Sei sulla pagina 1di 24

Ch006-X2998.

qxd

3/15/06

5:21 PM

Page 87

Section I: Pathophysiology of the Liver

HEPATIC FIBROSIS AND CIRRHOSIS


Don C. Rockey and Scott L. Friedman
Abbreviations ALT alanine aminotransferase AST aspartate aminotransferase AUROC area under the receiver operator characteristic BMI body mass index CINC cytokine-induced neutrophil chemoattractant CTGF connective tissue growth factor DDR discoidin domain receptors ECM extracellular matrix EGF epidermal growth factor ELF European liver brosis ET-1 endothelin-1 FGF broblast growth factor FPI brosis probability index GGT g-glutamyl transferase

6
MMP-9 NASH NGFR NO PDGF PELD PIIINP PPAR QTL ROC TGF-b1 TIMPs ULN VEGF matrix metalloproteinase 9 non-alcoholic steatohepatitis nerve growth factor receptor nitric oxide platelet-derived growth factor pediatric end-stage liver disease propeptide of type III collagen peroxisomal proliferator-activated receptor quantitative trait loci receiver operating characteristic transforming growth factor beta 1 tissue inhibitors of metalloproteinases upper limit of normal vascular endothelial growth factor

GnT-III HA HBV HCV HGF HIV HOMA-IR IL-10 JI LPS MCP-1 MEGX MELD MMPs MMP-2

N-acetylglucosaminyl transferase III hyaluronic acid hepatitis B virus hepatitis C hepatocyte growth factor human immunodeciency virus insulin resistance by the homeostasis model assessment interleukin-10 jejuno-ileal lipopolysaccharide monocyte chemotactic protein-1 monoethylglycinexylidide model for end-stage liver disease matrix metalloproteinases matrix metalloproteinase 2

INTRODUCTION
Hepatic brosis has emerged as a highly relevant aspect of liver biology because of the signicant progress in uncovering its mechanisms, combined with a growing realization that effective antibrotic therapies may soon alter the natural history of chronic liver disease. Thus, liver brosis can now be viewed as a clinical problem whose diagnosis and treatment will soon have rational, evidence-based approaches. This progress is very timely, as the continued aging of the HCV-infected cohort and the growing prevalence of obesityrelated liver diseases are leading to precipitous increases in the prevalence of advanced liver disease.1 With these issues in mind, this chapter will review clinical aspects of hepatic brosis, including natural history, pathophysiologic mechanisms, current and future tools for diagnosis, and emerging antibrotic strategies. In addition, several recent reviews highlight many of these aspects in greater detail.26 Hepatic brosis is the accumulation of extracellular matrix, or scar, in response to acute or chronic liver injury. Fibrogenesis represents a wound healing response to injury (Figure 6-1), and ultimately leads to cirrhosis. Cirrhosis is the end-stage consequence of brosis of the hepatic parenchyma, resulting in nodule formation that may lead to altered hepatic function and blood ow. Both brosis and cirrhosis are the consequences of a sustained wound-healing response to chronic liver injury from a range of causes, including viral, autoimmune, drug induced, cholestatic and metabolic diseases. The clinical manifestations of cirrhosis vary widely, from no symp-

toms at all to liver failure, and are determined by both the nature and severity of the underlying liver disease as well as the extent of hepatic brosis. Up to 40% of patients with cirrhosis are asymptomatic and may remain so for long periods, but progressive deterioration leading to death or liver transplantation is typical once complications (such as ascites, variceal hemorrhage or encephalopathy) develop. In such patients there is a 50% 5-year mortality, with approximately 70% of these deaths directly attributable to liver disease.7 In asymptomatic individuals cirrhosis may be rst suggested during routine examination, although histologic analysis may be required to establish the diagnosis. Cirrhosis affects hundreds of millions of patients worldwide. The overall burden of liver disease in the United States the vast majority of which is due to chronic disease with brosis continues to expand, exacting an increasing economic and social cost.1 Indeed, in the US cirrhosis is the most common non-neoplastic cause of death among hepatobiliary and digestive diseases, accounting for approximately 30 000 deaths per year. In addition, 10 000 deaths are due to liver cancer, the majority of which arise in cirrhotic livers, consistent with a steadily rising mortality rate from hepatic cancer.8 Notably, hepatocellular carcinoma is the most rapidly increasing neoplasm in the US and western Europe.9 Initial studies of hepatic brosis focused on the composition of extracellular matrix in liver, and continued incremental progress in this area is still anticipated. However, attention has gradually shifted towards exploring the cellular basis of brosis and the cellular mediators that drive brosis progression and regression (see Pathophysiology, below). In general, the molecular composition of the scar

87

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 88

Section I. Pathophysiology of the Liver

Normal liver Hepatocytes

Liver injury Loss of Hepatocyte microvilli

Space of Disse

Quiescent stellate cell Kupffer cell Hepatic sinusoid Endothelial cell

Activated stellate cell Deposition of scar matrix

Loss of fenestrae

Kupffer cell activation

Figure 6-1. Hepatic liver cells and the hepatic sinusoid in normal and injured liver. On the left panel is shown the multiple key liver-specic cellular elements in the normal liver, including hepatocytes, endothelial cells, Kupffer cells, and stellate cells. Stellate cells are located within the subendothelial space of Disse (i.e. between the sinusoidal endothelium and hepatocytes). The gure emphasizes the close physical relationships between the various cellular elements in the liver. After liver injury, changes in numerous cells occur; for example, stellate and Kupffer cells become activated (see Figure 6-3), hepatocytes lose their microvilli, and endothelial cells lose their characteristic fenestrae. All of these features contribute to continued cell activation and injury, as well as dysfunction at the whole organ level.

tissue in cirrhosis is similar regardless of etiology, and resembles that of other parenchymal scarring (e.g. kidney), consisting of the extracellular matrix constituents, collagen types I and III (i.e. brillar collagens), sulfated proteoglycans, and glycoproteins.10 However, some isoforms of extracellular matrix constituents, for example bronectin11 and proteoglycans,12 may be relatively enriched during progressive injury. These scar constituents accumulate from a net increase in their deposition in liver and not simply from the collapse of existing stroma.

CLINICAL ASPECTS OF HEPATIC FIBROSIS


NATURAL HISTORY AND RISK FACTORS
Fibrosis leading to cirrhosis can accompany virtually any chronic liver disease that is characterized by the presence of architectural disruption and/or inammation. The vast majority of patients with liver disease worldwide have chronic viral hepatitis, or steatohepatitis associated with either alcohol or obesity; other etiologies of liver disease include parasitic infestation (e.g. schistosomiasis), autoimmune attack on hepatocytes or biliary epithelium, neonatal liver disease, metabolic disorders including Wilsons disease,

hemochromatosis and a variety of storage diseases, chronic inammatory conditions (e.g. sarcoidosis), drug toxicity (e.g. methotrexate or hypervitaminosis A), and vascular derangements, either congenital or acquired. Of the many causes of chronic liver disease, our understanding of natural history of brosis is most complete in HCV, with some information about HBV and steatohepatitic diseases, including alcoholic liver disease and NASH. Information about brosis progression in other diseases is largely anecdotal, but the development of cirrhosis typically requires many years to decades. There are, however, some notable exceptions in which the development of cirrhosis can be greatly accelerated, possibly occurring within months rather than years: (1) neonatal liver disease infants with biliary atresia may present at birth with severe brosis and marked parenchymal distortion; (2) HCV-infected patients after liver transplantation a subset of patients who undergo liver transplantation for HCV cirrhosis may develop rapidly progressive cholestasis and recurrent cirrhosis within months, requiring retransplantation;13 (3) patients with HIV/HCV co-infection these patients have relatively rapid brosis compared to those with HCV alone,14 especially if the HIV is untreated (see below); (4) severe delta hepatitis;15 and (5) some cases of drug-induced liver disease. These examples of fulminant brosis probably reect dysregulation of several pathways, including defective immunity, massive inammation and necrosis, and/or

88

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 89

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

altered matrix resorption. Together, they highlight the highly dynamic nature of scar accumulation and degradation. Moreover, when matrix accumulation is unopposed because degradation is ineffective, more rapid brosis may ensue. Once cirrhosis and its complications develop the prognosis is predicted by widely used systems, including ChildPugh, PELD16 and MELD,17 which are predictive independent of the etiology of liver disease.

Hepatitis C Virus
The risk and natural history of brosis associated with HCV have been greatly claried as a result of several large clinical studies incorporating standardized assessments of brosis that combine detailed historical and clinical information.18 The disease can run a remarkably variable course, from decades of viremia with little brosis to a rapid onset of cirrhosis within 1015 years. It appears to be host factors rather than viral factors that correlate with brosis progression in HCV. The data supporting this conclusion include the following: (1) there is no relationship between viral load or genotype and severity of brosis even though these former factors affect the response to antiviral therapy; (2) human promoter polymorphisms (e.g. TGF-b1 and angiotensin) appear to correlate with brosis risk,19 with large-scale efforts currently under way to identify additional genetic markers of brosis risk;20 (3) host immune phenotype may be critical, as there is more rapid progression in immunosuppressed patients, whether due to HIV or to immunosuppressive drugs.14 In mice, a Th2 phenotype strongly correlates with brogenic potential,21 which has led to successful efforts to use quantitative trait loci (QTL) mapping to identify specic brosis risk genes in these animals.22 Other identied host risk factors for more rapid progression of HCV include: (1) older age at the time of infection; (2) concurrent liver disease due to HBV or alcohol (>50 g/day); it is uncertain, however, whether lesser amounts of alcohol intake are additive towards brosis progression: recent studies suggest that less than 50 g/day of alcohol results in a neglible increased risk of hepatic brosis;23 (3) male gender; (4) increased body mass index (BMI), associated with hepatic steatosis;24 (5) HIV infection or immunosuppression following liver transplantation. Because standard clinical indices cannot distinguish between minimal and even advanced brosis, knowledge about these risk factors and duration of infection can greatly inform clinical management. Thus, for chronic HCV, if the time of infection is known and a biopsy obtained at any time thereafter, the rate of progression per year based on either Ishak or METAVIR scoring can be estimated.25 Although initial analyses of this type suggested that brosis progression is truly linear, it is now increasingly clear that the progression rate accelerates as the disease advances,26 such that it takes less time to progress between Metavir stages 3 and 4, than from stage 1 to 2, for example. Assessment of brosis stage and rate of brosis progression can be valuable for at least three reasons: (1) the actual stage of brosis will indicate the likelihood of response to a-interferon or ainterferon/ribavirin, as the advanced stages of brosis (F3 or F4) generally have a lower response rate to antiviral therapy;27,28 (2) if little brosis progression has occurred over a long interval, then treatment with antiviral therapy may be deemed to be less urgent

and it may be safe to await more effective and/or better-tolerated therapy; (3) the approximate time to the development of cirrhosis can be estimated. This would not, however, indicate if or when clinical liver failure would occur, as the complications of liver disease may be delayed for up to a decade or more after the establishment of cirrhosis. As genetic risk markers that predict a rapid brosis progression rate are developed, this information, combined with the absolute stage of brosis, may enable more accurate identication of patients at risk for disease and thus in need of antibrotic therapy.

Hepatitis B Virus
Very few studies have assessed the progression rate of brosis in chronic HBV infection. In general, inammatory activity, as inuenced by viral factors, including e Ag status, that indicate active viral replication, correlates with brosis.29,30 Fibrosis progression has been correlated with HBV genotype in at least one study.31 In a subset of patients a rapidly progressive brosing cholestatic hepatitis may occur,32 but there are neither denitive risk factors for this condition nor unique etiologic, cellular or molecular determinants identied. In addition, delta hepatitis superinfection or co-infection may greatly accelerate the risk of advanced brosis and cirrhosis.15 What is striking, however, is that virologic suppression in response to potent antiviral regimens can effect remarkable improvement not only in serum alanine aminotransferase (ALT) levels and histologic inammation, but also in brosis.15,3335 Indeed, dramatic resolution of cirrhosis in a 10-year follow-up has been reported in patients with delta hepatitis who were successfully treated with a-interferon.15

Alcoholic Liver Disease


The clearest clinical determinant of brosis is continued alcohol abuse: patients with brosis who continue to drink are virtually assured of progression. In addition, two clinical features commonly seen in steatohepatitis, elevated BMI and serum glucose, also confer an increased risk of brosis in alcoholic liver disease.36 Pathologically, the presence of pericentral brosis (central hyaline sclerosis) carries a high risk of eventual panlobular cirrhosis, which is almost certain if alcohol intake continues.

Non-Alcoholic Steatohepatitis
There is a critical need for better data about natural history, risk factors for brosis, and rate of brosis progression in NASH, issues now being addressed in several multicenter studies. Patients with only steatosis and no inammation appear to have a benign course when followed for up to 19 years;37 however, it is unclear whether this lesion is completely distinct from steatohepatitis, or instead represents a precursor of NASH. It is instructive to remember that HCV brosis progression rates were underestimated shortly after the virus was rst identied, as many patients had a relatively early brosis stage. With continued infection, however, a sizeable fraction eventually have progressed to more advanced stages. In a parallel situation, the obesity epidemic in the US and the developed world is only now being fully appreciated, and a threshold level of obesity may have only begun to confer a risk of liver disease that will become clinically signicant in the next decade. In patients with sustained

89

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 90

Section I. Pathophysiology of the Liver

NASH spontaneous histologic improvement is very uncommon, but better longitudinal data are needed to understand the natural history of this disease; for example, data examining the evolution of NASH in patients undergoing bariatric surgery who have extensive weight loss and improvement in their metabolic syndrome are awaited. In three combined studies of 26 patients followed with sequential biopsies for up to 9 years, 27% had progression of brosis and 19% advanced to cirrhosis, but none had reversal of brosis.37 Of interest is the recurrence of NASH following liver transplantation in some patients with cryptogenic cirrhosis, implicating an underlying metabolic defect that may account for liver disease in both the native and the transplanted organs. Risk of brosis and rate of progression are critical issues that will inuence risk stratication and patient selection for clinical trials, as progression to cirrhosis is the most important clinical consequence of NASH. Recently developed systems to grade and stage liver disease in NASH38 should allow for improved, prospective collection of standardized data that can further address these vital questions. In general, increasing obesity (BMI) >28 kg/m2) correlates with severity of brosis and risk of cirrhosis. Other risk factors include necroinammatory activity with ALT >2 normal and/or AST/ALT >1, age, elevated triglycerides, insulin resistance and/or diabetes mellitus, and systemic hypertension.39 It is uncertain whether these features are comparable across the spectrum of disorders associated with NASH, including obesity with insulin-resistance, JI (jejuno-ileal) bypass, total parenteral nutrition and rapid weight loss, among others. Whether these factors represent surrogates for other risk factors (i.e. reduced antioxidant levels in older patients, increased reninangiotensin activity in hypertensives) is unknown. Ratziu and colleagues40 have reported a clinicobio-logical score that combines age, BMI, triglycerides and ALT and which reportedly has 100% negative predictive value for excluding signicant brosis.

It remains unclear what distinguishes those patients whose cirrhosis is reversible from those in whom it is xed. Potential factors inuencing reversibility probably include: (1) a prolonged period of established cirrhosis, which could reect a longer period of crosslinking of collagen, rendering this collagen less sensitive to degradation by enzymes over time. Animal studies now clearly support this possibility;47 (2) total content of collagen and other scar molecules, which might lead to a large mass of scar that is physically inaccessible to degradative enzymes; (3) reduced expression of enzymes that degrade matrix, or sustained elevation of proteins that inhibit the function of these degradative enzymes, in particular elevated levels of tissue inhibitors of metalloproteinases (TIMPs), which block matrix proteases and also prevent apoptosis of activated stellate cells.48,49 All three scenarios highlight the dynamic process of collagen deposition and degradation.

PATHOPHYSIOLOGY OF HEPATIC FIBROSIS AND CIRRHOSIS


EXTRACELLULAR MATRIX (ECM) IN THE NORMAL AND THE FIBROTIC LIVER
Extracellular matrix refers to the array of macromolecules that comprise the scaffolding of normal and brotic liver. These macromolecules consist of three main families: collagens, glycoproteins and proteoglycans (see 10 for review). The number of collagens identied in liver is rapidly growing, and includes collagen XVIII, which is a precursor to the molecule angiostatin. Glycoproteins include bronectin, laminin, merosin, tenascin, nidogen, and hyaluronic acid, among others. Proteoglycans include heparan, dermatan sulfates, chondroitin sulfates, perlecan dystroglycan syndecan, biglycan and decorin. There is tremendous heterogeneity of these matrix macromolecules with respect to their different isoforms, variable combinations within different tissue regions, and changes related to age. In normal liver the subendothelial space of Disse separates the epithelium (hepatocytes) from the sinusoidal endothelium. This space contains a basement membrane-like matrix which, unlike the typical basement membrane, is not electron dense. The hepatic basement membrane is composed of non-bril-forming collagens, including types IV, VI and XIV, glycoproteins and proteoglycans. This normal subendothelial ECM is critical for maintaining the differentiated functions of resident liver cells, including hepatocytes, stellate cells and sinusoidal endothelium. In contrast to basement membrane-type matrix, in normal liver the so-called interstitial ECM is largely conned to the capsule, around large vessels, and in the portal areas. It is composed of brilforming collagens (e.g. types I and III) together with cellular (EDA) bronectin, undulin, and other glycoconjugates. As the liver becomes brotic, the total content of collagens and non-collagenous components increases three- to vefold, accompanied by a shift in the type of ECM in the subendothelial space from the normal low-density basement membrane-like matrix to interstitial-type matrix (see 10 for review). This capillarization leads to the loss of hepatocyte microvilli and the disappearance of endothelial fenestration (Figure 6-1).

REVERSIBILITY OF FIBROSIS AND CIRRHOSIS


There is now clear evidence that brosis and even cirrhosis can be reversible. The feature common to all cases of cirrhosis improvement is the elimination of the underlying cause of liver disease, whether due to eradication of HBV,41 delta hepatitis15 or HCV,42 decompression of biliary obstruction in chronic pancreatitis,43 or to immunosuppressive treatment of autoimmune liver disease.44 Moreover, there is ample evidence of reversibility in animal models, which provide vital clues to underlying mechanisms.45 Earlier studies demonstrated that brosis improves with treatment of HCV,46 and even cirrhosis can regress following HCV eradication with a-interferon/ribavirin.42 Among a large cohort of patients successfully treated with this combination there were 150 with cirrhosis, half of whom had a reduction in their brosis score according to METAVIR staging, with several regressing by two or more stages. Because brosis in HCV typically progresses over three decades, one might anticipate an equally slow but steady regression of brosis following viral clearance.

90

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 91

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

The outcome of brogenesis is the conversion of normal lowdensity basement membrane-like matrix to high-density interstitialtype matrix. A number of components are responsible for ECM remodeling (see 49,50 for reviews) (Figure 6.2). These include a family of zinc-dependent enzymes matrix metalloproteinases (MMPs),51 their inhibitors (tissue inhibitor of metalloproteinases, TIMP), and several converting enzymes (MT1-MMP , and stromelysin, for example). In human liver diseases there is down-regulation of MMP1 (interstitial collagenase, collagenase I) and up-regulation of MMP2 (gelatinase A) and MMP9 (gelatinase B). Based on the differing substrate specicities of these enzymes, the result is increased degradation of basement membrane collagen and decreased degradation of interstitial collagens. These activated MMPs are regulated in part by their tissue inhibitors, the so-called TIMPs. TIMP1 and TIMP2 are upregulated relative to MMP1 in progressive experimental liver brosis, which may explain the decreased degradation of interstitial-type matrix observed in experimental and human liver injury. In contrast, during the resolution of experimental liver injury TIMP-1 and TIMP-2 expression is decreased whereas collagenase expression is unchanged, resulting in a net increase in collagenase activity and increased resorption of scar matrix. Stellate cells are a key source of MMP-2 and stromelysin. They also express TIMP-1 and TIMP-2 mRNAs and produce TIMP-1 and MT1-MMP MMP-9, which is a type IV collagenase locally secreted by Kupffer cells, and may also be produced by stellate cells in

response to interleukin-1.52 The source of MMP-1, which plays a crucial role in degrading the excess interstitial matrix in advanced liver disease, is still uncertain.53 However, interstitial collagenase activity in liver may be attributable to either MT1-MMP or even MMP-2, although further studies are required.

ECMCELL INTERACTIONS
Changes in the microenvironment of the space of Disse result in phenotypic changes in all resident liver cells. Hepatic stellate cells are activated by the surrounding increase in interstitial matrix.54 Sinusoidal endothelial cells produce cellular bronectin in very early liver injury, which also contributes to stellate cell activation. In addition, endothelial cells produce type IV collagen, proteoglycans and factors (e.g. urokinase-type plasminogen activator) that participate in the activation of latent cytokines such as TGF-b1. Activated Kupffer cells release cytokines and reactive oxygen intermediates that may stimulate stellate cells in a paracrine manner.55 Platelets are also an abundant source of cytokines upon injury, producing a rich array of important growth factors. Hepatocytes, the most abundant cells in the liver, generate lipid peroxides following injury that lead to stellate cell activation, a prerequisite for brogenesis (see below). The dynamic interactions between brogenic cells in liver and the ECM is an important determinant of brogenesis. The ECM is a reservoir for growth factors, for example platelet-derived growth factor (PDGF).10 Like all cytokines, PDGF signals by binding to

Activated stellate cell Early pathological degradation Regression Apoptotic stellate cell Normal ECM MT1-MMP MMP-2 TIMP-2

Kupffer cell MMP-1 MMP-13 Other MMPs Progression

TIMP-1 TIMP-2

Figure 6-2. Emerging Mechanisms of Early Pathologic Matrix Degradation, Fibrosis Progression & Fibrosis Resolution in Chronic Liver Disease. Activation of stellate cells (top left panel) is a key event in hepatic brosis, and is associated with pathologic matrix degradation due to increased production of membrane type matrix metalloproteinase 1 (MT1-MMP), matrix metalloproteinase-2 (MMP-2), and tissue inhibitors of metalloproteinases (TIMPs), leading to replacement by interstitial collagen or scar matrix. As brosis progresses (middle panel), sustained expression of TIMPs prevents matrix degradation and apoptosis of activated stellate cells. Regression of brosis (upper right panel) is associated with increased apoptosis of activated stellate cells. Apoptosis requires decreased expression of tissue inhibitor of metalloproteinase-1 (TIMP-1), yielding a net increase in protease activity. These events may occur coincident with production of matrix metalloproteinases, which could include MMP-1 (in humans) and/or MMP-13 (in rodents), although cellular sources of these enzymes (possibly including Kupffer cells), and clear evidence of their induction in vivo are still lacking. Validation of these events and further elucidation of mechanisms underlying brosis regression represent key challenges for future studies.147a

91

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 92

Section I. Pathophysiology of the Liver

membrane receptors. The PDGF receptor belongs to a receptor family known as receptor tyrosine kinases, which collectively are key transducers for many important cytokines, including hepatocyte growth factor (HGF), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF) and broblast growth factor (FGF). Interestingly, a new subclass of receptor tyrosine kinases, socalled discoidin domain receptors (DDR), has been identied; this group of receptors signal in response to brillar collagens rather than peptide ligands.56 Indeed, stellate cell activation is accompanied by up-regulation of DDR2 receptors, and increased signaling is associated with altered MMP-2 expression.57,58 Intracellular signaling cascades downstream of receptor tyrosine kinases and other receptors are pervasive (see 59 for review). Integrins are another type of membrane receptor that transduce extracellular signals in liver. These are heterodimeric transmembrane proteins composed of an a and a b subunit whose ligands are matrix molecules rather than cytokines. Several integrins and their downstream effectors have been identied in stellate cells, including a1b1, a2b1, a5b1, avb1, avb3 and a6b4.6,60,61 Integrins may also complex with other receptor families in mediating cell motility and brogenesis, for example the tetraspanin family of receptors.62

HEPATIC STELLATE CELL ACTIVATION THE COMMON PATHWAY LEADING TO HEPATIC FIBROSIS
The identication of stellate cells as the key cellular source of extracellular matrix in liver has been a major advance. This distinct cell population, located in subendothelial space of Disse between hepatocytes and sinusoidal endothelial cells (Figure 6-1), represents onethird of the non-parenchymal population or about 15% of the total number of resident cells in normal liver.63 In normal liver they are the principal storage site for retinoids (vitamin A metabolites),

which accounts for 4070% of retinoids in the body. Most of the retinoids are in the form of retinyl esters and are conned to cytoplasmic droplets. Preferential expression of ECM genes in stellate cells has been conrmed in mechanistically distinct experiment models of injury. Recent studies have emphasized the heterogeneity of mesenchymal populations in the liver, with variable expression of neural,64 angiogenic,65 contractile,66 and even bone marrow-derived67 markers. Moreover, experimental genetic marking of stellate cells by the expression of uorescent proteins downstream of either brogenic or contractile gene promoters illustrates the plasticity of brogenic populations in vivo.68 In view of this capacity for transdifferentiation between different mesenchymal cell lineages, and possibly even from epithelium,69 the key issue is whether brogenic cells express target molecules such as receptors or cytokines in sufcient concentrations in vivo to merit their targeting by diagnostic agents or antibrotic compounds. Following liver injury of any etiology, stellate cells undergo a process known as activation, which is characterized by the transition of quiescent vitamin A-rich cells into proliferative, brogenic, and contractile myobroblasts.54 Stellate cell activation is typically a result of complex interplay among ECM (Figure 6.2) and cellular (Figure 6.3) elements found in the local environment. It should be noted that activation most often occurs in the setting of hepatocellular injury and subsequent inammation. Activation can be conceptually viewed as a two-stage process: initiation (also referred to as preinammatory) and perpetuation54 (Figure 6-4). Initiation refers to early changes in gene expression and phenotype that render the cells responsive to other cytokines and stimuli, whereas perpetuation results from the effects of these stimuli on maintaining the activated phenotype and generating brosis. Initiation is largely due to paracrine stimulation, whereas perpetuation involves autocrine as well as paracrine loops.

Inciting injury Recruitment of inflammatory cells T-cell NK cell Hepatocyte Stellate cell Kupffer cell

Expression of cytokines

Figure 6-3. Cellular response to wound healing. Most forms of liver injury result in hepatocyte injury followed by inammation, which in turn leads to activation of hepatic stellate cells. Inammatory effectors are multiple and include T cells, NK and NKT cells as well as Kupffer cells. These cells produce growth factors, cytokines, and chemokines that play an important role in stellate cell activation. Additionally, injury leads to disruption of the normal cellular environment, and also to stellate cell activation (right upper panel). Once activated, stellate cells themselves produce a variety of compounds, including growth factors, cytokines, chemokines, and vasoactive peptides. These substances have pleotropic effects in the local environment, including many which have autocrine effects on stellate cells themselves. One of the major results of stellate cell activation is extracellular matrix synthesis, as well as the production of matrix degrading enzymes.

Activated stellate cell

Stellate cell activation

92

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 93

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

PERPETUATION PROLIFERATION INITIATION CONTRACTILITY

ET-1 PDGF TGF-1 FIBROGENESIS

RESOLUTION MMP-2 MATRIX DEGRADATION PDGF MCP-1 REVERSION? MCP-1 PDGF

CHEMOTAXIS APOPTOSIS LEUKOCYTE CHEMOTAXIS RETINOID LOSS

Figure 6-4. Stellate cell activation. Stellate cell activation is a key pathogenic feature underlying liver brosis and cirrhosis. Multiple and varied stimuli contribute to the induction and maintenance of activation, including (but not limited to) cytokines, peptides, and the extracellular matrix itself. Key phenotypic features of activation include the production of extracellular matrix, loss of retinoids, proliferation, of up-regulation of smooth muscle proteins, secretion of peptides and cytokines (which have autocrine effects), and up-regulation of various cytokine and peptide receptors. Reprinted with permission from ref 54a.

Initiation
Oxidant stress may be an early determinant of stellate cell activation. In hepatic injury, whether subclinical or overt, there is a perturbation of normal liver homeostasis, with extracellular release of either free radicals (i.e. oxidant stress), intracellular constituents, and/or cytokines and signaling molecules. Sources of these mediators may be circulating (i.e. endocrine), paracrine or autocrine. In particular, oxidant stress-mediated necrosis leading to stellate cell activation may underlie a variety of liver diseases, including hemachromatosis, alcoholic liver disease, viral hepatitis and nonalcoholic steatohepatitis (NASH).55,70,71 Liver injury is typically associated with inltration of inammatory cells, but even in their absence the liver contains sufcient resident macrophages (Kupffer cells) and natural killer cells (pit cells) to initiate local inammation prior to the arrival of extrahepatic cells. In addition to oxidant stress, following early injury endothelial cells produce a splice variant of cellular bronectin that is able to stimulate stellate activation.

Endothelial cells in early injury may also participate in the conversion of latent TGF-b1 to its active, probrogenic form through the activation of plasmin. Whereas necrosis is considered a classic inammatory and brogenic stimulus, recent ndings also suggest that apoptosis may provoke a brogenic response in stellate cells. Apoptotic fragments released from hepatocytes are brogenic towards cultured stellate cells,72 and Fas-mediated hepatocyte apoptosis in vivo in experimental animals is also brogenic.73 Platelets in injured liver are a potent source of paracrine stimuli by generating multiple potentially important mediators, including PDGF, TGF-b1, and epidermal growth factor (EGF). Additionally, activated stellate cells have also been observed in primary and metastatic human tumors, as well as a murine model of metastatic melanoma to liver.74 In recent years, increasing interest has been focused on the molecular regulation of gene expression during early stellate cell activa-

93

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 94

Section I. Pathophysiology of the Liver

tion. There have been many advances in dissecting pathways of membrane and intracellular signaling and transcriptional gene regulation in activated hepatic stellate cells that are too numerous to detail here.75 A growing number of transcription factors may regulate stellate cell behavior, including peroxisomal proliferator-activated receptors (PPAR) a, b and g,76 retinoid receptors,77 NF-kB,78,79 Jun D,75 Krppel-like factor 6 (previously called Zf9),80 Foxf1,81 and CRP282 among others.

The expression of smooth muscle a actin is increased during stellate cell activation. ET-1 and other vasoactive mediators increase their expression.83 Thus studies of contractile proteins in stellate cells may yield a therapeutic target for the treatment of intrahepatic portal hypertension.

Fibrogenesis
Fibrogenesis is perhaps the key component of the stellate cells contribution to hepatic brosis. TGF-b1 is the most potent brogenic factor, with some brogenic activity documented for interleukin-1b, TNF, lipid peroxides, acetaldehyde, and others (see 2,6 for reviews). Because of its importance, TGF-b1 regulation has received considerable attention. TGF-b1 is up-regulated in experimental and human hepatic brosis. Although sources of this cytokine are many, autocrine expression is among the most important (see 2 for review). Several mechanisms underlie the increase in TGF-b1 expression by stellate cells during liver injury, including TGF-b transcriptional upregulation, activation of latent TGF-b1, increased TGF-b receptor expression, and up-regulation of TGF-b signaling components.9296

Perpetuation
After initiation, activated stellate cells undergo a series of phenotypic changes that collectively lead to the accumulation of ECM. These include proliferation; contractility; brogenesis; chemotaxis; matrix degradation; retinoid loss; and proinammatory responses and cytokine release. The following sections detail the mechanisms underlying each of these events.

Proliferation
An increase in the number of stellate cells has been documented after both human and experimental liver injury, in large part due to local proliferation. Following liver injury, many mitogenic factors as well as their cognate tyrosine kinase receptors are unregulated, primariliy through receptor tyrosine kinases.59 PDGF is the bestcharacterized and most potent mitogen towards stellate cells. Upregulation of PDGF receptor following liver injury enhances the responsiveness to autocrine PDGF, whose expression is also increased. The downstream signaling pathways involve ERK/MAP kinase, phosphoinositol 3 kinase (PI 3-kinase) and STAT-1 (signal transducers and activators of transcription) (see 3 for review). PDGF-induced proliferation correlates with increased intracellular Ca2+ and pH, raising the possibility that calcium channel blockers might modulate stellate cell mitogenesis or activation. Other stellate cell mitogens include endothelin-1 (ET-1),83,84 thrombin,85 FGF,86 and IGF,87,88 among others (see 89,90 for reviews). A recent study has documented increased sensitivity to ET-1 during activation,91 suggesting potentiation of autocrine/paracrine stimulation.

Chemotaxis
Stellate cells may accumulate both through proliferation and via directed migration into regions of injury, or chemotaxis. PDGF, the leukocyte chemoattractant MCP-1, and a growing family of chemokines have been identied as key stellate cell chemoattractants.97 In addition to tyrosine kinase receptors, new agents have been implicated in stellate cell migration, in particular tetraspanin receptors.3,62

Matrix Degradation
A greater understanding of matrix degradation in liver is emerging. Quantitative and qualitative changes in the activity of MMPs and their inhibitors play a vital role in extracellular matrix remodeling in liver brogenesis (see ECM in the normal and brotic liver and Figure 6.2 above). As noted above, the net effect of changes in matrix degradation is the conversion of the low-density subendothelial matrix to one rich in interstitial collagens.

Retinoid Loss Contractility


Contraction by stellate cells may be a major determinant of early and late increases in portal resistance during liver brosis. Activated stellate cells impede hepatic blood ow both by constricting individual sinusoids and by contracting the cirrhotic liver, as the collagenous bands typical of end-stage cirrhosis contain large numbers of activated stellate cells (see 66 for review). A key contractile stimulus towards stellate cells is ET-1.66 Other contractile agonists include arginine vasopressin, adrenomedullin, and eicosanoids.66 The regulation of stellate cell contraction is complex. The endothelium-derived relaxing factor nitric oxide (NO) appears to be an important relaxing factor in the sinusoid (although other factors, such as carbon monoxide, also play a role). The net contractile activity of stellate cells in vivo therefore reects the relative strength of each of these opposing activities. Current evidence suggests that intrahepatic portal hypertension probably results from diminished NO (and/or other vasodilators) activity as well as increased stimulation by ET-1 (or other constrictors).66 Stellate cell activation is accompanied by loss of their characteristic perinuclear retinoid (vitamin A) droplets. Although the intracellular form is largely retinyl esters, when retinol is exported from the cell during activation it is primarily as retinol, suggesting the possibility of intracellular hydrolysis of esters before being exported. Several nuclear retinoid receptors that bind intracellular retinoid ligands have been identied and their effects characterized in stellate cells.77,98

Proinammatory Responses and Cytokine Release


Hepatic stellate cells and sinusoidal endothelial cells have emerged as inammatory effectors. Sinusoidal endothelial cells, normally fenestrated to allow rapid bidirectional transport of solutes between sinusoidal blood and parenchymal cells, may rapidly lose their fenestrations upon injury and express proinammatory molecules, including ICAM-1, VEGF and adhesion molecules.74,99 Together with stellate cells, they activate angiogenic pathways in response to hypoxia associated with local injury or malignancy.74,97,100,101

94

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 95

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

Key inammatory pathways converge on stellate cells, leading to brosis (see Figure 6-3). Thus, the cell type is a central mediator in inammation, rather than just a passive target. Upon activation, they release chemokines97,102 and other leukocyte chemoattractants, proteinase-activated receptors,103 and up-regulate expression of key inammatory receptors, including ICAM-1,104 chemokine receptors,105 and those mediating lipopolysaccharide (LPS) signaling, including Toll-like receptor 4.78 Stellate cells may also contribute to intrahepatic apoptosis of T lymphocytes.106 Remarkably, little attention has focused on the contribution of different lymphocyte subsets to hepatic brogenesis. Interest has increased recently, in part because of the observation that patients with HCV who are co-infected with HIV, as well as those who are immunosuppressed following liver transplantation, have accelerated brosis rates, implicating the immune system as a determinant of brogenesis. These observations have been supported by animal studies demonstrating that the immune phenotype regulates brogenesis independent of effects on injury, which in turn have led to efforts to map the genetic loci accounting for these differences.22 Most recently, CD8 lymphocytes have emerged as potential probrogenic cells, based on their ability to induce early brogenesis following adoptive transfer to nave SCID mice from animals with liver injury.107 Autocrine cytokines play vital roles in regulating the activation process of stellate cells. These cytokines include TGF-b1, PDGF, FGF, HGF, PAF, stem cell factor and ET-1, among others.59,90,97,108 Furthermore, stellate cells release neutrophil and monocyte chemoattractants, which can amplify inammation in liver injury. These chemokines include colony-stimulating factor, monocyte chemotactic protein-1 (MCP-1), and cytokine-induced neutrophil chemoattractant (CINC).97 Anti-inammatory cytokines produced by stellate cells have also been identied. Up-regulation of interleukin-10 (IL-10) occurs in early stellate cell activation. The anti-inammatory effects of this cytokine are demonstrated by its ability to down-regulate TNF-a production from macrophages. Knockout mice lacking IL-10 have more severe hepatic brosis following CCl4 administration, and transgenic mice expressing IL-10 in liver have reduced brosis.107 Based on the consistent antibrotic effect of IL-10 in experimental liver disease, a clinical trial was undertaken which failed to show an antibrotic effect in patients with HCV infection, possibly because of marked increases in HCV replication109 (see Therapy of hepatic brosis, below).

The increasing prevalence of obesity in the US and western Europe is associated with an alarming increase in NASH,39 leading to advanced brosis and cirrhosis. Leptin, a circulating adipogenic hormone that is proportionate to adipose mass in circulating blood, has been clearly linked to stellate cell brogenesis.113115 Sources are likely to be both endocrine and autocrine, associated with enhanced signaling through the leptin receptor, which is up-regulated during stellate cell activation.113 Concurrently, down-regulation of adiponectin, a counterregulatory hormone, in obesity may amplify the brogenic activity of leptin. This possibility is supported by ndings in mice lacking adiponectin, which have enhanced brosis following toxic liver injury.116

RESOLUTION OF LIVER FIBROSIS AND THE FATE OF ACTIVATED STELLATE CELLS


During recovery from acute human and experimental liver injury the number of activated stellate cells decreases as tissue integrity is restored. Either reversion of stellate cell activation, or selective clearance of activated stellate cells by apoptosis, may explain the loss of activated cells in resolving liver injury. To date, evidence is strongest for stellate cell apoptosis in this setting. Apoptosis of stellate cells probably accounts for the decrease of activated stellate cells during resolution of hepatic brosis.49 Following injury, apoptosis may be inhibited by soluble factors and matrix components that are present during injury, whereas an apoptotic pathway otherwise represents a default mode. Furthermore, cell death ligands, including TRAIL and fas, are expressed in liver injury, and activated stellate cells are more susceptible to TRAILmediated apoptosis.73,117,118 Another death receptor, nerve growth factor receptor (NGFR), is also expressed by activated stellate cells, and its stimulation with ligand drives apoptosis.119 Survival factors also regulate the net activity of stellate cell apoptosis. IGF-I promotes stellate cell survival via the PI3-K/c-Akt pathway and TNF-a has the same effect, but utilizes the NF-kB pathway.120,121 Molecules regulating matrix degradation appear closely linked to survival and apoptosis. Active MMP2 correlates closely with apoptosis, and in fact may be stimulated by it.122 Inhibition of MMP2 activity by TIMP-1 blocks apoptosis in response to a number of apoptotic stimuli.123 Interactions between stellate cells and the surrounding matrix also inuence their propensity towards apoptosis, and this might partly explain the antiapoptotic activity of TIMP-1. Moreover, the brotic matrix may provide important survival signals to activated stellate cells. For example, animals expressing a mutant collagen I resistant to degradation have more sustained brosis and less stellate cell apoptosis following liver injury,48 and transgenic animals expressing TIMP-1 in liver have delayed resolution of brosis.124 Studies using gliotoxin,125 a fungal toxin that induces apoptosis in stellate cells, emphasize the role of this pathway in stellate cell removal during resolution of liver brosis. It is unknown whether an activated stellate cell can revert to a quiescent state in vivo, although it has been observed in culture. When stellate cells are grown on a basement membrane substratum (Matrigel) they remain quiescent, and plating of highly activated cells on this substratum down-regulates stellate cell activation.58,126

DISEASE-SPECIFIC MECHANISMS REGULATING HEPATIC FIBROSIS HCV AND NASH


In addition to generic mechanisms of brogenesis common to all experimental and human liver disease, there has been progress in elucidating disease-specic mechanisms, in particular in hepatitis C (HCV) and NASH (non-alcoholic steatohepatitis). In HCV, stellate cells might be infectable by the virus because they express putative HCV receptors.104,110 Moreover, adenoviral transduction of HCV non-structural and core proteins induces stellate cell proliferation and the release of inammatory signals.104 In HCV-infected liver chemokines and their receptors are up-regulated, stimulating lymphocyte recruitment.111 HCV proteins may also interact directly with sinusoidal endothelium.112

95

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 96

Section I. Pathophysiology of the Liver

METHODS TO MEASURE FIBROSIS


OVERVIEW
Measurement of brosis not only helps to stage the severity of disease, it allows serial determination of disease progression. The level of brosis may play an important role in clinical management and determine patients prognosis. For example, aggressive therapy is more appropriate in HCV-infected patients with advanced brosis. Further, the brosis progression rate is an important predictor of the time to develop cirrhosis.18 It is essential to measure brosis accurately, given the growing prospect of antibrotic therapies and the need to track their efcacy. Moreover, with growing evidence that brosis is reversible, methods will need to assess both progression and regression accurately. For example, specic therapy leads to a reduction in brosis in a number of diseases, including autoimmune liver disease, hepatitis C, hepatitis B, and others.34,35,42,44,127129 Percutaneous liver biopsy has traditionally been considered to be the gold standard test to assess liver brosis. However, a variety of non-invasive tests have been advanced as potential alternatives to biopsy. These include clinical signs, routine laboratory tests, quantitative assays of liver function, markers of extracellular matrix synthesis and/or degradation, and radiologic imaging studies. In addition to individual indicators of brosis, combination tests, and a number of models for predicting liver brosis have been developed. Individual and combination tests are discussed below. The ideal method to measure brosis would be simple, noninvasive, reproducible, inexpensive, accurate, and readily available. Unfortunately, none of the currently available approaches fullls all of these criteria.

Another major category of test includes those that are based specically on the pathogenesis of brosis (see above). For example, proteins that are produced as a result of the brogenic process itself that have been studied as markers of brosis include procollagen I, bronectin, tenascin, laminin, hyaluronic acid and others. Other markers have included cytokines (i.e. TGF-b1), connective tissue growth factor (CTGF), PDGF and others, matrix degrading enzymes (i.e. TIMP1), and others (Table 6-1). Finally, groups of tests, including those that utilize markers of brosis in combination with each other or in combination with other types of test, have been advanced in an attempt to detect and measure brosis. Ideally, a blood-based test should have both high sensitivity and high specicity. Many of the available tests have a high specicity (>95%) for advanced brosis. However, few (including algorithms) have great sensitivity to detect moderate levels of brosis. Moreover, a serum-based assay ideally should be linear over the full range of brosis, follow the natural history, and accurately reect the effect of treatment.

Routine Laboratory Tests


A number of studies have used routine laboratory tests in an attempt to determine whether a patient may have advanced liver disease, in particular to exclude or conrm portal hypertension and/or esophageal varices.130,131 Although tests such as the prothrombin time, albumin level, and portal vein diameter (measured by ultrasound) have all been associated with varices, studies have been remarkably consistent in their identication of the platelet count as

Table 6-1. Cytokines, Growth Factors, Peptides, Proteases, and other Components Important in Hepatic Fibrogenesis Cytokines Transforming growth factor-b Transforming growth factor-a Interleukin-1 Interleukin-4 *Interleukin-6 Interleukin-10 Interleukin-13 *Monocyte chemotactic factor Growth factors Transforming growth factor-b Transforming growth factor-a *Insulin-like growth factor (I, II) *Platelet-derived growth factor *Fibroblast growth factor Vascular endothelial growth factor Hepatocyte growth factor Connective tissue growth factor Peptides Endothelin-1 Norepinephrine Angiotensin II

BEDSIDE DIAGNOSTIC TOOLS


Clinical signs and symptoms of liver disease are frequently highlighted in assessing patients with liver disease, but these are of little value in detecting early, precirrhotic stages of liver brosis. In contrast, a number of clinical features can be utilized to assess whether cirrhosis with portal hypertension may be present. Signs of cirrhosis include spider angiomata, distension of abdominal wall veins, ascites, splenomegaly, muscle wasting, Dupuytrens contractures (especially with ethanol-associated cirrhosis), gynecomastia and testicular atrophy in males, and palmar erythema. However, it is important to emphasize that even in patients with histologic cirrhosis, and in those with portal hypertension, these physical signs may not be present.

NON-INVASIVE MARKERS OF FIBROSIS Blood-Based Markers Overview


A wide variety of blood, serum, or plasma markers for brosis have been proposed. There are several categories of marker or test. For example, some detect abnormalities in serum chemistries. Included in these types of test are aspartate aminotransferase (AST), alanine aminotransferase (ALT), g-glutamyl transferase (GGT), bilirubin, albumin, and a2-macroglobulin, among others. Moreover, some of these individual tests have been incorporated into simple and/or complex mathematical models or algorithms (see below).

Proteases and their inhibitors Matrix-metalloproteinase-1 (interstitial collagenase) Matrix-metalloproteinase-2 (gelatinase A) Matrix-metalloproteinase-3 (stromelysin-1) Matrix-metalloproteinase-7 (matrilysin) Matrix-metalloproteinase-8 Matrix-metalloproteinase-9 (gelatinase B) Matrix-metalloproteinase-10 (stromelysin-2) Tissue inhibitor of metalloproteinase-1

Miscellaneous Thrombospondin (1,2) Leptin Activin A *Thrombin Osteopontin

Agents may have direct effects on hepatic stellate cells, or indirect effects in the wounding environment. *Compounds whose effect is largely via stimulation of proliferation.

96

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 97

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

the best single predictor of esophageal varices. For example, in one study, cirrhotics without splenomegaly on physical examination and with a platelet count >88 000/mm3 had a risk of large esophageal varices of 7.2%, whereas the risk was 28% if the platelet count was less than 88 000/mm3.130 An AST/ALT ratio >1 has been proposed to indicate the presence of cirrhosis.132 In one study of patients with HCV, a ratio >1 had 100% specicity and positive predictive value for distinguishing cirrhotic from non-cirrhotic patients, with a 53.2% sensitivity and 80.7% negative predictive value.133 In addition, the ratio correlated positively with the stage of brosis, but not with the grade of activity or other biochemical indices. Of cirrhotic patients, 17% had no clinical or biochemical evidence of chronic liver disease except for an elevated AST/ALT ratio. In another study, the AST/ALT ratio had 81.3% sensitivity and 55.3% specicity in identifying cirrhotic patients who died within 1 year of follow-up.132,134 In a further attempt to develop non-invasive tools for the measurement of liver brosis, Forns and coworkers developed a model using data from HCV patients that included age, GGT, cholesterol, and platelet count.135 This model was developed with the intention to differentiate patients with signicant brosis from those without. The sensitivity for detecting METAVIR F2F4 brosis was 94%, and the presence of signicant F2F4 brosis could be excluded with high accuracy (negative predictive value of 96%).135 Likewise, Wai et al.136 constructed a simple model utilizing routine laboratory data (Table 6-2). The authors devised a novel index, termed the AST to platelet ratio index, or APRI, which is the AST level/upper limit of normal (ULN) divided by the platelet count (109/l) multiplied by 100. The sensitivity and specicity for brosis of the APRI value depended on the cut-offs used. Using an APRI value of 1.50, the positive and negative predictive values for significant brosis (Ishak score = 3) were 91% and 65%, respectively, whereas for cirrhosis and an APRI of 2.00, the positive and negative predictive values were 65% and 95%, respectively. Thus for a hypothetical patient, if the AST was 90 IU/l (and the ULN 45) and platelet count was 100 109/l, then the APRI would be 2.00. This means that the patient has essentially a 90% chance of having signicant brosis, and somewhat less likelihood of having cirrhosis. However, cirrhosis could not be excluded with certainty. Although the APRI is attractive because of its simplicity, it can neither denitively diagnose nor exclude cirrhosis, and it will not identify patients with early brosis. Other simple quantitative systems based on routine laboratory values have been developed. One early example was the PGA index, which combined prothrombin time, GGT and apolipoprotein A1 (Table 6-2); this test was examined in patients with alcoholic cirrhosis.137 The diagnostic accuracy of this index was later improved by the addition of a2-macroglobulin (and hence termed the PGAA index).138 The test characteristics of many of these indirect assays have been derived from datasets, but have not been validated on independent datasets. More complicated algorithms based on commonly available laboratory tests include the Fibrotest, reported by the French MULTIVIRC group.25 This group used mathematical modeling to develop an algorithm including ve different markers to predict brosis (the markers selected were a2-macroglobulin, haptoglobin, GGT, apolipoprotein A1, and total bilirubin). This index predicted a spe-

Table 6-2. Combined Panels of Blood Markers used to Detect Liver Fibrosis Panel AST/ALT Forns APRI PGA index Fibrotest Fibrospect *ELF FPI Components AST/ALT Platelets, GGT, cholesterol AST, Platelets Platelets, GGT, apolipoprotein A GGT, haptoglobin, bilirubin, apolipoprotein A, a2-macroglobulin Hyaluronic acid, TIMP-1, a2-macroglobulin ECM proteins AST, cholesterol, HOMA-IR References 132134 135 136 137138 25, 139142 146 147

ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, g-glutamyl transpeptidase; APRI, AST to platelet ratio index; TIMP-1, tissue inhibitor of metalloproteinase 1; ECM, extracellular matrix; ELF, European liver brosis; FPI, brosis probability index; HOMA-IR, insulin resistance by the homeostasis model assessment. Also includes age in the panel. *Components tested include collagen IV, collagen VI, amino terminal propeptide of type III collagen (PIIINP), matrix metalloproteinase 2 (MMP-2), matrix metalloproteinase 9 (MMP-9), tissue inhibitor of matrix metalloproteinase 1 (TIMP-1), tenascin, laminin, and hyaluronic acid (HA).

cic biopsy category in 46% of patients139 and has been validated in a number of hepatitis C patient cohorts, having been found to have an area under the receiver operator characteristic (AUROC) curve of 0.730.87.140 The addition of ALT to the marker panel allows for prediction of METAVIR necroinammatory activity.140 The panel has also been examined in other liver disease cohorts.141,142 Limitations of this panel in brosis include false positive results due to increases in bilirubin or decreases in haptoglobin, for example from hemolysis secondary to ribavirin therapy. Likewise, false positive results may also occur in situations where there is hyperbilirubinemia, such as Gilberts disease and cholestasis. Acute inammation may also affect the results of the test owing to changes in a2macroglobulin or increases in haptoglobin. Currently, it is unclear whether the brotest assay meets sufciently rigorous criteria, given a predictive value of only 46%, for routine clinical use.

Tests Using Extracellular Matrix/Fibrosis Markers


Analyses of serum markers of extracellular matrix/brosis include many proteins important in brogenesis, ECM constituents (i.e. bronectin, collagen I, collagen IV, collagen VI, amino terminal propeptide of type III collagen (PIIINP), tenascin, and hyaluronic acid, metalloproteinases (including many of those listed in Table 6-1), inhibitors of matrix metalloproteinases (i.e. TIMP-1, TIMP-2), and other proteins, peptides, and cytokines, as highlighted in Table 6-1. Although many tests have been studied individually, they are generally not sensitive for detection of brosis143,144 (see 145 for review).

Tests Using Combinations of Extracellular Matrix and/or Routine Markers


A combination test including hyaluronic acid, TIMP1, and a2macroglobulin was examined in a cohort of 294 patients with HCV infection and subsequently validated in a second cohort of 402 patients146 (Fibrospect, Table 6-2). This had a combined AUROC of 0.831 for METAVIR F2F4 brosis. The positive and negative predictive values were 74.3% and 75.8%, respectively, with an accuracy of 75%. This three-marker panel thus may help differentiate

97

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 98

Section I. Pathophysiology of the Liver

patients with HCV infection with moderate/severe brosis from those with no/mild brosis, although it was not possible to differentiate specic stages accurately. Another combination test was developed by the European Liver Fibrosis (ELF) Study Group.147 This group examined collagen IV, collagen VI, PIIINP , matrix metalloproteinase 2 (MMP-2), matrix metalloproteinase 9 (MMP-9), TIMP-1, tenascin, laminin, and hyaluronic acid (HA). The study was unique in that it examined patients with a wide variety of liver diseases, including those with chronic hepatitis C virus infection (n = 496), alcoholic liver disease (n = 64), non-alcoholic fatty liver disease (n = 61), chronic hepatitis B virus infection (n = 61), primary biliary cirrhosis or primary sclerosing cholangitis (n = 53), recurrent liver disease post orthotopic liver transplantation (n = 48), autoimmune hepatitis (n = 45), hemochromatosis (n = 32), cryptogenic cirrhosis (n = 19), both hepatitis B and C (n = 4), and other or no known diagnosis (n = 138); the cohort also had a wide distribution of brosis stages (Scheuer brosis stages were as follows: stage 0 = 24.6%; stage 1 = 35.5%; stage 2 = 13.4%; stage 3 = 14.9%; and stage 4 = 11.8%). An algorithm was developed that detected the upper third of brosis groups (Scheuer stages 2, 3, and 4) with a sensitivity of 90% and accurately detected the absence of brosis (Scheuer stages 0, 1), with a negative predictive value for this level of brosis of 92%. The AUC of a receiver operating characteristic (ROC) plot was 0.804. Interestingly, the addition of clinical chemistry tests including liver function tests, or hematological indices including platelet count and prothrombin time, did not improve test performance. The test appeared to be best in patients with hepatitis C, non-alcoholic fatty liver disease and alcoholic liver disease. The inclusion of patients with multiple etiologies of liver disease, although appealing, has the potential to limit the accuracy of these and other panels, as the characteristics of specic assays may be disease specic. Another model, including AST, cholesterol, and insulin resistance (as well as age and an estimate of past alcohol intake) in patients with HCV147a found that the sensitivity for detection of advanced brosis depended on the index value used. At a low probability index, the sensitivity for predicting signicant brosis was high, but specicity was low, while at a high probability index, sensitivity for signicant brosis was low, but specicity was high.

this study, patients with advanced brosis had elevated levels of a2macroglobulin, haptoglobin, and albumin, but apolipoprotein AI, apolipoprotein A-IV, complement C4, and serum retinol-binding protein were reduced. Another approach has included measurement of labeled Nglycans found in serum.151 The technique exploits the ability to analyze the desialylated total serum N-glycome on a DNA analyzer. The authors focused on cirrhosis (primarily ethanol induced), demonstrating unique patterns of serum N-glycans in those with cirrhosis compared to those with chronic liver disease alone. It was postulated that in cirrhotic livers characteristic N-glycans with a bisecting GlcNAc residue were prominent. In normal liver, the enzyme responsible for this modication, N-acetylglucosaminyl transferase III (GnT-III), is found only in non-parenchymal cells, but in regenerating liver (two-thirds partial hepatectomy) this enzyme is produced in hepatocytes. Thus, GnT-III expression is presumably a manifestation of hepatocellular regeneration, reected by regenerative nodules. This approach was most sensitive for the detection of cirrhosis and was also able to exclude cirrhosis with great accuracy. When combined with the commercially available Fibrotest this test had 100% specicity and 75% sensitivity for diagnosing compensated cirrhosis.151

Summary of Blood-based Markers


A key advantage of serum markers to detect brosis is their noninvasiveness. Additionally, it has been argued that serum markers overcome sampling problems associated with liver biopsy. However, these approaches have several drawbacks. First, most of the studies examining serum markers have been performed in cohorts of patients that have been biased toward advanced brosis/cirrhosis. A further problem is that the currently proposed serum marker algorithms use dichotomous rather than continuous scales. The dichotomous nature of these variables would be less problematic if there were clear clinical associations, for example if prognosis or treatment response were highly linked to stage 01 versus stages 24. In the absence of clinical correlates between dichotomous variables and outcomes, it remains important to diagnose the different stages of brosis accurately (04). Unfortunately, current tests and algorithms are unable to do this, and perhaps most importantly, the tests do not differentiate between intermediate levels of brosis. Thus, although assessments of brosis with approaches that use serum markers have great appeal, and indeed, in some areas the tests have begun to replace liver biopsy. Further investigation is required to optimize these tests.

Proteomics
With the recent explosion in proteomics, proteomic approaches have attempted to identify unique protein ngerprints in patients with liver disease. Various platforms are available, including those that measure protein expression, proteinprotein interactions, or even enzymatic activity. The majority of approaches have used highthroughput technologies to identify novel protein expression patterns. For example, a recent study in 46 patients with chronic hepatitis B identied 30 proteomic features predictive of signicant brosis (Ishak stage = 3) and cirrhosis. The AUROC for this analysis was 0.906 and 0.921, for advanced brosis and cirrhosis, respectively.148 Another study in 193 patients with chronic hepatitis C identied eight peaks that differentiated METAVIR brosis stages with an AUROC of 0.88; this was compared to an AUROC 0.81 for the Fibrotest.149 Another report in patients with HCV brosis identied several serum proteins to be differentially regulated.150 In

Imaging Tests
A wide variety of radiographic tests have been used to image patients with brosis/cirrhosis. Included in this group are ultrasound, CT, and MRI. In general, these tests are capable of detecting evidence of portal hypertension, thus they have the ability to detect advanced disease. As currently used in clinical practice, however, they are insensitive for the detection of moderate degrees of brosis. Transient elastography, which uses pulse-echo ultrasound acquisitions to measure liver stiffness and predict brosis stage, has gained interest as a method to quantify brosis as it appears that liver stiffness may accompany the brogenic response.152 In a prospective multicenter study of 327 chronic HCV patients, the AUROCs for

98

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 99

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

METAVIR stage F2F4 and cirrhosis were 0.79 and 0.97, respectively.153 In a separate study of 183 chronic HCV patients, transient elastography compared favorably with the Fibrotest and APRI (AUROC for F2F4 = 0.83, 0.85 and 0.78, for transient transient elastography, Fibrotest and APRI, respectively).154 When transient elastography was combined with the Fibrotest, the predictive value for brosis stage F2F4 was improved, with an AUROC of 0.88.154 Transient elastography (Fibroscan) reportedly offers good reproducibility with low inter- and intraobserver variability. The procedure is performed by obtaining multiple validated measurements in each patient, further reducing the potential for sampling errors. The depth of measurement from the skin surface is between 25 and 65 mm, raising the possibility that this technique may be difcult to use in obese patients or those with ascites. However, newer probes are being developed for obese patients, and further investigation is expected. Finally, it would theoretically be desirable to utilize advances in the molecular understanding of liver brosis to image the liver. For example, the number of activated stellate cells, which reect brogenic activity, might be identied by tagging them with cell-specic markers.155 Alternatively, matrix or matrix turnover could be labeled using molecular tools. Although such approaches are appealing, they remain experimental at present.

in the other. Finally, in 10% of subjects, stage 02 disease was identied in one lobe and stage 34 brosis was found in the other. Similar variability was reported in another study in patients with fatty liver disease.161 There are several other limitations of liver biopsy. Quantication of brosis in biopsies is subject to signicant interobserver variation. In chronic hepatitis C, for example, standardized grading systems, including Knodell, METAVIR, Scheuer or Ishak, are concordant in only 7080% of samples. Specimen quality is very important, with smaller samples leading to an underestimation of disease severity.162 A recent study created digitized virtual image biopsy specimens of varying length from large liver sections, and revealed that 75% of 25-mm biopsy specimens were correctly classied using the METAVIR staging system, compared to only 65% for biopsies 15 mm long.163 Interestingly, a recent study noted that the experience of the pathologist may have more inuence on interobserver agreement than specimen length.164 Another major problem with using liver biopsy or serum markers to quantify brosis is that all of the currently utilized grading systems use a simple linear numerical scoring approach, implying that they represent linear changes in brosis content. Such an inference is highly inaccurate, as METAVIR stage 4 brosis does not represent twice as much brosis as stage 2, but rather a 520-fold difference.

Tests of Liver Function


A variety of bona de liver function tests have been used to assess liver brosis and cirrhosis. Such tests generally measure advanced disease and several depend on perfusion, such as indocyanine green, sorbitol and galactose clearance tests, or tests such as the 13C galactose breath test and the 13Caminopyrine breath test that depend on the functional capacity of the liver.156158 Another test, the MEGX test, which measures monoethylglycinexylidide (MEGX) formation after the administration of lidocaine, depends upon the activity hepatic cytochrome P450 3A4 isoenzyme (which catalyzes oxidative N-de-ethylation of lidocaine to MEGX.159 The MEGX test has a sensitivity and specicity in the 80% range for distinguishing chronic hepatitis from cirrhosis in comparison to standard liver tests.159 Unfortunately, although the MEGX test and other function tests may predict prognosis in cirrhotic patients, they are insensitive for quantifying brosis in patients with less advanced disease.156158

TREATMENT OF FIBROSIS
Specic therapy for the treatment of liver brosis is attractive because the scarring response leads to many if not all of the complications of chronic liver disease, in particular impaired synthetic function, liver failure, and perhaps hepatocellular cancer. Fibrosis, particularly in its advanced stages, may also contribute to portal hypertension, by preventing blood ow through brotic nodules. Although attempts have been made previously to treat specically the brosis component of liver disease, these approaches have generally been unsuccessful. Thus, there remains a major unmet need for novel and effective antibrotic therapy. Advances in elucidating the pathogenesis of brosis have led to renewed efforts in this area. Additionally, data indicating that brosis is reversible have helped fuel this effort (Figure 6.5). Although brosis is commonly accepted as the precursor to cirrhosis, it is not clear that mortality risk increases directly with the stage of brosis, until the patient actually becomes cirrhotic. Even with established cirrhosis, in a cohort of patients with chronic HCV infection Fattovich and colleagues demonstrated that complications of cirrhosis developed over prolonged periods, and only when complications occurred was mortality increased.7 The most effective antibrotic therapies are currently those that treat or remove the underlying stimulus to brogenesis (Table 6-3). In addition, preclinical and human clinical studies have highlighted a number of therapies that may abrogate brogenesis without affecting the underlying disease, by targeting specic steps in the brogenic response. Anti-inammatory therapies have been based on the knowledge that inammation drives the brogenic cascade. Other treatments have attempted to inhibit cellular injury or focused on stellate cell activation, whereas others have targeted collagen syn-

Liver Biopsy
Percutaneous liver biopsy has traditionally been considered to be the gold standard test to measure brosis. Although there is great experience with liver biopsy, this procedure is time consuming, inconvenient, uncomfortable, invasive, and makes both patients and physicians anxious. Further, liver biopsy can be associated with substantial sampling-error (see Chapter 12 for further details about liver biopsy). In a recent study in which 124 patients with chronic HCV infection underwent laparoscopy-guided biopsy of each the right and left hepatic lobes, 33.1% had a difference of at least one histologic stage (modied Scheuer system) between the two lobes.160 Furthermore, in 18 study subjects a stage consistent with cirrhosis was found in one lobe, whereas stage 3 brosis was reported

99

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 100

Section I. Pathophysiology of the Liver

Inset in A

Figure 6-5. Reversal of brosis. An example of reversal of advanced brosis (cirrhosis in this situation) is depicted. A liver biopsy prior to lamivudine treatment is shown (upper panel and left panel). After treatment with lamivudine, liver biopsy was repeated and reveals almost complete dissolution of brosis. Data similar to these have been published in autoimmune liver disease, hepatitis C, alcoholic hepatitis, hepatitis B, and others. (Reprinted with permission, Wanless, et al: Arch Pathol Lab Med 2001;124:15991607.)

Table 6-3. Approaches to Treat Liver Fibrosis Approach Remove injurious agent Anti-inammatory agents Antioxidants Cytoprotective agents Inhibit stellate cell activation Inhibit stellate cell activation phenotypes (brogenesis) Example Eradication of HBV Corticosteroids in AIH PPC in alcoholic hepatitis Ursodeoxycholic acid Interferon-g Colchicine

Table 6-4. Diseases and Therapies in which there is Strong Evidence that Treatment Reduces Liver Fibrosis Disease Hepatitis B Hepatitis C Bile duct obstruction Autoimmune hepatitis Hemochromatosis Alcoholic hepatitis Therapy Lamivudine Interferon-a* Surgical decompression Corticosteroids Iron depletion Corticosteroids References 3335,129 42 43 44 165,166 168,169

Note: some approaches have not been demonstrated to be successful. AIH, autoimmune hepatitis; PPC, polyenylphosphatidylcholine.

*or PEG-interferon-a, with or without ribavirin. MTX, methotrexate; PPAR, peroxisomal proliferator-activated receptor.

thesis and matrix deposition. The following section highlights human studies in these areas.

THERAPIES DIRECTED AT THE UNDERLYING DISEASE


In many forms of liver disease treatment of the underlying inciting lesion leads to an improvement in brosis (Table 6-4). For example, eradication or inhibition of HBV33,34,129 or HCV replication42 leads to reversion of brosis, even in patients with histological cirrhosis. Fibrosis reverts in patients with hemochromatosis during iron depletion,165,166 after corticosteroid therapy in autoimmune hepatitis,165,166 and in patients with secondary biliary cirrhosis after decompression of bile duct obstruction.43 In a preliminary report in patients with non-alcoholic steatohepatitis (NASH) treated with the peroxisomal proliferator active receptor (PPAR)-g agonist rosiglitazone both steatosis and brosis were reduced.167

atitis who respond to medical treatment (prednisone or equivalent) advanced brosis and cirrhosis are reversible.44 Fibrosis may improve in patients with alcoholic liver disease who respond to corticosteroids.168,169 Thus, corticosteroids appear to have antibrotic effects in patients with certain liver disorders.

Interleukin-10 (IL-10)
Interleukin (IL)-10 has both anti-inammatory and immunosuppressive effects. IL-10 has been shown to reduce the production of proinammatory cytokines, such as TNF-a, IL-1, interferon-g, and IL-2 from T cells. These cytokines belong to the Th1 family. Endogenous IL-10 reduces the intrahepatic inammatory response, shifts the cytokine milieu towards a Th2 predominance, and reduces brosis in several in vivo models of liver injury.170 It was hypothesized that in vivo administration of IL-10 in patients with hepatitis C virus infection may have an anti-inammatory and hence an antibrotic effect.109 Therefore, 30 patients with advanced HCV-mediated brosis who had failed standard interferon-a-based antiviral therapy were enrolled in a 12-month treatment trial of IL-10 given daily or thrice weekly subcutaneously. In 13 of 28 of these patients the hepatic inammation score decreased by at least two points (Ishak score) and 11 of 28 had a reduction in brosis score (mean change from 5.0 0.2 to 4.5 0.3, p < 0.05). However, serum HCV RNA levels increased during therapy (mean HCV RNA at day 0: 12.3 3.0 mEq/ml; and at 12 months: 38 mEq/ml; p < 0.05). Changes in liver histology and HCV RNA levels were accompanied by an apparent shift in toward a Th2-predominant lymphocyte phenotype, as had been originally hypothesized. Long-term therapy with

ANTI-INFLAMMATORY COMPOUNDS
Many liver diseases, such as HCV disease, have an important inammatory component. Inammation in these disorders typically drives stellate cell activation and brogenesis, and it is these diseases in particular that have been studied in order to evaluate the efcacy of anti-inammatory drugs.

Corticosteroids
Classic examples of the benets of steroids include autoimmune hepatitis and alcoholic hepatitis. In patients with autoimmune hep-

100

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 101

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

IL-10 decreased hepatic inammatory activity and appeared to have an inhibitory overall effect on brosis. Thus, although IL-10 appears to reduce inammation and brosis, it has not been pursued as an antibrotic compound because of putative detrimental virologic effects.

Miscellaneous Anti-inammatory Drugs


A number of other anti-inammatory approaches have gained attention as therapies for brosis. Because TNF-a drives inammation in many diseases, and because TNF-a is up-regulated in liver diseases (such as alcoholic hepatitis), an anti-TNF-a compound should theoretically reduce inammation and hence the stimulus for brosis.171174 Preliminary analyses from a study of anti-TNF-a therapy suggest an improvement in inammation which presumably precedes brosis in patients with alcoholic hepatitis,173 although there was little effect of anti-TNF-a on hepatic brosis. Such data, in addition to the favorable effects of the TNF-a inhibitor pentoxifylline on mortality, provide the rationale for future study in patients with alcoholic hepatitis. These approaches and others that broadly inhibit inammation must be considered cautiously because of the concern about disruption of the immune system, with increased risk of infection. Penicillamine is a heavy metal chelating compound that has been proposed to have anti-inammatory and thus antibrogenic effects.175 However, this compound had no effect on brogenesis in patients with primary biliary cirrhosis.176,177 Metrothrexate is thought to have anti-inammatory properties, but interestingly has typically been considered to be probrogenic in the liver for patients receiving methotrexate for treatment of rheumatologic diseases178 (although it is noteworthy that the risk of brosis progression may be less prominent than typically believed178,179). Metrothrexate has been studied in patients with primary biliary cirrhosis. Although some investigators have reported highly favorable effects in this disease, including improvement of the disease and reversion of brosis,180 the majority of the data on methotrexate have either been negative181,182 or show that the drugs effects have been marginal, either alone181 or in combination with colchicine.183 It is important to emphasize that if methotrexate is used to treat patients with primary biliary cirrhosis, this must be undertaken by an experienced hepatologist.

A VA cooperative multicenter clinical trial examined the effect of polyenylphosphatidylcholine in 789 patients with alcoholic hepatitis who had a very high daily average alcohol intake (16 drinks/day).185 Subjects were randomized to either polyenylphosphatidylcholine or placebo for 2 years. The long period of treatment is noteworthy as it is likely that long periods of treatment will be required to effect changes in the liver brosis. Many subjects substantially reduced their ethanol consumption during the trial, which probably accounted for improvement in brosis in the control group, making it difcult to demonstrate an improvement in brosis in the polyenylphosphatidylcholine group. Thus, overall, polyenylphosphatidylcholine failed to lead to signicant improvement in brosis.

Silymarin
Silymarin is derived from the milk thistle Silybum marianum. This extract has been shown to reduce lipid peroxidation and inhibit brogenesis in rodent animal models,186,187 as well as in baboons.188 It has been tested in several carefully performed human clinical trials, although brosis was not used as an endpoint. The compound has been found to be safe, but reportedly has mixed effects.189,190 In one study examining silymarin in alcoholics189 mortality was reduced; in addition, patients with early stages of cirrhosis also appeared to benet. However, in another study in alcoholics no survival benet could be identied.190 In both of these trials, silymarin appeared to be safe. Thus, although the agent is safe and is commonly used by patients with brosing liver disease, there is limited evidence of its efcacy.

Other Antioxidants
Antioxidants such as vitamin E have been examined in animal models191 as well as in humans.192195 A vitamin E precursor, D-atocopherol (1200 IU/day for 8 weeks), was studied in six patients with hepatitis C virus infection who failed to respond to interferon therapy,192 and was found to inhibit stellate cell activation but did not affect brosis. A randomized controlled trial examined vitamin E in patients with mild to moderate alcoholic hepatitis and found that vitamin E reduced serum hyaluronic acid, but did not lead to a change in type III collagen.194 Combined antioxidant therapy, including vitamin E, had no effect on outcome in patients with severe alcoholic hepatitis, although brosis was not specically addressed.195 Malotilate is another potential cytoprotective agent, perhaps acting via inhibition of cytochrome P450 2E1; in addition, this compound may have anti-inammatory properties. In patients with primary biliary cirrhosis, although it was found to diminish plasma cell and lymphocytic inltrate and piecemeal necrosis, it had no signicant effect on brogenesis.196 Another agent used to antagonize oxidative stress is S-adenosylmethionine; this compound is important in the synthesis of the antioxidant glutathione. The enzyme (methionine adenosyltransferase) responsible for its synthesis is reduced in the injured liver;197 thus it has been hypothesized that if S-adenosylmethionine were replaced, then injury and brogenesis might be slowed. S-adenosylmethionine has been tested in a large randomized trial in patients with alcoholic cirrhosis.198 There was an improvement in overall mortality/need for liver transplantation in the treatment arm, espe-

ANTIOXIDANT AGENTS
Oxidative stress is thought to play an important role in injury, stellate cell activation, and the stimulation of extracellular matrix production, as discussed above. Thus, a wide variety of antioxidants have received attention as potential antibrotics.

Polyenylphosphatidylcholine
Polyenylphosphatidylcholine is a mixture of polyunsaturated phosphatidylcholines, extracted from soybeans. This compound has antioxidant properties and oxidant stress (see above) is thought to be important in the inammatory and brogenic response to injury, particularly in alcoholic liver disease. As oxidative stress leads to lipid peroxidation, and lipid peroxidation is injurious at the level of the cell membrane, phosphatidylcholine has been proposed to be protective against injury to cell membranes, resulting in reduced cellular injury and brogenesis.184

101

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 102

Section I. Pathophysiology of the Liver

cially in patients with Childs A/B cirrhosis, although histologic assessment of brosis was not specically assessed.198 Propylthiouracil is an antithyroid drug that reacts with some of the oxidizing species derived from the respiratory burst and may thus be protective in alcoholic liver disease, a disease in which an increase in hepatic oxygen consumption may predispose the liver to ischemic injury. Thus, propylthiouracil has been tested in a number of randomized clinical trials in patients with alcoholic liver disease. Unfortunately, a systematic review and meta-analysis found that propylthiouracil had no benet in brosis, or in any other outcome measured.199

improve brogenesis. Additionally, a small series indicated that seven of 10 patients with cystic brosis treated with ursodeoxycholic acid had a reduction in liver brosis.204 Although these effects are promising, it should be emphasized that the numbers of patients studied has been small. Finally, in a large randomized controlled trial of ursodeoxycholic acid in patients with non-alcoholic steatohepatitis over 2 years, including 107 subjects who had paired biopsy data, there was no improvement in brosis.209

MISCELLANEOUS
Anabolicandrogenic steroids such as oxandrolone have been examined in randomized trials including patients with alcoholic liver disease, but have not been found to have signicant effects on brosis (or other outcomes).210

CYTOPROTECTIVE AGENTS Ursodeoxycholic Acid


Ursodeoxycholic acid binds to hepatocyte membranes, where it presumably stabilizes them and is thus cytoprotective. This cytoprotective action in turn theoretically reduces inammation and may in turn have a benecial effect on brogenesis.200 Neither experimental data nor human studies indicate a primary antibrotic effect of ursodeoxycholic acid in the liver, but the compound has been examined extensively.201209 Ursodeoxycholic acid has been studied in patients with cystic brosis, primary biliary injury (primary biliary cirrhosis, primary sclerosing cholangitis and progressive familial intrahepatic cholestasis), and miscellaneous liver diseases. Results with ursodeoxycholic acid in these conditions have been mixed. Both symptomatic and biochemical improvement has been observed in these diseases, in particular the biliary diseases, but data on histologic improvement (and survival) have not been consistent. For example, in a randomized controlled trial in patients with primary biliary cirrhosis, ursodeoxycholic acid led to reduced brosis in those with mild disease but had no effect on those with severe disease.202 In another study, survival was improved in patients treated with ursodeoxycholic acid but brosis was not improved.206 Further, in a histopathologic study of 54 patients with primary biliary cirrhosis and paired liver biopsies, 4 years of ursodeoxycholic acid therapy was associated with a signicant decrease in the prevalence of orid interlobular bile duct lesions, lobular inammation, and necrosis. Worsening of brosis was observed in 14 patients (the majority had only a onegrade progression in brosis score), whereas stabilization was noted in the 40 remaining patients.207 A recent combined analysis of the histologic effect of ursodeoxycholic acid on paired liver biopsies including a total of 367 patients (200 ursodeoxycholic acid and 167 placebo) revealed that subpopulations of patients with initial earlystage disease may benet from therapy.208 Results of meta-analyses examining ursodeoxycholic acid have been mixed, and have largely reported that ursodeoxycholic acid is not effective in primary biliary cirrhosis.205 The aggregate data suggest that ursodeoxycholic acid may impede the progression of brosis in primary biliary cirrhosis via effects on (bile duct) inammation, particularly if given early in the disease course. It should be emphasized that so far as we know, ursodeoxycholic acid is extremely safe. Thus, although it is also expensive, the available data justify its use as an antibrotic in patients with primary biliary cirrhosis. Ursodeoxycholic acid has also been studied in children with progressive familial intrahepatic cholestasis,203 where it appeared to

Stellate Cell-Specic Compounds Interferon-g


A wealth of data supports the antibrotic potential of interferon-g. The interferons consist of a family of three major isoforms, a, b and g. These isoforms are unique, not only in structure but also in their biologic actions. Interferons-a and -b bind to the same receptor, whereas interferon-g binds to a different receptor. Interferon-a has more potent antiviral effects than does interferon-g, and interferong has been shown to specically inhibit extracellular matrix synthesis in isolated cells, including stellate cells.211,212 Interferon-g potently inhibits multiple aspects of stellate cell activation,211,212 and appears to have antibrotic effects in patients with pulmonary brosis.213 Such data have generated considerable enthusiasm about the use of interferon-g in patients with hepatic brosis, although there is theoretical concern about its use because it is proinammatory, and moreover its overexpression in the liver leads to chronic hepatitis.214 None the less, it has now been tested in humans with brosing liver disease, and appears to be safe.215 Although this pilot study provides a rm foundation highlighting the potential use of interferon-g in patients, larger studies are needed to prove a therapeutic benet.

COMPOUNDS THAT INHIBIT FIBROGENESIS Colchicine


Colchicine is a plant alkaloid that inhibits polymerization of microtubules, a process that in turn is believed to be required for collagen secretion. Based on this concept, colchicine has been advanced as an antibrotic agent. A sizeable body of literature indicates that colchicine has antibrotic properties in experimental animal models.216 This work has led to a number of human clinical trials.217220 A wide variety of liver diseases has been studied, including primary biliary cirrhosis, alcoholic cirrhosis, cryptogenic cirrhosis, and miscellaneous other liver diseases. In a double-blind randomized controlled trial examining colchicine in primary biliary cirrhosis improvements were noted in a number of biochemical markers, but the drug failed to reduce brosis.217 In an often-cited popularized double-blind randomized controlled trial of colchicine versus placebo in patients with various liver diseases, colchicine led to improved brosis as well as a dramatic improvement in survival.218 However, this study has been extended to clinical practice with great caution because of a variety of methodological concerns. First,

102

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 103

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

Table 6-5. New Potential Antibrotic Targets in Humans Agent Anti-TGF-b Anti-PDGF Interferon-g PPAR ligands Comments Blocks stellate cell brogenesis Blocks stellate cell proliferation Inhibits multiple features of stellate cell activation ? Stellate cell-specic effects

thus could evolve into effective antibrotic compounds. Many others have been highlighted (see 5 for review).

SUMMARY AND FUTURE DIRECTIONS FOR ANTIFIBROTIC THERAPY


The explosion of information about the pathogenesis of brogenesis has spawned a eld dedicated to antibrotics focused on the activation of hepatic stellate cells. Stellate cell activation is characterized by a number of important features, including enhanced matrix synthesis and a prominent contractile phenotype, processes that each contribute to the dysfunction of the liver typically found in advanced disease. It should be emphasized that the control of activation is multifactorial, and thus several potential therapeutic interventions are possible. A further critical concept is that brosis, in particular the ECM component of brosis, is dynamic, and the accumulation of brosis may be inhibited. It is likely that brosis, including even advanced brosis, may be reversible under the appropriate conditions. Currently, effective therapy for hepatic brogenesis exists for several diseases in which the cause of the underlying disease is removed. In contrast, specic therapy directed only at the brotic lesion is not currently available; the most effective therapies will most likely be directed at the stellate cell. Additionally, approaches that regulate matrix remodeling (i.e. by enhancing matrix degradation or inhibiting factors that prevent matrix breakdown) will be attractive. Thus, multiple potential targets have been identied, and it is highly likely that candidates will emerge. The ideal antibrotic compound will be specic, effective, safe, and inexpensive.

many patients were lost to follow-up, and in addition there was substantial unexplained excess mortality in the control group (unrelated to liver disease). In a recent large VA cooperative multicenter study involving 549 patients comparing colchicine (0.6 mg orally b.i.d.) to placebo in patients with alcoholic liver disease, there was no apparent effect of active treatment on survival. Histologic data that might have provided information on the anti-inammatory effects of colchicine were not obtained.219 A meta-analysis including 1138 subjects found that colchicine had no effect on hepatic brosis or mortality.220 In summary, the data surrounding colchicine suggest that this compound is safe but likely to be ineffective.

FUTURE ANTIFIBROTICS
Given the major effort in understanding the biology of hepatic brogenesis, it is not surprising that numerous pathways have been targeted as having therapeutic potential. Many compounds have been studied in experimental models and have been shown to have antibrotic properties, including several with great potential in human liver disease (Table 6-5). Several important pathways merit discussion. One of the most important examples is the TGF-b pathway, as it plays a central role in the brogenic cascade. Several approaches to inhibit the action of TGF-g have been proposed and include the use of molecules such as decorin, the protein core component of proteoglycan, which binds and inactivates TGF-b,221 antibodies directed against TGF-b1, and soluble receptors which typically encode for sequences that bind active TGF-b and prevent it from binding to its cognate receptors. The concept has been well established experimentally; indeed, the effect of inhibition of TGF-b in animal models of liver injury and brogenesis has been striking.222,223 A limitation of approaches that target TGF-b is that the cytokine potently inhibits cellular proliferation, and inhibition of its effects in vivo could predispose to malignant transformation. Another critical pathway involves PDGF. PDGF is the most potent stellate cell mitogen known,3,224 and in addition stimulates stellate cell migration.225 A number of approaches have been used to inhibit the effect of PDGF. For example, kinase inhibitors that specically inhibit PDGF signaling might be useful,226 as could those with more general effects on tyrosine kinase receptors. Additionally, stellate cells express angiotensin and endothelin receptors and their cognate ligands appear to be overproduced in the liver; further, stimulation of stellate cells with their respective ligands leads to stellate cell activation.66 Thus, inhibition of their binding may be clinically benecial. Among others agents, compounds such as pirfenidone,227 peroxisomal proliferator-activated receptor (PPAR)-g ligands,76,228 and halofuginone229 appear to have direct effects on stellate cells and

REFERENCES
1. Kim WR, Brown RS Jr, Terrault NA, El-Serag H. Burden of liver disease in the United States: summary of a workshop. Hepatology 2002;36:227242. 2. Gressner AM, Weiskirchen R, Breitkopf K, Dooley S. Roles of TGF-beta in hepatic brosis. Front Biosci 2002;7:D793807. 3. Pinzani M. PDGF and signal transduction in hepatic stellate cells. Front Biosci 2002;7:d17201726. 4. Friedman SL. Liver brosis from bench to bedside. J Hepatol 2003;38(Suppl 1):S3853. 5. Rockey DC. Antibrotic therapy in chronic liver disease. Clin Gastroenterol Hepatol 2005;3:95107. 6. Bataller R, Brenner DA. Liver brosis. J Clin Invest 2005;115:209218. 7. Fattovich G, Giustina G, Degos F, et al. Morbidity and mortality in compensated cirrhosis type C: a retrospective follow-up study of 384 patients. Gastroenterology 1997;112:463472. 8. El-Serag HB. Hepatocellular carcinoma and hepatitis C in the United States. Hepatology 2002;36(Suppl 1):S7483. 9. Bruix J, Boix L, Sala M, Llovet JM. Focus on hepatocellular cancer. Cancer Cell 2004;5:215219. 10. Schuppan D, Ruehl M, Somasundaram R, Hahn EG. Matrix as modulator of stellate cell and hepatic brogenesis. Semin Liver Dis 2001;21:351372. 11. Jarnagin WR, Rockey DC, Koteliansky VE, et al. Expression of variant bronectins in wound healing: cellular source and

103

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 104

Section I. Pathophysiology of the Liver

12.

13.

14.

15.

16.

17.

18.

19.

20. 21.

22.

23.

24.

25.

26.

27. 28.

29.

30.

31.

32.

biological activity of the EIIIA segment in rat hepatic brogenesis. J Cell Biol 1994;127:20372048. Bedossa P , Ferlicot S, Paradis V, et al. Dystroglycan expression in hepatic stellate cells: role in liver brosis. Lab Invest 2002;82:10531061. Schiano TD, Kim-Schluger L, Gondolesi G, Miller CM. Adult living donor liver transplantation: the hepatologists perspective. Hepatology 2001;33:39. Benhamou Y, Di Martino V, Bochet M, et al. Factors affecting liver brosis in human immunodeciency virus- and hepatitis C virus-coinfected patients: impact of protease inhibitor therapy. Hepatology 2001;34:283287. Farci P , Roskams T, Chessa L, et al. Long-term benet of interferon alpha therapy of chronic hepatitis D: regression of advanced hepatic brosis. Gastroenterology 2004;126:17401749. Wiesner RH, McDiarmid SV, Kamath PS, et al. MELD and PELD: application of survival models to liver allocation. Liver Transpl 2001;7:567580. Kamath PS, Wiesner RH, Malinchoc M, et al. A model to predict survival in patients with end-stage liver disease. Hepatology 2001;33:464470. Poynard T, Ratziu V, Charlotte F, et al. Rates and risk factors of liver brosis progression in patients with chronic hepatitis C. J Hepatol 2001;34:730739. Powell EE, Edwards-Smith CJ, Hay JL, et al. Host genetic factors inuence disease progression in chronic hepatitis C. Hepatology 2000;31:828833. Huang H, et al. Hepatology 2004;40:230A. Shi Z, Wakil AE, Rockey DC. Strain-specic differences in mouse hepatic wound healing are mediated by divergent T helper cytokine responses. Proc Natl Acad Sci USA 1997;94:1066310668. Hillebrandt S, Goos C, Matern S, Lammert F. Genome-wide analysis of hepatic brosis in inbred mice identies the susceptibility locus Hb1 on chromosome 15. Gastroenterology 2002;123:20412051. Monto A, Patel K, Bostrom A, et al. Risks of a range of alcohol intake on hepatitis C-related brosis. Hepatology 2004;39: 826834. Patton HM, Patel K, Behling C, et al. The impact of steatosis on disease progression and early and sustained treatment response in chronic hepatitis C patients. J Hepatol 2004;40:484490. Poynard T, Bedossa P , Opolon P . Natural history of liver brosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 1997;349:825832. de Torres M, Poynard T. Risk factors for liver brosis progression in patients with chronic hepatitis C. Ann Hepatol 2003;2:511. Wright TL. Treatment of patients with hepatitis C and cirrhosis. Hepatology 2002;36(Suppl 1):S185194. Arenas JI, Vargas HE. Hepatitis C virus antiviral therapy in patients with cirrhosis. Gastroenterol Clin North Am 2004;33:549562, ix. Lindh M, Horal P , Dhillon AP , Norkrans G. Hepatitis B virus DNA levels, precore mutations, genotypes and histological activity in chronic hepatitis B. J Viral Hepatol 2000;7:258267. Merican I, Guan R, Amarapuka D, et al. Chronic hepatitis B virus infection in Asian countries. J Gastroenterol Hepatol 2000;15:13561361. Kobayashi M, Arase Y, Ikeda K, et al. Clinical characteristics of patients infected with hepatitis B virus genotypes A, B, and C. J Gastroenterol 2002;37:3539. Chen CH, Chen PJ, Chu JS, et al. Fibrosing cholestatic hepatitis in a hepatitis B surface antigen carrier after renal transplantation. Gastroenterology 1994;107:15141518.

33. Lai CL, Chien RN, Leung NW, et al. A one-year trial of lamivudine for chronic hepatitis B. Asia Hepatitis Lamivudine Study Group [see comments]. N Engl J Med 1998;339:6168. 34. Hadziyannis SJ, Tassopoulos NC, Heathcote EJ, et al. Adefovir dipivoxil for the treatment of hepatitis B e antigen-negative chronic hepatitis B. N Engl J Med 2003;348:800807. 35. Liaw YF, Sung JJ, Chow WC, et al. Lamivudine for patients with chronic hepatitis B and advanced liver disease. N Engl J Med 2004;351:15211531. 36. Raynard B, Balian A, Fallik D, et al. Risk factors of brosis in alcohol-induced liver disease. Hepatology 2002;35:635638. 37. Falck-Ytter Y, Younossi ZM, Marchesini G, McCullough AJ. Clinical features and natural history of nonalcoholic steatosis syndromes. Semin Liver Dis 2001;21:1726. 38. Brunt EM. Nonalcoholic steatohepatitis: denition and pathology. Semin Liver Dis 2001;21:316. 39. Angulo P . Nonalcoholic fatty liver disease. N Engl J Med 2002;346:12211231. 40. Ratziu V, Giral P , Charlotte F, et al. Liver brosis in overweight patients. Gastroenterology 2000;118:11171123. 41. Kweon YO, Goodman ZD, Dienstag JL, et al. Decreasing brogenesis: an immunohistochemical study of paired liver biopsies following lamivudine therapy for chronic hepatitis B. J Hepatol 2001;35:749755. 42. Poynard T, McHutchison J, Manns M, et al. Impact of pegylated interferon alfa-2b and ribavirin on liver brosis in patients with chronic hepatitis C. Gastroenterology 2002;122:13031313. 43. Hammel P , Couvelard A, OToole D, et al. Regression of liver brosis after biliary drainage in patients with chronic pancreatitis and stenosis of the common bile duct. N Engl J Med 2001;344:418423. 44. Dufour JF, DeLellis R, Kaplan MM. Reversibility of hepatic brosis in autoimmune hepatitis. Ann Intern Med 1997;127:981985. 45. Iredale JP , Benyon RC, Pickering J, et al. Mechanisms of spontaneous resolution of rat liver brosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest 1998;102:538549. 46. Shiratori Y, Imazeki F, Moriyama M, et al. Histologic improvement of brosis in patients with hepatitis C who have sustained response to interferon therapy. Ann Intern Med 2000;132:517524. 47. Issa R, Zhou X, Constandinou CM, et al. Spontaneous recovery from micronodular cirrhosis: Evidence for incomplete resolution associated with matrix cross-linking. Gastroenterology 2004;126:17951808. 48. Issa R, Williams E, Trim N, et al. Apoptosis of hepatic stellate cells: involvement in resolution of biliary brosis and regulation by soluble growth factors. Gut 2001;48:548557. 49. Iredale JP . Stellate cell behavior during resolution of liver injury. Semin Liver Dis 2001;21:427436. 50. Arthur MJ. Fibrogenesis II. Metalloproteinases and their inhibitors in liver brosis. Am J Physiol Gastrointest Liver Physiol 2000;279:G245249. 51. Schaefer B, Rivas-Estilla AM, Meraz-Cruz N, et al. Reciprocal modulation of matrix metalloproteinase-13 and type I collagen genes in rat hepatic stellate cells. Am J Pathol 2003;162:17711780. 52. Han YP , Zhou L, Wang J, et al. Essential role of matrix metalloproteinases in interleukin-1-induced myobroblastic activation of hepatic stellate cell in collagen. J Biol Chem 2004;279:48204828. 53. Benyon D, Arthur MJP . Extracellular matrix degradation and the role of stellate cells. Semin Liver Dis 2001;21:373384. 54. Friedman SL. Molecular regulation of hepatic brosis, an integrated cellular response to tissue injury. J Biol Chem 2000;275:22472250.

104

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 105

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

54a. Friedman SL, Arthur MJD. Reversing hepatic brosis. Science and Medicine 2002;8:194205. 55. Tsukamoto H. Redox regulation of cytokine expression in Kupffer cells. Antioxid Redox Signal 2002;4:741748. 56. Vogel W, Gish GD, Alves F, Pawson T. The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell 1997;1:1323. 57. Ikeda K, Wang LH, Torres R, et al. Discoidin domain receptor 2 interacts with Src and Shc following its activation by type I collagen. J Biol Chem 2002;277:1920619212. 58. Olaso E, Ikeda K, Eng FJ, et al. DDR2 receptor promotes MMP-2-mediated proliferation and invasion by hepatic stellate cells. J Clin Invest 2001;108:13691378. 59. Pinzani M, Marra F. Cytokine receptors and signaling in hepatic stellate cells. Semin Liver Dis 2001;21:397416. 60. Racine-Samson L, Rockey DC, Bissell DM. The role of alpha1beta1 integrin in wound contraction. A quantitative analysis of liver myobroblasts in vivo and in primary culture. J Biol Chem 1997;272:3091130917. 61. Gao R, Brigstock DR. Connective tissue growth factor (CCN2) induces adhesion of rat activated hepatic stellate cells by binding of its C-terminal domain to integrin alpha(v)beta(3) and heparan sulfate proteoglycan. J Biol Chem 2004;279:88488855. 62. Mazzocca A, Carloni V, Sciammetta S, et al. Expression of transmembrane 4 superfamily (TM4SF) proteins and their role in hepatic stellate cell motility and wound healing migration. J Hepatol 2002;37:322330. 63. Geerts A. History, heterogeneity, developmental biology, and functions of quiescent hepatic stellate cells. Semin Liver Dis 2001;21:311335. 64. Cassiman D, Libbrecht L, Desmet V, et al. Hepatic stellate cell/myobroblast subpopulations in brotic human and rat livers. J Hepatol 2002;36:200209. 65. Corpechot C, Barbu V, Wendum D, et al. Hypoxia-induced VEGF and collagen I expressions are associated with angiogenesis and brogenesis in experimental cirrhosis. Hepatology 2002;35:10101021. 66. Rockey DC. Vascular mediators in the injured liver. Hepatology 2003;37:412. 67. Forbes SJ, Russo FP , Rey V, et al. A signicant proportion of myobroblasts are of bone marrow origin in human liver brosis. Gastroenterology 2004;126:955963. 68. Magness ST, Bataller R, Yang L, Brenner DA. A dual reporter gene transgenic mouse demonstrates heterogeneity in hepatic brogenic cell populations. Hepatology 2004;40: 11511159. 69. Kalluri R, Neilson EG. Epithelialmesenchymal transition and its implications for brosis. J Clin Invest 2003;112: 17761784. 70. Tuma DJ. Role of malondialdehydeacetaldehyde adducts in liver injury. Free Radic Biol Med 2002;32:303308. 71. Bataller R, Schwabe RF, Choi YH, et al. NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic brosis. J Clin Invest 2003;112:13831394. 72. Canbay A, Taimr P , Torok N, et al. Apoptotic body engulfment by a human stellate cell line is probrogenic. Lab Invest 2003;83:655663. 73. Canbay A, Higuchi H, Bronk SF, et al. Fas enhances brogenesis in the bile duct ligated mouse: a link between apoptosis and brosis. Gastroenterology 2002;123:13231330. 74. Olaso E, Salado C, Egilegor E, et al. Proangiogenic role of tumor-activated hepatic stellate cells in experimental melanoma metastasis. Hepatology 2003;37:674685. 75. Mann DA, Smart DE. Transcriptional regulation of hepatic stellate cell activation. Gut 2002;50:891896. 76. Marra F, Efsen E, Romanelli RG, et al. Ligands of peroxisome proliferator-activated receptor gamma modulate probrogenic

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89. 90. 91.

92.

93.

94.

and proinammatory actions in hepatic stellate cells. Gastroenterology 2000;119:466478. Hellemans K, Verbuyst P , Quartier E, et al. Differential modulation of rat hepatic stellate phenotype by natural and synthetic retinoids. Hepatology 2004;39:97108. Paik YH, Schwabe RF, Bataller R, et al. Toll-like receptor 4 mediates inammatory signaling by bacterial lipopolysaccharide in human hepatic stellate cells. Hepatology 2003;37:10431055. Rippe RA, Schrum LW, Stefanovic B, et al. NF-kappaB inhibits expression of the alpha1(I) collagen gene. DNA Cell Biol 1999;18:751761. Ratziu V, Lalazar A, Wong L, et al. Zf9, a Kruppel-like transcription factor up-regulated in vivo during early hepatic brosis. Proc Natl Acad Sci USA 1998;95:95009505. Kalinichenko VV, Bhattacharyya D, Zhou Y, et al. Foxf1 +/ mice exhibit defective stellate cell activation and abnormal liver regeneration following CCl4 injury. Hepatology 2003;37:107117. Weiskirchen R, Moser M, Weiskirchen S, et al. LIM-domain protein cysteine- and glycine-rich protein 2 (CRP2) is a novel marker of hepatic stellate cells and binding partner of the protein inhibitor of activated STAT1. Biochem J 2001;359(Pt 3):485496. Rockey DC, Fouassier L, Chung JJ, et al. Cellular localization of endothelin-1 and increased production in liver injury in the rat: potential for autocrine and paracrine effects on stellate cells. Hepatology 1998;27:472480. Pinzani M, Milani S, De Franco R, et al. Endothelin 1 is overexpressed in human cirrhotic liver and exerts multiple effects on activated hepatic stellate cells. Gastroenterology 1996;110:534548. Marra F, Grandaliano G, Valente AJ, Abboud HE. Thrombin stimulates proliferation of liver fat-storing cells and expression of monocyte chemotactic protein-1: potential role in liver injury. Hepatology 1995;22:780787. Rosenbaum J, Blazejewski S, Preaux AM, et al. Fibroblast growth factor 2 and transforming growth factor beta 1 interactions in human liver myobroblasts. Gastroenterology 1995;109:19861996. Skrtic S, Wallenius V, Ekberg S, et al. Insulin-like growth factors stimulate expression of hepatocyte growth factor but not transforming growth factor beta1 in cultured hepatic stellate cells. Endocrinology 1997;138:46834689. Pinzani M, Abboud HE, Aron DC. Secretion of insulin-like growth factor-I and binding proteins by rat liver fat-storing cells: regulatory role of platelet-derived growth factor. Endocrinology 1990;127:23432349. Pinzani M, Marra F, Carloni V. Signal transduction in hepatic stellate cells. Liver 1998;18:213. Friedman SL. Cytokines and brogenesis. Semin Liver Dis 1999;19:129140. Reinehr RM, Kubitz R, Peters-Regehr T, et al. Activation of rat hepatic stellate cells in culture is associated with increased sensitivity to endothelin 1. Hepatology 1998;28:15661577. Schnabl B, Kweon YO, Frederick JP , et al. The role of Smad3 in mediating mouse hepatic stellate cell activation. Hepatology 2001;34:89100. Dooley S, Delvoux B, Lahme B, et al. Modulation of transforming growth factor beta response and signaling during transdifferentiation of rat hepatic stellate cells to myobroblasts. Hepatology 2000;31:10941106. Liu C, Gaca MD, Swenson ES, et al. Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem 2003;278: 1172111728.

105

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 106

Section I. Pathophysiology of the Liver

95. Wells RG. Fibrogenesis. V. TGF-beta signaling pathways. Am J Physiol Gastrointest Liver Physiol 2000;279:G845850. 96. Wells RG, Kruglov E, Dranoff JA. Autocrine release of TGFbeta by portal broblasts regulates cell growth. FEBS Lett 2004;559:107110. 97. Marra F. Chemokines in liver inammation and brosis. Front Biosci 2002;7:d1899914. 98. Ohata M, Yamauchi M, Takeda K, et al. RAR and RXR expression by Kupffer cells. Exp Mol Pathol 2000; 68:1320. 99. LeCouter J, Moritz DR, Li B, et al. Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science 2003;299:890893. 100. Yoshiji H, Kuriyama S, Yoshii J, et al. Vascular endothelial growth factor and receptor interaction is a prerequisite for murine hepatic brogenesis. Gut 2003;52:13471354. 101. Ankoma-Sey V, Wang Y, Dai Z. Hypoxic stimulation of vascular endothelial growth factor expression in activated rat hepatic stellate cells. Hepatology 2000;31:141148. 102. Schwabe RF, Bataller R, Brenner DA. Human hepatic stellate cells express CCR5 and RANTES to induce proliferation and migration. Am J Physiol Gastrointest Liver Physiol 2003;285:G949958. 103. Gaca MD, Zhou X, Benyon RC. Regulation of hepatic stellate cell proliferation and collagen synthesis by proteinase-activated receptors. J Hepatol 2002;36:362369. 104. Bataller R, Paik YH, Lindquist JN, et al. Hepatitis C virus core and nonstructural proteins induce brogenic effects in hepatic stellate cells. Gastroenterology 2004;126:529540. 105. Efsen E, Grappone C, DeFranco RM, et al. Up-regulated expression of fractalkine and its receptor CX3CR1 during liver injury in humans. J Hepatol 2002;37:3947. 106. Kobayashi S, Seki S, Kawada N, et al. Apoptosis of T cells in the hepatic brotic tissue of the rat: a possible inducing role of hepatic myobroblast-like cells. Cell Tissue Res 2003;311:353364. 107. Safadi R, Ohta M, Alvarez CE, et al. Immune stimulation of hepatic brogenesis by CD8 cells and attenuation by transgenic interleukin-10 from hepatocytes. Gastroenterology 2004;127: 870882. 108. Gaca MD, Pickering JA, Arthur MJ, Benyon RC. Human and rat hepatic stellate cells produce stem cell factor: a possible mechanism for mast cell recruitment in liver brosis. J Hepatol 1999;30:850858. 109. Nelson DR, Tu Z, Soldevila-Pico C, et al. Long-term interleukin 10 therapy in chronic hepatitis C patients has a proviral and anti-inammatory effect. Hepatology 2003;38:859868. 110. Mazzocca A, Sciammetta SC, Carloni V, et al. Binding of hepatitis C virus envelope protein E2 to CD81 up-regulates matrix metalloproteinase-2 in human hepatic stellate cells. J Biol Chem 2005;280:1132911339. 111. Bonacchi A, Petrai I, Defranco RM, et al. The chemokine CCL21 modulates lymphocyte recruitment and brosis in chronic hepatitis C. Gastroenterology 2003;125:10601076. 112. Pohlmann S, Zhang J, Baribaud F, et al. Hepatitis C virus glycoproteins interact with DC-SIGN and DC-SIGNR. J Virol 2003;77:40704080. 113. Ikejima K, Takei Y, Honda H, et al. Leptin receptor-mediated signaling regulates hepatic brogenesis and remodeling of extracellular matrix in the rat. Gastroenterology 2002;122:13991410. 114. Leclercq IA, Farrell GC, Schriemer R, Robertson GR. Leptin is essential for the hepatic brogenic response to chronic liver injury. J Hepatol 2002;37:206213. 115. Saxena NK, Saliba G, Floyd JJ, Anania FA. Leptin induces increased alpha2(I) collagen gene expression in cultured rat hepatic stellate cells. J Cell Biochem 2003;89:311320.

116. Kamada Y, Tamura S, Kiso S, et al. Enhanced carbon tetrachloride-induced liver brosis in mice lacking adiponectin. Gastroenterology 2003;125:17961807. 117. Canbay A, Friedman S, Gores GJ. Apoptosis: the nexus of liver injury and brosis. Hepatology 2004;39:273278. 118. Taimr P , Higuchi H, Kocova E, et al. Activated stellate cells express the TRAIL receptor-2/death receptor-5 and undergo TRAIL-mediated apoptosis. Hepatology 2003;37:8795. 119. Trim N, Morgan S, Evans M, et al. Hepatic stellate cells express the low afnity nerve growth factor receptor p75 and undergo apoptosis in response to nerve growth factor stimulation. Am J Pathol 2000;156:12351243. 120. Lang A, Schoonhoven R, Tuvia S, et al. Nuclear factor kappaB in proliferation, activation, and apoptosis in rat hepatic stellate cells. J Hepatol 2000;33:4958. 121. Reif S, Lang A, Lindquist JN, et al. The role of focal adhesion kinase-phosphatidylinositol 3-kinase-akt signaling in hepatic stellate cell proliferation and type I collagen expression. J Biol Chem 2003;278:80838090. 122. Preaux AM, DOrtho MP , Bralet MP , et al. Apoptosis of human hepatic myobroblasts promotes activation of matrix metalloproteinase-2. Hepatology 2002;36:615622. 123. Li L, Tao J, Davaille J, et al. 15-deoxy-delta 12,14prostaglandin J2 induces apoptosis of human hepatic myobroblasts. A pathway involving oxidative stress independently of peroxisome-proliferator-activated receptors. J Biol Chem 2001;276:3815238158. 124. Yoshiji H, Kuriyama S, Yoshii J, et al. Tissue inhibitor of metalloproteinases-1 attenuates spontaneous liver brosis resolution in the transgenic mouse. Hepatology 2002;36:850860. 125. Wright MC, Issa R, Smart DE, et al. Gliotoxin stimulates the apoptosis of human and rat hepatic stellate cells and enhances the resolution of liver brosis in rats. Gastroenterology 2001;121:685698. 126. Sohara N, Znoyko I, Levy MT, et al. Reversal of activation of human myobroblast-like cells by culture on a basement membrane-like substrate. J Hepatol 2002;37:214221. 127. Lin SM, Sheen IS, Chien RN, et al. Long-term benecial effect of interferon therapy in patients with chronic hepatitis B virus infection. Hepatology 1999;29:971975. 128. Wanless IR, Nakashima E, Sherman M. Regression of human cirrhosis. Morphologic features and the genesis of incomplete septal cirrhosis. Arch Pathol Lab Med 2000;124:15991607. 129. Marcellin P , Chang TT, Lim SG, et al. Adefovir dipivoxil for the treatment of hepatitis B e antigen-positive chronic hepatitis B. N Engl J Med 2003;348:808816. 130. Chalasani N, Imperiale TF, Ismail A, et al. Predictors of large esophageal varices in patients with cirrhosis. Am J Gastroenterol 1999;94:32853291. 131. Zaman A, Becker T, Lapidus J, Benner K. Risk factors for the presence of varices in cirrhotic patients without a history of variceal hemorrhage. Arch Intern Med 2001;161: 25642570. 132. Giannini E, Risso D, Botta F, et al. Validity and clinical utility of the aspartate aminotransferasealanine aminotransferase ratio in assessing disease severity and prognosis in patients with hepatitis C virus-related chronic liver disease. Arch Intern Med 2003;163:218224. 133. Sheth SG, Flamm SL, Gordon FD, Chopra S. AST/ALT ratio predicts cirrhosis in patients with chronic hepatitis C virus infection. Am J Gastroenterol 1998;93:4448. 134. Giannini E, Testa R. Noninvasive diagnosis of brosis: the truth is rarely pure and never simple. Hepatology 2003;38:13121313; author reply 3. 135. Forns X, Ampurdanes S, Llovet JM, et al. Identication of chronic hepatitis C patients without hepatic brosis by a simple predictive model. Hepatology 2002;36:986992.

106

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 107

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

136. Wai CT, Greenson JK, Fontana RJ, et al. A simple noninvasive index can predict both signicant brosis and cirrhosis in patients with chronic hepatitis C. Hepatology 2003;38: 518526. 137. Poynard T, Aubert A, Bedossa P , et al. A simple biological index for detection of alcoholic liver disease in drinkers. Gastroenterology 1991;100:13971402. 138. Naveau S, Poynard T, Benattar C, Bedossa P , Chaput JC. Alpha-2-macroglobulin and hepatic brosis. Diagnostic interest. Dig Dis Sci 1994;39:24262432. 139. Imbert-Bismut F, Ratziu V, Pieroni L, et al. Biochemical markers of liver brosis in patients with hepatitis C virus infection: a prospective study. Lancet 2001;357:10691075. 140. Poynard T, Imbert-Bismut F, Munteanu M, et al. Overview of the diagnostic value of biochemical markers of liver brosis (FibroTest, HCV FibroSure) and necrosis (ActiTest) in patients with chronic hepatitis C. Comp Hepatol 2004;3:8. 141. Myers RP , Tainturier MH, Ratziu V, et al. Prediction of liver histological lesions with biochemical markers in patients with chronic hepatitis B. J Hepatol 2003;39:222230. 142. Naveau S, Raynard B, Ratziu V, et al. Biomarkers for the prediction of liver brosis in patients with chronic alcoholic liver disease. Clin Gastroenterol Hepatol 2005;3:167174. 143. Colombo M, Annoni G, Donato MF, et al. Serum type III procollagen peptide in alcoholic liver disease and idiopathic hemochromatosis: its relationship to hepatic brosis, activity of the disease and iron overload. Hepatology 1985;5:475479. 144. Tsutsumi M, Takase S, Urashima S, et al. Serum markers for hepatic brosis in alcoholic liver disease: which is the best marker, type III procollagen, type IV collagen, laminin, tissue inhibitor of metalloproteinase, or prolyl hydroxylase? Alcohol Clin Exp Res 1996;20:15121517. 145. Afdhal NH, Nunes D. Evaluation of liver brosis: a concise review. Am J Gastroenterol 2004;99:11601174. 146. Patel K, Gordon SC, Jacobson I, et al. Evaluation of a panel of non-invasive serum markers to differentiate mild from moderate-to-advanced liver brosis in chronic hepatitis C patients. J Hepatol 2004;41:935942. 147. Rosenberg WM, Voelker M, Thiel R, et al. Serum markers detect the presence of liver brosis: a cohort study. Gastroenterology 2004;127:17041713. 147a. Sud A, Hui JM, Farrell GC, et al. Improved prediction of brosis in chronic hepatitis C using measures of insulin resistance in a probability index. Hepatology 2004;39:12391247. 148. Poon TC, Hui AY, Chan HL, et al. Prediction of liver brosis and cirrhosis in chronic hepatitis B infection by serum proteomic ngerprinting: a pilot study. Clin Chem 2005;51:328335. 149. Bora R, Ratziu V, Bedossa P , et al. Diagnostic value of biochemical markers, brotestactitestHCVbrosure compared with serum protein proling by seldi-TOF, for the diagnosis of bridging brosis in patients with chronic hepatitis C. Hepatology 2004;40:517A. 150. Dev A, White I, Symonds W, et al. Serum proteomic analysis in hepatitis C patients with and without brosis. J Hepatol 2005;42(suppl 2):117A. 151. Callewaert N, Van Vlierberghe H, Van Hecke A, et al. Noninvasive diagnosis of liver cirrhosis using DNA sequencerbased total serum protein glycomics. Nature Med 2004;10:429434. 152. Wells RG. The role of matrix stiffness in hepatic stellate cell activation and liver brosis. J Clin Gastroenterol 2005;39:S158161. 153. Ziol M, Handra-Luca A, Kettaneh A, et al. Noninvasive assessment of liver brosis by measurement of stiffness in patients with chronic hepatitis C. Hepatology 2005;41:48 54.

154. Castera L, Vergniol J, Foucher J, et al. Prospective comparison of transient elastography, Fibrotest, APRI, and liver biopsy for the assessment of brosis in chronic hepatitis C. Gastroenterology 2005;128:343350. 155. Beljaars L, Meijer DK, Poelstra K. Targeting hepatic stellate cells for cell-specic treatment of liver brosis. Front Biosci 2002;7:e214222. 156. Adler M, Verset D, Bouhdid H, et al. Prognostic evaluation of patients with parenchymal cirrhosis. Proposal of a new simple score. J Hepatol 1997;26:642649. 157. Giannini EG, Testa R. 13C-breath tests and liver brosis. Eur Rev Med Pharmacol Sci 2004;8:5154. 158. Giannini EG, Fasoli A, Borro P , et al. 13C-galactose breath test and 13C-aminopyrine breath test for the study of liver function in chronic liver disease. Clin Gastroenterol Hepatol 2005;3: 279285. 159. Testa R, Caglieris S, Risso D, et al. Monoethylglycinexylidide formation measurement as a hepatic function test to assess severity of chronic liver disease. Am J Gastroenterol 1997;92:22682273. 160. Regev A, Berho M, Jeffers LJ, et al. Sampling error and intraobserver variation in liver biopsy in patients with chronic HCV infection. Am J Gastroenterol 2002;97:26142618. 161. Ratziu V, Charlotte F, Heurtier A, et al. Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology 2005;128:18981906. 162. Colloredo G, Guido M, Sonzogni A, Leandro G. Impact of liver biopsy size on histological evaluation of chronic viral hepatitis: the smaller the sample, the milder the disease. J Hepatol 2003;39:239244. 163. Bedossa P , Dargere D, Paradis V. Sampling variability of liver brosis in chronic hepatitis C. Hepatology 2003;38: 14491457. 164. Rousselet MC, Michalak S, Dupre F, et al. Sources of variability in histological scoring of chronic viral hepatitis. Hepatology 2005;41:257264. 164a. Wanless IR, Nakashima E, Sherman M. Regression of human cirrhosis. Morphologic features and the genesis of incomplete septal cirrhosis. Arch Pathol Lab Med 2000;124:15991607. 165. Powell LW, Kerr JF. Reversal of cirrhosis in idiopathic haemochromatosis following long-term intensive venesection therapy. Australas Ann Med 1970;19:5457. 166. Blumberg RS, Chopra S, Ibrahim R, et al. Primary hepatocellular carcinoma in idiopathic hemochromatosis after reversal of cirrhosis. Gastroenterology 1988;95: 13991402. 167. Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, et al. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazone. Hepatology 2003;38:10081017. 168. Ramond MJ, Poynard T, Rueff B, et al. A randomized trial of prednisolone in patients with severe alcoholic hepatitis. N Engl J Med 1992;326:507512. 169. Spahr L, Rubbia-Brandt L, Pugin J, et al. Rapid changes in alcoholic hepatitis histology under steroids: correlation with soluble intercellular adhesion molecule-1 in hepatic venous blood. J Hepatol 2001;35:582589. 170. Thompson K, Maltby J, Falloweld J, et al. Interleukin-10 expression and function in experimental murine liver inammation and brosis [see comments]. Hepatology 1998;28:15971606. 171. Akriviadis E, Botla R, Briggs W, et al. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a doubleblind, placebo-controlled trial. Gastroenterology 2000;119:16371648. 172. Spahr L, Rubbia-Brandt L, Frossard JL, et al. Combination of steroids with iniximab or placebo in severe alcoholic hepatitis: a randomized controlled pilot study. J Hepatol 2002;37:448455.

107

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 108

Section I. Pathophysiology of the Liver

173. Tilg H, Jalan R, Kaser A, et al. Anti-tumor necrosis factor-alpha monoclonal antibody therapy in severe alcoholic hepatitis. J Hepatol 2003;38:419425. 174. Menon KV, Stadheim L, Kamath PS, et al. A pilot study of the safety and tolerability of etanercept in patients with alcoholic hepatitis. Am J Gastroenterol 2004;99:255260. 175. Schaff Z, Lapis K, Szende B, et al. The effect of Dpenicillamine on CCl4-induced experimental liver cirrhosis. Exp Pathol 1991;43:111120. 176. Bodenheimer HC Jr, Schaffner F, Sternlieb I, et al. A prospective clinical trial of D-penicillamine in the treatment of primary biliary cirrhosis. Hepatology 1985;5:11391142. 177. Dickson ER, Fleming TR, Wiesner RH, et al. Trial of penicillamine in advanced primary biliary cirrhosis. N Engl J Med 1985;312:10111015. 178. Aithal GP , Haugk B, Das S, et al. Monitoring methotrexateinduced hepatic brosis in patients with psoriasis: are serial liver biopsies justied? Aliment Pharmacol Ther 2004;19:391399. 179. Te HS, Schiano TD, Kuan SF, et al. Hepatic effects of longterm methotrexate use in the treatment of inammatory bowel disease. Am J Gastroenterol 2000;95:31503156. 180. Kaplan MM, DeLellis RA, Wolfe HJ. Sustained biochemical and histologic remission of primary biliary cirrhosis in response to medical treatment. Ann Intern Med 1997;126: 682688. 181. Hendrickse MT, Rigney E, Giaffer MH, et al. Low-dose methotrexate is ineffective in primary biliary cirrhosis: longterm results of a placebo-controlled trial [see comments]. Gastroenterology 1999;117:400407. 182. Bach N, Bodian C, Bodenheimer H, et al. Methotrexate therapy for primary biliary cirrhosis. Am J Gastroenterol 2003;98:187193. 183. Kaplan MM, Cheng S, Price LL, Bonis PA. A randomized controlled trial of colchicine plus ursodiol versus methotrexate plus ursodiol in primary biliary cirrhosis: ten-year results. Hepatology 2004;39:915923. 184. Aleynik SI, Leo MA, Ma X, et al. Polyenylphosphatidylcholine prevents carbon tetrachloride-induced lipid peroxidation while it attenuates liver brosis. J Hepatol 1997;27:554561. 185. Lieber CS, Weiss DG, Groszmann R, et al. II. Veterans Affairs Cooperative Study of Polyenylphosphatidylcholine in Alcoholic Liver Disease. Alcohol Clin Exp Res 2003;27:17651772. 186. Boigk G, Stroedter L, Herbst H, et al. Silymarin retards collagen accumulation in early and advanced biliary brosis secondary to complete bile duct obliteration in rats. Hepatology 1997;26:643649. 187. Jia JD, Bauer M, Cho JJ, et al. Antibrotic effect of silymarin in rat secondary biliary brosis is mediated by downregulation of procollagen alpha1(I) and TIMP-1. J Hepatol 2001;35:392398. 188. Lieber CS, Leo MA, Cao Q, et al. Silymarin retards the progression of alcohol-induced hepatic brosis in baboons. J Clin Gastroenterol 2003;37:336339. 189. Ferenci P , Dragosics B, Dittrich H, et al. Randomized controlled trial of silymarin treatment in patients with cirrhosis of the liver. J Hepatol 1989;9:105113. 190. Pares A, Planas R, Torres M, et al. Effects of silymarin in alcoholic patients with cirrhosis of the liver: results of a controlled, double-blind, randomized and multicenter trial [see comments]. J Hepatol 1998;28:615621. 191. Brown KE, Poulos JE, Li L, et al. Effect of vitamin E supplementation on hepatic brogenesis in chronic dietary iron overload. Am J Physiol 1997;272:G116123. 192. Houglum K, Venkataramani A, Lyche K, Chojkier M. A pilot study of the effects of d-alpha-tocopherol on hepatic stellate cell activation in chronic hepatitis C. Gastroenterology 1997;113:10691073.

193. Hasegawa T, Yoneda M, Nakamura K, et al. Plasma transforming growth factor-beta1 level and efcacy of alphatocopherol in patients with non-alcoholic steatohepatitis: a pilot study. Aliment Pharmacol Ther 2001;15:16671672. 194. Mezey E, Potter J, Rennie-Tankersley L, et al. A randomized placebo controlled trial of vitamin E in alcoholic hepatitis. Hepatology 2003;38:264A. 195. Stewart S, Prince M, Bassendine M, et al. A trial of antioxidant therapy alone or with corticosteroids in acute alcoholic hepatitis. J Hepatol 2002;36(S):16. 196. Anon. The results of a randomized double blind controlled trial evaluating malotilate in primary biliary cirrhosis. A European multicentre study group. J Hepatol 1993;17:227235. 197. Lu SC, Tsukamoto H, Mato JM. Role of abnormal methionine metabolism in alcoholic liver injury. Alcohol 2002;27:155 162. 198. Mato JM, Camara J, Fernandez de Paz J, et al. Sadenosylmethionine in alcoholic liver cirrhosis: a randomized, placebo-controlled, double-blind, multicenter clinical trial. J Hepatol 1999;30:10811089. 199. Rambaldi A, Gluud C. Meta-analysis of propylthiouracil for alcoholic liver disease a Cochrane Hepato-Biliary Group Review. Liver 2001;21:398404. 200. Nava-Ocampo AA, Suster S, Muriel P . Effect of colchiceine and ursodeoxycholic acid on hepatocyte and erythrocyte membranes and liver histology in experimentally induced carbon tetrachloride cirrhosis in rats. Eur J Clin Invest 1997;27:7784. 201. Stiehl A. Ursodeoxycholic acid in the treatment of primary sclerosing cholangitis. Ann Med 1994;26:345349. 202. Combes B, Carithers RL Jr, Maddrey WC, et al. A randomized, double-blind, placebo-controlled trial of ursodeoxycholic acid in primary biliary cirrhosis. Hepatology 1995;22:759766. 203. Jacquemin E, Hermans D, Myara A, et al. Ursodeoxycholic acid therapy in pediatric patients with progressive familial intrahepatic cholestasis. Hepatology 1997;25:519523. 204. Lindblad A, Glaumann H, Strandvik B. A two-year prospective study of the effect of ursodeoxycholic acid on urinary bile acid excretion and liver morphology in cystic brosis-associated liver disease. Hepatology 1998;27:166174. 205. Goulis J, Leandro G, Burroughs AK. Randomised controlled trials of ursodeoxycholic-acid therapy for primary biliary cirrhosis: a meta-analysis. Lancet 1999;354:10531060. 206. Poupon RE, Bonnand AM, Chretien Y, Poupon R. Ten-year survival in ursodeoxycholic acid-treated patients with primary biliary cirrhosis. The UDCA-PBC Study Group. Hepatology 1999;29:16681671. 207. Degott C, Zafrani ES, Callard P , et al. Histopathological study of primary biliary cirrhosis and the effect of ursodeoxycholic acid treatment on histology progression. Hepatology 1999;29:10071012. 208. Poupon RE, Lindor KD, Pares A, et al. Combined analysis of the effect of treatment with ursodeoxycholic acid on histologic progression in primary biliary cirrhosis. J Hepatol 2003;39:1216. 209. Lindor KD, Kowdley KV, Heathcote EJ, et al. Ursodeoxycholic acid for treatment of nonalcoholic steatohepatitis: results of a randomized trial. Hepatology 2004;39:770778. 210. Rambaldi A, Iaquinto G, Gluud C. Anabolicandrogenic steroids for alcoholic liver disease: a Cochrane review. Am J Gastroenterol 2002;97:16741681. 211. Rockey DC, Maher JJ, Jarnagin WR, et al. Inhibition of rat hepatic lipocyte activation in culture by interferon-gamma. Hepatology 1992;16:776784. 212. Rockey DC, Chung JJ. Interferon gamma inhibits lipocyte activation and extracellular matrix mRNA expression during experimental liver injury: implications for treatment of hepatic brosis. J Invest Med 1994;42:660670.

108

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 109

Chapter 6 HEPATIC FIBROSIS AND CIRRHOSIS

213. Ziesche R, Hofbauer E, Wittman K, et al. A preliminary study of long-term treatment with interferon gamma-1b and low-dose prednisolone in patients with idiopathic pulmonary brosis. N Engl J Med 1999;341:12641269. 214. Toyonaga T, Hino O, Sugai S, et al. Chronic active hepatitis in transgenic mice expressing interferon-gamma in the liver. Proc Natl Acad Sci USA 1994;91:614618. 215. Muir AJ, Sylvestre PB, Rockey DC. Interferon gamma-1b for the treatment of chronic hepatitis C infection. J Viral Hepatitis 2006; in press. 216. Rodriguez L, Cerbon-Ambriz J, Munoz ML. Effects of colchicine and colchiceine in a biochemical model of liver injury and brosis. Arch Med Res 1998;29:109116. 217. Kaplan MM, Alling DW, Zimmerman HJ, et al. A prospective trial of colchicine for primary biliary cirrhosis. N Engl J Med 1986;315:14481454. 218. Kershenobich D, Vargas F, Garcia-Tsao G, et al. Colchicine in the treatment of cirrhosis of the liver. N Engl J Med 1988;318:17091713. 219. Morgan TR, Nemchausky B, Schiff ER, et al. Colchicine does not prolong life in patients with advanced alcoholic cirrhosis: results of a prospective, randomized, placebo-controlled trial. Gastroenterology 2002;122:641A. 220. Rambaldi A, Gluud C. Colchicine for alcoholic and nonalcoholic liver brosis or cirrhosis. Liver 2001;21:129136. 221. Isaka Y, Brees DK, Ikegaya K, et al. Gene therapy by skeletal muscle expression of decorin prevents brotic disease in rat kidney. Nature Med 1996;2:418423. 222. George J, Roulot D, Koteliansky VE, Bissell DM. In vivo inhibition of rat stellate cell activation by soluble TGF beta

223.

224.

225.

226.

227.

228.

229.

type II receptor: a potential new therapy for hepatic brosis. Proc Natl Acad Sci USA 1999;96:1271912724. Yata Y, Gotwals P , Koteliansky V, Rockey DC. Dose-dependent inhibition of hepatic brosis in mice by a TGF-beta soluble receptor: implications for antibrotic therapy. Hepatology 2002;35:10221030. Pinzani M, Milani S, Herbst H, et al. Expression of plateletderived growth factor and its receptors in normal human liver and during active hepatic brogenesis. Am J Pathol 1996;148:785800. Ikeda K, Wakahara T, Wang YQ, et al. In vitro migratory potential of rat quiescent hepatic stellate cells and its augmentation by cell activation. Hepatology 1999;29:17601767. Kinnman N, Francoz C, Barbu V, et al. The myobroblastic conversion of peribiliary brogenic cells distinct from hepatic stellate cells is stimulated by platelet-derived growth factor during liver brogenesis. Lab Invest 2003;83:163173. Di Sario A, Bendia E, Svegliati Baroni G, et al. Effect of pirfenidone on rat hepatic stellate cell proliferation and collagen production. J Hepatol 2002;37:584591. Miyahara T, Schrum L, Rippe R, et al. Peroxisome proliferatoractivated receptors and hepatic stellate cell activation. J Biol Chem 2000;275:3571535722. Bruck R, Genina O, Aeed H, et al. Halofuginone to prevent and treat thioacetamide-induced liver brosis in rats. Hepatology 2001;33:379386.

109

Ch006-X2998.qxd

3/15/06

5:21 PM

Page 110

Potrebbero piacerti anche