Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Nimesulide-induced hepatotoxicity: A systematic review and meta-analysis

  • Jeongyoon Kwon,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Resources, Software, Writing – original draft, Writing – review & editing

    Affiliation College of Pharmacy & Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea

  • Seungyeon Kim,

    Roles Conceptualization, Data curation, Investigation, Methodology, Writing – review & editing

    Affiliation College of Pharmacy & Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea

  • Hyejin Yoo,

    Roles Conceptualization, Methodology, Supervision, Writing – review & editing

    Affiliation College of Pharmacy & Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea

  • Euni Lee

    Roles Conceptualization, Data curation, Funding acquisition, Supervision, Writing – review & editing

    eunilee@snu.ac.kr

    Affiliation College of Pharmacy & Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea

Abstract

Objective

This study aimed to evaluate the risk for hepatotoxicity with nimesulide, a non-steroidal anti-inflammatory drug (NSAID) available in Republic of Korea but withdrawn from the market in several countries.

Methods

A systematic review and meta-analysis were conducted of studies retrieved from PubMed, EMBASE, Cochrane, the Research Information Sharing Service and ClinicalTrials.gov up to September 2017. All studies reporting nimesulide-associated hepatotoxicity in patients as compared with the unexposed or the exposed to other NSAIDs were included. Studies using spontaneous reporting databases were included to estimate reporting odds ratio (ROR) of hepatotoxicity associated with nimesulide exposure. The association between nimesulide use and hepatotoxicity was estimated using relative risk (RR) and ROR with 95% confidence interval (CI).

Results

A total of 25 observational studies were eligible for review. In a meta-analysis of five observational studies, nimesulide was significantly associated with hepatotoxicity [RR 2.21, 95% CI 1.72–2.83]. From studies using spontaneous reporting databases (n = 6), rates of reported hepatotoxicity were significantly higher in patients using nimesulide, compared with those treated with other NSAIDs [pooled ROR 3.99, 95% CI 2.86–5.57]. Of a total of 33 patients from case studies and series, the majority (n = 28, 84.8%) were female, and the mean age (± standard deviation) was 56.8 (± 15.6) years. Almost half of the patients on nimesulide (45.5%) either required liver transplantation or died due to fulminant hepatic failure, of whom a third developed hepatotoxicity within less than 15 days of nimesulide administration.

Conclusions

Our study findings support previous reports of an increased risk for hepatotoxicity with nimesulide use and add to existing literature by providing risk estimates for nimesulide-associated hepatotoxicity. As the limited number of studies with primarily observational study designs were included in the analysis, more studies are needed to further describe the effects of dose and length of treatment on the risk for hepatotoxicity.

Introduction

Nimesulide is a non-steroidal anti-inflammatory drug (NSAID) with preferential inhibitory activity on cyclooxygenase 2 (COX-2) enzyme [1]. The drug was first launched in Italy in 1985 and was subsequently marketed in more than 50 countries, including South Korea [2]. It has potent analgesic, anti-inflammatory, and antipyretic properties, with a relatively low risk for gastrointestinal side effects, as demonstrated by numerous clinical trials [3, 4]. Moreover, nimesulide, when administered orally, is rapidly and extensively absorbed, thus allowing effective pain control [3, 5]. However, nimesulide induced hepatotoxicity was first reported in 1997 [6] and severe, and even fatal, cases of liver injury have been reported in patients who received nimesulide treatment [7]. Consequently, the use of nimesulide was restricted or withdrawn from the market in 2002 in Spain and Finland, followed by several other countries [8].

A number of observational studies that evaluated the safety profile of nimesulide were published [812] at around the time when nimesulide was initially banned in several countries. In 2004, the European Medicines Agency (EMA) recommended a restriction of nimesulide indications, as well as its maximal daily dose [13]. However, in May 2007, the Irish Medicines Board, the former regulatory agency of the Health Products Regulatory Authority, announced the marketing suspension of oral nimesulide-containing products due to a number of cases of fulminant hepatic failure requiring liver transplantation [14]. This prompted the EMA to undertake a further safety review of the drug, which, on completion in 2012, led the agency to support the continuous use of nimesulide, based on drug benefits outweighing the risks for liver toxicity [15]. However, this decision met with disagreement among some members of the Committee for Medicinal Products for Human Use within the EMA [15]. Subsequent widespread controversy surrounding the safety of nimesulide has led to varying regulatory decisions on restricting its use across different European countries.

To our knowledge, to date, there are no published studies using systematic evaluation methods to quantitatively assess the safety profile of nimesulide related to hepatotoxicity in peer-reviewed journals. The aim of this study was to evaluate hepatotoxic effects induced by nimesulide. We conducted a systematic review of the published literature, including case reports and series, on hepatotoxicity associated with the use of nimesulide in human patients and performed a meta-analysis of studies that assessed any hepatic adverse event outcomes.

Methods

Search strategy and data sources

A systematic review of the literature was performed in accordance with the Preferred Reporting of Systematic Reviews and Meta-Analyses (PRISMA) guideline (S1 Checklist) [16], using the following databases for studies published within the specified periods: PubMed (July 1998 to September 2017), Embase (August 1998 to September 2017), the Cochrane Central Register of Controlled Trials (November 1999 to September 2017), and the Research Information Sharing Service (Korean bibliographic database; April 1988 to September 2017). In addition, search in ClinicalTrials.gov database was also conducted to include unpublished trials. The following keywords were used to identify relevant articles on nimesulide-induced hepatotoxicity: ‘liver toxicity’, ‘hepatotoxicity’, ‘chemical and drug-induced liver injury’, ‘drug-induced hepatitis’, and ‘nimesulide’ (S1 Table). No restriction was imposed in terms of study design and publication language. Additionally, the reference lists of retrieved articles were also manually searched.

Study selection

Study designs for the selection included randomized controlled trials, observational studies, case reports, and case series. Hepatotoxicity outcomes were identified in patients exposed to nimesulide as compared with unexposed patients or those with exposure to other NSAIDs. Study participants were of either sex and any age. Of the identified articles, duplicates were removed using the bibliographic software EndNote X8.1 (Thomson Reuters, Philadelphia, PA, USA). One author (JK) identified potentially relevant articles for inclusion by titles and abstracts, while two authors (JK/SK) independently reviewed the entire manuscripts. Any disagreements between the authors were resolved by discussion or by a third author (EL). Studies were considered eligible for inclusion if they described hepatotoxicity associated specifically with the use of nimesulide. Exclusion criteria were: (1) non-human studies, (2) non-original research article type, (3) cases with pre-existing liver disease, and (4) same data also reported in another study.

Data extraction and quality assessment

We extracted information from each study, including study design, source of data, population characteristics, and study outcomes. Additional information from case series and case reports were gathered on the duration of nimesulide treatment prior to initial presentation of signs and symptoms of hepatotoxicity, concurrent medications, clinical features, evidence of hypersensitivity or autoimmune reaction, and laboratory results on admission. Where available, we recorded the odds ratios (ORs) and relative risks (RRs), as well as the proportion of patients who experienced liver injury that either was reported or could be calculated.

Since all included studies were non-randomized, we used the Newcastle-Ottawa Scale (NOS) to assess the quality of observational studies, except for case series or case reports [17]. The NOS uses a star system to assess the quality of a study based on three domains: selection, comparability, and outcome (cohort studies) or exposure (case-control studies), with the quality of the study rated as low (0–3 stars), medium (4–6 stars), or high (7–9 stars). Two authors (JK/SK) evaluated the quality of the studies, and any discrepancy was resolved by consensus reached including the third author (EL).

For quality assessment of case series and case reports, the Roussel Uclaf Causality Assessment Method (RUCAM) [18] was used to quantify the strength of the association between liver injury and use of nimesulide. Causality was classified as: highly probable, probable, possible, and unlikely. The RUCAM provides different subscales, depending on the pattern types of liver damage which are classified as hepatocellular, cholestatic, and mixed liver injury [18]. These three types of liver damage can be differentiated using the R value calculated as the alanine aminotransferase (ALT)/alkaline phosphatase (ALP) activity measured at the time liver injury is suspected, with both activities expressed as multiples of the upper limit of normal [18]. The hepatocellular pattern of liver damage was defined as R values ≥5, mixed pattern as R values >2 and <5, and cholestatic pattern as R values ≤2 [18]. If a study did not report the type of liver injury, we calculated the R value to determine the type of liver damage.

Statistical analysis

The primary analysis focused on assessing the risk for hepatotoxicity among patients treated with nimesulide. We used the ORs (adjusted, when available) or rate ratios reported in the case-control or cohort studies, respectively; we calculated the ORs or rate ratios, if not reported, using the proportion of patients reported with nimesulide-induced hepatotoxicity in each study. The association between nimesulide use and the risk for hepatotoxicity was estimated using relative risks (RRs) as well as 95% confidence intervals (CIs).

For studies using spontaneous reporting databases, we conducted a comprehensive disproportionality analysis by applying a case/non-case method. Cases included all studies reporting hepatotoxicity, whereas non-cases included all other reports recorded during the study period. Association between hepatotoxicity and use of nimesulide was estimated using reporting odds ratio (ROR) as a measure of disproportionality. The ROR is the ratio of the odds of nimesulide exposure among cases to the odds of nimesulide exposure among non-cases [19]. We pooled raw data of the proportion of reports for each NSAID, including nimesulide, from studies to compute the RORs, compared with other NSAIDs, and their corresponding 95% CIs.

The meta-analysis was conducted separately, depending on whether the measure of risk estimate was the RR or ROR. Sensitivity analyses were carried out to explain possible heterogeneity between studies by including studies in, or excluding them from, the meta-analysis, based on the study design and measure of the RR (ie., OR and rate ratio). Statistical heterogeneity across studies was assessed using the I2 statistic and Cochran’s Q test. An I2 value of ≥50% or a Cochran Q test P value of <0.10 indicated significant heterogeneity [20]. Overall estimate of the RR was obtained from a random effects model when statistic heterogeneity was present; otherwise, a fixed effects model was used. Data analyses were performed using SPSS version 23.0 (IBM SPSS Corp, Chicago, IL, USA) and Comprehensive Meta Analysis version 2.2 (Biostat, Englewood, NJ, USA).

Results

Search results

A total of 265 potentially eligible articles were identified by searching the three electronic databases using the keywords, as well as the relevant reference sections. Of these, 60 duplicate records were identified and removed. After screening the article abstracts and titles, 163 articles were excluded, and the remaining 42 articles underwent detailed full-text evaluation. Finally, 25 studies including 2 cohort studies, 1 case-control, 1 case-crossover study, 5 case/noncase studies, 3 case-population studies, 4 case series and 9 single-case reports were eligible for inclusion (Fig 1) and are summarized in Table 1. One [21] of the two listed cohort studies was analyzed as a case series study because only the number of cases and case descriptions were provided, without a clear measure of association. One [22] of the nine single-case reports performed an analysis generating the ROR using the World Health Organization Uppsala Monitoring Centre (WHO/UMC) pharmacovigilance database and was pooled with the studies using spontaneous reporting databases in our study. Of the included studies, 11 studies were included in quantitative analysis.

thumbnail
Fig 1. Flow chart showing study identification and selection.

https://doi.org/10.1371/journal.pone.0209264.g001

thumbnail
Table 1. Characteristics of included observational studies.

https://doi.org/10.1371/journal.pone.0209264.t001

Study characteristics and quality

The majority of observational studies included in our review were conducted in a number of European countries, including Italy, Spain, Switzerland, Ireland, France, Greece, the Netherlands, Portugal, and the UK, as well as in one Asian country, namely Taiwan (Table 1). Four of the studies identified patients from hospital medical records; five used administrative pharmacovigilance databases; two used both medical records and national health insurance databases, and one study used data from liver transplantation centers. In quality assessment, three studies were found to be of high quality, four studies of medium quality, and six of low quality (S2 and S3 Tables).

We identified 33 patients who were reported to have nimesulide-induced hepatic injury from the case reports and case series included in the study (Table 2). Cases were reported from 12 countries, including Israel, Belgium, France, Greece, Italy, Ireland, Iceland, Spain, Switzerland, Serbia, Singapore, and South Korea. The mean age (± standard deviation) of the patients was 56.8 ± 15.6 years (median 57 years; range 18–81 years). Age of ≥55 years was a risk factor found in 22 (66.7%) patients. The majority of patients with liver injury were female (n = 28, 84.8%), and the dose of nimesulide reported in the studies was either 100 mg or 200 mg daily, except for two patients who were given 150 mg or 600 mg daily, respectively, as well as an unreported dose for three patients. The duration of nimesulide treatment prior to initial presentation of signs and symptoms of hepatotoxicity ranged from 8 hours to 189 days (median 42 days).

thumbnail
Table 2. Characteristics of case series and case reports included in the analysis.

https://doi.org/10.1371/journal.pone.0209264.t002

Out of 33 identified patients, only two patients [29] had signs of hypersensitivity such as an increased eosinophilia or liver specific autoantibodies. The type of liver injury reported was hepatocellular in 14 cases (42.4%), cholestatic in six cases (18.2%), and mixed in four cases (12.1%), whereas the type was unknown in nine cases (27.3%) due to insufficient data. Eighteen (54.5%) of the patients recovered; of the remaining 15 (45.5%) patients who underwent liver transplantation, nine survived and six died. Moreover, 5 out of these 15 patients developed hepatotoxicity within less than 15 days of nimesulide administration. Using the RUCAM scoring system, nimesulide-attributable hepatotoxicity was probable in 18 cases, highly probable in 11 cases, and possible in four cases.

Meta-analysis

The findings of the meta-analysis are summarized in Figs 2 and 3. Five studies provided data suitable for analysis of hepatotoxicity outcomes. Use of nimesulide significantly increased the risk for hepatotoxicity (RR 2.21, 95% CI 1.72–2.83) (Fig 2). A fixed effects model was applied because heterogeneity across the studies was not statistically significant (I2 = 18.8%, P = 0.294). Sensitivity analysis showed no substantial change in pooled risk estimates upon exclusion of each of the included studies from the analysis (S4 Table). After excluding studies that reported rate ratios, the two remaining studies showed a statistically significant increased risk (fixed effects RR 2.43, 95% CI 1.82–3.26), with no evidence of heterogeneity (I2 = 0%, P = 0.474) (S4 Table). Excluding the two case-population studies had no effect on the overall risk for hepatotoxicity (fixed effects RR 2.23, 95% CI 1.76–3.00, I2 = 0%, P = 0.479) (S4 Table).

thumbnail
Fig 2. Forest plots of the risk for hepatotoxicity associated with nimesulide use.

RR, relative risk.

https://doi.org/10.1371/journal.pone.0209264.g002

thumbnail
Fig 3. Forest plots of reporting odds ratio for hepatotoxicity associated with nimesulide use relative to other NSAIDs.

ROR, reporting odds ratio.

https://doi.org/10.1371/journal.pone.0209264.g003

A total of six studies provided data suitable for the disproportionality analysis of hepatic adverse events in patients treated with nimesulide, based on the pharmacovigilance databases. Findings showed that use of nimesulide was associated with a significantly greater proportion of reported hepatic adverse events, compared to use of all other NSAIDs (random effects ROR 3.99, 95% CI 2.86–5.57), but the overall estimate was highly heterogeneous (I2 = 89.9%, P <0.001) (Fig 3). However, excluding the three analyses using the French [23, 24] and WHO/UMC [22] pharmacovigilance databases showed a statistically significant increased risk (fixed effects ROR 6.10, 95% CI 5.55–6.70), with no heterogeneity among the studies (I2 = 0.0%, P = 0.849) (S4 Table).

Discussion

Findings from this systematic review and meta-analysis indicated that nimesulide was associated with an increased risk for liver injury. While nimesulide-induced hepatotoxicity has been recognized in previously published studies [3, 41], we found no scientific reports quantifying the pooled risk, apart from an official report released by the EMA in 2012 that included two studies. We believe that our quantified measures as the pooled RR and ROR can be considered as one of the strengths of our systematic review that included all published studies until late 2017.

ROR measures for the evaluation of nimesulide-associated hepatotoxicity obtained from various databases and the patterns of NSAID use varied among countries, which could, at least in part, explain the differences in reporting rates. It has been previously shown that health care professionals’ reporting behavior for adverse drug reactions differed slightly across the European Union [42]. Media attention and publicity resulting in increased reporting, known as notoriety bias [43], could explain the differences in reported RORs in studies originating even from the same country [23, 24]. Withdrawal of nimesulide from Spain and Finland in 2002 [8] could also have contributed to the subsequent increased reporting rates in other European countries.

In our analysis of case reports and case series, the majority of cases of nimesulide-associated hepatotoxicity occurred in elderly and female patients. These findings are in agreement with a few published studies on drug-induced liver injury (DILI) related to NSAID use in particular [4446]. The increased incidence of DILI in the elderly carries biologic plausibility in terms of pharmacokinetic changes associated with aging. Conflicting reports, however, on gender-related occurrence of DILI were also published [23, 24]. A retrospective study from Spain showed an overall similar gender distribution in DILI cases [47], whereas a case-control study from France demonstrated a significant association of liver injury caused by NSAIDs in females [46]. Recent studies have shown a relationship between female sex and the hepatocellular pattern of DILI leading to poor outcomes [48]. Findings from our study showed a higher rate of hepatocellular DILI in females, compared to males, with the majority of patients with fatal outcomes directly related to DILI itself or as a result of liver transplantation for DILI being female. Although the same pattern of female preponderance for hepatocellular injury has been reported in various case series [21, 31], more epidemiologic assessments using well-validated study designs, as well as pathologic studies, are needed to explain this gender difference and its prevalence as well as the patterns and severity of nimesulide-associated hepatotoxicity.

Our study showed that almost half of patients required liver transplantation or died as a result of fulminant hepatic failure. Of importance, a third of these patients developed hepatotoxicity within less than 15 days of nimesulide administration, which is the maximum duration of nimesulide treatment as approved by the EMA. One study reported that the risk for liver injury increased with treatment duration, even when the treatment period is shorter than 15 days [9]. This results highlighted the needs for closer monitoring from the early phase of the nimesulide use process and healthcare professional should be aware of the nimesulide-induced hepatotoxicity. The majority of cases of DILI are idiosyncratic, occurring in most instances within 5–90 days after ingestion of the causative drug [49]. Similarly, in our study, nimesulide-induced hepatotoxicity generally occurred between 5 and 90 days after initiation of nimesulide treatment, suggesting an idiosyncratic mechanism is likely to be involved. Although the clinical signs of hypersensitivity were not observed in the majority of cases in our analysis, an increased eosinophil was presented in two patients. In addition, some studies suggested that their patients’ hepatotoxicity were related to metabolic idiosyncrasy [33]. Therefore, these findings indicated a potential mechanism of nimesulide-induced liver injury involving both immunologic and metabolic pathway. Further research is needed for elucidating biological plausibility of nimesulide-associated hepatotoxicity.

This systematic review has a few limitations. Firstly, only observational studies were included in the analysis, as no randomized controlled trials were available on the risk for hepatotoxicity with nimesulide use. While randomized controlled trials are superior in study design validity, they are usually underpowered when detecting rare events. Therefore, it is often inevitable to rely on observational study designs or secondary data analyses using heterogeneous data sources to evaluate safety outcomes at the expense of strong study validity. Secondly, our study had to apply less stringent inclusion criteria, as few published studies specifically investigated nimesulide-related safety outcomes as their primary research aim. In order to capture all potential adverse effects, our inclusion criteria were not limited to nimesulide-related liver injury as the primary research outcome. Despite our efforts to include as many studies as possible for evaluation, the limited number of studies available precluded any subgroup analysis to examine the effects of age, gender, dose, and length of treatment on the risk for liver injury. Despite the study design limitations, as well as the use of data sources such as spontaneous reporting databases, findings from our systematic review can be useful for the detection of rare adverse events, which has been recognized as a primary tool for pharmacovigilance reflecting the reality of clinical practice [19, 50]. In addition, research findings on drug safety such as ours should spur on further experimental studies aimed at investigating the underlying mechanism and degree of severity of nimesulide-induced hepatotoxicity.

This systematic review has important implications for clinical practice. Currently, nimesulide is still available on the market in many countries (e.g., Bulgaria, Czech Republic, Greece, Hungary, Italy, Poland, Portugal, Romania, Slovakia and South Korea) despite its market withdrawal in several countries. Clinicians should consider prescribing nimesulide only as a second-line medication for the treatment of acute pain or dysmenorrhea and should monitor those patients with an underlying risk for liver injury from very early phase, even with short-term use of nimesulide. Furthermore, an appropriate decision support system or vigilance teamwork including pharmacists would enable clinicians to better monitor nimesulide use and its associated adverse effects, especially in patients who concurrently use other potentially hepatotoxic drugs.

Conclusions

Our study indicates that nimesulide use is associated with an approximately twofold increased risk for hepatotoxicity. The association between nimesulide use and related hepatotoxicity is supported by our comprehensive disproportionality analysis, showing an increased rate of reported hepatic adverse events with nimesulide, compared with other NSAIDs. Further studies of nimesulide-induced hepatotoxicity are needed to evaluate the risk, as well as to better quantify the absolute risk, for hepatotoxicity associated with nimesulide by age, gender, and treatment dose and duration.

Supporting information

S2 Table. Newcastle-Ottawa Scale (NOS) for assessing the quality of cohort studies.

https://doi.org/10.1371/journal.pone.0209264.s003

(DOCX)

S3 Table. Newcastle-Ottawa Scale (NOS) for assessing the quality of case-control, case-population and case/non-case studies.

https://doi.org/10.1371/journal.pone.0209264.s004

(DOCX)

S4 Table. Sensitivity analysis for studies included in the analysis.

https://doi.org/10.1371/journal.pone.0209264.s005

(DOCX)

References

  1. 1. Hawkey CJ. COX-2 inhibitors. The Lancet. 1999;353(9149):307–14. https://doi.org/10.1016/S0140-6736(98)12154-2.
  2. 2. Chatterjee S, Pal J, Biswas N. Nimesulide-induced hepatitis and toxic epidermal necrolysis. Journal of postgraduate medicine. 2008;54(2):150–1. Epub 2008/05/16. pmid:18480536.
  3. 3. Kress HG, Baltov A, Basinski A, Berghea F, Castellsague J, Codreanu C, et al. Acute pain: a multifaceted challenge—the role of nimesulide. Current medical research and opinion. 2016;32(1):23–36. Epub 2015/09/29. pmid:26414386.
  4. 4. Wober W. Comparative efficacy and safety of nimesulide and diclofenac in patients with acute shoulder, and a meta-analysis of controlled studies with nimesulide. Rheumatology (Oxford, England). 1999;38(suppl_1):33–8.
  5. 5. Bennett A, Tavares IA. COX-2 inhibitors compared and contrasted. Expert Opinion on Pharmacotherapy. 2001;2(11):1859–76. pmid:11825322
  6. 6. Bessone F, Fay F, Vorobioff J, Passamonti ME, Godoy A, Tanno H. Nimesulide hepatotoxicity: Evidence from six cases. Hepatology. 1997;26:483A.
  7. 7. Zhou L, Pang X, Jiang J, Zhong D, Chen X. Nimesulide and 4'-Hydroxynimesulide as Bile Acid Transporters Inhibitors Are Contributory Factors for Drug-Induced Cholestasis. Drug metabolism and disposition: the biological fate of chemicals. 2017;45(5):441–8. Epub 2017/02/17. pmid:28202577.
  8. 8. Traversa G, Bianchi C, Da Cas R, Abraha I, Menniti-Ippolito F, Venegoni M. Cohort study of hepatotoxicity associated with nimesulide and other non-steroidal anti-inflammatory drugs. British Medical Journal. 2003;327(7405):18–22. pmid:12842950
  9. 9. Donati M, Conforti A, Lenti MC, Capuano A, Bortolami O, Motola D, et al. Risk of acute and serious liver injury associated to nimesulide and other NSAIDs: data from drug-induced liver injury case-control study in Italy. British journal of clinical pharmacology. 2016;82(1):238–48. Epub 2016/03/19. pmid:26991794; PubMed Central PMCID: PMCPMC4917796.
  10. 10. Gulmez SE, Larrey D, Pageaux GP, Lignot S, Lassalle R, Jove J, et al. Transplantation for acute liver failure in patients exposed to NSAIDs or paracetamol (acetaminophen): the multinational case-population SALT study. Drug safety. 2013;36(2):135–44. Epub 2013/01/18. pmid:23325533; PubMed Central PMCID: PMCPMC3568201.
  11. 11. Lee CH, Wang JD, Chen PC. Increased risk of hospitalization for acute hepatitis in patients with previous exposure to NSAIDs. Pharmacoepidemiology and Drug Safety. 2010;19(7):708–14. pmid:20582911
  12. 12. Sabate M, Ibanez L, Perez E, Vidal X, Buti M, Xiol X, et al. Risk of acute liver injury associated with the use of drugs: a multicentre population survey. Alimentary pharmacology & therapeutics. 2007;25(12):1401–9. Epub 2007/06/02. pmid:17539979.
  13. 13. EMEA. Committee for Proprietary Medicinal Products (CPMP) opinion following an article 31 referral: European Agency for the Evaluation of Medicinal Products; [updated 2004 May 7]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Referrals_document/Nimesulide_31/WC500013818.pdf.
  14. 14. IMB. IMB Suspension of Marketing of Nimesulide containing medicines: Irish Medicines Board; [updated 2007 July 1 ]. Available from: http://www.hpra.ie/docs/default-source/Safety-Notices/human-medicines—3rd-party-publications—hpra-suspension-of-marketing-of-nimesulide-containing-medicines-document.pdf?sfvrsn=0.
  15. 15. EMA. Assessment report for Nimesulide containing medicinal products for systemic use: European Medicines Agency; [updated 2012 Apr 19]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Referrals_document/Nimesulide_31/WC500125574.pdf.
  16. 16. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. Journal of Clinical Epidemiology. 2009;62(10):1006–12. pmid:19631508
  17. 17. Wells G, Shea B, O'Connell D, Peterson J, Welch V, Losos M, et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomized studies in meta-analyses 2014. Available from: http://www.ohri.ca/programs/clinical_epidemiology/oxford.htm.
  18. 18. Danan G, Teschke R. RUCAM in Drug and Herb Induced Liver Injury: The Update. International Journal of Molecular Sciences. 2016;17(1):14. PMC4730261. pmid:26712744
  19. 19. Egberts AC, Meyboom RH, van Puijenbroek EP. Use of measures of disproportionality in pharmacovigilance. Drug safety. 2002;25(6):453–8. pmid:12071783
  20. 20. Cochrane Handbook for Systematic Reviews of Interventions The Cochrane Collaboration; 2011. Available from: www.handbook.cochrane.org.
  21. 21. Walker SL, Kennedy F, Niamh N, McCormick PA. Nimesulide associated fulminant hepatic failure. Pharmacoepidemiology and Drug Safety. 2008;17(11):1108–12. pmid:18821716
  22. 22. Merlani G, Fox M, Oehen HP, Cathomas G, Renner EL, Fattinger K, et al. Fatal hepatotoxicity secondary to nimesulide. European Journal of Clinical Pharmacology. 2001;57(4):321–6. pmid:11549211
  23. 23. Lapeyre‐Mestre M, Grolleau S, Montastruc JL. Adverse drug reactions associated with the use of NSAIDs: a case/noncase analysis of spontaneous reports from the French pharmacovigilance database 2002–2006. Fundamental & clinical pharmacology. 2013;27(2):223–30.
  24. 24. Lapeyre-Mestre M, Rueda De Castro AM, Bareille MP, Garcia Del Pozo J, Alvarez Requejo A, Arias LM, et al. Non-steroidal anti-inflammatory drug-related hepatic damage in France and Spain: Analysis from national spontaneous reporting systems. Fundamental and Clinical Pharmacology. 2006;20(4):391–5. pmid:16867024
  25. 25. Licata A, Calvaruso V, Cappello M, Craxì A, Almasio PL. Clinical course and outcomes of drug-induced liver injury: Nimesulide as the first implicated medication. Digestive and Liver Disease. 2010;42(2):143–8. pmid:19625223
  26. 26. Motola D, Vargiu A, Leone R, Cocci A, Salvo F, Ros B, et al. Hepatic adverse drug reactions: a case/non-case study in Italy. European journal of clinical pharmacology. 2007;63(1):73–9. pmid:17119945
  27. 27. Sanchez-Matienzo D, Arana A, Castellsague J, Perez-Gutthann S. Hepatic disorders in patients treated with COX-2 selective inhibitors or nonselective NSAIDs: A case/noncase analysis of spontaneous reports. Clinical Therapeutics. 2006;28(8):1123–32. pmid:16982289
  28. 28. 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™. Drug safety. 2010;33(6):503–22. pmid:20486732
  29. 29. Van Steenbergen W, Peeters P, De Bondt J, Staessen D, Büscher H, Laporta T, et al. Nimesulide-induced acute hepatitis: Evidence from six cases. Journal of Hepatology. 1998;29(1):135–41. pmid:9696502
  30. 30. Cholongitas E, Koulenti D, Petraki K, Papatheodoridis GV. Nimesulide-induced acute hepatitis. Annals of Gastroenterology. 2003;16(4):359–62.
  31. 31. Dastis SN, Rahier J, Lerut J, Geubel AP. Liver transplantation for nonsteroidal anti-inflammatory drug-induced liver failure: nimesulide as the first implicated compound. European journal of gastroenterology & hepatology. 2007;19(11):919–22. Epub 2007/12/01. pmid:18049158.
  32. 32. Gallelli L, Ferraro M, Mauro GF, De Fazio S, De Sarro G. Nimesulide-induced hepatotoxicity in a previously healthy woman. Clinical Drug Investigation. 2005;25(6):421–4. pmid:17532683
  33. 33. Chae Hee Bok, Choi Won Jun, Lee Mun Woo, Park Seon Mee, Kim Hye young, Cho Myeong Chan, et al. A case of nimesulide induced hepatitis. Korean Journal of Internal Medicine. 2000;59(1):114–9.
  34. 34. Lukić S, Krstić M, Damjanov N, Boricić I, Popović D, Djuranović S, et al. Cholestatic hepatitis associated with nimesulide—a case report. Srpski arhiv za celokupno lekarstvo. 2009;137(9–10):550–3. pmid:19950766
  35. 35. Page M, Christin F, Hayi-Slayman D, Baillon JJ, Ber CE, Delafosse B, et al. Acute liver failure due to a treatment by nimesulide: Another case and review. Annales Francaises d'Anesthesie et de Reanimation. 2008;27(9):742–6. pmid:18760563
  36. 36. Rodrigo L, De Francisco R, Pérez-Pariente JM, Cadahía V, Tojo R, Rodriguez M, et al. Nimesulide-induced severe hemolytic anemia and acute liver failure leading to liver transplantation. Scandinavian Journal of Gastroenterology. 2002;37(11):1341–3. pmid:12465736
  37. 37. Sbeit W, Krivoy N, Shiller M, Farah R, Cohen HI, Struminger L, et al. Nimesulide-induced acute hepatitis. Annals of Pharmacotherapy. 2001;35(9):1049–52. pmid:11573855
  38. 38. Schattner A, Sokolovskaya N, Cohen J. Fatal hepatitis and renal failure during treatment with nimesulide. Journal of Internal Medicine. 2000;247(1):153–5. pmid:10672143
  39. 39. Tan HH, Ong WMC, Lai SH, Chow WC. Nimesulide-induced hepatotoxicity and fatal hepatic failure. Singapore Medical Journal. 2007;48(6):582–5. pmid:17538762
  40. 40. Weiss P, Mouallem M, Bruck R, Hassin D, Tanay A, Brickman CM, et al. Nimesulide-induced hepatitis and acute liver failure. The Israel Medical Association journal: IMAJ. 1999;1(2):89–91. pmid:10731303
  41. 41. Venegoni M, Da Cas R, Menniti-Ippolito F, Traversa G. Effects of the European restrictive actions concerning nimesulide prescription: A simulation study on hepatopathies and gastrointestinal bleedings in Italy. Annali dell'Istituto Superiore di Sanita. 2010;46(2):153–7. pmid:20567066
  42. 42. Varallo FR, Guimarães SdOP, Abjaude SAR, Mastroianni PdC. Causes for the underreporting of adverse drug events by health professionals: a systematic review. Revista da Escola de Enfermagem da USP. 2014;48(4):739–47.
  43. 43. van Hunsel F, van Puijenbroek E, de Jong-van den Berg L, van Grootheest K. Media attention and the influence on the reporting odds ratio in disproportionality analysis: an example of patient reporting of statins. Pharmacoepidemiol Drug Saf. 2010;19(1):26–32. Epub 2009/12/03. pmid:19953500.
  44. 44. Mitchell SJ, Hilmer SN. Drug-induced liver injury in older adults. Therapeutic Advances in Drug Safety. 2010;1(2):65–77. PMC4110803. pmid:25083196
  45. 45. Abboud G, Kaplowitz N. Drug-induced liver injury. Drug safety. 2007;30(4):277–94. Epub 2007/04/06. pmid:17408305.
  46. 46. Lacroix I, Lapeyre-Mestre M, Bagheri H, Pathak A, Montastruc JL. Nonsteroidal anti-inflammatory drug-induced liver injury: a case-control study in primary care. Fundam Clin Pharmacol. 2004;18(2):201–6. Epub 2004/04/07. pmid:15066135.
  47. 47. Lucena MI, Andrade RJ, Kaplowitz N, Garcia-Cortes M, Fernandez MC, Romero-Gomez M, et al. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and sex. Hepatology (Baltimore, Md). 2009;49(6):2001–9. Epub 2009/05/29. pmid:19475693.
  48. 48. Andrade RJ, Lucena MI, Fernández MC, Pelaez G, Pachkoria K, García-Ruiz E, et al. Drug-Induced Liver Injury: An Analysis of 461 Incidences Submitted to the Spanish Registry Over a 10-Year Period. Gastroenterology. 2005;129(2):512–21. https://doi.org/10.1053/j.gastro.2005.05.006. pmid:16083708
  49. 49. Hussaini SH, Farrington EA. Idiosyncratic drug-induced liver injury: an overview. Expert opinion on drug safety. 2007;6(6):673–84. Epub 2007/10/31. pmid:17967156.
  50. 50. Montastruc JL, Sommet A, Bagheri H, Lapeyre-Mestre M. Benefits and strengths of the disproportionality analysis for identification of adverse drug reactions in a pharmacovigilance database. British journal of clinical pharmacology. 2011;72(6):905–8. Epub 2011/06/11. pmid:21658092; PubMed Central PMCID: PMCPMC3244636.