Hepatotoxicity of Dermatologic Drug Therapy


Questions

  • Q62.1 Concerning various categories of liver disease, what percentage of patients likely have drug causation: (1) adults with hepatitis, (2) adults with hepatitis at least 50-years-old, (3) all patients with fulminant hepatitis? (Pg. 678, Table 62.1 )

  • Q62.2 What are several of the drugs removed from the United States market because of severe liver toxicity? (Pg. 678)

  • Q62.3 What is the purpose of the metabolic steps of biotransformation and detoxification? (Pgs. 678, 679 )

  • Q62.4 What are some of the enzymes involved with the phase I (primarily oxidation) and phase II (conjugation) metabolic steps of drug biotransformation (and which enzymes have polymorphisms)? (Pg. 679 , Table 62.2 )

  • Q62.5 What are several of the mechanisms involved in the pathogenesis of drug-induced liver injury (DILI) are caused by reactive metabolic intermediates? (Pg. 680 , Box 62.3 )

  • Q62.6 What is typical timing of DILI concerning (1) onset of the reaction after the drug therapy was initiated, and (2) time before resolution after drug is stopped? (Pg. 681)

  • Q62.7 Which cytochrome P-450 (CYP) isoforms currently have predictive tests for polymorphisms commercially available? (Pg. 682)

  • Q62.8 What are several of the reasons that clinical trials before a drug’s approval for marketing may not detect many of the relatively uncommon idiosyncratic adverse liver events caused by drugs? (Pg. 683)

  • Q62.9 What are four categories of DILI of greatest relevance to dermatologists (and what are several drugs that induce these patterns of liver injury)? (Pg. 683 , Table 62.7 )

  • Q62.10 For which systemic drugs prescribed by dermatologists is the need to monitor for liver toxicity not widely‘ known’? (Pg. 684x2)

  • Q62.11 What are the four steps of the basic algorithm proposed for causality determination in DILI (and why must the ‘rechallenge’ step be used very selectively)? (Pg. 685)

  • Q62.12 What do various liver ‘function’ tests truly evaluate in DILI? (Pg. 686 , Table 62.10 )

Abbreviations used in this chapter

AST a

Aspartate aminotransferase

ALT b

Alanine aminotransferase

Amox

Amoxicillin

CYP

Cytochrome P-450 enzymes

DAMP

Damage-associated molecular patterns

DHS

Drug hypersensitivity syndrome

DILI

Drug-induced liver injury

DRESS

Drug reaction with eosinophilia and systemic symptoms

EBV

Epstein-Barr virus

GGT

γ-Glutamyl transpeptidase

GST

Glutathione S -transferase

INR

International normalized ratio

LDH

Lactate dehydrogenase

LFT

Liver function tests

MCV

Mean corpuscular volume (for complete blood counts)

MHC

Major histocompatibility complex (antigens)

MTX

Methotrexate

NASH

Nonalcoholic steatohepatitis

NSAID

Nonsteroidal anti-inflammatory drug(s)

NAT 2

N -acetyl transferase 2

P3NP

Amino-terminus type III procollagen peptide

RA

Rheumatoid arthritis

RUQ

Right upper quadrant

SGOT a

Serum glutamate oxaloacetic transaminase

SGPT b

Serum glutamate pyruvate transaminase

SOD

Superoxide dismutase

TMP/SMX

Trimethoprim-sulfamethoxazole combination

UDP

Uridine diphosphate (UDP glucuronyl transferase)

Definitions Used in Chapter
Term Definition
Transaminases Collectively the enzymes AST/SGOT and ALT/SGPT
Transaminitis Elevation of AST/SGOT or ALT/SGOT levels up to twofold above upper normal values
Toxic hepatitis Either threefold elevation of transaminase levels, symptoms of hepatitis, or typically both
Liver failure Severely reduced synthetic and metabolic function of the liver
Polymorphism Differences in enzyme activity caused by genetic differences between various individuals
Enzyme variability Differences in enzyme activity in spite of similar genotypes
Isoform A specific cytochrome P-450 (CYP) enzyme; such as 2D6, 3A4, etc.
Idiosyncratic Drug reactions which are unpredictable, and which occur independent of drug dose
Toxicity Drug reactions which are predictable, given an adequately high dose of a few specific drugs
Neoantigens ‘New antigens’; because of covalent bonding of reactive intermediates to proteins, lipids, DNA
Steatosis Fat deposition in the liver; subtypes microvesicular and macrovesicular
Azalides Subset of macrolide antibiotics, including azithromycin
Statins HMG CoA-reductase inhibitors, such as lovastatin and atorvastatin
Threshold of concern Laboratory test elevation above a specified level, beyond which there is increased likelihood of a more serious reaction; abnormalities of this type are almost always reversible
Critical level Laboratory test elevation to an even greater level, beyond which there is dramatically increased likelihood of a severe reaction; lower likelihood of complete reversibility

Introduction

The importance of drug-induced liver injury (DILI) (drug hepatotoxicity) is illustrated by the following statistics: Q62.1

  • 1.

    Drugs are estimated to be responsible for 10% of cases of hepatitis in adults.

  • 2.

    Drugs are responsible for about 40% to 50% of cases of hepatitis in adults at least 50 years old.

  • 3.

    It is estimated that at least 25% of all cases of fulminant hepatitis are caused by drugs.

  • 4.

    Liver failure, when it is drug induced on an idiosyncratic basis, is fatal 75% to 80% of the time.

The vast majority of currently available systemic drugs have been reported, at least on rare occasions, to induce some degree of hepatotoxicity.

Several important questions must be addressed. Why is the liver a frequent site of important systemic drug reactions? Why do certain drugs induce significant hepatotoxicity on a relatively frequent basis? Why do individual patients selectively experience clinically significant hepatotoxicity from a given drug, which the vast majority of patients tolerate very well?

In this chapter, important general concepts regarding our current knowledge of DILI are summarized. Insightful answers to the previous questions and many more issues will be addressed as well. The focus is on four subsets of DILI most pertinent to practicing dermatologists, although physicians from all fields of medicine, who read this chapter, will glean drug safety principles of value to a wide variety of patients. These four most common subsets of DILI include: (1) acute hepatitis, (2) immunoallergic hepatitis or drug hypersensitivity syndrome (DHS), also known as drug reaction with eosinophilia and systemic symptoms (DRESS), (3) pure or bland cholestasis, and (4) nonalcoholic fatty liver disease, with associated fibrosis.

The ultimate motivation for writing this chapter involves sharing principles that will help prevent unnecessary morbidity and rare deaths from systemic drugs, while maximizing the overall safety and efficacy of important systemic drugs commonly used in dermatology. Q62.2 Several drugs that are no longer available to clinicians, because of severe hepatotoxicity, are listed in Table 62.1 . Liver transplantations performed because of severe, irreversible liver disease induced by drugs, such as ketoconazole and methotrexate (MTX), are overall avoidable. Avoiding the major cost of this life-saving technology is an important goal as well.

Table 62.1
Some Drugs Withdrawn From Market Caused By Severe Hepatotoxicity
Generic Name Trade Name Drug Category
Troglitazone Rezulin Insulin sensitizer; thiazolidinedione
Bromfenac Duract NSAID
Zomepirac Zomax NSAID
Ticrynafen Selacryn Diuretic with uricosuric properties
Benoxaprofen Oraflex NSAID
NSAID , Nonsteroidal anti-inflammatory drug.

The Liver and Drug Metabolism

Hepatic Drug Metabolism

The liver serves as the major site of metabolism for most drugs. Q62.3 The overall goal of this hepatic drug metabolism is to convert pharmacologically active, relatively lipophilic drugs, into inactive, relatively hydrophilic metabolites, to facilitate renal or biliary excretion. With these important functions, it should come as no surprise that the liver could be the ‘victim’ of aberrances in these metabolic processes. Furthermore, the hepatocyte is the hepatic cell of central importance in this drug metabolism; it follows that the hepatocyte is the most common liver cell ‘victimized’ by DILI. Important general concepts regarding hepatic drug metabolic systems are listed in Box 62.1 .

Box 62.1
Key Points About Hepatic Drug Metabolic Systems

Baseline Pharmacologic Principles

  • The vast majority of drugs have lipophilic properties

  • This lipophilicity is essential for drugs to effectively cross various lipid membranes to arrive at the site of desired pharmacologic activity

General Goals of Drug Metabolism

Biotransformation

  • Major goal of this process is conversion of the drug from lipophilic to hydrophilic properties

  • Increased hydrophilicity is essential for drugs to be excreted by either renal or biliary routes

Detoxification

  • Overall goal is to avoid local or distant damage from reactive intermediates created during the biotransformation process

  • These reactive metabolic intermediates are usually highly electrophilic compounds, which readily form covalent bonds with nearby proteins, DNA, and lipids

  • In the vast majority of patients receiving potentially hepatotoxic drugs, these detoxification systems are adequate to allow safe drug administration, without significant risk to the liver

Phases of Drug Metabolism

Phase I (predominantly Oxidative Reactions)

  • Cytochrome P-450 (CYP) enzymes—these reactions are largely accomplished by just 5–6 isoforms, which metabolize the vast majority of drugs used in humans

  • Majority of reactions are oxidative, adding a hydroxyl group, which provides a binding site for subsequent conjugation reactions

  • The aforementioned oxidative reactions result in only a small increase in hydrophilicity

  • Other phase I reactions include dealkylation and halogenation

Phase II (conjugation Reactions)

  • Enzymes involved include N-acetyl transferases, UDP glucuronyl transferase, and sulfotransferases

  • Major enzymes involved attach a large polar side chain to the oxidative site of phase I reaction

  • The phase II conjugation reactions may simultaneously result in a marked increase in hydrophilicity and may detoxify the reactive intermediates as well

DNA , Deoxyribonucleic acid; UDP , uridine diphosphate.

Q62.3 The human body has a complicated and intriguing system for biotransformation (conversion from lipophilic to hydrophilic molecules) and detoxification (conversion of reactive metabolic intermediates to more stable molecular compounds) of a multitude of very heterogeneous pharmaceutical molecules/drugs. These systems for biotransformation and detoxification of drugs logically evolved and developed long before all current drugs were developed. The same systems play an important role in the metabolism of a number of molecules (including steroid molecules, such as corticosteroids (CS), sex steroids, bile acids) and detoxification of various potential environmental insults (such as by ‘quenching’ free oxygen and superoxide radicals).

Increased activity of the biotransformation enzymes (because of cytochrome P-450 [CYP] inducers) can produce an increased quantity of reactive, electron-scavenging metabolic intermediates. The process of converting a pharmacologically active lipophilic drug to a reactive intermediate is known as bioactivation . Likewise, abnormalities of the detoxification systems may allow these reactive intermediates to wreak havoc locally in the liver, or to create problems through indirect mechanisms with important systemic consequences. As long as there is a reasonable balance between bioactivation and detoxification, no significant drug-induced liver toxicity occurs. Spielberg and associates developed the lymphocyte cytotoxicity assay as a potentially important laboratory method of assessing an individual’s detoxification capacity, for a limited number of drugs. The reader is encouraged to learn more about these hepatic metabolic systems from several excellent reviews.

Polymorphisms

Q62.4 The question regarding the varying probability of specific individuals experiencing DILI can in part be answered by the presence of genetic polymorphisms in important hepatic biotransformation and detoxification enzymes ( Table 62.2 ). The tremendous variability of enzyme activity is explained in part by distinct heritable deoxyribonucleic acid (DNA) mutations (polymorphisms) of both phase I and phase II hepatic enzymes. There are five CYP isoforms primarily involved in the pathogenesis of drug-induced hepatotoxicity (CYP1A2, 2C9, 2D6, 2E1, 3A4). CYP2C19 is not known to be responsible for producing reactive metabolites with hepatotoxic potential.

Table 62.2
Phase I/Phase II Metabolic Enzymes and Detoxification Systems
Enzyme Examples Polymorphism/Variability Comments
Phase I Enzymes
CYP1A2 a Variability up to ∼30-fold Induced by omeprazole, smoking, charbroiled meat
CYP2C9 Polymorphism Induced by various aromatic anticonvulsants, rifampin
CYP2C19 b Polymorphism Is not responsible for drug-induced liver disease
CYP2D6 c Polymorphism ∼10% Caucasians have essentially no 2D6 activity
CYP2E1 Variability up to ∼30-fold Induced by ethanol, isoniazid
CYP3A4 a Variability up to ∼30-fold Induced by aromatic anticonvulsants, rifampin
Phase II Enzymes
UDP glucuronyl transferases Polymorphism Result is glucuronidation, increased hydrophilicity
Sulfotransferases Polymorphism Result is sulfonation, increased hydrophilicity
N-acetyl transferase 1 and 2 Polymorphism NAT 2 50% Caucasians ‘slow acetylators’
Detoxification Systems
Epoxide hydrolases Polymorphism Possible role aromatic anticonvulsant-induced DRESS.
Glutathione S-transferases Polymorphism Critical detoxification enzyme for many drugs
Free radical quenchers Uncertain SOD, catalase, vitamin C, vitamin E
CYP , Cytochrome P-450, MEGX , monoethylglycinexylidide; NAT 2 , N-acetyl transferase 2; SOD , superoxide dismutase; UDP , uridine diphosphate.

a Variability in 1A2 is assessed by metabolism of caffeine; variability in 3A4 can be assessed by erythromycin breath test or MEGX assay, studying lidocaine metabolism.

b Polymorphism in 2C19 is assessed by metabolism of S-mephenytoin (an anticonvulsant).

c Polymorphism in 2D6 is assessed by metabolism of debrisoquine (an antiarrhythmic agent); overall with at least 50-fold variation in activity when comparing various individuals.

The prototype enzymes with the most significant polymorphisms are CYP2D6, CYP2C9, and CYP2C19. Individuals with ‘low activity’ (‘poor metabolizers’) for CYP2D6 may have at least 50 times less enzyme activity than individuals with the ‘high-activity’ (‘extensive metabolizers’ or ‘ultrarapid metabolizers’) phenotype for the same CYP isoform. Such wide variation in activity can help explain why some individuals experience excessive and prolonged daytime sedation following a single 10-mg dose of doxepin (a CYP2D6 substrate) taken at bedtime, whereas other individuals tolerate and have very little sedation with doxepin doses titrated up to 200 to 300 mg each night. By the same logic, other potentially hepatotoxic drugs that are substrates for CYP2D6 could have excessive drug levels in patients who inherited the low-activity phenotype. Even without genetic polymorphisms, other enzymes (such as CYP3A4) can have significant variability (up to 30-fold) of enzyme activity. There are rare allelic variation (polymorphism) in CYP3A4, but are too uncommon to be of clinical signicance.

Even more important to the topic of drug hepatotoxicity is the potential for CYP enzyme inducers to increase the enzyme activity of various CYP isoforms. Such increased activity of a given phase I oxidative CYP isoform will increase the quantities of various reactive intermediates derived from drug substrates. The risk of DILI is greatest when there is an accompanying genetic (such as N -acetyl transferase 2 (NAT 2 ) ‘slow acetylators’) or acquired (decreased glutathione levels caused by human immunodeficiency virus [HIV] infection) defect in a phase II conjugation and/or detoxification system.

Significant controversies and unanswered questions abound in this subject area. One such issue is the presence of the slow acetylator phenotype in up to 50% of Caucasians, whereas only a very small percentage of patients with this phenotype experience reactions thought to be caused by this polymorphism (such as drug-induced lupus erythematosus and isoniazid hepatotoxicity). DRESS has been postulated to be caused by genetically based reduced activity of epoxide hydrolase, leading to increased quantities of unstable epoxide intermediates from anticonvulsants, such as phenytoin, phenobarbital, and carbamazepine. This explanation has since been challenged.

Mechanisms of Drug Hepatotoxicity

General Mechanisms Involved

The most well-accepted hypothesis explaining idiosyncratic DILI has two components. First, there must be the presence of reactive metabolic intermediates, which are electrophilic compounds that rapidly form covalent bonds to various cellular components, if not equally rapidly detoxified. These covalent bonds result in various structural and functional changes, depending on the affected molecule ( Table 62.3 ). These molecular components (with which the reactive intermediates bind) include various proteins (including the CYP isoforms), DNA, and lipids, with consequences, as detailed in Table 62.4 . The presence of CYP inducers can significantly increase the quantities of these electrophilic reactive intermediates ( Table 62.5 ). Second, there generally must be a defect in the cellular detoxification systems for there to be an adequate presence or persistence of these reactive intermediates, to result in significant hepatic pathology. In the absence of the CYP inducers and/or with normal detoxification systems, significant DILI from these metabolic products is distinctly uncommon.

Table 62.3
Molecular Targets Involved with Drug-Induced Liver Disease
Molecule Consequence Comments
Various proteins Neoantigen formation Reactive metabolites induce a change in structure or conformation
CYP isoforms Neoantigen formation CYP especially vulnerable, caused by proximity to reactive metabolites
DNA Apoptosis or necrosis Either route results in cell death
Lipids Lose membrane integrity Result of lipid peroxidation by reactive metabolites
CYP , Cytochrome P-450, DNA , deoxyribonucleic acid.

Table 62.4
Cellular and Structural Targets Involved with Drug-Induced Liver Disease
Cell or Structure Category of Reaction Representative Drug Etiologies
Hepatocytes Hepatocellular necrosis Ketoconazole, minocycline
Bile ducts, bile canaliculi Cholestasis Erythromycin estolate (in adults)
Endothelial cells, sinusoids Veno-occlusive Cyclophosphamide (very high-doses)
Ito cells (fat storage cells) Steatosis → Fibrosis Methotrexate

Table 62.5
Several Cytochrome P-450 Inducers and Inhibitors That Also Have Hepatotoxic Potential
Drug Isoform Reaction Category
Cyp Inducers a
Phenytoin Various DRESS
Carbamazepine Various DRESS
Rifampin 3A4 Hepatocellular toxicity
Isoniazid 2E1 Hepatocellular toxicity
Ethanol 2E1 Hepatocellular toxicity
Cyp Inhibitors
Ketoconazole 3A4 Hepatocellular toxicity
Itraconazole b 3A4 Hepatocellular toxicity
Fluconazole b 2C9 Hepatocellular toxicity
Erythromycin estolate c 3A4 Cholestasis
CYP , Cytochrome P-450.

a In contrast to other enzyme inducers, which are anticonvulsants, phenobarbital has minimal risk for hepatotoxicity.

b Itraconazole and fluconazole have far less risk for hepatocellular toxicity compared with ketoconazole.

c Vary rare hepatotoxicity from other macrolides, including the azalide subset.

Probably the most important component of the detoxification enzyme repertoire is the glutathione and glutathione S -transferase system (GST). GST is an enzyme with a genetic polymorphism, with glutathione serving as a cofactor and various electrophilic drug metabolites serving as GST substrates. Glutathione can be depleted in the process of electron scavenging, particularly with an unusually heavy load of reactive intermediates, such as with an acetaminophen overdose. The electron scavenging capabilities of this GST/glutathione system can be further depleted by excessive ethanol consumption, HIV infection, and malnutrition. It stands to reason that drug hepatotoxicity, let alone drug reactions in general, will be increased in these clinical settings.

Q62.5 Two potential outcomes are thought to occur when there are excessive quantities of reactive, electrophilic intermediates. First, is the potential for direct toxicity to neighboring cells (most notably the hepatocyte) from protein, DNA, and lipid biomembrane alterations resulting from covalent binding to these reactive intermediates. An important final common pathway leading to cell necrosis or apoptosis is likely a disruption in calcium regulation, leading to alterations in the actin cytoskeleton and changes in cell membrane integrity. Binding of fas (on the cell membrane of ‘damaged’ hepatocytes) with fas ligand (on antigen-processing cells) leads to activation of the caspase cascade, and subsequent apoptosis, is an important pathway of cell destruction as well. This pathway may also be activated through the tumor necrosis factor (TNF)-α receptor. Second, the covalent binding of the reactive intermediates to various proteins leads to the development of new antigens (neoantigens). These neoantigens can include various CYP isoforms that logically are ‘in the neighborhood’, when the reactive intermediates are created. It is suggested that these reactive intermediates cause hepatocyte stress or death leading to release of Damage Associated Molecular Patterns (DAMP), which in turn activate innate immune cells, particularly Kupffer cells in the liver. The activated innate immune cells then release cytokines and chemokines that draw inflammatory cells into the liver, a process thought to be a prerequisite for a targeted adaptive immune attack on the liver. The end results are potentially either a direct humoral or cellular response to these neoantigens or an immunologic cross-reactivity, with structurally similar distant antigens, or both. The complexities of major histocompatibility complex (MHC) molecules involved with antigen presentation, along with the uncertainties, regarding the overall pathogenesis of autoimmune disorders, are beyond the scope of this chapter. Also, given the paucity of dermatologic drugs that induce cholestasis, the mechanisms involved with this process are not discussed in this chapter (see Erlinger and Church articles in the Bibliography).

Toxic Versus Idiosyncratic Reactions

It is worth reminding the reader of an important distinction at this point. Direct drug-induced toxicity to the liver results from excessive drug levels from very few specific drugs. The classic biochemistry example is the effect of carbon tetrachloride on the liver. The classic pharmacologic example is an acetaminophen overdose, although pharmacologic doses of acetaminophen in the presence of a CYP2E1 inducer (chronic excessive ethanol consumption) can produce a similar result. By definition, toxicity here means that virtually every individual will have a reaction to carbon tetrachloride or acetaminophen, if excessive quantities of these molecules are administered. In contrast, the term idiosyncratic suggests that only a distinct minority of individuals will experience DILI from selected drugs, and that the reaction will occur independent of drug dose. Under this heading is where the vast majority of hepatotoxicity caused by drugs occurs. What may be somewhat confusing is that the word toxicity is commonly used in the setting of idiosyncrasy as well. Idiosyncrasy can be divided into ‘metabolic’ idiosyncrasy (because of the local toxic effects of locally formed reactive intermediates) and ‘immunologic’ idiosyncrasy (local damage from an immunologic response, caused by neoantigens formed in response to these reactive intermediates). It is with these two dichotomies clearly in mind (‘toxic’ vs. ‘idiosyncratic’ reactions; ‘metabolic idiosyncrasy’ vs. ‘immunologic idiosyncrasy’) that the reader should continue through this chapter ( Q62.6 Box 62.2 ).

Box 62.2
Generalizations Regarding Drug-Induced Liver Disease

Mechanism (regarding Predictability)

  • Vast majority are idiosyncratic , that is unpredictable and without a clear relationship to drug dose

  • Small percentage are toxic , which are predictable and dose dependent (at doses above pharmacologic levels)

Timing

  • Onset—majority occur between days 15–90, after the initiation of drug therapy; generally have a much more gradual onset compared with potentially serious hematologic drug reactions

  • Resolution—most will have a marked improvement within 15 days after drug cessation

  • Aforementioned timing for hepatocellular toxicity, hypersensitivity syndrome/DRESS, and cholestasis type reactions

  • Steatosis progressing to fibrosis is much slower in onset (over years), and more insidious in progression

Reversibility

  • Most reactions are completely reversible if detected early (within days to a few weeks)

  • Some reactions are not fully reversible if detected relatively late (delay of many weeks to months)

  • Possible outcomes if significantly delayed diagnosis—death, liver failure requiring liver transplantation, severe fibrosis or cirrhosis, loss of some degree of liver function indefinitely a

    a In contrast to the kidneys, the liver has a tremendous reserve capacity—marked liver damage is possible before most reliable laboratory indicators of reduced liver function (↓ albumin, ↑ international normalized ratio) become abnormal.

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