Immunopathogenesis of hepatitis C virus infection

Immunology and Cell Biology (2001) 79, 515–536 Review Article Immunopathogenesis of hepatitis C virus infection A N T H O N Y J F R E E M A N, 1 G E...
Author: Ralph Doyle
3 downloads 0 Views 384KB Size
Immunology and Cell Biology (2001) 79, 515–536

Review Article

Immunopathogenesis of hepatitis C virus infection A N T H O N Y J F R E E M A N, 1 G E O R G E M A R I N O S , 1 RO S E M A RY A F F R E N C H 2 a n d A N D R E W R L L OY D 3 1

Viral Hepatitis Research, Gastrointestinal and Liver Unit, The Prince of Wales Hospital, 2Westfield Research Laboratories, Sydney Children’s Hospital, Randwick and 3Inflammation Research Unit, School of Pathology, The University of New South Wales, Kensington, New South Wales, Australia Summary Hepatitis C virus, a recently identified member of the family Flaviviridae, is an important cause of chronic viral hepatitis and cirrhosis. There are similarities in the nature of the immune response to this pathogen with immunity in other flavivirus and hepatotropic virus infections, such as hepatitis B. However, the high rate of viral persistence after primary hepatitis C infection, and the observation that neutralizing antibodies are not protective, would suggest that there are a number of important differences between hepatitis C, other flaviviruses, and hepatitis B. The phenomenon of quasispecies evolution and other viral factors have been proposed to contribute to immune evasion by hepatitis C virus. In the face of established persistent infection, virus-specific cytotoxic T lymphocytes may exert some control over viral replication. However, these same effectors may also be responsible for the progressive liver damage characteristic of chronic hepatitis C infection. The nature of protective immunity, including the role of innate immune responses early after hepatitis C exposure, remains to be defined. Key words: CD8, chronic hepatitis, cytotoxic T lymphocytes, Flaviviridae, hepatitis C virus, immunity, liver, viral hepatitis, viral persistence.

Hepatitis C pathogenesis remains an important scientific challenge 10 years after the discovery of the virus Hepatitis C virus (HCV) is a significant public health problem. Approximately 3% of the world’s population has persistent HCV infection.1 In 1989, the virus was identified as the major aetiological agent responsible for post-transfusion non-A and non-B hepatitis.2–6 Following primary HCV infection, persistent viraemia and chronic hepatitis develop in the majority of cases.7 Patients are at risk of progressive hepatic fibrosis, cirrhosis and death from liver failure, as well as the advent of hepatocellular carcinoma.8 Efforts to elucidate the mechanisms behind viral persistence and hepatocellular damage have been frustrated by the lack of a reliable cell culture system for viral propagation in vitro. In addition, as the chimpanzee is the only experimental animal susceptible to HCV infection, progress in research is hampered by the lack of a small animal model to facilitate pathophysiological studies as well as the evaluation of antiviral treatment and vaccine strategies. The epidemiology and clinical aspects of HCV infection are briefly outlined below, followed by a summary of aspects of the molecular virology of HCV. The review of immunity in HCV infection focuses on: (i) the role of viral diversity in immune evasion, and other potential mechanisms of HCV persistence; (ii) the contribution of immune responses to Correspondence: Dr AJ Freeman, Viral Hepatitis Research, Gastrointestinal and Liver Unit, Edmund Blacket Building, The Prince of Wales Hospital, Barker Street, Randwick, NSW 2031, Australia. Email: [email protected] Received 30 March 2001; accepted 7 June 2001.

hepatocellular injury; (iii) studies investigating potential components of protective immunity against HCV, notably HCV-specific cytotoxic T lymphocytes (CTL); and (iv) the unique immunological environment encountered in the liver. The pathogenesis of HCV-related fibrosis, hepatocellular carcinoma and autoimmune disease associated with HCV infection are not discussed in detail. Potential insights into the immunopathogenesis of HCV infection may be gained by considering the immune response to other members of the Flaviviridae family, and to the hepatitis B virus (HBV), another viral agent responsible for chronic liver disease.

Hepatitis C is an hepatotropic flavivirus Hepatitis C virus was cloned from the plasma of an experimentally infected chimpanzee and characterized as a positive sense, ssRNA virus.2 On the basis of genome homology and hydrophobicity patterns, which are similar to the pestiviruses and flaviviruses, HCV has been classified as the first member of a third genus called Hepacivirus within the family Flaviviridae.9,10 Pestiviruses, such as bovine viral diarrhoea virus (BVDV), are important animal pathogens. In contrast, flaviviruses are responsible for disease in humans: including acute encephalitic infections (e.g. Japanese B encephalitis); febrile syndromes with rash, exemplified by dengue; and viral haemorrhagic fevers, such as yellow fever.11 The latter two diseases may include acute hepatitis as a feature. Other than with HCV, persistent infection is rare after primary flavivirus infection. Successful flavivirus vaccines have been developed, including the live, attenuated yellow fever vaccine, and the formalin-inactivated Japanese B encephalitis vaccine.12 However, there is currently no effective immunization strategy for the prevention, or treatment, of HCV infection.

516

AJ Freeman et al.

Similar to HCV, HBV is an hepatotropic virus that can cause persistent infection, chronic hepatitis, cirrhosis and hepatocellular carcinoma.13 Hepatitis B virus is a noncytopathic virus with a small, circular dsDNA genome and a lipoprotein envelope.14 In contrast to HCV, 95% of adults with primary HBV infection experience a self-limited illness that is attributed to an effective immune response and viral clearance.15 Vaccination with hepatitis B surface antigen results in the generation of protective antibody and a longlived anamnestic response in the majority of cases.16

Hepatitis C is a common pathogen Hepatitis C infection has been documented in sera stored in the 1950s.17 To the end of 1997, 190 000 Australians (i.e. approximately 1% of the population) were estimated to have antibodies to HCV and the annual incidence of new infections was estimated at 11 000.18 In the United States approximately 2.7 million persons suffer from persistent HCV infection.19 The prevalence is considerably higher in developing countries, such as parts of Africa and the Middle East.20,21 Hepatitis C virus represents a major economic burden to the health-care system because of the substantial medical costs incurred in managing patients with persistent infection.22,23

Hepatitis C is spread through blood-to-blood exposure Hepatitis C virus transmission occurs principally through parenteral exposure to contaminated blood or blood products.24 Because of the introduction of blood product and organ donor screening (in 1990 in Australia) the main risk factor for HCV infection remains injecting drug use.25,26 Between 50% and 70% of Australian injecting drug users are infected with HCV.18,27,28 Other risk factors for HCV infection include acupuncture, tattooing, haemodialysis, occupational exposure in health-care workers, and the re-use of needles in mass vaccination programs.24,29 The rate of transmission from mother-to-infant is approximately 6%.30 The majority of vertical HCV transmission occurs at the time of delivery.31 Early studies suggest that transmission is more common in the setting of high maternal viral load or in the presence of concomitant HIV infection.32–34 However, a recent study demonstrated that the viral load was the same in transmitting and non-transmitting women, and that vertical transmission was associated with the presence of negativestrand intermediates in maternal peripheral blood mononuclear cells.35 This supports the hypothesis that the transfer of infected cells to the infant is the major cause of vertical HCV transmission. Breast-feeding is not thought to increase infection risk to babies.31 Hepatitis C virus is not found consistently in bodily secretions, thus sexual transmission is rare.36,37 A small proportion of patients with no identifiable risk factor for HCV hepatitis are designated as sporadic cases and presumably reflect unrecognized or unreported blood-to-blood exposure. The majority of flaviviruses are transmitted to vertebrates by infected arthropods.9 However, mosquitoes fed HCV-infected blood are only transiently positive for viral RNA and are, therefore, incompetent as a vector of HCV.38

The liver in chronic hepatitis C contains a lymphocytic infiltrate and develops progressive fibrosis The histological features of persistent HCV infection are those of chronic hepatitis, with expanded portal tracts and lobular inflammation of varying severity.39,40 The infiltrate contains predominantly CD8+ T cells (Fig. 1).41 Lymphocytes may form focal aggregates that occasionally take the form of follicles with germinal centres. The limiting plates of hepatocytes around portal tracts may be eroded by interface hepatitis, also known as piecemeal necrosis. In contrast to chronic hepatitis B, confluent ‘bridging’ necrosis is uncommon. Hepatocytes in chronic HCV hepatitis may show acidophilic degeneration within intact plates and form apoptotic (Councilman) bodies. Bile duct damage and proliferation may also occur. Microvesicular fat accumulation (steatosis) is common, but rarely severe. Iron deposition (siderosis) is also rarely severe. However, in patients with thalassaemia major who receive multiple blood transfusions, or those with the inherited iron storage disorder, haemochromatosis, HCV can act synergistically with iron to promote liver damage.42 Progressive fibrosis occurs in patients with persistent HCV infection with a generally increasing prevalence over decades of chronic hepatitis.43 Once cirrhosis has developed the histological changes are less characteristic. Liver biopsies in patients with chronic HCV hepatitis are generally evaluated in terms of grade and stage. Grading is used to describe the intensity of necro-inflammatory activity. Staging is a measure of the fibrosis and architectural distortion that are the precursors of cirrhosis. A numerical score, for example the Knodell score with modifications by Ishak,44,45 is commonly assigned to both grade and stage, thereby providing a semiquantitative assessment of the observed histological features.

Primary hepatitis C virus infection results in persistent infection and cirrhosis in a significant proportion of cases Primary HCV infection is predominantly asymptomatic and fulminant hepatic failure in this context is rare.46,47 Serum HCV RNA can be identified by PCR as early as 7–21 days after exposure.48,49 There is a decline in the level of viraemia around the time of initial alteration in biochemical tests of liver function, notably a rise in the level of serum transaminases.49 Seroconversion typically occurs after approximately 2–8 weeks.50 Patients who present with jaundice (thus indicating more severe liver injury) during primary infection are more likely to clear HCV than the majority who acquire HCV without apparent symptoms.51–55 Some estimates suggest that persistent infection develops in approximately 80% of cases of HCV.54,56,57 However, considering that primary infection is predominantly asymptomatic and that antibody disappears in the intermediate term in a proportion of those who clear the virus,58 the true figure may be closer to 50%. The majority of patients with persistent HCV will have ongoing biochemical and histological evidence of hepatitis, but are typically either asymptomatic or have only mild fatigue.47 The most readily available marker of liver injury, the serum transaminase levels, characteristically fluctuate widely over time, correlate poorly with histological

HCV immunity

517

Figure 1 Chronic hepatitis C virus (HCV) infection is characterized by a lymphocytic infiltrate. (a) Uninfected human liver, demonstrating a portal tract (PT) surrounded by hepatocytes (H) stained with haematoxylin and eosin (H&E). (b) Chronic HCV infected liver with an expanded portal tract (PT), interface hepatitis (IH) and lobular inflammation (LI) (H&E). (c) Chronic HCV infected liver stained with anti-CD3 antibody to detect T lymphocytes. (d) Chronic HCV infected liver stained with anti-CD8 antibody. Arrows identify antigen positive cells. Reproduced with kind permission of CE Harvey and JJ Post. Magnification ×400.

disease activity and may be normal in patients with severe hepatitis and cirrhosis.59–61 The major morbidity and mortality of persistent HCV infection occurs after the establishment of hepatic fibrosis and the subsequent occurrence of cirrhosis and hepatocellular carcinoma.8 In those patients who develop clinically apparent HCV-related liver disease, the evolution is typically very slow with an average time to presentation with chronic hepatitis, cirrhosis and hepatocellular carcinoma of approximately 10, 20 and 30 years, respectively.62 As the infection is typically asymptomatic in the majority of patients, and runs a protracted course, it has been difficult to define the true proportion of cases that will progress to cirrhosis.63 There have been a large number of published series of patients recruited through specialist clinics that report an HCV-related cirrhosis rate in excess of 30% after 20 years infection.64,65 These cross-sectional series of referred patients, while containing large numbers of subjects, give a distorted picture of the natural history of HCV infection due to a selection bias

towards patients with more severe disease. Data from population-based cohorts suggest that less than 10% of patients with persistent HCV infection, acquired in young adulthood, will progress to cirrhosis after 20 years infection.66 The situation in the third and fourth decades is less certain, however, there is evidence that the rate of progression of hepatic fibrosis beyond 20 years is linear.43 Consistent with this impression of a more favourable outcome, a recent 45 year follow up of 17 American male military recruits with persistent HCV infection demonstrated that only 2 (12%) had developed advanced liver disease.17 Factors shown to accelerate progression to cirrhosis in persistent HCV infection include older age at HCV infection, male gender, heavy alcohol intake and co-infection with either HBV or HIV.43,67–69 The annual incidence of hepatocellular carcinoma is approximately 3% among HCV positive patients with cirrhosis.70 With regard to mortality, subjects with posttransfusion HCV infection followed-up 25 years after acquiring HCV during open-heart surgery had no excess all-cause

518

AJ Freeman et al.

Figure 2 Schematic diagram of the hepatitis C virus (HCV) genome. The location of HCV genes (C, core; E1/2, envelope; NS2/3/4A/4B/5A/5B, non-structural), proposed functions of gene products, and the 5′ and 3′ untranslated regions (5′UTR and 3′XR) are shown. Numbering refers to nucleotide positions of genes. From: Liang et al.352

mortality and only a small increase in liver-related mortality compared with age-matched controls.71,72 The mortality attributable to community acquired HCV infection as a result of injecting drug use remains to be defined.73

Current therapies for hepatitis C are only partially effective The goal of treatment is to eradicate HCV and prevent progression of liver disease. The two drugs currently available are IFN-α, a cytokine with immunomodulatory and antiviral activity, and ribavirin, a synthetic guanosine nucleoside analogue with in vitro antiviral activity.74 After a 48-week course of parenteral IFN-α monotherapy, less than 20% of patients with persistent HCV infection will clear the virus and show a sustained normalization of serum transaminase levels.75,76 A four-year follow up of treatment responders suggests that patients with persistently undetectable HCV RNA are cured.77 Ribavirin monotherapy has been shown to transiently decrease serum transaminase concentrations, but has no effect on serum viral load.78 Combination therapy with IFN-α and ribavirin, however, increases sustained virological and biochemical response rates to between 40% and 50%.79,80 Patients with higher serum HCV RNA levels, severe hepatic fibrosis and significant siderosis, and men, are less likely to respond to treatment.76,81 Hepatitis C virus has become the commonest indication for liver transplantation in Australia and worldwide.82,83 Chronic hepatitis redevelops in the majority of transplant recipients, but rarely leads to severe fibrosis or graft failure within the first decade.84

Much remains to be learnt about the molecular virology of hepatitis C The RNA genome of HCV consists of a single uninterrupted open-reading frame, approximately 9400 nucleotides in length, bracketed by 5′- and 3′-non-coding regions.85,86 Protein translation begins at an internal ribosomal entry site (IRES) in the 5′-non-coding region.87 The full-length polyprotein of approximately 3000 amino acids undergoes post-translational cleavage by host and viral proteases into HCV core, envelope glycoproteins (E1, E2) and non-

structural proteins (NS2, NS3, NS4A/B, NS5A/B)(Fig. 2).88 The core protein is thought to interact with RNA to form the virion nucleocapsid,89 while the non-structural regions are likely to have a role in virus replication and encode for proteases (NS2, NS3), a helicase (NS3) and an RNA-dependant RNA polymerase (NS5B).90 The virion half-life is between 3 and 5 h, with a clearance and production rate of approximately 1012 particles per day, corresponding to 50 particles per infected hepatocyte per day.91 Similar to other Flaviviridae, HCV is a small spherical virus (diameter approximately 30–50 nm)92,93 with a lipophilic envelope.94 Hepatitis C virus has been shown to associate with low density lipoproteins (LDL) in human sera.95,96 It has been proposed that an interaction between HCV–LDL complexes with LDL receptor is responsible for HCV binding and cell entry.97–99 However, the specific mechanism by which HCV particles interact with LDL receptor prior to internalization remains unclear. In addition, HCV E2 protein has been shown to bind the major extracellular loop of human CD81, a cell surface molecule expressed on virtually all nucleated cells.100 However, recent evidence suggests that this interaction is not important for HCV entry into cells.99,101,102 Hepatitis C virus envelope proteins may be important for fusion with the endosomal membrane through binding alternative, unidentified, cell surface proteins.103

Diagnosis of hepatitis C is based on serology and detection of viral RNA by PCR Serological assays for diagnosis of HCV were initially based on the detection of circulating antibody reactive to C100-3, a recombinant epitope derived from NS4.3 These assays lacked sensitivity and specificity, particularly in samples collected early in the course of HCV infection.104 Subsequent second and third generation enzyme-linked immunoassays and recombinant immunoblot assays detect circulating antibodies to multiple HCV epitopes, including core, which have been shown to appear earlier in the illness.105 Consequently, serological diagnosis has a sensitivity and specificity that approaches 95% when compared with serum HCV RNA detection by PCR of reverse transcribed cDNA.106 There is however, a proportion of HCV-infected individuals who are HCV RNA positive, but negative by serology.107 This group

HCV immunity

includes a small group of patients who have a delay of over 12 months before seroconversion after primary infection.108 Despite this, because serum HCV RNA levels fluctuate significantly over time and may give a false-negative result if the rate of viral replication is low or if viral persistence is limited to the liver, PCR testing is generally used to confirm persistent HCV infection only following serological testing for anti-HCV antibodies.109

The liver is the principal site of hepatitis C replication Immunohistochemical studies of liver specimens from patients with persistent HCV infection have demonstrated structural and non-structural antigens located almost exclusively within the cytoplasm of hepatocytes.110 In situ hybridization to detect complementary negative-strand RNA confirms that hepatocytes are the primary target for intrahepatic HCV replication.111,112 Early reports suggested that only a small proportion of hepatocytes are positive for negative-strand RNA, however, more recent evidence suggests that 25%,113 or even up to 73%,114 of these cells may have replicating HCV within them. The potential for extrahepatic replication and the biological significance of negative-strand RNA intermediates in both liver-infiltrating mononuclear cells and PBMC is unresolved.115–123 Additional sites of apparent extra-hepatic replication in immunodeficient hosts include lymph nodes, spleen, pancreas, adrenal gland and thyroid.124

Hepatitis C has a highly variable genome The genomic sequences of individual HCV isolates vary by as much as 35%. This nucleotide sequence heterogenicity is unevenly distributed over the genome. The most highly conserved region is at the terminus of the 5′-non-coding region.125 The proximal 3′-non-coding region varies in nucleotide sequence and length, downstream of which most isolates have a poly(U) stretch followed by a highly conserved 98-nucleotide tail.126 The conserved non-coding sequences are believed to be important for regulation of viral replication and gene expression.127 The putative core is the most highly conserved HCV protein, followed by conserved sequences in NS3 and NS5.128 The envelope proteins contain conserved elements, but overall these proteins are highly variable. A domain of approximately 28 amino acids at the amino-terminal end of E2 is so diverse among different HCV isolates that it has been labelled the hypervariable region (HVR1).129 However, despite significant variation at the nucleotide level, it retains a conformational structure that suggests it is involved in interactions with negatively charged compounds such as glycosaminoglycans and phospholipids.130 This is consistent with the hypothesis that HVR1 is important in interactions between HCV and the cell surface.

Hepatitis C exists as a dynamic quasispecies Typically, RNA viruses show high mutation frequencies because of an error-prone viral RNA polymerase that lacks proof-reading 3′-5′ exonuclease activity.131 For HCV, the rate of nucleotide misincorporation has been calculated as approximately 10–3 base substitutions per genome site per

519

year.132,133 The resulting genetic diversity defines the evolution of a quasispecies: a complex population of closely related HCV variants circulating simultaneously in each individual patient.134 The quasispecies encompasses a predominant ‘master’ genome, which is believed to have a superior replicative capacity, and a multitude of distinct ‘minor’ genomes. Generally, HCV quasispecies vary by less than 5% in nucleotide sequence.135 The generation and evolution of the quasispecies is likely to have important implications for escape from immune surveillance, generation of drug resistance and vaccine failure.136 Considering the potential role of the HVR1 in cell entry, as discussed above, and its high rate of nucleotide diversity, it is conceivable that different components of the quasispecies within a single patient may display different cellular tropism. This may explain the observation that circulating quasispecies do not always reflect those of the intrahepatic virus.137,138

The six hepatitis C genotypes may vary in pathogenicity Phylogenetic analysis of HCV isolates worldwide has lead to the classification of HCV variants into six1–6 distinct genotypes based on the NS5 region.139 Hepatitis C virus genotypes vary by as much as 30% in nucleotide sequence.135 Multiple, closely related isolates within these six genetic groups have been identified and are classified into subtypes (for example, 1a, 1b). It is not yet resolved as to whether more recently identified isolates from South-East Asia should be classified within the established six genotypes or represent three distinct, additional genetic variants.140–142 Detection of viral nucleotide sequence variations can be achieved using polymerase chain reaction (PCR) sequencing, type-specific primers, heteroduplex mobility analysis, selective hybridization and restriction fragment length polymorphism techniques targeting well-conserved areas, typically the 5′-non-coding region, of the HCV genome.143 Alternatively, serological methods that rely on differential detection of antibodies to antigenic sites, for example NS4, can be used to type HCV isolates.144 Genotype-specific changes in one region of the genome predict changes in other areas, thus, detection of type-specific antibody to NS4 peptides has been shown to correspond with PCR-based genotyping in almost all cases.145,146 The prevalent HCV genotypes vary in different geographical regions. Genotype 1 is widely distributed; however, in the United States genotype 1a predominates, while in Japan type 1b and 2a are most common. Genotype 4 is essentially restricted to the Middle East and central Africa, genotype 5 to South Africa and genotype 6 to Hong Kong.20,21,147,148 The distribution of given genotypes can vary in a single area over time. Over the past 30 years in Australia there has been an emergence of genotype 3, which is endemic in South-East Asia and India, and a corresponding relative reduction in the prevalence of genotype 1.149,150 All currently recognized HCV genotypes are hepatotropic and pathogenic.151 However, it has been suggested that different genotypes do vary in their infectivity and pathogenicity, thereby influencing the rate of progression to cirrhosis and the risk of hepatocellular carcinoma.152,153 Specifically, genotype 1b has been reported to be associated with higher

520

AJ Freeman et al.

Figure 3 Components of the adaptive anti-hepatitis C virus (anti-HCV) immune response. (Ab, antibody). Adapted from: Liang et al.352 and Koziel et al.302

HCV RNA levels, more advanced disease and a poor response to treatment.154–159 However, this has not been a universal finding.160–164 The association between type 1b infection and a worse prognosis may be confounded by factors such as acquisition of HCV through blood transfusion, an older age at infection and a longer duration of disease.67,165–167

Hepatitis C persists despite a vigorous immune response In contrast to other flavivirus infections and HBV, the majority of adults with primary HCV infection develop persistent infection. In addition, HCV can be responsible for multiple episodes of acute disease in an individual. Repetitive exposure of chimpanzees to heterologous and homologous HCV strains results in the reappearance of viraemia, as well as histopathological evidence of acute hepatitis attributable to reinfection rather than reactivation of the original strain.168,169 Similar observations have been made in multiply transfused thalassaemic children.170 As a DNA replicative intermediate has never been demonstrated in the HCV life cycle, there is no evidence to suggest that persistent HCV infection is related to viral integration into the host genome. Persistence must, therefore, relate to the inability of the host to mount an effective immune response, to viral factors that facilitate immune evasion, or to a combination of these factors. There is no evidence to suggest that individuals with persistent HCV infection are globally immunosuppressed.171,172 There is, in fact, considerable evidence that the majority of HCV-infected patients are capable of mounting polyclonal cellular and humoral immune responses directed at both structural and non-structural viral proteins. The recognized components of the adaptive anti-HCV immune response are summarized in Figure 3 and are discussed below.

Immune responses may mediate liver injury in hepatitis C In persistent HCV infection the immune response may contribute to hepatocellular injury. Immunosuppression of patients is generally associated with a transient normalization of serum transaminase levels and a surge in viraemia, while removal of immunosuppression can lead to an acute exacerbation of hepatitis.173 In addition, immunohistochemical studies in HCV infection indicate that T lymphocytes predominate both in the expanded portal tracts and inflammatory lobular infiltrates, suggesting that these cells are important mediators of HCV-related liver disease.41 The mechanisms of accumulation of effector cells in the liver in the setting of HCV are largely unexplored. The T-cell chemoattractant, interferon-inducible protein 10 (IP-10) is strongly expressed in HCV-infected liver.174,175 Our recent studies have demonstrated that hepatocytes are the major source of this chemokine, and that levels correlate with the degree of lobular inflammation.176

CD4+ helper T cells are an important component of the immune response to hepatitis C Antigenic peptides derived from the cleavage of exogenous viral proteins are recognized by CD4+ T-helper cells in association with MHC class II molecules on the surface of APC.177 T-helper lymphocytes have an immunoregulatory function through the secretion of lymphokines that support either cytotoxic T lymphocyte (CTL) generation (the T-helper type 1 cytokine profile: IL-2, IFN-γ) or B-cell function and antibody production (the type 2 profile: IL-4, IL-5, IL-10).178 Studies of the anti-HCV T-helper cell response, as detected

HCV immunity

by measuring PBMC proliferation in vitro in response to recombinant HCV proteins, have revealed that between 50% and 75% of individuals with persistent HCV infection have CD4+ T-cell responses to core, NS3, NS4 or NS5 proteins.179–182 A hierarchy of T-cell responsiveness to HCV proteins has been defined with core being the most immunogenic followed by NS4.183,184 Rather than representing the lack of T-helper lymphocyte activity, the absence of detectable lymphoproliferative responses to HCV in a minority of individuals with persistent HCV infection, may either relate to antigen-specific CD4+ cells present in the circulation at a frequency below the sensitivity of the assay, or lymphocytes that recognize variable regions of HCV proteins.185 An alternative explanation is compartmentalization of HCV-specific CD4+ T cells to the liver, as demonstrated in one study.186 Viral persistence in the face of significant T-helper cell proliferation at the site of viral replication may relate to the observation that, despite 40–80% of liver-infiltrating CD4+ cells expressing activation markers, just 1% of these cells were specific for HCV antigens.186 However, healthy subjects with lower levels of viraemia exhibit more vigorous proliferative responses to HCV antigens than those with progressive disease, particularly against core protein.179 This suggests that CD4+ T cells are indeed likely to influence HCV replication in patients with persistent HCV, probably by facilitating other effector mechanisms. The notion that CD4+ T lymphocytes may either directly or indirectly mediate tissue destruction is supported by the observation that a greater parenchymal concentration of activated CD4+ lymphocytes corresponds with more severe hepatitis.187,188 However, in one study, in vitro proliferation of T-helper cells was associated with a lower grade of liver inflammation, suggesting that CD4+ cells may be involved in host protection rather than liver injury.189 Similarly, higher levels of PBMC proliferation in response to HCV antigens in patients after liver transplantation have been shown to predict less severe histological changes due to HCV recurrence.190 An association between HLA class II genotype (for example, HLA DRB1*01)191 and HCV clearance in primary infection, suggests that CD4+ cells help play a critical role in facilitating clearance after HCV exposure.192–197 Furthermore, a vigorous CD4+ T-cell proliferative response from PBMC to multiple HCV proteins has been shown to predict disease resolution.51,52,180,198–201 Similarly, viral clearance after a course of IFN-α therapy,181,182,202,203 and combination IFN-α and ribavirin treatment204 appears to relate to the induction of a vigorous T-helper response. Rather than directly achieving viral clearance, it is likely that T-cell help plays a critical role in facilitating other antiviral immune mechanisms (e.g. CTL) as discussed below. The CD4+ response to HCV antigens has frequently been shown to persist for up to several decades following viral clearance, without apparent re-exposure to HCV.58,201 The response is generally directed at highly conserved immunodominant epitopes within the NS3 and core proteins that bind multiple HLA class II alleles.205 These features of the host response provide an encouraging backdrop for the development of prophylactic HCV vaccines designed to elicit cellular immune responses.206

521

Neutralizing antibodies do not prevent hepatitis C infection During primary HBV infection, the humoral response to envelope antigens contributes to the clearance of circulating virus particles.15 Similarly, in acute flavivirus infections, such as dengue and yellow fever, viraemia is typically terminated by the appearance of conformation-dependent neutralizing antibodies specific for determinants located on envelope glycoproteins.11 Clearance of yellow fever virus-infected cells is achieved by antibody-dependent complement-mediated lysis of cells bearing non-structural proteins.207 An effective humoral response, induced either by natural infection or by vaccination, is thought to generate long-lived protective immunity against subsequent infection. With regard to dengue, neutralizing antibodies are virus serotype-specific, allowing reinfection with alternative genotypes, such as dengue-2, after previous dengue-1 infection. Antibodies to envelope glycoproteins are also responsible for the phenomenon of antibody-dependant enhancement, in which preexisting antibodies against one genotype (e.g. dengue-1) facilitate viral entry via Fc receptors in the context of infection with a new genotype (e.g. dengue-2). This leads to fulminant clinical illness in dengue haemorrhagic fever,208 and exaggerated neurovirulence in yellow fever and Japanese B encephalitis.209 This phenomenon of antibody-dependent enhancement has not been studied in the context of hepatitis C. Immunization of chimpanzees with recombinant envelope glycoproteins is associated with the generation of a vigorous humoral response and control of HCV viral replication following a low-dose challenge with the homologous virus (i.e. the strain from which the vaccine was developed), but does not protect against heterologous HCV challenge (i.e. with another HCV isolate).210 In primary HCV infection, a humoral immune response also typically results in the generation of isolate-specific neutralizing antibodies.211 These antibodies bind only native and not denatured proteins, suggesting that they recognize conformational epitopes.212 As well as opsonizing HCV for elimination, antibodies may block viral attachment and impair HCV entry into cells.213 However, data from chimpanzees and humans suggest that generation of anti-E2 antibodies is not associated with viral clearance.214,215 In addition, resolution of primary HCV infection in agammaglobulinaemic children occurs in a similar proportion to the general population.216–218 The fact is that most patients with anti-HCV antibodies harbour a replicating virus, are infectious, and have evidence of liver injury.219 This suggests that antibodies are not protective. However, E2-specific antibodies may be involved in antibody-dependent cellular cytotoxicity (ADCC) against infected host cells.220 Rather than facilitating viral clearance, ADCC may contribute to hepatocellular damage.

Evolution of the hepatitis C quasispecies may facilitate immune evasion It has been postulated that the effectiveness of neutralizing antibodies in achieving viral clearance is limited because they have a restricted spectrum of activity against a potentially broad and evolving viral quasispecies.211 If this is the case, viral persistence in HCV infection may relate to the

522

AJ Freeman et al.

emergence of neutralization-resistant escape variants present in the quasispecies.221 Studies of chimpanzees inoculated with RNA transcribed in vitro from a cDNA clone have demonstrated that escape mutants need not be present in the infectious dose to establish persistence.133 However, in natural infection the selection of pre-existing mutants may occur.222 The HVR1 of E2 has been shown to be a critical target for neutralizing antibodies.213,223 Emergence of variants in vivo with mutations in this region may lead to a loss of antibody recognition of HVR1 epitopes and result in persistent HCV infection.224–227 Resolution of primary HCV infection has been associated with stasis of the quasispecies, and, conversely, progression to persistent infection has been associated with sequence changes within the HVR1.228 However, other factors are likely to be involved in viral persistence as demonstrated by the observation that HCV can cause persistent infection in chimpanzees in the absence of mutation events in the HVR1.229 Evolution of the quasispecies may provide the basis of establishment of persistence by selection for mutations in other parts of the HCV genome, such as alternative envelope and non-structural regions,133 which may be the targets for other host response mechanisms as outlined below. Indeed it has been argued that the primary failure in subjects who develop persistent HCV infection is a poor cellular immune response and that quasispecies evolution is the result of persistence rather than the cause. In this model, a lack of T-cell help by CD4+ cells leads to poor early viral control by CTL, restricted generation of antibody specificities, high selective pressure and immune escape.230 Alternatively, the observation that anti-HCV antibodies are not protective may simply relate to the timing of the generation of the humoral immune response in primary infection. Hepatitis C virus clearance has been associated with early antibody generation.231 Although there is no evidence in HCV infection, a delay in the production of neutralizing antibodies, and subsequent persistent infection, may relate to the appearance of HCV-specific CTL and the selective destruction of antigen-specific B cells expressing viral peptides in association with MHC class I.232,233

Is the cellular immune response to hepatitis C similar to that seen in hepatitis B? In hepatitis B, in conjunction with an MHC class II-restricted CD4+ T-helper response, virally encoded peptides synthesized within infected host cells and presented in association with class I molecules, induce a CD8+ CTL response.177 Polyclonal MHC class I- and II-restricted responses to structural and non-structural proteins are crucial for elimination of HBVinfected cells. In vitro studies suggest that the cellular immune response is vigorous and encompasses a broad range of epitopes in acutely infected patients who clear HBV, while the response is more narrowly focused in the 5% of adults who develop persistent infection.234–237 In contrast to adults, approximately 90% of neonates develop persistent HBV infection following vertical transmission.15,238 Neonatal tolerance to maternally derived circulating HBV e antigen may facilitate persistent HBV infection after perinatal exposure.239 However, vertical transmission of HBV also results in milder hepatitis, suggesting that the immune response may contribute to liver damage as well as having a role in host

protection.15 This is supported by the observation that immunodeficient individuals have a higher rate of persistent HBV, but milder liver disease.240 Consistent with this notion of immunologically mediated hepatocyte injury is the observation that both spontaneous clearance and acute exacerbations in patients with chronic HBV hepatitis are associated with stronger CTL activity. The direct hepatocytolytic effect of HBV-specific CTL in vivo has been demonstrated by the development of dose-dependant severe necro-inflammatory liver disease following adoptive transfer of HBV-specific CD8+ T-cell clones into transgenic mice expressing HBV antigens.241 Experiments using cytokine-specific neutralizing antibodies have demonstrated that in addition to direct cell lysis, CTL may inhibit intracellular HBV replication and cause liver damage via non-cytolytic mechanisms such as the secretion of IFN-γ and TNF-α.242 Viral clearance without hepatocyte destruction has been demonstrated in chimpanzees with acute HBV infection.243 As the majority of hepatocytes become infected in acute HBV, antiviral cytokines may be important in achieving viral clearance, while avoiding massive hepatocyte necrosis and fulminant hepatic failure, thereby ensuring host survival.244 Noncytopathic clearance of hepatotropic pathogens also occurs in murine cytomegalovirus (CMV) and lymphocytic choriomeningitis virus (LCMV) infection.245,246 Cytotoxic T cells specific for non-structural proteins presented in association with MHC class I may also contribute to the termination of flavivirus infections, with Kunjin and West Nile virus.247 Flaviviruses can upregulate MHC class I expression on infected host cells, thereby augmenting CTL-mediated cell lysis.248

Different methods used to study cytotoxic T lymphocytes in hepatitis C have produced varied results Several groups have studied HCV-specific CTL responses in peripheral blood and liver from HCV-infected patients and chimpanzees. Two assay strategies have been employed. The first has concentrated on the peripheral blood CTL response after in vitro HCV-peptide stimulation to expand the HCVspecific effector population.249–251 HLA-A2-restricted CTL activity in peripheral blood has been studied extensively because of the ease with which peripheral blood T cells are sampled and because of the high frequency of the HLA-A2 haplotype. The HLA-B44-restricted HCV-specific CTL response has been studied in Japanese patients because of an association with persistent HCV infection.249 The second strategy has sought to characterize the intrahepatic CTL response after T-cell expansion using non-antigen specific proliferative stimuli, such as anti-CD3 antibodies.252,253 In persistent HBV infection, HBV-specific CTL are generally undetectable in peripheral blood. By contrast, at least one HLA-A2-restricted CTL epitope (of 10 studied) was recognized by expanded PBMC in 97% of patients with persistent HCV infection.254 In general, the anti-HCV response was multispecific, with an average of four CTL epitopes being recognized in each patient. These CTL epitopes were located both in the structural and non-structural proteins of the virus.171 Given that the major site of HCV replication is believed to be the hepatocyte, and that chronic hepatitis due to HCV is characterized by a CD8+ T-cell infiltrate, it is not

HCV immunity

523

with substantially higher CTL activity evident with liverderived mononuclear cells than in peripheral blood (Fig. 4). This suggests that HCV-specific CTL are concentrated at the site of maximal viral replication and tissue injury.

Cytotoxic T lymphocytes may control viral replication in subjects with persistent hepatitis C infection

Figure 4 Intrahepatic and peripheral blood hepatitis C virus (HCV)-specific CTL responses and natural killer activity expanded from a patient with chronic HCV infection. Liver and peripheral blood mononuclear cells were expanded for six days in vitro using autologous cells infected with recombinant vaccinia virus constructs encoding HCV genomic regions (HCV genotype 1a core to NS2 () and genotype 1a NS2 to NS5 ( )).88 Autologous B lymphoblastoid cell lines were infected with the HCV-vaccinia virus constructs and vaccinia virus expressing the Escherichia coli b-galactose gene (vv-Lac, control ()). Target cell lysis was assessed in a 51Cr-release assay. Natural killer activity was assessed against K562 targets ( ). Intrahepatic lymphocytes displayed significant HCV-specific CTL activity against both genomic regions. Significantly less HCV–CTL was seen from peripheral blood effectors. Natural killer activity was high in blood and absent in liver.

surprising that HCV-specific CTL are sufficiently abundant in the liver that they can be detected after antigen-nonspecific expansion through the T-cell receptor complex.255 This method avoids virus-specific stimulation so that cells with a particular specificity are not selectively expanded. Using this technique, intrahepatic HCV-specific CTL activity can be detected in approximately 50% of cases.256,257 Intrahepatic CTL responses directed at multiple epitopes, including up to five structural and non-structural viral proteins, were demonstrated in patients with persistent HCV infection.258 Many of the epitopes identified after non-specific stimulation have been shown to conform to predicted MHC binding motifs, however, CTL epitopes recognized by both intrahepatic and peripheral blood effectors have been identified for which no published motif exists, suggesting that predicted binding motifs are not always reliable for identifying immunodominant determinants.258,259 The degree to which the peripheral blood CTL response detected after antigen-specific stimulation reflects the intrahepatic response is unknown. Our group has investigated intrahepatic and peripheral blood CTL activity in patients with persistent hepatitis C after in vitro expansion using autologous cells infected with recombinant vaccinia viruses encoding HCV genes.260 A broad and vigorous HCV-specific CTL response was demonstrated in the majority of patients,

The mechanism by which HCV persists despite the presence of a sustained, multispecific CTL response is not clear. Such a finding is particularly surprising considering that MHC class I, intercellular adhesion molecules and Fas antigen are all upregulated in the liver during HCV infection, and should facilitate cell binding, antigen recognition, and hepatocyte lysis.261,262 Although there is no evidence in HCV, one explanation may be that infected hepatocytes are induced to express Fas-ligand and may protect themselves against CTL-mediated injury by destroying virus-specific CTL via the same pathway that CTL use to kill their targets.263,264 Cytotoxic T lymphocytes may play a role in controlling viral replication in patients with persistent HCV infection. Attempts to correlate CTL activity with viral load have produced conflicting results. Our experiments suggest that a vigorous intrahepatic CTL response is associated with lower levels of viraemia.260 In one other study, the presence of an HCV-specific CTL response in liver-infiltrating lymphocytes was also associated with lower viral load.256 However, other studies have been unable to confirm this observation.257 An analysis of peripheral blood responses demonstrated no correlation between HCV-specific CTL precursor frequencies and viral load.171 However, in a related study the overall combined peripheral blood CTL response against a panel of epitopes was higher in patients whose HCV RNA level was below the threshold of detection,254 suggesting that HCVspecific CTL may be able to exert some control over viral replication, but not enough to terminate infection. A similar inverse relationship has been demonstrated between viral load and the HLA-B44-restricted peripheral blood CTL response to HCV core antigens.172

Cytotoxic T lymphocytes may contribute to hepatocellular injury in chronic hepatitis C The lymphocytic infiltrate characteristic of chronic HCV hepatitis contains a predominance of CD8+ cells over CD4+ cells. While CD4+ cells are confined to the portal and periportal areas, CD8+ cells contribute to the lobular infiltrate.265 This may suggest that similar to HBV, CTL may contribute to the liver damage in patients with HCV. While some investigators have failed to show any distinct cellular infiltrate at the site of antigen-positive hepatocytes,266 others have demonstrated CD8+ cells in contact with apoptotic bodies and hepatocytes containing HCV antigens.261 In addition, studies have demonstrated a correlation between the number of lobular CD8+ cells, serum transaminase levels and the degree of histological inflammation.265 However, correlations between functional assessments of HCV-specific CTL activity in vitro from persistently infected individuals with liver injury have produced conflicting results. The presence of liverderived CTL activity was associated with a higher hepatic

524

AJ Freeman et al.

necro-inflammatory score in one study.256 In contrast, analyses of peripheral blood HCV-specific CTL responsiveness demonstrated an inverse correlation with serum transaminase levels.254,267 This may be because HCV-specific CD8+ cells home to the liver as intrahepatic viral load increases and, therefore, are less frequently detected in the peripheral circulation in the context of more severe hepatitis. At odds with the hypothesis that CTL are directly responsible for liver injury in chronic HCV hepatitis is the recent report that only 1–2% of the total CD8+ cells infiltrating the liver, as detected by flow cytometric analysis of T cells stained with biotinylated MHC class I-peptide tetrameric complexes (tetramers), were HCV-specific.268 Similarly in HBV infection, tetramer staining has demonstrated that the bulk of the intrahepatic CD8+ cells in patients with severe hepatic inflammation are not specific for viral antigens.269 However, these studies only identified virus-specific CD8+ cells for which tetramers were available. Nevertheless, non-specific cytotoxic cells recruited by IFN-γ may contribute to liver damage in chronic viral hepatitis.270 Considering the close interplay between CD4+ help and CTL function, it has been postulated that deficient CD4+ T-helper function, in association with ongoing CTL activity and IFN-γ production, may be responsible for maintaining the low-grade hepatic inflammation and injury associated with persistent viraemia. A comprehensive evaluation of intrahepatic and peripheral blood cellular immunity in conjunction with measurement of viral load and assessment of hepatic necro-inflammatory activity may help clarify the situation.

Hepatitis C virus clearance in primary infection may depend on the vigour, breadth and quality of the early host CTL response Immunity in self-limited HCV infection has been difficult to define as the illness is largely asymptomatic, and thus patients rarely present to medical attention with primary infection. Despite this, the role of cellular immune factors in viral clearance during primary HCV infection has recently been addressed using techniques such as enzyme-linked immunospot (ELISPOT) assessment of IFN-γ production, tetramer and intracellular cytokine staining. In two experimentally infected chimpanzees, a vigorous, multispecific, intrahepatic CTL response, which persisted for over 1 year, and a limited antibody response were associated with viral clearance.271 These data must be interpreted cautiously given that in contrast to humans, only a minority of chimpanzees develop persistent HCV infection. However, a follow-up study of women who cleared HCV after exposure to contaminated human Rhesus immunoglobulin similarly demonstrated that while circulating HCV-specific antibodies were undetectable in 42% of cases 18–20 years after exposure, memory HCV-specific helper and cytotoxic T-cell responses persisted in 79% and 92%, respectively.58 Other investigators have confirmed that greater numbers of HCV-specific CD8+ T cells are found in patients with self-limited primary HCV infection,272,273 that an early, broadly directed response is critical,274 and that T-helper and CTL responses persist after resolution of clinical hepatitis.200 This has not been a universal finding with one report of comparable numbers of HCV peptide-specific IFN-γ producing cells in the peripheral

blood of persistently infected patients and recovered subjects.275 It may be that the responses in the liver differ from activity in blood. In line with this, early homing of HCVspecific CD4+ and CD8+ cells from blood to liver has been shown to be critical in chimpanzees that clear HCV infection.276 The detection of HCV-specific T cells in anti-HCV negative blood donors,251 HCV-exposed seronegative healthcare workers,277 spouses278 and healthy family members279 of persistently HCV-infected patients raises the notion that a cellular immune response may remain after the loss of antiHCV antibodies, or persist in people who never generate a significant antibody response. It is possible that cellular immunity persists due to retained antigen, or even plausibly low-level viral replication in the liver of clinically recovered patients.58,280 Alternatively, the response may reflect prior sensitization to non-HCV antigens.281 The ability of HCV to evade cellular immunity may relate to the observation that such responses, similar to humoral responses, have a viral-isolate specific spectrum of activity that may be ineffective against divergent strains present in the HCV quasispecies. The emergence of CTL epitope variants early in the course of primary infection may contribute to HCV persistence.282 Following primary infection in a chimpanzee that subsequently developed chronic hepatitis, liverinfiltrating CTL were identified that recognized an epitope in NS3, however, mutation at a single amino acid within the epitope resulted in the loss of CTL recognition.283 Mutant epitopes that emerge in this context may function as CTL antagonists and prevent T-cell recognition of the wild-type epitope.282,284–286 However, because the HCV-specific CTL response is generally multispecific, the mere loss of a single epitope should not provide sufficient survival advantage to the mutant. In addition, other flaviviruses have the same error-prone RNA-dependant RNA polymerase, which is responsible for the quasispecies nature of HCV, and yet rarely establish persistent infection.287 It has been argued that the selection of escape mutants occurs in the setting of preexisting persistent infection, that is, viral persistence leads to the selection of escape variants, not the converse.288 The extent to which quasispecies complexity contributes to, or is a consequence of, persistent HCV infection remains to be determined.

Early vaccine strategies provide promise for the control of hepatitis C It is clear that the quasispecies nature of HCV and the multiple prevalent genotypes pose a major challenge for the development of a protective vaccine.289 It is generally believed that an effective HCV vaccine would have to induce a vigorous, multispecific cellular immune response in order to eradicate HCV before the selection of escape mutants. Identification of conserved CTL epitopes restricted by multiple HLA alleles may facilitate the design of a vaccine that is immunogenic in the general population.267 A priming immunization with naked HCV-E2 DNA followed by a booster immunization with recombinant E2 protein (the so-called ‘prime-boost’ regimen) induced CTL activity and protected mice against tumour-expressing E2.290 Two chimpanzees vaccinated with HCV-E2 DNA also generated strong CTL responses, and after a viral challenge both resolved

HCV immunity

primary HCV infection early.291 A DNA prime and canarypoxHCV boost strategy using a polycistronic construct from the HCV core to the NS3 region has been shown to broaden the range of structural and non-structural epitopes targeted by HCV-specific CTL and IFN-γ-producing cells in immunized mice.292

Anti-viral cytokines regulate the immune response to hepatitis C The observed pattern of a vigorous CTL response and relatively weaker humoral response predicting disease resolution suggests that HCV persistence also relates to the pattern of cytokine release.189 In primary HCV infection, viral clearance is more likely in patients displaying a T-helper 1 profile,51,58,184,198,199,293 while T-helper type 2 cytokines may downregulate HCV-specific cellular immunity and promote viral persistence.294 Similarly, while higher serum IL-10 levels correlate with a poor response to treatment,295 a sustained treatment response has been associated with decreased IL-4 and IL-10.204,296 Activated HCV-specific CTL early in primary infection may be unable to secrete IFN-γ, representing a ‘stunned’ phenotype.200,297 Recovery from this may be important in affecting viral clearance. Hepatitis C virus core protein has been shown to directly downregulate IFN-γ and IL-2 production in mice.298 A predominance of type 2 cytokines, and a lack of IL-2 production by peripheral blood T-helper cells proliferating in response to NS3 has been demonstrated in persistent HCV infection.299 A reservoir of HCV may thereby be maintained in peripheral blood that continually reinfects the liver.300 Modification of cytokine responses by immune mediators other than IFN-α, for example, to promote a shift to T-helper type 1 predominance in response to IFN-β, may have future therapeutic applications.301 Despite a lower proportion of infected hepatocytes than in HBV, the number of HCV-infected cells is still so large that the CTL response is not likely to be able to eradicate HCV, while avoiding massive hepatocyte necrosis simply by killing infected cells.302 Persistence may, therefore, simply relate to a relative insensitivity of HCV to antiviral cytokines such as TNF-α and IFN-γ.275,303 However, HCV core protein has been shown to bind the cytoplasmic domain of TNF receptor 1 and enhance, rather than prevent, TNF-induced apoptosis.304 Other cytokines are also likely to play a role in immunemediated liver damage.305 The levels of IL-2 and IFN-γ have been shown to correlate with the degree of hepatic inflammation and fibrosis.306,307 Liver damage may extend from virally infected target cells to uninfected bystander hepatocytes via the release of other apoptotic mediators, such as Fas-ligand (Fas-L) from activated CTL.258,308

Little is known about the role of innate immunity in hepatitis C Uninfected human liver contains a high proportion of natural killer (NK) cells,309 and recently liver has also been shown to contain a significant population of cells with both NK-cell and T-cell phenotypic and functional characteristics, the natural T (NT) cells.310 These cells feature heavily biased T-cell receptor (TCR) gene usage, CD1d restriction and high

525

levels of cytokine production, particularly IL-4 and IFN-γ.311 In several murine models of infection, liver NT cells appear to play a key role in the host response.311 Thus, both NK and NT cells may have significant bearing on the immune response to hepatotropic pathogens such as HCV. Early NK-cell expansion, prior to the proliferation of CD4+ and CD8+ virus-specific cells, has been shown to control viral replication in murine models of viral hepatitis, such as LCMV.312 The high rate of HCV persistence following primary infection may plausibly relate to the observation that patients who develop persistent HCV infection have impaired NK activity,313 and few intrahepatic NK cells.314 Studies of this cell type in the liver of humans or chimpanzees with primary HCV infection are yet to be performed to better inform this debate. Similarly, virtually no data exists examining the potential role of NT cells in initiation or propagation of the host response to HCV. Similar to CTL it has been suggested that lymphocytes typically found in innate immune responses may play a role in producing chronic liver damage in HCV infection. Migration of NK and NT cells from blood to liver has been demonstrated in patients with persistent HCV infection.315 However, flow cytometry and in situ immunophenotyping of intrahepatic cytotoxic cells has demonstrated that NK and NT cells are not expanded in number in patients with chronic HCV hepatitis, thus suggesting that these cells are unlikely to play a major role in hepatocellular injury.265,316

Hepatitis C may use liver tolerance to promote its own persistence The relatively low requirement for immunosuppression to avoid organ rejection after allogeneic liver transplantation suggests that the liver may be a site for the induction of peripheral tolerance.317 This predisposition towards immune tolerance may relate to the observation that the liver is exposed to numerous harmless foreign antigens, including food and bacteria, from the intestine.318 Oral tolerance appears to involve both T-cell inactivation and deletion.319 The hepatotropic nature of HCV may facilitate the induction of tolerance and viral persistence. Activated HCV-specific T cells may be ‘trapped’ in the liver after antigen recognition on hepatocytes. Cells of the innate immune system may mediate apoptosis of activated cells, thereby terminating their antiviral function.315,318 In transgenic mice, enhanced apoptosis has been shown to be induced in activated T cells exposed to hepatocytes expressing HCV structural proteins.320 Alternatively, tolerance induction may relate to the observation that hepatocytes lack expression of costimulatory molecules, such as B7, and recognition of peptide in association with MHC class II by CD4+ cells may result in antigen-specific non-responsiveness.319

A number of viral factors may facilitate immune evasion by hepatitis C Hepatitis C virus proteins, such as core,298,321 E2322 and NS5,323 may interfere with the immune response, for example, by inhibition of the interferon-induced protein kinase PKR or binding to the gC1q complement receptor.324 Persistence may also be based on the kinetics of infection

526

AJ Freeman et al.

relative to the induction of immunity. During the early stages of infection a high rate of production of HCV virions may simply exceed the capacity of the immune response.91 Immunological tolerance may subsequently develop due to ‘exhaustion’ of the response in persistently viraemic patients.325 Viral persistence may also occur in the face of low levels of HCV replication, small doses of antigen, suboptimal T-cell activation, impaired proliferation and poor IL-2 production.326 Suppression of viral gene expression by a suboptimal immune response may facilitate persistent infection by reducing viral antigen expression so that infected cells escape immune recognition.288 Another potential method of immune evasion is replication in an immunologically privileged extra-hepatic site. For example, there is evidence for infection of pluripotent haematopoietic CD34+ stem cells.327 Lymphoid cells may thereby act as a reservoir of the virus that continually reinfects the liver. As discussed earlier, due to variations in the HVR1 potentially altering the tropism of different components of the HCV quasispecies, and the observation that progression to persistent infection has been associated with evolution of the HVR1,228 viral persistence may relate to mutations in the HVR1 facilitating HCV sequestration in immunologically privileged sites. In addition, infection of cells of the immune system, for example, dendritic cells,328 may impair their function. Consistent with this is the observation that dendritic cells recovered from HCV-infected patients display impaired allostimulatory activity.329

Hepatitis C virus may be directly cytopathic In general in flavivirus infection, disease pathogenesis relates primarily to a direct cytocidal effect of the virus on host cells. Severe disease is characterized by high levels of viral replication, rapid accumulation of antigen and an aggressive inflammatory response.11 The observation that the degree of liver damage in HCV-associated fulminant hepatic failure parallels the level of viral replication supports the notion that HCV may directly contribute to hepatocyte injury.330 In addition, highly viraemic liver transplant patients on immunosuppression may suffer cholestatic liver disease attributed to a direct cytopathic effect of HCV.331 The more typical situation in the immunocompetent host with established infection is controversial. Reports in patients with chronic HCV hepatitis have suggested a correlation between more severe histological changes and higher serum332–334 and intrahepatic335,336 HCV RNA levels, and a greater proportion of hepatocytes carrying negative-strand replicative intermediates.113 Against the argument for HCV having a direct cytopathic role are other studies that demonstrate no correlation between either intrahepatic RNA levels or the proportion of infected hepatocytes and necro-inflammatory activity.109,337–341 In addition, significant hepatocellular death is not seen in transgenic mice that express HCV core or E2 in the liver, although steatosis (which is also commonly observed in human infection) is apparently induced.342 It may be that the process of replication, rather than simply the presence of individual HCV proteins, induces HCV-associated cytopathic damage.113 However, it is well recognized that HCV replication can occur in patients without significant liver damage.343

The role of viral complexity as a pathogenic mechanism in hepatocellular injury has also been investigated. The degree of diversity of the HCV quasispecies has been shown to correlate with disease severity, such that the within-patient variation in nucleotide sequence increases with the severity of liver disease.138,344–347 However, this has not been a universal finding and may simply relate to a longer duration of infection.348–350 Longitudinal analysis of HCV quasispecies after primary infection has demonstrated that the complexity of the quasispecies may predict the subsequent course of HCV. A decrease in diversity is seen in patients that clear primary infection, and a dramatic increase in patients with rapidly progressive chronic hepatitis.228,289 In contrast, greater rates of HCV quasispecies diversification are associated with less severe disease in immunocompromised patients after liver transplantation351. The different methods used to describe quasispecies diversity may explain such disparate findings.

Conclusion Despite a decade of research since the discovery of HCV as an important agent responsible for chronic viral hepatitis, much remains to be defined regarding pathogenesis. Considerable efforts are being directed towards the development of an in vitro system to support viral propagation. Such a system will assist in defining the role of various viral proteins in translation, replication and critical virus–host cell interactions, for example, the means of HCV entry into hepatocytes. With regard to disease pathogenesis, the conclusions to date are largely based on studies of small numbers of chimpanzees, or, because of the difficulties in recruiting patients with primary HCV infection, observations have predominantly been made in subjects followed up long after HCV clearance, or in those who have persistent infection. Thus, key information regarding the evolution of the host response and viral dynamics early after infection is lacking. An additional consideration is the difficulty in sampling human hepatic tissue. Anti-viral immunity in the liver appears likely to differ significantly from the immune response observed in peripheral blood. A prospective and systematic evaluation of immunity, in conjunction with the assessment of viral replication and quasispecies, in a number of patients with primary HCV infection will assist in defining the nature of protective immunity. A better understanding of these immunological correlates of viral clearance will then facilitate immunotherapeutic and vaccine strategies.

Acknowledgements AJ Freeman is supported by a scholarship from the National Health and Medical Research Council of Australia.

References 1 Alter MJ. Epidemiology of hepatitis C. Hepatology 1997; 26: S62–5. 2 Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M. Isolation of a cDNA clone derived from a bloodborne non-A, non-B viral hepatitis genome. Science 1989; 244: 359–62.

HCV immunity

3 Kuo G, Choo QL, Alter HJ et al. An assay for circulating antibodies to a major etiologic virus of human non-A, non-B hepatitis. Science 1989; 244: 362–4. 4 Alter HJ, Purcell RH, Shih JW et al. Detection of antibody to hepatitis C virus in prospectively followed transfusion recipients with acute and chronic non-A, non-B hepatitis. N. Engl. J. Med. 1989; 321: 1494–1500. 5 Esteban JI, Esteban R, Viladomiu L et al. Hepatitis C virus antibodies among risk groups in Spain. Lancet 1989; 2: 294–7. 6 van der Poel CL, Reesink HW, Lelie PN, Leentvaar-Kuypers A, Choo QL, Kuo G, Houghton M. Anti-hepatitis C antibodies and non-A, non-B post-transfusion hepatitis in The Netherlands. Lancet 1989; 2: 297–8. 7 Alter MJ, Margolis HS, Krawczynski K et al. The natural history of community-acquired hepatitis C in the United States. The Sentinel Counties Chronic non-A, non-B Hepatitis Study Team. N. Engl. J. Med. 1992; 327: 1899–1905. 8 Bruix J, Barrera JM, Calvet X et al. Prevalence of antibodies to hepatitis C virus in Spanish patients with hepatocellular carcinomahepatic cirrhosis. Lancet 1989; 2: 1004–6. 9 Chambers TJ, Hahn CS, Galler R, Rice CM. Flavivirus genome organization, expression, and replication. Annu. Rev. Microbiol. 1990; 44: 649–88. 10 Houghton M, Weiner A, Han J, Kuo G, Choo QL. Molecular biology of the hepatitis C viruses: implications for diagnosis, development and control of viral disease. Hepatology 1991; 14: 381–8. 11 Monath TP, Heinz FX. Flaviviruses. In: Fields BN, Knipe DM, Howley PM (eds) Fields Virology. Philadelphia: LippincottRaven, 1996; 961–1034. 12 Stephenson JR. Flavivirus vaccines. Vaccine 1988; 6: 471–480. 13 Di Marco V, Lo Iacono O, Camma C et al. The long-term course of chronic hepatitis B. Hepatology 1999; 30: 257–64. 14 Lee WM. Hepatitis B virus infection. N. Engl. J. Med. 1997; 337: 1733–45. 15 Chisari FV, Ferrari C. Hepatitis B virus immunopathogenesis. Annu. Rev. Immunol. 1995; 13: 29–60. 16 Are booster immunisations needed for lifelong hepatitis B immunity? European Consensus Group on Hepatitis B Immunity. Lancet 2000; 355: 561–5. 17 Seeff LBMD, Miller RNMD, Rabkin CSMD et al. 45-year follow-up of hepatitis C virus infection in healthy young adults. Ann. Intern. Med. 2000; 132: 105–111. 18 Hepatitis C Virus Projections Working Group. Estimates and projections of the hepatitis C virus epidemic in Australia. Canberra: National Centre in HIV Epidemiology and Clinical Research, 1998. 19 Alter MJ, Kruszon-Moran D, Nainan OV et al. The prevalence of hepatitis C virus infection in the United States, 1988 through 1994. N. Engl. J. Med. 1999; 341: 556–62. 20 Xu LZ, Larzul D, Delaporte E, Brechot C, Kremsdorf D. Hepatitis C virus genotype 4 is highly prevalent in central Africa (Gabon). J. Gen. Virol. 1994; 75: 2393–8. 21 Chamberlain RW, Adams N, Saeed AA, Simmonds P, Elliott RM. Complete nucleotide sequence of a type 4 hepatitis C virus variant, the predominant genotype in the Middle East. J. Gen. Virol. 1997; 78: 1341–7. 22 Brown K, Crofts N. Health care costs of a continuing epidemic of hepatitis C virus infection among injecting drug users. Aust. NZ J. Public Health 1998; 22: 384–8. 23 Wong JB, McQuillan GM, McHutchison JG, Poynard T. Estimating future hepatitis C morbidity, mortality, and costs in the United States. Am. J. Public Health 2000; 90: 1562–9.

527

24 Alter MJ, Hadler SC, Judson FN et al. Risk factors for acute non-A, non-B hepatitis in the United States and association with hepatitis C virus infection. JAMA 1990; 264: 2231–5. 25 Donahue JG, Munoz A, Ness PM et al. The declining risk of post-transfusion hepatitis C virus infection. N. Engl. J. Med. 1992; 327: 369–73. 26 Thomas DL, Vlahov D, Solomon L, Cohn S, Taylor E, Garfein R, Nelson KE. Correlates of hepatitis C virus infections among injection drug users. Medicine 1995; 74: 212–20. 27 Crofts N, Jolley D, Kaldor J, van Beek I, Wodak A. Epidemiology of hepatitis C virus infection among injecting drug users in Australia. J. Epidemiol. Community Health 1997; 51: 692–7. 28 MacDonald MA, Wodak AD, Dolan KA, van Beek I, Cunningham PH, Kaldor JM. Hepatitis C virus antibody prevalence among injecting drug users at selected needle and syringe programs in Australia, 1995–97. Med. J. Aust. 2000; 172: 57–61. 29 Post JJ, Dolan KA, Whybin LR, Carter IWJ, Haber PS, Lloyd AR. Acute hepatitis C virus infection in an Australian prison inmate: tattooing as a possible transmission route. Med. J. Aust. 2001; 174: 183–4. 30 Dore GJ, Kaldor JM, McCaughan GW. Systematic review of the role of polymerase chain reaction in defining infectiousness among people infected with hepatitis C virus. BMJ 1997; 315: 333–7. 31 Gibb DM, Goodall RL, Dunn DT, Healy M, Neave P, Cafferkey M, Bulter K. Mother-to-child transmission of hepatitis C virus: evidence for preventable peripartum transmission. Lancet 2000; 356: 904–7. 32 Roudot-Thoraval F, Pawlotsky JM, Thiers V et al. Lack of mother-to-infant transmission of hepatitis C virus in human immunodeficiency virus-seronegative women: a prospective study with hepatitis C virus RNA testing. Hepatology 1993; 17: 772–7. 33 Lin HH, Kao JH, Hsu HY et al. Possible role of high-titer maternal viremia in perinatal transmission of hepatitis C virus. J. Infect. Dis. 1994; 169: 638–41. 34 Ohto H, Terazawa S, Sasaki N et al. Transmission of hepatitis C virus from mothers to infants. The Vertical Transmission of Hepatitis C Virus Collaborative Study Group. N. Engl. J. Med. 1994; 330: 744–50. 35 Azzari C, Resti M, Moriondo M, Ferrari R, Lionetti P, Vierucci A. Vertical transmission of HCV is related to maternal peripheral blood mononuclear cell infection. Blood 2000; 96: 2045–8. 36 Fried MW, Shindo M, Fong TL, Fox PC, Hoofnagle JH, Di Bisceglie AM. Absence of hepatitis C viral RNA from saliva and semen of patients with chronic hepatitis C. Gastroenterology 1992; 102: 1306–8. 37 Bresters D, Mauser-Bunschoten EP, Reesink HW et al. Sexual transmission of hepatitis C virus. Lancet 1993; 342: 210–11. 38 Bellini R, Casali B, Carrieri M, Zambonelli C, Rivasi P, Rivasi F. Aedes albopictus (Diptera: Culicidae) is incompetent as a vector of hepatitis C virus. Apmis 1997; 105: 299–302. 39 Scheuer PJ, Ashrafzadeh P, Sherlock S, Brown D, Dusheiko GM. The pathology of hepatitis C. Hepatology 1992; 15: 567–71. 40 Bach N, Thung SN, Schaffner F. The histological features of chronic hepatitis C and autoimmune chronic hepatitis: a comparative analysis. Hepatology 1992; 15: 572–7. 41 Marrogi AJ, Cheles MK, Gerber MA. Chronic hepatitis C. Analysis of host immune response by immunohistochemistry. Arch. Pathol. Lab. Med. 1995; 119: 232–7. 42 Piperno A, Fargion S, Alba R et al. Liver damage in Italian patients with hereditary hemochromatosis is highly influenced by hepatitis B and C virus infection. J. Hepatol. 1992; 16: 364–8.

528

AJ Freeman et al.

43 Poynard T, Bedossa P, Opolon P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 1997; 349: 825–32. 44 Knodell RG, Ishak KG, Black WC et al. Formulation and application of a numerical scoring system for assessing histological activity in asymptomatic chronic active hepatitis. Hepatology 1981; 1: 431–5. 45 Ishak K, Baptista A, Bianchi L et al. Histological grading and staging of chronic hepatitis. J. Hepatol. 1995; 22: 696–9. 46 Wright TL, Hsu H, Donegan E et al. Hepatitis C virus not found in fulminant non-A, non-B hepatitis. Ann. Intern. Med. 1991; 115: 111–12. 47 Hoofnagle JH. Hepatitis C: the clinical spectrum of disease. Hepatology 1997; 26: S15–20. 48 Garson JA, Tuke PW, Makris M, Briggs M, Machin SJ, Preston FE, Tedder RS. Demonstration of viraemia patterns in haemophiliacs treated with hepatitis-C-virus-contaminated factor VIII concentrates. Lancet 1990; 336: 1022–5. 49 Farci P, Alter HJ, Wong D, Miller RH, Shih JW, Jett B, Purcell RH. A long-term study of hepatitis C virus replication in non-A, non-B hepatitis. N. Engl. J. Med. 1991; 325: 98–104. 50 Tremolada F, Casarin C, Tagger A, Ribero ML, Realdi G, Alberti A, Ruol A. Antibody to hepatitis C virus in posttransfusion hepatitis. Ann. Intern. Med. 1991; 114: 277–81. 51 Diepolder HM, Zachoval R, Hoffmann RM et al. Possible mechanism involving T-lymphocyte response to non-structural protein 3 in viral clearance in acute hepatitis C virus infection. Lancet 1995; 346: 1006–7. 52 Missale G, Bertoni R, Lamonaca V et al. Different clinical behaviors of acute hepatitis C virus infection are associated with different vigor of the anti-viral cell-mediated immune response. J. Clin. Invest. 1996; 98: 706–14. 53 Sachithanandan S, Fielding JF. Jaundice at onset signifies a good prognosis in anti-D-associated HCV infection. Eur. J. Gastroenterol. Hepatol. 1997; 9: 77–9. 54 Villano SA, Vlahov D, Nelson KE, Cohn S, Thomas DL. Persistence of viremia and the importance of long-term followup after acute hepatitis C infection. Hepatology 1999; 29: 908–14. 55 Wiese M, Berr F, Lafrenz M, Porst H, Oesen U. Low frequency of cirrhosis in a hepatitis C (genotype 1b) single-source outbreak in Germany: a 20-year multicenter study. Hepatology 2000; 32: 91–6. 56 Di Bisceglie AM, Goodman ZD, Ishak KG, Hoofnagle JH, Melpolder JJ, Alter HJ. Long-term clinical and histopathological follow-up of chronic posttransfusion hepatitis. Hepatology 1991; 14: 969–74. 57 Sansonno D, Dammacco F. Hepatitis-C-virus-related chronic liver disease of sporadic type: clinical, serological and histological features. Digestion 1992; 51: 115–120. 58 Takaki A, Wiese M, Maertens G et al. Cellular immune responses persist and humoral responses decrease two decades after recovery from a single-source outbreak of hepatitis C. Nat. Med. 2000; 6: 578–82. 59 Haber MM, West AB, Haber AD, Reuben A. Relationship of aminotransferases to liver histological status in chronic hepatitis C. Am. J. Gastroenterol. 1995; 90: 1250–7. 60 McCormick SE, Goodman ZD, Maydonovitch CL, Sjogren MH. Evaluation of liver histology, ALT elevation, and HCV RNA titer in patients with chronic hepatitis C. Am. J. Gastroenterol. 1996; 91: 1516–22. 61 Gholson CF, Morgan K, Catinis G, Favrot D, Taylor B, Gonzalez E, Balart L. Chronic hepatitis C with normal amino-

62

63 64

65 66

67

68

69

70

71

72

73

74 75 76 77

78

79

transferase levels: a clinical histologic study. Am. J. Gastroenterol. 1997; 92: 1788–92. Kiyosawa K, Sodeyama T, Tanaka E et al. Interrelationship of blood transfusion, non-A, non-B hepatitis and hepatocellular carcinoma: analysis by detection of antibody to hepatitis C virus. Hepatology 1990; 12: 671–5. Seeff LB. Natural history of hepatitis C. Hepatology 1997; 26: S21–8. Tong MJ, el-Farra NS, Reikes AR, Co RL. Clinical outcomes after transfusion-associated hepatitis C. N. Engl. J. Med. 1995; 332: 1463–6. Yano M, Kumada H, Kage M et al. The long-term pathological evolution of chronic hepatitis C. Hepatology 1996; 23: 1334–40. Freeman AJ, Dore GJ, Law MG et al. Estimating progression to cirrhosis in chronic hepatitis C virus infection. Hepatology 2001; 34: 809–16. Roudot-Thoraval F, Bastie A, Pawlotsky JM, Dhumeaux D. Epidemiological factors affecting the severity of hepatitis C virus-related liver disease: a French survey of 6,664; patients. The Study Group for the Prevalence and the Epidemiology of Hepatitis C Virus. Hepatology 1997; 26: 485–90. Benvegnu L, Fattovich G, Noventa F, Tremolada F, Chemello L, Cecchetto A, Alberti A. Concurrent hepatitis B and C virus infection and risk of hepatocellular carcinoma in cirrhosis. A prospective study. Cancer 1994; 74: 2442–8. Benhamou Y, Bochet M, Di Martino V et al. Liver fibrosis progression in human immunodeficiency virus and hepatitis C virus coinfected patients. The Multivirc Group. Hepatology 1999; 30: 1054–8. Serfaty L, Aumaitre H, Chazouilleres O, Bonnand AM, Rosmorduc O, Poupon RE, Poupon R. Determinants of outcome of compensated hepatitis C virus-related cirrhosis. Hepatology 1998; 27: 1435–40. Seeff LB, Buskell-Bales Z, Wright EC et al. Long-term mortality after transfusion-associated non-A, non-B hepatitis. The National Heart, Lung, and Blood Institute Study Group. N. Engl. J. Med. 1992; 327: 1906–11. Seeff LB, Hollinger FB, Alter HJ et al. Long-term mortality and morbidity of transfusion-associated non-A, non-B, and type C hepatitis: a national heart, lung, and blood insitute collaberative study. Hepatology 2001; 33: 455–63. Rodger AJ, Thomson JA, Thompson SC, Jolley D, Mijch AM, Lanigan A, Crofts N. Assessment of long-term outcomes of hepatitis C virus infection in a cohort of patients with acute hepatitis in 1971–1975: results of a pilot study. J. Gastroenterol. Hepatol. 1999; 14: 269–73. Patterson JL, Fernandez-Larsson R. Molecular mechanisms of action of ribavirin. Rev. Infect. Dis. 1990; 12: 1139–46. Carithers Jr RL, Emerson SS. Therapy of hepatitis C: metaanalysis of interferon alfa-2b trials. Hepatology 1997; 26: S83–8. Hoofnagle JH, Di Bisceglie AM. The treatment of chronic viral hepatitis. N. Engl. J. Med. 1997; 336: 347–56. Shindo M, Ken A, Okuno T. Varying incidence of cirrhosis and hepatocellular carcinoma in patients with chronic hepatitis C responding differently to interferon therapy. Cancer 1999; 85: 1943–50. Dusheiko G, Main J, Thomas H et al. Ribavirin treatment for patients with chronic hepatitis C: results of a placebo-controlled study. J. Hepatol. 1996; 25: 591–8. Poynard T, Marcellin P, Lee SS et al. Randomised trial of interferon alpha2b plus ribavirin for 48 weeks or for 24 weeks versus interferon alpha2b plus placebo for 48 weeks for treatment of chronic infection with hepatitis C virus. International Hepatitis Interventional Therapy Group (IHIT). Lancet 1998; 352: 1426–32.

HCV immunity

80 McHutchison JG, Gordon SC, Schiff ER et al. Interferon alfa2b alone or in combination with ribavirin as initial treatment for chronic hepatitis C. Hepatitis Interventional Therapy Group. N. Engl. J. Med. 1998; 339: 1485–92. 81 Van Thiel DH, Friedlander L, Fagiuoli S, Wright HI, Irish W, Gavaler JS. Response to interferon alpha therapy is influenced by the iron content of the liver. J. Hepatol. 1994; 20: 410–15. 82 Levy MT, Chen JJ, McGuinness PH, Koorey D, Sheil AG, McCaughan GW. Liver transplantation for hepatitis Cassociated cirrhosis in a single Australian centre: referral patterns and transplant outcomes. J. Gastroenterol. Hepatol. 1997; 12: 453–9. 83 Read AE, Donegan E, Lake J et al. Hepatitis C in patients undergoing liver transplantation. Ann. Intern. Med. 1991; 114: 282–4. 84 Boker KH, Dalley G, Bahr MJ et al. Long-term outcome of hepatitis C virus infection after liver transplantation. Hepatology 1997; 25: 203–10. 85 Kato N, Hijikata M, Ootsuyama Y, Nakagawa M, Ohkoshi S, Sugimura T, Shimotohno K. Molecular cloning of the human hepatitis C virus genome from Japanese patients with non-A, non-B hepatitis. Proc. Natl Acad. Sci. USA 1990; 87: 9524–8. 86 Choo QL, Richman KH, Han JH et al. Genetic organization diversity of the hepatitis C virus. Proc. Natl Acad. Sci. USA 1991; 88: 2451–5. 87 Tsukiyama-Kohara K, Iizuka N, Kohara M, Nomoto A. Internal ribosome entry site within hepatitis C virus RNA. J. Virol. 1992; 66: 1476–83. 88 Grakoui A, Wychowski C, Lin C, Feinstone SM, Rice CM. Expression and identification of hepatitis C virus polyprotein cleavage products. J. Virol. 1993; 67: 1385–95. 89 Yasui K, Wakita T, Tsukiyama-Kohara K et al. The native form and maturation process of hepatitis C virus core protein. J. Virol. 1998; 72: 6048–55. 90 Major ME, Feinstone SM. The molecular virology of hepatitis C. Hepatology 1997; 25: 1527–38. 91 Neumann AU, Lam NP, Dahari H, Gretch DR, Wiley TE, Layden TJ, Perelson AS. Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy. Science 1998; 282: 103–7. 92 Yuasa T, Ishikawa G, Manabe S, Sekiguchi S, Takeuchi K, Miyamura T. The particle size of hepatitis C virus estimated by filtration through microporous regenerated cellulose fibre. J. Gen. Virol. 1991; 72: 2021–4. 93 Shimizu YK, Feinstone SM, Kohara M, Purcell RH, Yoshikura H. Hepatitis C virus: detection of intracellular virus particles by electron microscopy. Hepatology 1996; 23: 205–9. 94 Feinstone SM, Mihalik KB, Kamimura T, Alter HJ, London WT, Purcell RH. Inactivation of hepatitis B virus and non-A, non-B hepatitis by chloroform. Infect. Immun. 1983; 41: 816–21. 95 Thomssen R, Bonk S, Propfe C, Heermann KH, Kochel HG, Uy A. Association of hepatitis C virus in human sera with betalipoprotein. Med. Microbiol. Immunol. 1992; 181: 293–300. 96 Prince AM, Huima-Byron T, Parker TS, Levine DM. Visualization of hepatitis C virions and putative defective interfering particles isolated from low-density lipoproteins. J. Viral Hepat. 1996; 3: 11–17. 97 Monazahian M, Bohme I, Bonk S, Koch A, Scholz C, Grethe S, Thomssen R. Low density lipoprotein receptor as a candidate receptor for hepatitis C virus. J. Med. Virol. 1999; 57: 223–9. 98 Agnello V, Abel G, Elfahal M, Knight GB, Zhang QX. Hepatitis C virus and other flaviviridae viruses enter cells via low density lipoprotein receptor. Proc. Natl Acad. Sci. USA 1999; 96: 12 766–71.

529

99 Wünschmann S, Medh JD, Klinzmann D, Schmidt WN, Stapleton JT. Characterization of hepatitis C virus (HCV) and HCV E2 interactions with CD81 and the low-density lipoprotein receptor. J. Virol. 2000; 74: 10 055–62. 100 Pileri P, Uematsu Y, Campagnoli S et al. Binding of hepatitis C virus to CD81. Science 1998; 282: 938–41. 101 Flint M, Thomas JM, Maidens CM, Shotton C, Levy S, Barclay WS, McKeating JA. Functional analysis of cell surface-expressed hepatitis C virus E2 glycoprotein. J. Virol. 1999; 73: 6782–90. 102 Meola A, Sbardellati A, Ercole BB et al. Binding of hepatitis C virus E2 glycoprotein to CD81 does not correlate with species permissiveness to infection. J. Virol. 2000; 74: 5933–8. 103 Takikawa S, Ishii K, Aizaki H, Suzuki T, Asakura H, Matsuura Y, Miyamura T. Cell fusion activity of hepatitis C virus envelope proteins. J. Virol. 2000; 74: 5066–74. 104 Esteban JI, Gonzalez A, Hernandez JM et al. Evaluation of antibodies to hepatitis C virus in a study of transfusionassociated hepatitis. N. Engl. J. Med. 1990; 323: 1107–12. 105 Van der Poel CL, Cuypers HT, Reesink HW et al. Confirmation of hepatitis C virus infection by new four-antigen recombinant immunoblot assay. Lancet 1991; 337: 317–19. 106 Nakagiri I, Ichihara K, Ohmoto K, Hirokawa M, Matsuda N. Analysis of discordant test results among five secondgeneration assays for anti-hepatitis C virus antibodies also tested by polymerase chain reaction-RNA assay and other laboratory and clinical tests for hepatitis. J. Clin. Microbiol. 1993; 31: 2974–80. 107 Schneeberger PM, Keur I, van der Vliet W et al. Hepatitis C virus infections in dialysis centers in The Netherlands: a national survey by serological and molecular methods. J. Clin. Microbiol. 1998; 36: 1711–15. 108 Beld M, Penning M, vann Putten M, van den Hoek A, Damen M, Klein MR, Goudsmit J. Low levels of hepatitis C virus RNA in serum, plasma, and peripheral blood mononuclear cells of injecting drug users during long antibodyundetectable periods before seroconversion. Blood 1999; 94: 1183–91. 109 Haydon GH, Jarvis LM, Blair CS, Simmonds P, Harrison DJ, Simpson KJ, Hayes PC. Clinical significance of intrahepatic hepatitis C virus levels in patients with chronic HCV infection. Gut 1998; 42: 570–5. 110 Scheuer PJ, Krawczynski K, Dhillon AP. Histopathology and detection of hepatitis C virus in liver. Springer Semin. Immunopathol. 1997; 19: 27–45. 111 Lanford RE, Sureau C, Jacob JR, White R, Fuerst TR. Demonstration of in vitro infection of chimpanzee hepatocytes with hepatitis C virus using strand-specific RT/PCR. Virology 1994; 202: 606–14. 112 Lau GK, Fang JW, Wu PC, Davis GL, Lau JY. Detection of hepatitis C virus genome in formalin-fixed paraffin-embedded liver tissue by in situ reverse transcription polymerase chain reaction. J. Med. Virol. 1994; 44: 406–9. 113 Chang M, Marquardt AP, Wood BL et al. In situ distribution of hepatitis C virus replicative-intermediate RNA in hepatic tissue and its correlation with liver disease. J. Virol. 2000; 74: 944–55. 114 Agnello V, Abel G, Knight GB, Muchmore E. Detection of widespread hepatocyte infection in chronic hepatitis C. Hepatology 1998; 28: 573–84. 115 Moldvay J, Deny P, Pol S, Brechot C, Lamas E. Detection of hepatitis C virus RNA in peripheral blood mononuclear cells of infected patients by in situ hybridization. Blood 1994; 83: 269–73.

530

AJ Freeman et al.

116 Lamas E, Baccarini P, Housset C, Kremsdorf D, Brechot C. Detection of hepatitis C virus (HCV) RNA sequences in liver tissue by in situ hybridization. J. Hepatol. 1992; 16: 219–23. 117 Qian C, Camps J, Maluenda MD, Civeira MP, Prieto J. Replication of hepatitis C virus in peripheral blood mononuclear cells. Effect of alpha-interferon therapy. J. Hepatol. 1992; 16: 380–3. 118 Nouri Aria KT, Sallie R, Sangar D et al. Detection of genomic and intermediate replicative strands of hepatitis C virus in liver tissue by in situ hybridization. J. Clin. Invest. 1993; 91: 2226–34. 119 Saleh MG, Tibbs CJ, Koskinas J et al. Hepatic and extrahepatic hepatitis C virus replication in relation to response to interferon therapy [published erratum appears in Hepatology 1995; 21: 900]. Hepatology 1993; 20: 1399–1404. 120 Lanford RE, Chavez D, Chisari FV, Sureau C. Lack of detection of negative-strand hepatitis C virus RNA in peripheral blood mononuclear cells and other extrahepatic tissues by the highly strand-specific rTth reverse transcriptase PCR. J. Virol. 1995; 69: 8079–83. 121 Zignego AL, De Carli M, Monti M et al. Hepatitis C virus infection of mononuclear cells from peripheral blood and liver infiltrates in chronically infected patients. J. Med. Virol. 1995; 47: 58–64. 122 Lerat H, Berby F, Trabaud MA, Vidalin O, Major M, Trepo C, Inchauspe G. Specific detection of hepatitis C virus minus strand RNA in hematopoietic cells. J. Clin. Invest. 1996; 97: 845–51. 123 Rodriguez-Inigo E, Casqueiro M, Navas S, Bartolome J, Pardo M, Carreno V. Fluorescent ‘in situ’ hybridization of hepatitis C virus RNA in peripheral blood mononuclear cells from patients with chronic hepatitis C. J. Med. Virol. 2000; 60: 269–74. 124 Laskus T, Radkowski M, Wang LF, Vargas H, Rakela J. Search for hepatitis C virus extrahepatic replication sites in patients with acquired immunodeficiency syndrome: specific detection of negative-strand viral RNA in various tissues. Hepatology 1998; 28: 1398–1401. 125 Smith DB, Mellor J, Jarvis LM et al. Variation of the hepatitis C virus 5′ non-coding region: implications for secondary structure, virus detection and typing. The International HCV Collaborative Study Group. J. Gen. Virol. 1995; 76: 1749–1761. 126 Tanaka T, Kato N, Cho MJ, Sugiyama K, Shimotohno K. Structure of the 3′ terminus of the hepatitis C virus genome. J. Virol. 1996; 70: 3307–12. 127 Kolykhalov AA, Mihalik K, Feinstone SM, Rice CM. Hepatitis C virus-encoded enzymatic activities and conserved RNA elements in the 3′ nontranslated region are essential for virus replication in vivo. J. Virol. 2000; 74: 2046–51. 128 Okamoto H, Kurai K, Okada S et al. Full-length sequence of a hepatitis C virus genome having poor homology to reported isolates: comparative study of four distinct genotypes. Virology 1992; 188: 331–41. 129 Weiner AJ, Brauer MJ, Rosenblatt J et al. Variable and hypervariable domains are found in the regions of HCV corresponding to the flavivirus envelope and NS1 proteins and the pestivirus envelope glycoproteins. Virology 1991; 180: 842–8. 130 Penin F, Combet C, Germanidis G, Frainais P, Deleage G, Pawlotsky J. Conformational conservation and basic character of the hypervariable region (HVR1) of hepatitis C virus (HCV) E2 envelope glycoprotein. Clues for HVR1 biological role. (abstract AO 05). In: 7th International Meeting on Hepatitis C and Related Viruses.

131 Holland J, Spindler K, Horodyski F, Grabau E, Nichol S, VandePol S. Rapid evolution of RNA genomes. Science 1982; 215: 1577–85. 132 Ogata N, Alter HJ, Miller RH, Purcell RH. Nucleotide sequence and mutation rate of the H strain of hepatitis C virus. Proc. Natl Acad. Sci. USA 1991; 88: 3392–96. 133 Major ME, Mihalik K, Fernandez J et al. Long-term follow-up of chimpanzees inoculated with the first infectious clone for hepatitis C virus. J. Virol. 1999; 73: 3317–25. 134 Martell M, Esteban JI, Quer J et al. Hepatitis C virus (HCV) circulates as a population of different but closely related genomes: quasispecies nature of HCV genome distribution. J. Virol. 1992; 66: 3225–9. 135 Davis GL. Hepatitis C virus genotypes and quasispecies. Am. J. Med. 1999; 107: S21–6. 136 Domingo E, Martinez-Salas E, Sobrino F et al. The quasispecies (extremely heterogeneous) nature of viral RNA genome populations: biological relevance – a review. Gene 1985; 40: 1–8. 137 Maggi F, Fornai C, Vatteroni ML et al. Differences in Hepatitis C virus quasispecies composition between liver, peripheral blood mononuclear cells and plasma. J. Gen. Virol. 1997; 78: 1521–5. 138 Cabot B, Martell M, Esteban JI et al. Nucleotide and amino acid complexity of hepatitis C virus quasispecies in serum and liver. J. Virol. 2000; 74: 805–11. 139 Simmonds P, Holmes EC, Cha TA et al. Classification of hepatitis C virus into six major genotypes and a series of subtypes by phylogenetic analysis of the NS-5 region. J. Gen. Virol. 1993; 74: 2391–9. 140 Tokita H, Okamoto H, Tsuda F et al. Hepatitis C virus variants from Vietnam are classifiable into the seventh, eighth, and ninth major genetic groups. Proc. Natl Acad. Sci. USA 1994; 91: 11 022–6. 141 Mellor J, Holmes EC, Jarvis LM, Yap PL, Simmonds P. Investigation of the pattern of hepatitis C virus sequence diversity in different geographical regions: implications for virus classification. The International HCV Collaborative Study Group. J. Gen. Virol. 1995; 76: 2493–507. 142 Mizokami M, Gojobori T, Ohba K et al. Hepatitis C virus types 7, 8 and 9 should be classified as type 6 subtypes. J. Hepatol. 1996; 24: 622–4. 143 White PA, Zhai X, Carter I, Zhao Y, Rawlinson WD. Simplified hepatitis C virus genotyping by heteroduplex mobility analysis. J. Clin. Microbiol. 2000; 38: 477–82. 144 Bhattacherjee V, Prescott LE, Pike I et al. Use of NS-4 peptides to identify type-specific antibody to hepatitis C virus genotypes 1, 2, 3, 4, 5 and 6. J. Gen. Virol. 1995; 76: 1737–48. 145 Lau JY, Mizokami M, Kolberg JA et al. Application of six hepatitis C virus genotyping systems to sera from chronic hepatitis C patients in the United States. J. Infect. Dis. 1995; 171: 281–9. 146 Leruez-Ville M, Nguyen QT, Cohen P, Cocco S, Nouyou M, Ferriere F, Deny P. Large-scale analysis of hepatitis C virus serological typing assay: effectiveness and limits. J. Med. Virol. 1998; 55: 18–23. 147 Chamberlain RW, Adams NJ, Taylor LA, Simmonds P, Elliott RM. The complete coding sequence of hepatitis C virus genotype 5a, the predominant genotype in South Africa. Biochem. Biophys. Res. Commun. 1997; 236: 44–9. 148 McOmish F, Yap PL, Dow BC et al. Geographical distribution of hepatitis C virus genotypes in blood donors: an international collaborative survey. J. Clin. Microbiol. 1994; 32: 884–92.

HCV immunity

149 Moaven LD, Crofts N, McCaw R, Bowden DS. The changing prevalence of hepatitis C virus types in injecting drug users between 1971 and 1994. Med. J. Aust. 1996; 165: 233. 150 Freeman AJ, Zekry A, Whybin LR et al. Hepatitis C prevalence among Australian injecting drug users in the 1970s and profiles of virus genotypes in the 1970s and 1990s. Med. J. Aust. 2000; 172: 588–91. 151 Simmonds P. Variability of hepatitis C virus. Hepatology 1995; 21: 570–83. 152 Dusheiko G, Schmilovitz-Weiss H, Brown D et al. Hepatitis C virus genotypes: an investigation of type-specific differences in geographic origin and disease. Hepatology 1994; 19: 13–18. 153 Simmonds P, Mellor J, Craxi A et al. Epidemiological, clinical and therapeutic associations of hepatitis C types in western European patients. J. Hepatol. 1996; 24: 517–24. 154 Bukh J, Miller RH, Purcell RH. Genetic heterogeneity of the hepatitis C virus. Princess Takamatsu Symposia 1995; 25: 75–91. 155 Zein NN, Poterucha JJ, Gross Jr JB et al. Increased risk of hepatocellular carcinoma in patients infected with hepatitis C genotype 1b. Am. J. Gastroenterol. 1996; 91: 2560–2. 156 Kobayashi M, Tanaka E, Sodeyama T, Urushihara A, Matsumoto A, Kiyosawa K. The natural course of chronic hepatitis C: a comparison between patients with genotypes 1 and 2 hepatitis C viruses. Hepatology 1996; 23: 695–9. 157 Silini E, Bottelli R, Asti M et al. Hepatitis C virus genotypes and risk of hepatocellular carcinoma in cirrhosis: a case-control study. Gastroenterology 1996; 111: 199–205. 158 Tanaka K, Ikematsu H, Hirohata T, Kashiwagi S. Hepatitis C virus infection and risk of hepatocellular carcinoma among Japanese: possible role of type 1b (II) infection. J. Natl Cancer Inst. 1996; 88: 742–6. 159 Nousbaum JB, Pol S, Nalpas B, Landais P, Berthelot P, Brechot C. Hepatitis C virus type 1b (II) infection in France and Italy. Collaborative Study Group. Ann. Intern. Med. 1995; 122: 161–8. 160 Lau JY, Davis GL, Prescott LE et al. Distribution of hepatitis C virus genotypes determined by line probe assay in patients with chronic hepatitis C seen at tertiary referral centers in the United States. Hepatitis Interventional Therapy Group. Ann. Intern. Med. 1996; 124: 868–76. 161 Smith DB, Davidson F, Yap P et al. Levels of hepatitis C virus in blood donors infected with different viral genotypes. International HCV Collaborative Study Group. J. Infect. Dis. 1996; 173: 727–30. 162 Romeo R, Colombo M, Rumi M et al. Lack of association between type of hepatitis C virus, serum load and severity of liver disease. J. Viral Hepat. 1996; 3: 183–90. 163 Benvegnu L, Pontisso P, Cavalletto D, Noventa F, Chemello L, Alberti A. Lack of correlation between hepatitis C virus genotypes and clinical course of hepatitis C virus-related cirrhosis. Hepatology 1997; 25: 211–15. 164 Verbaan H, Widell A, Bondeson L, Andersson K, Eriksson S. Factors associated with cirrhosis development in chronic hepatitis C patients from an area of low prevalence. J. Viral. Hepat. 1998; 5: 43–51. 165 Berg T, Hopf U, Stark K, Baumgarten R, Lobeck H, Schreier E. Distribution of hepatitis C virus genotypes in German patients with chronic hepatitis C: correlation with clinical and virological parameters. J. Hepatol 1997; 26: 484–91. 166 Haydon GH, Jarvis LM, Simmonds P, Harrison DJ, Garden OJ, Hayes PC. Association between chronic hepatitis C infection and hepatocellular carcinoma in a Scottish population. Gut 1997; 40: 128–32.

531

167 Kleter B, Brouwer JT, Nevens F et al. Hepatitis C virus genotypes: epidemiological and clinical associations. Benelux Study Group on Treatment of Chronic Hepatitis C. Liver 1998; 18: 32–8. 168 Farci P, Alter HJ, Govindarajan S et al. Lack of protective immunity against reinfection with hepatitis C virus. Science 1992; 258: 135–40. 169 Prince AM, Brotman B, Huima T, Pascual D, Jaffery M, Inchauspe G. Immunity in hepatitis C infection. J. Infect. Dis. 1992; 165: 438–43. 170 Lai ME, Mazzoleni AP, Argiolu F et al. Hepatitis C virus in multiple episodes of acute hepatitis in polytransfused thalassaemic children. Lancet 1994; 343: 388–90. 171 Rehermann B, Chang KM, McHutchison JG, Kokka R, Houghton M, Chisari FV. Quantitative analysis of the peripheral blood cytotoxic T lymphocyte response in patients with chronic hepatitis C virus infection. J. Clin. Invest. 1996; 98: 1432–40. 172 Hiroishi K, Kita H, Kojima M et al. Cytotoxic T lymphocyte response and viral load in hepatitis C virus infection. Hepatology 1997; 25: 705–12. 173 Gruber A, Lundberg LG, Bjorkholm M. Reactivation of chronic hepatitis C after withdrawal of immunosuppressive therapy. J. Intern. Med. 1993; 234: 223–25. 174 Narumi S, Tominaga Y, Tamaru M et al. Expression of IFNinducible protein-10 in chronic hepatitis. J. Immunol. 1997; 158: 5536–44. 175 Shields PL, Morland CM, Salmon M, Qin S, Hubscher SG, Adams DH. Chemokine and chemokine receptor interactions provide a mechanism for selective T cell recruitment to specific liver compartments within hepatitis C-infected liver. J. Immunol. 1999; 163: 6236–43. 176 Harvey CE, Post JJ, Kumar RK, Freeman AJ, Lloyd AR, Marinos G. IP-10 is expressed in the liver during chronic hepatitis C virus infection and correlates with lobular inflammation. In: 7th International Meeting on Hepatitis C and Related Viruses. 177 Koszinowski UH, Reddehase MJ, Jonjic S. The role of CD4 and CD8 T cells in viral infections. Curr. Opin. Immunol. 1991; 3: 471–75. 178 Mosmann TR, Sad S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol. Today 1996; 17: 138–46. 179 Botarelli P, Brunetto MR, Minutello MA et al. T-lymphocyte response to hepatitis C virus in different clinical courses of infection. Gastroenterology 1993; 104: 580–7. 180 Ferrari C, Valli A, Galati L et al. T-cell response to structural and nonstructural hepatitis C virus antigens in persistent and self-limited hepatitis C virus infections. Hepatology 1994; 19: 286–95. 181 Hoffmann RM, Diepolder HM, Zachoval R et al. Mapping of immunodominant CD4+ T lymphocyte epitopes of hepatitis C virus antigens and their relevance during the course of chronic infection. Hepatology 1995; 21: 632–8. 182 Leroux-Roels G, Esquivel CA, DeLeys R et al. Lymphoproliferative responses to hepatitis C virus core, E1, E2, and NS3 in patients with chronic hepatitis C infection treated with interferon alfa. Hepatology 1996; 23: 8–16. 183 Diepolder HM, Zachoval R, Hoffmann RM, Jung MC, Gerlach T, Pape GR. The role of hepatitis C virus specific CD4+ T lymphocytes in acute and chronic hepatitis C. J. Mol. Med. 1996; 74: 583–8. 184 Tsai SL, Liaw YF, Chen MH, Huang CY, Kuo GC. Detection of type 2-like T-helper cells in hepatitis C virus infection: implications for hepatitis C virus chronicity. Hepatology 1997; 25: 449–58.

532

AJ Freeman et al.

185 Abrignani S. Immune responses throughout hepatitis C virus (HCV) infection: HCV from the immune system point of view. Springer Semin. Immunopathol. 1997; 19: 47–55. 186 Minutello MA, Pileri P, Unutmaz D et al. Compartmentalization of T lymphocytes to the site of disease: intrahepatic CD4+ T cells specific for the protein NS4 of hepatitis C virus in patients with chronic hepatitis C. J. Exp. Med. 1993; 178: 17–25. 187 Khakoo SI, Soni PN, Savage K, Brown D, Dhillon AP, Poulter LW, Dusheiko GM. Lymphocyte and macrophage phenotypes in chronic hepatitis C infection. Correlation with disease activity. Am. J. Pathol. 1997; 150: 963–70. 188 Tran A, Yang G, Doglio A et al. Phenotyping of intrahepatic and peripheral blood lymphocytes in patients with chronic hepatitis C. Dig. Dis. Sci. 1997; 42: 2495–500. 189 Lechmann M, Ihlenfeldt HG, Braunschweiger I et al. T- and B-cell responses to different hepatitis C virus antigens in patients with chronic hepatitis C infection and in healthy antihepatitis C virus – positive blood donors without viremia. Hepatology 1996; 24: 790–5. 190 Rosen HR, Hinrichs DJ, Gretch DR et al. Association of multispecific CD4(+) response to hepatitis C and severity of recurrence after liver transplantation. Gastroenterology 1999; 117: 926–32. 191 Barrett S, Ryan E, Crowe J. Association of the HLA-DRB1*01 allele with spontaneous viral clearance in an Irish cohort infected with hepatitis C virus via contaminated anti-D immunoglobulin. J. Hepatol. 1999; 30: 979–83. 192 Congia M, Clemente MG, Dessi C et al. HLA class II genes in chronic hepatitis C virus-infection and associated immunological disorders. Hepatology 1996; 24: 1338–41. 193 Alric L, Fort M, Izopet J et al. Genes of the major histocompatibility complex class II influence the outcome of hepatitis C virus infection. Gastroenterology 1997; 113: 1675–81. 194 Cramp ME, Carucci P, Underhill J, Naoumov NV, Williams R, Donaldson PT. Association between HLA class II genotype and spontaneous clearance of hepatitis C viraemia. J. Hepatol. 1998; 29: 207–13. 195 Zavaglia C, Martinetti M, Silini E et al. Association between HLA class II alleles and protection from or susceptibility to chronic hepatitis C. J. Hepatol 1998; 28: 1–7. 196 Lechmann M, Schneider EM, Giers G, Kaiser R, Dumoulin FL, Sauerbruch T, Spengler U. Increased frequency of the HLADR15 (B1*15011) allele in German patients with self-limited hepatitis C virus infection. Eur. J. Clin. Invest. 1999; 29: 337–43. 197 Vejbaesya S, Songsivilai S, Tanwandee T, Rachaibun S, Chantangpol R, Dharakul T. HLA association with hepatitis C virus infection. Hum. Immunol. 2000; 61: 348–53. 198 Gerlach JT, Diepolder HM, Jung MC et al. Recurrence of hepatitis C virus after loss of virus-specific CD4(+) T-cell response in acute hepatitis C. Gastroenterology 1999; 117: 933–41. 199 Cramp ME, Carucci P, Rossol S, Chokshi S, Maertens G, Williams R, Naoumov NV. Hepatitis C virus (HCV) specific immune responses in anti-HCV positive patients without hepatitis C viraemia. Gut 1999; 44: 424–9. 200 Lechner F, Wong DK, Dunbar PR et al. Analysis of successful immune responses in persons infected with hepatitis C virus. J. Exp. Med. 2000; 191: 1499–512. 201 Chang KM, Thimme R, Melpolder JJ et al. Differential CD4+ and CD8+ T-cell responsiveness in hepatitis C virus infection. Hepatology 2001; 33: 267–76.

202 Missale G, Cariani E, Lamonaca V et al. Effects of interferon treatment on the antiviral T-cell response in hepatitis C virus genotype 1b- and genotype 2c-infected patients. Hepatology 1997; 26: 792–7. 203 Lasarte JJ, Garcia-Granero M, Lopez A et al. Cellular immunity to hepatitis C virus core protein and the response to interferon in patients with chronic hepatitis C. Hepatology 1998; 28: 815–22. 204 Cramp ME, Rossol S, Chokshi S, Carucci P, Williams R, Naoumov NV. Hepatitis C virus-specific T-cell reactivity during interferon and ribavirin treatment in chronic hepatitis C. Gastroenterology 2000; 118: 346–55. 205 Diepolder HM, Gerlach JT, Zachoval R et al. Immunodominant CD4+ T-cell epitope within nonstructural protein 3 in acute hepatitis C virus infection. J. Virol. 1997; 71: 6011–19. 206 Lamonaca V, Missale G, Urbani S et al. Conserved hepatitis C virus sequences are highly immunogenic for CD4(+) T cells: implications for vaccine development. Hepatology 1999; 30: 1088–98. 207 Schlesinger JJ, Brandriss MW, Putnak JR, Walsh EE. Cell surface expression of yellow fever virus non-structural glycoprotein NS1: consequences of interaction with antibody. J. Gen. Virol. 1990; 71: 593–9. 208 Halstead SB. Pathogenesis of dengue: challenges to molecular biology. Science 1988; 239: 476–81. 209 Gould EA, Buckley A. Antibody-dependent enhancement of yellow fever and Japanese encephalitis virus neurovirulence. J. Gen. Virol. 1989; 70: 1605–8. 210 Choo QL, Kuo G, Ralston R et al. Vaccination of chimpanzees against infection by the hepatitis C virus. Proc. Natl Acad. Sci. USA 1994; 91: 1294–8. 211 Farci P, Alter HJ, Wong DC et al. Prevention of hepatitis C virus infection in chimpanzees after antibody-mediated in vitro neutralization. Proc. Natl Acad. Sci. USA 1994; 91: 7792–6. 212 Harada S, Suzuki R, Ando A, Watanabe Y, Yagi S, Miyamura T, Saito I. Establishment of a cell line constitutively expressing E2 glycoprotein of hepatitis C virus and humoral response of hepatitis C patients to the expressed protein. J. Gen. Virol. 1995; 76: 1223–31. 213 Zibert A, Schreier E, Roggendorf M. Antibodies in human sera specific to hypervariable region 1 of hepatitis C virus can block viral attachment. Virology 1995; 208: 653–61. 214 Grellier L, Brown D, Power J, Dusheiko G. Absence of antienvelope antibodies and clearance of hepatitis C virus in a cohort of Irish women infected in 1977. J. Viral Hepat. 1997; 4: 379–81. 215 Bassett SE, Brasky KM, Lanford RE. Analysis of hepatitis C virus-inoculated chimpanzees reveals unexpected clinical profiles. J. Virol. 1998; 72: 2589–99. 216 Bjoro K, Froland SS, Yun Z, Samdal HH, Haaland T. Hepatitis C infection in patients with primary hypogammaglobulinemia after treatment with contaminated immune globulin. N. Engl. J. Med. 1994; 331: 1607–11. 217 Christie JM, Healey CJ, Watson J et al. Clinical outcome of hypogammaglobulinaemic patients following outbreak of acute hepatitis C: 2 year follow up. Clin. Exp. Immunol. 1997; 110: 4–8. 218 Adams G, Kuntz S, Rabalais G, Bratcher D, Tamburro CH, Kotwal GJ. Natural recovery from acute hepatitis C virus infection by agammaglobulinemic twin children. Pediatr. Infect. Dis. J. 1997; 16: 533–4. 219 Esteban JI, Lopez-Talavera JC, Genesca J et al. High rate of infectivity and liver disease in blood donors with antibodies to hepatitis C virus. Ann. Intern. Med. 1991; 115: 443–9.

HCV immunity

220 Schneiders AM, Schulz M, Leifeld L et al. E2 antibodies mediate antibody dependent cellular cytotoxicity in HCV. J. Hepatol 2000; 32 (Suppl.): 37. 221 Shimizu YK, Hijikata M, Iwamoto A, Alter HJ, Purcell RH, Yoshikura H. Neutralizing antibodies against hepatitis C virus and the emergence of neutralization escape mutant viruses. J. Virol. 1994; 68: 1494–500. 222 McAllister J, Casino C, Davidson F et al. Long-term evolution of the hypervariable region of hepatitis C virus in a commonsource-infected-cohort. J. Virol. 1998; 72: 4893–905. 223 Farci P, Shimoda A, Wong D et al. Prevention of hepatitis C virus infection in chimpanzees by hyperimmune serum against the hypervariable region 1 of the envelope 2 protein. Proc. Natl Acad. Sci. USA 1996; 93: 15 394–9. 224 Weiner AJ, Geysen HM, Christopherson C et al. Evidence for immune selection of hepatitis C virus (HCV) putative envelope glycoprotein variants: potential role in chronic HCV infections. Proc. Natl Acad. Sci. USA 1992; 89: 3468–72. 225 Kurosaki M, Enomoto N, Marumo F, Sato C. Rapid sequence variation of the hypervariable region of hepatitis C virus during the course of chronic infection. Hepatology 1993; 18: 1293–9. 226 Kato N, Sekiya H, Ootsuyama Y, Nakazawa T, Hijikata M, Ohkoshi S, Shimotohno K. Humoral immune response to hypervariable region 1 of the putative envelope glycoprotein (gp70) of hepatitis C virus. J. Virol. 1993; 67: 3923–30. 227 Kato N, Ootsuyama Y, Sekiya H, Ohkoshi S, Nakazawa T, Hijikata M, Shimotohno K. Genetic drift in hypervariable region 1 of the viral genome in persistent hepatitis C virus infection. J. Virol. 1994; 68: 4776–84. 228 Farci P, Shimoda A, Coiana A et al. The outcome of acute hepatitis C predicted by the evolution of the viral quasispecies. Science 2000; 288: 339–44. 229 Bassett SE, Thomas DL, Brasky KM, Lanford RE. Viral persistence, antibody to E1 and E2, and hypervariable region 1 sequence stability in hepatitis C virus-inoculated chimpanzees. J. Virol. 1999; 73: 1118–26. 230 Klenerman P, Lechner F, Kantzanou M, Ciurea A, Hengartner H, Zinkernagel R. Viral escape and the failure of cellular immune responses. Science 2000; 289: 2003. 231 Zibert A, Meisel H, Kraas W, Schulz A, Jung G, Roggendorf M. Early antibody response against hypervariable region 1 is associated with acute self-limiting infections of hepatitis C virus. Hepatology 1997; 25: 1245–9. 232 Barnaba V, Franco A, Alberti A, Benvenuto R, Balsano F. Selective killing of hepatitis B envelope antigen-specific B cells by class I-restricted, exogenous antigen-specific T lymphocytes. Nature 1990; 345: 258–60. 233 Planz O, Seiler P, Hengartner H, Zinkernagel RM. Specific cytotoxic T cells eliminate B cells producing virus-neutralizing antibodies [published erratum appears in Nature 1996; 384: 288]. Nature 1996; 382: 726–9. 234 Missale G, Redeker A, Person J et al. HLA-A31- and HLAAw68-restricted cytotoxic T cell responses to a single hepatitis B virus nucleocapsid epitope during acute viral hepatitis. J. Exp. Med. 1993; 177: 751–62. 235 Nayersina R, Fowler P, Guilhot S et al. HLA A2 restricted cytotoxic T lymphocyte responses to multiple hepatitis B surface antigen epitopes during hepatitis B virus infection. J. Immunol 1993; 150: 4659–71. 236 Rehermann B, Fowler P, Sidney J, Person J, Redeker A, Brown M, Moss B, Sette A, Chisari FV. The cytotoxic T lymphocyte response to multiple hepatitis B virus polymerase epitopes during and after acute viral hepatitis. J. Exp. Med. 1995; 181: 1047–58.

533

237 Penna A, Artini M, Cavalli A et al. Long-lasting memory T cell responses following self-limited acute hepatitis B. J. Clin. Invest. 1996; 98: 1185–94. 238 Stevens CE, Beasley RP, Tsui J, Lee WC. Vertical transmission of hepatitis B antigen in Taiwan. N. Engl. J. Med. 1975; 292: 771–4. 239 Schodel F, Peterson D, Milich D. Hepatitis B virus core and e antigen: immune recognition and use as a vaccine carrier moiety. Intervirology 1996; 39: 104–10. 240 Horvath J, Raffanti SP. Clinical aspects of the interactions between human immunodeficiency virus and the hepatotropic viruses. Clin. Infect. Dis. 1994; 18: 339–47. 241 Moriyama T, Guilhot S, Klopchin K et al. Immunobiology and pathogenesis of hepatocellular injury in hepatitis B virus transgenic mice. Science 1990; 248: 361–4. 242 Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 1996; 4: 25–36. 243 Guidotti LG, Rochford R, Chung J, Shapiro M, Purcell R, Chisari FV. Viral clearance without destruction of infected cells during acute HBV infection. Science 1999; 284: 825–9. 244 Guidotti LG, Chisari FV. To kill or to cure: options in host defense against viral infection. Curr. Opin. Immunol. 1996; 8: 478–83. 245 Tay CH, Welsh RM. Distinct organ-dependent mechanisms for the control of murine cytomegalovirus infection by natural killer cells. J. Virol. 1997; 71: 267–75. 246 Guidotti LG, Borrow P, Brown A, McClary H, Koch R, Chisari FV. Noncytopathic clearance of lymphocytic choriomeningitis virus from the hepatocyte. J. Exp. Med. 1999; 189: 1555–64. 247 Hill AB, Mullbacher A, Parrish C, Coia G, Westaway EG, Blanden RV. Broad cross-reactivity with marked fine specificity in the cytotoxic T cell response to flaviviruses. J. Gen. Virol. 1992; 73: 1115–23. 248 Lobigs M, Blanden RV, Mullbacher A. Flavivirus-induced up-regulation of MHC class I antigens; implications for the induction of CD8+ T-cell-mediated autoimmunity. Immunological Review 1996; 152: 5–19. 249 Kita H, Moriyama T, Kaneko T et al. HLA B44-restricted cytotoxic T lymphocytes recognizing an epitope on hepatitis C virus nucleocapsid protein. Hepatology 1993; 18: 1039–44. 250 Battegay M, Fikes J, Di Bisceglie AM et al. Patients with chronic hepatitis C have circulating cytotoxic T cells which recognize hepatitis C virus-encoded peptides binding to HLA-A2.1 molecules. J. Virol. 1995; 69: 2462–70. 251 Cerny A, McHutchison JG, Pasquinelli C et al. Cytotoxic T lymphocyte response to hepatitis C virus-derived peptides containing the HLA A2.1 binding motif. J. Clin. Invest. 1995; 95: 521–30. 252 Koziel MJ, Dudley D, Wong JT, Dienstag J, Houghton M, Ralston R, Walker BD. Intrahepatic cytotoxic T lymphocytes specific for hepatitis C virus in persons with chronic hepatitis [published erratum appears in J. Immunol. 1993; 150: 2563]. J. Immunol 1992. 149: 3339–44. 253 Erickson AL, Houghton M, Choo QL, Weiner AJ, Ralston R, Muchmore E, Walker CM. Hepatitis C virus-specific CTL responses in the liver of chimpanzees with acute and chronic hepatitis C. J. Immunol. 1993; 151: 4189–99. 254 Rehermann B, Chang KM, McHutchinson J, Kokka R, Houghton M, Rice CM, Chisari FV. Differential cytotoxic T-lymphocyte responsiveness to the hepatitis BC viruses in chronically infected patients. J. Virol. 1996; 70: 7092–102.

534

AJ Freeman et al.

255 Koziel MJ, Dudley D, Afdhal N, Choo QL, Houghton M, Ralston R, Walker BD. Hepatitis C virus (HCV)-specific cytotoxic T lymphocytes recognize epitopes in the core and envelope proteins of HCV. J. Virol. 1993; 67: 7522–32. 256 Nelson DR, Marousis CG, Davis GL, Rice CM, Wong J, Houghton M, Lau JY. The role of hepatitis C virus-specific cytotoxic T lymphocytes in chronic hepatitis C. J. Immunol. 1997; 158: 1473–81. 257 Wong DK, Dudley DD, Afdhal NH et al. Liver-derived CTL in hepatitis C virus infection: breadth and specificity of responses in a cohort of persons with chronic infection. J. Immunol. 1998; 160: 1479–88. 258 Koziel MJ, Dudley D, Afdhal N et al. HLA class I-restricted cytotoxic T lymphocytes specific for hepatitis C virus. Identification of multiple epitopes and characterization of patterns of cytokine release. J. Clin. Invest. 1995; 96: 2311–21. 259 Anthony DD, Post AB, Lehmann PV, Heeger PS. Comprehensive determinant mapping by elispot assay is capable of fully defining the HCV specific CD8 T cell repertoire in humans with self limited HCV related disease. Hepatology 2000; 32: 303A. 260 Freeman AJ, Harvey CE, Rawlinson WD, Law MG, Lloyd AR, Ffrench RA, Marinos G. Intrahepatic and peripheral blood CTL activity in patients with chronic HCV infection. In: 7th International Meeting on Hepatitis C and Related Viruses. 261 Ballardini G, Groff P, Pontisso P et al. Hepatitis C virus (HCV) genotype, tissue HCV antigens, hepatocellular expression of HLA-A,B,C, and intercellular adhesion-1 molecules. Clues to pathogenesis of hepatocellular damage and response to interferon treatment in patients with chronic hepatitis C. J. Clin. Invest. 1995; 95: 2067–75. 262 Okazaki M, Hino K, Fujii K, Kobayashi N, Okita K. Hepatic Fas antigen expression before and after interferon therapy in patients with chronic hepatitis C. Dig. Dis. Sci. 1996; 41: 2453–8. 263 Galle PR, Hofmann WJ, Walczak H et al. Involvement of the CD95 (APO-1/Fas) receptor and ligand in liver damage. J. Exp. Med. 1995; 182: 1223–30. 264 Xu XN, Screaton GR, Gotch FM et al. Evasion of cytotoxic T lymphocyte (CTL) responses by nef-dependent induction of Fas ligand (CD95L) expression on simian immunodeficiency virus-infected cells. J. Exp. Med. 1997; 186: 7–16. 265 Fiore G, Angarano I, Caccetta L, Serrone M, Jirillo E, Schiraldi O, Antonaci S. In-situ immunophenotyping study of hepatic-infiltrating cytotoxic cells in chronic active hepatitis C. Eur. J. Gastroenterol. Hepatol. 1997; 9: 491–6. 266 Krawczynski K, Beach MJ, Bradley DW et al. Hepatitis C virus antigen in hepatocytes: immunomorphologic detection and identification. Gastroenterology 1992; 103: 622–9. 267 Chang KM, Gruener NH, Southwood S, Sidney J, Pape GR, Chisari FV, Sette A. Identification of HLA-A3 and -B7restricted CTL response to hepatitis C virus in patients with acute and chronic hepatitis C. J. Immunol. 1999; 162: 1156–64. 268 He XS, Rehermann B, Lopez-Labrador FX et al. Quantitative analysis of hepatitis C virus-specific CD8(+) T cells in peripheral blood and liver using peptide-MHC tetramers. Proc. Natl Acad. Sci. USA 1999; 96: 5692–7. 269 Maini MK, Boni C, Lee CK et al. The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J. Exp. Med. 2000; 191: 1269–80. 270 Bertoletti A, Maini MK. Protection or damage: a dual role for the virus-specific cytotoxic T lymphocyte response in hepatitis B and C infection? Curr. Opin. Immunol. 2000; 12: 403–8.

271 Cooper S, Erickson AL, Adams EJ et al. Analysis of a successful immune response against hepatitis C virus. Immunity 1999; 10: 439–49. 272 Gruner NH, Gerlach TJ, Jung MC et al. Association of hepatitis C virus-specific CD8+ T cells with viral clearance in acute hepatitis C. J. Infect. Dis. 2000; 181: 1528–36. 273 Sobao Y, Tomiyama H, Nakamura S, Sekihara H, Tanaka K, Tskiguchi M. Visual demonstration of hepatitis C virusspecific memory CD8+ T-cell expansion in patients with acute hepatitis C. Hepatology 2001; 33: 287–94. 274 Cucchiarini M, Kammer AR, Grabscheid B et al. Vigorous peripheral blood cytotoxic T cell response during the acute phase of hepatitis C virus infection. Cell. Immunol. 2000; 203: 111–23. 275 Chang KM, Thimme R, Melpolder JJ et al. Differential CD4+ and CD8+ T-cell responsiveness in hepatitis C virus infection. Hepatology 2001; 33: 267–76. 276 Thimme R, Bukh J, Pemberton J, Guidotti LG, Sette A, Purcell RH, Chisari FV. Clearance of HCV infection is determined by the vigor of the antiviral T cell response and the intrahepatic cytokine profile. Hepatology 2000; 32: 385A. 277 Koziel MJ, Wong DK, Dudley D, Houghton M, Walker BD. Hepatitis C virus-specific cytolytic T lymphocyte and T helper cell responses in seronegative persons. J. Infect. Dis. 1997; 176: 859–66. 278 Bronowicki JP, Vetter D, Uhl G, Hudziak H, Uhrlacher A, Vetter JM, Doffoel M. Lymphocyte reactivity to hepatitis C virus (HCV) antigens shows evidence for exposure to HCV in HCV-seronegative spouses of HCV-infected patients. J. Infect. Dis. 1997; 176: 518–22. 279 Scognamiglio P, Accapezzato D, Casciaro MA et al. Presence of effector CD8+ T cells in hepatitis C virus-exposed healthy seronegative donors. J. Immunol. 1999; 162: 6681–89. 280 Haydon GH, Jarvis LM, Blair CS, Simmonds P, Harrison DJ, Simpson KJ, Hayes PC. Clinical significance of intrahepatic hepatitis C virus levels in patients with chronic HCV infection. Gut 1998; 42: 570–5. 281 Kammer AR, van der Burg SH, Grabscheid B et al. Molecular mimicry of human cytochrome P450 by hepatitis C virus at the level of cytotoxic T cell recognition. J. Exp. Med. 1999; 190: 169–76. 282 Chang KM, Rehermann B, McHutchison JG, Pasquinelli C, Southwood S, Sette A, Chisari FV. Immunological significance of cytotoxic T lymphocyte epitope variants in patients chronically infected by the hepatitis C virus. J. Clin. Invest. 1997; 100: 2376–85. 283 Weiner A, Erickson AL, Kansopon J et al. Persistent hepatitis C virus infection in a chimpanzee is associated with emergence of a cytotoxic T lymphocyte escape variant. Proc. Natl Acad. Sci. USA 1995; 92: 2755–9. 284 Bertoletti A, Sette A, Chisari FV et al. Natural variants of cytotoxic epitopes are T-cell receptor antagonists for antiviral cytotoxic T cells. Nature 1994; 369: 407–10. 285 Kaneko T, Moriyama T, Udaka K et al. Impaired induction of cytotoxic T lymphocytes by antagonism of a weak agonist borne by a variant hepatitis C virus epitope. Eur. J. Immunol. 1997; 27: 1782–7. 286 Tsai SL, Chen YM, Chen MH et al. Hepatitis C virus variants circumventing cytotoxic T lymphocyte activity as a mechanism of chronicity. Gastroenterology 1998; 115: 954–65. 287 Smith DB, McAllister J, Casino C, Simmonds P. Virus ‘quasispecies’: making a mountain out of a molehill? J. Gen. Virol. 1997; 78: 1511–19.

HCV immunity

288 Cerny A, Chisari FV. Pathogenesis of chronic hepatitis C: immunological features of hepatic injury and viral persistence. Hepatology 1999; 30: 595–601. 289 Farci P, Bukh J, Purcell RH. The quasispecies of hepatitis C virus and the host immune response. Springer Semin. Immunopathol. 1997; 19: 5–26. 290 Song MK, Lee SW, Suh YS, Lee KJ, Sung YC. Enhancement of immunoglobulin G2a and cytotoxic T-lymphocyte responses by a booster immunization with recombinant hepatitis C virus E2 protein in E2 DNA-primed mice. J. Virol. 2000; 74: 2920–5. 291 Forns X, Payette PJ, Ma X et al. Vaccination of chimpanzees with plasmid DNA encoding the hepatitis C virus (HCV) envelope E2 protein modified the infection after challenge with homologous monoclonal HCV. Hepatology 2000; 32: 618–25. 292 Pancholi P, Liu Q, Tricoche N, Zhang P, Perkus ME, Prince AM. DNA prime-canarypox boost with polycistronic hepatitis C virus (HCV) genes generates potent immune responses to HCV structural and nonstructural proteins. J. Infect. Dis. 2000; 182: 18–27. 293 Woitas RP, Lechmann M, Jung G, Kaiser R, Sauerbruch T, Spengler U. CD30 induction and cytokine profiles in hepatitis C virus core-specific peripheral blood T lymphocytes. J. Immunol. 1997; 159: 1012–18. 294 Reiser M, Marousis CG, Nelson DR, Lauer G, GonzalezPeralta RP, Davis GL, Lau JY. Serum interleukin 4 and interleukin 10 levels in patients with chronic hepatitis C virus infection. J. Hepatol. 1997; 26: 471–8. 295 Kuzushita N, Hayashi N, Katayama K et al. High levels of serum interleukin-10 are associated with a poor response to interferon treatment in patients with chronic hepatitis C. Scand. J. Gastroenterol. 1997; 32: 169–74. 296 Cacciarelli TV, Martinez OM, Gish RG, Villanueva JC, Krams SM. Immunoregulatory cytokines in chronic hepatitis C virus infection: pre- and post-treatment with interferon alfa. Hepatology 1996; 24: 6–9. 297 Klenerman P, Gruener N, Urbani S et al. Success and failure of antiviral CD8+ responses monitored by class I responses. In: 7th International Meeting on Hepatitis C and Related Viruses. 298 Large MK, Kittlesen DJ, Hahn YS. Suppression of host immune response by the core protein of hepatitis C virus: possible implications for hepatitis C virus persistence. J. Immunol. 1999; 162: 931–8. 299 Eckels DD, Tabatabail N, Bian TH et al. In vitro human Th-cell responses to a recombinant hepatitis C virus antigen: failure in IL-2 production despite proliferation. Hum. Immunol. 1999; 60: 187–99. 300 Eckels DD, Wang H, Bian TH, Tabatabai N, Gill JC. Immunobiology of hepatitis C virus (HCV) infection: the role of CD4 T cells in HCV infection. Immunol. Rev. 2000; 174: 90–7. 301 Martin J, Quiroga JA, Navas S, Pardo M, Carreno V. Modulation by biologic response modifiers of hepatitis C virus antigen-independent cytokine secretion in blood mononuclear cells. Cytokine 1999; 11: 267–73. 302 Chang KM, Rehermann B, Chisari FV. Immunopathology of hepatitis C. Springer Semin. Immunopathol. 1997; 19: 57–68. 303 Koziel MJ. Cytokines in viral hepatitis. Semin. Liver Dis. 1999; 19: 157–69. 304 Zhu N, Khoshnan A, Schneider R, Matsumoto M, Dennert G, Ware C, Lai MM. Hepatitis C virus core protein binds to the cytoplasmic domain of tumor necrosis factor (TNF) receptor 1 and enhances TNF-induced apoptosis. J. Virol. 1998; 72: 3691–7.

535

305 Bertoletti A, D’Elios MM, Boni C et al. Different cytokine profiles of intraphepatic T cells in chronic hepatitis B and hepatitis C virus infections. Gastroenterology 1997; 112: 193–9. 306 Napoli J, Bishop GA, McGuinness PH, Painter DM, McCaughan GW. Progressive liver injury in chronic hepatitis C infection correlates with increased intrahepatic expression of Th1-associated cytokines. Hepatology 1996; 24: 759–65. 307 Fukuda R, Ishimura N, Ishihara S et al. Intrahepatic expression of pro-inflammatory cytokine mRNAs and interferon efficacy in chronic hepatitis C. Liver 1996; 16: 390–9. 308 Ando K, Hiroishi K, Kaneko T et al. Perforin, Fas/Fas ligand, and TNF-alpha pathways as specific and bystander killing mechanisms of hepatitis C virus-specific human CTL. J. Immunol. 1997; 158: 5283–91. 309 Hata K, Van Thiel DH, Herberman RB, Whiteside TL. Phenotypic and functional characteristics of lymphocytes isolated from liver biopsy specimens from patients with active liver disease. Hepatology 1992; 15: 816–23. 310 Doherty DG, Norris S, Madrigal-Estebas L, McEntee G, Traynor O, Hegarty JE, Farrelly C. The human liver contains multiple populations of NK cells, T cells, and CD3+CD56+ natural T cells with distinct cytotoxic activities and Th1, Th2, and Th0 cytokine secretion patterns. J. Immunol. 1999; 163: 2314–21. 311 Godfrey DI, Hammond KJL, Poulton LD, Smyth MJ, Baxter AG. NKT cells: facts, functions and fallacies. Immunol. Today 2000; 21: 573–83. 312 McIntyre KW, Welsh RM. Accumulation of natural killer and cytotoxic T large granular lymphocytes in the liver during virus infection. J. Exp. Med. 1986; 164: 1667–81. 313 Corado J, Toro F, Rivera H, Bianco NE, Deibis L, De Sanctis JB. Impairment of natural killer (NK) cytotoxic activity in hepatitis C virus (HCV) infection. Clin. Exp. Immunol. 1997; 109: 451–7. 314 Leroy V, Vigan I, Baud M, Jouvin-Marche E, Zarski J-P, Marche P. Phenotypic and functional characteristics of intrahepatic lymphocytes during chronic hepatitis C. Hepatology 2000; 32: 267A. 315 Nuti S, Rosa D, Valiante NM et al. Dynamics of intra-hepatic lymphocytes in chronic hepatitis C: enrichment for Valpha24+ T cells and rapid elimination of effector cells by apoptosis. Eur. J. Immunol. 1998; 28: 3448–55. 316 Pernollet M, Leroy V, Vigan I, Zarski J-P, Marche PN, JouvinMarche E. Relation between CD3+CD56- lymphocytes and liver damage in chronic hepatitis C. In: 7th International Meeting on Hepatitis C and Related Viruses. 317 Starzl TE, Iwatsuki S, Van Thiel DH et al. Evolution of liver transplantation. Hepatology 1982; 2: 614–36. 318 Crispe IN, Dao T, Klugewitz K, Mehal WZ, Metz DP. The liver as a site of T-cell apoptosis: graveyard, or killing field? Immunol. Rev. 2000; 174: 47–62. 319 Lechler R, Marelli-Berg FM. Mechanisms of peripheral T-cell tolerance. J. Viral. Hepat. 1997; 4: 1–5. 320 Huang L, Fowler N, Schmidt EV, Koziel M. Enhanced apoptosis of activated T cells induced by hepatocytes expressing HCV structural proteins. Hepatology 2000; 32: 303A. 321 Chen CM, You LR, Hwang LH, Lee YH. Direct interaction of hepatitis C virus core protein with the cellular lymphotoxinbeta receptor modulates the signal pathway of the lymphotoxinbeta receptor. J. Virol. 1997; 71: 9417–26. 322 Taylor DR, Shi ST, Romano PR, Barber GN, Lai MMC. Inhibition of the interferon-inducible protein kinase PKR by HCV E2 protein. Science 1999; 285: 107–10.

536

AJ Freeman et al.

323 Gale Jr M, Kwieciszewski B, Dossett M, Nakao H, Katze MG. Anti-apoptotic and oncogenic potentials of hepatitis C virus are linked to interferon resistance by viral repression of the PKR protein kinase. J. Virol. 1999; 73: 6506–16. 324 Kittleson DJ, Chianese-Bullock KA, Yao ZQ, Braciale TJ, Hahn YS. Interactions between complement receptor gC1qR and hepatitis C virus core protein inhibits T-lymphocyte. J. Clin. Invest. 2000; 106: 1239–49. 325 Koup RA. Virus escape from CTL recognition. J. Exp. Med. 1994; 180: 779–82. 326 Rehermann B, Chisari FV. Cell mediated immune response to the hepatitis C virus. Curr. Microbiol. Immunol. 2000; 242: 299–325. 327 Sansonno D, Lotesoriere C, Cornacchiulo V, Fanelli M, Gatti P, Iodice G, Racanelli V, Dammacco F. Hepatitis C virus infection involves CD34(+) hematopoietic progenitor cells in hepatitis C virus chronic carriers. Blood 1998; 92: 3328–37. 328 Bain C, Fatmi A, Zoulim F, Zarski JP, Trepo C, Inchauspe G. Impaired allostimulatory function of dendritic cells in chronic hepatitis C infection. Gastroenterology 2001; 120: 512–24. 329 Kanto T, Hayashi N, Takehara T et al. Impaired allostimulatory capacity of peripheral blood dendritic cells recovered from hepatitis C virus-infected individuals. J. Immunol. 1999; 162: 5584–91. 330 Farci P, Alter HJ, Shimoda A et al. Hepatitis C virus-associated fulminant hepatic failure. N. Engl. J. Med. 1996; 335: 631–4. 331 Gretch DR, Bacchi CE, Corey L et al. Persistent hepatitis C virus infection after liver transplantation: clinical and virological features. Hepatology 1995; 22: 1–9. 332 Kato N, Yokosuka O, Hosoda K, Ito Y, Ohto M, Omata M. Quantification of hepatitis C virus by competitive reverse transcription-polymerase chain reaction: increase of the virus in advanced liver disease. Hepatology 1993; 18: 16–20. 333 Gordon SC, Kodali VP, Silverman AL, Dmuchowski CF, Urdea MS, Chan CS, Wilber JC. Levels of hepatitis C virus RNA and liver histology in chronic type C hepatitis. Am. J. Gastroenterol. 1994; 89: 1458–61. 334 Gretch D, Corey L, Wilson J et al. Assessment of hepatitis C virus RNA levels by quantitative competitive RNA polymerase chain reaction: high-titer viremia correlates with advanced stage of disease. J. Infect. Dis. 1994; 169: 1219–25. 335 Gonzalez-Peralta RP, Davis GL, Lau JY. Pathogenetic mechanisms of hepatocellular damage in chronic hepatitis C virus infection. J. Hepatol 1994; 21: 255–9. 336 Adinolfi LE, Andreana A, Utili R, Zampino R, Ragone E, Ruggiero G. HCV RNA levels in serum, liver, and peripheral blood mononuclear cells of chronic hepatitis C patients and their relationship to liver injury. Am. J. Gastroenterol. 1998; 93: 2162–2166. 337 Idrovo V, Dailey PJ, Jeffers LJ et al. Hepatitis C virus RNA quantification in right and left lobes of the liver in patients with chronic hepatitis C. J. Viral Hepat. 1996; 3: 239–246. 338 McGuinness PH, Bishop GA, Painter DM, Chan R, McCaughan GW. Intrahepatic hepatitis C RNA levels do not correlate with degree of liver injury in patients with chronic hepatitis C. Hepatology 1996; 23: 676–687.

339 De Moliner L, Pontisso P, De Salvo GL, Cavalletto L, Chemello L, Alberti A. Serum and liver HCV RNA levels in patients with chronic hepatitis C: correlation with clinical and histological features. Gut 1998; 42: 856–60. 340 Rodriguez-Inigo E, Bartolome J, De Lucas S et al. Histological damage in chronic hepatitis C is not related to the extent of infection in the liver. Am. J. Pathol 1999; 154: 1877–81. 341 Negro F, Krawczynski K, Quadri R, Rubbia-Brandt L, Mondelli M, Zarski JP, Hadengue A. Detection of genomicminus-strand of hepatitis C virus RNA in the liver of chronic hepatitis C patients by strand-specific semiquantitative reversetranscriptase polymerase chain reaction. Hepatology 1999; 29: 536–42. 342 Pasquinelli C, Shoenberger JM, Chung J et al. Hepatitis C virus core and E2 protein expression in transgenic mice. Hepatology 1997; 25: 719–27. 343 Brillanti S, Foli M, Gaiani S, Masci C, Miglioli M, Barbara L. Persistent hepatitis C viraemia without liver disease. Lancet 1993; 341: 464–5. 344 Honda M, Kaneko S, Sakai A, Unoura M, Murakami S, Kobayashi K. Degree of diversity of hepatitis C virus quasispecies and progression of liver disease. Hepatology 1994; 20: 1144–51. 345 Koizumi K, Enomoto N, Kurosaki M, Murakami T, Izumi N, Marumo F, Sato C. Diversity of quasispecies in various disease stages of chronic hepatitis C virus infection and its significance in interferon treatment. Hepatology 1995; 22: 30–35. 346 Yuki N, Hayashi N, Moribe T et al. Relation of disease activity during chronic hepatitis C infection to complexity of hypervariable region 1 quasispecies. Hepatology 1997; 25: 439–44. 347 Hayashi J, Kishihara Y, Yamaji K et al. Hepatitis C viral quasispecies and liver damage in patients with chronic hepatitis C virus infection. Hepatology 1997; 25: 697–701. 348 Naito M, Hayashi N, Moribe T et al. Hepatitis C viral quasispecies in hepatitis C virus carriers with normal liver enzymes and patients with type C chronic liver disease. Hepatology 1995; 22: 407–412. 349 Gonzalez-Peralta RP, Qian K, She JY, Davis GL, Ohno T, Mizokami M, Lau JY. Clinical implications of viral quasispecies heterogeneity in chronic hepatitis C. J. Med. Virol. 1996; 49: 242–7. 350 Lopez-Labrador FX, Ampurdanes S, Gimenez-Barcons M et al. Relationship of the genomic complexity of hepatitis C virus with liver disease severity and response to interferon in patients with chronic HCV genotype 1b infection [published erratum appears in Hepatology 1999; 29: 1915]. Hepatology 1999; 29: 897–903. 351 Sullivan DG, Wilson JJ, Carithers Jr RL, Perkins JD, Gretch DR. Multigene tracking of hepatitis C virus quasispecies after liver transplantation: correlation of genetic diversification in the envelope region with asymptomatic or mild disease patterns. J. Virol. 1998; 72: 10 036–43. 352 Liang TJ, Rehermann B, Seeff LB, Hoofnagle JH. Pathogenesis, natural history, treatment, and prevention of hepatitis C. Ann. Intern. Med. 2000; 132: 296–305.

Suggest Documents