Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Aug 17.
Published in final edited form as: Gastroenterology. 2010 Apr 12;138(7):2246–2259. doi: 10.1053/j.gastro.2010.04.001

Risk Factors for Idiosyncratic Drug-Induced Liver Injury

NAGA CHALASANI *,, EINAR BJÖRNSSON §
PMCID: PMC3157241  NIHMSID: NIHMS313390  PMID: 20394749

Abstract

Idiosyncratic drug-induced liver injury (DILI) is a rare disorder that is not related directly to dosage and little is known about individuals who are at increased risk. There are no suitable preclinical models for the study of idiosyncratic DILI and its pathogenesis is poorly understood. It is likely to arise from complex interactions among genetic, nongenetic host susceptibility, and environmental factors. Nongenetic risk factors include age, sex, and other diseases (eg, chronic liver disease or human immunodeficiency virus infection). Compound-specific risk factors include daily dose, metabolism characteristics, and propensity for drug interactions. Alcohol consumption has been proposed as a risk factor for DILI from medications, but there is insufficient evidence to support this. Many studies have explored genetic defects that might be involved in pathogenesis and focused on genes involved in drug metabolism and the immune response. Multicenter databases of patients with DILI (the United States Drug Induced Liver Injury Network, DILIGEN, and the Spanish DILI registry) are important tools for clinical and genetic research. A genome-wide association study of flucloxacillin hepatotoxicity has yielded groundbreaking results and many similar studies are underway. Nonetheless, DILI is challenging to investigate because of its rarity, the lack of experimental models, the number of medications that might cause it, and challenges to diagnosis.

Keywords: HLA, Amoxicillin-Clavulanate, Hy’s Law, DILI


The liver metabolizes xenobiotics, so it is not surprising that drug-induced liver injury (DILI) is a potential complication of many drugs. DILI broadly is classified into intrinsic and idiosyncratic types; intrinsic DILI generally is dose-dependent and predictable (eg, acetaminophen toxicity), whereas idiosyncratic DILI is unpredictable and does not depend directly on dose. This review focuses on the idiosyncratic type of DILI, which accounts for the majority of hepatotoxicity associated with medication use. Idiosyncratic DILI is rare even among individuals who are exposed to drugs that are known to be hepatotoxic. It occurs in 1 in 5000 to 1 in 100,000 individuals who take medication; the risk is lower for some drugs.1,2 The epidemiology of DILI is not well understood; most studies that assessed the risk of liver injury from different medications have been retrospective. 3,4 There are no surveillance mechanisms in place to monitor DILI, so adverse drug reactions, including DILI, are under-reported. Controlled clinical trials provide reliable information about abnormal liver test results that are associated with specific medications, but these generally do not detect rare adverse drug reactions, so most cases of idiosyncratic hepatotoxicity are not detected.1,2

To our knowledge, there is only one population-based prospective study that systematically assessed the incidence of DILI.5 The incidence of DILI in a French population was 13.9 cases per 100,000 inhabitants, a frequency that is 16-fold higher than that estimated from spontaneous reporting methods.4 Based on this incidence rate, it was estimated that more than 8000 cases of DILI might occur in France each year and lead to approximately 500 deaths.5 The United States Acute Liver Failure Study Group reported that acetaminophen and idiosyncratic drug reactions combined account for approximately 50% of cases of acute liver failure in the United States.6 Vuppalanchi et al7 reported that drug hepatotoxicity accounted for 4% of all cases of new-onset jaundice, but most cases of drug hepatotoxicity (24 patients) were attributable to acetaminophen toxicity and idiosyncratic DILI occurred in only 5 patients (0.7% of total study population). By using several different International Classification of Diseases 9th revision codes and the names of specific medications (amoxicillin/clavulanate, phenytoin, valproate, and isoniazid), Jinjuvadia et al8 identified an overall DILI frequency of 1.6% (119 DILI cases of 7395 total patients) using the most sensitive combination of an acute liver injury International Classification of Diseases 9th revision code plus a medical record search of the University of Michigan Health System database. Importantly, 36 of these DILI cases (0.5%) were attributed to acetaminophen overdose, whereas the remaining 83 were caused by other agents (1.1%).8

Studies of unselected patients with DILI revealed that their prognosis generally is favorable.911 Hyman Zimmerman, 12 a drug hepatotoxicity researcher, observed that mortality of patients with hepatocellular injury accompanied by jaundice was 10%–50%, depending on the drug involved. This observation, called Hy’s rule, is used by the United States Food and Drug Administration to assess hepatotoxicity in drug development.13 Recent studies from Spain, Sweden, and the United States confirmed Zimmerman’s observation—mortality among patients with hepatocellular jaundice is approximately 9%–12%.911

It is virtually impossible to predict an individual’s susceptibility to DILI from a specific compound (with the exception of flucloxacillin). Obviously, if an individual already has experienced DILI from a particular drug there is significant risk for recurrence.14 Genetic susceptibility is one of the most important risk factors for DILI, but the genetic basis of causes of liver injury are poorly understood for most drugs with documented hepatotoxicity.1518 Chemical properties of the drug, daily dose, drug metabolism, and other factors such as age, sex, nutritional factors, and underlying disease states might mediate the development of DILI. The pathogenic mechanisms of idiosyncratic DILI have been reviewed extensively1921 and are not covered in this review. We summarize our current understanding of nongenetic and genetic risk factors for idiosyncratic DILI in human beings (Table 1).

Table 1.

Factors That Cause Predisposition to Idiosyncratic DILI

Nongenetic factors Genetic variability
Age Phase 1 enzymes
    CYP 2C8
Sex     CYP 2C9
    CYP 2C19
Daily dose     CYP 2D6
    CYP 2E1
Metabolism profile Phase 2 and detoxifying enzymes
    NAT2
Drug interactions     GSTM1 and T1
    MnSOD
Alcohol     UGT2B7
Underlying comorbidities (pre-existing liver disease, HIV infection, diabetes) Drug transporters
    BSEP (ABCB11)
    MRP2 (ABCC2)
    MDR3 (ABCB4)
Immunologic
    HLA class antigen
    Cytokines (IL-10, IL-4, tumor necrosis factor-α)
Mitochondrial DNA mutations (POLG)

GST, glutathione S-transferase.

Nongenetic Risk Factors

Age

Age is a risk factor for DILI, but only from specific medications.1,2 Younger age is a risk factor for certain medications such as valproic acid and for Reye syndrome, associated with aspirin use.1,2 As age increases, so does the risk of liver injury from compounds such as erythromycin, halothane, isoniazid, nitrofurantoin, and flucloxacillin. 1,2,12 The risk of hepatotoxicity from isoniazid increases significantly with age.1,8 In a large study of patients in a US tuberculosis clinic, the age-specific incidence of isoniazid hepatotoxicity was 4.4 per 1000 patients age 25–34 years, whereas it increased to 20.83 per 1000 patients age 50 years and older.22 Increasing age also increases the risk for hepatotoxicity from amoxicillin/clavulanate.23 The cholestatic type of DILI is more common among the elderly, whereas hepatocellular DILI appears to be more common in younger individuals.10,24 The reasons that age affects DILI phenotypes are unclear. Although older age can affect the clearance of certain CYP3A substrates,25 older age does not significantly alter the activity or expression of phase I or phase II drug-metabolizing enzymes.26 Renal function is impaired in the elderly, which might increase drug concentrations in the liver; liver volume and liver blood flow have been correlated inversely with age.27 However, in the elderly these physiologic alterations would account for intrinsic DILI rather than idiosyncratic DILI. It is unclear if the elderly produce more reactive metabolites or have increased immune response to these metabolites. The increased risk of hepatotoxicity from some drugs might result from polypharmacy among the elderly.1,2 Although the incidence of certain adverse effects can increase with the use of multiple medications, there is little evidence to support polypharmacy as a predisposing factor for DILI. Combinations of 2 or more hepatotoxic drugs increased the risk for DILI by a factor of 6 in one study.4 However, a subsequent prospective study did not show a significant relationship between polymorbidity or polypharmacy and the risk for DILI.28

Sex

Women are believed to be at higher risk for idiosyncratic DILI than men, based on a higher prevalence of women in published DILI studies.1 However, recent studies have not shown that women are systematically at greater risk; in reviewing the published literature, Shapiro and Lewis29 did not find female sex to be a risk factor for DILI. In a landmark prospective study, Lucena24 reported that of 603 patients with DILI, 51% were male and 49% were female. Similarly, a population-based prospective study reported an annual incidence of 10.4 ± 3.0 per 100,000 women and 17.1 ± 3.6 per 100,000 men.5 In this study, the standardized female to male incidence ratio increased from 0.86 (0.26 –2.90) in women younger than 50 years of age to 2.62 (1.0 – 6.92) in women older than age 50. However, this observation was not reproduced by Lucena.24

Men and women might have differences in susceptibility to DILI caused by different medications. For example, women are more susceptible to liver injury associated with halothane, flucloxacillin, isoniazid, nitrofurantoin, chlorpromazine, or erythromycin,1,2,4,12 whereas men have an increased risk of azathioprine-induced liver injury.1,2 Zimmerman12 observed that autoimmune-type DILI occurred almost exclusively in women; this observation has been confirmed.30

Recent studies have shown a relationship between female sex, the hepatocellular pattern of DILI, and poor outcome (eg, acute liver failure, liver transplantation, and death).911 A prospective trial of the DILI network reported a significantly greater number of women with hepatocellular DILI than men (65% vs 35%; P < .05).11 A similar trend was observed by Lucena et al24 when the analysis of their cohort was restricted to patients younger than the age of 60 years. De Valle et al31 found hepatocellular injury to be more common among women, which could not be attributed to differences in the consumption of drugs that might have caused the reaction. It is not clear why hepatocellular DILI occurs more frequently in women. Lucena et al24 reported a relationship between female sex and DILI severity; nearly 90% of patients with fulminant liver failure from DILI were women. Similarly, more women patients with severe DILI underwent liver transplantation in the United States.32

Daily Dose

There is a traditional view that idiosyncratic DILI cannot be predicted based on dose,12 although a relationship to dose was observed for some medications, such as diclofenac, amoxicillin/clavulanate, and flucloxacillin.4 Idiosyncratic liver injury associated with bosentan also has been shown to have dose dependency.33 There are case reports in which dose reduction led to improvement and disappearance of hepatotoxicity caused by mianserin, 34 and an increased dose of duloxetine was associated with liver injury in a patient who had no signs of hepatotoxicity at a lower dose.35 The concept of idiosyncratic drug reactions of being entirely dose-independent might be incorrect. Uetrecht19 observed that idiosyncratic drug reactions were rare among patients given drug doses of less than 10 mg per day; conversely patients given drug doses of 1 gram or more per day were more likely to have idiosyncratic drug reactions.20 Therefore, some relationship exists between daily dose of a drug and the chances of liver injury. This hypothesis recently was tested in a systematic fashion.35 Data from 2 pharmaceutical databases were used to examine the relationship between daily dose of commonly prescribed oral medicines in the United States and their reported frequency of hepatotoxicity.35 Furthermore, previously reported cases of DILI and concomitant jaundice9 were analyzed for a relationship between daily dose and idiosyncratic DILI.35 Among US prescription medicines, daily doses of oral medications were associated significantly with liver failure, liver transplantation, and death from DILI (Table 2).35 In a study of approximately 600 DILI cases, only 9% of patients received less than 10 mg/day of medication, whereas 14% received 11–49 mg/day and 77% received more than 50 mg/day.15 In the Spanish Hepatotoxicity Registry, 77% of patients with DILI received medications with daily doses greater than 50 mg.24 Supporting these results, among nonacetaminophen cases of DILI that required liver transplantation in the United States from 1990 to 2002, 81% were reported to be caused by compounds that were taken in daily doses of greater than 50 mg/day.32 Further studies of the relationship between drug doses and DILI risk are important for the development of safe medications.

Table 2.

Relationship Between Daily Doses of Oral Medications and Hepatic Adverse Events

Outcome ≤10 mg
(n = 54)
10–50 mg
(n = 83)
≥50 mg
(n = 93)
P
value
ALT > 3 × ULN, n (%) 10 (19) 22 (27) 29 (31)   .10
Jaundice, n (%) 18 (33) 33 (40) 42 (45)   .16
Liver failure, n (%) 9 (17) 10 (12) 30 (32)   .009
Death, n (%) 6 (11) 9 (11) 26 (28)   .004
Transplant, n (%) 0 (0) 2 (2) 12 (13) <.001

ULN, upper limit of normal.

Reproduced from Lammert et al.35

Metabolism Characteristics

In associating the risk of DILI with hepatic metabolism of 207 of the most commonly prescribed oral medications in the United States, compounds with 50% or greater hepatic metabolism caused a significantly higher frequency (compared with drugs with less hepatic metabolism) of alanine aminotransferase (ALT) levels greater than 3 times the upper limit of normal (34% vs 10%; P = .007), liver failure (28% vs 9%; P = .001), liver transplantation (9% vs 1%; P = .045), and fatal DILI (23% vs 4%; P = .0003), but not jaundice (43% vs 34%; P = .2).36 Twelve compounds with no hepatic metabolism (risedronate, alendronate, hydrochlorothiazide, nadolol, cefdinir, cefprozil, gabapentin, metformin, cephalexin, benzonatate, cefuroxime, and sotalol) were not found to cause liver failure, liver transplantation, or fatal DILI.36 In analyses of the relationships between hepatic adverse events, hepatic metabolism, and daily dose, compounds with significant hepatic metabolism that were given at daily doses greater than 50 mg were significantly more hepatotoxic than compounds from all other groups.36 Compared with medications without biliary excretion, compounds with biliary excretion significantly increased the incidence of jaundice (67% vs 33%; P = .0006). There were potential differences among associations between cytochrome P450 enzymes (CYPs) and liver failure from DILI and fatal DILI; CYP 2C9 and 2C19 pathways generally were associated with more DILI than CYP3A and CYP2D6 pathways.36

Cross-Sensitization and Class Effect

Cross-sensitization means the risk of hepatotoxicity for agents with close chemical structure, whereas class-effect means hepatotoxicity risk for agents within a narrow therapeutic class (eg, cyclooxygenase-2 inhibitors or 3-hydroxy-3-methylglutaryl–coenzyme A reductase inhibitors). Although there are some examples of drug hepatotoxicity linked to cross-sensitivity and class effect, their overall impact on DILI is not well understood. Hypersensitivity from aromatic anticonvulsants might share common mechanisms and some patients could have cross-sensitivity to these drugs—cross-sensitivity frequently is observed between phenytoin and carbamazepine. 37 Cross-sensitivity has been reported from erythromycin derivatives,38 phenothiazines,39 haloalkane anesthetics,40 and anti-androgens.41 There is a case report of DILI that arose from cross-sensitivity between the propionic acid derivatives naproxen and fenoprofen.42 Cross-sensitivity also has been reported between tricyclic antidepressants such as amineptine and clomipramine43 as well as from trimipramine and despiramine.44 It might be reasonable to closely monitor for DILI in patients who restart tricyclic antidepressants who have had previous liver injury from tricyclic antidepressant agents.45 However, recent reports showed a lack of cross-hepatotoxicity among antifungal drugs; patients did not experience cross-hepatotoxicity between fluconazole and voriconazole46 or between voriconazole and posaconazole.47

Drug Interactions

The formation of toxic reactive metabolites during hepatic metabolism is believed to be the main pathogenic mechanism for DILI.12,48,49 Most drugs are bio-transformed by phase I and/or phase II metabolic reactions. Oxidation, reduction, and hydrolytic reactions are included in the phase I reactions, whereas phase II reactions include conjugation reactions and involve either esterification of the parent compound or a metabolite created by the phase I reactions. There are more than 20 different CYPs in the CYP450 family in human liver.50 CYPs are responsible for the most phase I reactions, and formation of reactive intermediates is more abundant in the centrilobular zone than in the periportal zone. Centrilobular necrosis is one of the characteristic features of severe DILI, so drug-metabolizing enzymes might mediate the pathogenesis of DILI.12,51 Certain drugs can modify the hepatotoxic potential of other drugs by enzyme induction and lead to formation of reactive metabolites. 52 Examples of CYP inducers are rifampicin, phenytoin, isoniazid, smoking, and ethanol. Some drug metabolizing and detoxification pathways might increase the risk for hepatotoxicity from other drugs, but there is little in vivo evidence to support this model. In a meta-analysis of studies of hepatotoxicity from isoniazid and rifampicin, the incidence of liver injury was significantly greater among patients who received this drug combination than those who received either as a single agent.53 Rifampicin, which can induce microsomal enzymes, seems to increase the risk of liver injury from isoniazid in frequency and latency.53,54 Pyrazinamide was reported to increase the hepatotoxicity of isoniazid.55

The neuroleptic agent thioridazine is a potent inhibitor of CYP2D6 that increases the plasma concentrations of the antidepressant trazodone.56 Interestingly, a combination of these drugs was reported to cause a case of fatal liver injury.57 Simvastatin is metabolized primarily by CYP3A4 and amiodarone is a recognized inhibitor of this enzyme; and interaction between these 2 might have accounted for hepatotoxicity in a 72-year-old man with concomitant rhabdomyolysis.58 Similarly, severe hepatotoxicity likely resulted from an interaction between raloxifene and fenofibrate because of CYP3A4 inhibition.59

The risk of valproate hepatotoxicity is increased by concomitant use of other anticonvulsants, presumably because of increased formation of reactive intermediaries during valproate metabolism.60,61 Covalent binding of these reactive metabolites normally is prevented by their conjugation with glutathione and subsequent urinary excretion because N-acetylation (NAC) conjugates (NAC I and II) in states of their increased formation.62 One study demonstrated high levels of NAC I and II in patients given valproate in combination with other anticonvulsants.61 Failure to detoxify these intermediates (eg, decreased glutathione pool or genetic variants) might lead to their accumulation, binding to liver proteins, and hepatotoxicity. However, another study did not associate serum or urine levels of 4-ene valproate with hepatotoxicity.62

Alcohol Consumption

Although the effects of acute and chronic alcohol use on the risk of acetaminophen hepatotoxicity are well established,63 the role of alcohol in idiosyncratic DILI is less clear. Alcohol consumption is one of the criteria in the RUCAM causality assessment instrument, although there is no evidence that alcohol consumption increases the risk of liver injury from medications other than methotrexate, isoniazid, or halothane.12 Consumption of large amounts of alcohol increases the risk of fibrosis/cirrhosis in long-term users of methotrexate.64,65 However, methotrexate alone might not cause severe liver fibrosis, other risk factors such as diabetes mellitus type 2, overweight, and heavy use of alcohol might contribute. 66,67 Chronic alcohol abuse might increase the hepatotoxicity of antituberculosis (anti-TB) drugs,68,69 possibly from alcohol-mediated induction of hepatic CYP2E1. However, other studies have shown that alcoholics might not have an increased risk of hepatotoxicity from isoniazid or other anti-TB drugs.22,70,71 Studies that associated alcohol with risk for idiosyncratic DILI12 might have been confounded by patients’ poor nutritional status and older age. Prospective registries did not find a significant association between alcohol consumption and the severity or chronicity of DILI.10,72 Although the package insert for duloxetine states that to prevent hepatotoxicity individuals with substantial alcohol consumption should not take the drug, there are no published data to show that alcoholism increases the risk of duloxetine hepatotoxicity. Additional studies are needed to determine the relationship between alcohol consumption and idiosyncratic DILI.

Underlying Disease States

There is controversy about whether chronic liver disease increases the risk for DILI. There is a belief that patients with chronic liver disease and cirrhosis are not necessarily prone to DILI,12,73,74 but patients with preexisting liver disease are at higher risk for complicated courses and adverse outcomes from DILI. A limited number of studies systematically have evaluated the safety of drugs in patients with pre-existing liver disease. Patients with increased baseline levels of aminotransferases have an increased risk of statin-induced hepatotoxicity.75 Several other studies confirmed the safety of statins in patients with chronic liver disease, including nonalcoholic fatty liver disease and hepatitis C.7678 One study reported that nonalcoholic fatty liver disease significantly increased the risk of DILI in middle-aged men,68 compared with men with hepatitis C (2.4% vs 0%; P < .05).79 Small sample sizes and other methodologic issues limit the validity of these observations. In a study of the clinical characteristics and outcome of disulfiram-associated liver injury, no differences in outcomes were related to pretreatment levels of enzymes.80 Some studies indicated that hepatitis B or C might be risk factors for hepatotoxicity of anti-TB agents,8183 whereas others did not confirm these findings.84 Many confounding factors must be considered in interpreting results from these studies; patient groups were not stratified according to hepatitis B virus (HBV) replication status and fluctuations in HBV-DNA levels and aminotransferase activities are common in these patients.85 The increased levels of ALT might have arisen from viral replication rather than hepatotoxicity. A greater proportion of patients with hepatitis C who were given anti-TB drugs developed hepatotoxicity than those without hepatitis C.86 Highly active antiretroviral therapy has been associated with hepatic adverse reactions in patients co-infected with hepatitis B or C and human immunodeficiency virus, compared with patients without chronic viral hepatitis. 8791 Furthermore, patients co-infected with human immunodeficiency virus and hepatitis C virus but cured of hepatitis C by interferon therapy were significantly less likely to show signs of hepatotoxicity to highly active antiretroviral therapy than nonresponders.92 However, patients with chronic hepatitis C have spontaneous fluctuations in levels of ALT and aspartate aminotransferase, so these factors are difficult to associated with drug-induced hepatotoxicity.93 Hepatitis B and C might increase susceptibility to DILI from anti-TB agents and highly active antiretroviral therapy, but studies are needed to specifically address this hypothesis. A recent case series illustrated the difficulties in interpreting abnormal liver test results in patients with human immunodeficiency virus and HBV co-infection.94 In these patients, aminotransferase levels increased significantly 6–12 weeks after highly active antiretroviral therapy began94; however, during the follow-up period, liver test results improved despite restarting or continuing the implicated medications.94

Other diseases might increase the risk of DILI, such as the higher risk of methotrexate-induced liver injury observed in patients with psoriasis, compared with patients with rheumatoid arthritis.95,96 However, confounding factors such as age, obesity, diabetes mellitus, and use of other potentially hepatotoxic drugs limit the validity of these observations. Obesity and diabetes mellitus are risk factors for methotrexate-induced hepatotoxicity.65,66 In 2 studies that tested rifampin for treating pruritus in patients with primary biliary cirrhosis, there was an increased frequency of hepatotoxicity (12.5% and 7.3%)97,98; similarly, rifampin might have caused DILI in 5% of patients when administered to treat brucellosis.99

Genetic Risk Factors

Because of their rarity and unpredictability, idiosyncratic DILI events are considered to have a strong genetic basis,12,1519 but significant association between certain genetic traits and DILI has been shown for only a few compounds.1518,100 Even when such an association has been observed, generally the odds ratio (OR) of a given haplotype to increase the risk of DILI has been rather low (with the exception of flucloxacillin).100 DILI is likely a complex genetic disorder in which multiple genetic variants along with environmental risk factors are responsible for liver injury. Unraveling the genetic basis for DILI has been and will remain challenging because of the vast number of compounds that can cause DILI, its rarity and variable clinical presentation, and diagnostic difficulties.

Genetic studies conducted to date largely have been hypothesis-driven and involved a candidate compound–candidate gene approach.1519 These studies predominantly have focused on drug disposition and immunologic mechanisms. More recently, a few genome-wide association studies of DILI caused by individual compounds have been published (vide infra). Several consortia (DILI network, DILIGEN, Spanish Hepatotoxicity Registry, and Serious Adverse Event Consortium) are collecting genomic DNA from patients with well-phenotyped DILI and novel observations are starting to emerge from their work.101103 A multistep model of the mechanistic and genetic basis for idiosyncratic DILI has been proposed.15,16,21 It involves upstream drug-specific pathways that generate reactive metabolites and downstream common pathways that cause cell stress and death, either directly or through immune-mediated mechanisms.

Variations in Phase 1 Drug-Metabolizing Enzymes

Formation of toxic reactive metabolites by cytochrome P450 enzymes (CYPs) is considered to be required for the pathogenesis of DILI.1921 There is considerable interindividual variation in the activity of different CYPs and many CYPs are polymorphic. Several studies have investigated their role in the pathogenesis of idiosyncratic DILI, but evidence is not strong to support their role, except for a few compounds.16

CYP3A is the most abundant and most important phase 1 enzyme; it consists of 3 isoforms: 3A4, 3A5, and 3A7. CYP3A4 is not polymorphic whereas CYP3A5 and CYP3A7 are. There is no evidence to associate variations in CYP3A with DILI.

CYP2C9 is an important phase 1 enzyme involved in the metabolism of several important therapeutic agents such as nonsteroidal anti-inflammatory drugs, phenytoin, and warfarin. CYP2C9 shows functional polymorphism (CYP2C9*1,*2, and *3)104; some polymorphisms initially were associated with diclofenac hepatotoxicity,105 but subsequent studies failed to confirm any significant association.106108

CYP2C19 is another important phase 1 enzyme that also has functional polymorphisms. It metabolizes proton pump inhibitors, antidepressants, and antiepileptics. A case-series study of 3 patients with atrium hepatotoxicity109 and a small case-series study of troglitazone hepatotoxicity indicated a role for CYP2C19 polymorphisms, 110 but there are no other strong data to implicate CYP2C19 genetic polymorphism in causing DILI. The Spanish registry failed to show significant enrichment of CYP2C9 and CYP2C19 polymorphisms in a well-characterized cohort of DILI patients.111

CYP2D6 is highly polymorphic and metabolizes opiates, antidepressants, β-blockers, and anti-arrhythmic agents. Polymorphisms in CYP2D6 have been associated with hepatotoxicity from perhexiline, senna, chlorpromazine, and other drugs.16 CYP2E1 is polymorphic but it is not clear how variants affect the function of the gene or its product. Nonetheless, there is some evidence that CYP2E1 mutant genotypes can lower the risk of isoniazid hepatotoxicity, compared with the CYP2E1*1A/*1A genotype (vide infra).112114 CYPs are the usual and first suspects in DILI pathogenesis, but studies do not support a major role for their variants in this process.

Variations in Phase 2 and Detoxifying Enzymes

N-acetylation is an important phase II reaction and N-acetyltransferase 2 (NAT2) has been implicated in DILI.115119 NAT2 is highly polymorphic and there is interindividual variability in its metabolic activity. NAT2*4 has the highest acetylation activity, whereas *5,*6, and *7 reduced enzymatic activity. Slow rates of acetylation have been associated with increased risk of hepatotoxicity from sulfonamides and isoniazid115,117 and also were associated with severe isoniazid hepatotoxicity. 112 Isoniazid is metabolized primarily in the liver, initially by NAT2 into acetyl-isoniazid; this is hydrolyzed rapidly into acetyl-hydrazine, which is either oxidized by CYP2E1 into toxic reactive metabolites or acetylated to form diacetyl-hydrazine.112,120 “Slow acetylators” presumably do not detoxify acetyl-hydrazine rapidly, promoting oxidation by CYP2E1 into toxic intermediaries. A recent meta-analysis showed a strong relationship between NAT2 genotype and isoniazid hepatotoxicity in Asians (OR, 2.52).114 NAT2 and CYP2E1 polymorphisms might act synergistically to affect predisposition to isoniazid hepatotoxicity.112 Huang et al112 showed that NAT2 slow acetylators (CYP2E1 c1/c1 genotype) were at significantly higher risk of isoniazid hepatotoxicity than rapid acetylators (CYP2E1 c1/c2 or c2/c2 genotypes; OR, 7.43).

Glutathione S-transferase and manganese superoxide dismutase (MnSOD) defend against cellular oxidative stress, so their roles in DILI have been explored. In human beings, cytosolic glutathione S-transferases T1 and M1 enzymes are polymorphic; their reduced expression could affect these pathways. This hypothesis recently was tested in 154 patients with idiosyncratic DILI caused by multiple agents, 250 age- and sex-matched population controls, and 88 medication-matched controls. 121 Individuals with glutathione S-transferase T- and glutathione S-transferase M1-null genotypes had significantly higher risk of DILI compared with individuals who expressed these gene products (OR, 2.70). This relationship persisted for DILI caused by antibacterials (n = 44; OR, 3.12) and nonsteroidal anti-inflammatory drugs (n = 19; OR, 5.61). However, it is possible that the relationship between these null genotypes and DILI could be accounted for by the relationship between null genotype and antibacterial and nonsteroidal anti-inflammatory drug DILI. When investigators reanalyzed the data, they found no significant association between null genotypes and DILI from agents other than antibacterials and nonsteroidal anti-inflammatory drugs (n = 92; OR, 1.97) (Dr Lucena, personal communication).

MnSOD is a mitochondrial enzyme that detoxifies reactive oxygen species into hydrogen peroxide, which is reduced to water by either catalase or glutathione peroxidase. MnSOD is functionally polymorphic and specific alleles (genotypes C/C or C/T) import greater levels of MnSOD protein into the mitochondrial matrix, with 30%–40% increases in activity. In a study of 115 patients with DILI caused by multiple agents (63 from anti-TB agents) and 115 matched controls, individuals with the MnSOD C/C or C/T genotypes had a significantly higher risk for overall DILI (OR, 2.44) or DILI from anti-TB medications (OR, 2.47).122 It is unclear how increased MnSOD activity might increase the risk of hepatotoxicity, but there might be increased production of hydrogen peroxide.122

Although glucuronidation is a detoxification process, it sometimes can generate toxic reactive intermediaries. Diclofenac undergoes glucuronidation by UGT2B7 to produce acyl glucuronide123; acyl glucuronides of diclofenac can undergo acyl migration and imine formation or nucleophilic displacement—either can cause irreversible binding to hepatic macromolecules.124 In a rat model of diclofenac hepatotoxicity, formation of diclofenac adducts was prevented by glucuronidation inhibitors, indicating that glucuronidation of diclofenac might induce formation of adducts.125 Furthermore, these adducts were localized mostly in zone 3 of the liver lobule, indicating that diclofenac adducts might have a role in the hepatotoxicity.125 Although diclofenac predominantly is metabolized by CYP2C9, polymorphisms do not appear to be a risk factor for diclofenac hepatotoxicity.126 In contrast, polymorphisms in UGT2B7 and CYP2C8, enzymes that generate reactive diclofenac metabolites, were risk factors for diclofenac hepatotoxicity.123 In the same study, an allelic variant of ABCC2, which encodes multidrug resistance protein (MRP2), which mediates biliary excretion of reactive metabolites, was associated with diclofenac hepatotoxicity.123 Patients with at least one UGT2B7*2 allele were at significantly higher risk for diclofenac-induced hepatotoxicity (OR, 8.5) compared with patients who took diclofenac but had no liver injury or healthy controls.123 It is conceivable that patients who develop diclofenac hepatotoxicity are rapid glucuronidators who have high concentrations of the unstable and reactive acyl glucuronide in their hepatocytes.

Hepatobiliary Transporters

Hepatic detoxification of xenobiotics results in their anionic conjugates with glutathione, sulfate, and glucuronate. These conjugated xenobiotics become substrates for hepatic drug transport, another potential step for drug hepatotoxicity.127,128 Drug metabolites actively are transported across hepatocyte membranes by transporters (uptake or efflux transporters) on the canalicular or the apical membranes.127,128 In the liver, transport at the basolateral membrane involves the organic anion-transporting polypeptide and in the organic anion transporter. 127 Limited data exist on defects in these uptake transporters and associated hepatotoxicity.127 Basolateral transport processes probably determine hepatic exposure to drugs and their metabolites, which reach the canalicular membrane,129 but their inhibition does not appear to increase the risk of hepatotoxicity.130 However, inhibition of efflux proteins can lead to cholestatic liver injury caused by certain compounds or their metabolites. 128 The efflux of drugs into bile involves canalicular transporters of the MRP family, which includes the glycoproteins MDR1 (ABCB1), MDR3 (ABCB4), MRP2 (ABCC2), and bile salt export pump (BSEP, ABCB11).128 Drugs that inhibit export on the canalicular side through inhibition of BSEP can lead to cholestasis in susceptible subjects.128 Cholestatic liver injury from sulindac, flucloxacillin, terbinafine, and bosentan have been associated with inhibition of the canalicular BSEP.131133 The endothelin antagonist, bosentan (used to treat pulmonary hypertension) inhibits BSEP, leading to accumulation of toxic bile acids within the hepatocytes.134,135 In a recent study of 110 human liver samples obtained at the time of resection for focal lesions, there was significant interindividual variability in canalicular levels of BSEP, MDR3, MDR2, and MRP2 proteins. For example, 30% of samples contained high levels of these transporters whereas 32% had low or very low levels.136 Limited data are available on the relationship between their polymorphisms and susceptibility to DILI.137 However, familial intrahepatic cholestasis and intrahepatic cholestasis of pregnancy have been associated with polymorphisms or mutations in BSEP and MDR3137,138; patients with intrahepatic cholestasis during pregnancy are at increased risk for cholestatic liver injury when they take oral contraceptives.139,140 Furthermore, mutations in BSEP have been associated with cholestatic episodes in patients taking antibiotics or nonsteroidal anti-inflammatory drugs.141 Mutations that disrupt BSEP function have been identified in patients with a history of drug-induced cholestasis.142 Patients who carry mutations in genes that encode BSEP or MDR3 have a 3-fold increase in risk of cholestatic liver injury from oral contraceptives, certain antibiotics, proton pump inhibitors, and psychotropic drugs.142 Further studies are needed to identify defects in the transporter proteins that can mediate pathogenesis of DILI.

Immunologic Mechanisms

The innate and acquired immune systems are each involved in the pathogenesis of idiosyncratic DILI; these might represent the most important events in the pathogenesis of DILI.1921 Eosinophilia in peripheral blood and/or in the liver of a patient with acute liver injury supports a diagnosis of DILI.51 Hypersensitivity features such as rash, fever, and eosinophilia are observed in 25%–30% of patients suspected of having DILI.10,143 However, liver injury might have an immunologic basis in patients without obvious clinical or biochemical evidence of hypersensitivity reactions.144,145 Most patients suspected to have DILI from hepatotoxic drugs have evidence of an inflammatory response in liver biopsy samples, 145 but this response could be a consequence of liver injury rather than a cause of DILI. A number of studies have shown a strong relationship between genetic polymorphisms that influence immune function (eg, HLA class II antigens or cytokines) and risk of DILI.1518 The hapten, danger, and pharmacologic interaction hypotheses all have been proposed to mediate DILI pathogenesis (for review see Uetrecht,19,20 Kaplowitz,21 and Pichler146). Idiosyncratic DILI from hepatotoxic drugs might arise from immune-mediated haptenization of reactive metabolites and subsequent danger signals that activate the immune response.

Association Between DILI and Specific HLA Haplotypes

There is evidence for an association between polymorphisms in HLA class II antigens and adverse drug reactions; specific HLA haplotypes have been associated with nonhepatic147,148 and hepatic adverse reactions (Table 3).144,149151 An association between HLA-B*5701 and abacavir hypersensitivity led to the concept that prospective testing for this allele might minimize the risk of abacavir hypersensitivity.152 In a prospective, doubleblind study of a predominantly white population, not administering abacavir to individuals with the HLA-B* 5701 allele virtually eliminated the immunologically confirmed hypersensitivity reactions (OR, 0.03).152

Table 3.

Summary of Various Studies That Investigated or Identified Relationships Between HLA Antigens and DILI

Study and
location
DILI
compound
Cases and
controls
Type of
study
Significant HLA allele(s) Frequency cases
vs controls
Result P value, OR (95% CI)
Hautekeete et al,149 1999
Belgium
Amox/clav 35 cases
60 bone marrow donors
CGAS HLAB1*1501 57% vs 12% Pc < .0002
O’Donohue et al,153 2000 UK Amox/clav 22 cases
134 controls
CGAS HLAB1*1501 70% vs 20% P = 2.5 × 10−6 OR, 9.25 (95% CI, N/A)
Donaldson et al,101 2008 UK Amox/clav 52 cases
39 controls
189 population controls
CGAS DRB1*15
DRB1*07 (protective allele)
50% vs 30%
N/A
P = .0032 OR, 2.45 (95% CI, 1.37–4.8)
Versus community controls 0.2 (0.02–0.59, Pc = .02)
Versus exposed controls 0.13 (0.04–0.44, Pc = .0062)
Andrade et al,154 2004 Spain Amox/clav 27 cases
635 controls
CGAS DQB1*06 74% vs 41% Pc = .015 OR, 4.14 (1.73–9.95)
Sharma et al,150 2002 India Anti-TB 56 cases
290 controls
CGAS DQB1*0201
DRA1*0103 (protective allele)
52% vs 23%
6% vs 39%
2.1 (1.0–4.18)
0.2 (0.04–0.69); Pc = .01
Donaldson,151 2008 UK Diclofenac 4 cases
189 population controls
GGAS DRB1*13 (protective)
DRB1*04
0% vs 18%
45% vs28%
OR, 0.19 (0.02–1.45)
Not significant
Berson et al,155 1994 France Different compounds 71
2163 population controls
CGAS HLA-A11 23% vs 12% <.01 but Pc = NS
No significant differences in HLA B antigen, HLD DR and DQ antigens tested
Andrade et al,154 2004 Spain Different compounds 140 cases
635 healthy controls
CGAS No differences in DRB1 and DQB antigens between cases and controls Compared with hepatocellular DILI, cholestatic/mixed had significantly higher prevalence of DRB1*15 and DQB1*06 and significantly lower prevalence of DQB1*02 antigens
Kindmark et al,144 2008 Northern
Europe
Ximelagatran 74 cases
130 treated controls
GWAS DRB1*07
DQA1*02
26% vs 8.5% 4.41 (2.22–8.87) (P = 9.1 × 10−6)
4.41 (2.21–8.8) (P = 1.3 × 10−5)
Daly et al,102 2009 UK Flucloxacillin 51 original and 21 replication cases
282 population controls
64 drug exposure controls
GWAS DRB1*5701 83% vs 6.3%
87% replication cases vs 6.3% controls
80.6 (22.8–284.9) P = 8.7 × 10−33
100.0 (20.6–485.8)

Amox/clav, amoxicillin/clavulanate; CGAS, candidate gene association study; GWAS, genome-wide association study; Pc, corrected P value.

One of the first HLA haplotypes associated with DILI, HLA B1*1501-DRB5*0101-DQB1*0602,149 is carried by approximately 57% of patients with amoxicillin/clavulanate-induced DILI, but in only 12% of healthy bone-marrow donors.149 Two subsequent studies from the United Kingdom confirmed the association between HLA DRB1*15 and liver injury amoxicillin/clavulanate.101,153 A more recent study based on the Spanish Registry did not confirm this association, but patients with amoxicillin/clavulanate-associated DILI had a significantly higher prevalence of HLA-DQR1*06 than controls.154 Liver injury from anti-TB reagents has been associated with specific HLA II alleles150; in a study of 56 North Indian patients with DILI and 290 controls (also exposed to anti-TB medicines), the presence of the HLA-DQB1*0201 allele (adjusted OR, 1.9) or a lack of HLA-DQA1*0102 (adjusted OR, 4.0) were associated independently with liver injury from anti-TB medicines.150 These observations require validation.

In 2 case series consisting of a relatively modest number of patients with DILI suspected to be caused by different agents, no specific HLA alleles were associated with DILI.154,155 However, Andrade et al154 reported a potential association between specific HLAs and DILI phenotype. Compared with patients with hepatocellular DILI, those with cholestatic/mixed DILI had a significantly higher frequency of HLA-DRB1*15 and HLA-DQB1* 06 alleles and a lower frequency of DRB1*07 and DQB1*02 alleles.154 Further studies are warranted to investigate the relationship between genetic variations and DILI phenotype.

Recent genome-wide association studies associated specific HLA alleles with DILI caused by ximelagatrin and flucloxacillin.102,144 Ximelagatran is an oral thrombin inhibitor that was withdrawn from the market because of serious hepatotoxicity—a somewhat surprising finding because it is not metabolized by the liver. Kindmark et al144 conducted genome-wide association studies and a large, candidate gene analysis study of DNA from 74 patients with ALT levels greater than 3 times the upper limit of normal with ximelagatran and 130 treated individuals without hepatotoxicity. Increased levels of ALT were associated with the major histocompatibility complex alleles DRB1*07 (OR, 4.41) and DQA1*02 (OR, 4.41) and this association was validated in a study of 10 cases and 16 controls.144 They also reported corroborative immunologic studies supporting the biological significance of DRB1*07 in ximelagatran-induced liver injury. Daly et al,102 under the auspices of the DILIGEN and International SAE consortium, recently reported the results of their landmark genome-wide association studies of flucloxacillin DILI (51 cases, 282 matched controls, and 64 controls who received flucloxacillin without liver injury). This study showed an association peak in the major histocompatibility complex region with the strongest association observed for rs2395029 [G] (P = 8.7 × 10−33), a marker in complete linkage disequilibrium with HLA-B* 570.1.102 Direct genotyping for HLA-B*5701 in 51 cases and 63 drug-exposed controls revealed a very strong relationship between this allele and flucloxacillin-induced liver injury (OR, 80.6). This finding was confirmed in 23 replication cases (OR, 100), but unfortunately a separate control group was not used for this comparison.102 There were additional significant single nucleotide polymorphisms (HLA DRB1*0701, tumor necrosis factor rs361525, tumor necrosis factor rs1799964, HSPAIL rs2227956), but their significance was deemed not independent of HLA-B* 5701. Although the association between HLA-B*5701 and DILI was a remarkable discovery, it was estimated that only 1 in every 500–1000 individuals who carry HLA-B*5701 will develop liver injury upon receiving flucloxacillin (an estimated population attributable fraction was 0.64).102 Many other undiscovered genetic variations are likely to be determinants of flucloxacillin-induced liver injury.

Dysregulation of Cytokines

Dysregulation of cytokine production also might mediate the pathogenesis of DILI in a non–medication-specific manner. Aithal et al125 investigated polymorphisms in interleukin (IL)-10, IL-4, and IL-4 receptors in 24 patients with diclofenac-induced liver injury, 48 diclofenac-exposed controls, and as many as 321 healthy controls; they observed a higher frequency of IL-10 and IL-4 variants in patients with diclofenac-induced DILI compared with controls. The investigators speculated that polymorphisms that increase IL-10 or reduce IL-4 expression might contribute to a T-helper cell 2–mediated antibody response to diclofenac-induced neoantigens. Pachkoria et al156 investigated IL-10, IL-4, and tumor necrosis factor-α genetic polymorphism in 140 patients with DILI from different agents and 268 healthy controls. IL-10 haplotypes that reduced expression levels were more common in patients with DILI without peripheral eosinophilia (OR, 5.29). All DILI cases with serious outcomes carried haplotypes that resulted in low or intermediate levels of IL-10 expression, and had normal or low numbers of eosinophils.156 The investigators attempted to correlate IL-10 genotypes with the severity of liver injury, but the study included too few cases of severe liver injury for this analysis.

Mitochondrial DNA Mutations

Some drugs (eg, valproate, salicylate, and antiretroviral agents) cause liver injury through mitochondrial toxicity; a recent preliminary study associated mitochondrial DNA mutations with liver injury caused by some compounds.103 Seventeen patients with suspected valproate hepatotoxicity enrolled in the DILI network studies were assessed for genetic variations in the mitochondrial DNA polymerase gamma (POLG) gene. POLG was sequenced and the frequency of genetic variants was compared between cases and historical controls. Common heterozygous genetic variants of POLG were associated significantly with valproate hepatotoxicity (OR, 23.6). The investigators speculated that impaired liver regeneration caused by genetically impaired POLG activity may play a role in the pathogenesis of valproate hepatotoxicity.103

Future Directions

DILI is a problem that is important not only to patients, but also to physicians, regulatory agencies, and drug developers. Although we have gained insight into its pathogenic mechanisms, we have much to learn about its clinical manifestations and factors that might be used in the diagnosis and determination of prognosis. Prospective registries are an important source of data, but firm and systematically established postmarketing surveillance methods urgently are required. Pharmaceutical companies and regulatory agencies have large amounts of data from early- and late-stage clinical trials, but because of confidentiality and proprietary issues it is difficult to evaluate and compare these data and there is no easy access for academic investigators. The annual workshops organized by the United States Food and Drug Administration/CDER-AASLD-PhRMA HepTox Steering Group might be a starting point for initiating discussions on data sharing and building partnerships beyond the current boundaries. Liver injury from dietary supplements is another area that requires more data collection and analysis to better understand its epidemiology and pathogenic mechanisms.

Abbreviations used in this paper

anti-TB

antituberculosis

BSEP

bile salt export pump

DILI

drug-induced liver injury

IL

interleukin

MnSOD

manganese superoxide dismutase

MRP

multidrug resistance protein

NAT2

N-acetyltransferase 2

OR

odds ratio.

Footnotes

Conflicts of interest

Dr Chalasani has served as a paid consultant within the preceding 12 months to Teva, Eli Lilly, Karobio, Salix, Debiovision, Amylin, Genentech, Abbott, and Gilead on issues related to drug safety, and has research support from Eli Lilly and Monarch LifeSciences; and Dr Björnsson has served as a paid consultant for Astellas Pharma Europe, AstraZeneca, and Karobio.

References

  • 1.Bell LN, Chalasani N. Epidemiology of idiosyncratic drug-induced liver injury. Semin Liver Dis. 2009;29:337–347. doi: 10.1055/s-0029-1240002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Larrey D. Epidemiology and individual susceptibility to adverse drug reactions affecting the liver. Semin Liver Dis. 2002;22:145–155. doi: 10.1055/s-2002-30105. [DOI] [PubMed] [Google Scholar]
  • 3.Garcia Rodriguez LA, Ruigomez A, Jick H. A review of epidemiologic research on drug-induced acute liver injury using the general practice research data base in the United Kingdom. Pharmacotherapy. 1997;17:721–728. [PubMed] [Google Scholar]
  • 4.de Abajo FJ, Montero D, Madurga M, et al. Acute and clinically relevant drug-induced liver injury: a population based case-control study. Br J Clin Pharmacol. 2004;58:71–80. doi: 10.1111/j.1365-2125.2004.02133.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Sgro C, Clinard F, Ouazir K, et al. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology. 2002;36:451–455. doi: 10.1053/jhep.2002.34857. [DOI] [PubMed] [Google Scholar]
  • 6.Ostapowicz G, Fontana RJ, Schiodt FV, et al. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med. 2002;137:947–954. doi: 10.7326/0003-4819-137-12-200212170-00007. [DOI] [PubMed] [Google Scholar]
  • 7.Vuppalanchi R, Liangpunsakul S, Chalasani N. Etiology of new-onset jaundice: how often is it caused by idiosyncratic drug-induced liver injury in the United States? Am J Gastroenterol. 2007;102:558–562. doi: 10.1111/j.1572-0241.2006.01019.x. [DOI] [PubMed] [Google Scholar]
  • 8.Jinjuvadia K, Kwan W, Fontana RJ. Searching for a needle in a haystack: use of ICD-9-CM codes in drug-induced liver injury. Am J Gastroenterol. 2007;102:2437–2443. doi: 10.1111/j.1572-0241.2007.01456.x. [DOI] [PubMed] [Google Scholar]
  • 9.Bjornsson E, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology. 2005;42:481–489. doi: 10.1002/hep.20800. [DOI] [PubMed] [Google Scholar]
  • 10.Andrade RJ, Lucena MI, Fernandez MC, et al. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology. 2005;129:512–521. doi: 10.1016/j.gastro.2005.05.006. [DOI] [PubMed] [Google Scholar]
  • 11.Chalasani N, Fontana RJ, Bonkovsky HL, et al. Causes, clinical features, and outcomes from a prospective study of drug-induced liver injury in the United States. Gastroenterology. 2008;135:1924–1934. doi: 10.1053/j.gastro.2008.09.011. e1921–1924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zimmerman H. Hepatotoxicity: the adverse effects of drugs and other chemicals on the liver. 2nd ed. Philadelphia: Lippincott, Williams & Wilkins; 1999. [Google Scholar]
  • 13.FDA’s Guidance for Industry. Drug induced liver injury: premarketing clinical evaluation. [Accessed: August 20, 2009];2009 July; Available at: http://www.fda.gov/ucm/groups/fdagov-public/fdagov-drugs-gen/documents/document/ucm174090.pdf.
  • 14.Papay JI, Clines D, Rafi R, et al. Drug-induced liver injury following positive drug rechallenge. Regul Toxicol Pharmacol. 2009;54:84–90. doi: 10.1016/j.yrtph.2009.03.003. [DOI] [PubMed] [Google Scholar]
  • 15.Russmann S, Kullak-Ublick GA, Grattagliano I. Current concepts of mechanisms in drug-induced hepatotoxicity. Curr Med Chem. 2009;16:3041–3053. doi: 10.2174/092986709788803097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Andrade RJ, Robles M, Ulzurrun E, et al. Drug-induced liver injury: insights from genetic studies. Pharmacogenomics. 2009;10:1467–1487. doi: 10.2217/pgs.09.111. [DOI] [PubMed] [Google Scholar]
  • 17.Daly AK, Day CP. Genetic association studies in drug-induced liver injury. Semin Liver Dis. 2009;29:400–411. doi: 10.1055/s-0029-1240009. [DOI] [PubMed] [Google Scholar]
  • 18.Wilke RA, Lin DW, Roden DM, et al. Identifying genetic risk factors for serious adverse reactions; current progress and challenges. Nat Rev Drug Discov. 2007;6:904–916. doi: 10.1038/nrd2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Uetrecht J. Idiosyncratic drug reactions: current understanding. Annu Rev Pharmacol Toxicol. 2007;47:513–539. doi: 10.1146/annurev.pharmtox.47.120505.105150. [DOI] [PubMed] [Google Scholar]
  • 20.Uetrecht JP. New concepts in immunology relevant to idiosyncratic drug reactions: the “danger hypothesis” and innate immune system. Chem Res Toxicol. 1999;12:387–395. doi: 10.1021/tx980249i. [DOI] [PubMed] [Google Scholar]
  • 21.Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov. 2005;4:489–499. doi: 10.1038/nrd1750. [DOI] [PubMed] [Google Scholar]
  • 22.Fountain FF, Tolley E, Chrisman CR, et al. Isoniazid hepatotoxicity associated with treatment of latent tuberculosis infection: a 7-year evaluation from a public health tuberculosis clinic. Chest. 2005;128:116–123. doi: 10.1378/chest.128.1.116. [DOI] [PubMed] [Google Scholar]
  • 23.Lucena MI, Andrade RJ, Fernandez MC, et al. Determinants of the clinical expression of amoxicillin-clavulanate hepatotoxicity: a prospective series from Spain. Hepatology. 2006;44:850–856. doi: 10.1002/hep.21324. [DOI] [PubMed] [Google Scholar]
  • 24.Lucena MI, Andrade RJ, Kaplowitz N, et al. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and gender. Hepatology. 2009;49:2001–2009. doi: 10.1002/hep.22895. [DOI] [PubMed] [Google Scholar]
  • 25.Cotreau MM, von Moltke LL, Greenblatt DJ. The influence of age and sex on the clearance of cytochrome P450 3A substrates. Clin Pharmacokinet. 2005;44:33–60. doi: 10.2165/00003088-200544010-00002. [DOI] [PubMed] [Google Scholar]
  • 26.Herrlinger C, Klotz U. Drug metabolism and drug interactions in the elderly. Best Pract Res Clin Gastroenterol. 2001;15:897–918. doi: 10.1053/bega.2001.0249. [DOI] [PubMed] [Google Scholar]
  • 27.Wynne HA, Cope LH, Mutch E, et al. The effect of age upon liver volume and apparent liver blood flow in healthy man. Hepatology. 1989;9:297–301. doi: 10.1002/hep.1840090222. [DOI] [PubMed] [Google Scholar]
  • 28.Meier Y, Cavallaro M, Roos M, et al. Incidence of drug-induced liver injury in medical inpatients. Eur J Clin Pharmacol. 2005;61:135–143. doi: 10.1007/s00228-004-0888-z. [DOI] [PubMed] [Google Scholar]
  • 29.Shapiro MA, Lewis JH. Causality assessment of drug-induced hepatotoxicity: promises and pitfalls. Clin Liver Dis. 2007;11:477–505. doi: 10.1016/j.cld.2007.06.003. [DOI] [PubMed] [Google Scholar]
  • 30.Bjornsson E, Davidsdottir L. The long-term follow-up after idiosyncratic drug-induced liver injury with jaundice. J Hepatol. 2008;50:511–517. doi: 10.1016/j.jhep.2008.10.021. [DOI] [PubMed] [Google Scholar]
  • 31.De Valle MB, Av Klinteberg V, Alem N, et al. Drug-induced liver injury in a Swedish University hospital out-patient hepatology clinic. Aliment Pharmacol Ther. 2006;24:1187–1195. doi: 10.1111/j.1365-2036.2006.03117.x. [DOI] [PubMed] [Google Scholar]
  • 32.Russo MW, Galanko JA, Shrestha R, et al. Liver transplantation for acute liver failure from drug induced liver injury in the United States. Liver Transpl. 2004;10:1018–1023. doi: 10.1002/lt.20204. [DOI] [PubMed] [Google Scholar]
  • 33.Kenyon KW, Nappi JM. Bosentan for the treatment of pulmonary arterial hypertension. Ann Pharmacother. 2003;37:1055–1062. doi: 10.1345/aph.1C256. [DOI] [PubMed] [Google Scholar]
  • 34.Otani K, Kaneko S, Tasaki H, et al. Hepatic injury caused by mianserin. BMJ. 1989;299:519. doi: 10.1136/bmj.299.6697.519-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lammert C, Einarsson S, Saha C, et al. Relationship between daily dose of oral medications and idiosyncratic drug-induced liver injury: search for signals. Hepatology. 2008;47:2003–2009. doi: 10.1002/hep.22272. [DOI] [PubMed] [Google Scholar]
  • 36.Lammert C, Niklasson A, Bjornsson E, et al. Oral medications with significant hepatic metabolism are at higher risk for hepatic adverse events. Hepatology. 2010;51:615–620. doi: 10.1002/hep.23317. [DOI] [PubMed] [Google Scholar]
  • 37.Shear NH, Spielberg SP. Anticonvulsant hypersensitivity syndrome: in vitro assessment of risk. J Clin Invest. 1988;82:1826–1832. doi: 10.1172/JCI113798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Keefe EB, Reis TC, Berland JE. Hepatotoxicity to both erythromycin estolate and erythromycin ethylsuccinate. Dig Dis Sci. 1982;27:701–704. doi: 10.1007/BF01393764. [DOI] [PubMed] [Google Scholar]
  • 39.Herron G, Bourda S. Jaundice secondary to promazine and an analysis of possible cross sensitivities between phenothiazines derivatives. Gastroenterology. 1960;38:87–90. [PubMed] [Google Scholar]
  • 40.Lewis JH, Zimmermn HJ, Ishak KG, et al. Enflurane hepatotoxicity. A clinicopathologic study of 24 cases. Ann Intern Med. 1983;98:984–992. doi: 10.7326/0003-4819-98-6-984. [DOI] [PubMed] [Google Scholar]
  • 41.Andrejak A, Davion T, Gineston JL, et al. Cross hepatotoxicity between non-steroidal anti-inflammatory. BMJ. 1987;295:180–181. doi: 10.1136/bmj.295.6591.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Larrey D, Rueff B, Pessayre D, et al. Cross hepatotoxicity between tricyclic antidepressants. Gut. 1986;27:726–727. doi: 10.1136/gut.27.6.726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Remy AJ, Larrey D, Pageaux GP, et al. Cross hepatotoxicity between tricyclic antidepressants and phenothiazines. Eur J Gastroenterol Hepatol. 1995;7:373–376. [PubMed] [Google Scholar]
  • 44.Lucena MI, Carvajal A, Andrade RJ, et al. Antidepressants-induced hepatotoxicity. Expert Opin Drug Saf. 2003;2:1–14. doi: 10.1517/14740338.2.3.249. [DOI] [PubMed] [Google Scholar]
  • 45.Miquel M, Soler A, Vaque A, et al. Suspected cross-hepatotoxicity of flutamide and cyproterone acetate. Liver Int. 2007;27:1144–1147. doi: 10.1111/j.1478-3231.2007.01549.x. [DOI] [PubMed] [Google Scholar]
  • 46.Spelberg B, Rieg G, Bayer A, et al. Lack of cross hepatotoxicity between fluconazole and voriconazole. Clin Infect Dis. 2003;36:1091–1093. doi: 10.1086/374255. [DOI] [PubMed] [Google Scholar]
  • 47.Foo H, Gottlieb T. Lack of cross-hepatotoxicity between voriconazole and posaconazole. Clin Infect Dis. 2007;45:803–805. doi: 10.1086/521174. [DOI] [PubMed] [Google Scholar]
  • 48.Knowles SR, Uetrecht J, Shear NH. Idiosyncratic drug reactions: the reactive metabolite syndromes. Lancet. 2000;356:1587–1591. doi: 10.1016/S0140-6736(00)03137-8. [DOI] [PubMed] [Google Scholar]
  • 49.Pessayre D. Role of reactive metabolites in drug-induced hepatitis. J Hepatol. 1995;23 Suppl 1:16–24. [PubMed] [Google Scholar]
  • 50.Hasler JA. Pharmacogenetics of cytochromes P450. Mol Aspects Med. 1999;20:12–137. doi: 10.1016/s0098-2997(99)00005-9. [DOI] [PubMed] [Google Scholar]
  • 51.Bjornsson E, Kalaitzakis E, Olsson R. The impact of eosinophilia and hepatic necrosis on prognosis in patients with drug-induced liver injury. Aliment Pharmacol Ther. 2007;25:1411–1421. doi: 10.1111/j.1365-2036.2007.03330.x. [DOI] [PubMed] [Google Scholar]
  • 52.Yun CH, Okerholm RA, Guengerich FP. Oxidation of the antihistaminic drug terfenadine in human liver microsomes. Role of cytochrome P-450 3A(4) in N-dealkylation and C-hydroxylation. Drug Metab Dispos. 1993;21:403–409. [PubMed] [Google Scholar]
  • 53.Steele MA, Burk RF, DesPrez RM. Toxic hepatitis with isoniazid and rifampin. A meta-analysis. Chest. 1991;99:465–471. doi: 10.1378/chest.99.2.465. [DOI] [PubMed] [Google Scholar]
  • 54.Pessayre D, Bentata M, Degott C, et al. Isoniazid-rifampin fulminant hepatitis. A possible consequence of the enhancement of isoniazid hepatotoxicity by enzyme induction. Gastroenterology. 1977;72:284–289. [PubMed] [Google Scholar]
  • 55.Durand F, Bernuau J, Pessayre D, et al. Deleterious influence of pyrazinamide on the outcome of patients with fulminant or subfulminant liver failure during antituberculous treatment including isoniazid. Hepatology. 1995;21:929–932. [PubMed] [Google Scholar]
  • 56.Yasui N, Otani K, Kaneko S, et al. Inhibition of trazodone metabolism by thioridazine in humans. Ther Drug Monit. 1995;17:333–335. doi: 10.1097/00007691-199508000-00003. [DOI] [PubMed] [Google Scholar]
  • 57.Hull M, Jones R, Bendall M. Fatal hepatic necrosis associated with trazodone and neuroleptic drugs. BMJ. 1994;309:378. [PMC free article] [PubMed] [Google Scholar]
  • 58.Ricaurte B, Guirguis A, Taylor HC, et al. Simvastatin-amiodarone interaction resulting in rhabdomyolysis, azotemia, and possible hepatotoxicity. Ann Pharmacother. 2006;40:753–757. doi: 10.1345/aph.1G462. [DOI] [PubMed] [Google Scholar]
  • 59.Lucena MI, Andrade RJ, Vicioso L, et al. Prolonged cholestasis after raloxifene and fenofibrate interaction: a case report. World J Gastroenterol. 2006;12:5244–5246. doi: 10.3748/wjg.v12.i32.5244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bryant AE, 3rd, Dreifuss FE. Valproic acid hepatic fatalities. III. U.S. experience since 1986. Neurology. 1996;46:465–469. doi: 10.1212/wnl.46.2.465. [DOI] [PubMed] [Google Scholar]
  • 61.Gopaul S, Farrell K, Abbott F. Effects of age and polytherapy, risk factors of valproic acid (VPA) hepatotoxicity, on the excretion of thiol conjugates of (E)-2,4-diene VPA in people with epilepsy taking VPA. Epilepsia. 2003;44:322–328. doi: 10.1046/j.1528-1157.2003.07202.x. [DOI] [PubMed] [Google Scholar]
  • 62.Siemes H, Nau H, Schultze K, et al. Valproate (VPA) metabolites in various clinical conditions of probable VPA-associated hepatotoxicity. Epilepsia. 1993;34:332–346. doi: 10.1111/j.1528-1157.1993.tb02419.x. [DOI] [PubMed] [Google Scholar]
  • 63.Schmidt LE, Dalhoff K, Poulsen HE. Acute versus chronic alcohol consumption in acetaminophen-induced hepatotoxicity. Hepatology. 2002;35:876–882. doi: 10.1053/jhep.2002.32148. [DOI] [PubMed] [Google Scholar]
  • 64.Whiting-O’Keefe QE, Fye KH, Sack KD. Methotrexate and histologic hepatic abnormalities: a meta-analysis. Am J Med. 1991;90:711–716. [PubMed] [Google Scholar]
  • 65.Malatjalian DA, Ross JB, Williams CN, et al. Methotrexate hepatotoxicity in psoriatics: report of 104 patients from Nova Scotia, with analysis of risks from obesity, diabetes and alcohol consumption during long term follow-up. Can J Gastroenterol. 1996;10:369–375. doi: 10.1155/1996/213596. [DOI] [PubMed] [Google Scholar]
  • 66.Rosenberg P, Urwitz H, Johannesson A, et al. Psoriasis patients with diabetes type 2 are at high risk of developing liver fibrosis during methotrexate treatment. J Hepatol. 2007;46:1111–1118. doi: 10.1016/j.jhep.2007.01.024. [DOI] [PubMed] [Google Scholar]
  • 67.Zachariae H. Have methotrexate-induced liver fibrosis and cirrhosis become rare? A matter for reappraisal of routine liver biopsies. Dermatology. 2005;211:307–308. doi: 10.1159/000088497. [DOI] [PubMed] [Google Scholar]
  • 68.Cross FS, Long MW, Banner AS, et al. Rifampin-isoniazid therapy of alcoholic and nonalcoholic tuberculous patients in a U.S. Public Health Service Cooperative Therapy Trial. Am Rev Respir Dis. 1980;122:349–353. doi: 10.1164/arrd.1980.122.2.349. [DOI] [PubMed] [Google Scholar]
  • 69.Pande JN, Singh SP, Khilnani GC, et al. Risk factors for hepatotoxicity from antituberculosis drugs: a case-control study. Thorax. 1996;51:132–136. doi: 10.1136/thx.51.2.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Dossing M, Wilcke JT, Askgaard DS, et al. Liver injury during antituberculosis treatment: an 11-year study. Tuber Lung Dis. 1996;77:335–340. doi: 10.1016/s0962-8479(96)90098-2. [DOI] [PubMed] [Google Scholar]
  • 71.Schaberg T, Rebhan K, Lode H. Risk factors for side-effects of isoniazid, rifampin and pyrazinamide in patients hospitalized for pulmonary tuberculosis. Eur Respir J. 1996;9:2026–2030. doi: 10.1183/09031936.96.09102026. [DOI] [PubMed] [Google Scholar]
  • 72.Andrade RJ, Lucena MI, Kaplowitz N, et al. Outcome of acute idiosyncratic drug-induced liver injury: long-term follow-up in a hepatotoxicity registry. Hepatology. 2006;44:1581–1588. doi: 10.1002/hep.21424. [DOI] [PubMed] [Google Scholar]
  • 73.Lewis JH. The rational use of potentially hepatotoxic medications in patients with underlying liver disease. Expert Opin Drug Saf. 2002;1:159–172. doi: 10.1517/14740338.1.2.159. [DOI] [PubMed] [Google Scholar]
  • 74.Russo MW, Watkins PB. Are patients with elevated liver tests at increased risk of drug-induced liver injury? Gastroenterology. 2004;126:1477–1480. doi: 10.1053/j.gastro.2004.03.030. [DOI] [PubMed] [Google Scholar]
  • 75.Chalasani N, Aljadhey H, Kesterson J, et al. Patients with elevated liver enzymes are not at higher risk for statin hepatotoxicity. Gastroenterology. 2004;126:1287–1292. doi: 10.1053/j.gastro.2004.02.015. [DOI] [PubMed] [Google Scholar]
  • 76.Ekstedt M, Franzen LE, Mathiesen UL, et al. Statins in nonalcoholic fatty liver disease and chronically elevated liver enzymes: a histopathological follow-up study. J Hepatol. 2007;47:135–141. doi: 10.1016/j.jhep.2007.02.013. [DOI] [PubMed] [Google Scholar]
  • 77.Lewis JH, Mortensen ME, Zweig S, et al. Efficacy and safety of high-dose pravastatin in hypercholesterolemic patients with well-compensated chronic liver disease: results of a prospective, randomized, double-blind, placebo-controlled, multicenter trial. Hepatology. 2007;46:1453–1463. doi: 10.1002/hep.21848. [DOI] [PubMed] [Google Scholar]
  • 78.Khorashadi S, Hasson NK, Cheung RC. Incidence of statin hepatotoxicity in patients with hepatitis C. Clin Gastroenterol Hepatol. 2006;4:902–907. doi: 10.1016/j.cgh.2006.03.014. quiz 806. [DOI] [PubMed] [Google Scholar]
  • 79.Tarantino G, Conca P, Basile V, et al. A prospective study of acute drug-induced liver injury in patients suffering from nonalcoholic fatty liver disease. Hepatol Res. 2007;37:410–415. doi: 10.1111/j.1872-034X.2007.00072.x. [DOI] [PubMed] [Google Scholar]
  • 80.Bjornsson E, Nordlinder H, Olsson R. Clinical characteristics and prognostic markers in disulfiram-induced liver injury. J Hepatol. 2006;44:791–797. doi: 10.1016/j.jhep.2005.12.016. [DOI] [PubMed] [Google Scholar]
  • 81.Wong WM, Wu PC, Yuen MF, et al. Antituberculosis drug-related liver dysfunction in chronic hepatitis B infection. Hepatology. 2000;31:201–206. doi: 10.1002/hep.510310129. [DOI] [PubMed] [Google Scholar]
  • 82.Lee BH, Koh WJ, Choi MS, et al. Inactive hepatitis B surface antigen carrier state and hepatotoxicity during antituberculosis chemotherapy. Chest. 2005;127:1304–1311. doi: 10.1378/chest.127.4.1304. [DOI] [PubMed] [Google Scholar]
  • 83.Patel PA, Voigt MD. Prevalence and interaction of hepatitis B and latent tuberculosis in Vietnamese immigrants to the United States. Am J Gastroenterol. 2002;97:1198–1203. doi: 10.1111/j.1572-0241.2002.05704.x. [DOI] [PubMed] [Google Scholar]
  • 84.Hwang SJ, Wu JC, Lee CN, et al. A prospective clinical study of isoniazid-rifampicin-pyrazinamide-induced liver injury in an area endemic for hepatitis B. J Gastroenterol Hepatol. 1997;12:87–91. doi: 10.1111/j.1440-1746.1997.tb00353.x. [DOI] [PubMed] [Google Scholar]
  • 85.Brunetto MR, Oliveri F, Coco B, et al. Outcome of anti-HBe positive chronic hepatitis B in alpha-interferon treated and untreated patients: a long term cohort study. J Hepatol. 2002;36:263–270. doi: 10.1016/s0168-8278(01)00266-5. [DOI] [PubMed] [Google Scholar]
  • 86.Ungo JR, Jones D, Ashkin D, et al. Antituberculosis drug-induced hepatotoxicity. The role of hepatitis C virus and the human immunodeficiency virus. Am J Respir Crit Care Med. 1998;157:1871–1876. doi: 10.1164/ajrccm.157.6.9711039. [DOI] [PubMed] [Google Scholar]
  • 87.den Brinker M, Wit FW, Wertheim-van Dillen PM, et al. Hepatitis B and C virus co-infection and the risk for hepatotoxicity of highly active antiretroviral therapy in HIV-1 infection. AIDS. 2000;14:2895–2902. doi: 10.1097/00002030-200012220-00011. [DOI] [PubMed] [Google Scholar]
  • 88.Bonfanti P, Landonio S, Ricci E, et al. Risk factors for hepatotoxicity in patients treated with highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2001;27:316–318. doi: 10.1097/00126334-200107010-00017. [DOI] [PubMed] [Google Scholar]
  • 89.Bonacini M. Liver injury during highly active antiretroviral therapy: the effect of hepatitis C coinfection. Clin Infect Dis. 2004;38 Suppl 2:S104–S108. doi: 10.1086/381453. [DOI] [PubMed] [Google Scholar]
  • 90.Kramer JR, Giordano TP, Souchek J, et al. Hepatitis C coinfection increases the risk of fulminant hepatic failure in patients with HIV in the HAART era. J Hepatol. 2005;42:309–314. doi: 10.1016/j.jhep.2004.11.017. [DOI] [PubMed] [Google Scholar]
  • 91.Sulkowski MS, Thomas DL, Mehta SH, et al. Hepatotoxicity associated with nevirapine or efavirenz-containing antiretroviral therapy: role of hepatitis C and B infections. Hepatology. 2002;35:182–189. doi: 10.1053/jhep.2002.30319. [DOI] [PubMed] [Google Scholar]
  • 92.Labarga P, Soriano V, Vispo ME, et al. Hepatotoxicity of antiretroviral drugs is reduced after successful treatment of chronic hepatitis C in HIV-infected patients. J Infect Dis. 2007;196:670–676. doi: 10.1086/520092. [DOI] [PubMed] [Google Scholar]
  • 93.Ito H, Yoshioka K, Ukai K, et al. The fluctuations of viral load and serum alanine aminotransferase levels in chronic hepatitis C. Hepatol Res. 2004;30:11–17. doi: 10.1016/j.hepres.2004.06.002. [DOI] [PubMed] [Google Scholar]
  • 94.Jain MK, Parekh NK, Hester J, et al. Aminotransferase elevation in HIV/hepatitis B virus co-infected patients treated with two active hepatitis B virus drugs. AIDS Patient Care STDS. 2006;20:817–822. doi: 10.1089/apc.2006.20.817. [DOI] [PubMed] [Google Scholar]
  • 95.Ujfalussy I, Koo E, Sesztak M, et al. Termination of disease-modifying antirheumatic drugs in rheumatoid arthritis and in psoriatic arthritis. A comparative study of 270 cases. Z Rheumatol. 2003;62:155–160. doi: 10.1007/s00393-003-0458-2. [DOI] [PubMed] [Google Scholar]
  • 96.Tilling L, Townsend S, David J. Methotrexate and hepatic toxicity in rheumatoid arthritis and psoriatic arthritis. Clin Drug Investig. 2006;26:55–62. doi: 10.2165/00044011-200626020-00001. [DOI] [PubMed] [Google Scholar]
  • 97.Bachs L, Pares A, Elena M, et al. Effects of long-term rifampicin administration in primary biliary cirrhosis. Gastroenterology. 1992;102:2077–2080. doi: 10.1016/0016-5085(92)90335-v. [DOI] [PubMed] [Google Scholar]
  • 98.Prince MI, Burt AD, Jones DE. Hepatitis and liver dysfunction with rifampicin therapy for pruritus in primary biliary cirrhosis. Gut. 2002;50:436–439. doi: 10.1136/gut.50.3.436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Cascio A, Scarlata F, Giordano S, et al. Treatment of human brucellosis with rifampin plus minocycline. J Chemother. 2003;15:248–252. doi: 10.1179/joc.2003.15.3.248. [DOI] [PubMed] [Google Scholar]
  • 100.Uetrecht J. Idiosyncratic drug reactions: past, present, and future. Chem Res Toxicol. 2008;21:84–92. doi: 10.1021/tx700186p. [DOI] [PubMed] [Google Scholar]
  • 101.Donaldson PT, Bhatnagar P, Graham J, et al. Susceptibility to drug induced liver injury determined by HLA class II genotype (abstr) Hepatology. 2008;48 S1:464A. [Google Scholar]
  • 102.Daly AK, Donaldson PT, Bhatnagar P, et al. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet. 2009;41:816–821. doi: 10.1038/ng.379. [DOI] [PubMed] [Google Scholar]
  • 103.Day CP, Stewart JD, Horvath R, et al. Common POLG genetic variants increase the risk of sodium valproate induced liver injury and failure. Hepatology. 2009;50:383A. [Google Scholar]
  • 104.Scordo MG, Aklillu E, Yasar U, et al. Genetic polymorphism of cytochrome P450 2C9 in a Caucasian and a black American population. Br J Clin Pharmacol. 2001;52:447–450. doi: 10.1046/j.0306-5251.2001.01460.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Morin S, Loriot MA, Poirier JM, et al. Is diclogenac a valuable CYP2C9 probe in humans? Eu J Clin Pharmacol. 2001;56:793–797. doi: 10.1007/s002280000240. [DOI] [PubMed] [Google Scholar]
  • 106.Aithal GP, Day CP, Leathart JB, et al. Relationship of polymorphism in CYP2C9 to genetic susceptibility to diclofenac-induced hepatitis. Pharmacogenetics. 2000;10:511–518. doi: 10.1097/00008571-200008000-00004. [DOI] [PubMed] [Google Scholar]
  • 107.Yasar U, Eliasson E, Forslund-Bergengren C, et al. The role of CYP2C9 genotype in the metabolism of CYP2C9 genotype in the metabolism of diclofenac in vivo and in vitro. Eur J Clin Pharmacol. 2001;57:729–735. doi: 10.1007/s00228-001-0376-7. [DOI] [PubMed] [Google Scholar]
  • 108.Kirchheiner J, Brockmoller J. Clinical consequences of cytochrome P450 2C9 polymorphisms. Clin Pharmacol Ther. 2005;77:1–16. doi: 10.1016/j.clpt.2004.08.009. [DOI] [PubMed] [Google Scholar]
  • 109.Horsmans Y, Lannes D, Pessayre D, et al. Possible association between poor metabolism of mephenytoin and hepatotoxicity caused by Atrium, a fixed combination preparation containing phenobarbital, febarbamate, and difebarbamate. J Hepatol. 1994;21:1075–1079. doi: 10.1016/s0168-8278(05)80620-8. [DOI] [PubMed] [Google Scholar]
  • 110.Kumashiro R, Kuboto T, Koga Y, et al. Association of troglitazone-induced liver injury with mutation of the cytochrome P4502C19 gene. Hepatol Res. 2003;26:337–342. doi: 10.1016/s1386-6346(03)00165-7. [DOI] [PubMed] [Google Scholar]
  • 111.Pachkoria K, Lucena MI, Ruiz-Cabello F, et al. Genetic polymorphisms of CYP2C9 and CYP2C19 are not related to drug-induced idiosyncratic liver injury (DILI) Br J Pharmacol. 2007;150:808–815. doi: 10.1038/sj.bjp.0707122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Huang YS, Chern HD, Su WJ, et al. Cytochrome P450 2E1 genotype and the susceptibility to antituberculosis drug-induced hepatitis. Hepatology. 2003;37:924–930. doi: 10.1053/jhep.2003.50144. [DOI] [PubMed] [Google Scholar]
  • 113.Vuilleumier N, Rossier MF, Chiape A, et al. CYP2E1 genotype and isoniazid-induced hepatotoxicity in patients treated for latent tuberculosis. Eur J Clin Pharmacol. 2006;62:423–429. doi: 10.1007/s00228-006-0111-5. [DOI] [PubMed] [Google Scholar]
  • 114.Sun F, Chen Y, Xiang Y, et al. Drug-metabolizing enzyme polymorphisms and predisposition to anti-tuberculosis drug-induced liver injury: a meta-analysis. Int J Tuberc Lung Dis. 2008;12:994–1002. [PubMed] [Google Scholar]
  • 115.Rieder MJ, Shear NH, Kanee A, et al. Prominence of slow acetylator phenotype among patients with sulfonamide hypersensitivity reactions. Clin Pharmacol Ther. 1991;49:13–17. doi: 10.1038/clpt.1991.3. [DOI] [PubMed] [Google Scholar]
  • 116.Mitchell JR, Thorgeirsson UP, Black M, et al. Increased incidence of isoniazid hepatitis in rapid acetylators: possible relation to hydranize metabolites. Clin Pharmacol Ther. 1975;18:70–79. doi: 10.1002/cpt197518170. [DOI] [PubMed] [Google Scholar]
  • 117.Yamamoto T, Suou T, Hirayama C. Elevated serum aminotransferase induced by isoniazid in relation to isoniazid acetylator phenotype. Hepatology. 1986;6:295–298. doi: 10.1002/hep.1840060223. [DOI] [PubMed] [Google Scholar]
  • 118.Huang YS, Chern HD, Su WJ, et al. Polymorphism of the N-acetyltransferase 2 gene as a susceptibility risk factor for anti-tuberculosis drug-induced hepatitis. Hepatology. 2002;35:883–889. doi: 10.1053/jhep.2002.32102. [DOI] [PubMed] [Google Scholar]
  • 119.Shimizu Y, Dobashi K, Mita Y, et al. DNA microarray genotyping of N-acetyltransferase 2 polymorphism using carbodiimide as the linker for assessment of isoniazid hepatotoxicity. Tuberculosis (Edinb) 2006;86:374–381. doi: 10.1016/j.tube.2005.09.002. [DOI] [PubMed] [Google Scholar]
  • 120.Huang YS. Genetic polymorphisms of drug-metabolizing enzymes and the susceptibility to antituberculosis drug-induced liver injury. Expert Opin Drug Metab Toxicol. 2007;3:1–8. doi: 10.1517/17425255.3.1.1. [DOI] [PubMed] [Google Scholar]
  • 121.Lucena MI, Andrade RJ, Martinez C, et al. Glutathione S-transferase m1 and t1 null genotypes increase susceptibility to idiosyncratic drug-induced liver injury. Hepatology. 2008;48:588–596. doi: 10.1002/hep.22370. [DOI] [PubMed] [Google Scholar]
  • 122.Huang YS, Su WJ, Huang YH, et al. Genetic polymorphisms of manganese superoxide dismutase, NAD(P)H: quinone oxidoreductase, glutathione S-transferase M1 and T1, and the susceptibility to drug-induced liver injury. J Hepatol. 2007;47:128–134. doi: 10.1016/j.jhep.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 123.Daly AK, Aithal GP, Leathart JB, et al. Genetic susceptibility to diclofenac-induced hepatotoxicity: contribution of UGT2B7, CYP2C8, and ABCC2 genotypes. Gastroenterology. 2007;132:272–281. doi: 10.1053/j.gastro.2006.11.023. [DOI] [PubMed] [Google Scholar]
  • 124.Boelsterli UA. Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity. Toxicol Appl Pharmacol. 2003;192:307–322. doi: 10.1016/s0041-008x(03)00368-5. [DOI] [PubMed] [Google Scholar]
  • 125.Aithal GP, Ramsay L, Daly AK, et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology. 2004;39:1430–1440. doi: 10.1002/hep.20205. [DOI] [PubMed] [Google Scholar]
  • 126.Aithal GP, Day CP, Leathart JB, et al. Relationship of polymorphism in CYP2C9 to genetic susceptibility to diclofenac-induced hepatitis. Pharmacogenetics. 2000;10:511–518. doi: 10.1097/00008571-200008000-00004. [DOI] [PubMed] [Google Scholar]
  • 127.Ho RH, Kim RB. Transporters and drug therapy: implications for drug disposition and disease. Clin Pharmacol Ther. 2005;78:260–277. doi: 10.1016/j.clpt.2005.05.011. [DOI] [PubMed] [Google Scholar]
  • 128.Pauli-Magnus C, Meier PJ. Hepatobiliary transporters and drug-induced cholestasis. Hepatology. 2006;44:778–787. doi: 10.1002/hep.21359. [DOI] [PubMed] [Google Scholar]
  • 129.Pauli-Magnus C, Stieger B, Meier Y, et al. Enterohepatic transport of bile salts and genetics of cholestasis. J Hepatol. 2005;43:342–357. doi: 10.1016/j.jhep.2005.03.017. [DOI] [PubMed] [Google Scholar]
  • 130.Vavricka SR, Van Montfoort J, Ha HR, et al. Interactions of rifamycin SV and rifampicin with organic anion uptake systems of human liver. Hepatology. 2002;36:164–172. doi: 10.1053/jhep.2002.34133. [DOI] [PubMed] [Google Scholar]
  • 131.Bolder U, Trang NV, Hagey LR, et al. Sulindac is excreted into bile by a canalicular bile salt pump and undergoes a cholehepatic circulation in rats. Gastroenterology. 1999;117:962–971. doi: 10.1016/s0016-5085(99)70356-2. [DOI] [PubMed] [Google Scholar]
  • 132.Lakehal F, Dansette PM, Becquemont L, et al. Indirect cytotoxicity of flucloxacillin toward human biliary epithelium via metabolite formation in hepatocytes. Chem Res Toxicol. 2001;14:694–701. doi: 10.1021/tx0002435. [DOI] [PubMed] [Google Scholar]
  • 133.Iverson SL, Uetrecht JP. Identification of a reactive metabolite of terbinafine: insights into terbinafine-induced hepatotoxicity. Chem Res Toxicol. 2001;14:175–181. doi: 10.1021/tx0002029. [DOI] [PubMed] [Google Scholar]
  • 134.Fattinger K, Funk C, Pantze M, et al. The endothelin antagonist bosentan inhibits the canalicular bile salt export pump: a potential mechanism for hepatic adverse reactions. Clin Pharmacol Ther. 2001;69:223–231. doi: 10.1067/mcp.2001.114667. [DOI] [PubMed] [Google Scholar]
  • 135.Leslie EM, Watkins PB, Kim RB, et al. Differential inhibition of rat and human Na+dependent taurocholate cotransporting polypeptide (NTCP/SLC10A1)by bosentan: a mechanism for species differences in hepatotoxicity. J Pharmacol Exp Ther. 2007;321:1170–1178. doi: 10.1124/jpet.106.119073. [DOI] [PubMed] [Google Scholar]
  • 136.Meier Y, Pauli-Magnus C, Zanger UM, et al. Interindividual variability of canalicular ATP-binding-cassette (ABC)-transporter expression in human liver. Hepatology. 2006;44:62–74. doi: 10.1002/hep.21214. [DOI] [PubMed] [Google Scholar]
  • 137.Strautnieks SS, Bull LN, Knisely AS, et al. A gene encoding a liver-specific ABC transporter is mutated in progressive familial intrahepatic cholestasis. Nat Genet. 1998;20:233–238. doi: 10.1038/3034. [DOI] [PubMed] [Google Scholar]
  • 138.Pauli-Magnus C, Lang T, Meier Y, et al. Sequence analysis of bile salt export pump (ABCB11) and multidrug resistance p-glycoprotein 3 (ABCB4, MDR3) in patients with intrahepatic cholestasis of pregnancy. Pharmacogenetics. 2004;14:91–102. doi: 10.1097/00008571-200402000-00003. [DOI] [PubMed] [Google Scholar]
  • 139.Leevy CB, Koneru B, Klein KM. Recurrent familial prolonged intrahepatic cholestasis of pregnancy associated with chronic liver disease. Gastroenterology. 1997;113:966–972. doi: 10.1016/s0016-5085(97)70193-8. [DOI] [PubMed] [Google Scholar]
  • 140.Meier Y, Zodan T, Lang C, et al. Increased susceptibility for intrahepatic cholestasis of pregnancy and contraceptive-induced cholestasis in carriers of the 1331T>C polymorphism in the bile salt export pump. World J Gastroenterol. 2008;14:38–45. doi: 10.3748/wjg.14.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Noe J, Kullak-Ublick GA, Jochum W, et al. Impaired expression and function of the bile salt export pump due to three novel ABCB11 mutations in intrahepatic cholestasis. J Hepatol. 2005;43:536–543. doi: 10.1016/j.jhep.2005.05.020. [DOI] [PubMed] [Google Scholar]
  • 142.Lang C, Meier Y, Stieger B, et al. Mutations and polymorphisms in the bile salt export pump and the multidrug resistance protein 3 associated with drug-induced liver injury. Pharmacogenet Genomics. 2007;17:47–60. doi: 10.1097/01.fpc.0000230418.28091.76. [DOI] [PubMed] [Google Scholar]
  • 143.Ibanez L, Perez E, Vidal X, et al. Prospective surveillance of acute serious liver disease unrelated to infectious, obstructive, or metabolic diseases: epidemiological and clinical features, and exposure to drugs. J Hepatol. 2002;37:592–600. doi: 10.1016/s0168-8278(02)00231-3. [DOI] [PubMed] [Google Scholar]
  • 144.Kindmark A, Jawaid A, Harbron CG, et al. Genome-wide pharmacogenetic investigation of a hepatic adverse event without clinical signs of immunopathology suggests an underlying immune pathogenesis. Pharmacogenomics J. 2008;8:186–195. doi: 10.1038/sj.tpj.6500458. [DOI] [PubMed] [Google Scholar]
  • 145.Bjornsson E, Kalaitzakis E, Olsson R. The impact of eosinophilia and hepatic necrosis on prognosis in patients with drug-induced liver injury. Aliment Pharmcol Ther. 2007;25:1411–1421. doi: 10.1111/j.1365-2036.2007.03330.x. [DOI] [PubMed] [Google Scholar]
  • 146.Pichler WJ. Pharmacological interaction of drugs with antigen-specific immune receptors: the p-i concept. Curr Opin Allergy Clin Immunol. 2002;2:301–305. doi: 10.1097/00130832-200208000-00003. [DOI] [PubMed] [Google Scholar]
  • 147.Mallal S, Nolan D, Witt C, et al. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet. 2002;359:727–732. doi: 10.1016/s0140-6736(02)07873-x. [DOI] [PubMed] [Google Scholar]
  • 148.Man CB, Kwan P, Baum L, et al. Association between HLA-B* 1502 allele and antiepileptic drug-induced cutaneous reactions in Han Chinese. Epilepsia. 2007;48:1015–1018. doi: 10.1111/j.1528-1167.2007.01022.x. [DOI] [PubMed] [Google Scholar]
  • 149.Hautekeete ML, Horsmans Y, Van Waeyenberge C, et al. HLA association of amoxicillin-clavulanate-induced hepatitis. Gastroenterology. 1999;117:1181–1186. doi: 10.1016/s0016-5085(99)70404-x. [DOI] [PubMed] [Google Scholar]
  • 150.Sharma SK, Balamurugan A, Saha PK, et al. Evaluation of clinical and immunogenetic risk factors for the development of hepatotoxicity during antituberculosis treatment. Am J Respir Crit Care Med. 2002;166:916–919. doi: 10.1164/rccm.2108091. [DOI] [PubMed] [Google Scholar]
  • 151.Donaldson PT. Flucloxacillin-induced liver injury, the extended MHC 57.1 haplotype as a major risk factor. Hepatology. 2008;48:396A–397A. [Google Scholar]
  • 152.Mallal S, Phillips E, Carosi G, et al. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med. 2008;358:568–579. doi: 10.1056/NEJMoa0706135. [DOI] [PubMed] [Google Scholar]
  • 153.O’Donohue J, Oien KA, Donaldson P, et al. Co-amoxiclav jaundice: clinical and histological features and HLA class II association. Gut. 2000;47:717–720. doi: 10.1136/gut.47.5.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Andrade RJ, Lucena MI, Alonso A, et al. HLA class II genotype influences the type of liver injury in drug-induced idiosyncratic liver disease. Hepatology. 2004;39:1603–1612. doi: 10.1002/hep.20215. [DOI] [PubMed] [Google Scholar]
  • 155.Berson A, Freneaux E, Larrey D, et al. Possible role of HLA in hepatotoxicity. An exploratory study in 71 patients with drug-induced idiosyncratic hepatitis. J Hepatol. 1994;20:336–342. doi: 10.1016/s0168-8278(94)80004-9. [DOI] [PubMed] [Google Scholar]
  • 156.Pachkoria K, Lucena MI, Crespo E, et al. Analysis of IL-10, IL-4 and TNF-alpha polymorphisms in drug-induced liver injury (DILI) and its outcome. J Hepatol. 2008;49:107–114. doi: 10.1016/j.jhep.2008.03.017. [DOI] [PubMed] [Google Scholar]

RESOURCES