35575737 drug induced liver disease 2007

Upload: eric-gibson

Post on 14-Apr-2018

221 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    1/226

    DRUG-INDUCED LIVER DISEASE

    CONTENTS

    Preface xiWilliam M. Lee

    Mechanisms of Drug-Induced Liver Disease 459Basuki K. Gunawan and Neil Kaplowitz

    Drug-induced liver injury depends initially on development ofhepatocyte stress and cell death, which can be induced directly byparent drugs or by toxic metabolites. Hepatocyte stress can lead to

    activation of built-in death programs for apoptosis or necrosis.Subsequently, the innate immune systems participation isrecruited. The interplay between proinflammatory and anti-inflammatory components of innate immune system determinesthe outcome of drug-induced liver injury. Both environmentalfactors and genetic differences in cellular responses to stress andthe innate immune response may account for different susceptibil-ities between individuals to drug-induced liver injury.

    Causality Assessment of Drug-Induced Hepatotoxicity:

    Promises and Pitfalls 477Max A. Shapiro and James H. Lewis

    Drug-induced liver injury is the leading cause of acute liver failurein the United States, but the ability to ascribe hepatic injuryconfidently to a specific drug remains a challenging and oftendifficult pursuit. This article explores the ongoing challengesinherent in what is currently a clinical process of eliminationmade in the attempt of assigning causality in drug-induced liverinjury. In particular, it points out the shortcomings and pitfalls thatoften limit the applicability of the causality-assessment method-

    ologies currently in use.

    VOLUME 11 NUMBER 3 AUGUST 2007 v

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    2/226

    Drug Hepatotoxicity from a Regulatory Perspective 507John R. Senior

    This article summarizes problems of drug-induced liver injury(DILI), as seen from the perspective of the Food and DrugAdministration (FDA). After brief consideration of the scope ofFDA activities and processes of new drug development and reviewfor possible approval of products for clinical use and marketing,some of the perceived current problems in detection, confirmation,close observation, differential diagnosis, and follow-up of cases ofpossible DILI in controlled clinical trials are described. Readers areinvited to consider possible solutions to the many problems of DILI,propose ways to support research in the field, and keep abreast ofprogress by visiting the web site at www.fda.gov/cder/livertox.

    Acetaminophen Hepatotoxicity 525Anne M. Larson

    Acetaminophen is a commonly used antipyretic and analgesicagent. It is safe when taken at therapeutic doses; however,overdose can lead to serious and even fatal hepatotoxicity. Theinitial metabolic and biochemical events leading to toxicity havebeen well described, but the precise mechanism of cell injury anddeath is unknown. Prompt recognition of overdose, aggressivemanagement, and administration ofN-acetylcysteine can minimize

    hepatotoxicity and prevent liver failure and death. Liver trans-plantation can be lifesaving for those who develop acute liverfailure.

    Hepatotoxicity Due to Antibiotics 549Julie E. Polson

    Antimicrobial drugs are important causative agents in idiosyn-cratic drug-induced liver injury (DILI). As with idiosyncratic DILIin general, antibiotic-induced liver injury is rare but difficult to

    diagnose and almost impossible to predict. Diagnosis requiresawareness of possible causal agents, vigilance in monitoringsymptoms and sometimes biochemical tests, attention to carefulhistory taking and establishing temporal association, and exclusionof competing etiologies. In most instances, patients with antibiotic-associated DILI recover if the offending agent is withdrawn in atimely fashion.

    Nonsteroidal Anti-Inflammatory DrugInduced Hepatotoxicity 563Guruprasad P. Aithal and Christopher P. Day

    Nonsteroidal anti-inflammatory drugs are among the mostcommon drugs associated with drug-induced liver injury, withan estimated incidence of between 3 and 23 per 100 000 patientyears. Nimesulide, sulindac, and diclofenac seem to be associatedwith the highest risk and the only risk factor consistently identified

    vi CONTENTS

    http://www.scielo.br/scielo.php?script=sci_arttext%26;;pid=S0066-782X2005002400007%26lng=en&nrm=isohttp://www.scielo.br/scielo.php?script=sci_arttext%26;;pid=S0066-782X2005002400007%26lng=en&nrm=iso
  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    3/226

    is the concomitant use of other hepatotoxic drugs. Diclofenac-induced liver injury is a paradigm for drug-related hepatotoxicity.Recent studies suggest that genetic factors favoring the formationand accumulation of the reactive acylglucuronide metabolite ofdiclofenac and an enhanced immune response to the metabolite-

    protein adducts are associated with increased susceptibility tohepatotoxicity.

    Herbal Hepatotoxicity 577Leonard B. Seeff

    There is appropriate concern about the potential risk forhepatotoxicity from herbal products because they are unregulatedand therefore not standardized with regard to their contents. Thisis particularly the case for the crude herbals that are commonlyformulated as a mixture, so that their ingredients may beambiguous and even contain harmful contaminants. Presentedhere is an overview of the more commonly recognized herbalproducts that have been reported to be associated with liver injury.Although many of them are clearly implicated, there are some,particularly those identified solely through an occasional casereport, for which the relationship is uncertain.

    Lipid-Lowering Agents That Cause Drug-Induced

    Hepatotoxicity 597Sidharth S. Bhardwaj and Naga Chalasani

    The effort to reduce cardiovascular risk factors, including hyper-lipidemia, has led to the increased use of lipid-lowering agents.Hyperlipidemic patients often have underlying fatty liver disease,however, and thus may have elevated and fluctuating liverbiochemistries. Therefore, caution should be applied beforeattributing elevated liver tests to lipid-lowering agents. Dataindicate that patients who have chronic liver disease andcompensated cirrhosis should not be precluded from receiving

    statins to treat hyperlipidemia. Several recent studies and expertopinion currently fully endorse statin use in patients who havenonalcoholic fatty liver disease and other chronic liver disease ifclinically indicated.

    Drug-Induced Liver Injury Associated with HIV Medications 615Mamta K. Jain

    Antiretroviral therapy (ART) for HIV infection frequently has beenassociated with elevated liver enzyme levels. Determining the

    cause of elevated liver enzyme levels in patients who have HIV isdifficult because ART usually consists of three different drugs,patients may be taking additional hepatotoxic medications andpatients who have HIV often suffer from other liver diseases.Several agents, however, are recognized as having noteworthy andspecific patterns of toxicity. This article reviews the different HIV

    CONTENTS vii

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    4/226

    drug classes, incidence of elevated liver enzyme values by classand by individual drug, risk factors, specific toxicities, and possiblemechanisms of injury.

    Cancer Chemotherapy I: Hepatocellular Injury 641Edmundo A. Rodriguez-Frias and William M. Lee

    Although hepatotoxicity is a frequent concern with all medica-tions, chemotherapeutic agents are more often implicated incausing liver damage than most other drug classes. In manyinstances, these reactions are considered dose related becausecytotoxic therapy directed at rapidly growing cancer cells mayreadily impact hepatocytes even though they are dividing moreslowly. Because the stakes (remission of cancer) are high, so arethe risks that the oncologist and the patient are willing to assume.The dose of many chemotherapeutic agents is limited by the toxiceffects on the lungs, bone marrow, kidneys, and gastrointestinalsystem, including the liver. An awareness of the toxic potential ofeach chemotherapeutic agent is necessary before initiation of newoncologic treatments.

    Cancer Chemotherapy II: Atypical Hepatic Injuries 663Edmundo A. Rodriguez-Frias and William M. Lee

    Although chemotherapy generally is accompanied by regulartesting for liver enzyme abnormalities, atypical reactions mayoccur that escape ordinary detection, because hepatocyte injury isnot the primary event. The presence of fatty liver, mitochondrialchanges, and even biliary abnormalities can be associated withnormal or nearly normal liver enzyme levels. This article discussesunique aspects of liver damage associated with cancer chemo-therapy. These unique reactions merit special attention and aspecial vigilance from clinicians.

    Index 677

    viii CONTENTS

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    5/226

    FORTHCOMING ISSUES

    November 2007

    Hepatitis B VirusIra Jacobson, MD,Guest Editor

    February 2008

    Cholestasis

    Donald Jensen, MD,Guest Editor

    May 2008

    Hepatitis C Virus

    K. Rajender Reddy, MD,and David E. Kaplan, MD,Guest Editors

    RECENT ISSUES

    May 2007

    Liver Transplantation

    Paul Martin, MD,Guest Editor

    February 2007

    Non-Alcoholic Steatohepatitis and Non-AlcoholicFatty Liver Diseases

    Zobair M. Younossi, MD,Guest Editor

    November 2006

    Hepatitis C Virus Update

    David R. Nelson, MD,Guest Editor

    THE CLINICS ARE NOW AVAILABLE ONLINE!

    Access your subscription at:www.theclinics.com

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    6/226

    Preface

    Guest Editor

    Drug-induced liver injury (DILI) continues to play an important role in

    the evolution of modern medical care. DILI cases are by their natureiatrogenic but also often represent self-inflicted wounds, whether as a drug

    overdose such as acetaminophen or as an idiosyncratic reaction. Hepatotox-

    icity caused by drugs continues to feature prominently in consideration for

    drug approval by the United States Food and Drug Administration (FDA)

    and is the primary reason for drugs failing to be approved or being with-

    drawn after initial approval. Recent examples include ximelagatran, a highly

    anticipated anticoagulant that probably would have replaced warfarin if not

    for its failure to be approved in the United States and its subsequent volun-

    tary withdrawal in Europe. Other drugs withdrawn or significantly limitedin recent years include troglitazone and bromfenac (both withdrawn),

    trovafloxacin, nefazodone, and telithromycin (limited use, strong warnings).

    These unexpected calamities affect patients because the typical DILI case is

    severe and often fatal or requires liver transplantation. These events also

    impact the pharmaceutical industry. Drug approval becomes a hard-won

    prize that is becoming ever more elusive and costly. Difficulties in obtaining

    approval affect the cost of products that are approved as well. Despite many

    recent advances in the understanding of drug-induced liver injury, it still is

    not possible to identify safe new products readily and effectively.Against this background, I present this edition ofClinics in Liver Disease,

    an in-depth look at many of the current hot topics in the field of DILI. Each

    is directed toward a particular area of current interest and provides new in-

    sights not available when this topic was covered previously. It begins with an

    William M. Lee, MD

    1089-3261/07/$ - see front matter 2007 Elsevier Inc. All rights reserved.

    doi:10.1016/j.cld.2007.06.011 liver.theclinics.com

    Clin Liver Dis 11 (2007) xixii

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    7/226

    incisive discussion of mechanisms of drug-induced liver injury from Dr. Neil

    Kaplowitz, followed by a careful review of the unwieldy topic of assessing

    causality by Dr. James Lewis. Analyzing the drug approval process andexplaining it to the lay public was the task of Dr. John Senior, the FDAs

    inhouse hepatology consultantdwho better to take on the task?

    After these introductory articles of general interest, I asked experts to re-

    view some of the specific drug classes of most interest and the drug-induced

    liver damage they cause. These are the problem drug classes most commonly

    encountered by the practitioner. Each class has its own signature form of in-

    jury and its own specific associated issues. Acetaminophen poisoning is the

    primary cause of acute liver failure in the United States, actually exceeding

    all idiosyncratic drugs by about threefold in terms of numbers of directlyrelated deaths. The drug classes most commonly associated with liver injury

    are antibiotics and nonsteroidal anti-inflammatory drugs (NSAIDS). DILI

    caused by these agents is nicely covered by Drs. Polson and Drs. Aithal

    and Day, respectively. The NSAID article includes much new information

    concerning pharmacogenomics. The 3-hydroxy-3-methylglutaryl coenzyme

    A reductase inhibitors, otherwise known as statins, is a topic of great

    current interest, and no one is better qualified to review this topic than

    Dr. Chalasani, who has written many of the primary articles in the field.

    Another topic confusing to the public is the use of herbal medicationsand their risks. Dr. Seeff kindly has taken on the task of reviewing all we

    know in this area. More drug toxicity issues are raised by cancer chemother-

    apy and highly active antiretroviral (HAART) drugs than by any other

    classes. Dr. Jain has developed a highly practical but detailed review of

    the issues surrounding HAART therapy, and Dr. Rodriguez and I have

    tackled the problem of liver injury during oncologic treatments.

    All in all, I think drug-induced liver injury has become, in the last 5 years,

    an exciting field as exemplified by the articles included here. I wish to thank

    all those who labored so diligently to help create this issue, including mostspecifically the authors and their assistants as well as Kerry Holland and

    Norman Gitlin. Any blame comes to me, while any credit goes to them

    for what I personally think is a dynamite issue of Clinics in Liver Disease

    that will be of interest to the primary care provider, specialists, and the

    public at large, in short, anyone trying to make sense of this fascinating

    field.

    William M. Lee, MD

    Division of Digestive & Liver DiseasesUniversity of Texas Southwestern Medical School

    5959 Harry Hines Boulevard, HP.4.420

    Dallas, TX 75390-8887

    E-mail address: [email protected]

    xii PREFACE

    mailto:[email protected]:[email protected]
  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    8/226

    Mechanisms of Drug-Induced

    Liver Disease

    Basuki K. Gunawan, MD*, Neil Kaplowitz, MDResearch Center for Liver Disease, Keck School of Medicine, University of Southern

    California, 2011 Zonal Avenue, HMR 101, Los Angeles, CA 90033, USA

    Drug-induced liver disease is of great importance because it is the leading

    cause of acute liver failure in the United States[1]. The most common cause

    for drug-induced liver disease resulting in acute liver failure is acetamino-

    phen (APAP), with about half the cases reported to be accidental [1].

    Drug-induced liver disease is also a major reason for withdrawal of drugs

    during drug development and clinical use with major medical and economic

    consequences [2]. The understanding of the mechanism of drug-inducedliver injury is of great importance and may lead to prevention and better

    treatments.

    Types of drug-induced liver injury

    Drug-induced liver injury can be predictable; it is normally dose-

    dependent, reproducible in animal models, and presents after a short latency

    (hours to a few days). Drugs that cause this type of liver injury are usually

    identified during initial toxicology studies. These findings typically preclude

    further use as medication. Most drug-induced liver injuries, however, are

    unpredictable or idiosyncratic, although they may or may not be dose-

    dependent or reproducible in animal models [24]. This idiosyncratic type

    can be classified as allergic, presenting with fever, rash, eosinophilia, and

    rapidly recurring positive challenge; or as nonallergic, presenting without

    allergic features[2,5]. Idiosyncratic drug reactions occur in only a small per-

    centage of individuals who are exposed to the drug, ranging from 0.01% to

    1%, and hence signify the uniqueness of the susceptible individuals [2,3,5].

    Both genetic and environmental factors likely play a role in determining

    * Corresponding author.

    E-mail address: [email protected](B.K. Gunawan).

    1089-3261/07/$ - see front matter 2007 Elsevier Inc. All rights reserved.

    doi:10.1016/j.cld.2007.06.001 liver.theclinics.com

    Clin Liver Dis 11 (2007) 459475

    mailto:[email protected]:[email protected]
  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    9/226

    the occurrence of this idiosyncratic type. The factors involved in drug-

    induced liver disease are listed in Box 1and can be viewed as a sequential

    cascade.Detailed discussion of all these factors is beyond the limits of this article.

    Some of the key pathogenetic factors are highlighted.

    Drug metabolism in the liver

    The liver is the principal organ for metabolism and elimination of

    many drugs. Even though some drugs cause hepatotoxicity when the parent

    compound directly targets specific organelles, such as mitochondria or

    nuclei, most toxic drugs require metabolism to toxic metabolites [2]. Thereare three phases of drug metabolism in the liver. In phase I, drugs are

    metabolized by cytochrome P-450 enzymes. This process can generate toxic

    electrophilic chemicals and free radicals. In phase II, the parent drug or

    metabolites are conjugated with glutathione (GSH), sulfate, or glucuronide

    to produce water-soluble compounds. Consequently, the compounds can

    then be excreted from the body in bile or urine. The route of elimination

    is mainly determined by excretory transporters in the hepatocyte canalicular

    and sinusoidal membrane (phase III). Genetic polymorphisms or environ-

    mental factors, such as concomitant drugs and alcohol, can account for dif-ferences in phase I, II, and III drug metabolism between individuals and

    Box 1. Factors involved in drug-induced liver disease

    Exposure to toxic metabolites

    Regulation and expression of phase I, II, and III drug

    metabolism

    Direct consequences of toxic metabolite-covalent binding,

    oxidative stress Intracellular stress

    Organelle stress: mitochondria, endoplasmic reticulum,

    nucleus

    Stress kinases

    Metabolic disturbance (eg, fat accumulation)

    Inhibition of bile salt export pump: either cholestasis or

    possible sensitization to apoptosis by bile acid induced

    targeting of death receptors to the plasma membrane

    Cell death: programmed apoptosis versus programmednecrosis

    Participation of mitochondrial outer membrane

    permeabilization or mitochondrial permeability transition

    Innate immune response

    Adaptive immune response

    460 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    10/226

    may be determinants of susceptibility to idiosyncratic drug-induced liver

    injury by influencing the hepatic exposure to toxic metabolites.

    Biochemical events leading to drug-induced liver injury

    The toxic metabolites from drug metabolism can then either directly affect

    the biochemistry of the liver cells leading to cell damage or elicit an immune-

    mediated attack on the liver [4]. Drug metabolites can covalently bind to

    proteins, lipids, and DNA, and mediate cell death by inciting biochemical

    events, such as oxidative stress, GSH depletion, and redox changes, and lipid

    peroxidation. Consequently, these events may directly affect the functions

    of mitochondria, endoplasmic reticulum, microtubules, cytoskeleton, andnucleus leading to an overwhelming direct insult [2,4]. Alternatively, these

    events may lead to activation or inhibition of signaling kinases, transcription

    factors, and gene expression profiles, which may sensitize hepatocytes or

    cholangiocytes to the toxic effects of the innate immune system, such as

    cytokines and chemokines, which are activated by the initial liver injury.

    The toxic metabolite-initiated immune-mediated attack on the liver is

    mainly achieved by cytotoxic T cells through an apoptotic mechanism

    mediated by Fas ligand (FasL) and granzyme B/porin[5]. The initial trigger

    for activity of cytotoxic T cells involves haptenization. In hepatocytes, thedrug metabolites can covalently bind to macromolecules, such as proteins, re-

    sulting in altered proteins. This process is called haptenization and it can

    incite an immune-mediated attack because the hapten-protein complexes

    are recognized as neoantigens[6], which are internalized and processed by

    antigen-presenting cells and subsequently presented on the cell surface with

    major histocompatibility complex class II, leading to activation of helper

    T cells. The activation of helper T cells results in release of cytokines, which

    activate cytotoxic T cells, natural killer (NK) cells, and B cells and sub-

    sequently generates autoantibodies and cell-mediated immune responses[5,7,8].

    Many drugs that form reactive metabolites and undergo haptenization,

    however, do not cause drug-induced liver disease. It seems that hapteniza-

    tion alone might be insufficient to trigger an immune-mediated attack.

    One hypothesis that may explain this phenomenon is the danger hypothesis

    [9]. For the immune-mediated attack to occur, a second costimulatory dan-

    ger signal must present. This danger signal primes the immune system

    against the haptens in genetically susceptible individuals. The danger signal

    may include concomitant infection or inflammatory conditions, becausethere have been reports of increased allergic hepatotoxicity in patients

    with concomitant hepatitis B and C or HIV infection or with AIDS

    [1014]. Alternatively, the danger signal may be the low-grade stress or in-

    jury in hepatocytes. Supporting this hypothesis is the more frequent occur-

    rence of mild alanine aminotransferase abnormalities in response to drugs

    that more rarely induce overt allergic injury.

    461MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    11/226

    The neoantigen formed because of haptenization can trigger the produc-

    tion of autoantibodies directed against both the native and modified cyto-

    chrome P-450 protein [6,15]. The autoantibodies formed in liver diseasecaused by several different drugs are normally specific to the particular

    cytochrome P-450 isoenzymes that metabolize the drugs (Table 1). It is

    unclear, however, whether the anticytochrome P-450 autoantibodies medi-

    ate an immune attack on hepatocytes. These autoantibodies are found in the

    serum when the drug-induced liver disease is diagnosed and they decline and

    may disappear after recovery[6]. In addition, in drug-induced liver disease

    caused by halothane or tienilic acid, an antibody-dependent cell-mediated

    cytotoxicity has been demonstrated in vitro [16,17]. Autoantibodies are

    commonly found in patients without evidence of drug-induced liver disease,however, suggesting that their presence is not always associated with liver

    injury.

    Mode of cell death

    The outcome of the events initiated by toxic metabolites either through

    directly affecting the biochemistry in hepatocytes or immune-mediated

    response is cell death. The mode of cell death may be apoptosis or necrosis.

    Apoptosis involves shrinkage, nuclear disassembly, and fragmentation of

    the cell into discrete bodies with intact plasma membranes. The apoptotic

    cells are then rapidly phagocytosed by neighboring cells. In contrast, necrosis

    involves cell swelling and lysis as a result of profound loss of mitochondrial

    function and resultant ATP depletion[4,5]. The selection between apoptosis

    versus necrosis depends on several factors, including ATP status [5]. A more

    severe injury to mitochondria might favor necrosis, whereas a less severe in-

    jury to mitochondria without profound ATP depletion might favor apoptosis

    [5,18].

    The initiation of cell death usually involves the participation of mitochon-

    dria. The interaction of proapoptotic Bcl2 family members (Bid, Bax, Bak,

    Bmf, and Bim) and antiapoptotic members of this family (Bcl-2 and Bcl-XL)

    regulates the permeability of mitochondria[4]. The immune-mediated attack

    on hepatocytes involves the participation of the extrinsic death system, such

    as tumor necrosis factor-a(TNF-a) or FasL, which then bind to their death

    receptors leading to caspase 8 activation [4,19,20]. Caspase 8 subsequently

    Table 1Autoantibody targets in drug-induced liver disease

    Autoantibody target Drug

    CYP2C9 Tienilic acid

    CYP1A2 Dihydralazine

    CYP3A1 Anticonvulsants (eg, phenytoin)

    CYP2E1 Halothane

    462 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    12/226

    cleaves Bid leading to translocation of Bax to the mitochondria and the

    aggregation of Bax and Bak[4,21]. In turn, Bax and Bak promote the perme-

    abilization of the mitochondria leading to cell death [4]. In contrast, theintrinsic death system is triggered by intracellular stress caused by covalent

    binding, GSH depletion, or oxidative stress, which can also activate Bcl2

    family members, but the precise mechanism is not entirely clear [4]. The

    key role of sustained c-Jun-N-terminal kinase (JNK) activation has been

    demonstrated in both extrinsic and intrinsic cell death.

    Role of innate immune system

    The innate immune system has been implicated in hepatotoxicity causedby various drugs, such as APAP, dihydralazine, and halothane[6,22]. Stud-

    ies have demonstrated that the liver injury caused by hepatotoxins can be

    associated with participation of increased numbers of proinflammatory

    mediators, such as cytokines, chemokines, reactive oxygen intermediates,

    and reactive nitrogen intermediates[22]. These proinflammatory mediators

    can be directly cytotoxic (eg, hydrogen peroxide, nitric oxide, peroxynitrite);

    can degrade the extracellular matrix (eg, collagenase, elastase); and can

    promote cell adhesion and infiltration (eg, interleukin [IL]-1, TNF-a,

    chemokines) [22]. Other mediators may indirectly damage hepatocytes bymodifying hepatocyte protein and nucleic acid biosynthesis, and cytochrome

    P-450mediated metabolism[22]. In addition, the liver is selectively enriched

    in Kupffer cells. Kupffer cells are the resident phagocytic macrophages in

    the liver and account for 20% of nonparenchymal cells in the liver [2224].

    They are well positioned to remove particulate and foreign materials from

    the portal circulation because of their location in hepatic sinusoids and

    predominantly in periportal and central regions of the liver lobule [22,25].

    Their functions are diverse and include phagocytosis, endocytosis, immuno-

    modulation, and synthesis and secretion of numerous biologically activemediators, which ultimately lead to liver injury [22,24,26]. Other cells in the

    liver, such as endothelial cells and stellate cells, have also been implicated to

    participate in drug-induced liver injury through the inflammatory mediators

    [22]. The innate immune system has also been demonstrated to be protec-

    tive; the role of the innate immune system seems to be complex (see section

    on APAP hepatotoxicity). The trigger for the participation of innate im-

    mune cells and mediators is believed to be caused by hepatocyte necrosis

    but the mechanism is unclear[24]. It is also conceivable that nonlethal stress

    in hepatocytes triggers activation of the innate immune system (eg, increasedhepatocytes expression of NKNKT cell activating ligands or decreased ex-

    pression of inhibitory ligands). NK-NKT cells, which reside in the liver, may

    play a key role in modulating the innate immune response by secreting in-

    terferon (IFN)-g, osteopontin, IL-4, and so forth and directly killing cells

    by FasL expression. An overview of the pathogenesis of drug-induced liver

    disease is shown inFig. 1.

    463MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    13/226

    Acetaminophen: an example of drug hepatotoxicity

    APAP is the most extensively studied hepatotoxin and provides the bulkof knowledge on drug hepatotoxicity. The current understanding of the

    mechanism of its hepatotoxicity is described next as an example, illustrating

    some of the key aspects of the pathogenesis of drug-induced liver injury.

    APAP is primarily metabolized in the liver by glucuronidation and sulfation

    pathways into nontoxic metabolites that are then excreted in the urine.

    A small amount of APAP is metabolized by oxidation, however, using

    cytochrome P-450, mainly CYP2E1, into a highly electrophilic metabolite,

    N-acetyl-p-benzoquinone-imine (NAPQI). With a small amount of APAP,

    hepatic GSH subsequently detoxifies NAPQI into a nontoxic mercapturicacid and cysteine derivatives thereby preventing hepatocyte damage. With

    a large amount of APAP, the glucuronidation and sulfation pathways are

    saturated leading to an increased production of NAPQI by cytochrome

    P-450 pathway. At the point when sufficient NAPQI is generated to deplete

    hepatic GSH severely in both cytosol and mitochondria of hepatocytes, cell

    death ensues[27,28].

    Parent Drug

    mitochondria reactive

    metabolite hapten

    Hepatocytes

    (upstream)covalent binding

    oxidative stress

    dangerStress/Cell Death

    (mild) adaptation

    Cytokines

    ChemokinesROS, RNS

    NK/NKT

    KupfferPMNs

    Innate Immune

    System

    (downstream)Pro-inflammatory Anti-inflammatory

    Adaptive Immune

    Response

    Recovery

    Overt Liver

    Injury

    Repair

    Recovery

    Fig. 1. Pathogenesis of drug-induced liver disease. The development of drug-induced liver

    injury depends initially on upstream development of stress and cell death, which can be induced

    directly by parent drugs or reactive metabolites. This stress or cell death can then lead into

    recovery, or activate downstream participation of innate immune system. The interplay between

    proinflammatory and anti-inflammatory components of innate immune system determines the

    ensuing outcome: overt liver injury versus repair or recovery.

    464 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    14/226

    Factors that affect NAPQI production and its detoxification conse-

    quently influence the threshold for APAP hepatotoxicity. CYP2E1 is the

    main isoenzyme for APAP metabolism; hence, increased activity of thisisoenzyme can lower the threshold for APAP hepatotoxicity and vice versa.

    CYP2E1-null mice are protected against APAP, whereas induction of the

    isoenzyme by isoniazid or ethanol has been shown to increase the suscepti-

    bility to APAP [2931]. It is not surprising that chronic alcoholic patients

    have been reported to develop hepatotoxicity at dosages only modestly

    higher than the maximum recommended[3237].

    The regulation of GSH synthesis also influences APAP hepatotoxicity.

    NF-E2-related factor 2 (Nrf2) is a transcription factor that regulates GSH

    synthetic and detoxification enzymes[38]. Nrf2-null mice have been shownto be more susceptible to APAP hepatotoxicity [39,40]. Nrf2 activity is

    repressed by a cytoplasmic protein Keap1, which binds to it and promotes

    its degradation [41]. Hepatocyte-specific deletion of the Keap1 gene has

    been shown to activate Nrf2 and protect against APAP hepatotoxicity

    [42]. In addition, it seems that GSH synthesis is dynamically regulated as

    Nrf2 is activated even with low-dose APAP [38]. The provision of GSH is

    the mechanism behind the use ofN-acetylcysteine for APAP hepatotoxicity

    because it is a cysteine precursor for GSH synthesis leading to replenishment

    of GSH in hepatocytes.Although one fact concerning the toxic mechanism of APAP is indisput-

    able, namely that as a prerequisite for toxicity, excessive NAPQI production

    (exposure) and marked hepatic GSH depletion need to occur, the mechanism

    of cell death downstream of NAPQI remains incompletely understood. It has

    been believed that unopposed covalent binding of NAPQI to cysteine groups

    on cellular proteins forming APAP-protein adducts, particularly in mito-

    chondria potentiated by mitochondrial GSH depletion, leads to dysfunction

    of mitochondria, oxidative stress, and nuclear damage, and ultimately cell

    death ensues[27,4348]. Evidence has emerged, however, that NAPQI cova-lent binding or GSH depletion, probably by mitochondrial oxidative stress,

    triggers intracellular metabolic processes (eg, signaling pathways), which

    recruit intrinsic cell death machinery and promote apoptosis or necrosis.

    JNK, a stress kinase protein, has been demonstrated to play a pivotal role

    in murine APAP hepatotoxicity [49]. APAP induces a sustained activation

    of JNK and inhibition of JNK markedly protects against APAP despite the

    comparable extent of covalent binding and GSH depletion [49]. Inhibition

    of JNK has also been shown to protect in one other hepatotoxicity model,

    ischemia-reperfusion injury[50,51].The JNK activation is likely induced by oxidative stress, either through

    redox alteration of the sequestration of JNK or other upstream kinases

    by thioredoxin and GSH S-transferase or through inhibition of JNK phos-

    phatase[5254]. It is unclear if the effect of APAP on this pathway is caused

    by an effect on protein thiols as a direct consequence of GSH depletion,

    change in the GSH/GSSG ratio, or the action of H2O2 released from

    465MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    15/226

    mitochondria. JNK is presumably not activated by a TNF receptor

    dependent mechanism in response to APAP because JNK inhibitor protected

    against APAP in TNF receptor-1null mice[49].There are two JNK isoenzymes in liver: JNK1 and JNK2. It is unclear

    which isoenzyme is responsible for the cell damage. Some work has suggested

    that JNK1 may promote cell death, whereas JNK2 may promote proliferation

    and survival [5557]. Recent evidence using JNK1 and JNK2 knockout

    mice or selective silencing of either isoenzyme with antisense oligonucleotide,

    however, shows that JNK2 plays a greater role in APAP hepatotoxicity[49].

    The precise target of JNK in the pathogenesis of APAP toxicity is un-

    clear. Strong candidate targets of JNK are the Bcl-2 family members and

    mitochondria proteins. JNK has been shown to influence both proapoptoticand antiapoptotic Bcl-2 family members[5861]. In addition, Bax transloca-

    tion to mitochondria has been observed in vivo after APAP treatment,

    which was blocked by JNK inhibitor[49]. Bax translocation alone is likely

    insufficient to be lethal to hepatocytes. APAP-induced mitochondrial GSH

    depletion and covalent binding, however, may render mitochondria more

    susceptible to JNK perturbations in the Bcl-2 family. Moreover, other

    Bcl-2 family members may contribute as targets of JNK because there is

    evidence that Bid, Bim, and Bax translocate to mitochondria in response

    to APAP treatment[62]. In their work, the authors did not see tBid forma-tion or Bim translocation. They did observe, however, that JNK itself and

    phospho-JNK translocate to mitochondria, suggesting other mitochondrial

    targets (Fig. 2). Phosphorylation of Bcl-XLhas been observed in response to

    JNK, which can inactivate a protective mechanism. Because the major form

    APAP

    CYP 2E1

    NAPQI

    Mitochondria ROS Sustained JNK

    activation

    MPT

    TranslocationBax

    ? otherNECROSIS

    mitochondrial GSH

    covalent binding

    P-Bcl-XL

    Fig. 2. The role of JNK in APAP toxicity in hepatocytes. Oxidative stress (increased ROS)

    occurs in response to effects of NAPQI on mitochondria, which leads to sustained activation

    of JNK. JNK then promotes translocation of death-inducing proteins to mitochondria, includ-

    ing JNK, and leads to mitochondrial permeability transition (MPT) and necrotic cell death.

    466 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    16/226

    of cell death in vitro and in vivo in APAP toxicity is necrosis [63], it is likely

    that the JNK-initiated events do not lead to apoptosis because of the severe

    injury to mitochondria, consequent oxidative stress, and insufficient ATPlevels required to sustain propagation of the apoptosis cascade. Irrespective

    of exactly how JNK leads to cell death, the fact that JNK inhibition protects

    against APAP, even when administration of inhibitor is delayed until after

    covalent binding and GSH depletion have reached maximal levels, strongly

    supports the concept that APAP induces a type of programmed necrosis

    downstream of APAP metabolism.

    Another very interesting aspect of cellular injury from APAP is the possi-

    ble role of toxic protein mediators (eg, calpains and DNase) from hepatocytes

    undergoing necrosis leading to the injury of adjacent hepatocytes, apparentinnocent bystander effect. Limaye and colleagues[64,65]have demonstrated

    that a cell-impermeable calpain inhibitor limited progression of liver injury

    from APAP and CCl4. Overexpression of calpastatin, an endogenous calpain

    inhibitor, also protected[65]. In addition, Dnase 1 knockout were protected

    because of inhibition of progression of necrosis from the pericentral-most

    hepatocytes [66]; Dnase 1 is released from wild-type hepatocytes, which

    undergo necrosis[66].

    Role of innate immune system in acetaminophen hepatotoxicity

    The severity of APAP liver injury may be influenced by the innate

    immune system. It is well established that hepatocellular necrosis can induce

    an inflammatory response [24]. Recent studies have demonstrated that

    chromatin protein high mobility group box-1 released from necrotic cells

    can trigger inflammatory response, and neutralization of high mobility

    group box-1 leads to decreased APAP-induced inflammatory cell infiltration

    in the liver [67,68]. It has also been shown that APAP-induced hepatocyte

    damage activates the innate immune system, which then releases inflamma-tory mediators, such as cytokines, chemokines, reactive oxygen, and nitro-

    gen species, and these events contribute significantly to the severity of the

    liver injury[6973]. Furthermore, different innate immune system knockout

    or mutant mice have been shown to have altered susceptibility to APAP

    hepatotoxicity. Knockout or mutant mice that lack IFN-g, lipopolysaccha-

    ride-binding protein, Fas or FasL, or CXC chemokine receptor 2 (CXCR2)

    are less susceptible to APAP hepatotoxicity[69,7476]. Knockout mice that

    lack IL-10, IL-6, or C-C chemokine receptor 2, however, are more suscep-

    tible [7779]. In most of these knockout mice, the altered susceptibilityto APAP hepatotoxicity is not associated with significant change in GSH

    depletion or APAP covalent binding. In addition, these mice demonstrate

    that in response to nontoxic dose of APAP, there is stimulation of the

    expression of both proinflammatory and anti-inflammatory cytokines, and

    the tendency of favoring expression of one type more than the other leads

    to increased or decreased susceptibility to APAP [5].

    467MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    17/226

    NK and NKT cells are examples of cells that are important components

    of the innate immune system and play a pivotal role in the APAP hepato-

    toxicity. These cells are abundant in the liver and account for 20% to50% of isolated liver leukocytes [24,8084]. NK cells are specialized in de-

    tecting aberrant cells, such as cells that have been infected, transformed,

    or stressed. NK cells subsequently destroy the cells directly or generate cy-

    tokines and chemokines, which activate other components of the immune

    system[25,85,86]. NKT cells recognize antigens in the context of major his-

    tocompatibility complex Ilike molecule CD1d and are capable of rapidly

    producing cytokines, including IFN-g and IL-4 when activated [24,87]. In

    APAP hepatotoxicity, depletion of both NK and NKT cells provided signif-

    icant protection[69]. This protection is associated with inhibition of mRNAexpression for IFN-g, FasL, and chemokines, and reduced neutrophil accu-

    mulation in the liver [69]. These results suggest that depletion of NK and

    NKT cells might prevent APAP hepatotoxicity not only by absence of direct

    cytotoxicity but also by preventing the production of proinflammatory cyto-

    kines and chemokines. The predominant source of IFN-g in liver injury is

    NK and NKT cells [24,69]. The signals that activate NK and NKT cells

    have not yet been identified but could include cytokines produced by

    Kupffer cells in response to initial hepatocyte damage or altered expression

    of activating or inhibitory ligands in hepatocytes in response to APAP [5].The role of Kupffer cells in APAP hepatotoxicity has been suggested by

    several studies but the results are controversial. APAP treatment causes an

    increase in Kupffer cell numbers in the liver [24,8890]. Two studies found

    that mice pretreated with macrophage inhibitors (gadolinium chloride and

    dextran surfate) are more resistant to APAP hepatotoxicity [88,89]. It is

    believed that Kupffer cells participate in injury through the production of

    cytokines and reactive oxygen and nitrogen species [88,89]. A recent study

    by Ju and colleagues [90], however, found that Kupffer celldepleted mice

    had significantly increased susceptibility to APAP-induced liver injury.These mice were pretreated with liposome-entrapped clodronate (liposome-

    clodronate), which has been shown to cause nearly complete depletion of

    Kupffer cells from the liver [9092]. The discrepancy may be caused by in-

    complete depletion of Kupffer cells by gadolinium chloride[90]. Ju and col-

    leagues [90] also found that Kupffer cell depletion by liposome-clodronate

    led to significant decreases in the levels of hepatic mRNA expression of

    several hepatoregulatory cytokines and mediators, including IL-6 and

    IL-10, suggesting that Kupffer cells are a significant source for production

    of these cytokines. The protective role of IL-6 and IL-10 in APAP hepato-toxicity is discussed further. Moreover, one study found that the protection

    afforded by Kupffer cell depletion only lasted for 2 hours [93].

    Neutrophils may also contribute to APAP hepatotoxicity. It is known

    that APAP treatment results in increased numbers of neutrophils in the liver

    [69,94,95]. It is unclear, however, if the neutrophils play a role in the liver

    injury itself, contributing to the severity of organ damage, or only serve

    468 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    18/226

    to remove the debris after the liver injury has occurred. Animals depleted of

    neutrophils with antibodies against neutrophils are more resistant to APAP

    hepatotoxicity [74,94,96]. This protection occurs despite unaltered GSHdepletion or APAP covalent binding[96]. The neutrophils are likely to dam-

    age hepatocytes through cytotoxicity and generation of reactive oxygen

    intermediates and FasL expression of hepatic leukocytes [9698]. Lawson

    and colleagues [95], however, demonstrated that neutrophils play a role in

    removal of cell debris and do not directly contribute to the damage to the

    hepatocytes. They recently demonstrated that neutrophil depletion with

    anti-Gr-1 may lead to increased expression of hepatocellular metallothio-

    neine, which may protect against APAP. The data available, although

    suggesting a role of neutrophils, are not definitive. More importantly,however, irrespective of the uncertainty regarding the pathophysiologic

    role of neutrophils, the participation of inflammatory mediators in APAP

    toxicity is strongly supported by existing data from multiple laboratories.

    Several proinflammatory cytokines have been demonstrated to play a role

    in APAP hepatotoxicity. IFN-gnull mice have decreased susceptibility to

    APAP implicating its critical role in APAP hepatotoxicity[69,74]. TNF-ais

    another proinflammatory cytokine whose role is more controversial. Some

    studies found that immunoneutralization of TNF-aor mouse lacking TNF

    receptor-1 were protected against liver injury induced by APAP [99,100].TNF-ais produced by activated Kupffer cells and its production is increased

    after APAP treatment[70,96]. The role of TNF-a in APAP hepatotoxicity

    seems to be complex and controversial, however, because recent studies did

    not demonstrate the protection against APAP in TNF-aor TNF receptor-1

    gene-deficient mice[101103]or in a study using TNF-ainhibition[104]. In

    addition, at high doses, TNF-a causes liver damage but mice lacking TNF

    receptor-1 are deficient in liver growth and regeneration [105]. Opposing

    injury and repair mechanisms may account for the varied findings.

    Other cytokines, such as IL-6 and IL-10, provide protection againstAPAP hepatotoxicity. IL-6 null mice and IL-10 null mice are more suscep-

    tible to APAP[77,78]. The susceptibility of IL-6 null mice is associated with

    a deficiency in the expression of cytoprotective hepatic heat-shock proteins

    [78], whereas in IL-10 null mice, the susceptibility is associated with elevated

    expression of TNF-a, IL-1, IFN-g, and iNOS, suggesting that IL-10 is

    anti-inflammatory[77].

    Chemokines, such as keratinocyte-derived chemokine, macrophage

    inflammatory protein-1a, macrophage inflammatory protein-2, monocyte

    chemoattractant protein 1, and IFN-ainducible protein (IP-10) are alsobelieved to play a significant role in APAP hepatotoxicity because these che-

    mokines are up-regulated in animals after APAP treatment [69,74,99,103].

    Similar to data on TNF-a, it is controversial whether they are proinflamma-

    tory or anti-inflammatory [24]. For example, CXCR2 is an important

    chemokine receptor in controlling neutrophil migration and Ishida and col-

    leagues [76] found that CXCR2-null mice are protected against APAP

    469MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    19/226

    hepatotoxicity. In addition, its protection is associated with reduced neutro-

    phil infiltration in the liver [76]. Mice lacking C-C chemokine receptor 2,

    however, a different chemokine receptor, were more sensitive to APAP [79].A key question regarding the innate immune response is whether it leads

    to modulation of direct killing of hepatocytes, perhaps sensitized by the

    effects of drugs (eg, GSH depletion), or the modulation of the toxic mech-

    anisms in the hepatocyte (eg, expression of protective cellular mechanisms).

    Summary

    It seems that the biochemical changes in hepatocytes (eg, GSH depletionand covalent binding) or immune-mediated response induced by toxic

    metabolites are required to cause hepatotoxicity. Toxic metabolites may

    induce intracellular stress (oxidative or organelle specific) leading to the

    activation of built-in death programs for apoptosis or necrosis. The partic-

    ipation of innate immune system downstream of the biochemical events

    induced by toxic metabolites contributes to the severity of liver injury.

    The interplay of proinflammatory and anti-inflammatory mediators may

    determine whether a specific drug causes severe injury or no injury. Both

    environmental factors and genetic differences in cellular responses to stressand the innate immune response may account for different susceptibilities

    between individuals to drug-induced liver injury.

    References

    [1] Ostapowicz G, Fontana RJ, Schiodt FV, et al. Results of a prospective study of acute liver

    failure at 17 tertiary care centers in the United States. Ann Intern Med 2002;137:94754.

    [2] Gunawan B, Kaplowitz N. Clinical perspectives on xenobiotic-induced hepatotoxicity.

    Drug Metab Rev 2004;36:30112.[3] Kaplowitz N. Causality assessment versus guilt by association in drug hepatotoxicity.

    Hepatology 2001;33:30810.

    [4] Kaplowitz N. Biochemical and cellular mechanisms of toxic liver injury. Semin Liver Dis

    2002;22:13744.

    [5] Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005;4:48999.

    [6] Liu ZX, Kaplowitz N. Immune-mediated drug-induced liver disease. Clin Liver Dis 2002;6:

    75574.

    [7] Uetrecht J. New concepts in immunology relevant to idiosyncratic drug reactions: the

    danger hypothesis and innate immune system. Chem Res Toxicol 1999;12:38795.

    [8] Knowles SR, Uetrecht J, Shear NH. Idiosyncratic drug reactions: the reactive metabolite

    syndrome. Lancet 2000;356:158791.[9] Matzinger P. Tolerance, danger, and extended family. Annu Rev Immunol 1994;12:

    9911045.

    [10] Ozick LA, Jacob L, Comer GM, et al. Hepatotoxicity from isoniazid and rifampin in

    inner-city AIDS patients. Am J Gastroenterol 1995;90:197880.

    [11] Wong WM, Wu PC, Yuen MF, et al. Antituberculous drug-related liver dysfunction in

    chronic hepatitis B infection. Hepatology 2000;31:2016.

    470 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    20/226

    [12] Ungo JE, Jones D, Ashkin D, et al. Antituberculous drug-induced hepatotoxicity: the role

    of hepatitis C virus and the human immunodeficiency virus. Am J Respir Crit Care Med

    1998;157:18716.

    [13] Gordin F, Simon G, Wofsy C, et al. Adverse reactions to trimethoprin-sulfamethoxazole

    in patients with the acquired immunodeficiency syndrome. Ann Intern Med 1984;100:

    4959.

    [14] Levy M. Role of viral infections in the induction of adverse drug reactions. Drug Saf 1996;

    16:18.

    [15] Robin MA, Le Roy M, Descatoire V, et al. Plasma membrane cytochromes P450 as

    neoantigens and autoimmune targets in drug-induced hepatitis. J Hepatol 1997;

    26(Suppl 1):2330.

    [16] Vergani D, Mieli-Vergani G, Alberti A, et al. Antibodies to the surface of halothane-altered

    rabbit hepatocytes in patients with severe halothane-associated hepatitis. N Engl J Med

    1980;303:6671.

    [17] Neuberger J, Williams R. Immune mechanisms in tienilic acid associated hepatotoxicity.

    Gut 1989;30:5159.

    [18] Kon K, Kim JS, Jaeschke H, et al. Mitochondrial permeability transition in acetamino-

    phen-induced necrosis and apoptosis of cultured mouse hepatocytes. Hepatology 2004;

    40:11709.

    [19] Ashkenazi A, Dixit V. Death receptors: signaling and modulation. Science 1998;281:

    13058.

    [20] Faubion WA, Gores G. Death receptors in liver biology and pathology. Hepatology 1999;

    29:14.

    [21] Wei MC, Zong WX, Cheng E, et al. Pro-apoptotic Bax and Bak: a requisite gateway to

    mitochondrial dysfunction and death. Science 2001;292:72730.[22] Laskin DL, Gardner CR. Nonparenchymal cells, inflammatory macrophages, and hepato-

    toxicity. In: Kaplowitz N, DeLeve LD, editors. Drug-induced liver diseases. New York:

    Marcel Dekker; 2003. p. 183212.

    [23] Mackay IR. Hepatoimmunology: a perspective. Immunol Cell Biol 2002;80:3644.

    [24] Liu ZX, Kaplowitz N. Role of innate immunity in acetaminophen-induced hepatotoxicity.

    Expert Opin Drug Metab Toxicol 2006;2:493503.

    [25] Bouwens L, Baekkeland M, De Zanger R, et al. Quantitation, tissue distribution and

    proliferation kinetics of Kupffer cells in normal rat liver. Hepatology 1986;6:71822.

    [26] Laskin DL, Laskin JD. Role of macrophages and inflammatory mediators in chemically

    induced toxicity. Toxicology 2001;160:1118.

    [27] Nelson SD, Bruschi SA. Mechanism of acetaminophen-inducedliver disease. In: KaplowitzN, DeLeve L, editors. Drug-induced liver disease. New York: Marcel-Decker, Inc.; 2003.

    p. 287325.

    [28] Mitchell JR, Jollow DJ, Potter WZ, et al. Acetaminophen-induced hepatic necrosis. IV.

    Protective role of glutathione. J Pharmacol Exp Ther 1973;187:2117.

    [29] Lee SS, Buters JT, Pineau T, et al. Role of CYP2e1 in the hepatotoxicity of acetaminophen.

    J Biol Chem 1996;217:120637.

    [30] Burk RF, Hill KE, Hunt RW, et al. Isoniazidpotentiation of acetaminophen hepatotoxicity

    in the rat and 4-methylpyrazole inhibition of it. Res Commun Chem Pathol Pharmacol

    1990;69:1158.

    [31] Thummel KE, Slattery JT, Ro H, et al. Ethanol and production of the hepatotoxic

    metabolite of acetaminophen in healthy adults. Clin Pharmacol Ther 2000;67:5919.[32] Gerber MA, Kaufmann H, Klion F, et al. Acetaminophen associated hepatic injury: report

    of two cases showing unusual portal tract reactions. Hum Pathol 1980;11:3742.

    [33] Leist MH, Lawrence EG, Payne JA. Enhanced toxicity of acetaminophen in alcoholics:

    report of three cases. J Clin Gastroenterol 1985;7:559.

    [34] ODell JR, Zetterman RK, Burnett DA. Centrilobular hepatic fibrosis following

    acetaminophen-induced hepatic necrosis in an alcoholic. JAMA 1986;255:26367.

    471MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    21/226

    [35] Denison H, Kaczynski J, Wallerstedt S. Paracetamol medication and alcohol abuse: a dan-

    gerous combination for the liver and the kidney. Scand J Gastroenterol 1987;22:7014.

    [36] Whitcomb DC, Block GD. Association of acetaminophen hepatotoxicity with fasting and

    ethanol use. JAMA 1994;272:184550.

    [37] Schidt FV, Rohling FA, Casey DL, et al. Acetaminophen toxicity in an urban county

    hospital. N Engl J Med 1997;337:11127.

    [38] Goldring CE, Kitteringham NR, Elsby R, et al. Activation of hepatic Nrf2 in vivo by acet-

    aminophen in CD-1 mice. Hepatology 2004;39:126776.

    [39] Chan K, Han XD, Kan YW. An important function of Nrf2 in combating oxidative stress:

    detoxification of acetaminophen. Proc Natl Acad Sci USA 2001;98:46116.

    [40] Enomoto A, Itoh K, Nagayoshi E, et al. High sensitivity of Nrf2 knockout mice to

    acetaminophen hepatotoxicity associated with decreased expression of ARE-regulated

    drug metabolizing enzymes and antioxidant genes. Toxicol Sci 2001;59:16977.

    [41] Itoh K, Wakabayashi N, Katoh Y, et al. Keap 1 represses nuclear activation of antioxidant

    responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes

    Dev 1999;13:7686.

    [42] Okawa H, Motohashi H, Kobayashi A, et al. Hepatocyte-specific deletion of the keap1 gene

    activates Nrf2 and confers potent resistance against acute drug toxicity. Biochem Biophys

    Res Commun 2006;339:7988.

    [43] Jollow DJ, Mitchell JR, Potter WZ, et al. Acetaminophen-induced hepatic necrosis. II.

    Role of covalent binding in vivo. J Pharmacol Exp Ther 1973;187:195202.

    [44] Meyers LL, Beierschmitt WP, Khairalla EA, et al. Acetaminophen-induced inhibition of

    mitochondrial respiration in mice. Toxicol Appl Pharmacol 1988;93:37887.

    [45] Knight TR, Kurtz A, Bajt ML, et al. Vascular and hepatocellular peroxynitrite formation

    during acetaminophen-induced liver injury: role of mitochondrial oxidant stress. ToxicolSci 2001;62:21220.

    [46] Jaeschke H, Knight TR, Bajt ML. The role of oxidant stress and reactive nitrogen species in

    acetaminophen hepatotoxicity. Toxicol Lett 2003;144:27988.

    [47] Burcham PC, Harman AW. Acetaminophen toxicity results in site-specific mitochondrial

    damage in isolated mouse hepatocytes. J Biol Chem 1991;266:504954.

    [48] Cohen SD, Pumford NR, Khairallah EA, et al. Selective protein covalent binding and

    target organ toxicity. Toxicol Appl Pharmacol 1997;143:112.

    [49] Gunawan B, Liu ZX, Han D, et al. c-Jun-N-terminal kinase plays a major role in murine

    acetaminophen hepatotoxicity. Gastroenterology 2006;131:16578.

    [50] Uehara T, Peng XX, Bennett B, et al. c-Jun-N-terminal kinase mediates hepatic injury after

    rat liver transplantation. Transplantation 2004;78:32432.[51] Uehara T, Bennett B, Sakata S, et al. JNK mediates hepatis ischemia perfusion injury.

    J Hepatol 2005;42:8509.

    [52] Adler V, Yin Z, Fuchs SY, et al. Regulation of JNK signaling by GSTp. EMBO J 1999;18:

    132134.

    [53] Saitoh M, Nishitoh H, Fuji M, et al. Mammalian thioredoxin is a direct inhibitor of

    apoptosis signal-regulating kinase (ASK) 1. EMBO J 1998;17:2596606.

    [54] Kamata H, Honda S, Maeda S, et al. Reactive oxygen species promote TNFa-induced

    death and sustained JNK activation by inhibiting MAP kinases phosphatases. Cell 2005;

    120:64961.

    [55] Qiao L, Han SI, Fang Y, et al. Bile acid regulation of C/EBPb, CREB, and c-Jun function,

    via the extracellular signal-regulated kinase and c-jun NH2-terminal kinase pathways,modulates the apoptotic response of hepatocytes. Mol Cell Biol 2003;23:305266.

    [56] Liu J, Minemoto Y, Lin A. c-Jun N-terminal protein kinase 1 (JNK1), but not JNK2, is

    essential for tumor necrosis factor alpha-induced c-jun kinase activation and apoptosis.

    Mol Cell Biol 2004;24:1084456.

    [57] Sabapathy K, Hochedlinger K, Nam SY, et al. Distinct roles for JNK1 and JNK2 in

    regulating JNK activity and c-Jun-dependent cell proliferation. Mol Cell 2004;15:71325.

    472 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    22/226

    [58] Tsuruta F, Sunayama J, Mori Y, et al. JNK promotes bax translocation to mitochondria

    through phosphorylation of 14-3-3 proteins. EMBO J 2004;23:188999.

    [59] Kharbanda S, Saxena S, Yoshida K, et al. Translocation of SAPK/JNK to mitochondria

    and interaction with Bcl-XL in response to DNA damage. J Biol Chem 2000;275:3227.

    [60] Fan M, Goodwin M, Vu T, et al. Vinblastine-induced phosphorylation of Bcl2 and Bcl-XL

    is mediated by JNK and occurs in parallel with inactivation of the Raf-1/MEK/ERK

    cascade. J Biol Chem 2000;275:299805.

    [61] Schroeter H, Boyd CS, Ahmed R, et al. c-Jun N-terminal kinase (JNK)-mediated modula-

    tion of brain mitochondria function: new target proteins for JNK signaling in mitochon-

    drion-dependent apoptosis. Biochem J 2003;372:35969.

    [62] El-Hassan H, Anwar K, Macanas-Pirard P, et al. Involvement of mitochondria in

    acetaminophen-induced apoptosis and hepatic injury: roles of cytochrome c, Bax, Bid,

    and caspases. Toxicol Appl Pharmacol 2003;191:11829.

    [63] Gujral JS, Knight TR, Farhood A, et al. Mode of cell death after acetaminophen overdose

    in mice: apoptosis or oncotic necrosis? Toxicol Sci 2002;67:3229.

    [64] Limaye PB, Apte UM, Shankar K, et al. Calpain released from dying hepatocytes mediates

    progression of acute liver injury induced by model hepatotoxicants. Toxicol Appl Pharma-

    col 2003;191:21126.

    [65] Limaye PB, Bhave VS, Palkar PS, et al. Upregulation of calpastatin in regenerating

    and developing rat liver: role in resistance against hepatotoxicity. Hepatology 2006;44:

    37988.

    [66] Napirei M, Basnakian AG, Apostolov EO, et al. Deoxyribonuclease 1 aggravates acet-

    aminophen-induced liver necrosis in male CD-1 mice. Hepatology 2006;43:297305.

    [67] Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells

    triggers inflammation. Nature 2002;418:1915.[68] Tsung A, Sahai R, Tanaka H, et al. The nuclear factor HMGB1 mediates hepatic injury

    after murine liver ischemia-reperfusion. J Exp Med 2005;201:113543.

    [69] Liu ZX, Govindarajan S, Kaplowitz N. Innate immune system plays a critical role in deter-

    mining the progression and severity of acetaminophen hepatotoxicity. Gastroenterology

    2004;127:176074.

    [70] Jaeschke H. Role of inflammation in the mechanism of acetaminophen hepatotoxicity.

    Expert Opin Drug Metab Toxicol 2005;1:38997.

    [71] Blazka ME, Wilmer JL, Holladay SD, et al. Role of proinflammatory cytokines in acet-

    aminophen hepatotoxicity. Toxicol Appl Pharmacol 1995;133:4352.

    [72] Ferret PJ, Hammoud R, Tulliez M, et al. Detoxification of reactive oxygen species by

    a nonpeptidyl mimic of superoxide dismutase cures acetaminophen-induced acute liverfailure in the mouse. Hepatology 2001;33:117380.

    [73] Dambach DM, Watson LM, Gray KR, et al. Role of CCR2 in macrophage migration into

    the liver during acetaminophen-induced hepatotoxicity in the mouse. Hepatology 2002;35:

    1093103.

    [74] Ishida Y, Kondo T, Ohshima T, et al. A pivotal involvement if IFN-gin the pathogenesis

    of acetaminophen-induced acute liver injury. FASEB J 2002;16:122736.

    [75] Su G, Gong KQ, Fan MH, et al. Lipopolysaccharide-binding protein modulates acetamin-

    ophen-induced liver injury in mice. Hepatology 2005;41:18795.

    [76] Ishida Y, Kondo T, Kimura A, et al. Opposite roles of neutrophils and macrophages in

    the pathogenesis of acetaminophen-induced acute liver injury. Eur J Immunol 2006;36:

    102838.[77] Bourdi M, Masubuchi Y, Reilly TP, et al. Protection against acetaminophen induced liver

    injury and lethality by interleukin 10: role of inducible nitric oxide synthase. Hepatology

    2002;35:28998.

    [78] Masubuchi Y, Bourdi M, Reilly TP, et al. Role of interleukin-6 in hepatic heat shock

    protein expression and protection against acetaminophen-induced liver disease. Biochem

    Biophys Res Commun 2003;304:20712.

    473MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    23/226

    [79] Hogaboam CM, Bone-Larson CL, Steinhauser ML, et al. Exaggerated hepatic injury due

    to acetaminophen challenge in mice lacking C-C chemokine receptor 2. Am J Pathol 2000;

    156:124552.

    [80] Wiltrout RH. Regulation and antimetastatic functions of liver-associated natural killer

    cells. Immunol Rev 2000;174:6376.

    [81] Hashimoto W, Takeda K, Anzai R, et al. Cytotoxic NK1.1 Ag ab T cells with intermedi-

    ate TCR induced in the liver of mice by IL-12. J Immunol 1995;154:433340.

    [82] Doherty DG, OFarrelly C. Innate and adaptive lymphoid cells in the human liver.

    Immunol Rev 2000;174:520.

    [83] Liu ZX, Govindarajan S, Okamoto S, et al. NK cells cause liver injury and facilitate the in-

    duction of T cell mediated immunity to a viral liver infection. J Immunol 2000;164:64806.

    [84] Crisp IN. Hepatic T cells and liver tolerance. Nat Rev Immunol 2003;3:5162.

    [85] Biron CA, Nguyen KB, Pien GC, et al. Natural killer cells in antiviral defense: function and

    regulation by innate cytokines. Annu Rev Immunol 1999;17:189220.

    [86] Cerwenka A, Lanier LL. Natural killer cells, viruses and cancer. Nat Rev Immunol 2001;1:

    419.

    [87] Taniguchi M, Harada M, Kojo S, et al. The regulatory role of Va14 NKT cells in innate and

    acquired immune response. Annu Rev Immunol 2003;21:483513.

    [88] Laskin DL, Gardner CR, Price VF, et al. Modulation of macrophage functioning abrogates

    the acute hepatotoxicity of acetaminophen. Hepatology 1995;21:104550.

    [89] Michael SL, Pumford NR, Mayeux PR, et al. Pretreatment of mice with macrophage

    inactivators decreases acetaminophen hepatotoxicity and the formation of reactive oxygen

    and nitrogen species. Hepatology 1999;30:18695.

    [90] Ju C, Reilly TP, Bourdi M, et al. Protective role of Kupffer cells in acetaminophen-induced

    hepatic injury in mice. Chem Res Toxicol 2002;15:150413.[91] Van Rooijen N, Sanders A. Liposome mediated depletion of macrophages: mechanism of

    action, preparation of liposomes and applications. J Immunol Methods 1994;174:8393.

    [92] Yamamoto T, Naito M, Moriyama H, et al. Repopulation of murine Kupffer cells after

    intravenous administration of liposome-encapsulated dichloromethylene diphosphonate.

    Am J Pathol 1996;149:127186.

    [93] Goldin RD, Ratnayak ID, Breach CS. Role of macrophages in acetaminophen

    (paracetamol)-induced hepatotoxicity. J Pathol 1996;179:4325.

    [94] Smith GS, Nadig DE, Kokoska ER, et al. Role of neutrophils in hepatotoxicity induced

    by oral acetaminophen administration in rats. J Surg Res 1998;80:2528.

    [95] Lawson JA, Farhood A, Hopper RD, et al. The hepatic inflammatory response after acet-

    aminophen overdose: role of neutrophils. Toxicol Sci 2000;54:50916.[96] Liu ZX, Han D, Gunawan B, et al. Neutrophil depletion protects against murine acetamin-

    ophen hepatotoxicity. Hepatology 2006;43:122030.

    [97] Jaeschke H, Smith CW. Mechanisms of neutrophil-induced parenchymal cell injury.

    J Leukoc Biol 1997;61:64753.

    [98] Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol

    2006;6:17382.

    [99] Ishida Y, Kondo T, Tsuneyama K, et al. The pathogenic roles of tumor necrosis factor

    receptor p55 in acetaminophen-induced liver injury in mice. J Leukoc Biol 2004;75:

    5967.

    [100] Blazka ME, Elwell MR, Holladay SD, et al. Histopathology of acetaminophen-induced

    liver changes: role of interleukin 1aand tumor necrosis factor-a. Toxicol Pathol 1996;24:1819.

    [101] Boess F, Bopst M, Althaus R, et al. Acetaminophen hepatotoxicity in tumor necrosis

    factor/lymphotoxin-agene knockout mice. Hepatology 1998;27:10219.

    [102] Chiu H, Gardner CR, Dambach DM, et al. Role of tumor necrosis factor receptor 1 (p55) in

    hepatocyte proliferation during acetaminophen-induced toxicity in mice. Toxicol Appl

    Pharmacol 2003;193:21827.

    474 GUNAWAN & KAPLOWITZ

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    24/226

    [103] Gardner CR, Laskin JD, Dambach DM, et al. Exaggerated hepatotoxicity of acetamino-

    phen in mice lacking tumor necrosis factor receptor-1: potential role of inflammatory

    mediators. Toxicol Appl Pharmacol 2003;192:11930.

    [104] Simpkon KJ, Lukacs NW, McGregor AH, et al. Inhibition of tumor necrosis factor alpha

    does not prevent experimental paracetamol-induced hepatic necrosis. J Pathol 2000;190:

    48994.

    [105] Yamada Y, Kirillova I, Peschon JJ, et al. Initiation of liver growth by tumor necrosis factor:

    deficient liver regeneration in mice lacking type I tumor necrosis factor receptor. Proc Natl

    Acad Sci USA 1997;94:14416.

    475MECHANISMS OF DRUG-INDUCED LIVER DISEASE

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    25/226

    Causality Assessment of Drug-Induced

    Hepatotoxicity: Promises and Pitfalls

    Max A. Shapiro, MD, James H. Lewis, MD*

    Hepatology Section, Division of Gastroenterology, Georgetown University Hospital,

    Georgetown University Medical Center, Washington, DC 20007, USA

    Art is science made clear.

    dJean Cocteau, 1926

    Drug-induced liver injury (DILI) remains an important clinical concern,

    accounting for 4% to 10% of all adverse drug reactions [1,2]. It has been

    diagnosed in about 1% of general medical inpatients [3], for 10% to 33%

    of patients presenting with acute hepatitis [4,5], and for 5% to 10% of con-

    sultations performed in hepatology practices[6,7]. DILI is the leading causeof acute liver failure in the United States[8]with acetaminophen also top-

    ping the list of drugs causing acute liver failure leading to emergency liver

    transplantation [9]. DILI is among the most frequent reasons for drug-re-

    lated regulatory actions [10], including nonapprovals, restriction of use,

    and removal of drugs from the market [11].

    Popper and colleagues [12] described DILI as a penalty for progress

    nearly half a century ago, but the ability to ascribe hepatic injury confidently

    to a specific drug remains a challenging and often difficult pursuit. The lack

    of a specific biomarker or characteristic histologic feature to identify a drugas causal further hampers this effort and has fostered reliance on clinical as-

    sessment techniques that are based largely on medical judgment and expert

    opinion rather than on a truly objective means of assessing causality accu-

    rately. As a result, depending on the knowledge and experience of the clini-

    cian performing the diagnostic evaluation, the final assessment may lack

    precision and seems at times to reduce the process to one of making a diag-

    nosis of exclusion based on circumstantial evidence. The consequences of

    erroneously attributing the cause of hepatic injury to a drug can be dire

    * Corresponding author. Georgetown University Hospital, Room M2408, 3800 Reservoir

    Road, NW, Washington, DC 20007.

    E-mail address: [email protected](J.H. Lewis).

    1089-3261/07/$ - see front matter 2007 Elsevier Inc. All rights reserved.

    doi:10.1016/j.cld.2007.06.003 liver.theclinics.com

    Clin Liver Dis 11 (2007) 477505

    mailto:[email protected]:[email protected]
  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    26/226

    for patients and health care providers and for the pharmaceutical industry

    and regulatory bodies as well.

    Although a number of thoughtful efforts have been undertaken duringthe past 2 decades to create causality-assessment instruments that attempt

    to objectify clinical findings and impressions using numeric scales [1316],

    none has proven infallible in creating a true reference standard to diagnose

    DILI in all of its forms and disguises[17,18]. This article explores the ongo-

    ing challenges inherent in what is currently a clinical process of elimination

    made in the attempt of assigning causality in DILI. In particular, it points

    out the shortcomings and pitfalls that often limit the applicability of the cau-

    sality-assessment methodologies (CAMs) currently in use around the globe.

    It is anticipated that ongoing advances in pharmacogenomics, toxicology,and understanding of the mechanisms of hepatocyte injury eventually will

    improve the ability to diagnose DILI in the not-too-distant future. For

    the time being, clinicians must make the best use of the available causality

    methods, imperfect though they may be.

    Historical basis of assessing causality

    Causality assessment began as more of an art than a science. A number of

    early studies explored the process by which a variety of nonorgan-specificadverse drug reactions were assessed [1924]. Arimone and colleagues [25]

    recently reviewed these methods and divided the nearly 2 dozen approaches

    into three main categories: expert judgment, probabilistic methodologies,

    and algorithms. In general, none was considered highly satisfactory. For ex-

    ample, they cited evidence that expert judgment was limited by subjectivity

    and a lack of standardization leading to poor reproducibility among the ex-

    perts. Probabilistic methods, mostly derived from Bayes theorem, require

    precise information to model probability and therefore are not considered

    appropriate for routine clinical use. The algorithmic approach to causality,although relatively simple to apply in theory, was thought to be less useful

    because of potential bias in the often arbitrary weighting of each criterion

    [25]. Many of the methods relying on an algorithm were based on guidelines

    developed by the World Health Organization (WHO) and others [25,26],

    which considered time to onset, the response to dechallenge and rechallenge,

    a search for nondrug-related causes, risk factors for the reaction, and pre-

    vious reports of the reaction to determine if a particular reaction is caused

    by a specific drug. Arimone and colleagues[25]recently proposed a weight-

    ing process based on statistical regression of modified WHO criteria thatthey applied to randomly selected adverse drug reactions affecting various

    organ systems from a French pharmacovigilance database. They were able

    to demonstrate good agreement between their weighted probability scores

    and expert opinion (which was based on a visual analogue scale) but ac-

    knowledged that further refinements in their weighting tool would be needed

    before such a method could be more widely applied [25].

    478 SHAPIRO & LEWIS

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    27/226

    As several investigators noted [21,2325], reliance on clinical judgment

    alone often is inadequate for determining causality, based on the low levels

    of agreement between evaluators (expert or not) and the poor correlationwith objective methods, as summarized in Table 1. As can be seen, signifi-

    cant differences often exist among evaluators: the rate of complete agree-

    ment between any two experts ranged from 41% to 57% [13,20]. With the

    use of a CAM, however, Naranjo and colleagues[20]showed that interrater

    agreement rose from 83% to 92%. Similarly, the use of a clinical diagnostic

    scale improved interrater agreement to 86% in a more recent study [16].

    Getting more than two evaluators to agree on the level of causality cer-

    tainty, however, has proven to be quite challenging: three evaluators were

    in agreement in 40% to 60% of cases [16,27], but five evaluators were inagreement in only 17% of cases [13]. Moreover, the majority of the agree-

    ment occurred for causality that was at the far ends of the assessment scale

    (ie, either excluded or definitely related). Classifying a reaction as pos-

    sibly related or probably related proved more difficult. When clinical

    judgment was compared with an objective scale, the correlation was even

    less accurate. In one study, there was complete agreement with physicians

    opinion in only 6% of adverse drug reactions [21]. Allowing for one level

    of discrepancy improved agreement somewhat (49%) but at the risk of di-

    luting the precision of the assessment[21]. These authors attributed the rel-atively poor level of agreement to the tendency of physicians in their study

    to form fairly strong opinions based on only a few clinical criteria [21].

    Agreement between clinical judgment and a CAM seems to improve over

    time as evaluators become more familiar with scoring criteria. For example,

    in a recent study of the Roussel Uclaf Causality Assessment Method (RU-

    CAM) in a clinical trial setting, the kappa statistic measuring interrater

    agreement improved from the first 50 cases assessed to the final 50 cases

    [28,29].

    Causality assessment method and drug-induced liver injury

    Zimmerman and other early pioneers in the field of hepatotoxicity

    employed many of the same components found in the current WHO

    guideline [26] and other authors criteria for causality assessment of

    drug reactions in general [19,20,25] and applied them to a common-sense

    clinical approach to establishing the cause of suspected DILI [30]. Exam-

    ining the circumstances of the liver injury, the host factors, the clinico-pathologic features of the reaction, its course and outcome, excluding

    other causes, and comparing the reaction with the known spectrum of

    injury associated with that agent formed the basis of DILI causality as-

    sessment based on expert opinion that has been increasingly refined over

    the years within the limitations of diagnostic tools and clinical acumen

    (Box 1).

    479CAUSALITY ASSESSMENT IN DILI

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    28/226

    Table 1

    Clinical judgment and expert opinion in causality assessment

    Study Comparison Type of reaction

    Method of assess

    and levels of caus

    Blanc et al[27] Experts assessments All-cause ADRs Questionnaire 5 l

    Naranjo et al [20] Clinicians assessments

    CAM

    All-cause ADRs Probability scale

    Arimone et al[13] Experts assessments All-cause ADRs VAS 7 levels

    Miremont et al [21] Physician opinion versus

    CAM

    All-cause ADRs French scale[22]

    Danan et al[14] Experts assessments Hepatotoxicity RUCAM[14,15]

    Maria and Victorino

    [16]

    Experts assessments Hepatotoxicity CDS[16]5 levels

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    29/226

    Aithal et al [40] Physician reports versus

    CAM

    Hepatotoxicity Modified CIOMS

    3 levels

    Aithal et al [42] Physician suspicion

    versus CAM

    Hepatotoxicity Modified CIOMS

    and CDS[16]

    Meier et al[3] Physician diagnosis

    versus CAM

    Inpatient hepatotoxicity WHO criteria[26

    2 levels

    Lewis et al [28] Physician opinion versus

    CAM

    Clinical trial

    of hepatotoxicity

    of a single agent

    RUCAM[14,15]

    Abbreviations:ADR, nonorgan-specific adverse drug reaction; CAM, causality method assessment; CDS, Clinica

    International Organizations of Medical Sciences; RUCAM, Roussel Uclaf Causality Assessment Method; VAS, visa See individual studies for definitions.

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    30/226

    In an effort to improve on the early efforts at ADR causality, a number of

    objective methodologic instruments have been introduced during the past 2

    decades to assess DILI specifically. The two most widely used of thesemethods are the RUCAM[14,15]and the Clinical Diagnostic Scale (CDS)

    [16]. A third method is being studied is the DILI Network approach to cau-

    sation, which represents the latest attempt to objectify expert opinion [10].

    Roussel Uclaf Causality Assessment Method

    The RUCAM was developed at the request of the Council for Interna-

    tional Organizations of Medical Sciences (CIOMS) by an internationally

    recognized panel of experts brought together by Danan and Benichou of

    the Drug Safety Department of the French pharmaceutical maker Roussel

    Box 1. Elements used in the clinical assessment of acute

    drug-induced liver injury

    1. High index of suspicion that a drug is responsible

    2. Time of onset (latency)

    3. Clinical features4. Biochemical injury pattern (hepatocellular, cholestatic,

    mixed); aspartate aminotransferase/alanine

    aminotransferase ratio and maximum absolute

    liver-associated enzyme values

    5. Mechanism of injury (eg, intrinsic, hypersensitivity,

    metabolic idiosyncrasy)

    6. Extrahepatic symptoms

    7. Clinical/biochemical course of the reaction (response to

    dechallenge, rechallenge; and adaptation [drug tolerance])8. Drug serum levels, drugprotein adducts, lymphocyte

    transformation tests, and others

    9. Histologic findings

    10. Genetic markers, polymorphisms

    11. Exclusion of other causes of acute drug-induced liver injury

    Serology for viral hepatitis A, B, C, D, E, cytomegalovirus,

    Epstein-Barr virus, other viral causes

    Metabolic, autoimmune causes

    Alcoholic liver disease Shock, sepsis

    Gallstones

    Muscle injury

    Postoperative jaundice

    Pregnancy-related liver injury

    482 SHAPIRO & LEWIS

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    31/226

    Uclaf in 1989 and 1990[14,15]. A major goal was to adapt existing methods

    for assessing nonorgan-specific drug reaction to well-defined hepatic reac-

    tions[31,32]. From these meetings, a consensus opinion emerged that culmi-nated in the construction of a causality assessment method based on seven

    major criteria that were in common use at the time: (1) time to onset, (2)

    course of the reaction, (3) risk factors for the reaction, (4) assessing the

    role of concomitant therapies, (5) screening for nondrug-related causes,

    (6) weighing the information known about the DILI in question, and (7) con-

    firmation of the reaction by positive rechallenge or in vitro assays (Table 2).

    Each criterion was assigned a score ranging from 3 to 3 corresponding

    to the probability of the involvement of the drug being evaluated. Maximum

    total scores ranged from 7 to 14, defining the following causal relation-ships: a causal relationship was excluded with a score of zero, unlikely with

    a score of 1 or 2, possible with a score of 3 to 5, probable with a score of 6 to

    8, and highly probable with a score higher than 8. The panel distinguished

    between hepatocellular reactions and those that were cholestatic or mixed

    based on the ratio of aminotransferase levels to alkaline phosphatase

    (AP). Reproducibility of the scoring system was assessed by an independent

    team that reviewed a series of 50 case reports of acute DILI received by the

    Bordeaux regional pharmacovigilance center. All four of these independent

    experts were in agreement in only 37% of cases, but three experts agreed in74% of cases, and two experts agreed in 99% of cases [14]. The scoring sys-

    tem then was validated by comparing 49 case reports of drug-induced acute

    liver injury with positive rechallenge from the literature and 28 well-matched

    controls. Sensitivity of 86% and specificity of 89%, a positive predictive

    value of 93%, and a negative predictive value of 78% were achieved, with

    no overlap between the cases and the controls [15].

    Since the RUCAM first appeared, it has been used most widely outside

    the United States to validate published case reports and case series

    [33,34]. Lee and colleagues[29]described the successful application of RU-CAM scoring to detect the hepatotoxicity of a new agent undergoing clinical

    trial testing when no prior information about the potential reaction existed.

    The Maria and Victorino clinical diagnostic scale

    The complexity of the RUCAM prompted Maria and Victorino[16]from

    Portugal to propose and validate a somewhat simpler scoring system to assess

    DILI. These authors constructed a CDS, based on a modification of the RU-CAM criteria, that also used the time to onset and time course of the reaction,

    the exclusion of alternative causes, the response to re-exposure (by intentional

    or accidental rechallenge), and previous reports in the literature implicating

    the drug. They added a fifth criterion based on the presence of extrahepatic

    manifestations of DILI, specifically fever, rash, eosinophilia, arthralgias,

    and cytopenia. The CDS then was validated against the opinion of three

    483CAUSALITY ASSESSMENT IN DILI

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    32/226

    Table 2

    Comparison of criteria used in RUCAM versus CDS for acute drug-induced liver injury

    Assessment component

    and injury pattern RUCAM[14,15]

    Points

    awarded CD

    Time to onset from start of drugHepatocellular type 590 days (IT), 115 days (ST) 2 45

    !5 or O90 days (IT), 15 (ST) 1 !4

    Cholestatic or mixed type 590 days (IT), 190 days (ST) 2 !4

    !5 or O90 days (IT),

    O15 days (ST)

    1

    Time to onset from cessation of drug

    Hepatocellular type !15 days 1 !7

    Cholestatic or mixed type !30 days 1 O

    Time to enzyme normalization

    after cessation of drug

    Hepatocellular type Decrease O 50% within 8 daysDecrease O 50% within 30 days

    Decrease O 50% after 30 days

    Persistence

    32

    0

    2

    VaU

    !6

    O6

    Cholestatic or mixed type Decrease O 50% within 180 days

    Decrease ! 50% within 180 days

    Persistence

    2

    1

    0

    Va

    U

    !

    O

    Risk factors

    Alcohol use 1 N/

    Pregnancy (in cholestatic/mixed only) 1

    Age O55 1

    Concomitant drug use

    None or incompatible timing 0 N/

    Compatible or suggestive timing 1

    Known hepatotoxin with suggestive timing 2

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    33/226

    Evidence of role (positive

    rechallenge or validated test)

    3

    Alternative causes (viral hepatitis,

    biliary obstruction, alcoholic

    or other liver disease, recent hypotension,

    other causes)All causes reasonably ruled out

    Most causes ruled out

    !3 causes not ruled out

    R3 causes not ruled out

    Nondrug cause highly probable

    2

    1

    0

    2

    3

    Co

    Pa

    Po

    d

    Pro

    d

    Extrahepatic manifestations

    Fever, rash, arthralgia,

    eosinophilia O6%, cytopenia

    N/A R

    23

    1

    NoPreviously reported

    hepatotoxicity of drug

    In product information

    Published reports

    Reaction unknown

    2

    1

    0

    Pu

    No

    No

    Response to re-exposure

    Intentional or accidental Positive

    Compatible

    Negative

    3

    1

    2

    Po

    Ne

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    34/226

    Table 2 (continued)

    Assessment component

    and injury pattern RUCAM[14,15]

    Points

    awarded CD

    Other

    Toxic plasma concentration of the drug

    or validated lab test

    Positive

    Negative

    3

    3

    N/

    Total point range 7 to 14

    Probability of drug causality Highly probable/definite O8

    Probable 68

    Possible 35

    Unlikely 12

    Excluded !1

    Abbreviations:IT, initial treatment; N/A, not assessed; ST, subsequent treatment; ULN, upper limit of normal.

  • 7/27/2019 35575737 Drug Induced Liver Disease 2007

    35/226

    experts in drug-induced hepatotoxicity, a decision-tree analysis, and the re-

    sults of immunologic testing of 50 cases of suspected DILI. Scores ranged

    from6 to20 and were broken down into subgroups based on the probabil-ity of a true causal relationship: a relationship was excluded by a score less

    than 6, was unlikely with a score of 6 to 9, possible with a score of 10 to 13,

    probable with a score of 14 to 17, and definite with a score above 17.

    Although the Maria and Victorino CDS is similar to the RUCAM, it

    varies in several significant ways, with differences in the time to onset of

    the reaction and the course after drug has been stopped. Also, it reduces

    the probability of a suspected drug