Hepatic Drug Metabolism

Role of the Liver in Drug Elimination

By virtue of the portal circulation, the liver is highly exposed to drugs and other toxins absorbed from the intestine. Most drugs tend to be lipophilic compounds that are readily taken up by the liver but cannot be easily excreted unchanged in bile or urine. The liver is well equipped to handle these agents by an adaptable (inducible) series of metabolic pathways. These pathways include those that alter the parent molecule (phase 1); synthesize conjugates of the drug or its metabolite with a more water-soluble moiety, such as a sugar, amino acid, or sulfate molecule (phase 2); and excrete in an energy-dependent manner the parent molecule, its metabolites, or conjugates into bile (phase 3). For any given compound, 1, 2, or all 3 steps may be necessary for drug elimination. Expression and subcellular location of the proteins (enzymes, membrane transporters) that mediate these steps are controlled by a set of nuclear receptors that function as transcriptional regulators and co-regulators, thereby accounting for coordinated regulation of the 3 phases of hepatic drug elimination.

Pathways of Drug Metabolism

Phase 1 and Cytochrome P450

Phase 1 pathways of drug metabolism include oxidation, reduction, and hydrolytic reactions. The products can be readily conjugated or excreted without further modification. ,

Most phase 1 reactions are catalyzed by microsomal drug oxidases, which contain a hemoprotein of the cytochrome P450 (CYP) gene superfamily as a key component. The apparent promiscuity of drug oxidases toward drugs, environmental toxins, steroid hormones, lipids, and bile acids results from the existence of multiple closely related CYP proteins. More than 20 CYP enzymes are present in the human liver. ,

The reaction cycle involves binding of molecular oxygen to the iron in the heme prosthetic group, with subsequent reduction of oxygen by acceptance of an electron from nicotinamide-adenine dinucleotide phosphate (NADPH) cytochrome P450 reductase, a flavoprotein reductase. The resulting “activated oxygen” is incorporated into the drug or another lipophilic compound. Reduction of oxygen and insertion into a drug substrate (“mixed function oxidation”) generates chemically reactive intermediates, including free radicals, electrophilic “oxy-intermediates” (e.g., unstable epoxides, quinone imines), and reduced (and therefore reactive) oxygen species (ROS). The quintessential example is the CYP2E1-catalyzed metabolite of acetaminophen, N -acetyl- p -benzoquinone imine (NAPQI), an oxidizing and arylating metabolite that is responsible for acetaminophen hepatotoxicity. Other examples of reactive quinone compounds include metabolites of troglitazone, quinine, and methyldopa. Likewise, hepatic metabolism of some plant toxins can generate potentially hepatotoxic epoxide metabolites of diterpenoids (see Chapter 89 ). ROS contribute significantly to tissue injury, particularly by generating oxidative stress and triggering tissue stress responses and cell death pathways, as discussed later.

The hepatic content of CYP proteins is higher in acinar zone 3 than in zone 1. Localization of CYP2E1 is usually confined to a narrow rim of hepatocytes 1 to 2 cells thick around the terminal hepatic venule. This explains in part the zonality of hepatic lesions produced by drugs and toxins, such as acetaminophen and carbon tetrachloride, which are converted to reactive metabolites.

Genetic and Environmental Determinants of Cytochrome P450 Enzymes

Pharmacogenetics and Polymorphisms of Cytochrome P450 Expression

The hepatic expression of each CYP enzyme is genetically determined. This finding largely explains the 4-fold or greater differences in rates of drug metabolism among healthy subjects. Some CYPs, particularly minor forms, are also subject to polymorphic inheritance, with some individuals lacking the encoded protein. One example is CYP2D6, which metabolizes debrisoquine and perhexiline. Poor metabolizers lack CYP2D6 and accumulate perhexiline with usual doses; lack of CYP2D6 is the critical determinant in serious adverse effects of perhexiline, including chronic hepatitis and cirrhosis. Other examples include CYPs 2C9 and 2C19, which affect the metabolism of S -warfarin, omeprazole, and phenytoin and of S -mephenytoin, respectively ; 3% of white populations and 15% of Asians are poor metabolizers of S -mephenytoin.

Developmental Regulation and Constitutive Expression

Expression of several CYPs is developmentally regulated. During adult life, the expression of some CYPs declines slightly (by up to 10%) with advancing age, but this change is minor compared with the effects of genetic variation, environmental influences, and liver disease. Gender differences in the expression of CYPs 3A4 and 2E1 may explain the slightly enhanced metabolism of certain drugs (erythromycin, chlordiazepoxide, midazolam) in women, but whether this difference contributes to the increased risk of hepatic drug reactions in women remains unclear.

Nutrition and Disease-Related Changes

A person’s nutritional status influences the expression of certain CYPs, both in health and with liver disease. , , CYP2E1 expression is increased by obesity, high fat intake, diabetes mellitus, and fasting. , Diseases that alter the expression of hepatic CYPs include hypothyroidism (decreased CYP1A), and hypopituitarism (decreased CYP3A4). Cirrhosis is associated with decreased levels of total cytochrome P450 and also with reduced hepatic perfusion; the result is a decrease in the clearance of drugs such as propranolol that are metabolized rapidly by the liver. The effects of cirrhosis vary, however, among individual CYP families ( Table 88.1 ) and with the type of liver disease (e.g., CYP3A4 levels are preserved with cholestatic but lowered with hepatocellular liver disease).

Table 88.1
Cytochrome P450 (CYP) Isoenzymes Involved in Phase I Drug Metabolism in Humans
CYP Isoenzymes Substrates Effect of Liver Disease on CYP Activity
CYP1A2 Caffeine, theophylline, clonazepam ↓↓↓
CYP2A6 Halothane, methoxyflurane ↓↓
CYP2C9 Diclofenac, losartan, warfarin
CYP2C19 Citalopram, diazepam, omeprazole ↓↓↓
CYP2D6 Codeine, haloperidol, metoprolol
CYP2E1 Enflurane, halothane, acetaminophen
CYP3A4 Amiodarone, carbamazepine, cyclosporine, terfenadine ↓↓↓

Adaptive Response and Enzyme Induction

Exposure to lipophilic substances generates an adaptive response that usually involves transient liver cell injury (discussed later) as well as synthesis of new enzyme protein, a process termed enzyme induction. The molecular basis for genetic regulation of constitutive and inducible expression of CYP3A4, the major human hepatic cytochrome P450, has been determined. Drugs such as rifampin interact with the pregnane X-receptor (PXR), a member of the orphan nuclear receptor family of transcriptional regulators. Activated PXR and the analogous constitutive androstane receptor (CAR) in turn bind to cognate nucleotide sequences upstream to the CYP3A4 structural gene within a xenobiotic-regulatory enhancer module. This interaction regulates the CYP3A4 promoter downstream and ultimately the transcription of CYP3A4 protein. Similar control mechanisms apply to several other CYP pathways, particularly those involved in bile acid synthesis. ,

Common examples of microsomal enzymes induced by environmental agents include cigarette and cannabis smoking (CYP1A2) 8 and alcohol (CYP2E1 and possibly CYP3A4). Several drugs are potent inducers of CYP enzymes. Isoniazid induces CYP2E1, whereas phenobarbital and phenytoin increase the expression of multiple CYPs. Rifampin is a potent inducer of CYP3A4, as is hypericum, the active ingredient of St. John’s wort, a commonly used herbal medicine, thereby causing interactions between conventional medicines and a complementary and alternative medicine (CAM) preparation. Regulation of hepatic drug metabolizing enzymes is reviewed elsewhere. ,

The implications for drug-induced liver disease are 2-fold. First, enzyme induction often extends beyond the CYP system, possibly due to PXR and CAR activation. This induction may influence bile acid metabolism and liver growth and could account for increases in serum alkaline phosphatase and GGTP levels, which reflect “hepatic adaptation” to chronic drug ingestion. Second, the influence of one drug on expression and activity of drug metabolizing enzymes and drug elimination (phase 3) pathways can alter the metabolism or disposition of other agents. Such drug-drug interactions may be relevant to mechanisms of drug-induced liver injury.

Inhibition of Drug Metabolism

Some chemicals inhibit drug metabolism. In persons taking more than one medication, for example, competition for phase 2 pathways such as glucuronidation and sulfation facilitates the presentation of unconjugated drug to the CYP system. This may explain in part why agents such as zidovudine and phenytoin lower the dose threshold for acetaminophen-induced hepatotoxicity.

Other Pathways of Drug Oxidation

In addition to CYP enzymes, mitochondrial electron transport systems can generate tissue-damaging reactive intermediates during drug metabolism. Examples include nitroradicals from nitrofuran derivatives (nitrofurantoin, cocaine). Subsequent electron transfer by flavoprotein reductases into molecular oxygen generates superoxide and other ROS. Some anticancer drugs (e.g., doxorubicin, imidazole antimicrobials) can participate in other oxidation-reduction (redox) cycling reactions that generate ROS.

Phase 2 (Conjugation)

Phase 2 reactions involve formation of ester links to the parent compound or to a drug metabolite to form hydrophilic conjugates that can be excreted readily in bile or urine. The responsible enzymes include glucuronyl transferases, sulfotransferases, glutathione S- transferases, and acetyl and amino acid N -transferases. Conjugation reactions are also regulated by CAR and other nuclear transcription factors, and can be retarded by depletion of their rate-limiting cofactors, such as glucuronic acid and inorganic sulfate; the relatively low capacity of these enzyme systems restricts the efficacy of drug elimination when substrate concentrations exceed enzyme saturation. In general, drug conjugates are nontoxic, and phase 2 reactions are considered to be detoxification reactions, with exceptions. For example, some glutathione conjugates can undergo cysteine S -conjugate beta-lyase–mediated activation to highly reactive intermediates. In general, conjugation reactions are minimally affected by liver disease, with the possible exception of some reduction of enzyme activity and resulting drug clearance in decompensated cirrhosis; this is relevant to selection of major analgesics (morphine rather than pethidine) and hypnotics (oxazepam rather than diazepam). Little is known about the regulation of such enzymes or their potential significance for DILI.

Phase 3

This phase involves secretion of drugs, drug metabolites, or their conjugates into bile. Several transporters participate in these pathways that involve ATP-binding cassette (ABC) proteins and are powered by energy from ATP hydrolysis (see Chapter 64 ). ABC transport proteins are widely distributed in nature and include the CF transmembrane conductance regulator and the canalicular and intestinal copper transporters (see Chapter 76 ).

Multidrug resistance protein 1 (MDR1, gene symbol ABCB1 ) is highly expressed on the apical (canalicular) plasma membrane of hepatocytes, where it transports cationic drugs, particularly anticancer agents, into bile. Another family of ABC transporters, the multidrug resistance-associated proteins (MRPs), is also expressed in the liver. At least 2 members of this family excrete drug (and other) conjugates from hepatocytes: MRP-3 (gene symbol ABCC3 ) on the basolateral surface facilitates passage of drug conjugate into the sinusoidal circulation and MRP-2 (gene symbol ABCC2 ), expressed on the canalicular membrane, pumps endogenous compounds (e.g., bilirubin diglucuronide, leukotriene-glutathionyl conjugates, glutathione) and drug conjugates into bile. The bile salt export pump (BSEP) and MDR3 (gene symbols ABCB4 in humans and Mdr2 in mice) are other canalicular transporters involved, respectively, in bile acid and phospholipid secretion into bile. Polymorphisms involving these genes are associated with human cholestatic liver diseases. BSEP interacts with several drugs.

Regulation of the membrane expression and activity of these drug elimination pathways is complex. Altered expression or impaired activity (by competition between agents, changes in membrane lipid composition, or damage from reactive metabolites or covalent binding) could lead to drug accumulation, impairment of bile flow, or cholestatic liver injury. This has been demonstrated for estrogens, , troglitazone, terbinafine, and flucloxacillin and has wider mechanistic implications for drug-induced cholestasis and other forms of liver injury.

Effect of Liver Disease on Drug Metabolism

In considering the safety of prescribing medications in patients with liver disease, physicians need to understand the hepatic extraction ratio of the drug (its rate of uptake and metabolism), its disposition (hepatic, renal, other), the pathways involved if it is subject to hepatic drug metabolism, and whether there are potential interactions between drug effects (pharmacodynamics) and disease complications. In light of the complexity of hepatic drug handling, it is fortunate that most drugs are safe to use in most patients with liver disease. The contexts that will give rise to concern are liver disease associated with reduced hepatic blood flow (cirrhosis and portal hypertension), in which hepatic clearance of drugs with high clearance is reduced, and poor metabolic (synthetic) function of the liver. Apart from subjects already awaiting LT, this category includes patients with alcohol-associated hepatitis and cirrhosis, severe autoimmune hepatitis (AIH), and viral hepatitis with hepatic decompensation. In such patients, oral doses of high-clearance compounds must be reduced substantially because systemic bioavailability may increase 2- to 10-fold as a result of the reduced “first-pass” hepatic clearance. The best example is propranolol, which is usually prescribed in this context to lower portal venous pressure and reduce the risk of variceal bleeding. Instead of doses used for cardiovascular indications (such as 160 to 320 mg daily), the usual starting dose in a patient with cirrhosis should be 10 to 20 mg daily. Other high-clearance compounds affected by severe liver disease include pethidine, tricyclic antidepressants, and salbutamol.

The pathways of hepatic drug metabolism and elimination most affected by liver disease are those involving CYP (see Table 88.1 ). As mentioned earlier, cholestatic forms of liver disease have little effect on CYP3A4 and therefore minimally affect hepatic metabolism of commonly used drugs, such as glucocorticoids, angiotensin-converting enzyme (ACE) inhibitors, cyclosporine, and HIV protease inhibitors. Drugs that rely on hepatic elimination through biliary excretion are minimally affected by liver disease, with the exception of cancer chemotherapeutic agents. Patients with jaundice are at increased risk of liver injury with such agents. By contrast, liver disease has much less effect on conjugation pathways (phase 2 drug metabolism), a property that can be exploited in the choice of sedatives or major analgesics (see later).

Drugs known to precipitate liver complications should be avoided. Patients with cirrhosis have impaired creatinine clearance and are at risk of developing gentamicin nephrotoxicity. Another challenge is the appropriate choice of a sedative to manage alcohol withdrawal in a patient with alcohol-associated cirrhosis. Diazepam is a poor choice in this setting because it is extensively metabolized by CYPs; its clearance is delayed, and hepatic encephalopathy may be precipitated by its use. An alternative benzodiazepine that is metabolized by conjugation alone (e.g., oxazepam) would be a safer choice. Other adverse effects that are not usually related to hepatic drug metabolism include exaggerated effects on clotting factor synthesis (even though warfarin metabolism is not usually affected by liver disease); sodium and water retention by NSAIDs, which also confer high risk of GI bleeding; metabolic acidosis or profound hypoglycemia by metformin and other oral hypoglycemic agents; and hypotension after administration of an ACE inhibitor or major tranquilizer. Acetaminophen appears to be the safest analgesic agent to use in cirrhosis (see later). In general, however, most commonly used agents (antimicrobials, DAAs, antiepileptics, antidepressants, antihypertensives, statins, and oral contraceptives) are safe to use in patients with liver disease.

Liver Disease Caused by Drugs

Definitions and Importance

Drugs are a relatively common cause of liver injury, which usually is defined by abnormalities of liver biochemical test levels, particularly an increase in the serum ALT, alkaline phosphatase, or bilirubin level to more than twice the upper limit of normal (ULN). DILI can be difficult to define in clinical practice because the biochemical tests used to detect liver injury may also be elevated as part of a hepatic adaptive response. Indeed, evidence indicates that some forms of hepatic adaptation to drugs follow an earlier transient process of self-limiting liver injury, followed in turn by operation of innate immunity. Further, the severity of DILI varies from minor nonspecific changes in hepatic structure and function to ALF, cirrhosis, and liver cancer.

The term drug-induced liver disease should be confined to cases in which the nature of the liver injury has been characterized histologically. With the exception of acetaminophen, anticancer drugs, and some botanical or industrial hepatotoxins, most cases of DILI represent adverse drug reactions or hepatic drug reactions. These effects are noxious and unintentional and occur at recommended doses. The latent period is longer (typically from 1 week to 3 to 6 months) than that for direct hepatotoxins (from hours to a few days), and extrahepatic features of drug hypersensitivity may be present.

Although DILI is a relatively uncommon cause of jaundice or acute hepatitis in the community, it is an important cause of more severe acute liver disease, particularly among older people. The overall mortality rate among patients hospitalized for DILI is approximately 10% but varies greatly for individual drugs. , Reported frequencies of individual hepatic drug reactions are underestimated because of the inadequacy of spontaneous reporting. , With reliable prospective and epidemiologic techniques, the frequency (or risk) of most types of drug-induced liver disease is between 1 per 10,000 and 1 per 100,000 persons exposed. Because these responses to drug exposure are clearly rare and unpredictable, they are often termed idiosyncratic drug reactions. Their rarity blunts diagnostic acumen because most clinicians will see few, if any, cases and therefore do not have an appropriate level of clinical suspicion. This concern applies especially to CAM preparations (see Chapter 89 ). Failure to withdraw the causative agent after the onset of symptoms of drug hepatitis or reexposure to such a drug is a common and avoidable factor in ALF attributable to DILI. , Another challenge is that DILI includes an array of clinical syndromes and pathologic findings that mimic known hepatobiliary diseases. Furthermore, although individual agents (and some drug classes) typically produce a characteristic “signature syndrome,” they can also be associated with other and sometimes multiple clinicopathologic syndromes.

DILI is one of the most common reasons for withdrawal of an approved drug. The subject therefore has medico-economic, legal, and regulatory ramifications. Because most types of idiosyncratic hepatic drug reactions are infrequent, serious hepatotoxicity is not usually detected until post-marketing surveillance is conducted. Historically, drugs with a reputation for potential hepatotoxicity have usually been replaced by more acceptable alternatives. Examples include troglitazone, the prototypic thiazolidinedione, and bromfenac, an NSAID, both of which were withdrawn due to fatal hepatotoxicity. , ,

The burgeoning number of available conventional medications and CAM preparations now includes many hundreds that are cited as rare causes of drug-induced liver disease. This poses several challenges to clinicians, , , including concern about what constitutes an adequate level of patient information at the time a drug is prescribed and the reliability of evidence linking an individual agent to a particular type of liver injury. , , Another development is the appreciation that in the context of a complex medical setting, drug toxicity can interact with other causes of liver injury. Noteworthy examples of such situations are bone marrow transplantation; cancer chemotherapy; antiretroviral therapy (ART) for HIV infection; use of antituberculous drugs in patients with chronic viral hepatitis; rifampin hepatitis in patients with PBC; and NAFLD—particularly NASH—precipitated by tamoxifen.

Epidemiology

Frequency or risk, the number of adverse reactions for a given number of persons exposed, is the best term for expressing how common a drug reaction is. Time-dependent terms such as incidence and prevalence are not appropriate because the frequency is not linearly related to the duration of exposure. For most reactions, the onset occurs within a relatively short exposure time, or latent period, although some forms of chronic liver disease occur months or years later. The frequency of drug-induced liver disease is derived from post-marketing surveillance reports submitted to the manufacturers or adverse drug reaction monitoring bodies. In the USA, following approval by the FDA, drug companies are required to report serious adverse events (any incident resulting in death, a threat to life, hospitalization, or permanent disability [Code of Federal Regulations]). Surveillance becomes a more passive process, however, when a drug is approved for marketing and physicians and pharmacists are encouraged to file voluntary written reports through the MedWatch program. Nevertheless, MedWatch receives reports for fewer than 10% of adverse drug reactions, similar to the rate of reporting in France (<6%). The electronic tool for drug-induced serious hepatotoxicity (eDISH) is a graphic instrument that can help identify participants in clinical trials who may be at risk of severe liver disease. Both at-a-glance laboratory data (especially serum aminotransferase and bilirubin levels) for all participants and time-course data for individual patients can be obtained for further scrutiny.

Case Definition: Which Agent?

Many drugs have been implicated in DILI. , The evidence for most drugs, however, is confined to individual or small numbers of case reports, especially in letters to scientific journals or to regulatory authorities, or small observational series. Therefore, for most agents, the evidence that they could cause liver injury is circumstantial and incomplete. Reports often lack pathologic definition, thorough exclusion of other disorders, and logistic imputation of causality, especially with respect to temporal associations (see later). , , Of the drugs listed in the LiverTox DILI database in 2016, ( livertox.nih.gov ), only about one half had convincing associations with DILI. Of these, 22% (76) and 13% (22) were based on cumulative reports of 12 to 50 and more than 50 cases, respectively. In general, agents used most commonly in clinical practice and in the community, including antimicrobials, antineoplastic agents, and NSAIDs, are those that have been implicated in causing DILI. The challenge of identifying the culprit drug among multiple candidates is discussed later.

Frequencies of Hepatic Drug Reactions

Because of incomplete reporting, frequencies of hepatic drug reactions are often underestimated. These estimated frequencies are also crude indicators of risk because of the inherent inaccuracies of case definitions (see later) and because case recognition and reporting depend on the skill and motivation of observers. , , The increased interest of prescribers when initial cases of drug-induced liver disease have been described, together with inappropriate prescribing (e.g., prolonged use of bromfenac, which was approved only for 7 days of use, and overprescribing of flucloxacillin and amoxicillin-clavulanic acid in some countries) can give rise to apparent “mini-epidemics.” More appropriate epidemiologic methods applied to hepatotoxicity have included prescription event monitoring, record linkage, and case-control studies. Prescription event monitoring and record linkage have been used to estimate the frequency of liver injury with some antimicrobials (erythromycins, sulfonamides, tetracyclines, flucloxacillin, amoxicillin-clavulanate) and NSAIDs.

Epidemiologic studies confirm the rarity of drug-induced liver disease with current drugs. For NSAIDs, the risk of liver injury is between 1 and 10 per 100,000 individuals exposed. Amoxicillin-clavulanic acid has been associated with cholestatic hepatitis in 1 to 2 per 100,000 exposed persons, and low-dose tetracyclines have caused hepatotoxicity in less than one case per million persons exposed. , , , The frequency of liver injury may be higher for agents that exert a metabolic type of hepatotoxicity. For example, isoniazid causes liver injury in up to 2% of persons exposed; the risk depends on the patient’s age and gender, concomitant exposure to other agents, and presence of HBV and HCV infections. For some drugs in which other host factors play a pathogenic role, case-control studies have been used to define attributable risk. Examples include the association of aspirin with Reye syndrome and oral contraceptives with liver tumors and hepatic vein thrombosis.

In the 1970s, the late Hyman Zimmerman hypothesized a relationship between the frequency and severity of serum ALT elevations that indicate liver injury and the risk of severe hepatotoxicity. According to “Hy’s rule,” elevations of serum ALT levels to 3-fold or more above the ULN with an associated increase in the serum bilirubin concentration (≥2 × ULN) without an elevation of the serum alkaline phosphatase level (<2 × ULN) indicate a potential for the drug to cause ALF at a rate of about 10% of the number of cases with jaundice. Therefore, if 2 cases of jaundice associated with DILI are observed in a phase 3 clinical trial of 2500 patients, one case of ALF would be expected for every 12,500 subjects who received the drug during the marketing phase. Modifications of Hy’s rule have been suggested to improve its specificity in identifying cases that may progress to ALF. , These include using an R ratio (ALT/ULN divided by ALP/ULN) or a modified nR ratio (which uses either ALT or AST depending on whichever parameter produces the higher R ratio) of greater than 5. Applying these approaches to a Spanish DILI cohort improved the specificity from 44% (Hy’s rule) to 67% (R ratio), and 63% (nR ratio) with similar sensitivities. These criteria await further validation.

Importance of Drugs as a Cause of Liver Disease

Hepatotoxicity accounts for less than 5% of cases of jaundice or acute hepatitis in the community and for even fewer cases of chronic liver disease. , However, drugs are an important cause of more severe types of liver disease and for liver disease in older people. They account for 10% of cases of severe hepatitis admitted to the hospital in France and for 43% of cases of hepatitis among patients 50 years of age or older. Drugs account for more than one half of the cases of ALF referred to a special unit in the USA. The pattern and frequency of agents incriminated varies among countries; for example, herbal and dietary supplements accounted for over a quarter of DILI cases in South Korea (see Chapter 89 ).

In most cases of DILI, drugs are the sole cause of hepatic damage. In other cases, drugs increase the relative risk for types of liver disease that may occur in the absence of drug exposure. Examples include salicylates in Reye syndrome, oral contraceptive steroids (OCS) in hepatic venous thrombosis, methotrexate in hepatic fibrosis associated with alcohol-associated liver disease and NAFLD, and tamoxifen in NAFLD and NASH. Predisposition of patients with preexisting liver disease to DILI is minimal, but potential interactions between chronic HCV infection and several groups of drugs and between chronic HBV infection and antituberculous chemotherapy are now reasonably established. On the other hand, liver failure may be more likely to develop if the patient with a hepatic drug reaction (e.g., to amoxicillin-clavulanic acid) that usually is associated with a good outcome has underlying chronic liver disease.

Risk Factors

For dose-dependent hepatotoxins such as acetaminophen and methotrexate and for some idiosyncratic reactions that are partly dose-dependent (e.g., bromfenac, tetracyclines, dantrolene, tacrine, oxypenicillins), the factors that influence the risk of drug-induced liver disease include the drug dose (DILI is more likely with a drug dose ≥ 50 mg daily), blood level of the drug, and duration of intake. For idiosyncratic reactions, however, host determinants are central to liver injury. The most critical determinant is likely to be genetic predisposition, but other “constitutional” and environmental factors can influence the risk of liver injury, as summarized in Table 88.2 . The most important factors are age, gender, exposure to other substances, a history or family history of previous drug reactions, other risk factors for liver disease, and concomitant medical disorders.

Table 88.2
Factors Influencing the Risk of Liver Diseases Caused by Drugs
Factor Examples of Drugs Affected Influence
Genetic factors Halothane, phenytoin, sulfonamides Multiple cases in families
Abacavir, amoxicillin-clavulanic acid, flucloxacillin Strong HLA association
Age Halothane, isoniazid, nitrofurantoin, troglitazone Age >60 yrs: increased frequency, increased severity
Valproic acid, salicylates More common in children
Gender Halothane, minocycline, nitrofurantoin More common in women, especially those with chronic hepatitis
Amoxicillin-clavulanic acid, azathioprine More common in men
Dose Acetaminophen, aspirin, some herbal and dietary supplements Blood levels are directly related to the risk of hepatotoxicity
Oxypenicillins, tacrine, tetracycline Idiosyncratic reactions, with partial relationship to dose
Methotrexate, vitamin A Total dose, dosing frequency, and duration of exposure are related to the risk of hepatic fibrosis
Valproic acid Familial cases, association with mitochondrial enzyme deficiencies
Other drugs Acetaminophen Isoniazid, phenytoin, and zidovudine lower the dose threshold and increase the severity of hepatotoxicity
Valproic acid Other antiepileptic drugs increase the risk of hepatotoxicity
Anticancer drugs Interactive vascular toxicity
History of other drug reactions Enflurane, halothane, isoflurane
Erythromycins
Diclofenac, ibuprofen, tiaprofenic acid
COX-2 inhibitors, sulfonamides
Instances of cross-sensitivity have been reported among members of each drug class but are rare
Excessive alcohol use Acetaminophen Lowered dose threshold, poorer outcome
Isoniazid, methotrexate Increased risk of liver injury, hepatic fibrosis
Nutritional Status
Obesity Halothane, methotrexate, tamoxifen, troglitazone Increased risk of liver injury, hepatic fibrosis
Fasting Acetaminophen Increased risk of hepatotoxicity
Preexisting liver disease Hycanthone, pemoline Increased risk of liver injury
Antituberculosis drugs, ibuprofen Increased risk of liver injury with chronic hepatitis B and C
Other Diseases/Conditions
Diabetes mellitus Methotrexate Increased risk of hepatic fibrosis
HIV/AIDS Sulfonamides Increased risk of hypersensitivity
Renal failure Methotrexate, tetracycline Increased risk of liver injury, hepatic fibrosis
Organ transplantation Azathioprine, busulfan, thioguanine Increased risk of vascular toxicity

Genetic Factors

Genetic determinants predispose to drug-induced liver disease, as they do for other types of drug reaction, such as penicillin allergy. The contention that atopic patients may be at increased risk of some types of drug hepatitis is unproven, however. Genetic factors determine the activity of drug-activating and antioxidant pathways, encode pathways of canalicular bile secretion, and modulate the immune response, tissue stress responses, and cell death pathways. Documented examples of drugs associated with a familial predisposition to adverse hepatic drug reactions are few and include valproic acid and phenytoin. Inherited mitochondrial diseases are a risk factor for valproic acid–induced hepatotoxicity. Some forms of drug-induced liver disease, particularly drug-induced hepatitis and granulomatous reactions, are associated with the reactive metabolite syndrome (see later). Initial studies showed only no or weak associations between specific HLA haplotypes and some types of drug-induced liver disease. Genome-wide association studies (GWAS) have revealed stronger associations between specific HLA haplotypes and several drugs, including flucloxacillin and amoxicillin-clavulanic acid ( Table 88.3 ).

Table 88.3
Genetic Variants Associated with DILI
Drug Category Allele(s) Odds Ratio (CI) for DILI
Flucloxacillin Antibiotic HLA-B 57:01; 80.6 (23-285)
Ticlopidine Antiplatelet HLA A 33:03 36.5 (7.3-184)
Minocycline Antibiotic HLA-B 35:02 30
Lumiracoxib NSAID HLA-DQA1 01:02; DRB1 1501 ; DQB1 06:02; DRB5∗01:01 6.3 (4.1-9.6)
Diclofenac NSAID ABCC2 [MRP2]
C-24T
6 (2.4-17)
Ximelagatran Thrombin inhibitor HLA-DRB1 07:01 4.4
Nevirapine Protease inhibitor HLA-B 58:01 3.5
Amoxicillin-clavulanic acid Antibiotic HLA-DRB1 1501 ;
DRB5 01:01; DQB1 06:02
2.3 (1.0-5.26)
Terbinafine Antifungal drug HLA-A 33:01 2.3
Sex hormones Various ABCB11 [BSEP] V444A 1.7-4
ABC, APT-binding cassette; BSEP, bile salt export pump; CI, confidence intervals; MRP2, multidrug resistance-associated protein 2.

Alleles in bold are the most important in the pathogenesis of DILI for the particular drug.

Age

Most hepatic drug reactions are more common in adults than in children. Exceptions include valproic acid hepatotoxicity, which is most common in children under 3 years of age but rare in adults, and Reye syndrome, in which salicylates play a key role. In adults, the risk of isoniazid-associated hepatotoxicity is greater in persons older than 40 years of age. Similar observations have been made for nitrofurantoin, halothane, etretinate, diclofenac, and troglitazone. , , , The increased frequency of adverse drug reactions in older subjects is largely the result of increased exposure, polypharmacy, and altered drug disposition. In addition, the clinical severity of hepatotoxicity increases strikingly with age, as exemplified by fatal reactions to isoniazid and halothane. , , ,

Gender

Women are particularly predisposed to drug-induced hepatitis, a difference that cannot be attributed simply to increased exposure. Examples include toxicity caused by halothane, nitrofurantoin, sulfonamides, flucloxacillin, minocycline, and troglitazone. , Drug-induced chronic hepatitis caused by nitrofurantoin, diclofenac, or minocycline has an even more pronounced female preponderance. , Conversely, equal sex frequency or even male preponderance is common for some cholestatic drug reactions (e.g., amoxicillin-clavulanic acid). Azathioprine-induced liver disease occurs more frequently in male renal transplant recipients than in female recipients.

Concomitant Exposure to Other Agents

Patients who are taking multiple drugs are more likely to experience an adverse reaction than those who are taking one agent. , , The mechanisms include enhanced CYP-mediated metabolism of the second drug to a toxic intermediate (see later). Examples include toxicity caused by acetaminophen, isoniazid, valproic acid, other anticonvulsants, and anticancer drugs. Alternatively, drugs may alter the disposition of other agents by reducing bile flow or competing with canalicular pathways for biliary excretion (phase 3 drug elimination). This mechanism may account for interactions between OCS and other drugs to produce cholestasis. Drugs or their metabolites may also interact through mechanisms of cellular toxicity and cell death that involve mitochondrial injury, intracellular signaling pathways, activation of transcription factors, and regulation of hepatic genes involved in controlling the response to stress and injury that triggers pro-inflammatory and cell death processes. ,

Previous Drug Reactions

A history of an adverse drug reaction generally increases the risk of reactions to the same drug as well as other agents. Nevertheless, instances of cross-sensitivity to related agents in cases of drug-induced liver disease are surprisingly uncommon. Examples of cross-sensitivity between drugs (or drug classes) include the haloalkane anesthetics (see Chapter 89 ), erythromycins, phenothiazines and tricyclic antidepressants, isoniazid and pyrazinamide, and some NSAIDs. A crucial point is that a previous reaction to the same drug is a major risk factor for an increase in the severity of DILI. A Spanish study examined the risk of DILI in persons with a history of DILI (with a different drug). Recurrent DILI was infrequent (1.2%) and was attributable most commonly to drugs that were structurally similar or had similar targets, whereas others exhibited features consistent with AIH, thereby raising the possibility that immune-mediated processes may be mechanistically involved or that the correct diagnosis was actually AIH.

Alcohol

Chronic excessive alcohol ingestion decreases the dose threshold for, and enhances the severity of, acetaminophen-induced hepatotoxicity and increases the risk and severity of isoniazid hepatitis, niacin (nicotinic acid, nicotinamide) hepatotoxicity, and methotrexate-induced hepatic fibrosis.

Nutritional Status

Obesity is strongly associated with the risk of halothane hepatitis and is an independent risk factor for NASH and hepatic fibrosis in persons taking methotrexate or tamoxifen. Fasting also predisposes to acetaminophen hepatotoxicity, and a role for undernutrition has been proposed in isoniazid hepatotoxicity.

Preexisting Liver Disease

In general, liver diseases such as alcohol-associated cirrhosis and cholestatic disorders do not predispose to adverse hepatic reactions. Exceptions include toxicity due to some anticancer drugs, niacin, pemoline, and hycanthone. Preexisting liver disease is a critical determinant of methotrexate-induced hepatic fibrosis. Patients with chronic HBV or HCV infection or HIV/AIDS appear to be at heightened risk of liver injury during antituberculous or ART therapy, after exposure to ibuprofen and possibly other NSAIDs, after myeloablative therapy in preparation for bone marrow transplantation (resulting in sinusoidal obstruction syndrome), and possibly after taking antiandrogens, such as flutamide and cyproterone acetate. A particularly strong association has been reported between HCV infection and the risk of liver injury during ART; the risk may be increased 2- to 10-fold. ,

Other Diseases

RA increases the risk of salicylate hepatotoxicity, and a curious, unexplained observation is that sulfasalazine hepatitis is more common in patients with RA than in those with IBD. Diabetes mellitus, obesity, and chronic kidney disease predispose to methotrexate-induced hepatic fibrosis, whereas HIV/AIDS confers a heightened risk of sulfonamide hypersensitivity. A retrospective cohort study found that the age- and sex-standardized incidence of drug-induced ALF in patients with diabetes mellitus was 0.08 to 0.15 per 1000 person-years, irrespective of the therapeutic agent used (the number using troglitazone was small); the incidence was highest (approximately 0.3 per 1000) during the first 6 months of exposure. Renal transplantation is a risk factor for azathioprine-associated vascular injury, whereas kidney disease predisposes to tetracycline-induced fatty liver. Finally, sinusoidal obstruction syndrome induced by anticancer drugs is more common after bone marrow transplantation and in persons with HCV infection. , , , ,

Pathophysiology

Toxic Mechanisms of Liver Injury

Direct Hepatotoxins and Reactive Metabolites

Highly hepatotoxic chemicals injure key subcellular structures, particularly mitochondria and the plasma membrane. The injury arrests energy generation, dissipates ionic gradients, and disrupts the physical integrity of the cell. This type of overwhelming cellular injury does not apply to currently relevant hepatotoxins, most of which require metabolic activation to mediate damage to liver cells. The resulting reactive metabolites can interact with critical cellular target molecules, particularly those with nucleophilic substituents such as thiol-rich proteins and nucleic acids. Together with ROS, they act as oxidizing species within the hepatocyte to establish oxidative stress, a state of imbalance between pro-oxidants and antioxidants. ROS are also key signaling molecules that mediate biological responses to stress, as discussed later. Alternatively, reactive metabolites bind irreversibly to macromolecules, particularly proteins and lipids. Such covalent binding may produce injury by inactivating key enzymes or by forming protein-drug adducts that could be targets for immunodestructive processes that cause liver injury. Notwithstanding these comments, there is increasing evidence that “direct hepatotoxins,” such as acetaminophen, activate innate immune mechanisms in the liver in response to stress with release of danger-activated molecular patterns; the latter (as well as bacterial products such as endotoxin, a pathogen-associated molecular pattern) trigger Toll-like receptors to activate pro-inflammatory and cell death pathways (see Chapter 72 ).

Oxidative Stress and the Glutathione System

The liver is exposed to oxidative stress by the propensity of hepatocytes to reduce oxygen, particularly in mitochondria and in microsomal electron transport systems (such as CYP2E1), and by NADPH-oxidase-catalyzed formation of ROS and nitroradicals in Kupffer cells, endothelial cells, and stimulated polymorphonuclear leukocytes (neutrophils) and macrophages. To combat oxidative stress, the liver is well-endowed with antioxidant mechanisms, including micronutrients, such as vitamin E and vitamin C, thiol-rich proteins (e.g., metallothionein, ubiquinone), metal-sequestering proteins (e.g., ferritin), and enzymes that metabolize reactive metabolites (e.g., epoxide hydrolases), ROS (e.g., catalase, superoxide dismutase), and lipid peroxides (e.g., glutathione peroxidases). Glutathione (L-δ-glutamyl-L-cysteine-glycine) is the most important antioxidant in the mammalian liver.

Hepatocytes are the exclusive site of glutathione synthesis. Hepatic levels of glutathione are high (5 to 10 mmol/L) and can be increased by enhancing the supply of cysteine for glutathione synthesis; this mechanism is the cornerstone of thiol antidote therapy for acetaminophen poisoning. Hepatocyte glutathione synthesis increases in response to pro-oxidants, as occurs when CYP2E1 is overexpressed as a result of signaling via the redox-sensitive transcription factor Nrf. , , , Glutathione synthesis, via expression of the rate-limiting enzyme glutamate cysteine ligase is also a response to mitochondrial injury by such agents as acetaminophen. Glutathione in its reduced form (GSH) is a critical cofactor for several antioxidant pathways, including thiol-disulfide exchange reactions and glutathione peroxidase. Glutathione peroxidase has a higher affinity for hydrogen peroxide than does catalase, and it disposes of lipid peroxides, free radicals, and electrophilic drug metabolites. GSH is also a cofactor for conjugation reactions catalyzed by the glutathione S -transferases involved with phase 3 transport of drug metabolites into bile. Other reactions proceed nonenzymatically. In turn, the products include glutathione-protein mixed disulfides and oxidized glutathione. The latter can be converted back to glutathione by proton donation catalyzed by glutathione reductase.

Normally, most glutathione within the hepatocyte is in the reduced state, indicating the importance of this pathway for maintenance of the redox capacity of the cell. The reduced form of NADPH is an essential cofactor for glutathione reductase; NADPH formation requires ATP, thereby illustrating a critical link between mitochondrial integrity and the energy-generating capacity of the liver and its ability to withstand oxidative stress. Glutathione is also compartmentalized within the hepatocyte, with the highest concentrations found in the cytosol. Adequate levels of glutathione are essential in mitochondria, where ROS are constantly being formed as a minor by-product of oxidative respiration and in response to some drugs or metabolites that interfere with the mitochondrial respiratory chain. Mitochondrial glutathione is maintained by active uptake from the cytosol via a transport system that is altered by chronic ethanol exposure and in some forms of lipotoxicity (e.g., with cholesterol) and is therefore another potential target for drug toxicity.

Biochemical Mechanisms of Cellular Injury

Mechanisms once thought to be central to hepatotoxicity, such as covalent binding to cellular enzymes and peroxidation of membrane lipids, are no longer regarded as exclusive pathways of cellular damage. Rather, oxidation of proteins, phospholipid fatty acyl side chains (lipid peroxidation), and nucleosides appears to be a component of the biochemical stress that characterizes toxic liver injury. In one experiment, healthy volunteers were administered a variety of low-molecular-weight heparins, known to cause transient serum ALT elevations. In addition to aminotransferase increases in >90% of cases, markers of subcellular injury (cytosolic, mitochondrial), apoptosis (M30 fragmentation product of cytokeratin [CK] 8/19), microRNA (miRNA)-122, DNA, and high-mobility group box-1 (HMGB1) increased; HMGB1 is a DAMP released in necrosis. The authors concluded that heparins as a class caused self-limited and mild necrosis with secondary activation of an innate immune response. Secondary reactions, including post-translational modification of proteins via adenosine diphosphate ribosylation or protease activation, cleavage of DNA by activation of endogenous endonucleases, and disruption of lipid membranes by activated phospholipases may also play a role in DILI. Some of these catabolic reactions could be initiated by a rise in the cytosolic ionic calcium concentration (Ca 2+ ) i , as a result of increased Ca 2+ entry or release from internal stores in the endoplasmic reticulum and mitochondria. , The potential role of endoplasmic reticulum stress in DILI is less well defined. ,

The concept that hepatotoxic chemicals cause hepatocyte cell death by a biochemical final common pathway (e.g., activation of catalytic enzymes by a rise in [Ca 2+ ] i ) has proved inadequate to explain the diverse processes that can result in lethal hepatocellular injury. Rather, a variety of processes can damage key organelles, thereby causing intracellular stress that activates signaling pathways and transcription factors. Mitochondrial injury, particularly that signaled via activation of the c-Jun N-terminal kinase (JNK), appears to be critically involved with acetaminophen and most likely several other hepatotoxins. , In turn, the balance between these factors can trigger the onset of cell death or facilitate protection of the cell, as discussed later.

Types of Cell Death

Apoptosis

Apoptosis is an energy-dependent, genetically programmed form of cell death that typically results in controlled deletion of individual cells. In addition to its major roles in developmental biology, tissue regulation, and carcinogenesis, apoptosis is important in toxic, viral, and immune-mediated liver injury. The ultrastructural features of apoptosis are cell and nuclear shrinkage, condensation and margination of nuclear chromatin, plasma membrane blebbing, and ultimately fragmentation of the cell into membrane-bound bodies that contain intact mitochondria and other organelles. Engulfment of these apoptotic bodies by surrounding epithelial and mesenchymal cells conserves cell fragments that contain nucleic acid and intact mitochondria. These fragments are then digested by lysosomes and recycled without release of bioactive substances. As a consequence, apoptosis in its purest form (usually found only in vitro) does not incite an inflammatory tissue reaction. The cellular processes that occur in apoptosis are often mediated by caspases, a family of proteolytic enzymes that contain a cysteine at their active site and cleave polypeptides at aspartate residues; non–caspase-mediated programmed cell death has also been described in experimental hepatotoxicity.

Apoptosis rarely if ever is the sole form of cell death in common forms of liver injury, such as ischemia-reperfusion injury, cholestasis, and toxic liver injury, all of which are typically associated with at least some necrosis and a hepatic inflammatory response. Whether activation of pro-death signals causes cell death depends on several factors, including pro-survival signals, the rapidity of the process, the availability of glutathione and ATP, and the role of other cell types. Some of these issues are discussed briefly here and are reviewed in more detail elsewhere. ,

The operation of hepatocellular apoptosis can be determined by detection of the caspase-3-cleaved fragmentation product (M30) of cytokeratins 8 and 18 that is specific to hepatocytes. Hepatocytes undergo apoptosis when pro-apoptotic intracellular signaling pathways are activated, either because of toxic biochemical processes within the cell (intrinsic pathway) or because cell surface receptors are activated to transduce cell death signals (external pathway). Pro-apoptotic receptors are members of the TNF receptor superfamily, which possess a so-called death domain. These receptors include Fas, for which the cognate ligand is Fas-ligand (Fas-L), TNF-R1 receptor (cognate ligand is TNF), and T NF- r elated a poptosis- i nducing l igand (TRAIL) receptors (cognate ligand is TRAIL). In addition to model hepatotoxins such as the quinone, menadione, and hydrogen peroxide, some drugs (e.g., acetaminophen, plant diterpenoids) have been shown to be converted into pro-oxidant reactive metabolites, thereby initiating the following sequence: CYP-mediated metabolism to form reactive metabolites → glutathione depletion → mitochondrial injury with release of cytochrome c and operation of the mitochondrial membrane permeability transition → caspase activation → apoptosis.

Mitochondria play a pivotal role in pathways that provoke or oppose apoptosis. , , , In the external pathway, activation of the death domain of pro-apoptotic receptors recruits adapter molecules, Fas-associated death domain and TNF receptor-associated death domain, which bind and activate procaspase 8 to form the death-inducing signaling complex. In turn, caspase 8 cleaves Bid, a pro-apoptotic member of the B cell lymphoma/leukemia (Bcl-2) family, to tBid. Then, tBid causes translocation of Bax to the mitochondria, where it aggregates with Bak to promote permeability of the mitochondria. Release of cytochrome c and other pro-death molecules, including Smac (which binds caspase inhibitor proteins, such as inhibitor of apoptosis proteins [IAPs]) and apoptosis-inducing factor (AIF, also known as Apaf) allows formation of the “apoptosome,” which activates caspase 9 and eventually caspase 3 to execute cell death ( Fig. 88.1 ). Intracellular stresses in various sites release other mitochondrial permeabilizing proteins (e.g., Bmf from the cytoskeleton and Bim from the endoplasmic reticulum), whereas members of the Bcl-2 family, Bcl-2 and Bcl-xL, antagonize apoptosis and serve as survival factors by regulating the integrity of mitochondria; the protective mechanism is not yet fully understood but involves myeloid cell leukemia sequence 1 (Mcl-1). Stress-activated protein kinases, particularly JNK, are also pro-apoptotic, targeting Mcl-1 degradation and phosphorylating and inactivating the mitochondrial protective protein Bcl-xL.

Fig. 88.1, Apoptosis and necrosis pathways in mammalian cells. See text for details. Bcl , B-cell lymphoma/leukemia family (Bax, Bid, and Bcl-xL are members); DISC , death-inducing signaling complex; FADD , Fas-associated death domain; FLIP , FLICE-inhibitory proteins; IAP , inhibitor of apoptosis proteins; MPT , mitochondrial permeability transition; RIP , receptor-interacting protein; TNF , tumor necrosis factor; TNF-R1 , TNF receptor-1; TRADD , TNF receptor-associated death domain; TRAF2 , TNF receptor-associated factor-2; TRAIL , TNF-related apoptosis ligand.

Execution of cell death by apoptosis usually occurs via activation of caspase 3, but more than one caspase-independent pathway of programmed cell death has been described. Stresses to the endoplasmic reticulum can bypass mitochondrial events by activation of caspase 12, which in turn activates caspase 9 independently of the apoptosome. The final steps of programmed cell death are energy dependent. Therefore, depletion of ATP abrogates the controlled attempt at “cell suicide,” resulting instead in necrosis (see later) or an overlapping pattern that has been designated as “apoptotic necrosis” or “necrapoptosis.” , Furthermore, when apoptosis is massive, the capacity for rapid phagocytosis can be exceeded, and “secondary” necrosis can occur.

Intracellular processes and activation of pro-apoptotic death receptors are not mutually exclusive pathways of cell death in toxic liver injury. In fact, drug toxicity could predispose the injured hepatocyte to apoptosis mediated by TNF-R or Fas-operated pathways by several mechanisms, including blockade of nuclear factorkappa B (NF-ĸB), which usually is a hepatoprotective transcription factor in hepatocytes, and inhibition of purine and protein synthesis. Furthermore, activation of Kupffer cells (e.g., by endotoxin) and recruitment of activated inflammatory cells can increase production of TNF.

Caspase inhibition is an important protective mechanism against cell death. Such anti-apoptotic pathways include chemical blockade of the cysteine thiol group by nitric oxide (NO) or ROS and cellular depletion of glutathione. Protein inhibitors include IAP family members, heat shock proteins, and FLICE (caspase-8)-inhibitory proteins (FLIP). FLIP inhibit caspase-8 activation as a decoy for Fas-associated death domain binding. Bcl-2 and Bcl-X L inhibit mitochondrial permeability, whereas phosphatidylinositol 3-kinase/Akt phosphorylates caspase 9 and activates NF-ĸB.

Necrosis

In contrast to apoptosis, necrosis has been conceptualized as a relatively uncontrolled process that can result from extensive damage to the plasma membrane with disturbance of ion transport, dissolution of membrane potential, cell swelling, and eventually rupture of the cell. Drug-induced injury to the mitochondrion can impair energy generation, whereas membrane permeability transition can release stored Ca 2+ into the cytosol and perturb other ionic gradients. Mitochondrial enzymes are a particular target of NAPQI, the reactive metabolite of acetaminophen. This has been clearly demonstrated both in rodent models and in human acetaminophen hepatotoxicity, in which mitochondrial injury with fragmentation of nuclear DNA by the released endonucleases has been documented. The initial mitochondrial injury can also activate various signaling pathways (JNK, glycogen synthase kinase -3β), thereby leading to further mitochondrial dysfunction. Reye syndrome–like disorders (e.g., toxicity caused by valproic acid; some nucleoside analogs, such as fialuridine, didanosine, zidovudine, zalcitabine; and possibly “ecstasy”) may also result from mitochondrial injury. Mitochondrial injury can result in cell death by either apoptosis or necrosis , ; the type of cell death pathway may depend primarily on the energy state of the cell, as well as the rapidity and severity of the injury process. In the presence of ATP, cell death can proceed by apoptosis, but when mitochondria are de-energized, the mechanism of cell death is necrosis. This apparent dichotomy between cell death processes is probably artificial, and apoptosis and necrosis more likely represent the morphologic and mechanistic ends of a spectrum of overlapping cell death processes. ,

One important way in which necrosis differs from apoptosis is that uncontrolled dissolution of the cell liberates danger-activated molecular patterns (e.g., HMGB1) and macromolecular breakdown products, including lipid peroxides, aldehydes, and eicosanoids. The latter products act as chemoattractants for circulating leukocytes, which then partake in the inflammatory response in the hepatic parenchyma. Even before cell death occurs, oxidative stress produced during drug toxicity can up-regulate adhesion molecules and chemokines that are expressed or secreted by endothelial cells. These processes contribute to recruitment of the hepatic inflammatory response, which is prominent in some types of drug-induced liver disease. Lymphocytes, polymorphonuclear leukocytes (neutrophils and eosinophils), and macrophages also may be attracted to the liver as part of a cell-mediated immune reaction.

Role of Oxidative Stress

Although severe oxidative stress in hepatocytes, particularly when focused on mitochondria, is likely to induce necrosis, lesser (or more gradual) exposure can trigger apoptosis because ROS and oxidative stress can activate Fas signaling, JNK and other kinases, p53, and microtubular assembly and impair protein folding, thereby resulting in an unfolded protein response by the endoplasmic reticulum.

Oxidative stress also may amplify cell death processes by uncoupling of the mitochondrial respiratory chain, release of cytochrome c, or massive oxidation and export of glutathione (intact glutathione is required for Fas signaling). Conversely, oxidative stress may protect against apoptosis in some circumstances through inhibition of caspase or activation of NF-ĸB. As a result of these opposing effects, predicting the consequences of hepatic oxidative stress in terms of liver injury is not straightforward.

Role of Hepatic Nonparenchymal Cells and the Innate Immune Response

In addition to migratory cells, activation of nonparenchymal liver cell types is likely to play an important role in drug and toxin-induced liver injury. Kupffer cells function as resident macrophages and antigen-presenting cells, whereas dendritic cells and natural killer (NK) T cells are also resident in the liver and play a role in antigen processing and innate immunity. Some of the toxic effects of activated Kupffer cells, as well as of recruited leukocytes, may be mediated by release of cytokines, such as TNFα, interleukin (IL)-1β and Fas-L, which under some circumstances can induce cell death in hepatocytes by apoptosis or necrosis. In addition, activated Kupffer cells release ROS, nitroradicals, leukotrienes, and proteases. It has been suggested, however, that the sterile inflammatory response may aid in clearing cell debris and pave the way for tissue repair.

Endothelial cells of the hepatic sinusoids or terminal hepatic veins are vulnerable to injury by some hepatotoxins because of their low glutathione content. Such hepatotoxins include the pyrrolizidine alkaloids, which are an important cause of the sinusoidal obstruction syndrome (hepatic veno-occlusive disease). Other types of drug-induced vascular injury may be caused primarily by involvement of the sinusoidal endothelial cells.

Hepatic stellate cells are the principal liver cell type involved in matrix deposition in hepatic fibrosis. Stellate cells are activated in methotrexate-induced hepatic fibrosis. The possibility that vitamin A, ROS, or drug metabolites can transform stellate cells into collagen-synthesizing myofibroblasts is of considerable interest.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here