Next Article in Journal
Heterogeneity in Heart Failure with Preserved Ejection Fraction: A Systematic Review of Phenotypic Classifications and Clinical Implications
Next Article in Special Issue
Impact of an Enhanced Disinfection Protocol on the Incidence of Clostridioides difficile Infections and Antibiotic Consumption in a Hospital Setting: A Retrospective Intervention Study
Previous Article in Journal
Reconstruction of the Extensor Apparatus After Total Patellectomy in Orthopedic Oncology: A Systematic Literature Review
Previous Article in Special Issue
Ventriculoperitoneal Shunt Infections: Causative Pathogens and Associated Outcomes from Multiple Hospitals in Saudi Arabia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of Adverse Events Associated with the Sulfamethoxazole/Trimethoprim Combination Drug

1
Department of Pharmacy, Kochi Medical School Hospital, 185-1 Kohasu, Oko-cho, Nankoku-shi 783-8505, Kochi, Japan
2
Graduate School of Integrated Arts and Sciences, Kochi University, Kohasu, Oko-cho, Nankoku-shi 783-8505, Kochi, Japan
3
Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
4
Department of Infection Prevention and Control, Kochi Medical School Hospital; 185-1 Kohasu, Oko-cho, Nankoku-shi 783-8505, Kochi, Japan
5
Division of Public Health, Department of Planning and Coordination, Bureau of Health Security and Management, Japan Institute for Health Security, 1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan
6
Department of Public Health Medicine, Institute of Medicine, and Health Services Research and Development Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
7
AMR Clinical Reference Center, National Center for Global Health and Medicine Japan Institute for Health Security, 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(14), 4819; https://doi.org/10.3390/jcm14144819
Submission received: 3 June 2025 / Revised: 29 June 2025 / Accepted: 4 July 2025 / Published: 8 July 2025

Abstract

Background/Objectives: The combination drug sulfamethoxazole/trimethoprim (ST) is a broad-spectrum antibiotic used against various infections; however, it is associated with several serious adverse events. The ST package inserts contain warnings about these adverse events. However, warnings vary internationally, and specific measures to address ST-related adverse events are unclear. Therefore, we aimed to comprehensively evaluate ST-related adverse events using the Japanese Adverse Drug Event Report (JADER) database and analyze the onset time for each event. Methods: Adverse events due to ST were analyzed using the JADER database between April 2004 and June 2023. The reported odds ratio and 95% confidence interval (95% confidence interval [CI]) were calculated, with a signal detected if the 95% CI lower limit exceeded 1. The Weibull distribution was used to characterize the onset time of adverse events with detected signals. Results: The total number of cases in the JADER database during the study period was 862,952, and the number of adverse events involving ST as a suspected drug was 4203. Adverse events associated with ST include hyperkalemia, syndrome of inappropriate antidiuretic hormone secretion, hematopoietic cytopenia, acute renal failure, hypoglycemia, disseminated intravascular coagulation syndrome, hepatic disorder, and the Stevens–Johnson syndrome/toxic epidermal necrolysis. Conclusions: Weibull analysis indicated an early failure-type onset time for all adverse events, suggesting the need for intensive adverse event monitoring of ST, especially in the first month of use. These findings may support revising drug package inserts in Japan to better reflect the identified risks.

1. Introduction

The sulfamethoxazole/trimethoprim (ST) combination drug comprises two antibiotics: sulfamethoxazole, a sulfonamide that inhibits dihydropteroate synthase, and trimethoprim, an aminopyrimidine that inhibits dihydrofolate reductase. Together, they disrupt tetrahydrofolate biosynthesis, inhibit thymidine synthesis, and exert bacteriostatic effects against bacteria and fungi [1,2]. Trimethoprim has been associated with dose-dependent bone marrow suppression through the inhibition of folate synthesis [3,4,5]. When combined with sulfamethoxazole, the two drugs exert a synergistic antimicrobial effect by sequentially inhibiting two key enzymes in the bacterial folate synthesis pathway: dihydropteroate synthase and dihydrofolate reductase, respectively. This dual blockade more effectively suppresses tetrahydrofolate synthesis, thereby enhancing bacteriostatic activity across a broad range of pathogens. The synergistic mechanism not only broadens the antimicrobial spectrum but also reduces the likelihood of resistance development and permits lower individual drug dosages compared with those in monotherapy [6,7]. ST exhibits a broad spectrum of antimicrobial activity against Gram-positive bacteria, Gram-negative bacteria, and certain fungi. According to guidelines from the Infectious Diseases Society of America (IDSA), ST is recommended as a treatment option for mild-to-moderate skin and soft tissue infections, including those caused by community-acquired methicillin-resistant Staphylococcus aureus [8]. Additionally, the Centers for Disease Control and Prevention, as well as the IDSA, recommend ST as a first-line agent for the treatment and long-term prophylaxis of Pneumocystis pneumonia, especially in immunocompromised individuals. These include individuals with HIV/AIDS, patients undergoing solid organ or hematopoietic stem cell transplantation, those receiving cytotoxic chemotherapy, and those on prolonged high-dose corticosteroids [9,10,11]. Furthermore, ST is widely used in the treatment of urinary tract infections and Nocardia infections, and as prophylaxis in patients with chronic granulomatous disease. While ST remains a central antimicrobial agent, recent studies have raised concerns regarding the emergence of Staphylococcus aureus-resistant strains, including small colony mutants [12,13], particularly in monotherapy, and caution should be exercised when using it long-term. However, ST use is associated with adverse events, which can make treatment continuation difficult. Notably, hematopoietic cytopenia and renal failure are frequently reported adverse events [3,14]. In addition, severe adverse events with a low incidence, such as Stevens–Johnson syndrome/toxic epidermal necrolysis (SJS/TEN), syndrome of inappropriate antidiuretic hormone secretion (SIADH), and severe hypoglycemia, have also been reported. Despite these risks, studies investigating ST-associated adverse events are limited, and the causal relationship remains unclear [15,16,17]. Against this background, side effect warnings have been included in the package inserts of various countries. In Japan, warnings for anaphylactic reactions and hematopoietic cytopenia are listed, while SJS/TEN is identified as a serious adverse effect in the United States and the United Kingdom [18,19,20]. These adverse events are severe and potentially fatal, necessitating measures for their prevention and early detection. To promptly identify these effects and take appropriate action, such as discontinuing or switching medications or providing symptomatic treatment, it is crucial to understand when these events are most likely to occur after treatment begins. However, to our knowledge, no study has statistically evaluated the onset timing of ST-related adverse events using a large database. Existing reports on onset timing are limited to isolated case reports, which do not permit generalizable conclusions or estimates of typical patterns of onset.
We hypothesized that ST-related adverse events would follow an early-onset pattern of clinical significance. To test this hypothesis, we conducted a Weibull analysis using the Japanese Adverse Drug Event Report (JADER), a national database of adverse drug reactions, to validate the onset pattern. Therefore, the aim of this study was to evaluate the detection of ST-related adverse events using the JADER database and determine their timing, thereby clarifying the appropriate monitoring period for each adverse event. Additionally, we aimed to generate information not currently included in the package inserts regarding adverse events associated with ST use in Japan and to provide information for revision of the package inserts.

2. Materials and Methods

2.1. Data Extraction

The JADER database, which is maintained by the Pharmaceuticals and Medical Devices Agency (PMDA), is a collection of voluntary adverse drug reaction reports. Data recorded in the JADER database between April 2004 and June 2023 were obtained from the PMDA website (http://www.pmda.go.jp/; accessed on 10 January 2024). The JADER dataset includes four tables containing the following: (1) patient information (sex, age, and weight); (2) drug information; (3) adverse events and outcomes; and (4) medical history and underlying conditions. These tables were merged using the Fund E-Z Backup Archive v11. 06. 01(Fund E-Z Development Corporation, White Plains, NY, USA) [21].
Overall, 862,952 cases in the JADER dataset were analyzed, and the reported odds ratios (RORs) and 95% confidence intervals (CIs) for ST-related adverse events were calculated using data from these cases. To analyze drug-induced adverse events, cases in which ST was administered orally or intravenously were selected from each dataset. Among these, we selected 4203 cases in which ST was identified as a suspected drug.

2.2. Analysis of the ROR

In JADER, each drug entry includes a classification for “drug involvement”, categorized as “suspect drug,” “concomitant drug,” and “drug interaction.” In this analysis, only cases classified as “suspect drug” were considered indicative of suspected adverse events, while cases involving “concomitant drug” and “drug interaction” were analyzed separately.
The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) and the Medical Dictionary for Regulatory Activities (MedDRA) v24.0 were used to extract adverse events and underlying diseases recorded in the JADER database. We extracted cases in which ST was the suspect drug and counted all adverse events at the MedDRA Preferred Term code (PT) level. The top 40 events were ranked by the number of reports, and included in the analysis. To avoid duplication of similar events, we merged the relevant PTs with reference to the Standardised MedDRA Queries (SMQ) and calculated the ROR for the merged events (Table A1).
RORs and 95% CIs were calculated to analyze ST-associated adverse events. RORs were determined by classifying cases into four categories: (a) individuals administered ST who developed the target adverse events; (b) individuals administered ST who developed other adverse events; (c) individuals administered other drugs who developed the target adverse events; and (d) individuals administered other drugs who developed other adverse events. RORs and 95% CIs were calculated using the following formulae:
R O R = ( a / c )   /   ( b / d ) ,
95 %   C I = e x p   ( l o g   ( R O R )   ±   1.96   ( ( 1 / a ) + ( 1 / b ) + ( 1 / c ) + ( 1 / d ) ) ) ,
RORs were expressed as point estimates with 95% CIs, and statistical analysis was performed using Fisher’s exact test. A signal was detected if the lower limit of the 95% CI of the crude ROR exceeded 1.
The adjusted ROR was calculated based on previous reports of adverse events with the ST combination drug as the suspect drug. Overall, 762,191 cases were included in the analysis, excluding those with missing age and gender reporting. Gender (female), reporting year, and stratified age groups (0–19, 20–29, 30–39, 40–49, 50–59, 60–69, 60–70, and 80+) were coded to construct the logistic model.
Log o d d s = β 0 + β 1 Y + β 2 S + β 3 A + β 4 D
Y = r e p o r t i n g   y e a r , | S = s e x , | A = s t r a t i f i e d   a g e   g r o u p , | D = d r u g
All analyses were conducted using EZR (version 1.51, Saitama Medical Center, Jichi Medical University, Saitama, Japan). Statistical significance was set at p < 0.05. To ensure transparency in annual variations, the total number of reports per year and the percentage of reports related to ST were presented (Table A2).

2.3. Analysis of Onset Time of Adverse Events

We selected 2359 cases (56% of 4203 total cases) in which ST was the suspected drug. Records of the occurrence of complete adverse events and prescription start dates were used for time-to-onset analysis. The onset of adverse events was calculated from the first prescription to the occurrence of adverse events. The median duration, interquartile range (IQR), and Weibull shape parameters were used to analyze the onset data [22,23]. The Kaplan–Meier method was used to illustrate the cumulative incidence of adverse drug events associated with ST [24]. The Weibull shape parameter alpha (α) is the scale of the distribution, with a larger scale indicating a stretched distribution, whereas smaller-scale values shrink the data distribution. The Weibull shape parameter beta (β) defines the shape of the distribution function. The shape parameter β value categorized failure into three groups: if β = 1, the hazard was estimated to be constant over time (random failure type); if β > 1 (with 95% CI greater than 1), the hazard was considered to increase with time (wear-out failure type); and if β and the 95% CI were lower than 1, the hazard was considered to decrease with time (early failure type) [25]. These analyses were conducted using JMP 14.0 (SAS Institute, Mumbai, India).

3. Results

3.1. RORs and Number of Cases of Each Adverse Event Associated with ST

The ROR (95% CI) for each adverse event associated with ST in the JADER database was calculated. Signals for adverse events with ST as the suspected drug were detected for hyperkalemia (ROR: 11.3; 95% CI: 9.85–13.0), SIADH (ROR: 4.67; 95% CI: 3.90–5.55), hematopoietic cytopenia (ROR: 3.53; 95% CI: 3.31–3.79), hypersensitivity (ROR: 2.78; 95% CI: 2.59–2.97), acute renal failure (ROR: 2.39; 95% CI: 2.12–2.68), hypoglycemia (ROR: 2.33; 95% CI: 1.89–2.84), and hepatic disorder (ROR: 2.07; 95% CI: 1.90–2.26) (Table 1). In contrast, signals for SIADH (ROR: 1.37; 95% CI: 1.18–1.58), hematopoietic cytopenia (ROR: 3.79; 95% CI: 3.67–3.91), acute renal failure (ROR: 1.13; 95% CI: 1.04–1.21), and hepatic disorder (ROR: 1.07; 95% CI: 1.01–1.12) were also detected as concomitant or interaction drug signals (Table 1).
Hematopoietic cytopenia was analyzed by subdividing the reduction in each type of hematopoietic cell. Signals for ST as the suspected drug were detected for pancytopenia (ROR: 4.22; 95% CI: 3.75–4.75), erythropenia (ROR: 4.06; 95% CI: 2.93–5.50), leukopenia (ROR: 2.80; 95% CI: 2.58–3.04), and thrombocytopenia (ROR: 2.56; 95% CI: 2.28–2.86), with similar signals observed for concomitant or interacting drugs (Table 1).
Hypersensitivity was analyzed by subdividing the data according to the allergy type. A signal for ST as the suspected drug was detected for SJS/TEN (ROR: 7.19; 95% CI: 6.41–8.04); however, no signal was identified for anaphylactic reactions. No signals were observed for ST as a concomitant or interacting drug in either anaphylactic reactions or SJS/TEN (Table 1).
The adjusted ROR was calculated for each adverse event with ST as the suspect drug using a logistic regression model adjusted for age, gender, and year of report as confounders. The results showed elevated adjusted RORs for several events, including hyperkalemia (adjusted ROR: 9.45; 95%CI: 8.10–1.10), SIADH (adjusted ROR: 4.75; 95%CI: 3.98–5.65), hematopoietic cytopenia (adjusted ROR: 3.56; 95%CI: 3.32–3.81), cytopenia affecting more (adjusted ROR: 4.35; 95%CI: 3.87–4.90), erythropenia (adjusted ROR: 3.65; 95%CI: 2.65–5.02), leukopenia (adjusted ROR: 2.79; 95%CI: 2.57–3.03), thrombocytopenia (adjusted ROR: 2.45; 95%CI: 2.19–2.76), hypersensitivity (adjusted ROR: 2.66; 95%CI: 2.48–2.86), SJS/ TEN (adjusted ROR: 6.74; 95%CI: 6.01–7.56), acute renal failure (adjusted ROR: 2.21; 95%CI: 1.95–2.50), hypoglycemia (adjusted ROR: 2.41; 95%CI: 1.96–2.96), and hepatic disorder (adjusted ROR: 1.98; 95%CI: 1.81–2.17). The same adverse events identified in the crude ROR were also signaled in this model (Table 2).

3.2. Time-to-Onset Analysis by the Weibull Distribution of Adverse Events

Records with complete dates of ST adverse events and prescription start dates (2359 cases, 56% of 4203 total cases) were used for the time-to-onset analysis.
The analysis of the time from the start of ST administration to the onset of adverse events is shown in Figure 1. The change in cumulative incidence over time for ST combination agents is illustrated in the Kaplan–Meier plot (Figure 2). The median (IQR) onset times (days) for ST-related adverse events were as follows: hyperkalemia (8 (5–13)), SIADH (11 (5–15)), hematopoietic cytopenia (24 (11–45)), SJS/TEN (23 (12–44)), acute renal failure (11 (6–40)), hypoglycemia (8 (4–23)), and hepatic disorder (21 (9–52)). All adverse events occurred within the first 2 months of treatment. Time-to-onset analysis of adverse events by ST administration is shown in Figure 1.
Weibull shape parameters for ST-related adverse events were as follows: hyperkalemia (α = 16, 95% CI: 11–21; β = 0.66, 95% CI: 0.59–0.73), SIADH (α = 28, 95% CI: 18–43; β = 0.50, 95% CI: 0.44–0.57), hematopoietic cytopenia (α = 46, 95% CI: 41–51; β = 0.71, 95% CI: 0.68–0.75), SJS/TEN (α = 38, 95% CI: 31–46; β = 0.76, 95% CI: 0.70–0.83), acute renal failure (α = 33, 95% CI: 25–45; β = 0.59, 95% CI: 0.53–0.65), hypoglycemia (α = 28, 95% CI: 16–47; β = 0.52, 95% CI: 0.43–0.61), and hepatic disorder (α = 45, 95% CI: 39–53; β = 0.72, 95% CI: 0.67–0.77). All events were categorized as early failures, with an upper limit of 95% CI and β values < 1, indicating a lower failure rate over time.

4. Discussion

The JADER database has been increasingly utilized to investigate rare and serious adverse drug reactions that are challenging to evaluate through single-center or prospective cohort studies. For instance, previous studies have used JADER to assess the risk of pseudoaldosteronism associated with the traditional Japanese medicine yokukansan, as well as central nervous system ischemia linked to bevacizumab use [21,26]. These studies highlight the utility of JADER in detecting safety signals for adverse events that may be underrepresented in clinical trials or routine observational studies. Building on this approach, the present study is the first to use the JADER to systematically evaluate adverse events associated with ST, with a particular focus on their frequency and onset timing. Signals were detected for several serious adverse events, including SJS/TEN, SIADH, and hypoglycemia. In a logistic regression model adjusted for age, gender, and year of report as confounders for each adverse event with ST as the suspected drug, the adverse events for which signals were detected in adjusted ROR were consistent with those identified in the crude ROR. This consistency suggests that the observed signals are unlikely to be substantially confounded by age, gender, or reporting year. An evaluation of the onset timing of adverse events using the Weibull distribution suggested that these events occur within the first 2 months of drug administration.
Through the inhibition of folate synthesis, trimethoprim has been linked to dose-dependent bone marrow suppression [3,4,5]. In this study, adverse event signals related to myelosuppression were also identified through analysis of the JADER database. The median time of occurrence of myelosuppression-related events was 24 days. Since folate deficiency occurs within 1 month of folate synthesis inhibition [27], the onset date of folate deficiency should coincide with that of thrombocytopenia. ST has also been associated with renal failure due to the inhibition of tubular transporters and hepatic disorders caused by immunogenicity. In this study, analysis of the JADER database similarly detected signals for hematopoietic cytopenia, renal failure, and hepatic disorders.
ST has been reported to cause side effects such as renal dysfunction, SIADH, and hyperkalemia [14,15]. However, trimethoprim is known to inhibit creatinine excretion by blocking the organic cation transporter expressed in the proximal tubules, and there are reports that creatinine blood levels are only transiently increased, independent of kidney function [28]. Therefore, it has remained unclear whether ST causes renal dysfunction. In this study, we investigated these adverse events and detected signals for renal dysfunction, SIADH, and hyperkalemia associated with ST use. Overall, 317 cases of renal dysfunction were reported with ST as the suspect drug. It is known that transient elevations in creatinine are unlikely to be reported in cases where actual renal function is unaffected [29]. Additionally, there have been reports that ST metabolites, owing to their low solubility, can precipitate in urine and cause hematuria, interstitial nephritis, and tubular necrosis [30,31,32]. Based on the above, it is possible that ST affects renal function through mechanisms that are not yet fully understood. The effect of ST on renal dysfunction and its underlying mechanism should be further investigated in future studies. In this study, the time of renal dysfunction onset was 11 (6–41) days. In previous reports, interstitial nephritis has been shown to develop within approximately 1 week of initiation of dosing, which is consistent with our findings [5,32]. It has also been reported that trimethoprim induces hyponatremia and hyperkalemia by inhibiting Na+/K+-ATPase in the distal tubule [33]. Furthermore, trimethoprim is structurally similar to amiloride and is thought to act on epithelial sodium channels in the distal nephron, leading to natriuresis and hyponatremia [15]. Therefore, the hyperkalemia caused by ST may not be secondary to impaired renal function but rather an electrolyte abnormality mediated by transporter inhibition. In the present study, hyperkalemia and SIADH occurred at 11 (5–15) days, most often shortly after taking the drug. Considering that these electrolyte abnormalities are due to transporter inhibition, we believe this onset time is reasonable [15].
Additionally, although case reports of ST-induced liver injury exist [34], the actual situation remains unclear because no large-scale investigative study has been conducted. Drug-induced liver injury can be broadly classified into predictable and unpredictable (idiosyncratic) types [34]. In the case of ST-induced liver injury, it has been suggested that idiosyncratic mechanisms, possibly related to genetic polymorphisms, may be involved [35]. Previous studies have reported that patients with ST-induced liver injury more commonly carry HLA gene polymorphisms, particularly HLA-B*14:01 and HLA-B*35:01, which are known to readily bind sulfamethoxazole [35]. This suggests that sulfamethoxazole forms immune complexes with HLA, activating T cells and leading to hepatocellular damage. Hepatocellular-type liver injury is known to require an incubation period ranging from several days to up to 90 days from the start of use of the suspect drug [34]. The time of onset of hepatocellular damage caused by the ST combination drug in this study was 21 days (9–52 days), which aligns with the presumed mechanism of action.
A case report suggested that ST use may induce severe adverse events, such as anaphylaxis and SJS/TEN, through immunogenic mechanisms [36]. However, few studies have been conducted, and measures to address this issue remain unclear.
Drug package inserts in various countries include warnings; however, the lack of sufficient research has led to differing views.
In Japan, the drug package insert includes warnings about anaphylactic reactions, while in the United States and the United Kingdom, SJS/TEN is listed as a serious adverse event [18,19,20]. In this study, SJS/TEN was detected as a suspected adverse drug reaction, whereas no signal was detected for anaphylactic reactions. These findings suggest that SJS/TEN is more likely to occur as an immunogenic adverse event induced by ST use. Furthermore, similar immunogenic mechanisms have also been reported with other antibiotic classes, such as β-lactams, macrolides, and fluoroquinolones, which have been implicated in severe cutaneous adverse reactions, including SJS/TEN. However, sulfonamides, particularly sulfamethoxazole, have shown a stronger association with these reactions, likely caused by the formation of highly reactive metabolites and specific HLA risk alleles such as HLA-B13:01 and HLA-A11:01, which are more prevalent in certain populations [8].
The proposed mechanism through which SJS/TEN occurs involves the binding of sulfamethoxazole nitroso metabolites to T cells, inducing an immune response based on previous in vitro findings [37]. Drug eruptions caused by type IV hypersensitivity reactions have a delayed onset, with symptoms appearing from one week to several months after the initiation of treatment once sensitization occurs [38]. The time of onset of SJS/TEN in this study was 23 days (IQR: 12–45 days), which is consistent with the hypothesis that SJS/TEN is caused by type IV hypersensitivity.
In addition, basic research using rats has reported that trimethoprim induces hyponatremia and hyperkalemia by inhibiting sodium-potassium ATPase activity in the distal tubules [33]. Trimethoprim, structurally similar to amiloride, has been suggested to affect epithelial sodium channels in the distal nephron, leading to natriuresis and hyponatremia [15]. In this study, signals for SIADH and hyperkalemia were also detected, suggesting that these adverse events can occur in clinical practice. The onset time for these adverse events was 11 days (IQR: 5–15 days) for both hyperkalemia and SIADH, with most cases occurring within the first week after administration. Given that these electrolyte abnormalities are caused by transporter inhibition, the onset time is considered reasonable [15].
The time-to-onset analysis in the 2359 patients with available data showed that serious adverse events, such as hyperkalemia, SIADH, and hypoglycemia, occurred mainly within 2 weeks after treatment initiation, with symptoms occurring as early as 5 days. These findings suggest that electrolyte monitoring, particularly of potassium and sodium levels, should begin within 7 days of therapy, especially in patients with renal dysfunction, older individuals, and those taking concomitant medications that affect electrolyte balance. Furthermore, since these serious events tend to occur early in the course of treatment, it is recommended that ST be prescribed with caution in outpatient settings, with prompt follow-up after the first week and regular monitoring of laboratory values to assess the patient’s condition.
Several case reports have linked the use of ST to severe and potentially fatal hypoglycemia [17,39]. However, no large-scale studies have been conducted, and the clinical characteristics remain unclear [17]. In this study, a signal for hypoglycemia was detected. Previous reports suggest that the sulfonamide structure of sulfamethoxazole induces adenosine triphosphate-sensitive potassium channels and insulin secretion in mice, which may explain the clinical hypoglycemia observed [40]. Sulfonamide derivatives typically cause hypoglycemia within a few hours of administration [41]; therefore, it is likely that ST-induced hypoglycemia can also occur rapidly. The results of this study showed an onset time of 8 days (4–23 days), which is consistent with the expected mechanism of action.
The causative factors of adverse events were classified into dose-dependent and immune-mediated categories. Hematopoietic cytopenia, SIADH, hyperkalemia, and hypoglycemia were more frequently observed in cases with higher daily or cumulative doses, suggesting a dose-dependent mechanism. In contrast, SJS/TEN and hepatic disorders tended to occur regardless of the administered dose, indicating an immune-mediated mechanism.
ST-related adverse events exhibit both regional consistency and variability across global data sources. Hematologic toxicities are consistently reported as common in cohort and pharmacovigilance studies conducted in Asia, Europe, and North America [42,43,44]. However, SIADH and hypoglycemia, which generated strong signals in our JADER analysis, have been rarely reported in Western literature, suggesting potential under-recognition outside Japan. Conversely, SJS/TEN, though rare, is a well-documented serious event across all regions [45,46]. Notably, anaphylaxis is reported more frequently in Japanese case series and reports but is less commonly emphasized in reports from Europe and North America. Pharmacogenomic studies have further identified HLA alleles, such as HLA-B13:01, HLA-A11:01, and HLA-B*15:02, as significant risk factors for sulfonamide-induced severe cutaneous adverse reactions in East Asian populations [47]. This genetic predisposition may partly account for the heightened incidence and regulatory emphasis on severe cutaneous reactions observed in East Asia. These findings underscore the importance of globally harmonized safety information and suggest that SIADH, hypoglycemia, and anaphylaxis may warrant closer epidemiological and pharmacovigilance assessment outside Japan.

Limitations

This study had certain limitations. First, although we used a spontaneous adverse event reporting database, owing to the nature of the database, information on pathogens prompting ST use, comorbidities, and clinical laboratory data was unavailable. The absence of these potential confounding factors may have affected the interpretation of our findings.
Second, the JADER database is subject to inherent limitations associated with spontaneous reporting systems, including underreporting, duplicate submissions, and selection bias [48,49]. Duplicate reports referring to the same case were excluded in this study; however, underreporting remains a significant limitation, as not all suspected adverse events are reported to the PMDA. Moreover, the database includes only reports from patients who experienced adverse events, which may result in a study population that does not fully represent the general clinical population. Additionally, this study did not involve a control group, limiting its ability to draw causal inferences through comparative analysis. These factors may introduce bias in signal detection and restrict the generalizability of the findings. Therefore, our results should be interpreted with caution and further validated through prospective studies that include case and control groups, such as cohort studies and randomized controlled trials.
Third, a high percentage of serious adverse drug reactions tend to be reported in JADER, including a high number of anaphylaxis reports [48]. In this study, 39,117 anaphylaxis reports were recorded in the entire JADER dataset, accounting for 4.5% of all reports. Therefore, the high number of anaphylaxis reports associated with drugs other than ST, coupled with the relatively low number of reports linked to ST, may explain why no signal was detected for ST. This suggests that anaphylaxis reports are relatively infrequent among ST-related adverse events but does not rule out the possibility that anaphylaxis can occur with ST. Therefore, it is necessary to investigate the frequency of anaphylaxis in actual clinical practice and compare its occurrence with that of comparable antimicrobial agents in future studies.
In this study, the identification of side effects specific to ST was enabled, as well as the timing of adverse events. Among them, no signal was detected for anaphylactic reactions, which is warned against in the Japanese package insert. On the other hand, hematopoietic cytopenia, for which a similar warning exists, was detected. As hematopoietic cytopenia is a dose-dependent side effect, it has recently been suggested that the incidence of hematopoietic cytopenia can be suppressed by adjusting the dosage of the drug. However, the JADER database contains many missing data points on dosage, making it challenging to investigate this possibility fully. Therefore, it is necessary to clarify the relationship between dosage and the occurrence of adverse effects through prospective studies.
Consequently, further validation through cohort studies and randomized controlled trials is required. Nevertheless, our study, by utilizing a spontaneous adverse event reporting database, contributes valuable insights into the frequency and timing of adverse events associated with ST.

5. Conclusions

In this study, adverse events such as hematopoietic cytopenia, acute renal failure, hypoglycemia, and SIADH were observed to occur shortly after ST administration. These findings emphasize the need for early monitoring and risk management during treatment. They also highlight the need to revise the Japanese ST package insert to include warnings for hypoglycemia and SIADH. Future prospective studies and international pharmacovigilance efforts incorporating genetic risk factors, such as HLA alleles, are warranted to validate these associations and support the global harmonization of safety information.

Author Contributions

Methodology and Formal analysis: T.I.; Supervision: Y.H.; Writing—original draft: T.S.; Writing—review and editing: K.J., S.M., Y.Y., T.K. (Tomomi Kihara), S.S., M.E. and N.M. Validation: K.A. Investigation: T.M. and T.K. (Takahiro Kato). All authors have read and agreed to the published version of the manuscript.

Funding

The authors declare that financial support was received for the research, authorship, and/or publication of this article. This study was supported by a grant from the Ministry of Health, Labour and Welfare, Japan (rant number 22HA1004).

Institutional Review Board Statement

The Institutional Review Board Statement and Informed Consent Statement are marked as “Not applicable” because this study was conducted using data from the Japanese Adverse Drug Event Report (JADER) database, which is a publicly available and anonymized database provided by the Pharmaceuticals and Medical Devices Agency (PMDA), Japan.

Informed Consent Statement

As the analysis was based on anonymized secondary data with no access to individual patient information, ethical review, and informed consent were not required in accordance with the ethical guidelines of our institution.

Data Availability Statement

The datasets generated and analyzed during the current study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest. The funding agency had no role in the design, execution, analysis, or interpretation of the data.

Abbreviations

The following abbreviations are used in this manuscript:
CIConfidence interval
IDSAInfectious Diseases Society of America
IQRInterquartile range
JADERJapanese Adverse Drug Event Report
MedDRAMedical Dictionary for Regulatory Activities
PMDAPharmaceuticals and Medical Devices Agency
PTPreferred term
RORReported odds ratio
SIADHSyndrome of inappropriate antidiuretic hormone secretion
SJS/TENStevens–Johnson syndrome/toxic epidermal necrolysis
STSulfamethoxazole/trimethoprim

Appendix A

Table A1. Top reported adverse effects of sulfamethoxazole/trimethoprim.
Table A1. Top reported adverse effects of sulfamethoxazole/trimethoprim.
Preferred Term CodeCase (n)
10019670 Hepatic function abnormal237
10035528 Platelet count decreased237
10020646 Hyperkalaemia211
10033661 Pancytopenia210
10037844 Rash199
10001507 Agranulocytosis198
10042033 Stevens–Johnson syndrome198
10073508 Drug reaction with eosinophilia and systemic symptoms187
10013687 Drug eruption187
10047942 White blood cell count decreased174
10062237 Renal impairment168
10022611 Interstitial lung disease165
10044223 Toxic epidermal necrolysis141
10024670 Liver disorder131
10015218 Erythema multiforme125
10069339 Acute kidney injury120
10029354 Neutropenia112
10021036 Hyponatraemia110
10029366 Neutrophil count decreased108
10043554 Thrombocytopenia100
10016288 Febrile neutropenia89
10020993 Hypoglycaemia84
10072268 Drug-induced liver injury77
10002034 Anaemia71
10038428 Renal disorder62
10015150 Erythema61
10028584 Myelosuppression52
10024384 Leukopenia47
10013442 Disseminated intravascular coagulation47
10012735 Diarrhoea45
10025256 Lymphocyte count decreased43
10028813 Nausea40
10003481 Aspartate aminotransferase increased35
10001551 Alanine aminotransferase increased32
10066274 Cytopenia30
10047700 Vomiting28
10048302 Tubulointerstitial nephritis27
10039020 Rhabdomyolysis26
10071583 Haemophagocytic lymphohistiocytosis26
10060795 Hepatic enzyme increased26
Table A2. Annual counts of total adverse event reports and reports related to sulfamethoxazole/trimethoprim.
Table A2. Annual counts of total adverse event reports and reports related to sulfamethoxazole/trimethoprim.
Reporting YearTotal Case (n)Suspected ST, n (%)Concomitant + Interaction ST, n (%)
200424,403118 (0.48)379 (1.55)
200524,168103 (0.43)454 (1.88)
200624,281110 (0.45)568 (2.34)
200725,610157 (0.61)543 (2.12)
200828,593149 (0.52)571 (2.00)
200929,297165 (0.56)843 (2.88)
201032,972169 (0.51)1167 (3.54)
201136,173175 (0.48)1364 (3.77)
201240,778194 (0.48)999 (2.45)
201337,927221 (0.58)1026 (2.71)
201448,451231 (0.48)904 (1.87)
201550,191241 (0.48)1282 (2.55)
201654,929272 (0.50)1174 (2.14)
201760,263365 (0.61)1303 (2.16)
201861,472320 (0.52)1219 (1.98)
201960,604298 (0.49)1108 (1.83)
202051,843253 (0.49)1100 (2.12)
202182,353318 (0.39)1374 (1.67)
202271,877283 (0.39)1530 (2.13)
2023 (first quarter)16,76761 (0.36)275 (1.64)
Total (2004–2023)862,9524203 (0.49)19,183 (2.22)
Abbreviation: ST, sulfamethoxazole/trimethoprim.

References

  1. Minato, Y.; Dawadi, S.; Kordus, S.L.; Sivanandam, A.; Aldrich, C.C.; Baughn, A.D. Mutual potentiation drives synergy between trimethoprim and sulfamethoxazole. Nat. Commun. 2018, 9, 1003. [Google Scholar] [CrossRef] [PubMed]
  2. Gleckman, R.; Blagg, N.; Joubert, D.W. Trimethoprim: Mechanisms of action, antimicrobial activity, bacterial resistance, pharmacokinetics, adverse reactions, and therapeutic indications. Pharmacotherapy 1981, 1, 14–20. [Google Scholar] [CrossRef] [PubMed]
  3. Elajez, R.; Nisar, S.; Adeli, M. Does trimethoprim-sulfamethoxazole prophylaxis induce myelosuppression in primary immune deficiency disease patients; A retrospective, 3 groups comparative study. Asian Pac. J. Allergy Immunol. 2023, 41, 353–360. [Google Scholar] [CrossRef] [PubMed]
  4. Kitazawa, T.; Seo, K.; Yoshino, Y.; Asako, K.; Kikuchi, H.; Kono, H.; Ota, Y. Efficacies of atovaquone, pentamidine, and trimethoprim/sulfamethoxazole for the prevention of Pneumocystis jirovecii pneumonia in patients with connective tissue diseases. J. Infect. Chemother. 2019, 25, 351–354. [Google Scholar] [CrossRef]
  5. Haseeb, A.; Abourehab, M.A.S.; Almalki, W.A.; Almontashri, A.M.; Bajawi, S.A.; Aljoaid, A.M.; Alsahabi, B.M.; Algethamy, M.; AlQarni, A.; Iqbal, M.S.; et al. Trimethoprim-sulfamethoxazole (bactrim) dose optimization in Pneumocystis jirovecii Pneumonia (PCP) management: A systematic review. Int. J. Environ. Res. Public Health 2022, 19, 2833. [Google Scholar] [CrossRef]
  6. Mikasa, K.; Aoki, N.; Aoki, Y.; Abe, S.; Iwata, S.; Ouchi, K.; Kasahara, K.; Kadota, J.; Kishida, N.; Kobayashi, O.; et al. The JAID/JSC Guideline to Clinical Management of Infectious Diseases (Respiratory Infections). Kansenshogaku Zasshi 2014, 88, 1–109. [Google Scholar] [CrossRef]
  7. Cockerill, F.R.; Edson, R.S. Trimethoprim-sulfamethoxazole. Mayo Clin. Proc. 1991, 66, 1260–1269. [Google Scholar] [CrossRef]
  8. Stevens, D.L.; Bisno, A.L.; Chambers, H.F.; Dellinger, E.P.; Goldstein, E.J.C.; Gorbach, S.L.; Hirschmann, J.V.; Kaplan, S.L.; Montoya, J.G.; Wade, J.C. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2014, 59, e10–e52. [Google Scholar] [CrossRef]
  9. Panel on Opportunistic Infections in Adults and Adolescents with HIV. Guidelines for the Prevention and Treatment of Opportunistic Infections in Adults and Adolescents with HIV; U.S. Department of Health and Human Services: Rockville, MD, USA, 2023. Available online: https://clinicalinfo.hiv.gov/en/guidelines/adult-and-adolescent-opportunistic-infection (accessed on 21 June 2025).
  10. Maschmeyer, G.; Carratalà, J.; Buchheidt, D.; Hamprecht, A.; Heussel, C.P.; Kahl, C.; Karthaus, M.; Kern, W.; Kolbe, K.; Krause, R.; et al. Diagnosis and antimicrobial therapy of lung infiltrates in febrile neutropenic patients—Guidelines of the Infectious Diseases Working Party of the German Society of Hematology and Oncology. Eur. J. Cancer 2009, 45, 2462–2472. [Google Scholar] [CrossRef]
  11. Dworkin, M.S.; Hanson, D.L.; Kaplan, J.E.; Jones, J.L.; Ward, J.W. Prophylaxis with trimethoprim-sulfamethoxazole for human immunodeficiency virus–infected patients: Impact on risk for infectious diseases. Clin. Infect. Dis. 2001, 33, 393–398. [Google Scholar] [CrossRef]
  12. Sato, T.; Ito, R.; Kawamura, M.; Fujimura, S. The Risk of Emerging Resistance to Trime-thoprim/Sulfamethoxazole in Staphylococcus aureus. Infect. Drug Resist. 2022, 15, 4779–4784. [Google Scholar] [CrossRef] [PubMed]
  13. Sato, T.; Kawamura, M.; Furukawa, E.; Fujimura, S. Screening method for trimethoprim/sulfamethoxazole-resistant small colony variants of Staphylococcus aureus. J. Glob. Antimicrob. Resist. 2018, 15, 1–5. [Google Scholar] [CrossRef]
  14. Shimizu, Y.; Hirai, T.; Ogawa, Y.; Yamada, C.; Kobayashi, E. Characteristics of risk factors for acute kidney injury among inpatients administered sulfamethoxazole/trimethoprim: A retrospective observational study. J. Pharm. Health Care Sci. 2022, 8, 20. [Google Scholar] [CrossRef]
  15. Marak, C.; Nunley, M.; Guddati, A.K.; Kaushik, P.; Bannon, M.; Ashraf, A. Severe hyponatremia due to trimethoprim-sulfamethoxazole-induced SIADH. SAGE Open Med. Case Rep. 2022, 10, 2050313X221132654. [Google Scholar] [CrossRef]
  16. Fukasawa, T.; Urushihara, H.; Takahashi, H.; Okura, T.; Kawakami, K. Risk of Stevens-Johnson syndrome and toxic epidermal necrolysis associated with antibiotic use: A case-crossover study. J. Allergy Clin. Immunol. Pract. 2023, 11, 3463–3472. [Google Scholar] [CrossRef]
  17. Wang, C.; Fang, W.; Li, Z.; Sun, L. Clinical features, risk factors, diagnosis, and treatment of trimethoprim-sulfamethoxazole-induced hypoglycemia. Front. Endocrinol. 2023, 14, 1059522. [Google Scholar] [CrossRef]
  18. BAKTAR® Combination Tablets/BAKTAR® Mini Combination Tablets/BAKTAR® Combination Granules. Published July 2023. Available online: https://pins.japic.or.jp/pdf/newPINS/00070053.pdf (accessed on 19 June 2024).
  19. DailyMed—Sulfamethoxazole and Trimethoprim Double Strength—Sulfamethoxazole and Trimethoprim Tablet. NPublished 2020. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=804f8363-8ea1-4a57-bbd1-7fd9b6b4d29b (accessed on 19 June 2024).
  20. Co-Trimoxazole 80 mg/400 mg Per 5 mL Adult Suspension—Summary of Product Characteristics (SmPC)-(emc). Published 2021. Available online: https://www.medicines.org.uk/emc/product/6996/smpc (accessed on 19 June 2024).
  21. Ishida, T.; Kawada, K.; Morisawa, S.; Jobu, K.; Morita, Y.; Miyamura, M. Risk factors for pseudoaldosteronism with yokukansan use: Analysis using the Japanese adverse drug report (JADER) database. Biol. Pharm. Bull. 2020, 43, 1570–1576. [Google Scholar] [CrossRef]
  22. Ishida, T.; Kawada, K.; Jobu, K.; Kawazoe, T.; Tamura, N.; Miyamura, M. Analysis of drug-induced liver injury from Bofutsushosan administration using Japanese Adverse Drug Event Report (JADER) database. Biol. Pharm. Bull. 2022, 45, 460–466. [Google Scholar] [CrossRef]
  23. Sauzet, O.; Carvajal, A.; Escudero, A.; Molokhia, M.; Cornelius, V.R. Illustration of the Weibull shape parameter signal detection tool using electronic healthcare record data. Drug Saf. 2013, 36, 995–1006. [Google Scholar] [CrossRef]
  24. Noguchi, Y.; Katsuno, H.; Ueno, A.; Otsubo, M.; Yoshida, A.; Kanematsu, Y.; Sugita, I.; Esaki, H.; Tachi, T.; Tsuchiya, T.; et al. Signals of gastroesophageal reflux disease caused by incretin-based drugs: A disproportionality analysis using the Japanese adverse drug event report database. J. Pharm. Health Care Sci. 2018, 4, 15. [Google Scholar] [CrossRef]
  25. Ohyama, K.; Iida, M.; Akiyama, S.; Yamazaki, H.; Hori, Y. Time-to-onset analysis of rhabdomyolysis due to different proton pump inhibitors using a pharmacovigilance database. In Vivo 2024, 38, 1285–1291. [Google Scholar] [CrossRef] [PubMed]
  26. Sugimoto, S.; Ishida, T.; Kawada, K.; Jobu, K.; Morisawa, S.; Tamura, N.; Takuma, D.; Yoshioka, S.; Miyamura, M. Central nervous system ischemia associated with bevacizumab: An analysis of the Japanese Adverse Drug Event Report database. Biol. Pharm. Bull. 2022, 45, 1805–1811. [Google Scholar] [CrossRef] [PubMed]
  27. Meidahl Petersen, K.; Eplov, K.; Kjær Nielsen, T.; Jimenez-Solem, E.; Petersen, M.; Broedbaek, K.; Daugaard Popik, S.; Kallehave Hansen, L.; Enghusen Poulsen, H.; Trærup Andersen, J. The effect of trimethoprim on serum folate levels in humans: A randomized, double-blind, placebo-controlled trial. Am. J. Ther. 2016, 23, e382–e387. [Google Scholar] [CrossRef] [PubMed]
  28. Delanaye, P.; Mariat, C.; Cavalier, E.; Maillard, N.; Krzesinski, J.M.; White, C.A. Trimethoprim, creatinine and creatinine-based equations. Nephron Clin. Pract. 2011, 119, c187–c193; discussion c193–c194. [Google Scholar] [CrossRef]
  29. Nomura, K.; Takahashi, K.; Hinomura, Y.; Kawaguchi, G.; Matsushita, Y.; Marui, H.; Anzai, T.; Hashiguchi, M.; Mochizuki, M. Effect of database profile variation on drug safety assessment: An analysis of spontaneous adverse event reports of Japanese cases. Drug Des. Devel. Ther. 2015, 9, 3031–3041. [Google Scholar] [CrossRef]
  30. Smith, E.J.; Light, J.A.; Filo, R.S.; Yum, M.N. Interstitial nephritis caused by trimethoprim-sulfamethoxazole in renal transplant recipients. JAMA 1980, 244, 360–361. [Google Scholar] [CrossRef]
  31. Linton, A.L.; Clark, W.F.; Driedger, A.A.; Turnbull, D.I.; Lindsay, R.M. Acute interstitial nephritis due to drugs: Review of the literature with a report of nine cases. Ann. Intern. Med. 1980, 93, 735–741. [Google Scholar] [CrossRef]
  32. Ju, M.; Zheng, M.; Yuan, J.; Lin, D.; Qian, Y. Prevalence and risk factors of trimethoprim/sulfamethoxazole-related acute kidney injury in pediatric patients: An observational study from a public database. Transl. Pediatr. 2022, 11, 1285–1291. [Google Scholar] [CrossRef]
  33. Velázquez, H.; Perazella, M.A.; Wright, F.S.; Ellison, D.H. Renal mechanism of trimethoprim-induced hyperkalemia. Ann. Intern. Med. 1993, 119, 296–301. [Google Scholar] [CrossRef]
  34. Chalasani, N.; Bonkovsky, H.L.; Fontana, R.; Lee, W.; Stolz, A.; Talwalkar, J.; Reddy, K.R.; Watkins, P.B.; Navarro, V.; Barnhart, H.; et al. Features and outcomes of 899 patients with drug-induced liver injury: The DILIN prospective study. Gastroenterology 2015, 148, 1340–1352.e7. [Google Scholar] [CrossRef]
  35. Li, Y.J.; Phillips, E.J.; Dellinger, A.; Nicoletti, P.; Schutte, R.; Li, D.; Ostrov, D.A.; Fontana, R.J.; Watkins, P.B.; Stolz, A.; et al. Human leukocyte antigen B14:01 and B35:01 are associated with trimethoprim-sulfamethoxazole induced liver injury. Hepatology 2021, 73, 268–281. [Google Scholar] [CrossRef] [PubMed]
  36. Khan, D.A.; Knowles, S.R.; Shear, N.H. Sulfonamide hypersensitivity: Fact and fiction. J. Allergy Clin. Immunol. Pract. 2019, 7, 2116–2123. [Google Scholar] [CrossRef]
  37. Illing, P.T.; Mifsud, N.A.; Purcell, A.W. Allotype specific interactions of drugs and HLA molecules in hypersensitivity reactions. Curr. Opin. Immunol. 2016, 42, 31–40. [Google Scholar] [CrossRef] [PubMed]
  38. Naisbitt, D.J.; Hough, S.J.; Gill, H.J.; Pirmohamed, M.; Kitteringham, N.R.; Park, B.K. Cellular disposition of sulphamethoxazole and its metabolites: Implications for hypersensitivity. Br. J. Pharmacol. 1999, 126, 1393–1407. [Google Scholar] [CrossRef]
  39. Rossio, R.; Arcudi, S.; Peyvandi, F.; Piconi, S. Persistent and severe hypoglycemia associated with trimethoprim-sulfamethoxazole in a frail diabetic man on polypharmacy: A case report and literature review. Int. J. Clin. Pharmacol. Ther. 2018, 56, 86–89. [Google Scholar] [CrossRef]
  40. Ogata, H.; Kitamura, S.; Fujiwara, M.; Shimizu, M.; Tan, C.; Zhao, S.; Maejima, Y.; Shimomura, K. Dose dependent effect of sulfamethoxazole on inhibiting KATP channel of mouse pancreatic β cell. Dose Response 2023, 21, 15593258231203611. [Google Scholar] [CrossRef]
  41. Klein-Schwartz, W.; Stassinos, G.L.; Isbister, G.K. Treatment of sulfonylurea and insulin overdose. Br. J. Clin. Pharmacol. 2016, 81, 496–504. [Google Scholar] [CrossRef]
  42. Asmar, B.I.; Maqbool, S.; Dajani, A.S. Hematologic abnormalities after oral trimethoprim-sulfamethoxazole therapy in children. Am. J. Dis. Child. 1981, 135, 1100–1103. [Google Scholar] [CrossRef]
  43. Doodnauth, A.V.; Sivakumar, S.; Mulatu, Y.; Alicea, E.; McFarlane, S.I. Acute severe throm-bocytopenia event associated with trimethoprim-sulfamethoxazole use. Am. J. Med. Case Rep. 2021, 9, 155–157. [Google Scholar] [CrossRef]
  44. Justice, J.; Mukherjee, E.; Martin-Poz, M.; Phillips, E. Updates in the pathogenesis of SJS/TEN. Allergol. Int. 2025, 74, 361–371. [Google Scholar] [CrossRef]
  45. Roujeau, J.C.; Kelly, J.P.; Naldi, L.; Rzany, B.; Stern, R.S.; Anderson, T.; Begaud, B.; Shapiro, S.; Kaufman, D.W.; Brup-pacher, R. Medication use and the risk of Stevens–Johnson syndrome or toxic epidermal necrolysis. N. Engl. J. Med. 1995, 333, 1600–1607. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, E.Y.; Knox, C.; Phillips, E.J. Worldwide prevalence of antibiotic-associated Stevens-Johnson syndrome and toxic epidermal necrolysis: A systematic review and meta-analysis. JAMA Dermatol. 2023, 159, 384–392. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, W.T.; Wang, C.W.; Lu, C.W.; Chen, C.B.; Lee, H.E.; Hung, S.I.; Choon, S.E.; Yang, C.H.; Liu, M.T.; Chen, T.J.; et al. The function of HLA-B*13:01 involved in the pathomechanism of dap-sone-induced severe cutaneous adverse reactions. JAMA Dermatol. 2018, 138, 1546–1554. [Google Scholar] [CrossRef]
  48. Matsuda, S.; Aoki, K.; Kawamata, T.; Kimotsuki, T.; Kobayashi, T.; Kuriki, H.; Nakayama, T.; Okugawa, S.; Sugimura, Y.; Tomita, M.; et al. Bias in spontaneous reporting of adverse drug reactions in Japan. PLoS ONE 2015, 10, e0126413. [Google Scholar] [CrossRef]
  49. Sakai, T. A checklist of important points in research using JADER. Jpn. J. Pharmacoepidemiol. 2020, 25, 64–73. [Google Scholar] [CrossRef]
Figure 1. Time-to-onset analysis of adverse events by sulfamethoxazole/trimethoprim administration. Histograms for (A) hyperkalemia, (B) syndrome of inappropriate secretion of antidiuretic hormone, (C) hematopoietic cytopenia, (D) Stevens–Johnson syndrome/toxic epidermal necrolysis, (E) acute renal failure, (F) hypoglycemia, and (G) hepatic disorders are shown. Abbreviations: 95% CI, 95% confidence interval; IQR, interquartile range; SIADH, syndrome of inappropriate antidiuretic hormone secretion; SJS/TEN, Stevens–Johnson syndrome/toxic epidermal necrolysis.
Figure 1. Time-to-onset analysis of adverse events by sulfamethoxazole/trimethoprim administration. Histograms for (A) hyperkalemia, (B) syndrome of inappropriate secretion of antidiuretic hormone, (C) hematopoietic cytopenia, (D) Stevens–Johnson syndrome/toxic epidermal necrolysis, (E) acute renal failure, (F) hypoglycemia, and (G) hepatic disorders are shown. Abbreviations: 95% CI, 95% confidence interval; IQR, interquartile range; SIADH, syndrome of inappropriate antidiuretic hormone secretion; SJS/TEN, Stevens–Johnson syndrome/toxic epidermal necrolysis.
Jcm 14 04819 g001aJcm 14 04819 g001b
Figure 2. Cumulative incidence of adverse events over time. Kaplan–Meier-like curves illustrate the time to reporting of (A) hyperkalemia, (B) syndrome of inappropriate secretion of antidiuretic hormone, (C) hematopoietic cytopenia, (D) Stevens–Johnson syndrome/toxic epidermal necrolysis, (E) acute renal failure, (F) hypoglycemia, (G) disseminated intravascular coagulation, and (H) hepatic disorder following drug administration, based on data from the spontaneous reporting system. The x-axis represents the number of days from the start of drug administration to adverse event onset, and the y-axis indicates the cumulative reported rate. Shaded areas represent 95% confidence intervals.
Figure 2. Cumulative incidence of adverse events over time. Kaplan–Meier-like curves illustrate the time to reporting of (A) hyperkalemia, (B) syndrome of inappropriate secretion of antidiuretic hormone, (C) hematopoietic cytopenia, (D) Stevens–Johnson syndrome/toxic epidermal necrolysis, (E) acute renal failure, (F) hypoglycemia, (G) disseminated intravascular coagulation, and (H) hepatic disorder following drug administration, based on data from the spontaneous reporting system. The x-axis represents the number of days from the start of drug administration to adverse event onset, and the y-axis indicates the cumulative reported rate. Shaded areas represent 95% confidence intervals.
Jcm 14 04819 g002aJcm 14 04819 g002b
Table 1. Signal detection of each adverse event with sulfamethoxazole/trimethoprim.
Table 1. Signal detection of each adverse event with sulfamethoxazole/trimethoprim.
Adverse EventSuspectedConcomitant + Interaction
CaseNo CaseRatio (%)ROR (95%CI)p ValueCaseNo CaseRatio (%)ROR
(95% CI)
p Value
Hyperkalemia23039735.511.3 (9.85–13.0)<0.00111019,0730.61.08 (0.88–1.31)0.42
SIADH13640673.24.67 (3.90–5.55)<0.00118818,9951.01.37 (1.18–1.58)<0.001
Hematopoietic cytopenia1232297129.33.53 (3.31–3.79)<0.001574413,43929.93.79 (3.67–3.91)<0.001
Cytopenia affecting more31138927.44.22 (3.75–4.75)<0.00190918,2744.72.68 (2.50–2.88)<0.001
Erythropenia4341601.04.06 (2.93–5.50)<0.00110219,0810.52.12 (1.72–2.59)<0.001
Leukopenia701350216.72.80 (2.58–3.04)<0.001414715,03621.64.05 (3.91–4.20)<0.001
Thrombocytopenia33438697.92.56 (2.28–2.86)<0.001226716,91611.84.20 (4.01–4.39)<0.001
Hypersensitivity1167303627.82.78 (2.59–2.97)<0.001165117,5328.60.67 (0.64–0.71)<0.001
SJS/TEN34338608.27.19 (6.41–8.04)<0.00114919,0800.80.61 (0.51–0.72)<0.001
Anaphylactic reaction4641571.10.23 (0.17–0.31)<0.00152218,6612.70.58 (0.53–0.64)<0.001
Acute renal failure31738867.52.39 (2.12–2.68)<0.00171318,4703.71.13 (1.04–1.21)0.002
Hypoglycemia10041032.42.33 (1.89–2.84)<0.0018418,0890.50.41 (0.33–0.51)<0.001
DIC4741561.12.25 (1.64–2.99)<0.00118818,9951.01.99 (1.71–2.31)<0.001
Hepatic disorder589361414.02.07 (1.90–2.26)<0.001148717,6967.81.07 (1.01–1.12)0.021
Interstitial lung disease16540383.90.88 (0.75–1.03)0.1285518,3284.51.01 (0.94–1.08)0.79
Nausea and vomiting symptoms5541481.30.81 (0.61–1.06)0.1432718,8561.71.06 (0.95–1.19)0.28
Noninfectious diarrhea4541581.10.78 (0.57–1.05)0.1138018,8032.01.47 (1.33–1.63)<0.001
Rhabdomyolysis2641770.60.69 (0.45–1.01)0.0595819,1250.30.33 (0.25–0.43)<0.001
Abbreviations: 95% CI, 95% confidence interval; DIC, disseminated intravascular coagulation; ROR, reporting odds ratio; SIADH, syndrome of inappropriate secretion of antidiuretic hormone; SJS/TEN, Stevens–Johnson syndrome/toxic epidermal necrolysis.
Table 2. Adjusted reporting odds ratio of each adverse event with sulfamethoxazole/trimethoprim.
Table 2. Adjusted reporting odds ratio of each adverse event with sulfamethoxazole/trimethoprim.
Adverse EventsAdjusted ROR (95% CI)p-Value
Hyperkalemia9.45 (8.10—1.10)<0.001
SIADH4.75 (3.98—5.65)<0.001
Hematopoietic cytopenia3.56 (3.32—3.81)<0.001
Cytopenia affecting more4.35 (3.87—4.90)<0.001
Erythropenia3.65 (2.65—5.02)<0.001
Leukopenia2.79 (2.57—3.03)<0.001
Thrombocytopenia2.45 (2.19—2.76)<0.001
Hypersensitivity2.66 (2.48—2.86)<0.001
SJS/TEN6.74 (6.01—7.56)<0.001
Anaphylactic reaction0.20 (0.15—0.28)<0.001
Acute renal failure2.21 (1.95—2.50)<0.001
Hypoglycemia2.41 (1.96—2.96)<0.001
Hepatic disorder1.98 (1.81—2.17)<0.001
Interstitial lung disease0.88 (0.75—1.03)0.11
Nausea and vomiting symptoms0.79 (0.60—1.04)0.092
Noninfectious diarrhea0.78 (0.58—1.05)0.099
Rhabdomyolysis0.58 (0.39—0.88)0.010
Abbreviations: 95% CI, 95% confidence interval; ROR, reporting odds ratio; SIADH, syndrome of inappropriate secretion of antidiuretic hormone; SJS/TEN, Stevens–Johnson syndrome/toxic epidermal necrolysis.
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

Sagawa, T.; Ishida, T.; Jobu, K.; Morisawa, S.; Akagaki, K.; Kato, T.; Maruyama, T.; Yagi, Y.; Kihara, T.; Suzuki, S.; et al. Evaluation of Adverse Events Associated with the Sulfamethoxazole/Trimethoprim Combination Drug. J. Clin. Med. 2025, 14, 4819. https://doi.org/10.3390/jcm14144819

AMA Style

Sagawa T, Ishida T, Jobu K, Morisawa S, Akagaki K, Kato T, Maruyama T, Yagi Y, Kihara T, Suzuki S, et al. Evaluation of Adverse Events Associated with the Sulfamethoxazole/Trimethoprim Combination Drug. Journal of Clinical Medicine. 2025; 14(14):4819. https://doi.org/10.3390/jcm14144819

Chicago/Turabian Style

Sagawa, Takaya, Tomoaki Ishida, Kohei Jobu, Shumpei Morisawa, Keita Akagaki, Takahiro Kato, Takumi Maruyama, Yusuke Yagi, Tomomi Kihara, Sanae Suzuki, and et al. 2025. "Evaluation of Adverse Events Associated with the Sulfamethoxazole/Trimethoprim Combination Drug" Journal of Clinical Medicine 14, no. 14: 4819. https://doi.org/10.3390/jcm14144819

APA Style

Sagawa, T., Ishida, T., Jobu, K., Morisawa, S., Akagaki, K., Kato, T., Maruyama, T., Yagi, Y., Kihara, T., Suzuki, S., Endo, M., Matsunaga, N., & Hamada, Y. (2025). Evaluation of Adverse Events Associated with the Sulfamethoxazole/Trimethoprim Combination Drug. Journal of Clinical Medicine, 14(14), 4819. https://doi.org/10.3390/jcm14144819

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