 · An adverse event can therefore be any unfavorable and unintended sign (for example, an abnormal laboratory finding), symptom, or disease.
  10. [10]
    Management of adverse effects and pharmacovigilance - NCBI - NIH
    Pharmacovigilance is defined by WHO as “the science and activities relating to the detection, assessment, understanding and prevention of adverse effects or ...Introduction · Monitoring for adverse effects... · Management of adverse effectsMissing: scope | Show results with:scope
  11. [11]
    [PDF] Definitions and Standards for Expedited Reporting - FDA
    Mar 1, 1995 · Definitions for the terms adverse event (or experience), adverse reaction, and unexpected adverse reaction have previously been agreed to by.
  12. [12]
    [PDF] Definitions and Standards for Expedited Reporting
    An adverse event (AE) can therefore be any unfavourable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease ...
  13. [13]
  14. [14]
    Adverse Drug Reactions - StatPearls - NCBI Bookshelf
    Jan 10, 2024 · An adverse drug reaction (ADR) refers to an untoward reaction to a medication. ADRs are common and constitute a significant healthcare burden.Introduction · Function · Clinical Significance · Other Issues
  15. [15]
    'Adverse Event,' Not the Same as 'Side Effect' | Pharmacy Times
    Feb 22, 2017 · Adverse events are unintended pharmacologic effects that occur when a medication is administered correctly while a side effect is a secondary unwanted effect.
  16. [16]
    Adverse Reactions vs Side Effects: Are They the Same Thing?
    Jun 15, 2025 · Side effects are generally expected and less concerning, while adverse reactions are unexpected and may pose significant health risks.
  17. [17]
    [PDF] Adverse Drug Events, Adverse Drug Reactions and Medication Errors
    An adverse drug reaction is a “response to a drug which is noxious and unintended and which occurs at doses normally used in man for prophylaxis, diagnosis, or ...
  18. [18]
    Medical Error, Adverse Events, and Complications in Interventional ...
    Dec 15, 2020 · An adverse event is defined as “an unintended injury or complication caused by medical management resulting in measurable disability, prolonged ...skipMainNavigation" · Introduction · Adverse Events in IR · Complications
  19. [19]
    Is it a complication or a consequence - CVIR Endovascular
    Jan 5, 2024 · In other words, the complication/adverse event is an unintended injury caused by medical management resulting in measurable disability, ...
  20. [20]
    Adverse Events, Near Misses, and Errors | PSNet
    Summary. In summary, adverse events refer to harm from medical care rather than an underlying disease. Important subcategories of adverse events include: ...
  21. [21]
    Medication errors | European Medicines Agency (EMA)
    A medication error is an unintended failure in the drug treatment process that leads to, or has the potential to lead to, harm to the patient.
  22. [22]
    Adverse Drug Reactions: Types and Treatment Options - AAFP
    Nov 1, 2003 · This classification system includes: Type I reactions (IgE-mediated); Type II reactions (cytotoxic); Type III reactions (immune complex); ...
  23. [23]
    Adverse Drug Reactions - Clinical Pharmacology - Merck Manuals
    One classification system categorizes ADRs into 6 types (1):. Type A, intrinsic (also called augmented): Related to the predictable pharmacological actions of a ...
  24. [24]
    [PDF] Definitions and Standards for Expedited Reporting E2A - ICH
    A serious adverse event (experience) or reaction is any untoward medical occurrence that at any dose: * results in death,. * is life-threatening,. NOTE: The ...
  25. [25]
    [PDF] Common Terminology Criteria for Adverse Events (CTCAE) - EORTC
    May 28, 2009 · The CTCAE displays Grades 1 through 5 with unique clinical descriptions of severity for each AE based on this general guideline:Missing: pharmacovigilance | Show results with:pharmacovigilance
  26. [26]
    DAIDS Adverse Event Grading Tables
    Feb 13, 2025 · The Division of AIDS (DAIDS) Table for Grading the Severity of Adult and Pediatric Adverse Events consists of parameters, or Adverse Events (AEs), with ...Missing: pharmacovigilance | Show results with:pharmacovigilance
  27. [27]
    Seriousness vs. Severity - Ennov
    'Severe' is used to describe the intensity (severity) of a specific event (mild, moderate or severe); the event itself, however, may be of relatively minor ...
  28. [28]
    Preventable Adverse Drug Events Among Inpatients: A Systematic ...
    Sep 1, 2018 · ADEs were further categorized as being preventable when due to error or not preventable when a drug was used as clinically indicated but the ...
  29. [29]
    Methods for evaluating adverse drug event preventability in ...
    Dec 4, 2018 · We compared three methods for determining preventability of adverse drug events in emergency department patients and explored their strengths and weaknesses.
  30. [30]
    Incidence and Preventability of Adverse Drug Events Among Older ...
    Mar 5, 2003 · Electrolyte/renal, gastrointestinal tract, hemorrhagic, and metabolic/endocrine events were the most common types of preventable adverse drug ...
  31. [31]
    Labeling an Adverse Drug Event “Preventable” - AMA Journal of Ethics
    These contributing factors can be broadly grouped into six categories: (1) patient-related causes, (2) physician-related causes, (3) medication-related ...
  32. [32]
    Study on the Classification, Causality, Preventability and Severity of ...
    The majority of ADRs were moderate to severe (n = 35, 68.6%), of which 37.3% were found to be potentially preventable.
  33. [33]
    Adverse Drug Reaction Probability Scale (Naranjo) in Drug Induced ...
    May 4, 2019 · The Naranjo Algorithm, or Adverse Drug Reaction Probability Scale, is a method by which to assess whether there is a causal relationship ...
  34. [34]
    Adverse drug reaction and causality assessment scales - PMC - NIH
    The Naranjo Probability Scale are the generally accepted and most widely used methods for causality assessment in clinical practice as they offer a simple ...
  35. [35]
    Causality assessment of adverse drug reactions by applying a ...
    Causality assessment of adverse drug reactions (ADRs) is an essential approach in pharmacovigilance. The World Health Organization-Uppsala Monitoring Center ...
  36. [36]
    A method for estimating the probability of adverse drug reactions
    This systematic method offers a sensitive way to monitor ADRs and may be applicable to postmarketing drug surveillance.
  37. [37]
    Algorithms for assessing the probability of an Adverse Drug Reaction
    When the same individuals used the Naranjo Algorithm it rose to between 83% and 92% (kappa = 0.69–0.86, r = 0.92), this rise being statistically significant.
  38. [38]
    [PDF] The use of the WHO-UMC system for standardised case causality ...
    An inherent problem in pharmacovigilance is that most case reports concern suspected adverse drug reactions. Adverse reactions are rarely specific for the ...Missing: attribution | Show results with:attribution
  39. [39]
    Agreement between WHO-UMC causality scale and the Naranjo ...
    In the present study, we assessed agreement between the two widely used causality assessment scales, that is, the WHO-UMC criteria and the Naranjo algorithm.
  40. [40]
    Development and inter-rater reliability of the Liverpool adverse drug ...
    The Liverpool ADR CAT, using 40 cases from an observational study, showed causality categories of 1 unlikely, 62 possible, 92 probable and 125 definite.
  41. [41]
    Interrater agreement of two adverse drug reaction causality ...
    A new method to assess causality of suspected adverse drug reactions, the Liverpool Adverse Drug Reaction Causality Assessment Tool (LCAT), showed high ...
  42. [42]
    Comparison of the Liverpool Causality Assessment Tool vs. the ...
    Feb 27, 2023 · The aim of this study is to compare the Liverpool Causality Assessment Tool vs. Naranjo Scale for screening suspected adverse drug reaction ...INTRODUCTION · METHODS · RESULTS · DISCUSSION
  43. [43]
    Causality assessment - Perspectives in Clinical Research
    Adverse reactions are rarely specific for the drug, diagnostic tests are usually absent and a re-challenge is rarely ethically justified.
  44. [44]
    Causality Assessment in Pharmacovigilance: Still a Challenge
    Causality in pharmacovigilance is a difficult and time consuming exercise. This paper presents the challenges in determining causation by drug therapy.
  45. [45]
    Inter-rater agreement between the three raters: WHO-UMC scale.
    Results: The agreement between the WHO-UMC criteria and Naranjo's algorithm was the highest (136), with a Kappa value of 0.511, suggesting a moderate level of ...
  46. [46]
    Evaluation of inter-rater agreement between three causality ...
    A majority of the ADRs found by WHO-UMC method were possible (39.06%), followed by probable (34.33%) and certain (26.18%). Naranjo's algorithm found 52.54% of ...
  47. [47]
    Dilemmas of the causality assessment tools in the diagnosis of ...
    Such approaches are quite skeptical and liable to generate cloudy causation results. Causation is quite intricate to ascertain than correlation in ...
  48. [48]
    Causality assessment: A brief insight into practices in ... - NIH
    An inherent problem in pharmacovigilance is that most case reports concern suspected ADR. Adverse reactions are rarely specific for the drug, diagnostic tests ...
  49. [49]
    Aspects to consider in causality assessment of safety signals
    In the field of drug safety, causality assessment aims to determine the level of plausibility of the relationship between an adverse event and the exposure ...Abstract · Introduction · Outline of the proposed... · Conclusion
  50. [50]
    CTCAE and AE Reporting - NCI
    Sep 2, 2025 · Name ("specify") the adverse event. The name must be explicit, brief (2-4 words) and provide sufficient detail to name the event.CTCAE · Adverse Event Reporting (AER... · Defining Adverse Events
  51. [51]
    [PDF] Common Terminology Criteria for Adverse Events (CTCAE) v6.0 ...
    Jul 22, 2025 · CTCAE Term: An Adverse Event (AE) is any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease ...
  52. [52]
    [PDF] Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers ...
    Adverse events in a clinical trial of an investigational vaccine must be recorded and monitored and, when appropriate, reported to FDA and others involved in an ...
  53. [53]
    Common terminology criteria for adverse events - UpToDate
    CTCAE stands for Common Terminology Criteria for Adverse Events; these criteria are also called "common toxicity criteria." In CTCAE, an adverse event (AE) is ...
  54. [54]
    Using CTCAE to Report Immunotherapy Adverse Events
    Aug 1, 2018 · Nurses can use CTCAE to grade symptoms and report it to providers to address an algorithm to further work up and treat the irAE,” Riemer says. “ ...
  55. [55]
    Adverse Event Recognition and Management in Practice and on ...
    Reviewing the CTCAE reminds us that, generally, the grading of adverse events is an attempt to describe the clinical severity of a sign, symptom, or disease ...
  56. [56]
    How Is CTCAE Improving Research and Patient Care?
    Aug 1, 2018 · CTCAE is an internationally accepted standard for defining and categorizing adverse events. It allows us to talk to our patients in a more useful way.
  57. [57]
    Toxicity Grading Scale for Volunteers in Vaccine Clinical Trials - FDA
    Feb 2, 2025 · Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials, CBER, Biiologics.
  58. [58]
    The use of and adherence to CTCAE v3.0 in cancer clinical trial ...
    In this study, we sought to comprehensively assess the use of CTCAE v3.0 for AE reporting in recent publications of cancer clinical trials.
  59. [59]
    Overview of the PRO-CTCAE - Healthcare Delivery Research Program
    Jun 27, 2024 · PRO-CTCAE is a patient-reported outcome (PRO) measurement system developed to evaluate symptomatic toxicity in patients on cancer clinical ...
  60. [60]
    Clinician and Patient Reporting of Symptomatic Adverse Events in ...
    Dec 8, 2022 · This review addresses the development of PRO-CTCAE with an emphasis on the differences between PRO-CTCAE scores and CTCAE severity grades.
  61. [61]
    CTEP Trial Development and Conduct - NCI
    Jun 25, 2025 · Looking for the Adverse Event Reporting Guidelines or CTCAE? Click here for the new site-focused adverse events page. Resources for Lead ...LOI and Concept Development · Protocol and Informed... · CTCAE and CTEP Codes<|separator|>
  62. [62]
    Adverse Event Reporting to IRBs — Improving Human Subject ... - FDA
    May 6, 2020 · This guidance is intended to assist the research community in interpreting requirements for submitting reports of unanticipated problems, including certain ...
  63. [63]
    Data Mining at the Center for Biologics Evaluation and Research | FDA
    Nov 29, 2022 · It involves the systematic examination of reported adverse events (AEs) to identify an excess of AEs reported for a product relative to other ...
  64. [64]
    [PDF] E2E Pharmacovigilance Planning - FDA
    Stimulated adverse event reporting in the early postmarketing phase can lead companies to notify healthcare professionals of new therapies and provide safety.
  65. [65]
    Text Mining of Adverse Events in Clinical Trials: Deep Learning ...
    Dec 24, 2021 · Early detection of unknown adverse events, reactions, interactions, and an increase in the frequency of (known) adverse events is a key element ...
  66. [66]
    standard pharmacovigilance pipeline for detection of adverse drug ...
    Mar 7, 2023 · The spontaneous reporting system is a widely used, effective, and relatively inexpensive method of collecting information on suspected ADRs. ...
  67. [67]
    How to Report Medical Device Problems - FDA
    Mar 27, 2025 · Requirements for reporting medical device problems, including malfunctions and adverse events (serious injuries or deaths) associated with ...
  68. [68]
    [PDF] Guideline on good pharmacovigilance practices (GVP) - Module VI
    Jul 28, 2017 · This Module of GVP addresses the legal requirements detailed in Directive 2001/83/EC [DIR] and. Regulation (EC) No 726/2004 [REG], ...
  69. [69]
    Pharmacovigilance - World Health Organization (WHO)
    Pharmacovigilance (PV) is defined as the science and activities relating to the detection, assessment, understanding and prevention of adverse effects or ...
  70. [70]
    The FDA Safety Information and Adverse Event Reporting Program
    MedWatch is made up of voluntary and mandatory reporting on prescription medicines, over-the-counter medicines, non-vaccine biologicals, medical devices, ...Reporting Serious Problems to ...FDA MedWatchMedical Product Safety ...FDA MedWatch/Safety AlertsAbout the MedWatch E-list
  71. [71]
    Vaccine Adverse Event Reporting System (VAERS)
    Report an Adverse Event using the VAERS online form or the downloadable PDF. New! Important: If you are experiencing a medical emergency, seek immediate ...Report an Adverse EventData
  72. [72]
    European database of suspected adverse drug reaction reports
    On this website you can view data on suspected side-effects, also known as suspected adverse drug reactions, for authorised medicines in the European Economic ...Switch to Veterinary · How to report a side effect · Eudravigilance · Search
  73. [73]
    Pharmacovigilance - Guidance and information
    Adverse events following immunization (AEFI). Brief overview of anaphylaxis as an adverse event following immunization (‎AEFI)‎ and practical guidance on its ...Missing: scope | Show results with:scope
  74. [74]
    EudraVigilance: electronic reporting - European Medicines Agency
    Marketing authorisation holders and sponsors of clinical trials must report and evaluate suspected adverse drug reactions during the development and ...<|separator|>
  75. [75]
    Pharmacovigilance: reporting requirements throughout a product's ...
    Drug manufacturers are required to collect, analyze, and report AEs to all countries where the product is marketed, working closely with the regulatory agencies ...
  76. [76]
    Factors Associated with Underreporting of Adverse Drug Reactions ...
    Jun 6, 2023 · Underreporting is a major limitation of spontaneous notification systems, as it is estimated that only 6–10% of all ADRs are reported [6, 7]. On ...
  77. [77]
    Why Clinicians Don't Report Adverse Drug Events: Qualitative Study
    Feb 27, 2018 · Our objectives were to identify barriers to adverse drug event documentation and factors contributing to underreporting.
  78. [78]
    Determinants of under-reporting of adverse drug reactions - PubMed
    Other factors associated with under-reporting were ignorance (only severe ADRs need to be reported) in 95%; diffidence (fear of appearing ridiculous for ...
  79. [79]
    Under-reporting of adverse drug reactions - PubMed Central - NIH
    The aim was to evaluate the extent and factors responsible for underreporting (UR) of adverse drug reactions (ADRs) in India.
  80. [80]
    Factors associated with underreporting of adverse drug reactions by ...
    May 29, 2023 · Ignorance (10/13), complacency (6/13), and lethargy (6/13) were the most frequent reasons for not reporting. Conclusion. This study highlighted ...
  81. [81]
    Underreporting and Triggering Factors for Reporting ADRs of Two ...
    The outcomes of the research show significant rates of underreporting (0.006029% for antiglaucoma and 0.003552% for artificial tears); this indicates a ...
  82. [82]
    FDA Adverse Event Reporting System (FAERS) - openFDA
    The FDA Adverse Event Reporting System (FAERS) is a database that contains information on adverse event and medication error reports submitted to FDA.
  83. [83]
    FDA Begins Real-Time Reporting of Adverse Event Data
    Aug 22, 2025 · FAERS is the FDA's primary database for collecting and analyzing adverse event reports, serious medication errors, and product quality ...
  84. [84]
    FDA Adverse Event Reporting System (FAERS) Public Dashboard
    Dec 7, 2023 · The FAERS Public Dashboard is a highly interactive web-based tool that will allow for the querying of FAERS data in a user friendly fashion.
  85. [85]
    EudraVigilance | European Medicines Agency (EMA)
    EudraVigilance is the system for managing and analysing information on suspected adverse reactions to medicines which have been authorised or being studied ...Electronic reporting · Access to EudraVigilance data · EudraVigilance training
  86. [86]
    EudraVigilance system overview | European Medicines Agency (EMA)
    The European database of suspected adverse drug reaction reports Adrreports.eu is a publicly accessible portal designed to search and view EudraVigilance data ...
  87. [87]
    About VigiBase | UMC - Uppsala Monitoring Centre
    Oct 17, 2025 · VigiBase, the World Health Organization global database of adverse event reports for medicines and vaccines, holds over 40 million reports ...
  88. [88]
    Programme for International Drug Monitoring
    In July 2023, there were over 35 million reports of adverse reactions in VigiBase. Data in VigiBase are recorded in a structured and comprehensive way to allow ...
  89. [89]
    VigiAccess
    A VigiAccess search on a medicinal product will return a basic list of reported potential side effects and the number of times each effect has been reported.
  90. [90]
    Exploring the FDA Adverse Event Reporting System (FAERS) to ...
    The FDA Adverse Event Reporting System (FAERS) is a database for post-marketing drug safety monitoring and influences FDA safety guidance documents.
  91. [91]
    VigiBase FAQ | UMC - Uppsala Monitoring Centre
    Oct 9, 2025 · VigiBase data is intended for use by pharmacovigilance professionals with expertise in assessing medicines safety. Gathering national data into ...
  92. [92]
    About Manufacturer and User Facility Device Experience (MAUDE)
    The MAUDE database contains medical device reports (MDRs) of adverse events, including device and patient problems, and patient demographic information.
  93. [93]
    Manufacturer and User Facility Device Experience (MAUDE) Database
    The MAUDE database is a searchable database of medical device reports of adverse events, submitted by manufacturers, importers, and user facilities, updated ...
  94. [94]
    Adverse Event Reporting Data Files - FDA
    Jun 21, 2019 · MAUDE data contain reports received by the FDA of adverse events involving medical devices. The data consists of voluntary reports since June 1993.
  95. [95]
    Adverse Events in Robotic Surgery: A Retrospective Study of 14 ...
    Mar 8, 2016 · The adverse events reports related to robotic surgical systems and instruments, submitted to the publicly available FDA MAUDE database ...
  96. [96]
    ACS National Surgical Quality Improvement Program
    In a study comparing ACS NSQIP data to claims data, ACS NSQIP identified 61% more complications, including 97% more surgical site infections. Risk-adjusted.ACS NSQIP Participant Use... · FAQs · (ACS NSQIP ® ) is at the... · Nsqip & qvpMissing: adverse devices
  97. [97]
    Surgical research using national databases - PMC - NIH
    The NSQIP database is peer-controlled and collects information on patients who undergo elective and select trauma surgical procedures. Most high energy ...
  98. [98]
    Registries for Medical Devices - NCBI - NIH
    Medical device registries are critical for the identification and study of medical devices outcomes. Device registries are used for many purposes.
  99. [99]
    EUDAMED Overview - Public Health - European Commission
    EUDAMED is the IT system established by Regulation (EU) 2017/745 on medical devices and Regulation (EU) 2017/746 on in vitro diagnosis medical devices.
  100. [100]
    The European Commission prepares stakeholders ... - Emergo by UL
    Oct 7, 2025 · The PMS Vigilance Module is not expected to become mandatory before Q2 2027. EUDAMED timeline from Q3 2025 to Q1 2027 showing key milestones, ...
  101. [101]
    WHO and UMC enhance data sharing clarity with new VigiBase ...
    Mar 20, 2025 · WHO and UMC introduce new VigiBase access conditions, enhancing data sharing clarity and stakeholder confidence in global pharmacovigilance.
  102. [102]
  103. [103]
    Building better data bridges between pharmacovigilance databases ...
    Mar 27, 2025 · This initiative aimed to strengthen the quality of transmission of individual case study reports between countries across the region and VigiBase.
  104. [104]
    An historical overview over Pharmacovigilance - PMC - NIH
    Pharmacovigilance started about 170 years ago, although it was not yet named as such at that time. It is structured activity in the professional health field.
  105. [105]
    The Dawn of Pharmacovigilance: An Historical Perspective
    Aug 7, 2025 · An early example of a systemic collection of adverse drug reactions occurred in the beginning of the 19th century when reactions were ...
  106. [106]
    Pharmacovigilance: the evolution of drug safety monitoring - PMC
    The first adverse drug reaction (ADR) related catastrophe in the United States of America (USA) occurred in 1937 after diethyl glycol was used as a solvent in ...
  107. [107]
    Pharmacovigilance: An Overview - ScienceDirect.com
    This experience was codified in the 1962 Kefauver-Harris Amendments to the Federal Food, Drug and Cosmetic Act as adverse event evaluation and reporting ...
  108. [108]
    Adverse Drug Event Surveillance and Drug Withdrawals in the ...
    Jun 27, 2005 · During the 33-year period from 1969 when adverse drug event reporting was initiated through 2002, about 2.3 million case reports of adverse ...Missing: conceptualization | Show results with:conceptualization
  109. [109]
    The FDA Adverse Event Reporting System (FAERS) Explained
    Aug 21, 2025 · Under this program, over 180 countries' regulatory agencies contribute their adverse event reports to a global database called VigiBase.
  110. [110]
    Promoting Safe & Effective Drugs for 100 Years - FDA
    Apr 23, 2019 · "Also critically, the 1962 amendments required that the FDA specifically approve the marketing application before the drug could be marketed, ...
  111. [111]
    Reform, Regulation, and Pharmaceuticals — The Kefauver–Harris ...
    Oct 18, 2012 · It was only by chance timing that the summer of 1962 also produced a highly visible tragedy (thalidomide), a hero (Frances Kelsey), and enough ...Missing: adverse | Show results with:adverse
  112. [112]
    Data Mining of the Public Version of the FDA Adverse Event ... - NIH
    The original system was started in 1969, but since the last major revision in 1997, reporting has markedly increased. Data mining algorithms have been developed ...
  113. [113]
    The evolution of pharmacovigilance ecosystems: Does Moore's law ...
    Oct 20, 2022 · With the increasing volume of legislation, pharmacovigilance systems today are inherently complex, and the emphasis has shifted from reactive ( ...Missing: reforms | Show results with:reforms
  114. [114]
    Underreporting of Adverse Drug Events: a Look into the Extent ...
    Organization-related factors that hinder effective reporting include lack of leadership support, insufficient resources, prioritization of other metrics over ...
  115. [115]
    The reporting sensitivity of the Vaccine Adverse Event ... - PubMed
    Nov 3, 2020 · VAERS sensitivity for capturing anaphylaxis after seven different vaccines ranged from 13 to 76%; sensitivity for capturing GBS after three different vaccines ...
  116. [116]
    Vaccine Adverse Event Reporting System (ESP:VAERS)
    The Electronic Support for Public Health - Vaccine Adverse Event Reporting System (ESP: VAERS) project sought to create a generalizable system to facilitate ...
  117. [117]
    False-positives in spontaneous reporting: should we ... - PubMed
    Spontaneous reporting remains the most used and, undoubtedly, the most cost-effective approach for the identification of adverse drug reactions (ADRs).Missing: signals | Show results with:signals
  118. [118]
    Duplicate Adverse Drug Reactions (ADRs) - a menace. How to ...
    Jun 12, 2025 · These duplicates, whether partial or exact, can distort signal detection, inflate drug-event frequency counts, and compromise the validity of ...
  119. [119]
    Performance of Pharmacovigilance Signal Detection Algorithms for ...
    We provide new insights into the diagnostic potential and characteristics of SDAs routinely applied to FDAs adverse event reporting system.
  120. [120]
    [PDF] Practical Aspects of Signal Detection in Pharmacovigilance - CIOMS
    Introduction and scope of CIOMS VIII​​ A signal in pharmacovigilance was defined by WHO in 2002 as “Reported information on a possible causal relationship ...
  121. [121]
    Conducting and interpreting disproportionality analyses derived ...
    Pilot evaluation of an automated method to decrease false-positive signals induced by co-prescriptions in spontaneous reporting databases. Pharmacoepidemiol ...
  122. [122]
    A New Era in Pharmacovigilance: Toward Real‐World Data ... - NIH
    We outline several different approaches to modern pharmacovigilance, including spontaneous reporting databases, electronic health record monitoring and ...
  123. [123]
    One-Third Of New Drugs Had Safety Problems After FDA Approval
    May 9, 2017 · More than 70 drugs approved by the FDA from 2001 to 2010 ran into safety concerns that prompted withdrawals from the market, ...
  124. [124]
    The hazards of rapid approval of new drugs - PMC - NIH
    Between 1998 and 2013, 27 drugs were approved on limited data and 11 (41%) subsequently received a safety warning or were withdrawn because of safety concerns.
  125. [125]
    Post-marketing withdrawal of 462 medicinal products because of ...
    Feb 4, 2016 · We have identified 462 medicinal products withdrawn because of adverse drug reactions between 1953 and 2013. Hepatotoxicity and immune-mediated ...
  126. [126]
    How FDA Failures Contributed to the Opioid Crisis | Journal of Ethics
    Even in safety trials for opioid drugs approved by the FDA, serious adverse events—including respiratory depression, death, and drug diversion—are common.
  127. [127]
    Potential Signals of Serious Risks/New Safety Information Identified ...
    Sep 30, 2025 · Discover potential signals of serious risks and new safety information identified by the FDA's Adverse Event Reporting System (FAERS).October - December 2024 · January - March 2024 · July - September 2024Missing: techniques | Show results with:techniques
  128. [128]
    Drug & Pharmaceutical Lawsuits | Dangerous Drug Injury Lawyers
    Merck paid a record $4.85 billion to settle thousands of lawsuits from patients who suffered heart attacks, strokes, or sudden cardiac death after using its ...Medication · Dangerous Drugs: Advocating... · Battling Big PharmaMissing: examples | Show results with:examples
  129. [129]
    Top 10 Largest Pharmaceutical Lawsuits & Settlement Amounts
    Criminal fines in this action were $700 million and civil settlements were $800 million. Abbott promoted Depakote as a drug to control agitation and aggression ...
  130. [130]
    Guide to Interpreting VAERS Data
    May 8, 2025 · The term, underreporting refers to the fact that VAERS receives reports for only a small fraction of actual adverse events. The degree of ...
  131. [131]
    The reporting sensitivity of the Vaccine Adverse Event Reporting ...
    Underreporting is a limitation common to passive surveillance systems, including the Vaccine Adverse Event Reporting System (VAERS) that monitors the safety ...
  132. [132]
    VAERS misuse: The zombie practice that keeps coming back
    Nov 15, 2024 · VAERS is a passive reporting system, meaning it is subject to underreporting and lacks the capacity to confirm causation. Use of proportional ...
  133. [133]
    Real-World pharmacovigilance analysis of drug-related ... - Nature
    Apr 18, 2025 · To identify potential adverse event signals, we employed four disproportionality analysis methods: the Reporting Odds Ratio (ROR), Proportional ...
  134. [134]
    2 Current Adverse Event Reporting Systems
    Reports of adverse drug events (ADEs) make their way to the FDA through various means—through pharmaceutical companies, which collect and report adverse events ...
  135. [135]
    Biases in reporting of adverse effects in clinical trials, and potential ...
    Sep 20, 2022 · This MiniReview presents and discusses biases in reporting of safety data in clinical trials and the implications for systematic reviews and guidelines.
  136. [136]
    Biases in reporting of adverse effects in clinical trials, and potential ...
    This MiniReview presents and discusses biases in reporting of safety data in clinical trials and the implications for systematic reviews and guidelines.
  137. [137]
    [PDF] Pharmaceutical advertising biases media reports on drug safety
    Oct 30, 2020 · Our results indicate that pharmaceutical advertising reduces the extent to which the media reports on potential harms of their advertising ...
  138. [138]
    Media coverage of drug regulatory agencies' safety advisories - NIH
    Previous research has shown significant shortcomings in media coverage of medicines, including overstating of benefits and downplaying of harms.
  139. [139]
    Ideological differences in COVID-19 vaccine intention: the effects of ...
    Jan 29, 2024 · We found that leftist ideology and trust in the healthcare system positively predicted vaccination intention, whereas CAM negatively predicted this intention.
  140. [140]
    Vaccine hesitancy: evidence from an adverse events following ...
    Sep 16, 2021 · Our second objective is to identify the potential cognitive biases connecting vaccine hesitancy concerns to vaccine-hesitant behaviors and ...
  141. [141]
    Full article: Entangled evidence: epistemic injustice, and systemic ...
    This paper emphasizes that structural bias in healthcare, health research, and policy accumulates at different stages of pharmacovigilance, shaping the evidence ...