Next Article in Journal
Semaglutide in Diabetic Kidney Disease: Integrating Clinical Evidence with Mechanistic Insights
Previous Article in Journal
Clinical Efficacy of Different Therapies for Painful Shoulder Conditions: A Network Meta-Analysis of Randomized Controlled Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pharmacovigilance Signal Detection of Drug-Induced Hospitalizations and Mortality: A 5-Year Nationwide Study

1
Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
2
Institute of Regulatory Innovation through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
3
College of Pharmacy and Institute of Integrated Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
4
College of Arts and Science, New York University, New York, NY 10003, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Healthcare 2025, 13(22), 2921; https://doi.org/10.3390/healthcare13222921
Submission received: 4 October 2025 / Revised: 3 November 2025 / Accepted: 6 November 2025 / Published: 14 November 2025

Abstract

Background/Objectives: This study aimed to comprehensively characterize the prevalence and patterns of drug-induced hospitalizations and death and to identify predictors strongly associated with drug-induced death. Methods: This study analyzed 29,438 serious adverse event (SAE) reports submitted to the Korea Adverse Event Reporting System (KIDS KAERS DB) database between January 2019 and December 2023. Disproportionality analysis was conducted to detect drug–event associations, and multiple logistic regression was performed to identify independent predictors of mortality. Results: Mortality accounted for 7.53% (n = 2217) and hospitalization for 93.53% (n = 27,532). The strong signals for drug-induced death were observed with steroids (ROR 3.81, 95% CI 3.39–4.27), antidotes (ROR 3.65, 95% CI 2.15–6.18), and anticoagulants (ROR 2.01, 95% CI 1.73–2.34). Immunosuppressants (ROR 9.17, 95% CI 4.75–17.70), diuretics (ROR 3.83, 95% CI 1.42–10.31), and antihyperlipidemics (ROR 3.65, 95% CI 1.72–7.69) were strongly associated with hospitalizations. In multivariate regression, men, aging (OR 1.02, 95% CI 1.02–1.03), use of antidotes (OR 11.37, 95% CI 6.59–19.62), steroids (OR 5.78, 95% CI 4.71–7.08), and anticoagulants (OR 3.60, 95% CI 2.90–4.46) were independent predictors of drug-induced mortality. Conclusions: This study emphasizes the need for targeted surveillance and risk-mitigation strategies focusing on anticoagulants, steroids and immunosuppressants, particularly among elderly and multimorbid populations.

1. Introduction

Adverse drug events (ADEs) represent a major global public health concern, contributing substantially to patient morbidity and mortality [1]. Recent international data indicate that the annual mortality rate attributable to ADEs increased from 2.05 per 100,000 population (95% CI 0.92–3.18) in 2001 to 6.86 (95% CI 5.76–7.95) in 2019, representing more than a threefold rise over two decades [2]. Moreover, VigiBase, the global spontaneous ADE reporting system, contained 36,406,358 ADE reports as of December 2023, marking an 8.68% increase (3,159,452 additional reports) compared with the previous year, implying a continuous global rise in ADEs [3]. Drug-induced hospitalizations account for a considerable proportion of emergency admissions, and severe cases may result in fatal outcomes [4]. Previous study has further reported that ADE-related hospitalizations range from 9.7 to 383 per 100,000 population, whereas mortality rates range from 0.1 to 7.88 per 100,000 [5]. These findings indicate that ADEs are not only frequent occurrences but also clinically significant outcomes, warranting targeted investigation in vulnerable populations, particularly the elderly. Older adults are particularly vulnerable to ADEs due to the increasing complexity of pharmacotherapy associated with multimorbidity, polypharmacy and age-related physiological changes, which collectively amplifies the risk of drug-related hospitalizations and death [6]. With the continued growth of the aging population, the clinical burden of ADE-related hospitalization and mortality is expected to increase further.
Pharmacovigilance, defined as the science and activities related to the detection, assessment, understanding, and prevention of ADEs or other drug-related problems, plays an essential role in ensuring patient safety and optimizing therapeutic outcomes [7]. Among various types of ADEs, those resulting in death, requires hospitalization or the prolongation of existing hospitalizations, persistent or significant disability or incapacity, or birth defect are classified as serious adverse events (SAEs) [8]. The U.S. Food and Drug Administration (FDA) reported that 6.7% of hospitalized patients experience SAEs, with a fatality rate of 0.32%, making drug-related ADEs the 4th leading cause of death, surpassing diabetes, pneumonia, acquired immune deficiency syndrome (AIDS) and pulmonary disease [9]. Despite growing recognition of the clinical and societal burden of ADEs, substantial evidence gaps remain regarding SAEs [10]. In recent years, the utilization of real-world data (RWD) and real-world evidence (RWE) has emerged as an essential approach to bridge the limitations of clinical trials by capturing safety outcomes in real clinical practice. RWD-based pharmacovigilance studies, unlike randomized controlled trials (RCTs), enables identification of rare, delayed, or population-specific adverse events that may not be detected in pre-approval studies [11]. As a regulatory agency, the FDA has released a framework to support RWD use and RWE in regulatory decision-making, thereby reinforcing the legal basis for RWD and its global significance [12]. 1Clinical trials provide important pre-marketing safety data but are limited by relatively small sample sizes, short follow-up periods and narrowly defined patient populations, thereby failing to capture rare yet clinically meaningful events. Therefore, this study was designed to perform signal detection of drug-induced hospitalizations and deaths using a nationwide database. This study aimed to characterize the prevalence and patterns of drug-induced hospitalization and death, and to identify the patient- and treatment-related predictors strongly associated with SAEs.

2. Materials and Methods

2.1. Study Design and Data Collection

This was a retrospective pharmacovigilance database study employing cross-sectional analyses of spontaneous ADE reports, conducted in accordance with the Strengthening the reporting of observational studies in epidemiology (STROBE) guideline [13]. This study analyzed SAEs reports, primarily drug-induced hospitalizations or death, recorded in the Korean Adverse Event Reporting System database (KIDS KAERS DB) between 1 January 2019, and 31 December 2023. KIDS KAERS DB was constructed by the Korean Institute of Drug Safety and & Risk Managements (KIDS, Ministry of Food and Drug Safety). KIDS KAERS DB collects spontaneous ADE reports from healthcare professionals, the general public, hospitals, community pharmacies, pharmaceutical companies, and regional pharmacovigilance centers [14]. In the KIDS KAERS DB, serious outcomes are coded according to predefined International Council for Harmonisation (ICH) E2D categories, including “hospitalization required” and “prolonged hospitalization” [15]. However, these subcategories are coded under a single outcome variable labeled as “hospitalization”. Therefore, this study included both new hospital admissions and prolonged hospitalizations. All medications reported with drug-induced hospitalization or death and assess as ‘certain’, ‘probable/likely’, and ‘possible; causality criteria according to World Health Organization-Uppsala Monitoring Centre (WHO-UMC) criteria, were included in the analysis [16]. To ensure the accuracy and completeness of submitted reports, KIDS performs in-depth investigations to verify the causality. These investigations include interviewing patients and healthcare professionals, reviewing patients’ medical records, and collecting scientific pharmacovigilance data from pharmaceutical manufacturers [14]. Any ADEs classified as ‘unlikely’, ‘conditional/unclassified”, or ‘assessable/unclassifiable’, and those with masked (MSK-coded) etiologic medications, were excluded from the analysis. MSK codes are assigned to medication products that are marketed by fewer than 2 pharmaceutical companies. Duplicate reports were identified and removed prior to analysis, with only the most recent record retained when multiple versions existed. Recurrent reports referring to the same patient but involving distinct ADEs were treated as separate cases. Reports finalized as invalid, lacking any serious outcomes, or in which the adverse events preceded drug administration were excluded. In addition, reports involving non-medicinal products such as vitamins, dietary supplements, and herbal preparations were also removed. ADEs were coded according to the Medical Dictionary for Regulatory Activity (MedDRA) terminology and were further classified into system organ class (SOC) for the statistical analysis. The prespecified criteria for data extraction included (1) patient demographic information (age and sex), (2) ADE information (etiologic medications, occurrence and resolution dates, causality assessment, and seriousness), (3) reporter information, and (4) medical history and concomitant medications. The protocol of utilizing KIDS KAERS DB was approved by KIDS (Ministry of Food and Drug Safety) (KIDS KAERS DB 2503A0002), and the study was exempted from review by the Kyung Hee University Institutional Review Board (IRB) (KHSIRB-25-185).

2.2. Statistical Analysis

Descriptive statistics were used to summarize the demographic and clinical characteristics of patients with SAE. The normality of continuous variables was evaluated using Kolmogorov–Smirnov normality test. As the age did not follow a normal distribution, it was presented as median and interquartile range (IQR). Categorical variables were expressed as frequencies and percentages. Disproportionality analyses were conducted to evaluate the associations between (1) specific drug class and SAE reports related to death and hospitalization, (2) SOC-type and SAE reports related to death and hospitalizations, and (3) sex and SOC categories. Effect sizes were estimated using reporting odds Ratio (ROR) and 95% confidence interval (CI). To ensure the validity and stability of the disproportionality analysis, we restricted inclusion to drug-event pairs with at least 4 reports in both outcome categories: death compared with non-death, and hospitalization compared with non-hospitalizations [17]. This threshold was based on prior methodological validation demonstrating that the reliability and concordance of disproportionality measures become robust when four or more reports constitute a drug-ADE combination [18]. Univariate logistic regression analysis was performed to identify patient- and treatment-specific predictors related to drug-induced death, including variables such as sex, age, hospitalization, and drug class, and predictors associated with significant associations were subsequently entered into multivariate logistic regression model using a forward selection method. Effect size was presented as odds ratios (OR) with corresponding 95% CIs. Sensitivity analyses were performed by excluding chemotherapeutic agents to minimize potential confounding from underlying malignancy-related mortality. In addition, subgroup analyses were conducted according to age (<60 vs. ≥60 years) to explore potential differences in drug-related hospitalization and mortality across age groups, given that studies have reported a substantial increase in polypharmacy prevalence among patients aged 60 years or older [19,20]. Reports with missing age or sex information were excluded from subgroup analyses but retained in overall descriptive and signal-detection analyses to preserve statistical power and minimize selection bias. All statistical analyses were performed using SPSS Statistics 29.0 (IBM Corp., Armonk, NY, USA) and R 4.5.0 (R Foundation for Statistical Computing, Vienna, Austria), and a two-sided p-value of <0.05 was considered statistically significant

3. Results

3.1. Patient Demographic

A total of 23,976,563 ADE cases were initially retrieved from the KIDS KAERS DB between January 2018 and December 2023. After excluding duplicates (n = 8,956,482), inappropriate causality (n = 14,814,661), MSK-coded cases (n = 121,734), invalidated reports (n = 1256), non-SAE cases (n = 19,253) lacking clear temporal relationship (n = 33,426), and non-pharmaceutical product cases (n = 313), a total of 29,438 drug-induced hospitalization or death cases were included in the analysis (Figure 1). The baseline characteristics of included cases are summarized in Table 1. The median age was 65 years (IQR 21 years). The largest proportions were observed among patients aged 60–69 (27.49%) and 70–79 (23.34%) age groups, while those aged ≥80 years accounted for 9.74% of cases. Men represented 54.02% (n = 15,901) of the reports. Drug-induced hospitalization, including new admissions and prolonged hospital stays, comprised 93.53% (n = 27,532), whereas death accounted for 7.53% (n = 2217). By therapeutic class, anticancer drugs were the most frequently implicated (n = 15,196; 51.62%), followed by antibiotics (n = 2632; 8.94%), steroids (n = 2119; 7.20%), and anticoagulants (n = 1566; 5.32%). Other notable contributors included NSAIDs (n = 1182; 4.02%) and immunosuppressants (n = 1157; 3.93%).

3.2. Disproportionality Analysis of Drug-Induced Death or Hospitalizations by Drug Class

The strongest signals for drug-induced death were observed for steroids (ROR 3.81, 95% CI 3.39–4.27), followed by antidotes (ROR 3.65, 95% CI 2.15–6.18), and anticoagulants (ROR 2.01, 95% CI 1.73–2.34) (Table 2). In contrast, markedly lower reporting odds were found for antihyperlipidemic drugs (ROR 0.17, 95% CI 0.07–0.40), diuretics (ROR 0.22, 95% CI 0.08–0.60), analgesics other than nonsteroidal anti-inflammatory drugs (NSAIDs) and opioids (ROR 0.32, 95% CI 0.13–0.77), NSAIDs (ROR 0.51, 95% CI 0.38–0.68), anticonvulsants (ROR 0.60, 95% CI 0.43–0.83), immunosuppressants (ROR 0.69, 95% CI 0.53–0.89), and anticancer drugs (ROR 0.73, 95% CI 0.67–0.80).
For drug-induced hospitalizations, the strongest positive signals were observed for immunosuppressants (ROR 9.17, 95% CI 4.75–17.70), followed by diuretics (ROR 3.83, 95% CI 1.42–10.31), antihyperlipidemic drugs (ROR 3.64, 95% CI 1.72–7.69), antiulcer agents (ROR 3.14, 95% CI 1.40–7.05), other analgesics (ROR 2.68, 95% CI 1.10–6.53), anticoagulants (ROR 1.81, 95% CI 1.39–2.36), anticonvulsants (ROR 1.81, 95% CI 1.26–2.62), and NSAIDs (ROR 1.63, 95% CI 1.22–2.17) (Table 2). On contrary, significantly lower odds of hospitalization were observed for antiviral drugs (ROR 0.41, 95% CI 0.21–0.81), steroids (ROR 0.41, 95% CI 0.36–0.47), antifungals (ROR 0.47, 95% CI 0.23–0.94), and anticancer drugs (ROR 0.70, 95% CI 0.64–0.77).

3.3. Disproportionality Analysis of Drug-Induced Death or Hospitalizations by SOC

The results of disproportionality analysis between drug-induced deaths or hospitalizations and SOC-class are summarized in Table 3. The strongest signals related to drug-induced deaths were observed in myo-, endo-, pericardial & valve disorders (ROR 5.52, 95% CI 3.63–8.39), resistance mechanism disorders (ROR 4.66, 95% CI 4.07–5.32), and respiratory system disorders (ROR 4.44, 95% CI 3.99–4.94). On the other hand, the strongest positive signals for drug-induced hospitalizations were observed for metabolic and nutritional disorders (ROR 3.05, 95% CI 1.85–5.01) and psychiatric disorders (ROR 2.80, 95% CI 1.54–5.11). Additional elevated associations were identified for central and peripheral nervous system disorders (ROR 1.88, 95% CI 1.35–2.62), gastrointestinal disorders (ROR 1.73, 95% CI 1.46–2.05), skin and appendage disorders (ROR 1.72, 95% CI 1.42–2.08), and white cell and reticuloendothelial system (RES) disorders (ROR 1.15, 95% CI 1.02–1.31).

3.4. Predictors of Drug-Induced Mortality

Patient- and treatment-specific predictors associated with increased risk of drug-induced mortality included men, aging (OR 1.02, 95% CI 1.02–1.03) and use of antidotes (OR 11.37, 95% CI 6.59–19.62), steroids (OR 5.78, 95% CI 4.71–7.08) or anticoagulants (OR 3.60, 95% CI 2.90–4.46) (Figure 2). In contrast, predictors associated with decreased risk of drug-induced death involved women (OR 0.56, 95% CI 0.49–0.63), previously hospitalized status (OR 0.02, 95% CI 0.02–0.02), and use of antiulcer agents (OR 0.13, 95% CI 0.03–0.60), antihyperlipidemic drugs (OR 0.25, 95% CI 0.09–0.65), diuretics (OR 0.30, 95% CI 0.10–0.91), and NSAIDs (OR 0.44, 95% CI 0.28–0.69).
Among women, the likelihood of reporting death was the highest for neoplasms (ROR 2.70, 95% CI 1.68–4.33), musculoskeletal system disorders (ROR 1.95, 95% CI 1.59–2.38), and vascular (extracardiac) disorders (ROR 1.79, 95% CI 1.55–2.07) (Figure 3). In contrast, reduced reporting odds were observed for myo-, endo-, pericardial & valve disorders (ROR 0.38, 95% CI 0.24–0.60), vision disorders (ROR 0.54, 95% CI 0.37–0.78), and psychiatric disorders (ROR 0.56, 95% CI 0.45–0.68).

3.5. Sensitivity Analysis

The highest likelihood of reporting drug-induced death was observed with steroids (ROR 3.79, 95% CI 3.34–4.30), followed by antidotes (ROR 3.14, 95% CI 1.85–5.33) and anticoagulants (ROR 1.77, 95% CI 1.51–2.08) (Table 4). On the other hand, the strongest signal for drug-induced hospitalization was observed with immunosuppressants (ROR 7.82, 95% CI 4.04, 15.13), followed by diuretics (ROR 3.16, 95% CI 1.17–8.51), antihyperlipidemic drugs (ROR 3.01, 95% CI 1.42–6.37), antiulcer drugs (ROR 2.59, 95% CI 1.15–5.83), anticoagulant (ROR 1.51, 95% CI 1.15–1.98) and anticonvulsants (ROR 1.50, 95% CI 1.03–2.17).
The highest likelihood of reporting drug-induced death was observed for body as a whole disorders (ROR 3.26, 95% CI 2.84–3.73), followed by respiratory system disorders (ROR 2.01, 95% CI 1.69–2.37), and platelet, bleeding and clotting disorders (ROR 1.70, 95% CI 1.28–2.27) (Table 5). In contrast, the likelihood of reporting drug-induced hospitalization was the highest for central and peripheral nervous system disorders (ROR 12.73, 95% CI 4.75–34.08), followed by urinary system disorders (ROR 1.84, 95% CI 1.24–2.74) and skin and appendages disorders (ROR 1.30, 95% CI 1.07–1.60).

3.6. Subgroup Analysis

In patients aged <60 years, the likelihood of reporting death was highest with anticoagulants (ROR 29.24, 95% CI 22.63–37.78), followed by steroids (ROR 25.61, 95% CI 19.53–33.58) (Table 6). Among patients aged ≥60 years, elevated reporting odds of death were observed with antidotes (ROR 6.29, 95% CI 3.51–11.27), antivirals (ROR 3.15, 95% CI 1.35–7.29), steroids (ROR 2.79, 95% CI 2.36–3.28), metabolic disorder drugs (ROR 1.87, 95% CI 1.18–2.99), opioids (ROR 1.77, 95% CI 1.01–3.13), and immunosuppressants (ROR 1.77, 95% CI 1.01–3.13). Immunosuppressants (ROR 3.76, 95% CI 1.77–7.98) and NSAIDs (ROR 3.42, 95% CI 1.27–9.21) were associated with increased odds of hospitalizations in patients aged <60 years. In contrast, antihyperlidemics (ROR 4.16, 95% CI 1.71–10.09), diuretics (ROR 3.20, 95% CI 1.19–8.64), anticoagulants (ROR 2.40, 95% CI 1.70–3.40), and NSAIDs (ROR 2.27, 95% CI 1.51–3.43) were associated with substantially higher odds of hospitalizations in elderly patients aged 60 years and older.
The reporting odd of drug-induced death in patients aged <60 years was substantially higher with resistance mechanism disorders (ROR 29.41, 95% CI 22.57–38.34), neoplasms (ROR 5.81, 95% CI 2.67–12.65), and metabolic and nutritional disorders (ROR 5.65, 95% CI 3.68–8.69). On the other hand, elderly patients (≥60 years) were more likely to report drug-induced death involving myo-, endo-, pericardial & valve disorders (ROR 6.85, 95% CI 4.39–10.69), respiratory disorders (ROR 3.32, 95% CI 2.92–3.77), urinary system disorders (ROR 1.27, 95% CI 1.03–1.57), and cardiovascular disorders (ROR 1.46, 95% CI 1.05–2.04) (Table 7). On the other hand, the odds of reporting drug-induced hospitalizations were significantly higher with urinary system disorders (ROR 5.35, 95% CI 1.99–14.39) and skin and appendage disorders (ROR 3.02, 95% CI 1.87–4.87) in patients aged <60 years. Elderly patients were more likely to report hospitalizations in red blood cell disorders (ROR 8.33, 95% CI 4.14–16.76), vascular disorders (ROR 5.74, 95% CI 2.56–12.88), respiratory disorders (ROR 3.64, 95% CI 2.65–12.88), metabolic and nutritional disorders (ROR 2.96, 95% CI 1.74–5.05), central and peripheral nervous system disorders (ROR 1.49, 95% CI 1.06–2.09), and gastrointestinal disorders (ROR 1.74, 95% CI 1.42–2.13).

4. Discussion

4.1. Key Findings and Interpretations

This study comprehensively evaluated SAEs reported to the nationwide pharmacovigilance reporting system, KIDS KAERS DB, between 1 January 2019 and 31 December 2023. The majority of cases occurred in patients aged over 60 years, confirming that the elderly population is particularly vulnerable to drug-related complications. ADE cases involving drug-induced hospitalizations accounted for over 90% of events. Strong signals for drug-induced death were observed with steroids, antidotes, and anticoagulants. In contrast, immunosuppressants, diuretics, and antihyperlipidemic agents were strongly associated with hospitalizations. At the SOC-level, the strongest signals related to drug-induced deaths were observed for myocardial, endocardial, pericardial and valve disorders, resistance mechanism disorders, and respiratory system disorders. Conversely, metabolic and nutritional disorders and psychiatric disorders demonstrated the strongest positive signals for drug-induced hospitalizations. Predictors associated with an increased risk of drug-induced mortality included aging, and use of antidotes, steroids or anticoagulants.
Corticosteroids and immunosuppressants were associated with an increased likelihood of serious drug-related outcomes in this study (Table 2 & Figure 2). The observed associations between corticosteroid use and mortality may reflect both drug-related toxicities and the vulnerability of patients for whom steroids are prescribed [21]. Corticosteroids are typically indicated for patients who have chronic inflammatory or autoimmune diseases, and prolonged exposure to corticosteroids is associated with impaired immune function, subsequently increasing susceptibility to severe infections [21]. Furthermore, long-term corticosteroids can exacerbate underlying comorbidities, including metabolic and cardiovascular disorders, thereby further elevating the risk of hospitalizations and death [22,23]. Importantly, drug-induced mortality signals for corticosteroid-associated mortality remained robust in sensitivity analyses, suggesting that this association was not solely attributable confounding from the underlying malignancy.
In contrast, immunosuppressants demonstrated the strongest association with drug-induced hospitalization, consistent with findings from a Spanish pharmacovigilance study [24]. The increased likelihood of hospitalizations among immunosuppressant users can be explained by both the pharmacological properties of these agents and the characteristics of patient populations in which they are used [25]. Immunosuppressants are commonly prescribed in transplant recipients or patients with severe autoimmune disorders, who require intensive prolonged immunosuppression [26]. This trend has been consistently observed across other large-scaled pharmacovigilance datasets, including FDA Adverse Event Reporting System (FAERS), where immunosuppressant related reports often involve severe infections or systemic complications, resulting in hospitalizations [27]. These patients are consequently more susceptible to opportunistic infections, organ dysfunction, and drug–drug interactions, all of which can collectively contribute to higher hospitalization rates [28]. Moreover, in clinical practice, concomitant use of corticosteroids and immunosuppressants can synergistically amplify immunosuppressive effects, thereby further heightening the risk of severe infection and other complications [29]. Hence, considering the heightened vulnerability to severe infections, metabolic complications, and organ dysfunctions, healthcare professionals should adopt active monitoring strategies, including routine assessment of infection risk, metabolic parameters, and comorbidity status, to mitigate preventable adverse outcomes in these high-risk populations. Furthermore, from a regulatory perspective, these findings highlight the need for targeted formulary-based monitoring programs and safety alerts for patients receiving corticosteroids or immunosuppressants.
Similar to the previous study utilizing VigiBase, which also reported a predominance of anticancer drug-related adverse event reports, anticancer agents accounted for 51.62% of all serious cases in our dataset [30]. However, anticoagulant use had the strongest signals associated with serious drug-related outcomes in this study, and this signal remained consistent in the sensitivity analyses (Table 2 and Table 4 & Figure 2). Subgroup analyses further demonstrated that anticoagulant-related mortality was particularly pronounced in patients aged <60 years, whereas elevated hospitalization risks were observed in older adults (≥60 years) (Table 6). Previous studies have reported that approximately 70% of deaths among anticoagulants users were attributable to cardiovascular disease, while only 6% resulted from stroke or embolism [30,31]. This finding suggests that underlying disease burden plays a major role in mortality risk, implying the need for drug safety management strategies that extend beyond bleeding prevention to include comprehensive comorbidity management. The major adverse effects of anticoagulants are well established and primarily include major bleeding events such as intracranial hemorrhage and gastrointestinal bleeding, which represent the leading causes of anticoagulant-associated morbidity and mortality [32]. Bleeding risk is particularly elevated among older adults due to age-related physiological changes, polypharmacy, and the high prevalence of comorbid cardiovascular disease, all of which collectively increase susceptibility to bleeding [33]. Despite the widespread use of anticoagulants, routine bleeding risk monitoring is not uniformly implemented in clinical practice, with the exception of warfarin, which requires regular international normalized ratio (INR) monitoring. Considering the substantial comorbidity burdens of patients prescribed with anticoagulants, this study indicated the need for careful patient selection, individualized dosing, and regular monitoring of anticoagulant therapy, especially in older adults and patients with multiple comorbidities. Moreover, integration of validated bleeding risk assessment tools, standardized bleeding-risk checklists, and patient education on early signs of bleeding should be prioritized to minimize preventable adverse outcomes.
In this study, most antidote-related cases were attributed to naloxone, which is primarily used for the reversal of opioid overdose (Table 2). The strong mortality signal observed with antidote use likely reflects the underlying clinical circumstances in which naloxone is administered, a life-threatening opioid intoxication [34]. Patients receiving naloxone often present with profound respiratory depression, multi-substance overdose, delayed medical presentation, or comorbid organ dysfunction, all of which contribute to high baseline mortality risk [35]. Importantly, the association between antidote use and mortality persisted even in sensitivity analyses excluding chemotherapeutic agents, suggesting that this signal is not fully explained by confounding from malignancy (Table 4). Naloxone remains the standard of care for opioid overdose reversal, with a well-established safety profile [34]. Its primary mechanism of benefit lies in rapidly reversing opioid-induced respiratory depression, thereby preventing mortality. The rapid reversal of opioid-induced respiratory depression is the primary mechanism by which naloxone reduces mortality. However, naloxone administration can occasionally precipitate acute withdrawal symptoms such as vomiting, tachycardia or agitation, especially in opioid-dependent individuals, which in rare cases may contribute to adverse outcomes [36]. Therefore, mortality signal associated with naloxone use should be interpreted with caution, as it is more likely to serve as a marker of the severity of the underlying overdose rather than a direct drug effect. Hence, recognition of opioid overdose, prompt naloxone administration, and close post-overdose monitoring are critical in clinical practice. Moreover, enhanced pharmacovigilance monitoring and registry-based monitoring of naloxone use may further elucidate patient characteristics and outcomes, providing a clearer understanding of overdose severity and improving strategies to reduce mortality among high-risk populations. Furthermore, regulatory policies such as national opioid safety programs, mandatory naloxone distribution protocols, and integration of opioid overdose monitoring into pharmacovigilance system are warranted to mitigate opioid-related hospitalizations and mortalities.
Aging and sex-related differences were identified as significant predictors of drug-induced mortality in this study (Figure 2 and Figure 3). In the context of aging, previous studies have reported consistent findings. For example, a pharmacovigilance study utilizing VigiBase, reported fatal ADEs were predominantly observed in patients over 75 years of age [30]. Older adults are particularly vulnerable to ADEs due to multimorbidity, polypharmacy and age-related physiological changes, including reduced renal and hepatic clearance, and altered pharmacodynamics [6]. These factors contribute to increased susceptibility to drug toxicity. Our subgroup analyses revealed that elderly patients were more likely to report drug-induced deaths related to cardiovascular, respiratory, and urinary system disorders, which involve vital organ systems essential for survival (Table 6 and Table 7). Age-related decline in cardiac reserve, pulmonary function, and renal capacity, in alignment with underlying comorbidities, may amplify the lethality of drug-induced injuries in older adults [6]. Women were associated with a significantly lower likelihood of mortality compared with men, consistent with findings from previous studies [37,38]. The precise underlying mechanisms of sex differences remain incompletely understood, but several biological and clinical factors have been proposed. Men generally differ in body composition, including lower fat mass but higher muscle mass [39]. Moreover, sex-related variations in cytochrome P450 enzyme activity and drug transporter function may also affect pharmacokinetics of drug, leading to differential toxicity risks [40]. Nevertheless, considering that most patients in this population are elderly, the observed sex differences are likely influenced by complex interactions between biological sex, comorbidity burden, and age-related vulnerability. Therefore, further studies are warranted to perform stratified analyses by age, sex, comorbidities and concomitant medication use to determine population-specific risks. Meanwhile, healthcare professionals should adopt clinical risk assessment and monitoring strategies that incorporate patient age, sex, and comorbidity profiles.

4.2. Limitation

This study has several limitations to be acknowledged. First, due to the inherent nature of the voluntary reporting system, reporting bias is likely to be present. Spontaneous reporting depends on the awareness and motivation of healthcare professionals as well as patients, which can lead to both underreporting of less severe cases and overrepresentation of unusual or severe adverse events. Consequently, the database may not fully capture the true incidence or distribution of adverse drug events in the patient populations. Moreover, differences in reporting practices across institutions and over time may have further influenced the observed patterns. Second, the results of this study primarily demonstrate statistical associations rather than causal relationships due to the nature of observational study design. We identified significant association between certain drug classes and mortality or hospitalization; however, definite causal inferences from drug exposure cannot be established. Moreover, several important confounding variables could not be fully addressed due to limited clinical detail in the database. Information on drug dosage, treatment duration, route of administration, and concomitant medications was not consistently available, making it difficult to assess dose–response relationships or drug–drug interactions. Similarly, the severity of underlying comorbidities and the presence of acute illness were not captured, which could have influenced both prescribing decisions and patient outcomes. Fourth, the database did not distinguish between “new hospital admission” and “prolonged hospitalizations” as both were aggregated under a single outcome category of “hospitalization”. Consequently, it was not possible to determine whether drug-related hospitalization represented a new admission event or an extension of an existing hospital stay. Moreover, detailed clinical information such as reason for hospitalization, duration of inpatient care, or outcomes after discharge was not available, which may limit the interpretability of hospitalization related findings. Moreover, the true incidence of ADE could not be calculated as the database lacks exposure denominator. Nevertheless, minimal bias from ADE cases was expected because the Korea Institute of Drug Safety and Risk Management (Ministry of Food and Drug Safety) performs in-depth investigations of ADE reports by collecting scientific pharmacovigilance data from manufacturers, reviewing patients’ medical charts, and consulting with healthcare professionals appointed by the institution. Despite these limitations, this study provides valuable real-world evidence regarding drug-induced hospitalizations and mortality at the nationwide level. This study emphasizes the importance of strengthening pharmacovigilance systems in identifying drug classes and patient subgroups at elevated risk for drug-induced hospitalizations and mortality. Furthermore, routine implementation of clinical monitoring strategies to minimize drug-related risks is crucial for early detection of ADEs, thereby improving patient safety.

5. Conclusions

This nationwide drug surveillance study comprehensively analyzed SAEs involving drug-induced hospitalizations and death from 2019 to 2023. Disproportionality analyses highlighted distinct safety signals: steroids, antidotes, and anticoagulants were most strongly associated with drug-induced deaths, whereas immunosuppressants, diuretics, and antihyperlipidemic drugs had strong associations with drug-induced hospitalizations. SOC analysis further revealed strong signals for cardiovascular, respiratory, and resistance mechanism disorders in fatal cases, and for metabolic, psychiatric, and nervous system disorders in hospitalizations. Age-stratified subgroup analyses demonstrated notable differences in drug classes and clinical conditions contributing to severe outcomes, with younger patients more frequently associated with anticoagulants and steroids, whereas elderly patients exhibited higher reporting frequencies with antifungals, diuretics, anticoagulants, and NSAIDs. Multivariate analysis confirmed that men, aging, use of certain drug classes such as antidotes, steroids, and anticoagulants were independent predictors of drug-induced mortality. These results emphasize the importance of strengthening pharmacovigilance systems for early detection of safety signals in high-risk populations, particularly elderly patients and those receiving drug classes such as antidotes, steroids, anticoagulants, and immunosuppressants for early detection of safety signals. Further studies are warranted to perform stratified analyses by age, sex, comorbidities and concomitant medication use to determine population-specific risks. These results emphasize the importance of integrating pharmacovigilance evidence into national regulatory frameworks to enhance proactive risk monitoring strategies to advance pharmacovigilance policy development, prioritizing surveillance efforts toward high-risk drug classes and population through evidence-based risk assessment and targeted safety interventions.

Author Contributions

Conceptualization, J.M., J.E.L. and Y.J.C.; methodology, J.M., J.E.L. and Y.J.C.; software, J.M. and J.E.L.; validation, E.C. and J.I.; formal analysis, J.M., E.C., J.I. and J.E.L.; investigation, Y.J.C.; resources, J.M.; data curation, J.M. and J.E.L.; writing—original draft preparation, J.M. and J.E.L.; writing—review and editing, Y.J.C.; visualization, E.C. and J.I.; supervision, Y.J.C.; project administration, J.M. and J.E.L.; funding acquisition, Y.J.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study is funded by Ministry of Education [No. 2021R1I1A1A01044500] and Ministry of Food and Drug Safety in 2025 (21153MFDS601).

Institutional Review Board Statement

The protocol of utilizing KIDS KAERS DB was approved by KIDS (Ministry of Food and Drug Safety) (KIDS KAERS DB 2503A0002), and the study was exempted from review by the Kyung Hee University Institutional Review Board (IRB) (KHSIRB-25-185).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author and KIDS due to the institutional policy and ethical concerns.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADEAdverse drug events
CIConfidence interval
NSAIDsNonsteroidal anti-inflammatory drugs
OROdds ratio
RORReporting odds ratio
SAESerious adverse events
SOCSystem Organ Class
WHO-UMCWorld Health Organization-Uppsala Monitoring Centre

References

  1. Sunshine, J.E.; Meo, N.; Kassebaum, N.J.; Collison, M.L.; Mokdad, A.H.; Naghavi, M. Association of adverse effects of medical treatment with mortality in the United States: A secondary analysis of the global burden of diseases, injuries, and risk factors study. JAMA Netw. Open 2019, 2, e187041. [Google Scholar] [CrossRef]
  2. Koyama, T.; Iinuma, S.; Yamamoto, M.; Niimura, T.; Osaki, Y.; Nishimura, S.; Harada, K.; Zamami, Y.; Hagiya, H. International trends in adverse drug event-related mortality from 2001 to 2019: An analysis of the World Health Organization mortality database from 54 Countries. Drug Saf. 2024, 47, 237–249. [Google Scholar] [CrossRef]
  3. Uppsala Monitoring Centre. Annual Report 2023. 2024. Available online: https://who-umc.org/media/yhunvrcz/annual-report-2023.pdf (accessed on 3 November 2025).
  4. Bayoumi, I.; Dolovich, L.; Hutchison, B.; Holbrook, A. Medication-related emergency department visits and hospitalizations among older adults. Can. Fam. Physician 2014, 60, e217–e222. [Google Scholar]
  5. Silva, L.T.; Modesto, A.C.F.; Amaral, R.G.; Lopes, F.M. Hospitalizations and deaths related to adverse drug events worldwide: Systematic review of studies with national coverage. Eur. J. Clin. Pharmacol. 2022, 78, 435–466. [Google Scholar] [CrossRef] [PubMed]
  6. Zazzara, M.B.; Palmer, K.; Vetrano, D.L.; Carfì, A.; Onder, G. Adverse drug reactions in older adults: A narrative review of the literature. Eur. Geriatr. Med. 2021, 12, 463–473. [Google Scholar] [CrossRef]
  7. Campbell, J.E.; Gossell-Williams, M.; Lee, M.G. A review of pharmacovigilance. West. Indian. Med. J. 2014, 63, 771–774. [Google Scholar]
  8. Jensen, M.Q.; Munch, M.W.; Granholm, A.; Møller, M.H.; Bahrenkova, M.; Perner, A. Serious adverse events reporting in recent randomised clinical trials in intensive care medicine—A methodological study protocol. Acta Anaesthesiol. Scand. 2024, 68, 1581–1587. [Google Scholar] [CrossRef]
  9. U.S. Food & Drug Administration. Preventable Adverse Drug Reactions: A Focus on Drug Interactions. 2018. Available online: https://www.fda.gov/drugs/drug-interactions-labeling/preventable-adverse-drug-reactions-focus-drug-interactions (accessed on 3 November 2025).
  10. Jo, H.G.; Jeong, K.; Ryu, J.Y.; Park, S.; Choi, Y.S.; Kwack, W.G.; Choi, Y.J.; Chung, E.K. Fatal events associated with adverse drug reactions in the Korean national pharmacovigilance database. J. Pers. Med. 2021, 12, 5. [Google Scholar] [CrossRef] [PubMed]
  11. Ho, Y.F.; Hu, F.C.; Lee, P.I. The advantages and challenges of using real-world data for patient care. Clin. Transl. Sci. 2019, 13, 4–7. [Google Scholar] [CrossRef] [PubMed]
  12. U.S. Food and Drug Administration. Framework for FDA’s Real-World Evidence Program. 2018. Available online: https://www.fda.gov/media/120060/download?attachment (accessed on 1 November 2025).
  13. von Elm, E.; Altman, D.G.; Egger, M.; Pocock, S.J.; Gøtzsche, P.C.; Vandenbroucke, J.P. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: Guidelines for reporting observational studies. J. Clin. Epidemiol. 2008, 61, 344–349. [Google Scholar] [CrossRef]
  14. Shin, J.Y.; Jung, S.Y.; Ahn, S.H.; Lee, S.H.; Kim, S.J.; Seong, J.M.; Chung, S.Y.; Park, B.J. New initiatives for pharmacovigilance in South Korea: Introducing the Korea Institute of Drug Safety and Risk Management (KID). Pharmacoepidemiol. Drug Saf. 2014, 23, 1115–1122. [Google Scholar] [CrossRef]
  15. International Conference on Harmonisation. Post-Aproval Safety Data Management: Definitions and Standard for Expedites Reporting E2D. Available online: https://database.ich.org/sites/default/files/E2D_Guideline.pdf (accessed on 25 October 2024).
  16. The Uppsala Monitoring Centre. The Use of the WHO-UMC System for Standardised Case Causality Assessment; World Health Organization: Geneva, Switzerland, 2013; Available online: https://www.who.int/docs/default-source/medicines/pharmacovigilance/whocausality-assessment.pdf (accessed on 25 March 2025).
  17. Choi, Y.J.; Choi, C.Y.; Kim, C.U.; Shin, S. A nationwide pharmacovigilance investigation on trends and seriousness of adverse events induced by anti-obesity medication. J. Glob. Health 2023, 13, 04095. [Google Scholar] [CrossRef]
  18. Van Puijenbroek, E.P.; Bate, A.; Leufkens, H.G.M.; Lindquist, M.; Orre, R.; Egberts, A.C.G. A comparison of measures of disproportionality for signal detection in spontaneous reporting systems for adverse drug reactions. Pharmacoepidemiol. Drug Saf. 2002, 11, 3–10. [Google Scholar] [CrossRef] [PubMed]
  19. Michalik-Marcinkowska, U.; Kieltyka-Stowik, A.; Myrcik, D. Is polypharmacy an everyday occurrence for older adults with multiple diseases? J. Public Health, 2025; online ahead of publication. [Google Scholar] [CrossRef]
  20. Lau, S.R.; Waldorff, F.; Holm, A.; Frolich, A.; Andersen, J.S.; Sallerup, M.; Christensen, S.E.; Clausen, S.S.; Due, T.D.; Holmkjaer, P. Disentangling concepts of inappropriate polypharmacy in old age: A scoping review. BMC Public Health. 2023, 23, 245. [Google Scholar] [CrossRef] [PubMed]
  21. Shaikh, S.; Verma, H.; Yadav, N.; Jauhari, M.; Bullangowda, J. Applications of steroid in clinical practice: A review. ISRN Anesthesiol. 2012, 2012, 985495. [Google Scholar] [CrossRef]
  22. Kulkarni, S.; Durham, H.; Glover, L.; Ather, O.; Phillips, V.; Nemes, S.; Cousens, L.; Blomgran, P.; Ambery, P. Metabolic adverse events associated with systemic corticosteroid therapy-a systematic review and meta-analysis. BMJ Open 2022, 12, e061476. [Google Scholar] [CrossRef]
  23. Pujades-Rodriguez, M.; Morgan, A.W.; Cubbon, R.M.; Wu, J. Dose-dependent oral glucocorticoid cardiovascular risks in people with immune-mediated inflammatory diseases: A population-based cohort study. PLoS Med. 2020, 17, e1003432. [Google Scholar] [CrossRef] [PubMed]
  24. Carrasco-Garrido, P.; de Andrés, L.A.; Barrera, V.H.; de Miguel, G.Á.; Jiménez-García, R. Trends of adverse drug reactions related-hospitalizations in Spain (2001–2006). BMC Health Serv. Res. 2010, 10, 287. [Google Scholar] [CrossRef]
  25. Lee, A.Y.; Jeong, J.; Heo, K.N.; Park, S.; Ah, Y.M.; Han, J.M.; Lee, J.Y.; Min, S.I. Complications associated with immunosuppressive agents in solid organ transplant recipients: A nationwide analysis. J. Clin. Med. 2025, 14, 3602. [Google Scholar] [CrossRef]
  26. Opałka, B.; Żołnierczuk, M.; Grabowska, M. Immunosuppressive Agents-Effects on the Cardiovascular System and Selected Metabolic Aspects: A Review. J. Clin. Med. 2023, 12, 6935. [Google Scholar] [CrossRef] [PubMed]
  27. Nakao, S.; Maezawa, M.; Yamashita, M.; Miyasaka, K.; Hirofuji, S.; Ichihara, N.; Nokura, Y.; Sugishita, K.; Yamazaki, T.; Tamaki, H.; et al. Pharmacovigilance study of immunomodulatory drug-related adverse events using spontaneous reporting system databases. Int. J. Immunopathol. Pharmacol. 2025, 39, 3946320251327618. [Google Scholar] [CrossRef] [PubMed]
  28. Bhagat, V.; Pandit, R.A.; Ambapurkar, S.; Sengar, M.; Kulkarni, A.P. Drug interactions between antimicrobial and immunosuppressive agents in solid organ transplant recipients. Indian. J. Crit. Care Med. 2021, 25, 67–76. [Google Scholar]
  29. Marzbani, C.; Bhimaraj, A. Corticosteroids in Immunosuppression. Handb. Exp. Pharmacol. 2022, 272, 73–84. [Google Scholar]
  30. Montastruc, J.L.; Lafaurie, M.; de Canecaude, C.; Durrieu, G.; Sommet, A.; Montastruc, F.; Bagheri, H. Fatal adverse drug reactions: A worldwide perspective in the World Health Organization pharmacovigilance database. Br. J. Clin. Pharmacol. 2021, 87, 4334–4340. [Google Scholar] [CrossRef]
  31. Pokorney, S.D.; Piccini, J.P.; Stevens, S.R.; Patel, M.R.; Pieper, K.S.; Halperin, J.L.; Breithardt, G.; Singer, D.E.; Hankey, G.J.; Hacke, W.; et al. Cause of death and predictors of all-cause mortality in anticoagulated patients with nonvalvular atrial fibrillation: Data from ROCKET AF. J. Am. Heart Assoc. 2016, 5, e002197. [Google Scholar] [CrossRef]
  32. Kocjan, M.; Kosowski, M.; Mazurkiewicz, M.; Muzyk, P.; Nowakowski, K.; Kawecki, J.; Morawiec, B.; Kawecki, D. Bleeding complications of anticoagulation therapy in clinical practice-epidemiology and management: Review of the literature. Biomedicines 2024, 12, 2242. [Google Scholar]
  33. Yamashita, T.; Group, A.R. Impact of polypharmacy on clinical outcomes in elderly patients with nonvalvular atrial fibrillation: Sub-analysis of the ANAFIE Registry. Eur. Heart J. 2021, 42 (Suppl. S1), ehab724.0467. [Google Scholar] [CrossRef]
  34. Wermeling, D.P. Review of naloxone safety for opioid overdose: Practical considerations for new technology and expanded public access. Ther. Adv. Drug Saf. 2015, 6, 20–31. [Google Scholar] [CrossRef]
  35. Dahan, A.; Franko, T.S.; Carroll, J.W.; Craig, D.S.; Crow, C.; Galinkin, J.L.; Garrity, J.C.; Peterson, J.; Rausch, D.B. Fact vs. fiction: Naloxone in the treatment of opioid-induced respiratory depression in the current era of synthetic opioids. Front. Public Health 2024, 12, 1346109. [Google Scholar] [CrossRef]
  36. Go, S.I.; Kim, J.H.; Kang, J.H. Treatment of Opioid Withdrawal Syndrome Triggered by Oxycodone/Naloxone with Dexmedetomidine. J. Hosp. Palliat. Care 2023, 26, 18–21. [Google Scholar] [CrossRef]
  37. Choi, Y.J.; Yang, S.W.; Kwack, W.G.; Lee, J.K.; Lee, T.H.; Jang, J.Y.; Chung, E.K. Comparative safety profiles of sedatives commonly used in clinical practice: A 10-year nationwide pharmacovigilance study in Korea. Pharmaceuticals 2021, 14, 783. [Google Scholar] [CrossRef]
  38. Lee, S.H.; Sung, D.H.; Cho, E.; Min, J.; Shin, S.; Choi, Y.J. Investigation into safety profiles of antiepileptic drugs and identification of predictors for serious adverse events: Insights from national pharmacovigilance data. Pharmaceuticals 2025, 18, 1013. [Google Scholar] [CrossRef] [PubMed]
  39. Bredella, M.A. Sex Differences in Body Composition. Adv. Exp. Med. Biol. 2017, 1043, 9–27. [Google Scholar] [PubMed]
  40. Campesi, I.; Franconi, F. Sex-gender differences in pharmacokinetics. Expert. Opin. Drug Metab. Toxicol. 2025, 21, 491–493. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Data acquisition process.
Figure 1. Data acquisition process.
Healthcare 13 02921 g001
Figure 2. Predictors of drug-induced death risks. The red dotted line indicates a ROR of 1.
Figure 2. Predictors of drug-induced death risks. The red dotted line indicates a ROR of 1.
Healthcare 13 02921 g002
Figure 3. Disproportionality analysis on the association of SOC-based death with women. The red dotted line indicates an ROR of 1.
Figure 3. Disproportionality analysis on the association of SOC-based death with women. The red dotted line indicates an ROR of 1.
Healthcare 13 02921 g003
Table 1. Baseline demographic characteristics of patients.
Table 1. Baseline demographic characteristics of patients.
CharacteristicsNo. of Cases (% Relative Frequency) or Median (IQR)
Age (years) aMedian: 65 IQR: 21
0~91448 (4.92%)
10~191511 (5.13%)
20~29492 (1.67%)
30~39940 (3.19%)
40~491846 (6.27%)
50~593811 (12.95%)
60~698092 (27.49%)
70~796872 (23.34%)
80~892698 (9.17%)
90~169 (0.57%)
Sex b
Men15,901 (54.02%)
Women13,227 (44.93%)
Causality
Certain3540 (12.03%)
Probable/Likely6417 (21.80%)
Possible19,481 (66.18%)
Death
Death2217 (7.53%)
Non-death27,221 (92.47%)
Hospitalization
Hospitalization27,532 (93.53%)
Non-hospitalization1906 (6.47%)
Reporting individuals
Doctors15,727 (53.42%)
Pharmacists2458 (8.35%)
Other healthcare professionals10,178 (34.57%)
General Public642 (2.18%)
Unknown433 (1.47%)
Medication class
Anesthetics8 (0.03%)
Antibiotics2632 (8.94%)
Anticancer drugs15,196 (51.62%)
Anticholinergics9 (0.03%)
Anticoagulants1566 (5.32%)
Anticonvulsants806 (2.74%)
Antidepressants55 (0.19%)
Antidiabetic170 (0.58%)
Antidotes79 (0.27%)
Antiemetic drugs16 (0.05%)
Antifungals70 (0.24%)
Antihistamines56 (0.19%)
Antihormonal drugs39 (0.13%)
Antihyperlipidemic drugs371 (1.26%)
Antiparkinson drugs10 (0.03%)
Antiprotozoal drugs73 (0.25%)
Antipsychotics242 (0.82%)
Antituberculosis drugs468 (1.59%)
Antiulcer276 (0.94%)
Antiviral drugs70 (0.24%)
ARBs147 (0.50%)
Beta blockers73 (0.25%)
Calcium channel blockers94 (0.32%)
Cognitive enhancers79 (0.27%)
Contrast agents87 (0.30%)
Diuretics224 (0.76%)
Genitourinary system drugs59 (0.20%)
Hormonal therapy drugs83 (0.28%)
Hypnotics56 (0.19%)
Immunostimulants27 (0.09%)
Immunosuppressants1157 (3.93%)
Metabolic disorder drugs246 (0.84%)
Muscle relaxants88 (0.30%)
NSAIDs1182 (4.02%)
Opioids147 (0.50%)
Osteoporosis drugs21 (0.07%)
Other analgesic drugs198 (0.67%)
Other blood and blood-forming drugs218 (0.74%)
Other cardiovascular drugs142 (0.48%)
Other drugs412 (1.40%)
Other Gastrointestinal drugs125 (0.42%)
Other neurological drugs5 (0.02%)
Other psychiatric drugs62 (0.21%)
Respiratory system drugs125 (0.42%)
Rheumatoid arthritis and osteoarthritis drugs21 (0.07%)
Steroids2119 (7.20%)
Vaccines29 (0.10%)
a missing in 1559 cases (5.30%). b missing in 310 cases (1.05%).
Table 2. Association of medications class with Drug-induced death or hospitalizations.
Table 2. Association of medications class with Drug-induced death or hospitalizations.
ClassDeath ROR (95%CI)p-ValueHospitalization ROR (95%CI)p-Value
Antibiotics0.96 (0.82–1.12)0.5760.99 (0.84–1.16)0.895
Anticancer drugs0.73 (0.67–0.80)<0.0010.70 (0.64–0.77)<0.001
Anticoagulants2.01 (1.73–2.34)<0.0011.81 (1.39–2.36)<0.001
Anticonvulsants0.60 (0.43–0.83)0.0021.81 (1.26–2.62)0.002
Antidotes3.65 (2.15–6.18)<0.001N/AN/A
Antifungals1.82 (0.90–3.66)0.0960.47 (0.23–0.94)0.034
Antihyperlipidemic drugs0.17 (0.07–0.40)<0.0013.64 (1.72–7.69)<0.001
Antipsychotics0.64 (0.36–1.14)0.1311.80 (0.92–3.51)0.085
Antituberculosis drugs1.02 (0.73–1.44)0.8941.56 (0.99–2.45)0.053
Antiulcer drugsN/AN/A3.14 (1.40–7.05)0.006
Antiviral drugs1.37 (0.63–2.99)0.4350.41 (0.21–0.81)0.010
Diuretics0.22 (0.08–0.60)0.0033.83 (1.42–10.31)0.008
Immunosuppressants0.69 (0.53–0.89)0.0059.17 (4.75–17.70)<0.001
Metabolic disorder drugs1.21 (0.78–1.88)0.400.74 (0.47–1.16)0.189
Muscle relaxants0.74 (0.30–1.82)0.5121.15 (0.47–2.84)0.762
NSAIDs0.51 (0.38–0.68)<0.0011.63 (1.22–2.17)0.001
Opioids1.61 (0.97–2.68)0.0660.66 (0.38–1.14)0.135
Other analgesic drugs0.32 (0.13–0.77)0.0112.68 (1.10–6.53)0.03
Other cardiovascular drugs0.73 (0.36–1.50)0.3920.91 (0.48–1.74)0.783
Respiratory system drugs0.62 (0.27–1.41)0.2511.01 (0.49–2.08)0.973
Steroids3.81 (3.39–4.27)<0.0010.41 (0.36–0.47)<0.001
Table 3. Association between System Organ Class (SOC)-based Death caused by Medications.
Table 3. Association between System Organ Class (SOC)-based Death caused by Medications.
System Organ ClassDeath ROR (95% CI)p-ValueHospitalization ROR (95% CI)p-Value
Skin and appendages disorders0.46 (0.38–0.56)<0.0011.72 (1.42–2.08)<0.001
Central & peripheral nervous system disorders0.57 (0.42–0.77)<0.0011.88 (1.35–2.62)<0.001
Psychiatric disorders0.33 (0.19–0.59)<0.0012.80 (1.54–5.11)<0.001
Gastro-intestinal system disorders0.46 (0.39–0.55)<0.0011.73 (1.46–2.05)<0.001
Liver and biliary system disorders0.44 (0.32–0.62)<0.0011.11 (0.85–1.44)0.448
Metabolic and nutritional disorders0.56 (0.39–0.80)0.0023.05 (1.85–5.01)<0.001
Cardiovascular disorders, general1.37 (0.99–1.91)0.0610.64 (0.46–0.89)0.008
Heart rate and rhythm disorders1.19 (0.67–2.11)0.5460.71 (0.40–1.26)0.244
Vascular (extracardiac) disorders1.01 (0.78–1.32)0.9191.06 (0.79–1.42)0.684
Respiratory system disorders4.44 (3.99–4.94)<0.0010.76 (0.65–0.88)<0.001
Red blood cell disorders0.09 (0.05–0.16)<0.0011.27 (1.00–1.60)0.052
White cell and RES disorders0.37 (0.32–0.43)<0.0011.15 (1.02–1.31)0.027
Platelet, bleeding & clotting disorders0.81 (0.67–0.99)0.0360.66 (0.56–0.78)<0.001
Urinary system disorders1.10 (0.92–1.31)0.3180.86 (0.71–1.04)0.123
Neoplasms2.58 (1.45–4.59)0.001N/AN/A
Body as a whole—general disorders1.58 (1.40–1.78)<0.0010.50 (0.44–0.57)<0.001
Resistance mechanism disorders4.66 (4.07–5.32)<0.0010.57 (0.48–0.69)<0.001
Secondary terms—events0.26 (0.10–0.70)0.0080.25 (0.18–0.36)<0.001
Abbreviation: RES: reticuloendothelial system.
Table 4. Sensitivity analysis on drug-induced death and hospitalization.
Table 4. Sensitivity analysis on drug-induced death and hospitalization.
ClassDeath Reports
(n = 1234)
Death ROR (95% CI)p-ValueHospitalization Reports
(n = 13,477)
Hospitalization ROR (95% CI)p-Value
Antibiotics1910.79 (0.68–0.93)0.00524600.77 (0.65–0.92)0.003
Anticoagulants2111.77 (1.51–2.08)<0.00115071.51 (1.15–1.98)0.003
Anticonvulsants380.51 (0.36–0.71)<0.0017761.50 (1.03–2.17)0.034
Antidotes183.14 (1.85–5.33)<0.00179N/AN/A
Antifungals91.56 (0.77–3.15)0.215610.38 (0.19–0.77)0.007
Antihyperlipidemic drugs50.14 (0.06–0.34)<0.00173.01 (1.42–6.37)0.004
Antipsychotics120.55 (0.30–0.98)0.04291.48 (0.76–2.89)0.253
Antituberculosis drugs360.88 (0.62–1.24)0.447201.28 (0.81–2.02)0.285
Antiulcer drugs2N/AN/A62.59 (1.15–5.83)0.022
Antiviral drugs71.17 (0.54–2.57)0.691100.34 (0.17–0.66)0.002
Diuretics40.19 (0.07–0.51)<0.00143.16 (1.17–8.51)0.023
Immunosuppressants620.58 (0.44–0.75)<0.00197.82 (4.04–15.13)<0.001
Metabolic disorder drugs221.04 (0.67–1.61)0.876210.60 (0.38–0.95)0.028
Muscle relaxants50.63 (0.26–1.57)0.32350.94 (0.38–2.33)0.897
NSAIDs480.42 (0.32–0.57)<0.001491.34 (1.00–1.80)0.052
Opioids171.38 (0.83–2.30)0.211140.54 (0.31–0.93)0.027
Other analgesic drugs50.27 (0.11–0.66)0.00452.21 (0.91–5.38)0.081
Other cardiovascular drugs80.63 (0.31–1.28)0.201100.75 (0.39–1.43)0.376
Respiratory system drugs60.53 (0.23–1.20)0.25180.83 (0.40–1.70)0.609
Steroids4433.79 (3.34–4.30)<0.0012820.27 (0.23–0.32)<0.001
Table 5. Association between System Organ Class (SOC)-based drug induced death and hospitalization.
Table 5. Association between System Organ Class (SOC)-based drug induced death and hospitalization.
System Organ ClassDeath Reports
(n = 1234)
Death ROR (95% CI)p-ValueHospitalization Reports
(n = 13,477)
Hospitalization
ROR (95% CI)
p-Value
Skin and appendages disorders1100.40 (0.32–0.48)<0.00125691.30 (1.07–1.60)0.01
Central & peripheral nervous system disorders50.06 (0.02–0.14)<0.00184512.73 (4.75–34.08)<0.001
Psychiatric disorders110.66 (0.36–1.22)0.1891750.99 (0.52–1.89)0.984
Gastro-intestinal system disorders1010.69 (0.56–0.85)<0.00114960.84 (0.68–1.04)0.117
Liver and biliary system disorders280.50 (0.34–0.73)<0.0015851.46 (0.96–2.24)0.078
Metabolic and nutritional disorders280.54 (0.37–0.79)0.0025542.69 (1.51–4.79)<0.001
Cardiovascular disorders, general40.20 (0.07–0.54)0.0012061.68 (0.79–3.58)0.179
Heart rate and rhythm disorders121.09 (0.60–1.98)0.7741160.55 (0.30–0.99)0.047
Vascular (extracardiac) disorders590.96 (0.73–1.26)0.748470.79 (0.58–1.07)0.126
Respiratory system disorders1862.01 (1.69–2.37)<0.00111550.71 (0.57–0.90)0.004
Red blood cell disorders110.21 (0.12–0.38)<0.001548N/AN/A
White cell and RES disorders250.42 (0.28–0.63)<0.0016071.40 (0.93–2.10)0.108
Platelet, bleeding & clotting disorders561.70 (1.28–2.27)<0.0013891.11 (0.70–1.75)0.661
Urinary system disorders470.61 (0.45–0.82)0.0018201.84 (1.24–2.74)0.003
Neoplasms81.60 (0.76–3.36)0.2261N/AN/A
Body as a whole—general disorders3383.26 (2.84–3.73)<0.00114730.31 (0.26–0.37)<0.001
Resistance mechanism disorders2018.1 (6.71–9.79)<0.0014530.47 (0.35–0.63)<0.001
Abbreviation: RES: reticuloendothelial system.
Table 6. Subgroup analysis on likelihood off reporting drug-induced death or hospitalization per drug class.
Table 6. Subgroup analysis on likelihood off reporting drug-induced death or hospitalization per drug class.
ClassAge < 60 Death Reports (n = 409)Age < 60 Death ROR (95%CI)
(n = 409)
p-ValueAge ≥ 60 Death Reports
(n = 1521)
Age ≥ 60 Death ROR (95%CI)
(n = 1521)
p-ValueAge < 60 Hospitalization Reports
(n = 9727)
Age < 60 Hospitalization ROR (95%CI)
(n = 9727)
p-ValueAge ≥ 60 Hospitalization Reports
(n = 16,597)
Age ≥ 60 Hospitalization ROR (95%CI)p-Value
Antibiotics511.07 (0.79–1.44)0.671401.19 (0.99–1.43)0.06611340.68 (0.5–0.93)0.01413030.79 (0.65–0.96)0.019
Anticancer drugs560.16 (0.12–0.21)<0.0018310.94 (0.84–1.04)0.2146520.50 (0.04–0.63)<0.00192730.84 (0.75–0.95)0.004
Anticoagulants13829.24 (22.63–37.78)<0.001480.48 (0.36–0.64)<0.001300N/AN/A10572.40 (1.70–3.40)<0.001
Anticonvulsants0N/AN/A381.23 (0.88–1.73)0.227425N/AN/A3430.94 (0.64–1.40)0.773
Antidotes0N/AN/A186.29 (3.51–11.27)<0.00130N/AN/A49N/AN/A
Antifungals85.47 (2.52–11.88)<0.0011N/AN/A350.14 (0.07–0.31)<0.00124N/AN/A
Antihyperlipidemic drugs0N/AN/A50.19 (0.08–0.47)<0.00187N/AN/A2764.16 (1.71–10.09)<0.001
Antipsychotics121.52 (0.84–2.75)0.1660N/AN/A1910.69 (0.35–1.37)0.29242N/AN/A
Antituberculosis drugs0N/AN/A361.46 (1.02–2.07)0.036165N/AN/A2831.05 (0.67–1.66)0.825
Antiulcer0N/AN/A2N/AN/A199N/AN/A710.88 (0.38–2.03)0.763
Antiviral drugs0N/AN/A73.14 (1.35–7.29)0.00830N/AN/A230.21 (0.10–0.48)<0.001
Diuretics0N/AN/A40.25 (0.09–0.67)0.00644N/AN/A1713.20 (1.19–8.64)0.022
Immunosuppressants120.36 (0.20–0.64)<0.001391.40 (1.00–1.96)0.037523.76 (1.77–7.98)<0.001339N/AN/A
Metabolic disorder drugs1N/AN/A211.87 (1.18–2.99)0.00894N/AN/A1230.48 (0.29–0.78)0.003
Muscle relaxants0N/AN/A51.01 (0.40–2.54)0.9830N/AN/A530.79 (0.31–1.97)0.610
NSAIDs2N/AN/A250.36 (0.24–0.55)<0.0014023.42 (1.27–9.21)0.0157162.27 (1.51–3.43)<0.001
Opioids3N/AN/A141.77 (1.01–3.13)0.04848N/AN/A850.45 (0.25–0.79)0.006
Other analgesic drugs0N/AN/A50.56 (0.23–1.37)0.20387N/AN/A961.43 (0.58–3.52)0.436
Other cardiovascular drugs0N/AN/A80.88 (0.43–1.82)0.73837N/AN/A950.71 (0.37–1.36)0.294
Respiratory system drugs0N/AN/A60.95 (0.41–2.18)0.89646N/AN/A680.84 (0.37–1.94)0.687
Steroids11525.61 (19.53–33.58)<0.0012002.79 (2.36–3.28)<0.0012562.14 (0.79–5.79)0.1338730.35 (0.30–0.42)<0.001
Table 7. Subgroup analysis on the association between System Organ Class (SOC)-based Death caused by Medications.
Table 7. Subgroup analysis on the association between System Organ Class (SOC)-based Death caused by Medications.
System Organ ClassAge < 60 Death Reports (n = 409)Age < 60 Death ROR (95% CI)p-ValueAge < 60 Hospitalization Reports
(n = 9727)
Age < 60 Hospitalization ROR (95% CI)p-ValueAge ≥ 60 Death Reports
(n = 1521)
Age ≥ 60 Death ROR (95% CI)p-ValueAge ≥ 60 Hospitalization Reports
(n = 16,597)
Age ≥ 60 Hospitalization ROR (95% CI)p-Value
Skin and appendages disorders190.27 (0.17–0.43)<0.00114783.02 (1.87–4.87)<0.001910.75 (0.60–0.93)0.00812770.97 (0.78–1.20)0.753
Central & peripheral nervous system disorders2N/AN/A211N/AN/A440.66 (0.49–0.90)0.0097111.49 (1.06–2.09)0.022
Psychiatric disorders60.82 (0.36–1.86)0.6341731.14 (0.47–2.80)0.76860.24 (0.11–0.55)<0.0012603.26 (1.45–7.33)0.004
Gastro-intestinal system disorders320.68 (0.47–0.98)0.03910570.77 (0.56–1.06)0.1101130.47 (0.39–0.58)<0.00123711.74 (1.42–2.13)<0.001
Liver and biliary system disorders181.03 (0.64–1.68)0.8933990.42 (0.28–0.61)<0.001180.34 (0.21–0.54)<0.0015501.38 (0.95–2.00)0.093
Metabolic and nutritional disorders275.65 (3.68–8.69)<0.001144N/AN/A40.08 (0.03–0.20)<0.0015452.96 (1.74–5.05)<0.001
Cardiovascular disorders, general0N/AN/A45N/AN/A401.46 (1.05–2.04)0.0263000.61 (0.43–0.87)0.006
Myo-, endo-, pericardial & valve disorders0N/AN/A21N/AN/A326.85 (4.39–10.69)<0.00181N/AN/A
Heart rate and rhythm disorders0N/AN/A51N/AN/A101.54 (0.79–2.99)0.026700.52 (0.27–1.01)0.053
Vascular (extracardiac) disorders0N/AN/A263N/AN/A160.38 (0.23–0.63)<0.0014535.74 (2.56–12.88)<0.001
Respiratory system disorders181.9 (1.16–3.10)0.010245N/AN/A3833.32 (2.92–3.77)<0.00118433.64 (2.65–12.88)<0.001
Red blood cell disorders0N/AN/A456N/AN/A110.13 (0.07–0.24)<0.0018568.33 (4.14–16.76)<0.001
White cell and RES disorders80.07 (0.04–0.15)<0.00120050.65 (0.51–0.83)<0.0011640.58 (0.49–0.69)<0.00127650.98 (0.84–1.14)0.745
Platelet, bleeding & clotting disorders340.81 (0.67–0.99)0.8797901.05 (0.69–1.58)0.829770.93 (0.73–1.18)0.5628200.41 (0.34–0.49)<0.001
Urinary system disorders311.26 (0.87–1.84)0.2246155.35 (1.99–14.39)<0.0011051.27 (1.03–1.57)0.0248840.53 (0.43–0.64)<0.001
Neoplasms85.81 (2.67–12.65)<0.00141N/AN/A0N/AN/A33N/AN/A
Body as a whole—general disorders761.70 (1.31–2.20)<0.00111851.21 (0.84–1.75)0.3052731.93 (1.68–2.23)<0.00116040.30 (0.26–0.34)<0.001
Application site disorders0N/AN/A59N/AN/A0N/AN/A33N/AN/A
Resistance mechanism disorders12829.41 (22.57–38.34)<0.0012570.46 (0.28–0.75)0.0021312.20 (1.81–2.68)<0.0017581.36 (0.99–1.86)0.058
Secondary terms—events1N/AN/A670.05 (0.03–0.08)<0.0010N/AN/A71N/AN/A
Abbreviation: RES: reticuloendothelial system.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Min, J.; Lee, J.E.; Cho, E.; Im, J.; Choi, Y.J. Pharmacovigilance Signal Detection of Drug-Induced Hospitalizations and Mortality: A 5-Year Nationwide Study. Healthcare 2025, 13, 2921. https://doi.org/10.3390/healthcare13222921

AMA Style

Min J, Lee JE, Cho E, Im J, Choi YJ. Pharmacovigilance Signal Detection of Drug-Induced Hospitalizations and Mortality: A 5-Year Nationwide Study. Healthcare. 2025; 13(22):2921. https://doi.org/10.3390/healthcare13222921

Chicago/Turabian Style

Min, Jeongah, Jeong Eon Lee, Eunah Cho, Jayoung Im, and Yeo Jin Choi. 2025. "Pharmacovigilance Signal Detection of Drug-Induced Hospitalizations and Mortality: A 5-Year Nationwide Study" Healthcare 13, no. 22: 2921. https://doi.org/10.3390/healthcare13222921

APA Style

Min, J., Lee, J. E., Cho, E., Im, J., & Choi, Y. J. (2025). Pharmacovigilance Signal Detection of Drug-Induced Hospitalizations and Mortality: A 5-Year Nationwide Study. Healthcare, 13(22), 2921. https://doi.org/10.3390/healthcare13222921

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop