Skip to main content
Full access
Perspectives
Published Online: 1 April 2014

Antidepressant-Induced Liver Injury: A Review for Clinicians

Abstract

There are no strategies yet for preventing antidepressant-induced adverse hepatic events, but early detection by aminotransferase surveillance is essential, as drug-induced liver injury from antidepressants, although rare, may be irreversible in at-risk patients. Age and polypharmacy are two of several critical factors to consider when prescribing antidepressants.

Abstract

Objective

Antidepressant drugs can cause drug-induced liver injury (DILI). The authors review clinical data relevant to antidepressant-induced liver injury and provide recommendations for clinical practice.

Method

A PubMed search was conducted for publications from 1965 onward related to antidepressant-induced liver injury. The search terms were “liver injury,” “liver failure,” “DILI,” “hepatitis,” “hepatotoxicity,” “cholestasis,” and “aminotransferase,” cross-referenced with “antidepressant.”

Results

Although data on antidepressant-induced liver injury are scarce, 0.5%−3% of patients treated with antidepressants may develop asymptomatic mild elevation of serum aminotransferase levels. All antidepressants can induce hepatotoxicity, especially in elderly patients and those with polypharmacy. Liver damage is in most cases idiosyncratic and unpredictable, and it is generally unrelated to drug dosage. The interval between treatment initiation and onset of liver injury is generally between several days and 6 months. Life-threatening antidepressant-induced liver injury has been described involving fulminant liver failure or death. The underlying lesions are often of the hepatocellular type and less frequently of the cholestatic and mixed types. The antidepressants associated with greater risks of hepatotoxicity are iproniazid, nefazodone, phenelzine, imipramine, amitriptyline, duloxetine, bupropion, trazodone, tianeptine, and agomelatine. The antidepressants that seem to have the least potential for hepatotoxicity are citalopram, escitalopram, paroxetine, and fluvoxamine. Cross-toxicity has been described, mainly for tricyclic and tetracyclic antidepressants.

Conclusions

Although an infrequent event, DILI from antidepressant drugs may be irreversible, and clinicians should be aware of it. Aminotransferase surveillance is the most useful tool for detecting DILI, and prompt discontinuation of the drug responsible is essential. The results of antidepressant liver toxicity in all phases of clinical trials should be available and published. Further research is needed before any new and rigorously founded recommendations can be made.
Drug-induced liver injury (DILI), the fourth leading cause of liver damage in Western countries, is a matter of concern in the context of increasing drug availability and prescription (1). DILI is the most frequent cause of market withdrawal of a drug and rejection of applications for a marketing license in the United States (2). The U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) proposed guidelines for DILI in 2009 and 2010, respectively (3, 4).
The incidence of DILI is between 1 per 10,000 and 1 per 100,000 patient-years (5, 6), and therefore the first cases are generally described after commercialization of the drug, when a large number of patients have been exposed. In such situations, instances of DILI are often underdeclared, thus underestimating the true frequency (7, 8).
DILI can be classified as hepatocellular, cholestatic, or mixed, depending on the underlying liver injury. Hepatocellular injury is characterized by abnormally high serum alanine aminotransferase (ALT) titers with a small or no increase in alkaline phosphatase (ALP) titers; an associated high serum bilirubin level, found in cases of severe hepatocellular damage, is a marker of poor prognosis (9). Cholestatic liver injury is characterized by high serum ALP titers associated with only slightly higher than normal ALT levels; serum bilirubin concentrations may also be high. In cases of mixed injury, both ALT and ALP levels are abnormally high. Slightly higher than normal serum aminotransferase titers (less than 3 times the upper limit of normal; <3×ULN) are found in 1%−5% of the general population (10). Therefore, ALT values >3×ULN or ALP values >2×ULN are indicative of DILI. These thresholds, however, are sensitive but not specific markers (11). To avoid unnecessary withdrawal of drugs erroneously identified to be hepatotoxic, a new threshold of 5×ULN for ALT has been proposed (9).
Two pathophysiological types of DILI have been identified. The most common type is idiosyncratic, dose independent, and unpredictable (12). It is the consequence either of immune-mediated liver damage (immuno-allergic idiosyncratic DILI) or of direct cellular injury (metabolic idiosyncratic DILI) (13). Intrinsic DILI, related to drug accumulation, has also been described; it is dose dependent and predictable and is generally observed during preclinical and clinical trials, leading to early drug withdrawal.
Our aim in this study was to review clinical data on antidepressant-induced liver injury and to provide recommendations for clinical practice against a background of increasing numbers of antidepressant prescriptions, increasing polypharmacy, a significant frequency of liver injury associated with antidepressants, and the potential risks of antidepressant-induced liver injury.

Method

We conducted a PubMed search of articles published from 1965 through September 2013, using the search terms “liver injury,” “liver failure,” “DILI,” “hepatitis,” “hepatotoxicity,” “cholestasis,” and “aminotransferase,” cross-referenced with “antidepressant.” Case reports, letters, original articles, and reviews, in English and other languages, were identified. We also noted all relevant articles in the reference lists. We carefully analyzed a total of 378 articles, from which 158 (88 case reports, 38 original papers, and 32 reviews) were selected, by author consensus, as being suitable for review based on exclusion of other causes of liver injury and the use of validated scales for drug imputability assessment.
The available data on antidepressant-induced hepatic toxicity are mostly from reported cases and to a lesser extent from results of clinical trials and other studies, especially for the most recent drugs (nefazodone, venlafaxine, duloxetine, bupropion, and agomelatine). It is therefore difficult to draw conclusions about the prevalence and severity of antidepressant-induced liver injury. In our review, the potential for causing DILI was assessed for each antidepressant on the basis of five criteria: total number of published cases of antidepressant-induced liver injury, number of published cases of severe liver injury leading to death or liver transplantation, significant abnormalities of liver function tests in clinical trials, the existence of studies describing cases of antidepressant-induced liver injury, and hepatotoxicity demonstrated by published experimental studies.

Results

Epidemiology

Asymptomatic mild abnormal liver function is detected in 0.5%−1% of patients treated with second-generation antidepressants such as selective serotonin reuptake inhibitors (SSRIs) (1416) and serotonin-norepinephrine reuptake inhibitors (SNRIs) (17, 18), and up to 3% of patients treated with monoamine oxidase (MAO) inhibitors or tricyclic and tetracyclic antidepressants (19, 20). Results of clinical trials evaluating liver function during antidepressant treatment are available for duloxetine, venlafaxine, and agomelatine. For the other antidepressants, the estimated rates of asymptomatic mild abnormal liver function are based on data reported by the manufacturer or on empiric evidence. The incidence of DILI is estimated to be 4 per 100,000 patient-years for tricyclic/tetracyclic antidepressants (5, 14). Overall, the incidence of antidepressant-induced liver toxicity requiring hospitalization is only 1.28–4 cases per 100,000 patient-years, except for nefazodone, for which the incidence can be estimated to be 29 cases per 100,000 patient-years (6, 14).

Risk Factors

The risk factors for antidepressant-induced liver injury are poorly known. In particular, no gene polymorphisms associated with greater susceptibility to idiosyncratic DILI from antidepressant agents have been described.
Coprescription of more than one drug targeting the same cytochrome P450 (CYP450) isoenzyme pathway may increase the risk of DILI (Table 1). The CYP450 enzyme system is responsible for phase I oxidative reactions involving drugs (21). Some antidepressants can inhibit or induce CYP450 enzyme activity, thus affecting the serum concentrations of antidepressants or their metabolites and thereby potentially increasing the risk of hepatic toxicity. Furthermore, other therapeutic compounds may compete with antidepressants for the same CYP450 metabolic pathway. Caution is required when using such combinations of drugs because of the potential for hepatotoxic reactions. Case reports describing possible drug-drug interactions involving antidepressants are summarized in Table 1. It should be noted that cross-toxicity has been described for tricyclic/tetracyclic antidepressants (40, 41) and, to a lesser extent, for SSRIs (fluvoxamine and citalopram) (42). In the context of increasing polypharmacy in major depressive disorders, drug-drug interactions must be considered for possible liver toxicity.
TABLE 1. Potential Antidepressant Drug-Drug Interactionsa
InteractionCYP450 MetabolismCYP450 InhibitionPotential Enzymatic MechanismReported Liver ToxicityReferences
Moclobemide-fluoxetineMoclobemide: 2C19; fluoxetine: 2D6, 2CMoclobemide: 2D6, 2C9, 1A2; fluoxetine: 2D6, 2C9/19, 3A4InhibitionDeath: 121, 22
Amitriptyline-diazepamAmitriptyline: 2C19, 3A4, 1A2, 2C9, 2D6; diazepam: 2C19, 3A4Amitriptyline: none; diazepam: 3A4Competition and inhibitionDeath: 12325
Venlafaxine-trazodoneVenlafaxine: 2D6, 3A4; trazodone: 2D6Venlafaxine: 2D6; trazodone: 3A4, 1A2Competition and inhibitionLiver transplant: 1; spontaneous resolution: 121, 2628
Duloxetine-mirtazapineDuloxetine: 1A2, 2D6; mirtazapine: 1A2, 2D6Duloxetine: none; mirtazapine: noneCompetitionDeath: 1; spontaneous resolution: 121, 29, 30
Duloxetine-fluoxetineDuloxetine: 1A2, 2D6; fluoxetine: 2D6, 2CDuloxetine: none; fluoxetine: 2D6, 2C9/19, 3A4Competition and inhibitionSevere hepatocellular injury with spontaneous resolution: 121, 31
Duloxetine-clorazepateDuloxetine: 1A2, 2D6; clorazepate: 3A4Duloxetine: none; clorazepate: noneCompetition and inhibitionSevere hepatocellular injury with spontaneous resolution: 121, 31
Duloxetine-trazodoneDuloxetine: 1A2, 2D6; trazodone: 2D6Duloxetine: none; trazodone: 3A4, 1A2Competition and inhibitionFulminant hepatic failure with spontaneous resolution: 121, 32
Sertraline-donepezilSertraline: 2D6, 3A4, 2C9/19; donepezil: 2D6, 3A4, 1A2Sertraline: 2D6, 3A4, 1A2; donepezil: noneCompetitionFulminant hepatic failure with spontaneous resolution: 121, 33, 34
Sertraline-diazepamSertraline: 2D6, 3A4, 2C9/19; diazepam: 2C19, 3A4Sertraline; 2D6, 3A4, 1A2; diazepam: 3A4Competition and inhibitionDeath: 121, 24, 35
Paroxetine-trazodoneParoxetine: 2D6; trazodone: 2D6Paroxetine: 2D6; trazodone: 3A4, 1A2Competition and inhibitionSpontaneous resolution: 121, 36
Nefazodone-clonazepamNefazodone: 3A4, 2D6; clonazepam: 3A4Nefazodone: 3A4; clonazepam: noneCompetitionSpontaneous resolution: 137, 38
Trazodone-thioridazineTrazodone: 2D6; thioridazine: noneTrazodone: 3A4, 1A2; thioridazine: 2D6InhibitionDeath: 121, 28, 39
a
CYP450=cytochrome P450.
Antidepressant-induced liver injury is generally considered to be dose independent. However, cases of hepatic toxicity following antidepressant dosage escalation have been reported for duloxetine, nefazodone, mianserin, and sertraline (30, 38, 43, 44) (Table 2); in one case report, reduction of the daily dose was followed by reversal of mianserin-associated liver injury (43) (Table 2). In clinical trials of agomelatine, aminotransferase values >3×ULN were observed in 1.4% of patients treated with 25 mg/day and 2.5% of those treated with 50 mg/day, compared with 0.6% of the placebo group (147, 151). Although idiosyncratic drug reactions tend to be dose independent, a retrospective analysis of reported cases of DILI in the United States found that drugs with daily doses ≥50 mg were associated with a higher risk of liver failure, liver failure leading to death, and liver transplantation than those with lower doses (12). Conversely, drugs administered at daily doses ≤10 mg rarely caused DILI (12). Moreover, a recent study indicated that the association of high daily doses and high lipophilicity is predictive of a risk of liver injury (152).
TABLE 2. Hepatotoxicity of the Main Antidepressant Drugsa
Antidepressant Class and AgentEpidemiologyType of LesionMechanismLatencyCoprescriptionsOutcomeOtherRisk of Liver Injury
MAO inhibitors
IproniazidAbnormal LFT: 3% of treated patients (20)HepatocellularMetabolic; immuno-allergic (autoantibodies: antimitochondrial type 6) (45)4 days–6 monthsFluoxetineDeath or LT in 20% of patients developing jaundice (4548)Hepatic focal necrosis in rats (100–400 mg/kg) (49); withdrawn in 1978 due to hepatotoxicity (available in France until 1999)++++
PhenelzineAbnormal LFT: 3% of treated patients (20)Cholestatic cirrhosisMetabolic2–4 months LT: 2; cirrhosis: 1 (50, 51) +++
MoclobemideAbnormal LFT: 3% of treated patients (20)Cholestatic; hepatocellularImmuno-allergic1–3 weeksFluoxetineDeath: 1; recovery: 1 (22, 52) ++
Tricyclic and tetracyclic drugs
Imipramine, desipramineCholestatic jaundice: 0.5%–1% of treated patients (53); no DILI in clinical trials (54, 55); DILI: 4/100,000 patient-years (5, 14)Hepatocellular; cholestatic; VBDSImmuno-allergic1 week–5 months Death: 1; LT: 1; VBDS: 1; recovery: 4; (53, 5660)Cross-toxicity between tricyclic/tetracyclic drugs (6, 40, 41)+++
AmitriptylineAbnormal LFT: 3% of treated patients (19)Cholestatic; hepatocellular; VBDSImmuno-allergic1–8 months Deaths: 3; VBDS: 1; recovery: 4; (25, 6165)Cross-toxicity between tricyclic/tetracyclic drugs (6, 40, 41)+++
MaprotilineUnknownHepatocellular; cholestaticMetabolic25 days–4 years Recovery: 5 (6670) +
ClomipramineUnknownHepatocellularImmuno-allergic∼1 month Recovery: 2 (40, 71)Cross-toxicity between tricyclic/tetracyclic drugs (6, 40)+
Serotonin-norepinephrine reuptake inhibitors
VenlafaxineALT >3×ULN: 0.4% of treated patients (17)Hepatocellular; cholestaticMetabolic; immuno-allergic10 days–6 monthsTrazodoneLT: 1; recovery: 9 (26, 27, 7278)Non-dose-dependent abnormal LFT in clinical trials (17); 1 case of DILI following venlafaxine dosage escalation (75)++
DuloxetineALT >3×ULN: 1.1% of treated patients (placebo: 0.3%) (18); ALT >5×ULN: 0.6% of treated patients (placebo: 0%) (79); ALT increase leading to discontinuation in 0.4% of treated patients (80); Hy’s law: 0 (79); DILI: 26.2/100,000 patient-years (80, 81)Hepatocellular; cholestatic; mixedMetabolic; immuno-allergic2 weeks–3 monthsMirtazapine and fluoxetineDeath: 1; recovery: 12 (2932, 8082)Dose-dependent abnormal LFT in clinical trials (80); 1 case of DILI following duloxetine dosage escalation (32); risk factor for DILI: preexisting chronic liver disease (18, 8183) (cautious use in the U.S. [84] and contraindication in Europe [85])+++
Selective serotonin reuptake inhibitors
SertralineALT >3×ULN: 0.5%–1.3% of treated patients (14); DILI: 1.28/100,000 patient-years (14); no DILI in clinical trials (86, 87)Hepatocellular; cholestatic; mixedImmuno-allergic; metabolic2 weeks–6 months Death: 1; recovery: 10 (34, 35, 44, 8892) ++
ParoxetineALT >3×ULN: 1% of treated patients (16); no DILI in clinical trials (93, 94)Hepatocellular; cholestatic; chronic hepatitisMetabolic1 day–10 months Complete recovery: 12 (36, 9599) +
FluoxetineALT >3×ULN: 0.5% of treated patients (15); no DILI in clinical trials (100, 101)Hepatocellular; mixed; cholestatic; chronic hepatitisMetabolic2.5 months–1 year Chronic hepatitis: 1; recovery: 5; (102106) +
Citalopram, escitalopramNo difference in LFT versus placebo (107, 108)HepatocellularMetabolic4 days–8 weeks Recovery: 4 (109112) +
FluvoxamineUnknownHepatocellularMetabolic9 days Recovery: 1 (42) +
Other antidepressants
NefazodoneDILI: 28.96/100,000 patient-years (14); severe DILI: 81.3% of cases (113); death or LT: 1/250,000–300,000 patient-years (6); no DILI in clinical trials (114)HepatocellularMetabolic4 weeks–8 months Deaths: 2; LT: 3; severe liver dysfunction: 2; recovery: 2 (14, 38, 115119)Discontinuation of branded formulation in 2003, but generic formulations available; FDA black box warning regarding hepatotoxicity++++
TrazodoneUnknownHepatocellular; cholestaticImmuno-allergic4 days–18 monthsThioridazineDeath: 1; chronic hepatitis: 1; recovery: 5 (39, 120126) ++
BupropionALT >3×ULN: 0.1%–1% of treated patients (127); no DILI in clinical trials (128)Hepatocellular; cholestaticImmuno-allergic20 days–6 monthsParoxetineDeaths: 2; recovery: 5 (127, 129133)DILI possible after bupropion discontinuation (134)+++
MianserinUnknownCholestatic; mixed; hepatocellularImmuno-allergic12–28 days Recovery: 4 (43, 135)1 case of DILI following mianserin dosage escalation (43); positive drug rechallenge (43)++
MirtazapineALT >3×ULN: 2% of treated patients (136); no DILI in clinical trials (136)Cholestatic; mixed; hepatocellularMetabolic2 weeks–3 yearsDuloxetineRecovery: 4(137139) ++
TianeptineNo difference in LFT versus placebo (140); no DILI in clinical trials (140)Cholestatic; hepatocellularImmuno-allergic6 days–2 months Recovery: 4 (141145)Major metabolic: beta oxidation; microvesicular steatosis in mice (146)++
AgomelatineALT >3×ULN: 1.4% with 25 mg/day and 2.5% with 50 mg/day (placebo: 0.6%) (147, 151)HepatocellularUnknownFirst months of treatment LT: 1; recovery: 1; (148150)Assessment of LFT recommended before treatment and then after 3, 6, 12, and 24 weeks of treatment (151); dose-dependent abnormal LFT in clinical trials (147); postmarketing settings: normalization of LFT in most cases (148)+++
a
DILI=drug-induced liver injury; FDA=U.S. Food and Drug Administration; LFT=liver function tests; LT=liver transplantation; recovery=full recovery; ULN=upper limit of normal; VBDS=vanishing bile duct syndrome.
Preexisting hepatic disease is usually not considered to be a risk factor for the development of antidepressant-induced liver injury, except in the case of duloxetine, for which DILI seems to be more frequent in patients with previous chronic liver disease or those who are at risk of liver disease (18, 31, 81). Furthermore, hepatic reserve is reduced in patients with cirrhosis or chronic hepatic failure, and when DILI occurs in such patients, it may be more severe (31, 153). Therefore, it has been suggested that agomelatine should be contraindicated in cases of preexisting liver disease (148).

Clinical Presentation and Diagnosis

Antidepressant-induced liver injury includes various biological and clinical presentations, ranging from isolated increases in liver enzyme levels to nonspecific symptoms such as fatigue, asthenia, anorexia, nausea, vomiting, and upper right abdominal pain, and also to more specific symptoms such as jaundice, dark urine or pale stool, progressive or even fulminant liver failure with hepatic encephalopathy, loss of hepatocellular functions, acute liver failure, and death. In most cases, however, patients are clinically asymptomatic and the biological alterations identified by abnormal results on liver function tests are the only elements that may raise a suspicion of antidepressant-induced liver injury.
Thus, the diagnosis of DILI often relies on the detection of an increase in ALT levels. In this context, it is important to note that there are physiological variations of ALT levels in healthy subjects. Indeed, in phase 1 clinical trials, aminotransferase activities between 1×ULN and 3×ULN can be observed in about 20% of patients treated with placebo during 2 weeks of follow-up (154). Also, although hospitalization is believed to eliminate confounding factors, it can itself be associated with an increase in ALT level (155). Similarly, changes in diet involving increased carbohydrate or fat intake can lead to a threefold increase in baseline ALT levels in only 3 days (156). Abnormal liver function test results for antidepressant-treated patients should therefore be interpreted with caution.
The mechanism of liver injury associated with antidepressants is metabolic or immuno-allergic (Table 2). A hypersensitivity syndrome (fever, rash, eosinophilia, autoantibodies) and a short latency period (1 to 6 weeks) (13) suggests immune-mediated hepatic injury, whereas the absence of any hypersensitivity syndrome and a longer latency period (1 month to 1 year) suggests an idiosyncratic metabolic mechanism (157). In most cases, the onset of DILI is between several days and 6 months after the beginning of antidepressant treatment (Table 2). Note that some of the clinical symptoms of antidepressant-induced liver injury, particularly fatigue, asthenia, anorexia, and nausea, may be misdiagnosed as symptoms of depression or anxiety and thus may lead to an increase in the antidepressant dosage or even to coprescriptions.
Antidepressant-related hepatic injury is an exclusion diagnosis: it can mimic almost all known liver diseases (viral hepatitis, alcoholic liver disease, nonalcoholic fatty liver disease, autoimmune hepatitis, metabolic diseases, biliary tract obstruction, hepatic ischemia, vascular obstruction) and consequently is diagnosed by ruling out all other possible causes (9). Several criteria are used to assess drug imputability: the patient’s age, the nature of the drug, previous case reports of antidepressant-induced liver injury, drug dosage, event chronology, description of first clinical signs, alcohol consumption, concomitant medication (including self-medication, phytotherapy products, and illicit drugs), other potential causes, and the results of liver function tests after drug withdrawal (a 50% decrease in liver enzyme levels following withdrawal of the suspected culprit drug is highly suggestive of DILI). Although drug rechallenge should be avoided because of the risk of severe hepatic failure, deliberate or inadvertent drug rechallenge resulting in a deterioration of liver function test findings provides strong evidence for drug imputability.

Outcome

Antidepressant-associated DILI is generally of the hepatocellular type and less frequently of the cholestatic or mixed types (Table 2). Generally, in the hepatocellular type, there is an increase in aminotransferase levels that can rapidly normalize with drug withdrawal. In severe cases, an increase in bilirubin concentrations associated with a decrease in prothrombin time is observed. In these rare cases, the drug must be withdrawn immediately because of the risk of fulminant hepatitis and liver failure. The cholestatic pattern of DILI occurs less commonly (20, 157, 158). The antidepressants most frequently associated with a cholestatic pattern are phenelzine, moclobemide, amitriptyline, mianserin, mirtazapine, and tianeptine. Regression of cholestatic DILI is slower than that of hepatocellular DILI (159). Cases of prolonged cholestasis and vanishing bile duct syndrome (disappearance of interlobular bile ducts) leading to biliary fibrosis have been described for amitriptyline (61) and imipramine (56).
For most patients, liver test results normalize after antidepressant withdrawal. However, in some cases there may be clinical symptoms and severe DILI, such as fulminant liver failure leading to death or liver transplantation. Data on the frequency of severe antidepressant-induced liver injury are not available except for nefazodone: the incidence of severe liver failure leading to death or liver transplantation is estimated to be 1 per 250,000–300,000 patient-years of treatment (6, 115, 116). Cases of fulminant hepatic failure leading to liver transplantation or death have also been reported for other antidepressants, including phenelzine, imipramine, amitriptyline, venlafaxine, duloxetine, sertraline, bupropion, trazodone, and agomelatine (27, 31, 35, 39, 50, 57, 62, 129, 149) (Table 2). In terms of prognosis, the presence of a hepatocellular pattern of DILI with jaundice (serum bilirubin level >2×ULN)—meeting the criteria of Hy’s law on DILI—is associated with a mortality rate of at least 10% (20, 157, 158). When the criteria of Hy’s law are met, the culprit drug should be withdrawn immediately. There is evidence suggesting that women and patients with concomitant stable chronic liver disease have more severe forms of DILI (160). However, there is currently no satisfactory method to assess the likelihood of developing liver failure.
Results for individual drugs are summarized in Table 2.

Discussion

Antidepressant liver toxicity has been underestimated in the scientific literature, and data on DILI from antidepressant agents are scarce. Nevertheless, cases of life-threatening hepatic failure and death have been reported in patients treated with antidepressants.
For the older drugs, notably MAO inhibitors, tricyclic/tetracyclic antidepressants, fluoxetine, and mianserin, data are scarce because the results of clinical trials are not available, and only case reports have been published. With more recent drugs, such as duloxetine, venlafaxine, bupropion, and agomelatine, data from both clinical trials and published case reports are available.
The available data show that all antidepressants are associated with a risk of hepatotoxicity. However, the evidence is insufficient for rigorous conclusions to be drawn about the prevalence and severity of antidepressant-induced liver injury (161). The drugs for which the frequency of reported DILI appears to be highest are MAO inhibitors, tricyclic/tetracyclic antidepressants, nefazodone, bupropion, duloxetine, and agomelatine. Those with apparently lower risks are citalopram, escitalopram, paroxetine, and fluvoxamine. Life-threatening or severe DILI has been reported for some antidepressants, including MAO inhibitors, tricyclic/tetracyclic antidepressants, venlafaxine, duloxetine, sertraline, bupropion, nefazodone, trazodone, and agomelatine.
In most cases, liver injury associated with antidepressants emerges between several days and 6 months after the beginning of treatment. Life-threatening DILI can occur, in some patients involving fulminant liver failure requiring liver transplantation and even leading to death. Cross-toxicity has been described for tricyclic/tetracyclic antidepressants and, to a lesser extent, SSRIs.
The main limitation of this review is related to publication biases that must be considered in the analysis of the literature. Any analysis involving case reports is subject to the inherent bias toward the publication of more severe cases. Also, the number of reported cases of DILI is inevitably higher for the most frequently used antidepressants, which may tend to indicate, falsely, a higher hepatotoxicity rate. By contrast, most clinical studies do not report the effects of antidepressant treatment on liver function or liver function markers, and the numbers of patients exposed to antidepressants in these studies are mostly insufficient to detect any potential liver toxicity.
The strengths of this review include the exhaustive analysis of published cases of DILI from antidepressant drugs and the evaluation of liver function test anomalies reported in clinical trials.
Detection of DILI during premarketing clinical trials is a difficult challenge because of the small numbers of patients treated and the short duration of the majority of antidepressant clinical trials (6–12 weeks) relative to the latency of DILI (54, 55). Therefore, indicators of the potential for severe DILI have been proposed by the FDA: an excess of aminotransferase values increasing to >3×ULN in the treatment group relative to the placebo group; any marked elevations of aminotransferase values to >5×ULN in the treatment group without a corresponding increase in the placebo group; and one or more cases of bilirubin levels increasing to >2×ULN associated with aminotransferase levels >3×ULN (Hy’s law) with no other explanation (3). The presence of one of these criteria may indicate a significant risk of DILI (at least 1 per 10,000 patient-years); consequently, shortly after marketing approval, cases of severe DILI may appear as the number of patients exposed increases. For newer antidepressants, such as agomelatine, the EMA recommends regular monitoring of liver function. Although serum aminotransferase activities are probably a poor indicator of DILI for most antidepressants, it remains the gold standard that should be used to monitor patients. Studies of various individual gene polymorphisms may allow the identification of more reliable indicators of the risk of DILI for use in the future.

Recommendations for Clinical Practice

Risk factors for DILI include age and polypharmacy. These risk factors should be checked systematically before prescribing antidepressants. It remains unclear whether existing liver damage favors, or is a risk factor for, antidepressant-induced liver injury.
Antidepressants suspected to have a higher potential for hepatotoxicity should be used with caution in elderly patients, in patients with coprescriptions, and in patients with substantial alcohol use, illicit substance use, or evidence of chronic liver disease, because in these patients the development of a severe form of DILI could have devastating consequences.
Although no dose-response relationship has been clearly demonstrated, the prescription of minimum effective dosages of antidepressant should be recommended to reduce the risk of DILI. Similarly, coprescriptions with antidepressants should be avoided as far as possible, to decrease the risk of DILI and especially the risk of severe DILI. Cases of possible drug-drug interaction have been described, so it would be advisable to avoid coprescriptions that may target the same CYP450 pathway in patients treated with antidepressants.
Ordinarily, treatments presumed to be associated with a greater risk of hepatotoxicity (iproniazid, nefazodone, tianeptine, phenelzine, imipramine, amitriptyline, duloxetine, bupropion, trazodone, and agomelatine) should not be initiated in individuals with preexisting liver failure. Although there is no clear evidence that preexisting chronic liver disease increases the likelihood of developing liver toxicity, baseline abnormalities can complicate monitoring of the patient. Moreover, in patients with a history of DILI associated with tricyclic/tetracyclic antidepressants, it is recommended that clinicians avoid prescribing these agents or other therapeutic compounds with a similar tricyclic structure, such as phenothiazines (41).
DILI is difficult to prevent, and consequently its early detection is a major goal. Physicians should therefore be aware of the possibility of antidepressant-associated liver injury. For antidepressants with a high potential for hepatotoxicity and for patients with known risk factors, systematic pretherapeutic screening and regular assessment of hepatic enzymes while under treatment may be useful. Baseline assessment of ALT, ALP, and bilirubin levels can provide an estimation of reference values and identify any liver disease. Given the physiological variations of ALT levels, a single determination of ALT before or during treatment may not be sufficiently informative. Moreover, an increase in ALT level (before any antidepressant treatment or even during treatment) may be related to an underlying liver disease, not necessarily severe, such as weight gain (nonalcoholic fatty liver disease) or active hepatitis C virus infection, that does not contraindicate antidepressant treatment. Patients with alcohol abuse may also have increased ALT levels and need antidepressants. Overall, baseline ALT values are useful as they help in the interpretation of abnormal liver function test results during antidepressant treatment; such abnormal results may be a manifestation of either underlying liver disease or antidepressant-induced hepatotoxicity.
For safety reasons, when using an antidepressant associated with a greater risk of hepatotoxicity (iproniazid, nefazodone, phenelzine, imipramine, amitriptyline, duloxetine, bupropion, trazodone, tianeptine, or agomelatine) or prescribing an antidepressant for a patient with an underlying liver disease, regular monitoring of serum ALT should be discussed, even though there is no formal recommendation for such investigations. For agomelatine, assessment of liver function is recommended for all patients before treatment and again after 3, 6, 12, and 24 weeks of treatment (151). Nevertheless, further studies are needed both to confirm the efficacy of follow-up liver function tests for the early detection of DILI and to identify patients at greater risk of DILI.
Patients should also be informed that antidepressant therapy can be associated with liver abnormalities ranging from asymptomatic reversible increases in serum aminotransferase levels to signs and symptoms of liver dysfunction (jaundice, anorexia, gastrointestinal complaints, malaise, and so on) and even liver failure resulting in death or liver transplantation. They should be informed that the risk of severe liver abnormalities may be increased by alcohol consumption, illicit drug use, and over-the-counter medications. Consumption of such agents should be discouraged in patients receiving antidepressants. Patients should be encouraged to refer to their primary care physician or psychiatrist should any clinical symptoms emerge suggesting the possibility of DILI, and to stop treatment if jaundice develops.
Indeed, antidepressants should be discontinued immediately in any patient with suspected DILI. It has been suggested that these drugs should be promptly discontinued when serum ALT levels are >3×ULN (or >5×ULN, as recently suggested) (9) or if there is an unexplained increase in bilirubin levels to >2×ULN. For patients with high baseline ALT levels, drug discontinuation is recommended when serum ALT levels are >3 times the baseline value. In patients with a concomitant increase in ALT and bilirubin, treatment continuation (particularly of imipramine/desipramine, amitriptyline, paroxetine, trazodone, or agomelatine) despite hepatotoxicity could lead to severe hepatic failure or chronic hepatocellular dysfunction that may progress to liver cirrhosis.
Investigations should be performed to exclude potential causes of liver injury and to confirm the diagnosis of DILI, including serological tests for hepatotropic viruses (hepatitis A, B, C, and E viruses, Epstein-Barr virus, cytomegalovirus, and herpes simplex virus), autoantibody tests, iron and copper levels, and abdominal ultrasonography. Alcohol consumption should also be evaluated, as alcoholism is common among depressed patients and may be the cause of liver injury in some cases. A recent study showed that acute hepatitis E is the cause of some cases of liver disease that were initially suspected to be DILI (162). Hepatitis E testing should therefore be performed in all cases of suspected antidepressant-induced liver injury, particularly if the clinical features are compatible with acute viral hepatitis. Abdominal ultrasonography should also be carried out to search for fatty liver, bile duct lithiasis, and permeability of the portal vein and suprahepatic veins.
As many antidepressants may lead to weight gain, an increase in ALT levels during antidepressant treatment may be related to an antidepressant-induced metabolic syndrome (nonalcoholic fatty liver disease), but not to a direct antidepressant-induced liver toxicity. Moreover, the depression itself may be associated with a metabolic syndrome (163, 164). Studies are needed to explore the relationship between metabolic syndrome (nonalcoholic fatty liver disease), antidepressant-induced weight gain, and abnormal liver function tests in antidepressant-treated patients.
Evaluation of liver function (ALT, ALP, and bilirubin tests) is essential once DILI is suspected and until normalization or return to baseline values. Liver function in most cases improves after drug discontinuation, but hepatic injury may persist for several months, particularly in cases with the cholestatic pattern.
Finally, patients with antidepressant-induced liver injury should be presumed to be at increased risk of liver injury if the same antidepressant is reintroduced. Accordingly, such patients should not be considered for retreatment with the same antidepressant or with any other antidepressant that may display cross-toxicity.

Conclusions

All antidepressant drugs may potentially cause liver injury, even at therapeutic doses. Nevertheless, DILI from antidepressant agents is a rare event, although in some cases it is irreversible. As there is still no strategy available to prevent antidepressant-induced adverse hepatic events, early detection and prompt drug discontinuation remain critical. Surveillance of liver function in clinical trials and careful evaluation of reported abnormalities could make a major contribution to the early detection of antidepressants associated with a high risk of causing DILI. Finally, further research is required before rigorously founded recommendations can be established for clinical practice.

References

1.
Schuster D, Laggner C, Langer T: Why drugs fail: a study on side effects in new chemical entities. Curr Pharm Des 2005; 11:3545–3559
2.
Temple RJ, Himmel MH: Safety of newly approved drugs: implications for prescribing. JAMA 2002; 287:2273–2275
3.
US Food and Drug Administration, Center for Drug Evaluation and Research: Guidance for Industry: Drug-Induced Liver Injury: Premarketing Clinical Evaluation. July 2009. http://www.fda.gov/downloads/Drugs/.../Guidances/UCM174090.pdf
4.
European Medicines Agency, Committee for Medicinal Products for Human Use: Reflection paper on non-clinical evaluation of drug-induced liver injury. June 24, 2010. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2010/07/WC500094591.pdf
5.
Selim K, Kaplowitz N: Hepatotoxicity of psychotropic drugs. Hepatology 1999; 29:1347–1351
6.
DeSanty KP, Amabile CM: Antidepressant-induced liver injury. Ann Pharmacother 2007; 41:1201–1211
7.
Sgro C, Clinard F, Ouazir K, Chanay H, Allard C, Guilleminet C, Lenoir C, Lemoine A, Hillon P: Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology 2002; 36:451–455
8.
Lasser KE, Allen PD, Woolhandler SJ, Himmelstein DU, Wolfe SM, Bor DH: Timing of new black box warnings and withdrawals for prescription medications. JAMA 2002; 287:2215–2220
9.
Aithal GP, Watkins PB, Andrade RJ, Larrey D, Molokhia M, Takikawa H, Hunt CM, Wilke RA, Avigan M, Kaplowitz N, Bjornsson E, Daly AK: Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther 2011; 89:806–815
10.
Neuschwander-Tetri BA, Caldwell SH: Nonalcoholic steatohepatitis: summary of an AASLD Single Topic Conference. Hepatology 2003; 37:1202–1219
11.
Verma S, Kaplowitz N: Diagnosis, management, and prevention of drug-induced liver injury. Gut 2009; 58:1555–1564
12.
Lammert C, Einarsson S, Saha C, Niklasson A, Bjornsson E, Chalasani N: Relationship between daily dose of oral medications and idiosyncratic drug-induced liver injury: search for signals. Hepatology 2008; 47:2003–2009
13.
Kaplowitz N: Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005; 4:489–499
14.
Carvajal García-Pando A, García del Pozo J, Sánchez AS, Velasco MA, Rueda de Castro AM, Lucena MI: Hepatotoxicity associated with the new antidepressants. J Clin Psychiatry 2002; 63:135–137
15.
Cooper GL: The safety of fluoxetine: an update. Br J Psychiatry Suppl 1988; 3(Sep):77–86
16.
Boyer WF, Blumhardt CL: The safety profile of paroxetine. J Clin Psychiatry 1992; 53(suppl):61–66
17.
Rudolph RL, Derivan AT: The safety and tolerability of venlafaxine hydrochloride: analysis of the clinical trials database. J Clin Psychopharmacol 1996; 16:54S–59S
18.
Xue F, Strombom I, Turnbull B, Zhu S, Seeger JD: Duloxetine for depression and the incidence of hepatic events in adults. J Clin Psychopharmacol 2011; 31:517–522
19.
Holmberg MB, Jansson B: A study of blood count and serum transaminase in prolonged treatment with amitriptyline. J New Drugs 1962; 2:361–365
20.
Zimmerman HJ, Ishak KG: The hepatic injury of monoamine oxidase inhibitors. J Clin Psychopharmacol 1987; 7:211–213
21.
Wille SM, Cooreman SG, Neels HM, Lambert WE: Relevant issues in the monitoring and the toxicology of antidepressants. Crit Rev Clin Lab Sci 2008; 45:25–89
22.
Timmings P, Lamont D: Intrahepatic cholestasis associated with moclobemide leading to death. Lancet 1996; 347:762–763
23.
Iribarne C, Dréano Y, Bardou LG, Ménez JF, Berthou F: Interaction of methadone with substrates of human hepatic cytochrome P450 3A4. Toxicology 1997; 117:13–23
24.
Andersson T, Miners JO, Veronese ME, Birkett DJ: Diazepam metabolism by human liver microsomes is mediated by both S-mephenytoin hydroxylase and CYP3A isoforms. Br J Clin Pharmacol 1994; 38:131–137
25.
Cunningham ML: Acute hepatic necrosis following treatment with amitriptyline and diazepam. Br J Psychiatry 1965; 111:1107–1109
26.
Horsmans Y, De Clercq M, Sempoux C: Venlafaxine-associated hepatitis. Ann Intern Med 1999; 130:944
27.
Detry O, Delwaide J, De Roover A, Hans MF, Delbouille MH, Monard J, Honoré P: Fulminant hepatic failure induced by venlafaxine and trazodone therapy: a case report. Transplant Proc 2009; 41:3435–3436
28.
Yasui N, Otani K, Kaneko S, Ohkubo T, Osanai T, Ishida M, Mihara K, Kondo T, Sugawara K, Fukushima Y: Inhibition of trazodone metabolism by thioridazine in humans. Ther Drug Monit 1995; 17:333–335
29.
Park YM, Lee BH, Lee HJ, Kang SG: Cholestatic jaundice induced by duloxetine in a patient with major depressive disorder. Psychiatry Investig 2010; 7:228–230
30.
Hanje AJ, Pell LJ, Votolato NA, Frankel WL, Kirkpatrick RB: Case report: fulminant hepatic failure involving duloxetine hydrochloride. Clin Gastroenterol Hepatol 2006; 4:912–917
31.
Vuppalanchi R, Hayashi PH, Chalasani N, Fontana RJ, Bonkovsky H, Saxena R, Kleiner D, Hoofnagle JH; Drug-Induced Liver Injury Network (DILIN): Duloxetine hepatotoxicity: a case-series from the drug-induced liver injury network. Aliment Pharmacol Ther 2010; 32:1174–1183
32.
Yuan W, Williams B: Acute hepatic failure involving duloxetine hydrochloride. J Neuropsychiatry Clin Neurosci 2012; 24:E48–E49
33.
Cacabelos R: Donepezil in Alzheimer’s disease: from conventional trials to pharmacogenetics. Neuropsychiatr Dis Treat 2007; 3:303–333
34.
Verrico MM, Nace DA, Towers AL: Fulminant chemical hepatitis possibly associated with donepezil and sertraline therapy. J Am Geriatr Soc 2000; 48:1659–1663
35.
Fartoux-Heymann L, Hézode C, Zafrani ES, Dhumeaux D, Mallat A: Acute fatal hepatitis related to sertraline. J Hepatol 2001; 35:683–684
36.
Azaz-Livshits T, Hershko A, Ben-Chetrit E: Paroxetine associated hepatotoxicity: a report of 3 cases and a review of the literature. Pharmacopsychiatry 2002; 35:112–115
37.
Seree EJ, Pisano PJ, Placidi M, Rahmani R, Barra YA: Identification of the human and animal hepatic cytochromes P450 involved in clonazepam metabolism. Fundam Clin Pharmacol 1993; 7:69–75
38.
Conway CR, McGuire JM, Baram VY: Nefazodone-induced liver failure. J Clin Psychopharmacol 2004; 24:353–354
39.
Hull M, Jones R, Bendall M: Fatal hepatic necrosis associated with trazodone and neuroleptic drugs. BMJ 1994; 309:378
40.
Larrey D, Rueff B, Pessayre D, Danan G, Algard M, Geneve J, Benhamou JP: Cross hepatotoxicity between tricyclic antidepressants. Gut 1986; 27:726–727
41.
Remy AJ, Larrey D, Pageaux GP, Ribstein J, Ramos J, Michel H: Cross hepatotoxicity between tricyclic antidepressants and phenothiazines. Eur J Gastroenterol Hepatol 1995; 7:373–376
42.
Solomons K, Gooch S, Wong A: Toxicity with selective serotonin reuptake inhibitors. Am J Psychiatry 2005; 162:1225
43.
Otani K, Kaneko S, Tasaki H, Fukushima Y: Hepatic injury caused by mianserin. BMJ 1989; 299:519
44.
Li Wai Suen CF, Boyapati R, Simpson I, Dev A: Acute liver injury secondary to sertraline. BMJ Case Rep, Sep 26, 2013;
45.
Danan G, Homberg JC, Bernuau J, Roche-Sicot J, Pessayre D: [Iproniazid-induced hepatitis: the diagnostic value of a new antimitochondrial antibody anti-M6.] Gastroenterol Clin Biol 1983; 7:529–532 (French)
46.
Rosenblum LE, Korn RJ, Zimmerman HJ: Hepatocellular jaundice as a complication of iproniazid therapy. Arch Intern Med 1960; 105:583–593
47.
Lefebure B, Castot A, Danan G, Elmalem J, Jean-Pastor MJ, Efthymiou ML: [Hepatitis from antidepressants: evaluation of cases from the French Association of Drug Surveillance Centers and the Technical Committee.] Therapie 1984; 39:509–516 (French)
48.
Maille F, Duvoux C, Cherqui D, Radier C, Zafrani ES, Dhumeaux D: [Auxiliary hepatic transplantation in iproniazid-induced subfulminant hepatitis: should iproniazid still be sold in France?] Gastroenterol Clin Biol 1999; 23:1083–1085 (French)
49.
Nelson SD, Mitchell JR, Timbrell JA, Snodgrass WR, Corcoran GB: Isoniazid and iproniazid: activation of metabolites to toxic intermediates in man and rat. Science 1976; 193:901–903
50.
Gómez-Gil E, Salmerón JM, Mas A: Phenelzine-induced fulminant hepatic failure. Ann Intern Med 1996; 124:692–693
51.
Bonkovsky HL, Blanchette PL, Schned AR: Severe liver injury due to phenelzine with unique hepatic deposition of extracellular material. Am J Med 1986; 80:689–692
52.
García-Díaz JD, Gila A, Fraguas C, Castillo I: [Cholestatic hepatitis associated with the use of moclobemide.] Med Clin (Barc) 1998; 111:77 (Spanish)
53.
Klerman GL, Cole JO: Clinical pharmacology of imipramine and related antidepressant compounds. Pharmacol Rev 1965; 17:101–141
54.
Van Amerongen AP, Ferrey G, Tournoux A: A randomised, double-blind comparison of milnacipran and imipramine in the treatment of depression. J Affect Disord 2002; 72:21–31
55.
Koran LM, Gelenberg AJ, Kornstein SG, Howland RH, Friedman RA, DeBattista C, Klein D, Kocsis JH, Schatzberg AF, Thase ME, Rush AJ, Hirschfeld RM, LaVange LM, Keller MB: Sertraline versus imipramine to prevent relapse in chronic depression. J Affect Disord 2001; 65:27–36
56.
Horst DA, Grace ND, LeCompte PM: Prolonged cholestasis and progressive hepatic fibrosis following imipramine therapy. Gastroenterology 1980; 79:550–554
57.
Powell WJ, Koch-Weser J, Williams RA: Lethal hepatic necrosis after therapy with imipramine and desipramine. JAMA 1968; 206:642–645
58.
Shaefer MS, Edmunds AL, Markin RS, Wood RP, Pillen TJ, Shaw BW: Hepatic failure associated with imipramine therapy. Pharmacotherapy 1990; 10:66–69
59.
Morrow PL, Hardin NJ, Bonadies J: Hypersensitivity myocarditis and hepatitis associated with imipramine and its metabolite, desipramine. J Forensic Sci 1989; 34:1016–1020
60.
Ilan Y, Samuel D, Reynes M, Tur-Kaspa R: Hepatic failure associated with imipramine therapy. Pharmacopsychiatry 1996; 29:79–80
61.
Larrey D, Amouyal G, Pessayre D, Degott C, Danne O, Machayekhi JP, Feldmann G, Benhamou JP: Amitriptyline-induced prolonged cholestasis. Gastroenterology 1988; 94:200–203
62.
Randeva HS, Bangar V, Sailesh S, Hillhouse EW: Fatal cholestatic jaundice associated with amitriptyline. Int J Clin Pract 2000; 54:405–406
63.
Danan G, Bernuau J, Moullot X, Degott C, Pessayre D: Amitriptyline-induced fulminant hepatitis. Digestion 1984; 30:179–184
64.
Morgan DH: Jaundice associated with amitriptyline. Br J Psychiatry 1969; 115:105–106
65.
Yon J, Anuras S: Hepatitis caused by amitriptyline therapy. JAMA 1975; 232:833–834
66.
Aleem A, Lingam V: Hepatotoxicity following treatment with maprotiline. J Clin Psychopharmacol 1987; 7:54–55
67.
Moldawsky RJ: Hepatotoxicity associated with maprotiline therapy: case report. J Clin Psychiatry 1984; 45:178–179
68.
Prudent A, Marchetti B, Pignol D, Lacroix G, Bouckson G, Legré M: [Acute benign hepatitis probably due to maprotiline.] Gastroenterol Clin Biol 1988; 12:80 (French)
69.
Weinstein RP, Gosselin JY: Case report of hepatotoxicity associated with maprotiline. Can J Psychiatry 1988; 33:233–234
70.
Braun JS, Geiger R, Wehner H, Schäffer S, Berger M: Hepatitis caused by antidepressive therapy with maprotiline and opipramol. Pharmacopsychiatry 1998; 31:152–155
71.
Alderman CP, Atchison MM, McNeece JI: Concurrent agranulocytosis and hepatitis secondary to clomipramine therapy. Br J Psychiatry 1993; 162:688–689
72.
Maroy B: [Acute cytolytic hepatitis after taking venlafaxine.] Gastroenterol Clin Biol 2002; 26:804 (French)
73.
Phillips BB, Digmann RR, Beck MG: Hepatitis associated with low-dose venlafaxine for postmenopausal vasomotor symptoms. Ann Pharmacother 2006; 40:323–327
74.
Yildirim B, Tuncer C, Ergun M, Unal S: Venlafaxine-induced hepatotoxicity in a patient with ulcerative colitis. Ann Hepatol 2009; 8:271–272
75.
Stadlmann S, Portmann S, Tschopp S, Terracciano LM: Venlafaxine-induced cholestatic hepatitis: case report and review of literature. Am J Surg Pathol 2012; 36:1724–1728
76.
Kim KY, Hwang W, Narendran R: Acute liver damage possibly related to sertraline and venlafaxine ingestion. Ann Pharmacother 1999; 33:381–382
77.
Pradeep RJ, Victor G, Iby N, Kurpad SS, Galgali RB, Srinivasan K: Venlafaxine induced hepatitis. J Assoc Physicians India 2004; 52:340
78.
Cardona X, Avila A, Castellanos P: Venlafaxine-associated hepatitis. Ann Intern Med 2000; 132:417
79.
Choy EH, Mease PJ, Kajdasz DK, Wohlreich MM, Crits-Christoph P, Walker DJ, Chappell AS: Safety and tolerability of duloxetine in the treatment of patients with fibromyalgia: pooled analysis of data from five clinical trials. Clin Rheumatol 2009; 28:1035–1044
80.
McIntyre RS, Panjwani ZD, Nguyen HT, Woldeyohannes HO, Alsuwaidan M, Soczynska JK, Lourenco MT, Konarski JZ, Kennedy SH: The hepatic safety profile of duloxetine: a review. Expert Opin Drug Metab Toxicol 2008; 4:281–285
81.
Wernicke J, Acharya N, Strombom I, Gahimer JL, D’Souza DN, DiPietro N, Uetrecht JP: Hepatic effects of duloxetine, II: spontaneous reports and epidemiology of hepatic events. Curr Drug Saf 2008; 3:143–153
82.
Kang SG, Park YM, Lee HJ, Yoon B: Duloxetine-induced liver injury in patients with major depressive disorder. Psychiatry Investig 2011; 8:269–271
83.
Suri A, Reddy S, Gonzales C, Knadler MP, Branch RA, Skinner MH: Duloxetine pharmacokinetics in cirrhotics compared with healthy subjects. Int J Clin Pharmacol Ther 2005; 43:78–84
84.
US Food and Drug Administration: Label approved for Cymbalta. Oct 2012. http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/021427s040s041lbl.pdf
85.
European Medicines Agency: Cymbalta: European Public Assessment Report: Product Information. Aug 30, 2011. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR__Product_Information/human/000572/WC500036781.pdf
86.
Greist JH, Jefferson JW, Kobak KA, Chouinard G, DuBoff E, Halaris A, Kim SW, Koran L, Liebowtiz MR, Lydiard B, et al.: A 1 year double-blind placebo-controlled fixed dose study of sertraline in the treatment of obsessive-compulsive disorder. Int Clin Psychopharmacol 1995; 10:57–65
87.
Leombruni P, Pierò A, Lavagnino L, Brustolin A, Campisi S, Fassino S: A randomized, double-blind trial comparing sertraline and fluoxetine 6-month treatment in obese patients with binge eating disorder. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32:1599–1605
88.
Hautekeete ML, Colle I, van Vlierberghe H, Elewaut A: Symptomatic liver injury probably related to sertraline. Gastroenterol Clin Biol 1998; 22:364–365
89.
Menon R, Howard R: Sertraline and liver toxicity in the elderly. Int J Geriatr Psychiatry 1994; 9:332–334
90.
Persky S, Reinus JF: Sertraline hepatotoxicity: a case report and review of the literature on selective serotonin reuptake inhibitor hepatotoxicity. Dig Dis Sci 2003; 48:939–944
91.
Tabak F, Gunduz F, Tahan V, Tabak O, Ozaras R: Sertraline hepatotoxicity: report of a case and review of the literature. Dig Dis Sci 2009; 54:1589–1591
92.
Collados Arroyo V, Plaza Aniorte J, Hallal H, Pérez-Cuadrado E: [Sertraline-induced hepatotoxicity.] Farm Hosp 2008; 32:60–61 (Spanish)
93.
Lecrubier Y, Judge R; Collaborative Paroxetine Panic Study Investigators: Long-term evaluation of paroxetine, clomipramine, and placebo in panic disorder. Acta Psychiatr Scand 1997; 95:153–160
94.
Nemeroff CB, Evans DL, Gyulai L, Sachs GS, Bowden CL, Gergel IP, Oakes R, Pitts CD: Double-blind, placebo-controlled comparison of imipramine and paroxetine in the treatment of bipolar depression. Am J Psychiatry 2001; 158:906–912
95.
Benbow SJ, Gill G: Paroxetine and hepatotoxicity. BMJ 1997; 314:1387
96.
Colakoglu O, Tankurt E, Unsal B, Ugur F, Kupelioglu A, Buyrac Z, Akpinar Z: Toxic hepatitis associated with paroxetine. Int J Clin Pract 2005; 59:861–862
97.
Guzmán Ruiz O, Ramírez Martín del Campo M, Fernandez López I, Romero Gómez M: [Hepatotoxicity induced by paroxetine.] Med Clin (Barc) 2005; 124:399 (Spanish)
98.
Odeh M, Misselevech I, Boss JH, Oliven A: Severe hepatotoxicity with jaundice associated with paroxetine. Am J Gastroenterol 2001; 96:2494–2496
99.
Pompili M, Tittoto P, Mascianà R, Gasbarrini G, Rapaccini GL: Acute hepatitis associated with use of paroxetine. Intern Emerg Med 2008; 3:275–277
100.
Tollefson GD, Rampey AH, Potvin JH, Jenike MA, Rush AJ, Kominguez RA, Koran LM, Shear MK, Goodman W, Genduso LA: A multicenter investigation of fixed-dose fluoxetine in the treatment of obsessive-compulsive disorder. Arch Gen Psychiatry 1994; 51:559–567
101.
Marcus MD, Wing RR, Ewing L, Kern E, McDermott M, Gooding W: A double-blind, placebo-controlled trial of fluoxetine plus behavior modification in the treatment of obese binge-eaters and non-binge-eaters. Am J Psychiatry 1990; 147:876–881
102.
Bobichon R, Bernard G, Mion F: [Acute hepatitis during treatment with fluoxetine.] Gastroenterol Clin Biol 1993; 17:406–407 (French)
103.
Cai Q, Benson MA, Talbot TJ, Devadas G, Swanson HJ, Olson JL, Kirchner JP: Acute hepatitis due to fluoxetine therapy. Mayo Clin Proc 1999; 74:692–694
104.
Friedenberg FK, Rothstein KD: Hepatitis secondary to fluoxetine treatment. Am J Psychiatry 1996; 153:580
105.
Johnston DE, Wheeler DE: Chronic hepatitis related to use of fluoxetine. Am J Gastroenterol 1997; 92:1225–1226
106.
Mars F, Dumas de la Roque G, Goissen P: [Acute hepatitis during treatment with fluoxetine.] Gastroenterol Clin Biol 1991; 15:270–271 (French)
107.
Gleason OC, Yates WR, Isbell MD, Philipsen MA: An open-label trial of citalopram for major depression in patients with hepatitis C. J Clin Psychiatry 2002; 63:194–198
108.
Schaefer M, Sarkar R, Knop V, Effenberger S, Friebe A, Heinze L, Spengler U, Schlaepfer T, Reimer J, Buggisch P, Ockenga J, Link R, Rentrop M, Weidenbach H, Fromm G, Lieb K, Baumert TF, Heinz A, Discher T, Neumann K, Zeuzem S, Berg T: Escitalopram for the prevention of peginterferon-α2a-associated depression in hepatitis C virus-infected patients without previous psychiatric disease: a randomized trial. Ann Intern Med 2012; 157:94–103
109.
López-Torres E, Lucena MI, Seoane J, Verge C, Andrade RJ: Hepatotoxicity related to citalopram. Am J Psychiatry 2004; 161:923–924
110.
Neumann H, Csepregi A, Evert M, Malfertheiner P: Drug-induced liver disease related to citalopram. J Clin Psychopharmacol 2008; 28:254–255
111.
Merino MI, Carrero AA, García JF, Martínez JA: [Citalopram hepatotoxicity.] Med Clin (Barc) 2011; 136:270–271 (Spanish)
112.
Del Val Antoñana A, Ortiz Polo I, Roselló Sastre E, Moreno-Osset E: [Hepatotoxicity related to escitalopram.] Med Clin (Barc) 2008; 131:798 (Spanish)
113.
Stewart DE: Hepatic adverse reactions associated with nefazodone. Can J Psychiatry 2002; 47:375–377
114.
Gelenberg AJ, Trivedi MH, Rush AJ, Thase ME, Howland R, Klein DN, Kornstein SG, Dunner DL, Markowitz JC, Hirschfeld RM, Keitner GI, Zajecka J, Kocsis JH, Russell JM, Miller I, Manber R, Arnow B, Rothbaum B, Munsaka M, Banks P, Borian FE, Keller MB: Randomized, placebo-controlled trial of nefazodone maintenance treatment in preventing recurrence in chronic depression. Biol Psychiatry 2003; 54:806–817
115.
Schirren CA, Baretton G: Nefazodone-induced acute liver failure. Am J Gastroenterol 2000; 95:1596–1597
116.
Aranda-Michel J, Koehler A, Bejarano PA, Poulos JE, Luxon BA, Khan CM, Ee LC, Balistreri WF, Weber FL: Nefazodone-induced liver failure: report of three cases. Ann Intern Med 1999; 130:285–288
117.
Eloubeidi MA, Gaede JT, Swaim MW: Reversible nefazodone-induced liver failure. Dig Dis Sci 2000; 45:1036–1038
118.
Lucena MI, Andrade RJ, Gomez-Outes A, Rubio M, Cabello MR: Acute liver failure after treatment with nefazodone. Dig Dis Sci 1999; 44:2577–2579
119.
Tzimas GN, Dion B, Deschênes M: Early onset, nefazodone-induced fulminant hepatic failure. Am J Gastroenterol 2003; 98:1663–1664
120.
Beck PL, Bridges RJ, Demetrick DJ, Kelly JK, Lee SS: Chronic active hepatitis associated with trazodone therapy. Ann Intern Med 1993; 118:791–792
121.
Chu AG, Gunsolly BL, Summers RW, Alexander B, McChesney C, Tanna VL: Trazodone and liver toxicity. Ann Intern Med 1983; 99:128–129
122.
Fernandes NF, Martin RR, Schenker S: Trazodone-induced hepatotoxicity: a case report with comments on drug-induced hepatotoxicity. Am J Gastroenterol 2000; 95:532–535
123.
Rettman KS, McClintock C: Hepatotoxicity after short-term trazodone therapy. Ann Pharmacother 2001; 35:1559–1561
124.
Sheikh KH, Nies AS: Trazodone and intrahepatic cholestasis. Ann Intern Med 1983; 99:572
125.
Sánchez Ruiz JC, González López E, Aparicio Tijeras C, Ezquerra Gadea J: [Hepatotoxicity due to trazodone.] Aten Primaria 2004; 34:104 (Spanish)
126.
Longstreth GF, Hershman J: Trazodone-induced hepatotoxicity and leukonychia. J Am Acad Dermatol 1985; 13:149–150
127.
Hu KQ, Tiyyagura L, Kanel G, Redeker AG: Acute hepatitis induced by bupropion. Dig Dis Sci 2000; 45:1872–1873
128.
Tsoi DT, Porwal M, Webster AC: Efficacy and safety of bupropion for smoking cessation and reduction in schizophrenia: systematic review and meta-analysis. Br J Psychiatry 2010; 196:346–353
129.
Humayun F, Shehab TM, Tworek JA, Fontana RJ: A fatal case of bupropion (Zyban) hepatotoxicity with autoimmune features: case report. J Med Case Reports 2007; 1:88
130.
Alvaro D, Onetti-Muda A, Moscatelli R, Atili AF: Acute cholestatic hepatitis induced by bupropion prescribed as pharmacological support to stop smoking: a case report. Dig Liver Dis 2001; 33:703–706
131.
Carlos Titos-Arcos J, Hallal H, Collados V, Plaza-Aniorte J: [Acute hepatitis secondary to bupropion.] Gastroenterol Hepatol 2008; 31:549 (Spanish)
132.
Oslin DW, Duffy K: The rise of serum aminotransferases in a patient treated with bupropion. J Clin Psychopharmacol 1993; 13:364–365
133.
Bagshaw SM, Cload B, Gilmour J, Leung ST, Bowen TJ: Drug-induced rash with eosinophilia and systemic symptoms syndrome with bupropion administration. Ann Allergy Asthma Immunol 2003; 90:572–575
134.
Khoo AL, Tham LS, Lee KH, Lim GK: Acute liver failure with concurrent bupropion and carbimazole therapy. Ann Pharmacother 2003; 37:220–223
135.
Barbare JC, Biour M, Cadot T, Latrive JP: [Hepatotoxicity of mianserin: a case with positive reintroduction.] Gastroenterol Clin Biol 1992; 16:486–488 (French)
136.
Montgomery SA: Safety of mirtazapine: a review. Int Clin Psychopharmacol 1995; 10(suppl 4):37–45
137.
Hui CK, Yuen MF, Wong WM, Lam SK, Lai CL: Mirtazapine-induced hepatotoxicity. J Clin Gastroenterol 2002; 35:270–271
138.
Adetunji B, Basil B, Mathews M, Osinowo T: Mirtazapine-associated dose-dependent and asymptomatic elevation of hepatic enzymes. Ann Pharmacother 2007; 41:359
139.
Kang SG, Yoon BM, Park YM: Mirtazapine-induced hepatocellular-type liver injury. Ann Pharmacother 2011; 45:825–826
140.
Kasper S, Olié JP: A meta-analysis of randomized controlled trials of tianeptine versus SSRI in the short-term treatment of depression. Eur Psychiatry 2002; 17(suppl 3):331–340
141.
Fromenty B, Freneaux E, Labbe G, Deschamps D, Larrey D, Letteron P, Pessayre D: Tianeptine, a new tricyclic antidepressant metabolized by beta-oxidation of its heptanoic side chain, inhibits the mitochondrial oxidation of medium and short chain fatty acids in mice. Biochem Pharmacol 1989; 38:3743–3751
142.
Le Bricquir Y, Larrey D, Blanc P, Pageaux GP, Michel H: Tianeptine: an instance of drug-induced hepatotoxicity predicted by prospective experimental studies. J Hepatol 1994; 21:771–773
143.
Macaigne G, Auriault ML, Chayette C, Cheiab S, Deplus R: [Tianeptine induced acute hepatitis.] Gastroenterol Clin Biol 2003; 27:348–349 (French)
144.
Rifflet H, Vuillemin E, Rifflet I, Oberti F, Pilette C, Calès P: [Acute painful and febrile hepatic involvement related to ingestion of tianeptine.] Gastroenterol Clin Biol 1996; 20:606–607 (French)
145.
Balleyguier C, Stérin D, Ziol M, Trinchet JC: [Acute mixed hepatitis caused by tianeptine.] Gastroenterol Clin Biol 1996; 20:607–608 (French)
146.
Fromenty B, Pessayre D: Impaired mitochondrial function in microvesicular steatosis: effects of drugs, ethanol, hormones, and cytokines. J Hepatol 1997; 26(suppl 2):43–53
147.
Howland RH: A benefit-risk assessment of agomelatine in the treatment of major depression. Drug Saf 2011; 34:709–731
148.
Gahr M, Freudenmann RW, Connemann BJ, Hiemke C, Schönfeldt-Lecuona C: Agomelatine and hepatotoxicity: implications of cumulated data derived from spontaneous reports of adverse drug reactions. Pharmacopsychiatry 2013; 46:214–220
149.
Gruz F, Raffa S, Santucci C, Papale RM, Videla MG, Gimena Fernandez M, Yantorno S, Descalzi VI: [Agomelatine: fulminant liver failure in a patient with fatty liver.] Gastroenterol Hepatol (Epub ahead of print, July 9, 2013) (Spanish)
150.
Štuhec M: Agomelatine-induced hepatotoxicity. Wien Klin Wochenschr 2013; 125:225–226
151.
European Medicines Agency: Valdoxan: European Public Assessment Report: Product Information. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/000915/WC500046227.pdf
152.
Chen M, Borlak J, Tong W: High lipophilicity and high daily dose of oral medications are associated with significant risk for drug-induced liver injury. Hepatology 2013; 58:388–396
153.
Andrade RJ, Lucena MI, Fernández MC, Vega JL, Camargo R: Hepatotoxicity in patients with cirrhosis, an often unrecognized problem: lessons from a fatal case related to amoxicillin/clavulanic acid. Dig Dis Sci 2001; 46:1416–1419
154.
Rosenzweig P, Miget N, Brohier S: Transaminase elevation on placebo during phase I trials: prevalence and significance. Br J Clin Pharmacol 1999; 48:19–23
155.
Narjes H, Nehmiz G: Effect of hospitalisation on liver enzymes in healthy subjects. Eur J Clin Pharmacol 2000; 56:329–333
156.
Purkins L, Love ER, Eve MD, Wooldridge CL, Cowan C, Smart TS, Johnson PJ, Rapeport WG: The influence of diet upon liver function tests and serum lipids in healthy male volunteers resident in a Phase I unit. Br J Clin Pharmacol 2004; 57:199–208
157.
Andrade RJ, Lucena MI, Fernández MC, Pelaez G, Pachkoria K, García-Ruiz E, García-Muñoz B, González-Grande R, Pizarro A, Durán JA, Jiménez M, Rodrigo L, Romero-Gomez M, Navarro JM, Planas R, Costa J, Borras A, Soler A, Salmerón J, Martin-Vivaldi R; Spanish Group for the Study of Drug-Induced Liver Disease: Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology 2005; 129:512–521
158.
Björnsson E, Olsson R: Outcome and prognostic markers in severe drug-induced liver disease. Hepatology 2005; 42:481–489
159.
Bjornsson ES, Jonasson JG: Drug-induced cholestasis. Clin Liver Dis 2013; 17:191–209
160.
Lucena MI, Andrade RJ, Kaplowitz N, García-Cortes M, Fernández MC, Romero-Gomez M, Bruguera M, Hallal H, Robles-Diaz M, Rodriguez-González JF, Navarro JM, Salmeron J, Martinez-Odriozola P, Pérez-Alvarez R, Borraz Y, Hidalgo R; Spanish Group for the Study of Drug-Induced Liver Disease: Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and sex. Hepatology 2009; 49:2001–2009
161.
Gartlehner G, Gaynes BN, Hansen RA, Thieda P, DeVeaugh-Geiss A, Krebs EE, Moore CG, Morgan L, Lohr KN: Comparative benefits and harms of second-generation antidepressants: background paper for the American College of Physicians. Ann Intern Med 2008; 149:734–750
162.
Davern TJ, Chalasani N, Fontana RJ, Hayashi PH, Protiva P, Kleiner DE, Engle RE, Nguyen H, Emerson SU, Purcell RH, Tillmann HL, Gu J, Serrano J, Hoofnagle JH; Drug-Induced Liver Injury Network (DILIN): Acute hepatitis E infection accounts for some cases of suspected drug-induced liver injury. Gastroenterology 2011; 141:1665-72.e1–1665-72.e19
163.
Koponen H, Jokelainen J, Keinänen-Kiukaanniemi S, Kumpusalo E, Vanhala M: Metabolic syndrome predisposes to depressive symptoms: a population-based 7-year follow-up study. J Clin Psychiatry 2008; 69:178–182
164.
Pyykkönen AJ, Räikkönen K, Tuomi T, Eriksson JG, Groop L, Isomaa B: Association between depressive symptoms and metabolic syndrome is not explained by antidepressant medication: results from the PPP-Botnia Study. Ann Med 2012; 44:279–288

Information & Authors

Information

Published In

Go to American Journal of Psychiatry
Go to American Journal of Psychiatry
American Journal of Psychiatry
Pages: 404 - 415
PubMed: 24362450

History

Received: 31 May 2013
Revision received: 17 October 2013
Accepted: 25 October 2013
Published online: 1 April 2014
Published in print: April 2014

Authors

Affiliations

Cosmin Sebastian Voican, M.D., Ph.D.
From the Hepatogastroenterology Service, AP-HP (Public Assistance-Hospitals of Paris) Hôpital Antoine Béclère, DHU (university hospital department) Hépatinov, Clamart, France; INSERM (National Institute of Health and Medical Research) U996, IPSIT (Institut Paris-Sud d’Innovation Thérapeutique), Clamart; INSERM U669, CHU (university hospital center) de Bicêtre, Kremlin-Bicêtre, France; the Psychiatric Service, AP-HP Hôpital Bicêtre, Kremlin-Bicêtre; and the Faculty of Medicine, Université Paris-Sud, Kremlin-Bicêtre.
Emmanuelle Corruble, M.D., Ph.D.
From the Hepatogastroenterology Service, AP-HP (Public Assistance-Hospitals of Paris) Hôpital Antoine Béclère, DHU (university hospital department) Hépatinov, Clamart, France; INSERM (National Institute of Health and Medical Research) U996, IPSIT (Institut Paris-Sud d’Innovation Thérapeutique), Clamart; INSERM U669, CHU (university hospital center) de Bicêtre, Kremlin-Bicêtre, France; the Psychiatric Service, AP-HP Hôpital Bicêtre, Kremlin-Bicêtre; and the Faculty of Medicine, Université Paris-Sud, Kremlin-Bicêtre.
Sylvie Naveau, M.D., Ph.D.
From the Hepatogastroenterology Service, AP-HP (Public Assistance-Hospitals of Paris) Hôpital Antoine Béclère, DHU (university hospital department) Hépatinov, Clamart, France; INSERM (National Institute of Health and Medical Research) U996, IPSIT (Institut Paris-Sud d’Innovation Thérapeutique), Clamart; INSERM U669, CHU (university hospital center) de Bicêtre, Kremlin-Bicêtre, France; the Psychiatric Service, AP-HP Hôpital Bicêtre, Kremlin-Bicêtre; and the Faculty of Medicine, Université Paris-Sud, Kremlin-Bicêtre.
Gabriel Perlemuter, M.D., Ph.D.
From the Hepatogastroenterology Service, AP-HP (Public Assistance-Hospitals of Paris) Hôpital Antoine Béclère, DHU (university hospital department) Hépatinov, Clamart, France; INSERM (National Institute of Health and Medical Research) U996, IPSIT (Institut Paris-Sud d’Innovation Thérapeutique), Clamart; INSERM U669, CHU (university hospital center) de Bicêtre, Kremlin-Bicêtre, France; the Psychiatric Service, AP-HP Hôpital Bicêtre, Kremlin-Bicêtre; and the Faculty of Medicine, Université Paris-Sud, Kremlin-Bicêtre.

Notes

Address correspondence to Dr. Perlemuter ([email protected]).

Author Contributions

The first two authors contributed equally to this work.

Funding Information

Dr. Corruble has received consulting fees from AstraZeneca, Bristol-Myers Squibb, Eisai, Lundbeck, Otsuka, Sanofi, and Servier. Dr. Naveau has received travel funds from Janssen, Gilead, and Bristol-Myers Squibb. Dr. Perlemuter has received travel funds from Janssen, Gilead, and Roche, consulting fees from Bayer, Biocodex, Physiogenex, and Servier, and royalties from Elsevier-Masson. Dr. Voican reports no financial relationships with commercial interests.

Metrics & Citations

Metrics

Citations

Export Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format
Citation style
Style
Copy to clipboard

There are no citations for this item

View Options

View options

PDF/ePub

View PDF/ePub

Get Access

Login options

Already a subscriber? Access your subscription through your login credentials or your institution for full access to this article.

Personal login Institutional Login Open Athens login
Purchase Options

Purchase this article to access the full text.

PPV Articles - American Journal of Psychiatry

PPV Articles - American Journal of Psychiatry

Not a subscriber?

Subscribe Now / Learn More

PsychiatryOnline subscription options offer access to the DSM-5-TR® library, books, journals, CME, and patient resources. This all-in-one virtual library provides psychiatrists and mental health professionals with key resources for diagnosis, treatment, research, and professional development.

Need more help? PsychiatryOnline Customer Service may be reached by emailing [email protected] or by calling 800-368-5777 (in the U.S.) or 703-907-7322 (outside the U.S.).

Media

Figures

Other

Tables

Share

Share

Share article link

Share