Skip to main content

Liver injury in children: signal analysis of suspected drugs based on the food and drug administration adverse event reporting system

Abstract

Background

Evidence of drug-induced liver injury is abundant in adults but is lacking in children. Our aim was to identify suspected drug signals associated with pediatric liver injury.

Methods

Hepatic adverse events (HAEs) among children reported in the Food and Drug Administration Adverse Event Reporting System were analyzed. A descriptive analysis was performed to summarize pediatric HAEs, and a disproportionality analysis was conducted by evaluating reporting odds ratios (RORs) and proportional reporting ratios to detect suspected drugs.

Results

Here, 14,143 pediatric cases were reported, specifically 49.6% in males, 45.1% in females, and 5.2% unknown. Most patients (68.8%) were 6–18 years old. Hospitalization ranked first among definite outcomes (7,207 cases, 37.2%). In total, 264 disproportionate drug signals were identified. The top 10 drugs by the number of reports were paracetamol (1,365; ROR, 3.6; 95% confidence interval (CI), 3.4–3.8), methotrexate (878; ROR, 2.5; 95% CI, 2.3–2.7), vincristine (649; ROR, 3.0; 95% CI, 2.8–3.3), valproic acid (511; ROR, 3.2; 95% CI, 2.9–3.6), cyclophosphamide (490; ROR, 2.4; 95% CI, 2.2–2.6), tacrolimus (427; ROR, 2.4; 95% CI, 2.2–2.7), prednisone (416; ROR, 2.1; 95% CI, 1.9–2.3), prednisolone (401; ROR, 2.3; 95% CI, 2.1–2.5), etoposide (378; ROR, 2.3; 95% CI, 2.1–2.6), and cytarabine (344; ROR, 2.8; 95% CI, 2.5–3.2). After excluding validated hepatotoxic drugs, six were newly detected, specifically acetylcysteine, thiopental, temazepam, nefopam, primaquine, and pyrimethamine.

Conclusions

The hepatotoxic risk associated with 264 signals needs to be noted in practice. The causality of hepatotoxicity and mechanism among new signals should be verified with preclinical and clinical studies.

Peer Review reports

Background

Drug-induced liver injury (DILI) refers to liver injury caused by the direct toxicity of a drug or its metabolites or by idiosyncratic reactions after exposure to drugs [1,2,3]. More than 1,100 drugs have been confirmed to be hepatotoxic worldwide, including non-steroidal anti-inflammatory drugs, anti-infective drugs (including anti-tuberculosis drugs), anti-tumor drugs, cardiovascular system drugs, and biological agents [3]. The annual incidence of DILI is estimated to be approximately 23.80 per 100,000 in the general population of China and 12 per 100,000 in Korea [4, 5]. In a prospective study in the US, 11.1% of cases of acute liver failure (ALF) were attributed to DILI [6]. DILI is also an important cause of failures in pharmaceutical research and development, black box warnings added to post-marketing drugs, and drug withdrawals [7].

Children are vulnerable to hepatotoxicity owing to the immaturity of their liver, lack of exclusive drugs, shortage of research data, and poor self-reporting [8,9,10,11,12]. A multicenter study conducted 17 years before this study showed that DILI accounts for 19% of ALF cases in children and that it is the leading cause of pediatric ALF [13]. It is thus essential to conduct studies on pediatric DILI to prevent potential risks for children. In one study, data from 2000 to 2006 were analyzed based on VigiBase and hepatotoxic drugs were compared between children and adults, this information seems insufficient to guide current practice as pediatric medications have been markedly changed [8]. From the implementation of the Pediatric Research Equity Act in 2003 to the end of 2018, the Food and Drug Administration (FDA) has accepted over 500 changes to drug labels, most of which expand the population to children [14]. Moreover, most current evidence of pediatric DILI still focuses on case series, and studies based on a large sample size are scarce. This study aimed to characterize hepatic adverse events (HAEs) in children based on the Food and Drug Administration’s Adverse Event Reporting System (FAERS) and to detect suspected drugs associated with liver injury.

Methods

Data source

We collected HAEs that might be associated with drugs among people under 18 years of age from 2004Q1 to 2020Q2 in FAERS. FAERS is a large database that supports the FDA’s post-marketing surveillance program for drugs and therapeutic biologicals and has been publicly accessible since 2004 [15]. This database contains adverse event (AE) reports, medication error reports, and product quality complaints submitted by healthcare professionals, consumers, and manufacturers worldwide. For consistency among heterogeneous reporters, all AEs in FAERS were encoded based on the Medical Dictionary for Regulatory Activities (MedDRA) [15]. With a magnitude of reports from the real world, FAERS has the advantage of being widely utilized to detect pharmacovigilant signals and support safe medication in clinical practice [16,17,18].

Procedures

In this study, we adopted OpenVigil 2.1 to query liver injury reports from FAERS. OpenVigil 2.1 is a publicly-available and validated tool used for extracting, cleaning, mining, and analyzing data from FAERS [19]. The advantages of this tool, such as being free of charge, its use of clean data, and its convenience for precise results, have made it widely applied in pharmacovigilance studies [20,21,22]. By summarizing terms associated with liver injury, which might be drug-related, from previous literature, MedDRA 21.1, and standardized MedDRA queries [23,24,25], we completed our query (Supplementary Table 1). We then unified the drug names into generic names, removed duplicate records, and excluded non-drug reports (iron, aluminum hydroxide, amino acids, etc.).

Statistical analysis

A descriptive analysis was performed to summarize all pediatric HAEs in terms of age, sex, country, and outcomes. Disproportionality analysis was conducted to quantitatively assess the association between drugs and target AEs by calculating the ratio of target AEs to other AEs in a database and providing the putative relevance from a statistical perspective, which has been extensively applied in pharmacovigilance to evaluate drug–event or vaccine–event relationships for decades [26,27,28]. We completed a disproportionality analysis based on a 2 × 2 contingency table (Supplementary Table 2), calculated the reporting odds ratio (ROR) and proportional reporting ratio (PRR), and identified a significantly positive drug signal when both the ROR and PRR were greater than the cut-off values (Supplementary Table 3) [29,30,31].

All statistical analyses were completed using SPSS 26.0 and Excel 2016. We sorted suspected drugs according to the World Health Organization anatomical therapeutic chemical (ATC) classification and reviewed prescribing information and published studies to determine whether HAEs had been verified and whether the drug was metabolized via the liver. The results were ranked according to the number of reports and the ROR.

Results

Descriptive analysis

From 2004 to 2020, 14,143 pediatric cases of suspected liver injury were submitted to FAERS, of which 49.6% were males, 45.1% were females, and 5.2% were of unknown sex (Table 1). There were 2,358 cases (16.7%) aged 0–2 years, 2,048 cases (45.1%) aged 3–5 years, and 9,737 cases (68.8%) aged 6–18 years. More than 90 countries contributed reports during 2004–2020, mainly in North America and Europe (Supplementary Table 4). The US (5,073 cases, 35.9%), Japan (1,029 cases, 7.3%), France (1,013 cases, 7.2%), the United Kingdom (UK) (964 cases, 6.8%), and Germany (531 cases, 3.8%) were ranked as the top 5 based on the number of reports. The annual number of HAEs in children in 2004–2020 ranged from 422 to 1,556. Peaks were reported in 2018 and 2019 (Fig. 1). The definite outcomes were high-risk, predominantly hospitalization, with 7,207 cases (37.2%) reported, followed by death, life threatening, disability, congenital anomaly, required intervention, and generating injury.

Table 1 Characteristics of hepatic adverse events in children in the period 2004–2020
Fig. 1
figure 1

Annual cases of hepatic adverse events in children from 2004 to 2020

Suspected drug signals associated with liver injury in children

We found 2,496 reports, with 586 positive signals suspected to cause liver injury. After unifying the generic names, excluding duplicates, and eliminating non-drug signals, 264 disproportional signals were obtained. The top 10 drugs based on the number of liver injury reports in children were paracetamol (1,365), methotrexate (878), vincristine (649), valproic acid (511), cyclophosphamide (490), tacrolimus (427), prednisone (416), prednisolone (401), etoposide (378), and cytarabine (344), as listed in Table 2. The top 10 drugs by ROR were teceleukin (ROR, 179.8; 95% confidence interval (CI), 22.5–1,437.4), cefpirome (ROR, 67.4; 95% CI, 7.0–647.8), trabectedin (ROR, 56.2; 95% CI, 10.9–289.5), ciprofloxacin (ROR, 44.9; 95% CI, 11.2–179.7), propoxyphene napsylate (ROR, 44.9; 95% CI, 8.2–245.3), vandetanib (ROR, 35.3; 95% CI, 13.7–91.1), calcium levofolinate (ROR, 33.7; 95% CI, 5.6–201.6), valsartan and amlodipine (ROR, 25.3; 95% CI, 9.8–65.5), aclarubicin (ROR, 22.5; 95% CI, 4.5–111.3), and sovaprevir (ROR, 22.5; 95% CI, 10.7–47.2), as shown in Table 3.

Table 2 Top 10 drugs by number of hepatic injury reports in children
Table 3 Top 10 drugs in terms of ROR in children

The 264 positive signals were then classified according to the ATC 1st level class, that is, the anatomical class, as shown in Table 4. Anti-infectives for systemic use were the most reported class, comprising 75 suspected drugs, followed by anti-neoplastic and immunomodulatory agents, with 69 signals. Each drug was checked to determine whether hepatic injury information had been provided on the package inserts and whether the drug was metabolized via the liver. The details of all positive signals are listed in Supplementary Table 5. After excluding drugs with hepatotoxic information provided in the package inserts or declared in published studies, six of 264 were identified, all of which were disproportionately associated with liver injury in children, namely acetylcysteine, thiopental, temazepam, nefopam, primaquine, and pyrimethamine (Table 5).

Table 4 Suspected drugs associated with liver injury in children by anatomical class
Table 5 Suspected drugs associated with liver injury in children but not labeled as such in medicine specifications

Discussion

In this study, suspected drug signals related to pediatric liver injury were identified by conducting a disproportional analysis of data from FAERS in the period 2004–2020 to provide information on safe medication for children. To the best of our knowledge, this study confirms the issues raised in the previous literature and provides new insights into pediatric liver injury associated with drugs. In 2010, Ferrajolo et al. explored drug-induced hepatic injury in children based on VigiBase from 2000 to 2006 in a case/non-case study [8]. Both paracetamol and methotrexate were among the top 10 drugs according to the number of reports in previous and current studies. Forty-two suspected drugs identified in the former (e.g., basiliximab, caspofungin, isoniazid) were also identified in our results. Basiliximab, a novel selective anti-human interleukin-2 receptor monoclonal antibody used for immunosuppressive therapy, was the only novel signal detected in children in a previous study, which suggested future monitoring, and this was confirmed herein. Liver injury associated with basiliximab has not been included in the FDA’s prescribing information, but the incidence of abnormal liver biochemical parameters associated with basiliximab, which is greater than 5%, has been indicated by Japanese package inserts [32, 33]. A randomized controlled trial of adults found an increase in serum alkaline phosphatase within the first month after the administration of basiliximab, but it was considered to be “functional cholestasis” [34, 35]. No elevated liver enzyme levels or hepatotoxicity were reported in clinical trials among children treated with basiliximab [36, 37].

Forty-three signals found in Ferrajolo’s study (chlorprothixene, methylphenidate, infliximab, etc.) were not included here. The reasons for this could be as follows. (1) Different data sources. VigiBase is the world’s largest database of individual case safety reports, having collected data on AEs from more than 130 countries since the late 1960s, whereas FAERS has collected data reported to the FDA only since 2004 [38]. (2) Distinct algorithms have been used for signal detection. Only ROR was adopted in the former, whereas ROR and PRR were combined here. (3) Some drugs were withdrawn from the market in their early days and their AE reporting ratios were diluted. Compared with the 2010 study, 222 suspicious drug signals were newly detected in the current study, including drugs marketed before 2006, such as ganciclovir and amlodipine, and after 2006, such as vandetanib and gemtuzumab.

Anti-infectives, especially antibacterial drugs, are extensively applied, and their irrational use can cause hepatotoxicity. The immature liver function in children and increased hepatotoxicity caused by the concomitant administration of multiple antibacterial drugs make children more vulnerable to hepatic injury than adults. The results of anti-infectives for systemic use, as the most reported class in our study, were consistent with the published literature [39, 40]. Representative drugs included trimethoprim-sulfamethoxazole (TMP-SMX), ceftriaxone, fluconazole, and isoniazid.

TMP-SMX exerts its antimicrobial effect by blocking the metabolism of folic acid through a dual pathway and can cause DILI via allergic reactions (fever, rash, etc.) [41]. Bell et al. [42] reported a case of DILI induced by TMP-SMX; specifically, a 9-year-old boy with a community-acquired methicillin-resistant Staphylococcus aureus skin and soft tissue infection received TMP-SMX, and 14 days after administration, he developed fever, vomiting, and abdominal pain, with poor appetite and mentality. He was diagnosed with TMP-SMX-induced liver injury based on biochemical tests and the exclusion of other factors contributing to hepatic injury.

Ceftriaxone is a semi-synthetic, third-generation cephalosporin that mainly causes cholelithiasis or bile stasis [43]. One retrospective study showed that 3.2% of patients treated with ceftriaxone might develop liver injury [44]. Liver injury caused by ketoconazole in children mostly presents as elevated levels of direct bilirubin and liver enzymes [45]. Liver functions usually recover after drug cessation, but severe cases can also develop into liver failure [46, 47]. Animal studies have confirmed the dose-dependent liver injury induced by ketoconazole [48].

Isoniazid is a first-line anti-tuberculosis drug. Approximately 10–20% of patients administered isoniazid develop the transient elevation of alanine aminotransferase, and fewer than 1–3% develop severe liver injury or even liver failure [49, 50]. A greater risk of isoniazid- and rifampicin-related liver injury in children (6.9% in children and 2.7% in adults) was reported in a retrospective study [51]. In the US, the incidence of isoniazid-associated hepatotoxicity was determined to be 1% in children treated for latent tuberculosis infection, and there was a synergistic harmful effect on the liver when isoniazid and rifampin were used in one prescription [52, 53]. It is generally believed that isoniazid-induced liver injury is attributed to the toxicity of the metabolites acetylhydrazine and hydrazine [54]. Moreover, mitochondrial damage caused by isoniazid-induced oxidative stress and lipid peroxidation stimulated by isoniazid and its metabolites has also been discussed [55].

In this study, six signals were found to be disproportionally associated with pediatric liver injury for the first time, namely acetylcysteine, thiopental, temazepam, nefopam, primaquine, and pyrimethamine. Acetylcysteine (also known as N-acetylcysteine, NAC) was detected as a disproportional signal associated with pediatric hepatic injury (ROR, 3.0; 95% CI 1.9–4.8). None of these HAEs had been reported in either package inserts or the published literature since NAC was marketed. NAC is the only drug approved by the FDA for the treatment of DILI caused by acetaminophen (APAP) overdose and might prevent hepatic injury by restoring glutathione levels [56]. A dosage of NAC for children greater than 5 kg is set based on clinical practice; however, the safety and effectiveness of NAC have not been verified in adequate and well-controlled studies. The ACG Clinical Guideline: Diagnosis and Management of Idiosyncratic Drug-Induced Liver Injury suggests the use of NAC for children with ALF caused by severe DILI, as a significantly lower 1-year spontaneous survival rate was associated with the intravenous infusion of NAC in children with non-APAP ALF in a placebo-controlled clinical trial [2, 57]. In our study, we found that NAC was a secondary suspected or concomitant drug in most reports and a primary suspicious drug in one report where APAP was secondary. Therefore, as a disproportional signal, NAC is used to treat or prevent HAEs, especially APAP-induced HAEs. However, whether NAC has a negative effect on children with non-APAP ALF is unclear.

We also found an association between temazepam use and pediatric liver injury (ROR, 2.4; 95% CI, 1.2–4.7). No hepatotoxic risk is indicated in the prescribing information, and there is a lack of evidence to ensure the effectiveness and safety of temazepam among children. Temazepam is a benzodiazepine, and it is uncommon to observe elevated hepatic enzymes with benzodiazepine used and to report hepatoxic cases in practice. However, it has been reported that benzodiazepines (alprazolam, chlordiazepoxide, clonazepam, diazepam, flurazepam, and triazolam) are associated with rare cholestatic liver injuries [58, 59]. We have not retrieved any literature directly reporting temazepam-induced liver injury and postulated that this is due to its lower frequency of medication and shorter duration [2]. In addition, there is a possibility that cross-sensitivity to temazepam might occur with other benzodiazepines [58].

In our study, nefopam was disproportionally associated with liver injury in children (ROR, 18.7; 95% CI, 5.7–61.3). It is unclear whether nefopam is effective and safe for children as a painkiller. In this study, we identified five nefopam-related pediatric hepatotoxic AEs. Four patients reported nefopam as a secondary suspected drug, and one reported nefopam as a concomitant drug. The primary suspected drugs in these reports were ketoprofen, esomeprazole, and pregabalin, all of which have adverse effects on the liver, as specified in the package inserts. Whether it is safe to use nefopam alone in children merits attention in clinical practice.

In this study, thiopental was detected as a disproportional signal (ROR, 3.5; 95% CI, 2.0–5.9). Thiopental is an intravenous anesthetic without hepatotoxic information provided in the inserts. In our findings, most reports of HAEs in children associated with thiopental recorded it as a concomitant drug, and the suspected drugs were mainly propofol, lamotrigine, phenytoin, propranolol, valproic acid, carbamazepine, lacosamide, and clonazepam, all of which have hepatotoxicity labeled in their package inserts. The risk of hepatotoxicity associated with this drug in children when applied alone is unclear. Bedir et al. indicated that oxidative stress and inflammation develop in the liver tissue of rats injected with thiopental alone, but further clinical studies are needed to explore this effect in humans [60].

Pyrimethamine was also detected as a disproportional signal (ROR, 3.5; 95% CI, 1.2–9.9). Four reports of pyrimethamine-associated hepatic injury in children have been identified. One patient recorded pyrimethamine as the primary suspected drug and sulfadiazine as the secondary drug, of which liver injury was a definite adverse effect. In the other three reports, the primary suspected drugs associated with liver injury were clindamycin, calcium folinate, and isotretinoin, all of which were reported to have a risk of hepatotoxicity. Pyrimethamine is typically combined with sulfonamides in practice. Many case reports of liver injury, such as granulomatous hepatitis, hypersensitivity to liver injury, and fatal hepatic necrosis, were determined to be induced by pyrimethamine-sulfadoxine [61,62,63]. Whether pyrimethamine alone can cause liver injury requires further investigation.

We found that primaquine is associated with pediatric HAEs (ROR, 14.0; 95% CI, 4.6–42.9). Among the five liver injury records associated with primaquine, one reported primaquine as the primary suspected drug, and four reported primaquine as a secondary suspected or concomitant drug, with chloroquine, tocilizumab, and malarone as the primary suspected drugs, all of which are toxic to the liver. The safety of primaquine when applied alone in children is not clear, as current studies have only presented an elevation of biochemical indicators of the liver when primaquine is used with chloroquine [64, 65].

This study has both limitations and strengths. Firstly, due to the differences in physiological characteristics, biochemical parameters, and disease spectrum between children and adults, and the lack of prospective data on cases in FAERS, such as viral antibody tests and re-exposure results, it is not appropriate to use tools that aid in the diagnosis of DILI to establish a robust causal relationship indicating that a drug ‘caused’ the liver injury in children. For example, the Roussel-Uclaf Causality Assessment Method is the most commonly used tool to assess DILI in adults, but it is not available as an independent assessment for children and is more appropriate for prospective data [66]. In this study, the association between suspected drugs and HAEs was derived from a statistical perspective. Secondly, HAEs in children may be under-reported due to the wide variation in the clinical presentation of HAEs, ranging from asymptomatic elevated liver enzymes to ALF, and the weak autonomic expression in children. For example, specific hepatotoxic drugs for children may be ignored if liver injury is not manifest. Despite its limitations, this study has the following non-negligible advantages. Firstly, this study uses a robustly validated and easily accessible real-world database, combined with advanced tools and commonly recommended pharmacovigilance methods, to statistically detect the association between drugs and HAEs in children, and to supplement clinical information with prescribing information and published literature. Secondly, unlike previous studies in children with small sample sizes, such as case reports and case series, this study reports the characteristics of HAEs and suspected drugs in a specific population of children based on a large volume of data (~ 14,000 cases) spanning a long time period (2004–2020), which can provide information for clinical drug safety in children and guide future independent studies in this important area of DILI for the pediatric population.

Conclusion

In our study, HAEs were analyzed in children from 2004 to 2020 based on FAERS, and suspected drug signals were discovered. In total, 264 drugs were identified as having a disproportional association with pediatric liver injury, six of which were detected for the first time. These findings call for attention in clinical practice to reduce potential risks, and preclinical and clinical studies are expected to verify the signals, determine the hepatotoxic mechanism, and promote the safe use of medication in pediatric patients.

Data Availability

The authors confirm the data and materials supporting the findings of this study are available within the article and its supplementary materials. FAERS reports are publicly available on the FDA website (https://fis.fda.gov/extensions/FPD-QDE-FAERS/FPD-QDE-FAERS.html).

Abbreviations

AE:

Adverse event

ALF:

Acute liver failure

APAP:

Acetaminophen

ATC:

Anatomical therapeutic chemical

DILI:

Drug-induced liver injury

FAERS:

Food and Drug Administration’s Adverse Event Reporting System

FDA:

Food and Drug Administration

HAEs:

Hepatic adverse events

MedDRA:

Medical Dictionary for Regulatory Activities

NAC:

N-acetylcysteine

PRR:

Proportional reporting ratio

ROR:

Reporting odds ratio

TMP-SMX:

Trimethoprim-sulfamethoxazole

References

  1. Andrade RJ, Chalasani N, Björnsson ES, Suzuki A, Kullak-Ublick GA, Watkins PB, et al. Drug-induced liver injury. Nat reviews Disease primers. 2019;5(1):58.

    PubMed  Google Scholar 

  2. Chalasani NP, Maddur H, Russo MW, Wong RJ, Reddy KR. ACG Clinical Guideline: diagnosis and management of Idiosyncratic Drug-Induced Liver Injury. Am J Gastroenterol. 2021;116(5):878–98.

    PubMed  CAS  Google Scholar 

  3. Yu Y, Mao Y, Chen C. Guidelines for diagnosis and treatment of drug-induced liver injury. J Practical Hepatol. 2017;20(02):257–74.

    Google Scholar 

  4. Shen T, Liu Y, Shang J, Xie Q, Li J, Yan M, et al. Incidence and etiology of Drug-Induced Liver Injury in Mainland China. Gastroenterology. 2019;156(8):2230–41e11.

    PubMed  Google Scholar 

  5. Suk KT, Kim DJ, Kim CH, Park SH, Yoon JH, Kim YS, et al. A prospective nationwide study of drug-induced liver injury in Korea. Am J Gastroenterol. 2012;107(9):1380–7.

    PubMed  Google Scholar 

  6. Reuben A, Koch DG, Lee WM. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology (Baltimore MD). 2010;52(6):2065–76.

    PubMed  Google Scholar 

  7. Mao Y. Strengthening the scientific research and supervision of drug-induced liver injury based on big data. J Clin Hepatol. 2018;34(6):1166–8.

    Google Scholar 

  8. Ferrajolo C, Capuano A, Verhamme KM, Schuemie M, Rossi F, Stricker BH, et al. Drug-induced hepatic injury in children: a case/non-case study of suspected adverse drug reactions in VigiBase. Br J Clin Pharmacol. 2010;70(5):721–8.

    PubMed  PubMed Central  Google Scholar 

  9. Fontana RJ, Seeff LB, Andrade RJ, Björnsson E, Day CP, Serrano J, et al. Standardization of nomenclature and causality assessment in drug-induced liver injury: summary of a clinical research workshop. Hepatology (Baltimore MD). 2010;52(2):730–42.

    PubMed  Google Scholar 

  10. Gan Y, Dong Y, zhang H, Xu Z, Chen D, Wang L et al. Clinical characteristics and outcomes of drug-induced liver injury in children: a study of 184 cases. J Clin Hepatol. 2015(08 vo 31):1244–7.

  11. Danan G, Teschke R. RUCAM in Drug and Herb Induced Liver Injury: the Update. Int J Mol Sci. 2015;17(1).

  12. Thakkar N, Salerno S, Hornik CP, Gonzalez D. Clinical Pharmacology Studies in critically Ill Children. Pharm Res. 2017;34(1):7–24.

    PubMed  CAS  Google Scholar 

  13. Squires RH Jr, Shneider BL, Bucuvalas J, Alonso E, Sokol RJ, Narkewicz MR, et al. Acute liver failure in children: the first 348 patients in the pediatric acute liver failure study group. J Pediatr. 2006;148(5):652–8.

    PubMed  PubMed Central  Google Scholar 

  14. Li F, Yu Q. International development and domestic situation of pediatric medication development and clinical trials. Chin J New Drugs. 2020;29(17):1933–8.

    Google Scholar 

  15. FDA, Questions, and Answers on FDA’s Adverse Event Reporting System (FAERS). 2018. https://www.fda.gov/drugs/surveillance/questions-and-answers-fdas-adverse-event-reporting-system-faers

  16. Cirmi S, El Abd A, Letinier L, Navarra M, Salvo F. Cardiovascular toxicity of tyrosine kinase inhibitors used in chronic myeloid leukemia: an analysis of the FDA adverse event reporting System Database (FAERS). Cancers. 2020;12(4).

  17. Wichelmann TA, Abdulmujeeb S, Ehrenpreis ED. Bevacizumab and gastrointestinal perforations: a review from the FDA adverse event reporting System (FAERS) database. Aliment Pharmacol Ther. 2021;54(10):1290–7.

    PubMed  CAS  Google Scholar 

  18. Zhang KW, Reimers MA, Calaway AC, Fradley MG, Ponsky L, Garcia JA, et al. Cardiovascular events in men with prostate Cancer receiving hormone therapy: an analysis of the FDA adverse event reporting System (FAERS). J Urol. 2021;206(3):613–22.

    PubMed  PubMed Central  Google Scholar 

  19. Böhm R, von Hehn L, Herdegen T, Klein HJ, Bruhn O, Petri H, et al. OpenVigil FDA - Inspection of U.S. american adverse drug events Pharmacovigilance Data and Novel Clinical Applications. PLoS ONE. 2016;11(6):e0157753.

    PubMed  PubMed Central  Google Scholar 

  20. Tian X, Zheng S, Wang J, Yu M, Lin Z, Qin M et al. Cardiac disorder-related adverse events for aryl hydrocarbon receptor agonists: a safety review. Exp Opin Drug Saf. 2022:1–6.

  21. Wu Q, Fan X, Hong H, Gu Y, Liu Z, Fang S, et al. Comprehensive assessment of side effects in COVID-19 drug pipeline from a network perspective. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 2020;145:111767.

    PubMed  CAS  Google Scholar 

  22. Böhm R, Höcker J, Cascorbi I, Herdegen T. OpenVigil–free eyeballs on AERS pharmacovigilance data. Nat Biotechnol. 2012;30(2):137–8.

    PubMed  Google Scholar 

  23. Suzuki A, Andrade RJ, Bjornsson E, Lucena MI, Lee WM, Yuen NA, et al. Drugs associated with hepatotoxicity and their reporting frequency of liver adverse events in VigiBase: unified list based on international collaborative work. Drug Saf. 2010;33(6):503–22.

    PubMed  CAS  Google Scholar 

  24. Raschi E, Poluzzi E, Koci A, Salvo F, Pariente A, Biselli M, et al. Liver injury with novel oral anticoagulants: assessing post-marketing reports in the US Food and Drug Administration adverse event reporting system. Br J Clin Pharmacol. 2015;80(2):285–93.

    PubMed  PubMed Central  Google Scholar 

  25. ICH. Introductory Guide for Standardised MedDRA Queries (SMQs) 21.1. 2018. https://admin.meddra.org/sites/default/files/guidance/file/smq_intguide_21_1_english.pdf

  26. Chuma M, Nakamoto A, Bando T, Niimura T, Kondo Y, Hamano H, et al. Association between statin use and daptomycin-related musculoskeletal adverse events: a mixed approach combining a meta-analysis and a disproportionality analysis. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America; 2022.

    Google Scholar 

  27. Hansen RA, Gartlehner G, Powell GE, Sandler RS. Serious adverse events with infliximab: analysis of spontaneously reported adverse events. Clin Gastroenterol hepatology: official Clin Pract J Am Gastroenterological Association. 2007;5(6):729–35.

    Google Scholar 

  28. Arai M, Shirakawa J, Konishi H, Sagawa N, Terauchi Y. Bullous Pemphigoid and Dipeptidyl Peptidase 4 inhibitors: a disproportionality analysis based on the japanese adverse drug Event Report Database. Diabetes Care. 2018;41(9):e130–e2.

    PubMed  CAS  Google Scholar 

  29. Bate A, Lindquist M, Orre R, Edwards IR, Meyboom RH. Data-mining analyses of pharmacovigilance signals in relation to relevant comparison drugs. Eur J Clin Pharmacol. 2002;58(7):483–90.

    PubMed  CAS  Google Scholar 

  30. van Puijenbroek E, Diemont W, van Grootheest K. Application of quantitative signal detection in the dutch spontaneous reporting system for adverse drug reactions. Drug Saf. 2003;26(5):293–301.

    PubMed  Google Scholar 

  31. Evans SJ, Waller PC, Davis S. Use of proportional reporting ratios (PRRs) for signal generation from spontaneous adverse drug reaction reports. Pharmacoepidemiol Drug Saf. 2001;10(6):483–6.

    PubMed  CAS  Google Scholar 

  32. FDA. Package Insert-Simulect® (basiliximab). 2003. https://www.accessdata.fda.gov/drugsatfda_docs/label/2003/basnov010203LB.htm

  33. PMDA. Simulect i.v. injection 10 mg for pediatric. 2021. http://zy.yaozh.com/data/pdf/00055862.pdf

  34. Zu C, Li B, Wei Y, Fan W, Huo Y, Deng Z, et al. Application of simulect in liver transplantation patients. Chin J Organ Transplantation. 2005;26(12):745–7.

    Google Scholar 

  35. Wu J, Wu M. Clinical liver transplantation. Shanghai, China: Second Military Medical University Press; 1998.

    Google Scholar 

  36. Shemshadi M, Hoseini R, Zareh R, Otukesh H. Use of Basiliximab with the Standard Immunosuppressive Protocol in Pediatric Renal transplantation: a double-blind Randomized Clinical Trial. Int J Organ Transplantation Med. 2020;11(1):8–14.

    CAS  Google Scholar 

  37. Mocarquer A, Pinto V, Buckel E, Lagos E, Pefaur J, Ramirez K et al. Basiliximab: efficacy and tolerability in adults and children. Transplantation Proceedings. 2003;35(7):2518-9.

  38. Center UM, VigiBase. WHO’s global database signalling harm and pointing to safer use 2022 [Available from: https://who-umc.org/vigibase/vigibase-who-s-global-database/

  39. Zhang H, Xu L, Xie Y, Geng T, Su F. 201 cases of drug-induced Liver Injury in Children. Chin J Pharmacovigil. 2020;17(10):715–9.

    Google Scholar 

  40. Xie X. A single-center cross-sectional study of drug-induced liver injury in children. Shanxi, China: Shanxi Medical University; 2020.

    Google Scholar 

  41. Frank DP, Molleston JP. Drug-Induced Liver Injury in Children. Curr Hepatol Rep. 2018;17(3):283–91.

    Google Scholar 

  42. Bell TL, Foster, Jennifer N, Townsend ML. Trimethoprim-Sulfamethoxazole–Induced Hepatotoxicity in a Pediatric patient. Pharmacotherapy. 2010.

  43. Devarbhavi H, Andrade RJ. Drug-induced liver injury due to antimicrobials, central nervous system agents, and nonsteroidal anti-inflammatory drugs. Semin Liver Dis. 2014;34(2):145–61.

    PubMed  CAS  Google Scholar 

  44. Nakaharai K, Sakamoto Y, Yaita K, Yoshimura Y, Igarashi S, Tachikawa N. Drug-induced liver injury associated with high-dose ceftriaxone: a retrospective cohort study adjusted for the propensity score. Eur J Clin Pharmacol. 2016;72(8):1003–11.

    PubMed  Google Scholar 

  45. Crerar-Gilbert A, Boots R, Fraenkel D, Macdonald GA. Survival following fulminant hepatic failure from fluconazole induced hepatitis. Anaesth Intensive Care. 1999;27(6):650–2.

    PubMed  CAS  Google Scholar 

  46. 46, Gayam V, Khalid M, Dahal S, Garlapati P, Gill A. Hyperacute liver injury following intravenous fluconazole: a rare case of dose-independent hepatotoxicity. J Family Med Prim Care. 2018;7(2):451–4.

    Google Scholar 

  47. Wen Y, Guo J, Lv L, Qi X. A case of acute liver failure caused by fluconazole. China Pharmacist. 2018;21(05):882–3.

    Google Scholar 

  48. Bronstein JA, Gros P, Hernandez E, Larroque P, Molinié C. Fatal acute hepatic necrosis due to dose-dependent fluconazole hepatotoxicity. Clin Infect diseases: official publication Infect Dis Soc Am. 1997;25(5):1266–7.

    CAS  Google Scholar 

  49. Pengcheng W, Komal P, Xiao-bo Z, Xiaochao M. Isoniazid metabolism and hepatotoxicity. Acta Pharmaceutica Sinica B. 2016(05 vo 6):384 – 92.

  50. CDC. Severe isoniazid-associated liver injuries among persons being treated for latent tuberculosis infection-United States, 2004–2008. MMWR Morbidity and mortality weekly report. 2010;59(8):224–9.

  51. Devarbhavi H, Karanth D, Prasanna KS, Adarsh CK, Patil M. Drug-Induced liver injury with hypersensitivity features has a better outcome: a single-center experience of 39 children and adolescents. Hepatology (Baltimore MD). 2011;54(4):1344–50.

    PubMed  CAS  Google Scholar 

  52. Hayashi PH, Fontana RJ. Clinical features, diagnosis, and natural history of drug-induced liver injury. Semin Liver Dis. 2014;34(2):134–44.

    PubMed  Google Scholar 

  53. Chang SH, Nahid P, Eitzman SR. Hepatotoxicity in children receiving isoniazid therapy for latent tuberculosis infection. J Pediatr Infect Dis Soc. 2014;3(3):221–7.

    Google Scholar 

  54. Metushi IG, Cai P, Zhu X, Nakagawa T, Uetrecht JP. A fresh look at the mechanism of isoniazid-induced hepatotoxicity. Clin Pharmacol Ther. 2011;89(6):911–4.

    PubMed  CAS  Google Scholar 

  55. Wang Y, Bao J. Research progress on the pathogenesis of isoniazid induced liver injury. J Pharm Pract. 2019;37(04):289–93.

    Google Scholar 

  56. Björnsson ES, Bergmann OM, Björnsson HK, Kvaran RB, Olafsson S. Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology. 2013;144(7):1419–25. 25.e1-3; quiz e19-20.

    PubMed  Google Scholar 

  57. Squires RH, Dhawan A, Alonso E, Narkewicz MR, Shneider BL, Rodriguez-Baez N, et al. Intravenous N-acetylcysteine in pediatric patients with nonacetaminophen acute liver failure: a placebo-controlled clinical trial. Hepatology (Baltimore MD). 2013;57(4):1542–9.

    PubMed  CAS  Google Scholar 

  58. LiverTox: Clinical and Research Information on Drug-. Induced Liver Injury [Internet]: Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases; 2012- [updated January 24, 2017. Available from: https://www.ncbi.nlm.nih.gov/books/NBK548556/

  59. Baumgardner JN, Shankar K, Hennings L, Albano E, Badger TM, Ronis MJ. N-acetylcysteine attenuates progression of liver pathology in a rat model of nonalcoholic steatohepatitis. J Nutr. 2008;138(10):1872–9.

    PubMed  CAS  Google Scholar 

  60. Bedir Z, Erdem KTO, Ates I, Karakurt TCO, Gursul C, Onk D, et al. Effects of ketamine, thiopental and their combination on the rat liver: a biochemical evaluation. Advances in clinical and experimental medicine: official organ. Wroclaw Med Univ. 2022;31(3):285–92.

    Google Scholar 

  61. van Everdingen-Bongers JJ, Janssen P, Lammens M, Stricker BH, Bravenboer B. Granulomatous hepatitis attributed to the combination pyrimethamine-chloroquine. Ned Tijdschr Geneeskd. 1996;140(6):320–2.

    PubMed  Google Scholar 

  62. Okazaki Y, Watanabe N, Uchiyama J, Nakano A, Nishizaki Y, Kagawa T, et al. A case of hypersensitivity type of liver injury induced by pyrimetamine and sulfadoxin (Fansidar). Nihon Shokakibyo Gakkai zasshi = The Japanese journal of gastro-enterology. 1997;94(2):129–32.

    PubMed  CAS  Google Scholar 

  63. Zitelli BJ, Alexander J, Taylor S, Miller KD, Howrie DL, Kuritsky JN, et al. Fatal hepatic necrosis due to pyrimethamine-sulfadoxine (Fansidar). Ann Intern Med. 1987;106(3):393–5.

    PubMed  CAS  Google Scholar 

  64. Hu K, Tang J, Feng C, Yang L, Wang X. Toxicity characteristics analysis on overdose of chloroquine phosphate/primaquine phosphate in children. Practical Pharm Clin Remedies. 2014;17(07):904–6.

    CAS  Google Scholar 

  65. Hu K, Tang J, He B, Wang X, Ma D. Analysis of adverse reactions of chloroquine combined with primaquine. Pharm J Chin People’s Liberation Army. 2011;27(06):564–80.

    Google Scholar 

  66. Danan G, Teschke R. RUCAM in Drug and Herb Induced Liver Injury: the Update. Int J Mol Sci 2015 Dec 24;17(1):14.

Download references

Acknowledgements

We would like to thank Editage (www.editage.cn) for English language editing.

Funding

This study was supported by the National Natural Science Foundation for Young Scholars of China (Grant No. 72004151) and the Science and Technology Plan Project of Sichuan Province (Grant No. 2020YFS0035).

Author information

Authors and Affiliations

Authors

Contributions

Yan Liu and Hailong Li conceptualized and designed the study, collected and analyzed the data, and drafted the manuscript. Liang Huang, Chaomin Wan, Guo Cheng, and Huiqing Wang interpreted the results and provided professional suggestions from clinical perspectives. Xuefeng Jiao and Linan Zeng collected the data and made critical revisions to the manuscript for important intellectual content. Zhijun Jia, Lei Zhang, Wei Zhang, and Lingli Zhang critically reviewed and revised the manuscript, and offered administrative support. All authors approved the final manuscript as submitted and agree to be accountable for all aspects of the work.

Corresponding authors

Correspondence to Wei Zhang or Lingli Zhang.

Ethics declarations

Competing interests

The authors declare no conflicts of interest.

Ethics approval and consent to participate

Ethics approval and consent to participate were not obtained for this study because the FAERS database collects spontaneous reports and is publicly available.

Consent for publication

Not applicable.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Li, H., Huang, L. et al. Liver injury in children: signal analysis of suspected drugs based on the food and drug administration adverse event reporting system. BMC Pediatr 23, 492 (2023). https://doi.org/10.1186/s12887-023-04097-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12887-023-04097-9

Keywords