Wednesday 19 March 2014

Drug Induced Liver Disease (Hepatotoxicity)-Chapter 8-MECHANISM OF DRUG-INDUCED LIVER INJURY



               Drug-induced liver injury (DILI) is a major health problem that challenges not only health care professionals but also the pharmaceutical industry and drug regulatory agencies. According to the United States Acute Liver Failure Study Group, 1 DILI accounts for more than 50% of acute liver failure, including hepatotoxicity caused by overdose of acetaminophen (APAP, 39%) and idiosyncratic liver injury triggered by other drugs (13%). Because of the significant patient morbidity and mortality associated with DILI, 2 the U.S. Food and Drug Administration (FDA) has removed several drugs from the market, including bromfenac, ebrotidine, and troglitazone. Other hepatotoxic drugs, such as risperi-done, trovafloxacin, and nefazodone, have been assigned  “ black box ” warnings. DILI is the most common cause for the withdrawal of drugs from the pharmaceutical market. 

              Although the exact mechanism of DILI remains largely unknown, it appears to involve 2 pathways — direct hepato-toxicity and adverse immune reactions. In most instances, DILI is initiated by the bioactivation of drugs to chemically reactive metabolites, which have the ability to interact with cellular macromolecules such as proteins, lipids, and nucleic acids, leading to protein dysfunction, lipid peroxidation, DNA damage, and oxidative stress. Additionally, these reactive metabolites may induce disruption of ionic gradients and intracellular calcium stores, resulting in mitochondrial dysfunction and loss of energy production. This impairment of cellular function can culminate in cell death and possible liver failure. 

               Hepatic cellular dysfunction and death also have the ability to initiate immunological reactions, including both innate and adaptive immune responses. Hepatocyte stress and/or damage could result in the release of signals that stimulate activation of other cells, particularly those of the innate immune system, including Kupffer cells (KC), natural killer (NK) cells, and NKT cells. These cells contribute to the progression of liver injury by producing proinflammatory mediators and secreting chemokines to further recruit inflammatory cells to the liver. It has been demonstrated that various inflammatory cytokines, such as tumor necrosis factor (TNF)- , interferon (IFN)- , and interleukin (IL)-1, produced during DILI are involved in pro moting tissue damage.



However, innate immune cells are also the main source of IL-10, IL-6, and certain postglandins, all of which have been shown to play a hepatoprotective role. Thus, it is the delicate balance of inflammatory and hepatoprotective mediators produced after activation of the innate immune system that determines an individual ’ s susceptibility and adaptation to DILI.
                 In addition to the innate immune responses, clinical features of certain DILI cases strongly suggest that the adaptive immune system is activated and involved in the pathogenesis of liver injury. With regard to the involvement of the adaptive immune system in DILI, our current understanding is based on the hapten hypothesis and the p-i ( p harmacological interaction of drugs with i mmune receptors) concept. Evidence to support these hypotheses is gained by the detection of drug-specific antibodies and T cells in some patients with DILI. 
 





Figure.8.1:-Pathogenesis of drug-induced liver diseases. Upstream events in the hepatocytes affect viability of individual cells but sensitize to downstream processes leading to clinically overt organ damage. The latter involves a balance of effects of cytokines, chemokines, and inflammatory mediators, mainly produced by nonparenchymal cells and the effects on repair processes a such as regeneration.




Figure.8.2:- Role of cytokine balance in determining susceptibility to toxins. Drugs or metabolites may directly injure hepatocytes to a minor extent, but may markedly sensitize to the lethal effects of TNFa and IFNg. The latter are modulated by cytokines that promote or inhibit their production or actions. Abbreviations: TNFa, tumor necrosis factor a; IFNg, interferon g.



DILI

           DILI can affect both parenchymal and nonparenchymal cells of the liver, leading to a wide variety of pathological con ditions, including acute and chronic hepatocellular hepatitis, fibrosis/cirrhosis, cholestasis, steatosis, as well as sinusoidal and hepatic artery/vein damage. The predominant forms of DILI include acute hepatitis, cholestasis, and a mixed pattern. Acute hepatitis is defined as a marked increase in aminotransferases coinciding with hepatocellular necrosis. Cholestasis is characterized by jaundice with a concurrent elevation in alkaline phosphatase, conjugated bilirubin, and -glutamyl transpeptidase. Mixed-pattern DILI includes clinical manifestations of both hepatocellular and cholestatic injury. 


            The clinical diagnosis and prediction of DILI remain a major challenge due to various confounding factors. These factors include preexisting liver disease, multiple drug usage by patients, and most important, lack of reliable screening methods and diagnostic standards. As a general rule, alanine transaminase (ALT) levels greater than 3 times the upper limits of normal (ULN) have been identified as a marker for liver injury. Hyman Zimmerman noted that elevated ALT accompanied by jaundice was associated with a mortality between 5% and 50%.  This observation has since been referred to as “ Hy ’ s rule ” and is currently employed by the FDA in the evaluation of hepatotoxicity for newly developed drugs. However, it must be noted that 3 × ULN of ALT levels are not necessarily predictive of overt severe liver toxicity or acute liver failure.

           

MECHANISMS OF DILI



Drug-Induced Direct Hepatotoxicity



           Direct hepatotoxicity is often caused by the direct action of a drug, or more often a reactive metabolite of a drug, against hepatocytes. One classically studied drug used to examine the mechanisms of hepatotoxicity is APAP. APAP is a popular over-the-counter analgesic that is safe at therapeutic doses but at overdose can produce centrilobular hepatic necrosis, which may lead to acute liver failure. APAP is metabolized to a minor electrophilic metabolite, N-acetyl-p-benzoquinoneimine (NAPQI), which during APAP overdose depletes glutathione and initiates covalent binding to cellular proteins. These events lead to the disruption of calcium homeostasis, mitochondrial dysfunction, and oxi-dative stress and may eventually culminate in cellular damage and death. Fortunately, drug candidates that induce significant direct hepatotoxicity at therapeutic doses are more likely to be detected during preclinical toxicity screening and thus rarely reach the pharmaceutical market.

          In most instances of DILI, it appears that hepatocyte damage triggers the activation of other cells, which can initiate an inflammatory reaction and/or an adaptive immune re -sponse. These secondary events may overwhelm the capacity of the liver for adaptive repair and regeneration, thereby contributing to the pathogenesis of liver injury.



Drug-Induced Immune-Mediated Liver Injury



Innate and Adaptive Immune System of the Liver

           The innate immune system provides a first line of defense against microbial infection, but it is not sufficient in eliminating infectious organisms. The lymphocytes (T and B cells) of the adaptive immune system provide a more versatile means of defense and possess “ memory, ” which is the ability to respond more vigorously to repeated exposure to the same microbe. Moreover, cells of the innate immune system play an integral role in the initiation of adaptive immunity by presenting antigens and are crucial in determining the subsequent T-cell- or antibody-mediated immune response. Because of the liver ’ s continuous exposure to pathogens, toxins, tumor cells, and harmless dietary antigens, it possesses a range of local immune mechanisms to cope with these challenges. The liver contains large numbers of both innate and adaptive immune cells, including the largest populations of tissue macrophages (KC), NK cells, and NKT cells. The liver also possesses a unique combination of intrahepatic lymphocytes, which include not only the conventional CD4 + and CD8 + T cells but also high percentages of T cells and CD4 − CD8 − T cells. Collectively, the innate and adaptive immune cells contribute to the unique immune responses of the liver, including removal of pathogenic microorganisms, clearance of particles and soluble molecules from circulation, deletion of activated T cells, and induction of tolerance to food antigens derived from the gastrointestinal tract. 


           KC play an essential role in the phagocytosis and removal of pathogens entering the liver via portal-venous blood. Upon activation, KC produce various cytokines and other mediators, including prostanoids, nitric oxide, and reactive oxygen intermediates. These KC products play prominent roles in promoting and regulating hepatic inflammation, as well as modulating the phenotype of other cells in the liver, such as NK and NKT cells. Furthermore, KC represent a major population of antigen-presenting cells (APCs) within the liver. It has been demonstrated that KC can activate T cells in vitro, but they do so less efficiently than peritoneal-exudate macrophages. Studies of organ transplantation using animal models have further shown that inhibition of KC abrogated the prolonged survival of allografts induced by portal vein infusion of allogeneic donor cells. Collectively, this evidence suggests that KC play an important role in the delicate balance between the induction of immunity and tolerance within the liver. 


          Unique to the liver are the remarkably high frequencies of NK and NKT cells, which account for ~50% of intrahepatic leukocytes. These cells act as a first line of defense against certain pathogens and invading tumor cells prior to the adaptive immune response of B and T lymphocytes. One characterized function of hepatic NK and NKT cells is their cytotoxic capacity against other cells. It has been demonstrated that freshly isolated liver NK cells spontaneously induce the cyto-toxicity of various cell lines, whereas NKT cells are cytotoxic in the presence of IL-2. This cytotoxicity is further enhanced by IL-12 and IL-18, which are produced by activated KC. Another function ascribed to NK and NKT cells is their ability to produce high levels of T helper (Th) 1 and Th2 cyto-kines upon stimulation. NK cells have been shown to represent a major source of IFN- in many types of liver disease. NKT cells produce either IFN- or IL-4, or in some cases both cytokines, depending on the differentiation state of the cells and the stimuli. It has also been demonstrated that IL-4 produced by NKT cells may be associated with the initiation and regulation of Th2 responses.  


           The liver ’ s adaptive immune responses are unique in that the liver is known to favor induction of immunological tolerance rather than immunity. This is supported by numerous studies demonstrating that (1) dietary antigens derived from the gastrointestinal tract are tolerized in the liver; (2) allogeneic liver organ transplants are accepted across major histocompatibility complex (MHC) barriers  (3) preexposure to donor cells through the portal vein of recipient animals increased their acceptance of solid tissue allografts and (4) preexposure of soluble antigens via the portal vein leads to systemic immune tolerance. Several mechanisms have been suggested to account for this tolerance, including apoptosis of activated T cells, immune deviation, and active suppression. 


          The liver has been dubbed the “ elephant ’ s graveyard ” for activated T cells. These cells accumulate in the liver before undergoing apoptosis. Studies using T-cell receptor transgenic models have demonstrated that following antigen exposure, the activated T cells undergo apoptosis after a transient accumulation within the liver. Immune deviation may account for liver-induced tolerance, as it has been shown that Th2 cytokine production is preferentially maintained when adoptively transferred Th1 and Th2 cells are recovered from the liver. It has also been reported that liver sinusoidal endothelial cells (LSEC) are capable of selectively suppressing IFN- -producing Th1 cells while concurrently promoting the outgrowth of IL-4-expressing Th2 cells. Active suppression of T-cell activation resulting in liver-induced tolerance is also likely to occur within the liver because of its unique anatomy and composition of “ tolerogenic ” APCs. Within the liver, blood flow slows down through the narrow sinusoids (7-12 µm) and is temporarily obstructed by KC, which reside in the sinusoidal lumen. Because of this reduction in blood flow, circulating T cells can interact with LSEC and KC. Consequently, naïve T cells, which would normally encounter APCs in lymphoid tissues, could be primed directly by LSEC and/or KC within the liver. Current evidence suggests that LSEC and KC as well as hepatic dendritic cells are important in the induction of tolerance, rather than the activation of T-cell responses. It has been further demonstrated that although LSEC are capable of presenting antigen to T cells, LSEC-activated CD4 + or CD8 + T cells fail to differentiate into Th1 cells or cytotoxic effector cells, respectively. In addition, studies have shown that KC and hepatic dendritic cells are not effective APCs when compared with their counterparts in lymphoid tissues.

           In summary, it is the unique composition of innate and adaptive immune cells in the liver and the characteristic response of the liver to endogenous and exogenous antigens that may account for the mechanism of immune-mediated DILI, as well as the low occurrence and unpredictable nature of these reactions.



Role of Innate Immunity in DILI

               Drug-induced stress and/or damage of hepatocytes may trigger activation and inflammatory responses of the innate immune system within the liver. Evidence to support this idea has been mainly obtained from studies of liver injury induced by overdose of APAP, which is one of the few drugs that provide an experimental animal model of DILI. There is growing evidence that the initial NAPQI-induced hepatocyte damage may lead to activation of innate immune cells within the liver, thereby stimulating hepatic infiltration of inflammatory cells. Activated cells of the innate immune system produce a range of inflammatory mediators, including cytokines, chemokines, and reactive oxygen and nitrogen species that contribute to the progression of liver injury. Some of these mediators, such as IFN- , Fas, or Fas ligand, are directly involved in causing liver damage; mutant mice lacking these factors are resistant to APAP hepatotoxicity. On the other hand, the innate immune cells also represent a major source of hepatoprotective factors, as it has been demonstrated that transgenic mice deficient in IL-10, IL-6, or COX-2 are more susceptible to APAP-induced liver injury.


            The innate immune cells reported to participate in APAP hepatotoxicity include NK and NKT cells, macrophages, and neutrophils. A recent study demonstrated that depletion of NK and NKT cells protected mice from APAP-induced liver injury. This protective mechanism seems to involve eliminating the production of IFN- and various other pro-inflammatory chemokines as well as decreasing neutrophil accumulation within the liver. APAP hepatotoxicity has also been attributed in part to the activation of KC secondary to hepatocyte damage. KC activation results in the release of a wide range of proinflammatory mediators, such as TNF- , which may directly induce tissue damage, and IL-12 and IL-18, which are important activators of NK and NKT cells. However, other studies suggest that KC may play a protective role in addition to their protoxicant effect, as KC are the predominant source of IL-10 and IL-6, which are important in counteracting inflammatory responses and/or stimulating liver regeneration. Although many studies have shown neutrophil accumulation in the liver of APAP-treated animals, the pathogenic role of these cells remains controversial. One study has shown that in vivo injection of rabbit antineutrophil antiserum protected rats against APAP toxicity, while another study suggested that neutrophils are recruited into the liver to remove cellular debris and do not directly cause tissue damage.


          Our understanding of APAP-induced liver injury underscores the role of the innate immune system as an important regulator in the progression and severity of tissue damage. However, the temporal activation of different cell populations and their production of various mediators, as well as the interplay among these cells, warrant further investigation. A proposed mechanism of DILI involving parenchymal cell damage and subsequent activation of the innate immune system is presented in Figure 1 .

Role of Adaptive Immune Response in DILI

            The clinical features of some cases of DILI strongly suggest an involvement of the adaptive immune system. These clinical characteristics include (1) concurrence of rash, fever, and eosinophilia; (2) delay of the initial reaction (1-8 weeks) or requirement of repeated exposure to the culprit drug; (3) rapid recurrence of toxicity on reexposure to the drug; and (4) presence of antibodies specific for native or drug-modified hepatic proteins. (5) Drugs suspected to induce these types of reactions include halothane, tienilic acid, dihydralazine, diclofenac, phenytoin, and carbamazepine.


          Our current understanding of drug-induced adaptive immune responses is largely based on the hapten hypothesis and the p-i concept. The hapten hypothesis proposes that drugs, or more often reactive metabolites of the drugs, act as haptens and covalently bind to endogenous proteins to form immunogenic drug-protein adducts. These immunogenic adducts elicit either antibody or cytotoxic T-cell responses. The  hapten hypothesis is supported by the detection of antibodies that recognize drug-modified hepatic proteins in the sera of DILI patients. For example, antibodies that recognize trifluoroacetate-altered hepatic proteins have been detected in the sera of patients with halothane-induced hepatitis. Such drug-specific antibodies or autoantibodies that recognize native liver proteins have also been found in patients with liver injury caused by other drugs, such as tienilic acid, dihydralazine, and diclofenac. The p-i concept suggests that certain drugs can bind to T-cell receptors, mimicking a ligand and its receptor interaction, and cause T-cell activation in an MHC-dependent fashion. In patients who developed drug-induced systemic reactions of the liver and other organs, drug-specific T cells have been detected, and in some cases, T-cell clones were generated.


            Despite the detection of drug-specific antibodies and T cells, it has been difficult to directly prove the pathogenic role of the adaptive immune system in DILI, in part because of the lack of animal models. An important reason for the difficulty in developing animal models is that the default response of the liver to antigens is immunological tolerance. This tolerogenic response could also explain the low occurrence of this type of DILI in humans. As described in the above section, the anatomy, cellular composition, and microenvironment of the liver favor tolerance rather than pathogenic immunity. Therefore, DILI mediated by adaptive immune reactions against a drug can occur only when the tolerance mechanism is deficient or abrogated in susceptible individuals. As such, animal models of this type of DILI can be developed only after the barrier of such tolerance is overcome.
 






Figure.8.3:- General interrelationship between toxicity (including immune response; “hapten hypothesis”) anddisposition of a drug with reactive metabolites (left ). Scheme focusing on major processes relevant for the dispositionof reactive acyl glucuronides (right ), for which an antigen character of adducts was shown and antibodies have beendetected, also in vivo, in some cases. Acyl glucuronides are generating increasing interest as potential mediators ofhypersensitivity reactions and cellular toxicity.




Figure.8.4:- Illustration of the proposed mechanism of DILI, which involves drug metabolism, hepatocyte damage, activation of innate immune cells, and production of tissue-damaging and tissue-protective mediators. CYP indicates cytochrome P450; IFN, interferon; IL, interleukin; NK, natural killer cell; NKT, natural killer T cell; TNF, tumor necrosis factor.




Figure.8.5:- A 3-step mechanistic working model of hepatotoxicity. First, initial injury is exerted through direct cell stress, direct mitochondrial inhibition and/or specific immune reactions. Second, initial injury can lead to mitochondrial permeability transition (MPT). Direct cell stress causes MPT via the intrinsic pathway. The intrinsic pathway involves activation of intracellular stressor cascades and pro-apoptotic proteins including Bax. Alternatively MPT can be initiated through the death receptor-mediated extrinsic pathway that is activated by immune reactions and/or after sensitization to TNF and FasL binding to death receptors. Cytokines modulate the sensitivity of its activation. Third, MPT leads to necrosis or apoptosis depending on the availability of ATP. In hepatocytes activation of initiator caspase 8 through the extrinsic pathway is not sufficient to directly activate apoptosis, but amplification through pro-apoptotic factors including Bid and ceramides lead to MPT, which will then lead to the apoptotic pathway that is activated in the presence of sufficient remaining ATP production. Necrosis occurs if there is no ATP available, which is required for energy-consuming apoptotic pathways. Several highlighted amplification mechanisms (A) may play an important role at different levels for the idiosyncratic occurrence of hepatotoxicity.




Figure.8.6:- Risk factors for hepatotoxicity. First, risk factors can be classified into environmental vs. genetic factors. From a mechanistic point of view risk factors can further affect all different levels of events leading to the final outcome of drug-induced liver injury, which is mostly dichotomous, i.e. full recovery vs. acute liver failure. Note that risk factors affecting events downstream of initial injury are rather unspecific regarding different hepatotoxins. The figure presents a selection of well-described risk factors, but one must assume that other factors also play a role, of which many currently remain unknown.

  


Figure.8.7:- Six Mechanisms of Liver Injury. Injury to liver cells occurs in patterns specific to the intracellular organelles affected. The normal hepatocyte shown in the center of the figure may be affected in at least six ways, labeled A through F. Disruption of intracellular calcium homeostasis leads to the disassembly of actin fibrils at the surface of the hepatocyte, resulting in blebbing of the cell membrane, rupture, and cell lysis. In cholestatic diseases, disruption of actin filaments (B) may occur next to the canaliculus, the specialized portion of the cell responsible for bile excretion. Loss of villous processes and the interruption of transport pumps such as multidrug-resistance–associated protein 3 (MRP3) prevent the excretion of bilirubin and other organic compounds. Many hepatocellular reactions involve the heme-containing cytochrome P-450 system (C), generating high-energy reactions that can lead to the covalent binding of drug to enzyme, thus creating new, nonfunctioning adducts. These enzyme–drug adducts migrate to the cell surface (D) in vesicles to serve as target immunogens for cytolytic attack by T cells, stimulating a multifaceted immune response involving both cytolytic T cells and cytokines. Activation of apoptotic pathways by tumor necrosis factor a (TNF-a) receptor or Fas may trigger the cascade of intercellular caspases (E), which results in programmed cell death with loss of nuclear chromatin. Certain drugs inhibit mitochondrial function by a dual effect on both b-oxidation (affecting energy production by inhibition of the synthesis of nicotinamide adenine dinucleotide and flavin adenine dinucleotide, resulting in decreased ATP production) and the respiratory-chain enzymes (F). Free fatty acids cannot be metabolized, and the lack of aerobic respiration results in the accumulation of lactate and reactive oxygen species. The presence of reactive oxygen species may further disrupt mitochondrial DNA. This pattern of injury is characteristic of a variety of agents, including nucleoside reverse-transcriptase inhibitors, which bind directly to mitochondrial DNA, as well as valproic acid, tetracycline, and aspirin. Toxic metabolites excreted in bile may damage bile-duct epithelium (not shown). DD denotes death domain.




No comments:

Post a Comment