ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Drugs and the liver: Metabolism and mechanisms of injury

Drugs and the liver: Metabolism and mechanisms of injury
Literature review current through: Jan 2024.
This topic last updated: Jun 07, 2023.

INTRODUCTION — Multiple drugs, both prescription and over-the-counter, herbal products, or toxins can cause hepatotoxicity through a variety of mechanisms [1,2]. A high index of suspicion is often necessary to expeditiously establish the diagnosis.

The hepatic metabolism of drugs and the mechanisms by which drugs might injure the liver will be reviewed here. The different clinical patterns of drug-induced hepatotoxicity and the use of medications in patients with liver disease are discussed separately. (See "Drug-induced liver injury" and "Overview of the management of chronic hepatitis C virus infection", section on 'Dose adjustments of medications' and "Cirrhosis in adults: Overview of complications, general management, and prognosis", section on 'General management'.)

EPIDEMIOLOGY — Drug-induced liver injury (DILI) and herbal-induced liver injury are well-recognized problems and symptomatically can mimic both acute and chronic liver diseases. Over 1000 medications and herbal products have been implicated in the development of DILI [3,4]. The probability of an individual drug causing liver injury ranges from 1 in 10,000 to 100,000, with some drugs reported as having an incidence of 100 in 100,000 (eg, chlorpromazine, isoniazid) [5,6]. DILI has a worldwide annual incidence ranging from 1.3 to 19.1 per 100,000 persons and 30 percent of cases will develop jaundice [6-16]. The prevalence and causes of DILI vary geographically [17]. DILI is the cause of acute jaundice in up to 50 percent of patients who present with new onset jaundice and accounts for 3 to 5 percent of hospital admissions for jaundice [8,9,13,18-23]. DILI accounts for up to half of the cases of acute liver failure in Western countries [24,25]. Finally, DILI was the most frequently cited reason for the withdrawal of medications from the marketplace (up to 32 percent) [26-33]. Following publication of the US Food and Drug Administration guidance statement for DILI in drug development, awareness of the issue increased and drug withdrawal subsequently decreased [6,34].

Database of drugs, herbs, and supplements associated with hepatotoxicity — The National Institutes of Health (NIH) maintains a searchable database of drugs, herbal medications, and dietary supplements that have been associated with DILI [35]. In addition, in 2004 the DILI Network initiated a prospective, observational, longitudinal study of individuals two years of age or older with suspected DILI [36]. The DILI Network continues to gather data and provide insight into the condition [37], and other DILI databases have been developed as well [30,38-40].

ROLE OF THE LIVER IN DRUG METABOLISM — The liver is responsible for the selective uptake, concentration, metabolism, and excretion of the majority of drugs and toxins that are introduced into the body. While some parent drugs can directly cause hepatotoxicity, it is generally the metabolites of these compounds that lead to drug-induced liver injury (DILI). These compounds are processed by a variety of soluble and membrane-bound enzymes, especially those related to the hepatocyte endoplasmic reticulum. Each drug has its specific enzyme disposal pathway(s) of biotransformation involving one or more of these enzyme systems. Genetic variation in drug metabolism is increasingly being recognized as a factor in the development of DILI [41]. Although less clear, environmental factors (eg, alcohol use) may also alter the processing of drugs and toxins.

The majority of drugs absorbed from the gastrointestinal tract are lipophilic and water-insoluble. They are rendered water-soluble via hepatic metabolism and thus, more easily excreted in the bile or renally filtered. Exogenous products are hepatically metabolized predominantly through two mechanisms: phase I and phase II reactions (figure 1) [42]. The subsequent products are then excreted via excretory transporters on either the canalicular or sinusoidal membranes (phase III reactions) [43].

Phase I reactions — Phase I reactions transform lipophilic molecules into more polar, hydrophilic molecules via oxidation, reduction, or hydrolysis. These reactions are catalyzed by the membrane-bound cytochrome P450 superfamily of mixed function oxidases (CYP) [44-47]. These hemoproteins are composed of an apoprotein and a heme prosthetic group (the oxidizing center).

Approximately 60 genes coding for CYP proteins have been identified in humans. These enzymes are organized into families (eg, CYP2) and subfamilies (eg, CYP2D), and further identified by the isoenzyme (eg, CYP2D6) [48,49]. The vast majority of these enzymes are located on the cytoplasmic side of the membrane of the endoplasmic reticulum (microsomal type) or the mitochondria (mitochondrial type) [48]. The microsomal type is responsible for phase I drug metabolism.

Hepatic metabolism of exogenous drugs and toxins is performed mainly by the CYP1, CYP2, and CYP3 families, with a smaller contribution from CYP4 [46,48,50,51]. Five isoenzymes are involved in 90 percent of all drug metabolism (CYP3A4, CYP2D6, CYP2C9, CYP2C19, and CYP1A2) [52,53]. The remaining families are often highly specific for the metabolism of endogenous compounds and are not inducible by exogenous compounds [48,54]. CYP3A is the major subfamily of hepatic CYP enzymes, and the most important drug metabolizing member is CYP3A4, comprising approximately 60 percent of all hepatic cytochromes and catalyzing the biotransformation of over 50 percent of commonly used drugs [48,55]. CYP2C9, 2C19, and 2D6 metabolize 37 percent of commonly used drugs [53]. Free radicals and toxic electrophilic compounds can be produced during this process. These reactive metabolites can then lead to cellular injury and are important to the development of idiosyncratic DILI [56,57].

CYP activity — Cytochrome activity varies considerably depending in part upon the concentration of the enzymes and the degree of induction by exogenous factors (table 1). (See 'Factors influencing drug metabolism and the risk of DILI' below.)

Factors that alter the activity of an enzyme have the potential to increase the toxicity of a compound (either by reducing its conversion to nontoxic metabolites or by increasing its conversion to toxic metabolites) or to decrease its therapeutic effectiveness (eg, by increasing the rate of metabolism of active drug).

In some cases, alternate detoxification routes may become overloaded, leading to the development of hepatotoxicity. This may in part explain why some drugs (such as acetaminophen) are not toxic in normal therapeutic doses but are toxic when increased amounts are ingested. (See "Acetaminophen (paracetamol) poisoning in adults: Pathophysiology, presentation, and evaluation" and "Acetaminophen (paracetamol) poisoning: Management in adults and children".)

Phase II reactions — Phase II reactions occur either directly with a parent compound (rare) or with a metabolite formed by a phase I reaction that is still not adequately hydrophilic for excretion. These reactions conjugate the drug or metabolic byproducts to highly polar ligands such as glucuronate, sulfate, acetate, glycine, glutathione, or a methyl group (figure 1). As a general rule, phase II reactions result in the formation of readily excreted, nontoxic substances [42,50]. Phase II reactions occur predominantly within the hepatocyte cytoplasm via the uridine diphospho (UDP)-glucuronyl transferases (UGT), sulfotransferases, and glutathione S-transferases [58]. The UGT1 and UGT2 families are the most important in human glucuronidation [59]. Conjugation ordinarily leads to a decrease in pharmacologic activity with enhanced clearance of the compound (eg, acetaminophen, furosemide, and bilirubin), and these enzymes are rarely responsible for toxic metabolite formation. However, exceptions do occur [59], particularly with some cancer chemotherapeutic medications (eg, irinotecan). In some cases, conjugation leads to increased pharmacologic activity. For example, glucuronidation of morphine leads to increased analgesic potency and sulfation of minoxidil is required for its antihypertensive effect.

Phase III reactions — Phase III reactions lead to the transport of drugs and drug products across the canalicular membranes and into the bile. The biliary transporters are members of the adenosine triphosphate-binding cassette (ABC) superfamily and include multidrug resistant P-glycoprotein 1 (MDR1/ABCB1), MDR3 (ABCB4), multidrug resistant protein 2 (MRP2/ABCC2), and the bile salt export protein (BSEP/ABCB11) [60,61]. The predominant role of these transporters is that of regulation of bile formation and the excretion of xenobiotics [62]. Altered activity of these transporters may lead to hepatotoxicity [62-65].

Factors influencing drug metabolism and the risk of DILI — Several factors may alter the activity of any of these drug-metabolizing reactions and influence drug metabolism, contributing to hepatotoxicity [66]. These factors potentially alter the level of exposure to toxic drug products.

Genetics — Numerous genetic polymorphisms in the CYP isoenzymes, human leukocyte antigens (HLA) alleles, and other drug-processing enzymes have been identified and associated with DILI [46,54,67-76]. Genetic alterations may contribute to diminished metabolism, lack of metabolism, or excessive metabolism of a compound [77]. CYP polymorphisms have been shown to have five metabolic phenotypes: poor metabolizers, intermediate metabolizers, normal metabolizers, rapid metabolizers, and ultra-rapid metabolizers [78]. This has been particularly well studied in the alcohol-metabolizing CYP2E1 subfamily and in the CYP2D6 subfamily, which are responsible for the metabolism of drugs such as metoprolol, quinidine, and desipramine [1,79]. This genetic variability explains some of the individual hypersensitivity reactions to specific drugs.

Genetic polymorphisms in the phase II and the phase III enzymes also lead to both decreased and increased activity. This is seen in glutathione S-transferase, N-acetyltransferase 2, and UDP-glucuronosyltransferase B7, and has been associated with development of idiosyncratic DILI [59,80-86]. Certain genetic variations in the hepatobiliary transporters (BSEP, MDR3) may also predispose to drug-induced cholestasis or injury [62-64,87]. As an example, variable drug pharmacokinetics have been reported with digoxin and cyclosporine depending upon which genetic variant of the phase III hepatobiliary enzyme was present [60].

Another factor influencing the individual response to drugs is genetic polymorphisms of the major histocompatibility molecules (HLA), several of which have been identified [75,88-92]. Certain HLA types may favor presentation of the offending drug. As an example, a predominance of HLA-DR6 is seen in hepatitis caused by chlorpromazine, and a predominance of HLA-A11 is seen in hepatitis from tricyclic antidepressants [93]. In one study, patients with HLA B*5701 had an 80-fold increased risk of hepatotoxicity due to flucloxacillin [89]. There is a strong association of hepatotoxicity with HLA DRB1*07 and the use of ximelagatran. Multiple HLA haplotypes have been associated with amoxicillin-clavulanate-induced DILI (HLA-A*0201, HLA-B*1801, DRB1*1501, DRB5*0101, and DQB1*0602) [14,71,88,89,91,94,95]. HLA-A*3301 has been associated with all causes of DILI, particularly from terbinafine, fenofibrate, and ticlopidine [41,96]. Cholestatic DILI is more common in those with HLA DRB1*15 and HLA DQB1*06 [91,97,98].

Idiosyncratic DILI is most often immune-mediated [99-101]. The regulation of the immune response is genetically determined, which may play a role in an individual's susceptibility to hepatotoxicity. In addition, genetic alterations in the hepatocytes themselves may contribute to risk of injury [102].

Alcohol ingestion — The importance of alcohol in the development of DILI remains controversial [103-105]. Chronic alcohol ingestion increases CYP2E1 and CYP4A activity, an effect that lasts for up to 10 days following ingestion [103,106-110]. Alcohol also inhibits the rate of glutathione synthesis, leading to accelerated glutathione turnover, and it impairs mitochondrial transport of glutathione (with sequestration within the mitochondria) [103,106,111]. In a cohort study including 300 patients, any alcohol use within 12 months prior to the onset of DILI was a negative predictor of severe DILI [112]. Drugs that have been reported to have increased hepatotoxicity when associated with alcohol intake are acetaminophen, isoniazid, cocaine, methotrexate, and vitamin A [113].

Nutrition — Induction of CYP enzymes, particularly CYP1A2, has been observed with the ingestion of Brussels sprouts, cabbage, cruciferous vegetables (such as broccoli), and charcoal-broiled beef [48,114-116]. In contrast, grapefruit juice inhibits CYP3A activity, primarily acting on the intestinal form of the enzyme [117-119]. An excellent example is the interaction of grapefruit juice with the immunosuppressive agents cyclosporine or tacrolimus.

CYP activity may be increased by high-protein diets and reduced by low-protein diets and severe malnutrition [120-123]. Thus, states of severe malnutrition or chronic alcohol use disorder with poor nutrition may influence certain detoxifying cofactors such as glutathione.

Presence of other drugs — The concomitant use of two or more drugs may be one of the most important factors affecting components of the CYP system and influencing drug metabolism. A drug may act either as a cytochrome P450 inhibitor and slow another drug's metabolism, or it may induce CYP450 and enhance another drug's metabolism [48,53,124-126]. The adage that "alcohol and drugs do not mix" is based in part upon this phenomenon.

The list of drug interactions with the CYP system is vast. Even the aryl hydrocarbons in cigarette smoke can induce CYP1A2 [127]. Competitive inhibition of CYP can lead to clinically important drug interactions which are most pronounced when there is no alternative pathway for the metabolism of a potentially toxic drug or its metabolite. A striking example of this is the development of torsade de pointes during the administration of terfenadine or cisapride (both no longer available in the United States) to a patient taking a CYP3A4 inhibitor such as erythromycin or ketoconazole (table 1).

Drug-induced induction and inhibition of phase II enzymes are not uniformly seen [59]. However, reduced phase II reactions have been described with chlorpromazine and valproate use. There are many reports of both inhibition (eg, atorvastatin, carvedilol, clarithromycin, sertraline) and induction (eg, amiodarone, diltiazem, erythromycin, St. John's wort) of the phase III transporters [61]. These drug-drug interactions will significantly alter the transport and secretion activity of transport enzymes.

Patient demographics — An overall decrease in CYP activity has been reported to develop with increasing age [128-130]. Additionally, conjugation (phase II) reactions have been showed to be reduced in frail older adults, although this is not consistently seen [131].

It is therefore possible that older age increases the risk of developing DILI, but this is not uniformly supported by the data and appears to be drug-specific [11,12,97,132-134]. There still remain many interindividual differences that affect susceptibility, including genetic polymorphisms, biologic sex, volume of distribution, underlying liver disease, and nutritional status [134]. Older individuals do appear to have greater risk of toxicity from amoxicillin-clavulanate, isoniazid, and nitrofurantoin [12,135-137]. Older individuals are also more likely to develop cholestatic, rather than hepatocellular, DILI [12,97,132].

Infants may show considerable immaturity of the supply of drug-metabolizing CYP enzymes, which may be low to undetectable at birth and develop over time [138]. There are also major differences in the activity of the phase II enzymes in children when compared with adults [138]. Examples of medications that appear to have a greater risk of hepatotoxicity in children include acetylsalicylic acid and valproate [66]. There are no studies of the effects of aging on the hepatic phase III enzymes [129].

Women have been thought to be more susceptible to DILI than men [139]. Differences between men and women include a higher glucuronidation rate of acetaminophen in men and a greater expression of CYP3A4 in women [140,141]. Not all studies support a female preponderance and the increased susceptibility is most likely drug-specific. Drugs associated with DILI in woman include nitrofurantoin, erythromycin, flucloxacillin, minocycline, and isoniazid [96,142].

Racial differences in DILI presentation and prognosis have been reported. In one study of nearly 1000 patients with DILI, African American patients were more likely to have severe cutaneous reactions, severe liver injury, and worse outcomes (liver transplantation or liver-related death) compared with White patients [143]. This is attributed predominantly to variations in single nucleotide polymorphisms in different ethnic groups. A meta-analysis in patients treated for tuberculosis showed that in persons of East Asian and Middle Eastern descent, slow NAT2 genotype was associated with increased risk of DILI [144].

Underlying liver disease — Underlying liver disease and its role in DILI remains debated. Both acute and chronic liver diseases have a variable effect on the metabolism of many drugs [137,145]. Enzyme activities are generally reduced with increasing liver disease severity [145]. As a result, CYP activity may be increased (rare), unaltered, or in most cases reduced, or greatly reduced depending upon the severity of liver dysfunction [145,146]. The type of liver disease does not appear to be important. Reduction in the CYP activity leads to elevated plasma drug concentrations and increased risk of adverse drug reaction.

Phase II enzyme activity appears to be altered to a lesser degree [145,147,148]. The presence of cholestasis leads to decreased secretion (phase III) of both endogenous and exogenous substances [145]. This is seen not only in the setting of intrahepatic cholestasis but also secondary to biliary obstruction.

There has been considerable concern about the safety of statins in the setting of chronic liver disease. There are several trials that have confirmed their safety in this patient population [12,149-152]. However, there is more variation in serum levels of statins in the setting of cirrhosis; therefore, the drugs should be used more cautiously in this group [151]. (See "Statins: Actions, side effects, and administration", section on 'Chronic liver disease'.)

There is a growing body of evidence that nonalcohol-associated fatty liver disease may increase the risk or severity of DILI [153,154].

The data regarding the use of antituberculosis agents or antiretroviral agents used to treat HIV are more conflicting [66,149,150,155-157]. (See "Overview of antiretroviral agents used to treat HIV".)

Although patients with advanced liver disease are not generally felt to be at increased risk for all-cause DILI, those who develop DILI are more likely to have a worse outcome due to decreased functional hepatic reserve with mortality of 16 percent compared with 5.2 percent in those without chronic liver disease [11,12].

Dose — Liver damage from intrinsic hepatotoxins is clearly dose-related. While it has been thought that idiosyncratic DILI is dose-independent, this may not be entirely true [158-160]. There appears to be a threshold dose that may be necessary in most settings. Severe DILI rarely occurs from drugs taken at doses less than 10 mg, and drugs administered at daily doses of ≥50 mg are significantly more likely to cause DILI [25,66,97,99,126,160-165]. In addition, compounds are more likely to cause DILI if they are extensively hepatically metabolized when compared with drugs that have less extensive metabolism [160,161,166].

Drug lipophilicity — It has been shown that the lipophilicity of a drug/toxin is associated with increased risk for DILI [165,167]. The lipophilic nature of a drug affects its absorption, distribution, metabolism, and excretion. Medications at doses over 100 mg with high lipophilicity are significantly associated with severe DILI with an odds ratio of 14 [135,165,167].

MECHANISMS OF DRUG-INDUCED HEPATOTOXICITY — Drug-induced liver injury (DILI) is a complicated process involving the parent drug, its metabolites, and the host immune system [158]. Most hepatotoxic drug effects lead to hepatocyte necrosis or apoptosis and subsequent cell death. However, some drugs predominantly damage the bile ducts, biliary export proteins or bile canaliculi (cholestasis), vascular endothelial cells (sinusoidal obstruction syndrome), or the stellate cells. There may also be mixed patterns of injury (algorithm 1) [62,168-170]. (See "Drug-induced liver injury".)

Toxic hepatocellular injury may be classified by either clinical features (pattern of liver injury) or the underlying pathogenic mechanism (intrinsic hepatotoxins versus idiosyncratic reactions) (table 2). For the purposes of this review, we will use the pathogenic classification.

Direct Hepatotoxicity — Direct hepatotoxicity is caused by intrinsic hepatotoxins which reproducibly and predictably lead to dose-dependent hepatocellular necrosis ("toxic" hepatitis) in most mammalian species (algorithm 1). The latent period between the exposure and onset of hepatotoxicity is generally brief (ie, hours to a few days) and fairly consistent from person to person and among animal models. In addition to acute hepatitis, direct hepatotoxicity can lead to other forms of liver injury, including nodular regenerative hyperplasia, lactic acidosis and sinusoidal obstruction syndrome. (See "Drug-induced liver injury".)

In most instances, the drug itself or one of its active metabolites interacts with one or more cellular constituents (eg, proteins, lipids, DNA) to produce a sequence of events that often results in cell death [171]. Production of an active metabolite often yields free radicals, electrophilic radicals, or reactive oxygen species. Alternatively, covalent binding of the toxic metabolite to structures within the cell may interfere with their function or their regulation. These events may incite oxidative stress, glutathione depletion, redox changes, and/or lipid peroxidation, which may further affect cellular functions (eg, mitochondria, endoplasmic reticulum, cytoskeleton, etc) [43,172,173].

Drugs known to be intrinsically hepatotoxic are often removed from clinical use by regulatory agencies or fail to make it into the marketplace [30]. Examples include carbon tetrachloride, chloroform, and tannic acid [11]. However, some drugs with intrinsic hepatotoxic potential are still used clinically, either because they show hepatotoxicity only in large doses (eg, acetaminophen, niacin, iron sulfate), or because they show known dose-related toxicity (eg, ethanol, intravenous tetracycline, L-asparaginase, and phosphorus) [171]. Other direct hepatotoxins occur naturally, such as mushrooms (Amanita phalloides). (See "Acetaminophen (paracetamol) poisoning in adults: Pathophysiology, presentation, and evaluation" and "Acetaminophen (paracetamol) poisoning: Management in adults and children" and "Hepatotoxicity associated with chronic low-dose methotrexate for nonmalignant disease".)

Acute hepatitis — The most common type of injury is acute hepatitis. Serum aminotransferases are typically 8 to 500 times normal. Serum alkaline phosphatase levels are only mildly elevated and jaundice is less common [113]. The risk of mortality is high in severe cases. Although hepatocellular damage may be the primary effect, some of these compounds can also damage other organs (especially the kidneys). (See "Acetaminophen (paracetamol) poisoning in adults: Pathophysiology, presentation, and evaluation", section on 'Acute kidney injury (acute renal failure)'.)

The liver injury generally resolves when the drug is discontinued, or the dosage is decreased. Alternatively, adaptation can develop in which the injury resolves spontaneously despite continuation of the medication [174]. Severe cases will worsen despite drug discontinuation. This progression leads to acute liver failure, often necessitating liver transplantation [25,27].

Nodular regenerative hyperplasia — Nodular regenerative hyperplasia has been linked to chemotherapeutic agents and first-generation nucleoside antiretroviral agents. Its pathogenesis is poorly understood. However, it may be linked to chronic injury to the hepatic microvasculature [174].

Sinusoidal obstruction syndrome — Sinusoidal obstruction syndrome (SOS) was previously known as veno-occlusive disease. SOS develops following injury to the sinusoidal endothelial cells leading to extravasation of red cells and nonthrombotic obstruction of the sinusoids and central veins [175,176]. The most common drug-related cause of SOS is myeloablative-conditioning therapy in hematopoietic stem cell transplant. It can also be seen with immunosuppressive drugs (eg, azathioprine) or pyrrolizidine alkaloids [177].

Idiosyncratic hepatotoxicity — The majority of cases of DILI are related to idiosyncratic reactions (eg, amoxicillin-clavulanate, isoniazid, diclofenac, trimethoprim-sulfamethoxazole) [12]. These reactions occur in about 0.01 to 1 percent of individuals who are taking the drug (<1 to 100 of every 10,000 individuals exposed) [43,172,178]. Less than 1 percent of these drugs were associated with a predicted risk of developing DILI in preclinical or clinical trials [179]. The principal characteristic of this type of reaction is the apparent unpredictability of the liver injury in humans. The reactions tend to be species-specific and often cannot be reproduced experimentally in laboratory animals [172,173,178,180]. The latent period between exposure to the drug and the sensitivity reaction is highly variable; it may be one to three months, but reactions have been reported up to a year after starting the medication [101]. A threshold dose may be required. (See 'Dose' above.)

The individual's response to the drug/metabolite also contributes to the risk of DILI [165]. Although the parent compound may directly lead to idiosyncratic DILI, it is most often a toxic metabolite that is at fault [172,181]. Idiosyncratic drug toxicity may be either nonimmune (metabolic) or immune (allergic); however, overlap is believed to exist and leads to cellular injury [43,173,178].

In nonimmune injury, covalent binding of drug metabolites to cellular structures can incite cell death by interfering with protein/enzyme activity and disrupting critical cellular function. Immune-mediated idiosyncratic reactions depend upon a complex interaction of the drug and its metabolites with the host immune system, with resultant downstream effects that cause hepatocyte necrosis and/or apoptosis and unleash cytokines that can lead to secondary cell damage or have immune-modulating effects [43,100]. A comprehensive review of the biochemical and molecular mechanism of idiosyncratic DILI is beyond the scope of this review, and the reader is referred elsewhere [6].

Nonimmune — Nonimmune DILI is probably due to genetically determined aberrant metabolism of the drug in susceptible patients. The duration of exposure before the development of toxicity varies from weeks to one year, and reactions can develop several weeks after drug discontinuation. The injury may not recur or may recur many days to weeks after rechallenge. Features of an allergic or hypersensitivity reaction are absent. Drugs in this category include amiodarone, diclofenac, disulfiram, isoniazid, ketoconazole, troglitazone, and valproate. (See "Isoniazid hepatotoxicity" and "Amiodarone: Adverse effects, potential toxicities, and approach to monitoring".)

Local accumulation of toxic metabolites (electrophilic species) results in covalent binding of the metabolite to cellular proteins, enzymes, lipids, and DNA. The hepatocyte undergoes significant oxidative stress, leading to redox changes and lipid peroxidation, which results in cellular necrosis [43,113]. As cellular stress worsens, the function of cellular structures such as mitochondria, receptor signaling, microtubules, and the endoplasmic reticulum is directly affected [172,173,182]. Drugs may also inhibit bile acid amidation and alter regulation of hepatic transporters (eg, bile salt export pump) that actively pump drug metabolites out of the hepatocytes [11,183,184]. This further increases the oxidative stress within the cell. Immunologic injury may play some role in metabolic DILI. Dead and dying hepatocytes can release endogenous damage-associated molecular patterns (DAMPs) [185]. DAMPs are recognized by the immune system, triggering or worsening sterile inflammation [185]. Neoantigens may also be formed by the reaction of the metabolite with the hepatocyte, leading to activation of the immune system [186].

Immune-mediated — Immune-mediated DILI is the least well understood form of DILI. It may be accompanied by clinical and histologic evidence of classic hypersensitivity. The duration of exposure is generally about one to eight weeks [172]. Rash, fever, joint pain and inflammation, lymphadenopathy, eosinophilic leukocytosis and, in severe cases, Stevens-Johnson syndrome or toxic epidermal necrolysis may occur. However, these symptoms may be mild or absent. In some cases, the presentation may be similar to infectious mononucleosis (with atypical lymphocytes) (see "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis"). There is a prompt recurrence of symptoms in response to drug rechallenge of one or two doses [11,113,168]. This type of injury is believed to be a metabolite-initiated, immune-mediated attack on the liver [43]. Drugs that have been associated with this type of injury include amoxicillin-clavulanate, diclofenac, phenytoin, and allopurinol [142].

While cellular stress can activate an immune reaction to a drug, the drug can also directly lead to an immune response. The initial factor appears to be the modification of "self" proteins due to covalent binding of the active metabolite with host tissues (haptenization). These drug-protein products (adducts) can then cause direct toxicity or behave as neoantigens and trigger immune-mediated reactions [43,187]. The neoantigens are then presented by major histocompatibility complex class II, which leads to the activation of helper T-lymphocytes that subsequently recruit cytotoxic T-lymphocytes, natural killer cells, and B-cells [43,99,168,178,181]. Cytotoxic T-lymphocytes then mediate apoptosis via Fas ligand and granzyme B/porin [178]. The cells develop oxidative stress, which leads to the formation of reactive oxygen species that damage cellular DNA, proteins, and lipids [188]. This further induces immune-mediated liver damage. Mitochondrial dysfunction is seen and plays a pivotal role in the pathogenesis of DILI [164,170,188,189]. The drug or its reactive metabolite can also directly noncovalently bind to HLA alleles leading to immune system activation [190].

Haptenization alone does not appear to trigger immune-mediated responses, and the danger hypothesis has been proposed as an explanation for further injury. This hypothesis states that for the immune-mediated attack to develop, a second costimulatory factor must be present [99,191]. These factors appear to prime the immune system in susceptible individuals and may include infection or inflammatory conditions (eg, hepatitis B virus, hepatitis C virus, HIV infection), hepatocyte stress, or cellular damage (eg, DAMPs, heat shock proteins, S100 proteins) [43,185,192-194].

Neoantigens also prompt the development of autoantibodies against the cytochromes P450 (CYP), which can be identified in the serum, although it is unclear if these participate in the immune attack [43,172,187,195]. These autoantibodies are generally specific to the cytochrome that metabolizes the offending drug. The unique immunologic response of an individual to a drug and the specific T-cell receptor range may explain why hepatotoxicity occurs in some but not all individuals.

Both the innate/adaptive immune systems participate. Hepatocyte necrosis activates the innate system through proinflammatory mediators (eg, cytokines, chemokines, death signal pathways, etc) [187,196]. These mediators can be directly cytotoxic or lead to recruitment of cells of the innate immune system. The innate immune system may also have a role in recovery via anti-inflammatory components [43,187].

Idiosyncratic drug reactions of this type are becoming more frequently recognized, and the list of drugs implicated is sizable (eg, abacavir, nitrofurantoin, phenytoin, amoxicillin-clavulanate, dihydralazine, sulfonamides, minocycline, halothane, dapsone, diclofenac, carbamazepine, and sulindac) [11,197,198]. Liver biopsy often reveals eosinophilic or granulomatous inflammation of the liver with hepatocyte necrosis and cholestasis [199,200]. (See "Sulfonamide allergy in HIV-uninfected patients".)

Drug-induced autoimmune-like hepatitis — DILI can also present as an autoimmune-like hepatitis (DI-AIH) [142]. These patients generally have elevated levels of gamma globulins, antinuclear antibodies, and/or antismooth muscle antibodies. Drugs that have been associated with this pattern of injury include minocycline, nitrofurantoin, fluoroquinolones, and antitumor necrosis factor alpha inhibitors [201-204]. It can be difficult to differentiate DI-AIH from classic autoimmune hepatitis. However, it is imperative to distinguish DI-AIH from idiosyncratic DILI because DI-AIH responds to corticosteroid therapy. In addition, once it has resolved, there is no relapse after withdrawal of treatment [142]. This feature helps to differentiate it from classic AIH.

Indirect hepatotoxicity — Indirect hepatotoxicity has been proposed as a mechanism of drug injury in which the action of the drug leads to the hepatoxicity [174,205]. The drug induces or exacerbates liver disease. This type of injury is not dose-dependent and has a delayed latency (ie, generally months). The types of injury seen include fatty liver, acute hepatitis, immune-mediated hepatitis, and chronic hepatitis [174].

Examples of this would be the de novo development of fatty liver or its worsening with tamoxifen [206]. Glucocorticoids can lead to worsening of fatty liver secondary to their effect on insulin sensitivity. Antineoplastic agents can induce reactivation or worsening of viral hepatitis [207]. An immune-mediated response can be seen with tumor necrosis factor inhibitors, protein kinase inhibitors, and newer monoclonal antibodies (ie, checkpoint inhibitors) [203,208,209].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Drug-induced liver injury".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Drug-induced hepatitis (The Basics)")

SUMMARY

Drug-induced liver injury (DILI) is a well-recognized problem and symptomatically can mimic acute and chronic liver diseases. Most patients with DILI will recover following withdrawal of the offending agent; however, in some cases, DILI can lead to liver transplantation or liver-related death. (See 'Epidemiology' above.)

The liver is exposed to nearly all drugs absorbed from the intestinal tract and is the predominant site of drug metabolism. (See 'Role of the liver in drug metabolism' above.)

Reactive drug metabolites, genetic susceptibility, and environmental factors all appear to play a role in development of DILI. (See 'Mechanisms of drug-induced hepatotoxicity' above.)

Idiosyncratic hepatotoxicity is the most common type of DILI seen, accounting for >90 percent of cases. However, our understanding of the pathogenesis of DILI remains incomplete. (See 'Idiosyncratic hepatotoxicity' above.)

A searchable database of drugs, herbal medications, and dietary supplements has been developed by the National Institutes of Health (NIH).

  1. Lee WM. Drug-induced hepatotoxicity. N Engl J Med 1995; 333:1118.
  2. Chang CY, Schiano TD. Review article: drug hepatotoxicity. Aliment Pharmacol Ther 2007; 25:1135.
  3. Suzuki A, Andrade RJ, Bjornsson E, et al. Drugs associated with hepatotoxicity and their reporting frequency of liver adverse events in VigiBase: unified list based on international collaborative work. Drug Saf 2010; 33:503.
  4. Stirnimann G, Kessebohm K, Lauterburg B. Liver injury caused by drugs: an update. Swiss Med Wkly 2010; 140:w13080.
  5. Haque T, Sasatomi E, Hayashi PH. Drug-Induced Liver Injury: Pattern Recognition and Future Directions. Gut Liver 2016; 10:27.
  6. Garcia-Cortes M, Robles-Diaz M, Stephens C, et al. Drug induced liver injury: an update. Arch Toxicol 2020; 94:3381.
  7. Shapiro MA, Lewis JH. Causality assessment of drug-induced hepatotoxicity: promises and pitfalls. Clin Liver Dis 2007; 11:477.
  8. Sgro C, Clinard F, Ouazir K, et al. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology 2002; 36:451.
  9. Larrey D. Epidemiology and individual susceptibility to adverse drug reactions affecting the liver. Semin Liver Dis 2002; 22:145.
  10. Bell LN, Chalasani N. Epidemiology of idiosyncratic drug-induced liver injury. Semin Liver Dis 2009; 29:337.
  11. Björnsson ES, Bergmann OM, Björnsson HK, et al. Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology 2013; 144:1419.
  12. Chalasani N, Bonkovsky HL, Fontana R, et al. Features and Outcomes of 899 Patients With Drug-Induced Liver Injury: The DILIN Prospective Study. Gastroenterology 2015; 148:1340.
  13. Hussaini SH, O'Brien CS, Despott EJ, Dalton HR. Antibiotic therapy: a major cause of drug-induced jaundice in southwest England. Eur J Gastroenterol Hepatol 2007; 19:15.
  14. Ibáñez L, Pérez 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.
  15. Suk KT, Kim DJ, Kim CH, et al. A prospective nationwide study of drug-induced liver injury in Korea. Am J Gastroenterol 2012; 107:1380.
  16. Vega M, Verma M, Beswick D, et al. The Incidence of Drug- and Herbal and Dietary Supplement-Induced Liver Injury: Preliminary Findings from Gastroenterologist-Based Surveillance in the Population of the State of Delaware. Drug Saf 2017; 40:783.
  17. Hassan A, Fontana RJ. The diagnosis and management of idiosyncratic drug-induced liver injury. Liver Int 2019; 39:31.
  18. Larrey D. Drug-induced liver diseases. J Hepatol 2000; 32:77.
  19. Norris W, Paredes AH, Lewis JH. Drug-induced liver injury in 2007. Curr Opin Gastroenterol 2008; 24:287.
  20. 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.
  21. Akhtar AJ, Shaheen M. Jaundice in African-American and Hispanic patients with AIDS. J Natl Med Assoc 2007; 99:1381.
  22. 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.
  23. Wai CT, Tan BH, Chan CL, et al. Drug-induced liver injury at an Asian center: a prospective study. Liver Int 2007; 27:465.
  24. Wei G, Bergquist A, Broomé U, et al. Acute liver failure in Sweden: etiology and outcome. J Intern Med 2007; 262:393.
  25. Reuben A, Koch DG, Lee WM, Acute Liver Failure Study Group. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology 2010; 52:2065.
  26. Ostapowicz G, Fontana RJ, Schiødt 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.
  27. Larson AM, Polson J, Fontana RJ, et al. Acetaminophen-induced acute liver failure: results of a United States multicenter, prospective study. Hepatology 2005; 42:1364.
  28. Regev A. How to avoid being surprised by hepatotoxicity at the final stages of drug development and approval. Clin Liver Dis 2013; 17:749.
  29. Watkins PB. Drug safety sciences and the bottleneck in drug development. Clin Pharmacol Ther 2011; 89:788.
  30. Chen M, Zhang J, Wang Y, et al. The liver toxicity knowledge base: a systems approach to a complex end point. Clin Pharmacol Ther 2013; 93:409.
  31. Wysowski DK, Swartz L. Adverse drug event surveillance and drug withdrawals in the United States, 1969-2002: the importance of reporting suspected reactions. Arch Intern Med 2005; 165:1363.
  32. Babai S, Auclert L, Le-Louët H. Safety data and withdrawal of hepatotoxic drugs. Therapie 2021; 76:715.
  33. Stevens JL, Baker TK. The future of drug safety testing: expanding the view and narrowing the focus. Drug Discov Today 2009; 14:162.
  34. Drug-Induced Liver Injury: Premarketing Clinical Evaluation. US Food and Drug Administration. Available at: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/drug-induced-liver-injury-premarketing-clinical-evaluation (Accessed on March 16, 2021).
  35. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. Available at: http://www.ncbi.nlm.nih.gov/books/NBK547852/ (Accessed on July 29, 2023).
  36. Fontana RJ, Watkins PB, Bonkovsky HL, et al. Drug-Induced Liver Injury Network (DILIN) prospective study: rationale, design and conduct. Drug Saf 2009; 32:55.
  37. Drug-Induced Liver Injury Network. Available at: http://www.dilin.org (Accessed on March 16, 2021).
  38. Igarashi Y, Nakatsu N, Yamashita T, et al. Open TG-GATEs: a large-scale toxicogenomics database. Nucleic Acids Res 2015; 43:D921.
  39. Xing L, Wu L, Liu Y, et al. LTMap: a web server for assessing the potential liver toxicity by genome-wide transcriptional expression data. J Appl Toxicol 2014; 34:805.
  40. Mao Y. Hepatox: a professional web platform for the study of clinical and translational research on drug-induced liver injury in China. Chin Hepatol 2014; 19.
  41. Nicoletti P, Aithal GP, Bjornsson ES, et al. Association of Liver Injury From Specific Drugs, or Groups of Drugs, With Polymorphisms in HLA and Other Genes in a Genome-Wide Association Study. Gastroenterology 2017; 152:1078.
  42. Park BK, Pirmohamed M, Kitteringham NR. The role of cytochrome P450 enzymes in hepatic and extrahepatic human drug toxicity. Pharmacol Ther 1995; 68:385.
  43. Gunawan BK, Kaplowitz N. Mechanisms of drug-induced liver disease. Clin Liver Dis 2007; 11:459.
  44. Rendic S, Guengerich FP. Survey of Human Oxidoreductases and Cytochrome P450 Enzymes Involved in the Metabolism of Xenobiotic and Natural Chemicals. Chem Res Toxicol 2015; 28:38.
  45. Guengerich FP, Waterman MR, Egli M. Recent Structural Insights into Cytochrome P450 Function. Trends Pharmacol Sci 2016; 37:625.
  46. Ingelman-Sundberg M. Human drug metabolising cytochrome P450 enzymes: properties and polymorphisms. Naunyn Schmiedebergs Arch Pharmacol 2004; 369:89.
  47. Werck-Reichhart D, Feyereisen R. Cytochromes P450: a success story. Genome Biol 2000; 1:REVIEWS3003.
  48. Danielson PB. The cytochrome P450 superfamily: biochemistry, evolution and drug metabolism in humans. Curr Drug Metab 2002; 3:561.
  49. Al Omari A, Murry DJ. Pharmacogenetics of the cytochrome P450 enzyme system: review of current knowledge and clinical significance. J Pharm Pract 2007; 20.
  50. Murray M. Mechanisms and significance of inhibitory drug interactions involving cytochrome P450 enzymes (review). Int J Mol Med 1999; 3:227.
  51. Lehmann DE. Enzymatic shunting: resolving the acetaminophen-warfarin controversy. Pharmacotherapy 2000; 20:1464.
  52. Pinto N, Dolan ME. Clinically relevant genetic variations in drug metabolizing enzymes. Curr Drug Metab 2011; 12:487.
  53. Sychev DA, Ashraf GM, Svistunov AA, et al. The cytochrome P450 isoenzyme and some new opportunities for the prediction of negative drug interaction in vivo. Drug Des Devel Ther 2018; 12:1147.
  54. DeLeve LD, Kaplowitz N. Mechanisms of drug-induced liver disease. Gastroenterol Clin North Am 1995; 24:787.
  55. Goldstein I, Rivlin N, Shoshana OY, et al. Chemotherapeutic agents induce the expression and activity of their clearing enzyme CYP3A4 by activating p53. Carcinogenesis 2013; 34:190.
  56. Stachulski AV, Baillie TA, Park BK, et al. The generation, detection, and effects of reactive drug metabolites. Med Res Rev 2013; 33:985.
  57. Leone A, Nie A, Brandon Parker J, et al. Oxidative stress/reactive metabolite gene expression signature in rat liver detects idiosyncratic hepatotoxicants. Toxicol Appl Pharmacol 2014; 275:189.
  58. Sugatani J. Function, genetic polymorphism, and transcriptional regulation of human UDP-glucuronosyltransferase (UGT) 1A1. Drug Metab Pharmacokinet 2013; 28:83.
  59. Sanchez-Dominguez CN, Gallardo-Blanco HL, Salinas-Santander MA, Ortiz-Lopez R. Uridine 5'-diphospho-glucronosyltrasferase: Its role in pharmacogenomics and human disease. Exp Ther Med 2018; 16:3.
  60. Zhou SF. Structure, function and regulation of P-glycoprotein and its clinical relevance in drug disposition. Xenobiotica 2008; 38:802.
  61. Dean M. The human ATP-binding cassette (ABC) transporter superfamily. National Center for Biotechnology Information (NCBI), National Library of Medicine, Bethesda, MD, 2002.
  62. Pauli-Magnus C, Meier PJ. Hepatobiliary transporters and drug-induced cholestasis. Hepatology 2006; 44:778.
  63. 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.
  64. Choi JH, Ahn BM, Yi J, et al. MRP2 haplotypes confer differential susceptibility to toxic liver injury. Pharmacogenet Genomics 2007; 17:403.
  65. Ulzurrun E, Stephens C, Crespo E, et al. Role of chemical structures and the 1331T>C bile salt export pump polymorphism in idiosyncratic drug-induced liver injury. Liver Int 2013; 33:1378.
  66. Chalasani N, Björnsson E. Risk factors for idiosyncratic drug-induced liver injury. Gastroenterology 2010; 138:2246.
  67. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome P450 enzymes and its clinical impact. Drug Metab Rev 2009; 41:89.
  68. Hasler JA. Pharmacogenetics of cytochromes P450. Mol Aspects Med 1999; 20:12.
  69. Dorne JL, Walton K, Renwick AG. Human variability in xenobiotic metabolism and pathway-related uncertainty factors for chemical risk assessment: a review. Food Chem Toxicol 2005; 43:203.
  70. Pachkoria K, Lucena MI, Molokhia M, et al. Genetic and molecular factors in drug-induced liver injury: a review. Curr Drug Saf 2007; 2:97.
  71. Daly AK, Day CP. Genetic association studies in drug-induced liver injury. Semin Liver Dis 2009; 29:400.
  72. Vuilleumier N, Rossier MF, Chiappe A, et al. CYP2E1 genotype and isoniazid-induced hepatotoxicity in patients treated for latent tuberculosis. Eur J Clin Pharmacol 2006; 62:423.
  73. 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.
  74. Aithal GP, Ramsay L, Daly AK, et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology 2004; 39:1430.
  75. Kaliyaperumal K, Grove JI, Delahay RM, et al. Pharmacogenomics of drug-induced liver injury (DILI): Molecular biology to clinical applications. J Hepatol 2018; 69:948.
  76. Meitei HN, Pandey A, Haobam R. Polymorphisms in drug metabolism genes as a risk factor for first-line anti-tuberculosis drug-induced liver injury. Mol Biol Rep 2023; 50:2893.
  77. Ueshima Y, Tsutsumi M, Takase S, et al. Acetaminophen metabolism in patients with different cytochrome P-4502E1 genotypes. Alcohol Clin Exp Res 1996; 20:25A.
  78. Song Y, Li C, Liu G, et al. Drug-Metabolizing Cytochrome P450 Enzymes Have Multifarious Influences on Treatment Outcomes. Clin Pharmacokinet 2021; 60:585.
  79. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 2013; 138:103.
  80. Ohno M, Yamaguchi I, Yamamoto I, et al. Slow N-acetyltransferase 2 genotype affects the incidence of isoniazid and rifampicin-induced hepatotoxicity. Int J Tuberc Lung Dis 2000; 4:256.
  81. Cho HJ, Koh WJ, Ryu YJ, et al. Genetic polymorphisms of NAT2 and CYP2E1 associated with antituberculosis drug-induced hepatotoxicity in Korean patients with pulmonary tuberculosis. Tuberculosis (Edinb) 2007; 87:551.
  82. Possuelo LG, Castelan JA, de Brito TC, et al. Association of slow N-acetyltransferase 2 profile and anti-TB drug-induced hepatotoxicity in patients from Southern Brazil. Eur J Clin Pharmacol 2008; 64:673.
  83. Roy B, Chowdhury A, Kundu S, et al. Increased risk of antituberculosis drug-induced hepatotoxicity in individuals with glutathione S-transferase M1 'null' mutation. J Gastroenterol Hepatol 2001; 16:1033.
  84. Leiro V, Fernández-Villar A, Valverde D, et al. Influence of glutathione S-transferase M1 and T1 homozygous null mutations on the risk of antituberculosis drug-induced hepatotoxicity in a Caucasian population. Liver Int 2008; 28:835.
  85. Lucena MI, Andrade RJ, Martínez C, et al. Glutathione S-transferase m1 and t1 null genotypes increase susceptibility to idiosyncratic drug-induced liver injury. Hepatology 2008; 48:588.
  86. Du H, Chen X, Fang Y, et al. Slow N-acetyltransferase 2 genotype contributes to anti-tuberculosis drug-induced hepatotoxicity: a meta-analysis. Mol Biol Rep 2013; 40:3591.
  87. 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.
  88. 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.
  89. 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.
  90. Larrey D, Pageaux GP. Genetic predisposition to drug-induced hepatotoxicity. J Hepatol 1997; 26 Suppl 2:12.
  91. 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.
  92. Fan WL, Shiao MS, Hui RC, et al. HLA Association with Drug-Induced Adverse Reactions. J Immunol Res 2017; 2017:3186328.
  93. Berson A, Fréneaux 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.
  94. Stephens C, López-Nevot MÁ, Ruiz-Cabello F, et al. HLA alleles influence the clinical signature of amoxicillin-clavulanate hepatotoxicity. PLoS One 2013; 8:e68111.
  95. Lucena MI, Molokhia M, Shen Y, et al. Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011; 141:338.
  96. García-Cortés M, Ortega-Alonso A, Lucena MI, et al. Drug-induced liver injury: a safety review. Expert Opin Drug Saf 2018; 17:795.
  97. Lucena MI, Andrade RJ, Kaplowitz N, et al. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and sex. Hepatology 2009; 49:2001.
  98. Hautekeete ML, Horsmans Y, Van Waeyenberge C, et al. HLA association of amoxicillin-clavulanate--induced hepatitis. Gastroenterology 1999; 117:1181.
  99. Uetrecht JP. New concepts in immunology relevant to idiosyncratic drug reactions: the "danger hypothesis" and innate immune system. Chem Res Toxicol 1999; 12:387.
  100. Uetrecht J. Immune-mediated adverse drug reactions. Chem Res Toxicol 2009; 22:24.
  101. Uetrecht J. Immunoallergic drug-induced liver injury in humans. Semin Liver Dis 2009; 29:383.
  102. Strnad P, Zhou Q, Hanada S, et al. Keratin variants predispose to acute liver failure and adverse outcome: race and ethnic associations. Gastroenterology 2010; 139:828.
  103. Prescott LF. Paracetamol, alcohol and the liver. Br J Clin Pharmacol 2000; 49:291.
  104. Rumack BH. Acetaminophen hepatotoxicity: the first 35 years. J Toxicol Clin Toxicol 2002; 40:3.
  105. Dakhoul L, Ghabril M, Gu J, et al. Heavy Consumption of Alcohol is Not Associated With Worse Outcomes in Patients With Idiosyncratic Drug-induced Liver Injury Compared to Non-Drinkers. Clin Gastroenterol Hepatol 2018; 16:722.
  106. Schiødt FV, Lee WM, Bondesen S, et al. Influence of acute and chronic alcohol intake on the clinical course and outcome in acetaminophen overdose. Aliment Pharmacol Ther 2002; 16:707.
  107. Zimmerman HJ, Maddrey WC. Acetaminophen (paracetamol) hepatotoxicity with regular intake of alcohol: analysis of instances of therapeutic misadventure. Hepatology 1995; 22:767.
  108. Black M, Raucy J. Acetaminophen, alcohol, and cytochrome P-450. Ann Intern Med 1986; 104:427.
  109. Perrot N, Nalpas B, Yang CS, Beaune PH. Modulation of cytochrome P450 isozymes in human liver, by ethanol and drug intake. Eur J Clin Invest 1989; 19:549.
  110. Lucas D, Ménez C, Girre C, et al. Decrease in cytochrome P4502E1 as assessed by the rate of chlorzoxazone hydroxylation in alcoholics during the withdrawal phase. Alcohol Clin Exp Res 1995; 19:362.
  111. Lauterburg BH, Velez ME. Glutathione deficiency in alcoholics: risk factor for paracetamol hepatotoxicity. Gut 1988; 29:1153.
  112. 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.
  113. Lewis JH. Medication-related and other forms of toxic liver injury. In: Clin Prac Gastroenterol, Brandt LJ (Ed), Churchill Livingstone, Philadelphia 1998. p.855.
  114. Alvares AP, Pantuck EJ, Anderson KE, et al. Regulation of drug metabolism in man by environmental factors. Drug Metab Rev 1979; 9:185.
  115. Pantuck EJ, Pantuck CB, Garland WA, et al. Stimulatory effect of brussels sprouts and cabbage on human drug metabolism. Clin Pharmacol Ther 1979; 25:88.
  116. Hakooz N, Hamdan I. Effects of dietary broccoli on human in vivo caffeine metabolism: a pilot study on a group of Jordanian volunteers. Curr Drug Metab 2007; 8:9.
  117. Daković-Svajcer K, Samojlik I, Rasković A, et al. The activity of liver oxidative enzymes after single and multiple grapefruit juice ingestion. Exp Toxicol Pathol 1999; 51:304.
  118. Hukkanen J, Jacob P 3rd, Benowitz NL. Effect of grapefruit juice on cytochrome P450 2A6 and nicotine renal clearance. Clin Pharmacol Ther 2006; 80:522.
  119. Brill S, Zimmermann C, Berger K, et al. In vitro interactions with repeated grapefruit juice administration--to peel or not to peel? Planta Med 2009; 75:332.
  120. Zhang W, Parentau H, Greenly RL, et al. Effect of protein-calorie malnutrition on cytochromes P450 and glutathione S-transferase. Eur J Drug Metab Pharmacokinet 1999; 24:141.
  121. Whitcomb DC, Block GD. Association of acetaminophen hepatotoxicity with fasting and ethanol use. JAMA 1994; 272:1845.
  122. Price VF, Miller MG, Jollow DJ. Mechanisms of fasting-induced potentiation of acetaminophen hepatotoxicity in the rat. Biochem Pharmacol 1987; 36:427.
  123. Kurtovic J, Riordan SM. Paracetamol-induced hepatotoxicity at recommended dosage. J Intern Med 2003; 253:240.
  124. Flockhart DA, Oesterheld JR. Cytochrome P450-mediated drug interactions. Child Adolesc Psychiatr Clin N Am 2000; 9:43.
  125. Kalgutkar AS, Obach RS, Maurer TS. Mechanism-based inactivation of cytochrome P450 enzymes: chemical mechanisms, structure-activity relationships and relationship to clinical drug-drug interactions and idiosyncratic adverse drug reactions. Curr Drug Metab 2007; 8:407.
  126. Stepan AF, Walker DP, Bauman J, et al. Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: a perspective based on the critical examination of trends in the top 200 drugs marketed in the United States. Chem Res Toxicol 2011; 24:1345.
  127. Poellinger L. Mechanistic aspects--the dioxin (aryl hydrocarbon) receptor. Food Addit Contam 2000; 17:261.
  128. Le Couteur DG, McLean AJ. The aging liver. Drug clearance and an oxygen diffusion barrier hypothesis. Clin Pharmacokinet 1998; 34:359.
  129. Hilmer SN, Shenfield GM, Le Couteur DG. Clinical implications of changes in hepatic drug metabolism in older people. Ther Clin Risk Manag 2005; 1:151.
  130. Wrighton SA, Stevens JC. The human hepatic cytochromes P450 involved in drug metabolism. Crit Rev Toxicol 1992; 22:1.
  131. Mitchell SJ, Hilmer SN. Drug-induced liver injury in older adults. Ther Adv Drug Saf 2010; 1:65.
  132. Hunt CM, Yuen NA, Stirnadel-Farrant HA, Suzuki A. Age-related differences in reporting of drug-associated liver injury: data-mining of WHO Safety Report Database. Regul Toxicol Pharmacol 2014; 70:519.
  133. Abboud G, Kaplowitz N. Drug-induced liver injury. Drug Saf 2007; 30:277.
  134. George N, Chen M, Yuen N, et al. Interplay of gender, age and drug properties on reporting frequency of drug-induced liver injury. Regul Toxicol Pharmacol 2018; 94:101.
  135. Lucena MI, Andrade RJ, Fernández MC, et al. Determinants of the clinical expression of amoxicillin-clavulanate hepatotoxicity: a prospective series from Spain. Hepatology 2006; 44:850.
  136. 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.
  137. Tostmann A, Boeree MJ, Aarnoutse RE, et al. Antituberculosis drug-induced hepatotoxicity: concise up-to-date review. J Gastroenterol Hepatol 2008; 23:192.
  138. Benedetti MS, Whomsley R, Canning M. Drug metabolism in the paediatric population and in the elderly. Drug Discov Today 2007; 12:599.
  139. Amacher DE. Female gender as a susceptibility factor for drug-induced liver injury. Hum Exp Toxicol 2014; 33:928.
  140. Hunt CM, Westerkam WR, Stave GM. Effect of age and gender on the activity of human hepatic CYP3A. Biochem Pharmacol 1992; 44:275.
  141. Morgan ET. Regulation of cytochromes P450 during inflammation and infection. Drug Metab Rev 1997; 29:1129.
  142. Leise MD, Poterucha JJ, Talwalkar JA. Drug-induced liver injury. Mayo Clin Proc 2014; 89:95.
  143. Chalasani N, Reddy KRK, Fontana RJ, et al. Idiosyncratic Drug Induced Liver Injury in African-Americans Is Associated With Greater Morbidity and Mortality Compared to Caucasians. Am J Gastroenterol 2017; 112:1382.
  144. Cai Y, Yi J, Zhou C, Shen X. Pharmacogenetic study of drug-metabolising enzyme polymorphisms on the risk of anti-tuberculosis drug-induced liver injury: a meta-analysis. PLoS One 2012; 7:e47769.
  145. Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol 2008; 64:1147.
  146. Stavropoulou E, Pircalabioru GG, Bezirtzoglou E. The Role of Cytochromes P450 in Infection. Front Immunol 2018; 9:89.
  147. Debinski HS, Lee CS, Danks JA, et al. Localization of uridine 5'-diphosphate-glucuronosyltransferase in human liver injury. Gastroenterology 1995; 108:1464.
  148. Elbekai RH, Korashy HM, El-Kadi AO. The effect of liver cirrhosis on the regulation and expression of drug metabolizing enzymes. Curr Drug Metab 2004; 5:157.
  149. 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.
  150. Vuppalanchi R, Teal E, Chalasani N. Patients with elevated baseline liver enzymes do not have higher frequency of hepatotoxicity from lovastatin than those with normal baseline liver enzymes. Am J Med Sci 2005; 329:62.
  151. Russo MW, Scobey M, Bonkovsky HL. Drug-induced liver injury associated with statins. Semin Liver Dis 2009; 29:412.
  152. 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.
  153. Massart J, Begriche K, Moreau C, Fromenty B. Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity. J Clin Transl Res 2017; 3:212.
  154. Lammert C, Imler T, Teal E, Chalasani N. Patients With Chronic Liver Disease Suggestive of Nonalcoholic Fatty Liver Disease May Be at Higher Risk for Drug-Induced Liver Injury. Clin Gastroenterol Hepatol 2019; 17:2814.
  155. Chalasani N. Statins and hepatotoxicity: focus on patients with fatty liver. Hepatology 2005; 41:690.
  156. Cohen DE, Anania FA, Chalasani N, National Lipid Association Statin Safety Task Force Liver Expert Panel. An assessment of statin safety by hepatologists. Am J Cardiol 2006; 97:77C.
  157. Browning JD. Statins and hepatic steatosis: perspectives from the Dallas Heart Study. Hepatology 2006; 44:466.
  158. Chen M, Suzuki A, Borlak J, et al. Drug-induced liver injury: Interactions between drug properties and host factors. J Hepatol 2015; 63:503.
  159. Senior JR. What is idiosyncratic hepatotoxicity? What is it not? Hepatology 2008; 47:1813.
  160. 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.
  161. Uetrecht J. Idiosyncratic drug reactions: current understanding. Annu Rev Pharmacol Toxicol 2007; 47:513.
  162. Russmann S, Kullak-Ublick GA, Grattagliano I. Current concepts of mechanisms in drug-induced hepatotoxicity. Curr Med Chem 2009; 16:3041.
  163. 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.
  164. Han D, Dara L, Win S, et al. Regulation of drug-induced liver injury by signal transduction pathways: critical role of mitochondria. Trends Pharmacol Sci 2013; 34:243.
  165. 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.
  166. Lammert C, Bjornsson E, Niklasson A, Chalasani N. Oral medications with significant hepatic metabolism at higher risk for hepatic adverse events. Hepatology 2010; 51:615.
  167. McEuen K, Borlak J, Tong W, Chen M. Associations of Drug Lipophilicity and Extent of Metabolism with Drug-Induced Liver Injury. Int J Mol Sci 2017; 18:1335.
  168. Holt MP, Ju C. Mechanisms of drug-induced liver injury. AAPS J 2006; 8:E48.
  169. Morgan RE, Trauner M, van Staden CJ, et al. Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development. Toxicol Sci 2010; 118:485.
  170. Aleo MD, Luo Y, Swiss R, et al. Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump. Hepatology 2014; 60:1015.
  171. McGill MR, Williams CD, Xie Y, et al. Acetaminophen-induced liver injury in rats and mice: comparison of protein adducts, mitochondrial dysfunction, and oxidative stress in the mechanism of toxicity. Toxicol Appl Pharmacol 2012; 264:387.
  172. Gunawan B, Kaplowitz N. Clinical perspectives on xenobiotic-induced hepatotoxicity. Drug Metab Rev 2004; 36:301.
  173. Kaplowitz N. Biochemical and cellular mechanisms of toxic liver injury. Semin Liver Dis 2002; 22:137.
  174. Hoofnagle JH, Björnsson ES. Drug-Induced Liver Injury - Types and Phenotypes. N Engl J Med 2019; 381:264.
  175. Zeng L, Jia L, Xu S, et al. Vascular endothelium changes after conditioning in hematopoietic stem cell transplantation: role of cyclophosphamide and busulfan. Transplant Proc 2010; 42:2720.
  176. Bonifazi F, Barbato F, Ravaioli F, et al. Diagnosis and Treatment of VOD/SOS After Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2020; 11:489.
  177. Yang XQ, Ye J, Li X, et al. Pyrrolizidine alkaloids-induced hepatic sinusoidal obstruction syndrome: Pathogenesis, clinical manifestations, diagnosis, treatment, and outcomes. World J Gastroenterol 2019; 25:3753.
  178. Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005; 4:489.
  179. Tujios S, Fontana RJ. Mechanisms of drug-induced liver injury: from bedside to bench. Nat Rev Gastroenterol Hepatol 2011; 8:202.
  180. Kaplowitz N. Causality assessment versus guilt-by-association in drug hepatotoxicity. Hepatology 2001; 33:308.
  181. Knowles SR, Uetrecht J, Shear NH. Idiosyncratic drug reactions: the reactive metabolite syndromes. Lancet 2000; 356:1587.
  182. Pessayre D, Fromenty B, Berson A, et al. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev 2012; 44:34.
  183. Funk C, Ponelle C, Scheuermann G, Pantze M. Cholestatic potential of troglitazone as a possible factor contributing to troglitazone-induced hepatotoxicity: in vivo and in vitro interaction at the canalicular bile salt export pump (Bsep) in the rat. Mol Pharmacol 2001; 59:627.
  184. Ogimura E, Nakagawa T, Deguchi J, et al. Troglitazone Inhibits Bile Acid Amidation: A Possible Risk Factor for Liver Injury. Toxicol Sci 2017; 158:347.
  185. Yang R, Tonnesseen TI. DAMPs and sterile inflammation in drug hepatotoxicity. Hepatol Int 2019; 13:42.
  186. Spielberg SP, Gordon GB, Blake DA, et al. Predisposition to phenytoin hepatotoxicity assessed in vitro. N Engl J Med 1981; 305:722.
  187. Liu ZX, Kaplowitz N. Immune-mediated drug-induced liver disease. Clin Liver Dis 2002; 6:755.
  188. Jaeschke H, McGill MR, Ramachandran A. Oxidant stress, mitochondria, and cell death mechanisms in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Drug Metab Rev 2012; 44:88.
  189. Labbe G, Pessayre D, Fromenty B. Drug-induced liver injury through mitochondrial dysfunction: mechanisms and detection during preclinical safety studies. Fundam Clin Pharmacol 2008; 22:335.
  190. Stephens C, Andrade RJ. Genetic Predisposition to Drug-Induced Liver Injury. Clin Liver Dis 2020; 24:11.
  191. Li J, Uetrecht JP. The danger hypothesis applied to idiosyncratic drug reactions. Handb Exp Pharmacol 2010; :493.
  192. Wong WM, Wu PC, Yuen MF, et al. Antituberculosis drug-related liver dysfunction in chronic hepatitis B infection. Hepatology 2000; 31:201.
  193. Levy M. Role of viral infections in the induction of adverse drug reactions. Drug Saf 1997; 16:1.
  194. Iorga A, Dara L, Kaplowitz N. Drug-Induced Liver Injury: Cascade of Events Leading to Cell Death, Apoptosis or Necrosis. Int J Mol Sci 2017; 18:1018.
  195. Robin MA, Le Roy M, Descatoire V, Pessayre D. Plasma membrane cytochromes P450 as neoantigens and autoimmune targets in drug-induced hepatitis. J Hepatol 1997; 26 Suppl 1:23.
  196. Liu ZX, Kaplowitz N. Role of innate immunity in acetaminophen-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2006; 2:493.
  197. Shear NH, Spielberg SP. Anticonvulsant hypersensitivity syndrome. In vitro assessment of risk. J Clin Invest 1988; 82:1826.
  198. Larrey D, Vial T, Micaleff A, et al. Hepatitis associated with amoxycillin-clavulanic acid combination report of 15 cases. Gut 1992; 33:368.
  199. Kleckner HB, Yakulis V, Heller P. Letter: Severe hypersensitivity to diphenylhydantoin with circulating antibodies to the drug. Ann Intern Med 1975; 83:522.
  200. Pohl LR. Drug-induced allergic hepatitis. Semin Liver Dis 1990; 10:305.
  201. Czaja AJ. Drug-induced autoimmune-like hepatitis. Dig Dis Sci 2011; 56:958.
  202. Orman ES, Conjeevaram HS, Vuppalanchi R, et al. Clinical and histopathologic features of fluoroquinolone-induced liver injury. Clin Gastroenterol Hepatol 2011; 9:517.
  203. Ghabril M, Bonkovsky HL, Kum C, et al. Liver injury from tumor necrosis factor-α antagonists: analysis of thirty-four cases. Clin Gastroenterol Hepatol 2013; 11:558.
  204. Björnsson E, Talwalkar J, Treeprasertsuk S, et al. Drug-induced autoimmune hepatitis: clinical characteristics and prognosis. Hepatology 2010; 51:2040.
  205. Corsini A, Ganey P, Ju C, et al. Current challenges and controversies in drug-induced liver injury. Drug Saf 2012; 35:1099.
  206. Yoo JJ, Lim YS, Kim MS, et al. Risk of fatty liver after long-term use of tamoxifen in patients with breast cancer. PLoS One 2020; 15:e0236506.
  207. Di Bisceglie AM, Lok AS, Martin P, et al. Recent US Food and Drug Administration warnings on hepatitis B reactivation with immune-suppressing and anticancer drugs: just the tip of the iceberg? Hepatology 2015; 61:703.
  208. Malnick SD, Abdullah A, Neuman MG. Checkpoint Inhibitors and Hepatotoxicity. Biomedicines 2021; 9:101.
  209. Yamamoto A, Yano Y, Ueda Y, et al. Clinical features of immune-mediated hepatotoxicity induced by immune checkpoint inhibitors in patients with cancers. J Cancer Res Clin Oncol 2021; 147:1747.
Topic 3568 Version 31.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟