icon star paper   Articles  
Back grey_arrow_rt.gif
 
 
The differential immune responses during HBV and HCV infection: mechanisms of the diseases and response
 
 
  Hepatology July 2003, Vol 38, Number 1
 
Antonio Bertoletti1, Carlo Ferrari2. 1Institute of Hepatology, University College London, Royal Free and University College of London Medical School, London, UK; and the 2Divisione Malattie Infettive ed Epatologia, Azienda Ospedaliera di Parma, Parma, Italy.
 
The innate immune system has a role not only in protecting the host during the initial period of virus infection, but also in shaping the nature of the adaptive immune response. In this review, we follow the kinetics of the virologic and immunologic events occurring from the time of hepatitis B virus (HBV) and hepatitis C virus (HCV) infection. We primarily discuss how the early events after infection might influence the development of the adaptive immune response in these 2 important viral infections and how new strategies for more efficient preventive and therapeutic vaccines can be derived from this knowledge.
 
Hepatitis B (HBV) and hepatitis C (HCV) viruses are the 2 major causes of chronic liver inflammation worldwide.1,2 Despite distinct virologic features, both viruses are preferentially hepatotropic, not directly cytopathic, and elicit liver diseases that share several aspects of their natural history. HBV and HCV infections share also some important features of the adaptive immune response. Multispecific antiviral CD4 and CD8 responses with a T-helper type 1 profile of cytokine production are detectable in the blood of subjects with a self-limited infection.3-16 These responses are stronger than those found in patients with chronic infection.17-21 Based on these observations, it has been proposed that the ability to mount an efficient cellular immune response is the main mechanism responsible for HBV and HCV control, whereas a defect in this response leads to chronicity.2,22 However, the lack of suitable animal models to study the pathogenesis of HBV and HCV infections has so far hampered a definitive demonstration that associations between different infection outcomes and different vigor and breadth of T-cell responses have a causative effect. A recent report in HCV-infected chimpanzees, in which a clear association between T-cell response and virus clearance was not found,23 added a further note of caution. Even so, it is difficult to argue against the importance of the cellular immune response in HBV and HCV control. In chimpanzees infected with HCV, expansion of a multispecific and sustained HCV-specific CD8+ T-cell-mediated response was observed in 2 of 2 animals that cleared the virus, but not in the 4 animals that developed a chronic infection.24 These results have been confirmed by recent reports showing that expansion of interferon (IFN-)+, CD8+, and CD4+ T cells precedes viral clearance in patients studied during the incubation phase of acute HCV infection in man10 and in HCV-infected chimpanzees.25 Similar findings have been reported in animal models of HBV infection26 and in patients studied during the incubation phase of HBV infection,27 in whom reduced early expansion of virus-specific T cells was associated with virus persistence. Moreover, the importance of virus-specific CD8+ T cells in HBV control has been confirmed further by CD8+ T-cell deletion experiments performed in HBV-infected chimpanzees.28
 
The humoral immune response plays also an essential role during HBV and HCV infections. HBV and HCV clearances are associated temporally with production of anti-envelope antibodies29,30 and sera with high levels of antiviral antibodies (specific for the viral envelope) can prevent HBV31 and HCV32 infections. Therefore, the integrated activation of both the cellular and humoral arms of the adaptive immune response appears to ultimately allow control of both infections. The different components of the adaptive immune system are so interconnected that the failure of one of them clearly affects the expansion and protective efficacy of the others. Lack of CD4+ T-cell help can impair CD8+ T-cell activity and antibody production,33 whereas the inability to mount a virus-specific CD8+ T-cell response results in a level of circulating virus that cannot be cleared by antibodies alone.34 Thus, instead of making an artificial hierarchy of the most important protective arms of the immune response, we will try to dissect the mechanisms that regulate the induction of a fully integrated innate and adaptive immune response from the early stages of HBV and HCV infection.
 
Profiles of adaptive immune responses early after HBV and HCV infections
 
The viral and immunologic events that occur in the first weeks after HBV and HCV infections are likely to influence substantially the final outcome of infection. Nevertheless, a comprehensive immunologic analysis during this period has been performed so far in a very limited number of patients.10,27,35 In addition, the study of the CD8-mediated T-cell response has been restricted to few immunodominant epitopes.10,27 Despite these limitations, it is interesting to note that available studies in human and animal models of HBV and HCV infection appear to be consistent with the interpretation that HBV and HCV are ignored by the adaptive immune response for about 2 months after primary infection. After needle-stick exposure to HCV-positive blood, patients who subsequently controlled the infection showed antiviral-specific CD8+ and CD4+ T-cell responses only after 7 to 10 weeks from exposure.10 The appearance of HBV-specific CD8+ and CD4+ T cells during the incubation time of acute hepatitis B follows the same pattern.27 Activation of the virus-specific cellular immunity is followed by the humoral response, which appears at least 10 to 12 weeks after HBV36 and HCV37 infections. These data are in line with results obtained in animal models. During primary HCV infection of chimpanzees, virus-specific antibody responses are detectable 8 to 12 weeks after infection and HCV-specific T-cell responses appear with identical delays.25,38-41 This long interval of time between infection and induction of the adaptive immunity to HBV and HCV seems to contrast with the reported kinetics of virus-specific T-cell responses to other virus infections. Although comparison of different studies must be interpreted with caution because of possible sensitivity variations in the assays, it is intriguing that although T-cell responses to simian immunodeficiency virus, human immunodeficiency virus, and cytomegalovirus already can be detectable 2 weeks after infection,42,43 this finding is only occasional in HCV-infected chimpanzees because only 10 of the 12 animals reported in the literature24,25,38-41 showed a clear HCV-specific T-cell response by 4 weeks after infection.
 
The pathogenetic implications of the apparent delay of virus-specific adaptive immune responses are likely to be different in HCV and HBV infections. The methods used for viral quantification are often different, implying a careful interpretation of the results, yet available data appear to be consistent with a different kinetic pattern of HBV and HCV replication (see Fig. 1). While HCV-RNA levels increase rapidly in the first few days of HCV infection,10,25,38,41,44 HBV-DNA and HBV antigens are not detectable in serum and liver for about 4 to 7 weeks after HBV infection,36,45,46 even using methods with a level of sensitivity comparable with the ones used for HCV detection.28 Thus, in HCV infection the adaptive response seems to really ignore for several weeks a substantial quantity of virus (at least 106 copies/mL), whereas the limited quantity of HBV antigens in the early phases of HBV infection is likely responsible for the delayed induction of the HBV-specific adaptive immunity.
 
In addition to the late detection, quantitative and functional defects of the adaptive immune responses are present during primary HBV and HCV infections. The peak frequency of HBV-specific CD8+ T cells during the acute phase of disease in patients with self-limited infections generally does not exceed 1% of circulating CD8+ T cells.47 Higher frequencies (up to 7% of CD8+ T cells) have been observed in acute HCV-infected patients, but most of these cells show impaired production of IFN-, low perforin content and defective capacity for expansion, and lysis of target cells.16,48 These functional defects are detectable in the acutely infected population, irrespective of the subsequent outcome toward recovery or chronic viral persistence, and are transient only in patients who succeed in clearing the virus spontaneously.48-50
 
Interestingly, CD8+ T-cell frequencies in patients acutely infected with HCV and HBV are generally lower than those observed during acute Epstein-Barr virus or human immunodeficiency virus infections, where virus-specific IFN--producing CD8+ T cells easily can represent up to 10% of the circulating CD8+ T-cell population, with some cases in which single epitope-specific CD8+ T cells can account for more than 40% of the total CD8+ T-cell pool.51,52 The possibility that this quantitative difference reflects the use of tetramers able to detect CD8+ T cells restricted to selected human leukocyte antigen class I molecules cannot be excluded totally,53 but recent analysis performed using broad peptide libraries did not detect higher HCV-specific CD8+ T-cell frequencies.53,54 In addition, quantitative defects are not limited to the cellular arm of the adaptive response because antibody production of limited quantity and restricted to the immunoglobulin G1 isotype has been reported in acute HCV infections.37 Several factors likely are responsible for these defects of the adaptive immunity, including intrahepatic recruitment and subsequent deletion of activated virus-specific CD8+ T cells,55 antigen-driven T- or B-cell exhaustion,56 the ability of liver sinusoidal endothelial cells to suppress the expansion of T-helper 1-type cells,57 and the production of viral proteins with immunomodulatory effects.58-60 In addition, late appearance, reduced size, and functional T-cell defects detected in both primary HBV and HCV infections also might be a consequence of the inability of the innate immune response to provide an efficient containment of early virus replication and to promote timely and appropriate T-cell priming. Do available data allow support of this concept?
 
The innate immune response in HCV and HBV infections
 
One of the first defense mechanisms that an infected host is able to mount against viral infections is the production of type I IFNs. In addition to antiviral functions, these cytokines possess important immunomodulatory effects.61 The contribution and importance of type I IFNs have been deduced primarily from mouse models of different viral infections (lymphocytic choriomeningitis virus and murine cytomegalovirus).61,62 However, mice cannot be infected by HBV or HCV, and indications about the role of type I IFNs derive exclusively from recent studies in the chimpanzee model of HCV infection. Changes in the transcription of IFN- response genes have been shown to occur as early as 2 days after HCV infection in the liver of a chimpanzee acutely infected with HCV and these changes parallel the viral replication kinetics.63,64 HCV starts to replicate immediately after infection and viremia generally is detectable 1 to 2 weeks after inoculation in humans10 and within a few days in chimpanzees (Fig. 1).38,41,44 It is likely that HCV replication is the direct cause of type I IFN production because double-stranded RNA is a strong IFN- inducer.65 Although HCV is sensitive in vitro to IFNs,66,67 recent data raise the hypothesis that the virus may have developed strategies to interfere with the IFN type I antiviral activity.68 For example, the HCV E2 protein can inhibit in vitro the kinase activity of double-stranded RNA-activated protein kinase, one of the antiviral proteins induced by IFNs.69 Because of this interference, the initial production of type I IFNs only may be able to slow, but not block, virus replication. HCV viral kinetics are in line with this possibility. The initial rapid and early peak of viral replication can be followed by a period of about 4 to 6 weeks when HCV RNA can increase slowly or remain at stable levels10,38,41,44,63,64 in the absence of virus-specific T- and B-cell induction and liver inflammation.
 
The coexistence of HCV replication with the absence of signs of liver inflammation is not only indicative of the noncytopathic nature of HCV, but also calls into question the pathogenic contribution of natural killer (NK) and NK-T cells in HCV infection. NK and NK-T cells are components of the innate immune system that respond within minutes or hours after infection by producing IFN- and by killing infected cells.70 Although NK-T cells are a resident cellular population of the liver (high frequency of around 20% in mice and around 1% in humans),70 the majority of NK cells need to be recruited actively from the spleen.70 IFN- production by these cells not only has antiviral effects but also mediates the intrahepatic recruitment of inflammatory cells.71,72 Direct evidence of the activation state of NK and NK-T cells during the initial phase of HCV infection is lacking. Nevertheless, the absence of signs of liver inflammation in the first 4 to 6 weeks after HCV infection10,38,41,44,63 suggests that the contribution of NK and/or NK-T cells in this early phase may be minimal. This possibility also is supported by recent data showing that the HCV envelope protein E2 can inhibit natural killer cell functions directly73,74 by interacting with CD81, a tetraspannin molecule expressed on the surface of different cell lineages that is a putative HCV receptor.75 The direct inhibitory effect of the HCV envelope protein on global NK cell function may contribute to the absence of liver inflammation, despite the early signs of HCV replication and the early production of IFN-, which has been shown to promote NK cell recruitment into the liver.76 In line with this possibility, results obtained with DNA micro-array analysis of the chimpanzee liver63 show that the expression levels of IFN regulatory factor-1, a transcription factor essential for NK cell activity in vivo,77 does not change during acute HCV infection.
 
Is the pattern of innate immune response activation similar in HBV infection? As we have discussed previously, available data show a different replicative pattern after HBV and HCV infections . After an apparent initial quiescent phase of about 4 to 7 weeks,28,36,45,46 HBV starts to replicate vigorously, reaching levels of around 109 to 1010 copies/mL in 1 to 2 weeks,27,78 and infecting most hepatocytes.28,79-81 In animal models of HBV infection, this rapid peak of viral replication is accompanied in self-limited infection by a typical acute phase of disease with IFN- and TNF- production, followed by the triggering of the adaptive immunity.28,81 Animals that develop chronicity lack the acute phase response and fail to prime an adequate adaptive response.26,82,83 Thus, activation of innate components of the immune system seems to represent a key element in the control of the initial HBV burst. Available data do not allow the contribution of the individual components of the innate immunity to be dissected out. However, their overall efficiency is remarkable because the HBV-DNA quantity decreases by more than 80% within 2 to 3 weeks after the peak of viral replication,79,80 and before the peak of the antigen-specific CD8 response and liver damage.27,28,81
 
Is this initial IFN- production primarily sustained by NK and NK-T cells? Although we lack direct evidence of the role of NK and NK-T cells during natural HBV infection, a number of recent reports in animal models may be consistent with this hypothesis. NK-T cells in the transgenic mouse model of HBV infection have been shown to inhibit virus replication through the production of IFN-.72,84 In this system, the activation of NK-T cells was a consequence of -galactoceramide stimulation rather than a response to the natural infection. Nevertheless, recent results indicate that a population of nonclassic NK-T cells can be activated directly when injected into mice expressing HBV antigens in the liver.85 Thus, NK-T cells potentially could be triggered during natural HBV infection, by direct recognition of viral antigens, or, possibly, by the expression of stress signals either by infected hepatocytes or by liver dendritic cells. Strong evidence that NK and NK-T cells actually contribute to the initial control of HBV replication derives from work in acutely infected chimps. A rapid drop in viral replication occurs in the presence of intrahepatic IFN- production, before the massive intrahepatic recruitment of T cells. In this setting, NK or resident NK-T cells seem to be the principal source of IFN- production causing rapid inhibition of HBV replication and recruitment of virus-specific and nonspecific cells.72,86 A similar sequence of events has been reported in patients studied during the incubation phase of hepatitis B. An increased number of circulating NK cells precedes the peak of HBV replication, and is followed, 2 to 4 weeks later, by the appearance of HBV-specific CD8+ T cells, when viral replication has already dropped.27
 
Role of the dendritic cells in virus control
 
Dendritic cells (DCs) act as a link between innate and adaptive immunity. Signals delivered by the innate immune system (type I IFN production, interactions with NK cells) lead to the proper maturation of DCs, which are critical for triggering the antigen-specific immune response.87 The ability of DCs to induce T-cell responses also is influenced by their tissue localization, the antigen dose, and the interaction with helper T cells.87 Activation of cytotoxic T cells in lymph nodes draining the site of infection occurs hours after the initial challenge88 and is mediated by DCs. CD8+ T cells (but also CD4+ T cells) stimulated for only 24 hours by mature DCs proliferate extensively.89
 
Given the crucial role played by DCs in T-cell priming, we raise the hypothesis that an alteration of the DC function and/or maturation during primary HCV infection can contribute to the delayed appearance of HCV-specific CD8+ and CD4+ T cells after infection. Although no conclusive data are available about the function of DCs during acute or chronic HCV infection, it has been shown in vitro that the E2 protein of HCV can inhibit NK cell function directly.73,74 Because NK cell activation contributes to DC maturation,90 it is possible to speculate that this inhibitory effect may influence indirectly the function of DCs to trigger the adaptive response. Furthermore, experiments performed with monocyte-derived DCs indicate that these cells show an altered maturation process in chronic HCV infection,91-93 which may be related to the presence of HCV93 or to the effect of HCV proteins.94 Thus, it is possible, even though not directly proven, that the ability of DCs to pick up HCV antigens, to migrate to draining lymph nodes, or to induce proper T-cell activation is influenced by the high HCV levels present also during the early phase of HCV infection.
 
The situation seems to be different in primary HBV infection. Infection of DCs by HBV is even more controversial than in HCV, and the described functional defects are minimal.95-97 The delayed appearance of HBV-specific immune responses after primary infection seems to depend more on the kinetics of HBV replication (i.e., HBV persists for at least 5-8 weeks in the newly infected host without detectable signs of efficient replication), than on immune dysfunctions. The evidence that HBV-specific CD4+ and CD8+ T cells become detectable a short time after the exponential increase in HBV replication27,28 suggests that the DC function is not affected by the virus at the early phases of self-limited HBV infection.
 
The collapse of the adaptive immunity
 
When resolution of HBV and HCV infections is achieved, virus-specific antibodies as well as multispecific and T-helper 1-oriented CD4- and CD8-mediated T-cell responses remain detectable for several years.5,98,99 In contrast, the inability to control the infection and the establishment of chronicity lead to a progressive decline of the adaptive immunity with a lower number of circulating and intrahepatic virus-specific CD8+ and CD4+ T cells,100-103 as well as low and restricted production of virus-specific antibodies.37 Patients who develop chronic HCV infection show persistently defective CD8+ T-cell function,48-50 which probably is maintained by the combined effect of different mechanisms, including prolonged exposure of T cells to viral antigens,104 immunosuppression by viral gene products,58,59 and impaired DC accessory function.91-93 In chronic HBV patients, persistently high production of viral antigens can delete or tolerize antigen-specific T105 and B56 cells. HBV typically is able to produce large quantities of noninfectious subviral particles containing only envelope antigens and a secretory form of the nucleocapsid protein, called hepatitis B e antigen, that induces in transgenic mice the preferential production of T-helper 2 cytokines by deletion of T-helper 1 cells.106,107 Chronic exposure to HBV antigens has been shown to delete HBV-specific B cells56 and to cause an alteration of the hierarchy and function of HBV-specific CD8+ T cells, with a repertoire skewed toward subdominant epitopes. CD8+ T cells escaping deletion caused by large quantities of persisting antigen are characterized by an inability to bind specific human leukocyte antigen tetramers, and can be detected by intracellular cytokine staining.105 This CD8+ T-cell phenotypic alteration is not associated with a lower T-cell receptor affinity but potentially can reflect an altered T-cell receptor organization that can affect human leukocyte antigen-tetramer binding efficiency.108 The in vivo function of these tetramer/negative antigen-specific CD8+ T cells is unknown, but the inability to exert selective pressure on the infecting virus may suggest that they are anergic in vivo.105
 
The progressive qualitative and quantitative collapse of the adaptive immunity in both chronically HBV- and HCV-infected patients has a profound effect on the composition of the virus populations infecting individual patients. Because chronic patients are unable to control the infection rapidly, progressive selection of T- and B-cell escape variant viruses can occur. HBV and HCV variants carrying mutations in helper,109,110 cytotoxic,111-113 and B-cell114-117 epitopes have been detected in chronic hepatitis B and C and this selection is postulated to take place principally during the initial phases of infections.113,116,118 In HCV-infected chimpanzees, viral mutants that are able to escape CD8+ T-cell responses emerge early after infection.119-121 Furthermore, a longitudinal study of patients with chronic hepatitis C revealed that the sequences of CD8+ T-cell epitopes of the infecting viral populations did not diversify during several years of follow-up, despite the relatively high frequency of escape mutations initially present.118 Also, the selective pressure exerted by the humoral response acts early because mutations in B-cell epitope regions found in chronic HCV patients have been shown to develop in the initial phases of infection.116 At later stages, T- and B-cell epitopes appear to be fixed within the viral populations. This is a further indication of the progressive collapse of the immune response in persistent HCV and HBV infections resulting in the inability of the residual T- and B-cell activity to exert selective pressure on the virus.
 
FINAL REMARKS
 
Clarification of the virologic and immunologic events occurring during HBV and HCV infections not only allows us to better define the different mechanisms potentially involved in viral persistence, but also to understand why preventive HBV and HCV vaccines differ in the protection they provide against infection. Although the profile of the adaptive immune response against HBV and HCV infections is similar, the antiviral immune response can provide protection against HBV122 but not against HCV reinfection.123,124 Moreover, although a vaccine based on the envelope protein of HBV, which induces envelope-specific antibody122 and helper T-cell responses,125-127 can elicit a sterilizing anti-HBV immunity, different HCV vaccine preparations able to prime similar profiles of T- and B-cell responses have so far failed to protect animals from HCV infection.128,129 The different kinetics of HCV and HBV replication may represent one of the factors responsible for these differences. If HCV can survive the neutralizing effect of anti-HCV envelope antibodies32,130 that persist at low levels after natural HCV infection37 or immunization,129 a reservoir of replicating virus can be established immediately that may outpace the development of recall HCV-specific antibody and T-cell responses. It is interesting to note that HCV-specific cellular immune responses are detectable earlier after rechallenge (2 weeks) than after primary infection (8-10 weeks).39,41,128,131 This acceleration of the adaptive immune response, although insufficient to prevent infection, can influence positively the course of the HCV-induced liver disease in vaccinated animals, which usually develop low viremia and mild hepatitis. These findings in HCV rechallenge experiments and in HCV-vaccinated animals reinforce the hypothesis that the long interval of time between infection and activation of the HCV-specific adaptive immune response represents a major factor that influences the rate of chronicity and the profile of the disease. In contrast to HCV, HBV is controlled more easily by vaccination122 because its replication kinetics allows sufficient time for adaptive T- and B-cell responses to develop. This may explain why vaccination with HBV envelope antigens can prevent liver disease even if administered after HBV infection.132
 
In both infections, once chronicity develops, the common outcome is a progressive collapse of the immune system, which makes it unlikely that the host can spontaneously reacquire the capacity to control virus replication. At present, the treatment of chronic HBV and HCV infections relies on the use of drugs with direct antiviral effects. However, the different vigor and quality of the virus-specific immunity in self-limited versus chronic hepatitis suggests that therapeutic restoration of the antiviral immunity could lead to the control of virus replication and disease in chronically infected subjects. The real problem with the therapeutic vaccine approach is that the immune system of chronically HBV- and HCV-infected patients does not have the same functional efficiency and cell repertoire of subjects who have been able to control the infection. Therefore, strategies to optimize the efficacy of therapeutic vaccination are needed. In this perspective, efforts should be directed to develop vaccines not only designed to prime efficiently naive T and B cells89 but also to boost the antiviral efficiency of T cells co-existing with high antigen concentrations. For this purpose, inhibition of viral replication by antiviral drugs may be a strategy to restore the antiviral efficiency of the immune system. This is suggested by the observation that lamivudine treatment of chronically infected HBV patients can increase the frequency and improve the function of circulating HBV-specific helper and cytotoxic T cells.133,134 Data supporting the rationale of this approach recently have been reported in animal models of HBV infection, in which the efficacy of therapeutic vaccination is enhanced by previous antiviral treatment.135,136
 
REFERENCES
 
1. Lok AS, McMahon BJ. Chronic hepatitis B. HEPATOLOGY 2001;34:1225-1241.MEDLINE FULL TEXT
 
2. Lauer GM, Walker BD. Hepatitis C virus infection. N Engl J Med 2001;345:41-52.MEDLINE CROSSREF
 
3. Ferrari C, Penna A, Bertoletti A, Valli A, Degli Antoni A, Giuberti T, Cavalli A, et al. Cellular immune response to hepatitis B virus encoded antigens in acute and chronic hepatitis B virus infection. J Immunol 1990;145:3442-3449. MEDLINE
 
4. Jung M, Spengler U, Schraut W, Hoffman R, Zachoval R, Eisemburg J, Eichenlaub D, et al. Hepatitis B virus antigen-specific T-cell activation in patients with acute and chronic hepatitis B. J Hepatol 1991;13:310-317. MEDLINE
 
5. Penna A, Artini M, Cavalli A, Levrero M, Bertoletti A, Pilli M, Chisari FV, et al. Long-lasting memory T cell responses following self-limited acute hepatitis B. J Clin Invest 1996;98:1185-1194. MEDLINE
 
6. Penna A, Del Prete G, Cavalli A, Bertoletti A, D'Elios MM, Sorrentino R, D'Amato M, et al. Predominant T-helper 1 cytokine profile of hepatitis B virus nucleocapsid-specific T cells in acute self-limited hepatitis B. HEPATOLOGY 1997;25:1022-1027.MEDLINEBSTRACT FULL TEXT
 
7. Rehermann B, Fowler P, Sidney J, Person J, Redeker A, Brown M, Moss B, et al. The cytotoxic T lymphocyte response to multiple hepatitis B virus polymerase epitopes during and after acute viral hepatitis. J Exp Med 1995;181:1047-1058. MEDLINE
 
8. Penna A, Chisari F, Bertoletti A, Missale G, Fowler P, Giuberti T, Fiaccadori F, et al. Cytotoxic T lymphocytes recognize an HLA-A2 restricted epitope within the hepatitis B virus nucleocapsid antigen. J Exp Med 1991;174:1565-1570. MEDLINE
 
9. Jung M, Hartmann B, Gerlach J, Diepolder H, Gruber R, Schraut W, Gruner N, et al. Virus-specific lymphokine production differs quantitatively but not qualitatively in acute and chronic hepatitis B infection. Virology 1999;261:165-172.MEDLINE CROSSREF
 
10. Thimme R, Oldach D, Chang KM, Steiger C, Ray SC, Chisari FV. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J Exp Med 2001;194:1395-1406.MEDLINE CROSSREF
 
11. Bottarelli P, Brunetto M, Minutello M, Calvo P, Unutmaz D, Weiner A, Choo Q, et al. T-lymphocyte response to hepatitis C virus in different clinical courses of infection. Gastroenterology 1993;104:580-587.MEDLINE ABSTRACT
 
12. Missale G, Bertoni R, Lamonaca V, Valli A, Massari M, Mori C, Rumi M, 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;98:706-714. MEDLINE
 
13. Diepolder HM, Zachoval R, Hoffmann RM, Wierenga E, Santantonio T, Jung MC, Eichenlaub D, et al. Possible mechanism involving T-lymphocyte response to non-structural protein 3 in viral clearance in acute hepatitis C virus infection. Lancet 1995;346:1006-1007. MEDLINE
 
14. Cramp M, Carucci P, Rossol S, Chokshi S, Maertens G, Williams R, Naoumov N. Hepatitis C virus (HCV) specific immune responses in anti-HCV positive patients with hepatitis C viremia. Gut 1999;44:424-429. MEDLINE
 
15. Gruner NH, Gerlach TJ, Jung MC, Diepolder HM, Schirren CA, Schraut WW, Hoffmann R, et al. Association of hepatitis C virus-specific CD8+ T cells with viral clearance in acute hepatitis C. J Infect Dis 2000;181:1528-1536.MEDLINE CROSSREF
 
16. Lechner F, Wong D, Dunbar P, Chapman R, Chung R, Dohrenwend P, Robbins G, et al. Analysis of successful immune responses in person infected with hepatitis C virus. J Exp Med 2000;191:1499-1512.MEDLINE CROSSREF
 
17. Lechmann M, Ihlenfeldt HG, Braunschweiger I, Giers G, Jung G, Matz B, Kaiser R, et al. T- and B-cell responses to different hepatitis C virus antigens in patients with chronic hepatitis C infection and in healthy anti-hepatitis C virusÑpositive blood donors without viremia. HEPATOLOGY 1996;24:790-795.MEDLINEABSTRACT FULL TEXT
 
18. Chang K-M, Thimme R, Melpolder J, Oldach D, Pemberton J, Moorhead-Lodis J, McHutchison J, et al. Differential CD4+ and CD8+ T-cell responsiveness in hepatitis C virus infection. HEPATOLOGY 2001;33:267-276.MEDLINECROSSREFABSTRACT FULL TEXT
 
19. 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;25:449-458.MEDLINEABSTRACT FULL TEXT
 
20. 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;159:1012-1018. MEDLINE
 
21. Kamal SM, Rasenack JW, Bianchi L, Al Tawil A, El Sayed Khalifa K, Peter T, Mansour H, et al. Acute hepatitis C without and with schistosomiasis: correlation with hepatitis C-specific CD4(+) T-cell and cytokine response. Gastroenterology 2001;121:646-656.MEDLINEABSTRACT FULL TEXT
 
22. Chisari F, Ferrari C. Hepatitis B virus immunopathogenesis. Ann Rev Immunol 1995;13:29-60. CROSSREF
 
23. Thomson M, Nascimbeni M, Havert MB, Major M, Gonzales S, Alter H, Feinstone SM, et al. The clearance of hepatitis C virus infection in chimpanzees may not necessarily correlate with the appearance of acquired immunity. J Virol 2003;77:862-870.MEDLINE CROSSREF
 
24. Cooper S, Erickson A, Adams E, Kansopon J, Weiner A, Chien D, Houghton M, et al. Analysis of a successful immune response against hepatitis C virus. Immunity 1999;10:439-449. MEDLINE
 
25. Thimme R, Bukh J, Spangenberg HC, Wieland S, Pemberton J, Steiger C, Govindarajan S, et al. Viral and immunological determinants of hepatitis C virus clearance, persistence and disease. Proc Natl Acad Sci U S A 2002;99:15661-15668.MEDLINE CROSSREF
 
26. Menne S, Roneker CA, Roggendorf M, Gerin JL, Cote PJ, Tennant BC. Deficiencies in the acute-phase cell-mediated immune response to viral antigens are associated with development of chronic woodchuck hepatitis virus infection following neonatal inoculation. J Virol 2002;76:1769-1780. MEDLINE CROSSREF
 
27. Webster G, Reignat S, Maini M, Whalley S, Ogg G, King A, Brown D, et al. Incubation phase of acute hepatitis B in man: dynamic of cellular immune mechanism. HEPATOLOGY 2000;32:1117-1124.MEDLINECROSSREFABSTRACT FULL TEXT
 
28. Thimme R, Wieland S, Steiger C, Ghrayeb J, Reimann KA, Purcell RH, Chisari FV. CD8(+) T cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infection. J Virol 2003;77:68-76.MEDLINE CROSSREF
 
29. Alberti A, Diana S, Sculard GH, Eddleston AL, Williams R. Detection of a new antibody system reacting with Dane particles in hepatitis B virus infection. Br Med J 1978;2:1056-1058. MEDLINE
 
30. 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;25:1245-1249.MEDLINEABSTRACT FULL TEXT
 
31. Grady GF, Lee VA, Prince AM, Gitnick GL, Fawaz KA, Vyas GN, Levitt MD, et al. Hepatitis B immune globulin for accidental exposures among medical personnel: final report of a multicenter controlled trial. J Infect Dis 1978;138:625-638. MEDLINE
 
32. Farci P, Shimoda A, Wong D, Cabezon T, De Gioannis D, Strazzera A, Shimizu Y, 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 U S A 1996;93:15394-15399.MEDLINE CROSSREF
 
33. Kalams SA, Walker BD. The critical need for CD4 help in maintaining effective cytotoxic T lymphocyte responses. J Exp Med 1998;188:2199-2204.MEDLINE CROSSREF
 
34. Ciurea A, Hunziker L, Klenerman P, Hengartner H, Zinkernagel RM. Impairment of CD4(+) T cell responses during chronic virus infection prevents neutralizing antibody responses against virus escape mutants. J Exp Med 2001;193:297-305.MEDLINE CROSSREF
 
35. Vento S, Ranieri S, Williams R, Rondanelli E, O'Brien C, Eddleston A. Prospective study of cellular immunity to hepatitis B virus antigens from the early incubation phase of acute hepatitis B. Lancet 1987;2:119-122. MEDLINE
 
36. Fong TL, Di Bisceglie AM, Biswas R, Waggoner JG, Wilson L, Claggett J, Hoofnagle JH. High levels of viral replication during acute hepatitis B infection predict progression to chronicity. J Med Virol 1994;43:155-158. MEDLINE
 
37. Chen M, Sallberg M, Sonnerborg A, Weiland O, Mattsson L, Jin L, Birkett A, et al. Limited humoral immunity in hepatitis C virus infection. Gastroenterology 1999;116:135-143.MEDLINE FULL TEXT
 
38. Major ME, Mihalik K, Fernandez J, Seidman J, Kleiner D, Kolykhalov AA, Rice CM, et al. Long-term follow-up of chimpanzees inoculated with the first infectious clone for hepatitis C virus. J Virol 1999;73:3317-3325. MEDLINE
 
39. Major ME, Mihalik K, Puig M, Rehermann B, Nascimbeni M, Rice CM, Feinstone SM. Previously infected and recovered chimpanzees exhibit rapid responses that control hepatitis C virus replication upon rechallenge. J Virol 2002;76:6586-6595.MEDLINE CROSSREF
 
40. Bassett SE, Brasky KM, Lanford RE. Analysis of hepatitis C virus-inoculated chimpanzees reveals unexpected clinical profiles. J Virol 1998;72:2589-2599. MEDLINE
 
41. Bassett SE, Guerra B, Brasky K, Miskovsky E, Houghton M, Klimpel GR, Lanford RE. Protective immune response to hepatitis C virus in chimpanzees rechallenged following clearance of primary infection. HEPATOLOGY 2001;33:1479-1487.MEDLINECROSSREF ABSTRACTFULL TEXT
 
42. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, et al. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol 1994;68:4650-4655. MEDLINE
 
43. Rentenaar RJ, Gamadia LE, van DerHoek N, van Diepen FN, Boom R, Weel JF, Wertheim-van Dillen PM, et al. Development of virus-specific CD4(+) T cells during primary cytomegalovirus infection. J Clin Invest 2000;105:541-548. MEDLINE
 
44. Shimizu YK, Weiner AJ, Rosenblatt J, Wong DC, Shapiro M, Popkin T, Houghton M, et al. Early events in hepatitis C virus infection of chimpanzees. Proc Natl Acad Sci U S A 1990;87:6441-6444. MEDLINE
 
45. Korba BE, Cote PJ, Wells FV, Baldwin B, Popper H, Purcell RH, Tennant BC, et al. Natural history of woodchuck hepatitis virus infections during the course of experimental viral infection: molecular virologic features of the liver and lymphoid tissues. J Virol 1989;63:1360-1370. MEDLINE
 
46. Berquist KR, Peterson JM, Murphy BL, Ebert JW, Maynard JE, Purcell RH. Hepatitis B antigens in serum and liver of chimpanzees acutely infected with hepatitis B virus. Infect Immun 1975;12:602-605. MEDLINE
 
47. Maini MK, Boni C, Ogg GS, King AS, Reignat S, Lee CK, Larrubia JR, et al. Direct ex vivo analysis of hepatitis B virus-specific CD8+ T cells associated with the control of infection. Gastroenterology 1999;117:1386-1396.MEDLINEABSTRACT FULL TEXT
 
48. Urbani S, Boni C, Missale G, Elia G, Cavallo C, Massari M, Raimondo G, et al. Virus-specific CD8(+) lymphocytes share the same effector-memory phenotype but exhibit functional differences in acute hepatitis B and C. J Virol 2002;76:12423-12434.MEDLINE CROSSREF
 
49. Gruener NH, Lechner F, Jung MC, Diepolder H, Gerlach T, Lauer G, Walker B, et al. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J Virol 2001;75:5550-5558. MEDLINE CROSSREF
 
50. Wedemeyer H, He XS, Nascimbeni M, Davis AR, Greenberg HB, Hoofnagle JH, Liang TJ, et al. Impaired effector function of hepatitis C virus-specific CD8+ T cells in chronic hepatitis C virus infection. J Immunol 2002;169:3447-3458. MEDLINE
 
51. Callan MF, Tan L, Annels N, Ogg GS, Wilson JD, O'Callaghan CA, Steven N, et al. Direct visualization of antigen-specific CD8(+) T cells during the primary immune response to Epstein Barr virus in vivo. J Exp Med 1998;187:1395-1402.MEDLINE CROSSREF
 
52. Ogg GS, Jin X, Bonhoeffer S, Dunbar PR, Nowak MA, Monard S, Segal JP, et al. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science 1998;279:2103-2106.MEDLINE CROSSREF
 
53. Lauer GM, Ouchi K, Chung RT, Nguyen TN, Day CL, Purkis DR, Reiser M, et al. Comprehensive analysis of CD8(+)-T-cell responses against hepatitis C virus reveals multiple unpredicted specificities. J Virol 2002;76:6104-6113.MEDLINE CROSSREF
 
54. Anthony DD, Valdez H, Post AB, Carlson NL, Heeger PS, Lehmann PV. Comprehensive determinant mapping of the hepatitis C-specific CD8 cell repertoire reveals unpredicted immune hierarchy. Clin Immunol 2002;103:264-276.MEDLINE CROSSREF
 
55. 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.MEDLINE CROSSREF
 
56. 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;345:258-260.MEDLINE CROSSREF
 
57. Klugewitz K, Blumenthal-Barby F, Schrage A, Knolle PA, Hamann A, Crispe IN. Immunomodulatory effects of the liver: deletion of activated CD4+ effector cells and suppression of IFN-gamma-producing cells after intravenous protein immunization. J Immunol 2002;169:2407-2413. MEDLINE
 
58. Large M, Kittlesen D, Hahn Y. Suppression of host immune response by the core protein of hepatitis C virus: possible implications for hepatitis C virus persistence. J Immunol 1999;162:931-938. MEDLINE
 
59. Kittlesen DJ, Chianese-Bullock KA, Yao ZQ, Braciale TJ, Hahn YS. Interaction between complement receptor gC1qR and hepatitis C virus core protein inhibits T-lymphocyte proliferation. J Clin Invest 2000;106:1239-1249. MEDLINE
 
60. Vanlandschoot P, Van Houtte F, Roobrouck A, Farhoudi A, Leroux-Roels G. Hepatitis B virus surface antigen suppresses the activation of monocytes through interaction with a serum protein and a monocyte-specific receptor. J Gen Virol 2002;83:1281-1289. MEDLINE
 
61. Biron CA. Interferons alpha and beta as immune regulatorsÑa new look. Immunity 2001;14:661-664. MEDLINE CROSSREF
 
62. Ou R, Zhou S, Huang L, Moskophidis D. Critical role for alpha/beta and gamma interferons in persistence of lymphocytic choriomeningitis virus by clonal exhaustion of cytotoxic T cells. J Virol 2001;75:8407-8423.MEDLINE CROSSREF
 
63. Bigger CB, Brasky KM, Lanford RE. DNA microarray analysis of chimpanzee liver during acute resolving hepatitis C virus infection. J Virol 2001;75:7059-7066.MEDLINE CROSSREF
 
64. Su AI, Pezacki JP, Wodicka L, Brideau AD, Supekova L, Thimme R, Wieland S, et al. Genomic analysis of the host response to hepatitis C virus infection. Proc Natl Acad Sci U S A 2002;99:15669-15674.MEDLINE CROSSREF
 
65. Cella M, Salio M, Sakakibara Y, Langen H, Julkunen I, Lanzavecchia A. Maturation, activation, and protection of dendritic cells induced by double-stranded RNA. J Exp Med 1999;189:821-829. MEDLINECROSSREF
 
66. Blight KJ, Kolykhalov AA, Rice CM. Efficient initiation of HCV RNA replication in cell culture. Science 2000;290:1972-1974.MEDLINE CROSSREF
 
67. Castet V, Fournier C, Soulier A, Brillet R, Coste J, Larrey D, Dhumeaux D, et al. Alpha interferon inhibits hepatitis C virus replication in primary human hepatocytes infected in vitro. J Virol 2002;76:8189-8199.MEDLINE CROSSREF
 
68. Tan SL, Katze MG. How hepatitis C virus counteracts the interferon response: the jury is still out on NS5A. Virology 2001;284:1-12.MEDLINE CROSSREF
 
69. Taylor D, Shi S, Romano P, Barber G, Lai M. Inhibition of interferon-inducible protein kinase PKR by HCV E2 protein. Science 1999;285:107-110. MEDLINECROSSREF
 
70. Biron CA, Brossay L. NK cells and NKT cells in innate defense against viral infections. Curr Opin Immunol 2001;13:458-464.MEDLINE CROSSREF
 
71. Liu ZX, Govindarajan S, Okamoto S, Dennert G. NK cells cause liver injury and facilitate the induction of T cell-mediated immunity to a viral liver infection. J Immunol 2000;164:6480-6486. MEDLINE
 
72. Kakimi K, Lane TE, Chisari FV, Guidotti LG. Cutting edge: inhibition of hepatitis B virus replication by activated NK T cells does not require inflammatory cell recruitment to the liver. J Immunol 2001;167:6701-6705. MEDLINE
 
73. Tseng CT, Klimpel GR. Binding of the hepatitis C virus envelope protein E2 to CD81 inhibits natural killer cell functions. J Exp Med 2002;195:43-49.MEDLINE CROSSREF
 
74. Crotta S, Stilla A, Wack A, D'Andrea A, Nuti S, D'Oro U, Mosca M, et al. Inhibition of natural killer cells through engagement of CD81 by the major hepatitis C virus envelope protein. J Exp Med 2002;195:35-41. MEDLINE CROSSREF
 
75. Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, Weiner AJ, et al. Binding of hepatitis C virus to CD81. Science 1998;282:938-941. MEDLINECROSSREF
 
76. Salazar-Mather TP, Lewis CA, Biron CA. Type I interferons regulate inflammatory cell trafficking and macrophage inflammatory protein 1alpha delivery to the liver. J Clin Invest 2002;110:321-330.MEDLINE CROSSREF
 
77. Duncan GS, Mittrucker HW, Kagi D, Matsuyama T, Mak TW. The transcription factor interferon regulatory factor-1 is essential for natural killer cell function in vivo. J Exp Med 1996;184:2043-2048. MEDLINE
 
78. Whalley SA, Murray JM, Brown D, Webster GJ, Emery VC, Dusheiko GM, Perelson AS. Kinetics of acute hepatitis B virus infection in humans. J Exp Med 2001;193:847-854.MEDLINE CROSSREF
 
79. Jilbert A, Wu T, England J, Hall P, Carp N, O'Connel A, Mason W. Rapid resolution of duck hepatitis B virus infections occurs after massive hepatocellular involvement. J Virol 1992;66:1377-1388. MEDLINE
 
80. Kajino K, Jilbert AR, Saputelli J, Aldrich CE, Cullen J, Mason WS. Woodchuck hepatitis virus infections: very rapid recovery after prolonged viremia and infection of virtually every hepatocyte. J Virol 1994;68:5792-5803. MEDLINE
 
81. 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;284:825-829.MEDLINE CROSSREF
 
82. Cote PJ, Toshkov I, Bellezza C, Ascenzi M, Roneker C, Ann Graham L, Baldwin BH, et al. Temporal pathogenesis of experimental neonatal woodchuck hepatitis virus infection: increased initial viral load and decreased severity of acute hepatitis during the development of chronic viral infection. HEPATOLOGY 2000;32:807-817.MEDLINEABSTRACT FULL TEXT
 
83. Nakamura I, Nupp JT, Cowlen M, Hall WC, Tennant BC, Casey JL, Gerin JL, et al. Pathogenesis of experimental neonatal woodchuck hepatitis virus infection: chronicity as an outcome of infection is associated with a diminished acute hepatitis that is temporally deficient for the expression of interferon gamma and tumor necrosis factor-alpha messenger RNAs. HEPATOLOGY 2001;33:439-447.MEDLINECROSSREFABSTRACT FULL TEXT
 
84. Kakimi K, Guidotti LG, Koezuka Y, Chisari FV. Natural killer T cell activation inhibits hepatitis B virus replication in vivo. J Exp Med 2000;192:921-930.MEDLINE CROSSREF
 
85. Baron JL, Gardiner L, Nishimura S, Shinkai K, Locksley R, Ganem D. Activation of a nonclassical NKT cell subset in a transgenic mouse model of hepatitis B virus infection. Immunity 2002;16:583-594. MEDLINE
 
86. Kakimi K, Lane TE, Wieland S, Asensio VC, Campbell IL, Chisari FV, Guidotti LG. Blocking chemokine responsive to gamma-2/interferon (IFN)-gamma inducible protein and monokine induced by IFN-gamma activity in vivo reduces the pathogenetic but not the antiviral potential of hepatitis B virus-specific cytotoxic T lymphocytes. J Exp Med 2001;194:1755-1766. MEDLINE CROSSREF
 
87. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, et al. Immunobiology of dendritic cells. Annu Rev Immunol 2000;18:767-811.MEDLINE CROSSREF
 
88. Mueller SN, Jones CM, Smith CM, Heath WR, Carbone FR. Rapid cytotoxic T lymphocyte activation occurs in the draining lymph nodes after cutaneous herpes simplex virus infection as a result of early antigen presentation and not the presence of virus. J Exp Med 2002;195:651-656.MEDLINE CROSSREF
 
89. Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol 2002;2:251-262.MEDLINE CROSSREF
 
90. Piccioli D, Sbrana S, Melandri E, Valiante NM. Contact-dependent stimulation and inhibition of dendritic cells by natural killer cells. J Exp Med 2002;195:335-341.MEDLINE CROSSREF
 
91. Kanto T, Hayashi N, Takehara T, Tatsumi T, Kuzushita N, Ito A, Sasaki Y, et al. Impaired allostimulatory capacity of peripheral blood dendritic cells recovered from hepatitis C virus-infected individuals. J Immunol 1999;162:5584-5591. MEDLINE
 
92. 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;120:512-524.MEDLINEABSTRACT FULL TEXT
 
93. Auffermann-Gretzinger S, Keeffe EB, Levy S. Impaired dendritic cell maturation in patients with chronic, but not resolved, hepatitis C virus infection. Blood 2001;97:3171-3176. MEDLINE CROSSREF
 
94. Sarobe P, Lasarte JJ, Casares N, Lopez-Diaz de Cerio A, Baixeras E, Labarga P, Garcia N, et al. Abnormal priming of CD4(+) T cells by dendritic cells expressing hepatitis C virus core and E1 proteins. J Virol 2002;76:5062-5070.MEDLINE CROSSREF
 
95. Wang FS, Xing LH, Liu MX, Zhu CL, Liu HG, Wang HF, Lei ZY. Dysfunction of peripheral blood dendritic cells from patients with chronic hepatitis B virus infection. World J Gastroenterol 2001;7:537-541. MEDLINE
 
96. Beckebaum S, Cicinnati VR, Dworacki G, Muller-Berghaus J, Stolz D, Harnaha J, Whiteside TL, et al. Reduction in the circulating pDC1/pDC2 ratio and impaired function of ex vivo-generated DC1 in chronic hepatitis B infection. Clin Immunol 2002;104:138-150.MEDLINE CROSSREF
 
97. Lohr HF, Pingel S, Bocher WO, Bernhard H, Herzog-Hauff S, Rose-John S. Reduced virus specific T helper cell induction by autologous dendritic cells in patients with chronic hepatitis BÑrestoration by exogenous interleukin-12. Clin Exp Immunol 2002;130:107-114.MEDLINE CROSSREF
 
98. Rehermann B, Ferrari C, Pasquinelli C, Chisari FV. The hepatitis B virus persists for decades after patients' recovery from acute viral hepatitis despite active maintenance of a cytotoxic T-lymphocyte response. Nat Med 1996;2:1104-1108. MEDLINE
 
99. Takaki A, Wiese M, Maertens G, Depla E, Seifert U, Liebetrau A, Miller J, et al. Cellular immune responses persist and humoral responses decreases two decades after recovery from a single-source outbreak of hepatitis C. Nat Med 2000;6:578-582.MEDLINE CROSSREF
 
100. Maini MK, Boni C, Lee CK, Larrubia JR, Reignat S, Ogg GS, King AS, et al. The role of virus-specific CD8+ cells in viral control and liver damage during persistent hepatitis B virus (HBV) infection. J Exp Med 2000;191:1269-1280.MEDLINE CROSSREF
 
101. He X-S, Rehermann B, Lopez-Labrador FX, Boisvert J, Cheung R, Mumm J, Wedemeyer H, 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 U S A 1999;96:5692-5697.MEDLINE CROSSREF
 
102. 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;98:1432-1440. MEDLINE
 
103. Rosen HR, Miner C, Sasaki AW, Lewinsohn DM, Conrad AJ, Bakke A, Bouwer HG, et al. Frequencies of HCV-specific effector CD4+ T cells by flow cytometry: correlation with clinical disease stages. HEPATOLOGY 2002;35:190-198.MEDLINECROSSREFABSTRACT FULL TEXT
 
104. Welsh R. Assessing CD8 T cell number and dysfunction in the presence of antigen. J Exp Med 2001;193:F19-F22. MEDLINECROSSREF
 
105. Reignat S, Webster GJ, Brown D, Ogg GS, King A, Seneviratne SL, Dusheiko G, et al. Escaping high viral load exhaustion: CD8 cells with altered tetramer binding in chronic hepatitis B virus infection. J Exp Med 2002;195:1089-1101.MEDLINE CROSSREF
 
106. Milich D, Chen M, Hughes J, Jones J. The secreted hepatitis B precore antigen can modulate the immune response to the nucleocapsid: a mechanism for persistence. J Immunol 1998;160:2013-2021. MEDLINE
 
107. Milich DR, Schodel F, Hughes JL, Jones JE, Peterson DL. The hepatitis B virus core and e antigens elicit different Th cell subsets: antigen structure can affect Th cell phenotype. J Virol 1997;71:2192-2201. MEDLINE
 
108. Drake D III, Braciale T. Cutting edge: lipid raft integrity affects the efficiency of MHC class I tetramer binding and cell surface TCR arrangement on CD8+ T cells. J Immunol 2001;166:7009-7013. MEDLINE
 
109. Wang H, Eckels DD. Mutations in immunodominant T cell epitopes derived from the nonstructural 3 protein of hepatitis C virus have the potential for generating escape variants that may have important consequences for T cell recognition. J Immunol 1999;162:4177-4183. MEDLINE
 
110. Frasca L, Del Porto P, Tuosto L, Marinari B, Scotta C, Carbonari M, Nicosia A, et al. Hypervariable region 1 variants act as TCR antagonists for hepatitis C virus-specific CD4+ T cells. J Immunol 1999;163:650-658. MEDLINE
 
111. Rehermann B, Pasquinelli C, Mosier SM, Chisari FV. Hepatitis B virus (HBV) sequence variation of cytotoxic T lymphocyte epitopes is not common in patients with chronic HBV infection. J Clin Invest 1995;96:1527-1534. MEDLINE
 
112. Bertoletti A, Costanzo A, Chisari FV, Levrero M, Artini M, Sette A, Penna A, et al. Cytotoxic T lymphocyte response to a wild type hepatitis B virus epitope in patients chronically infected by variant viruses carrying substitutions within the epitope. J Exp Med 1994;180:933-943. MEDLINE
 
113. Tsai S-L, Chen Y-M, Chen M-H, Huang C-Y, Sheen I-S, Yeh C-T, Huang J-H, et al. Hepatitis C virus variants circumventing cytotoxic T lymphocyte activity as a mechanism of chronicity. Gastroenterology 1998;115:954-966.MEDLINEABSTRACT FULL TEXT
 
114. Weiner AJ, Geysen HM, Christopherson C, Hall JE, Mason TJ, Saracco G, Bonino F, 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 U S A 1992;89:3468-3472. MEDLINE
 
115. 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;68:1494-1500. MEDLINE
 
116. Farci P, Shimoda A, Coiana A, Diaz G, Peddis G, Melpolder JC, Strazzera A, et al. The outcome of acute hepatitis C predicted by the evolution of the viral quasispecies. Science 2000;288:339-344.MEDLINE CROSSREF
 
117. Booth JC, Kumar U, Webster D, Monjardino J, Thomas HC. Comparison of the rate of sequence variation in the hypervariable region of E2/NS1 region of hepatitis C virus in normal and hypogammaglobulinemic patients. HEPATOLOGY 1998;27:223-227.MEDLINE ABSTRACTFULL TEXT
 
118. 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;100:2376-2385. MEDLINE
 
119. Weiner A, Erickson AL, Kansopon J, Crawford K, Muchmore E, Hughes AL, Houghton M, 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 U S A 1995;92:2755-2759. MEDLINE
 
120. Erickson AL, Kimura Y, Igarashi S, Eichelberger J, Houghton M, Sidney J, McKinney D, et al. The outcome of hepatitis C virus infection is predicted by escape mutations in epitopes targeted by cytotoxic T lymphocytes. Immunity 2001;15:883-895. MEDLINE
 
121. Thomson M, Nascimbeni M, Gonzales S, Murthy KK, Rehermann B, Liang TJ. Emergence of a distinct pattern of viral mutations in chimpanzees infected with a homogeneous inoculum of hepatitis C virus. Gastroenterology 2001;121:1226-1233.MEDLINEABSTRACT FULL TEXT
 
122. Purcell RH, Gerin JL. Hepatitis B subunit vaccine: a preliminary report of safety and efficacy tests in chimpanzees. Am J Med Sci 1975;270:395-399. MEDLINE
 
123. Farci P, Alter HJ, Govindarajan S, Wong DC, Engle R, Lesniewski RR, Mushahwar IK, et al. Lack of protective immunity against reinfection with hepatitis C virus. Science 1992;258:135-140. MEDLINE
 
124. Lai ME, Mazzoleni AP, Argiolu F, De Virgilis S, Balestrieri A, Purcell RH, Cao A, et al. Hepatitis C virus in multiple episodes of acute hepatitis in polytransfused thalassaemic children. Lancet 1994;343:388-390. MEDLINE
 
125. Celis E, Ou D, Otvos L Jr. Recognition of hepatitis B surface antigen by human T lymphocytes. Proliferative and cytotoxic responses to a major antigenic determinant defined by synthetic peptides. J Immunol 1988;140:1808-1815. MEDLINE
 
126. Ferrari C, Penna A, Bertoletti A, Cavalli A, Valli A, Schianchi C, Fiaccadori F. The preS1 antigen of hepatitis B virus is highly immunogenic at the T cell level in man. J Clin Invest 1989;84:1314-1319. MEDLINE
 
127. Desombere I, Gijbels Y, Verwulgen A, Leroux-Roels G. Characterization of the T cell recognition of hepatitis B surface antigen (HBsAg) by good and poor responders to hepatitis B vaccines. Clin Exp Immunol 2000;122:390-399.MEDLINE CROSSREF
 
128. Weiner AJ, Paliard X, Selby MJ, Medina-Selby A, Coit D, Nguyen S, Kansopon J, et al. Intrahepatic genetic inoculation of hepatitis C virus RNA confers cross-protective immunity. J Virol 2001;75:7142-7148.MEDLINE CROSSREF
 
129. Forns X, Payette PJ, Ma X, Satterfield W, Eder G, Mushahwar IK, Govindarajan S, 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;32:618-625. MEDLINECROSSREFABSTRACTFULL TEXT
 
130. Farci P, Alter HJ, Wong DC, Miller RH, Govindarajan S, Engle R, Shapiro M, et al. Prevention of hepatitis C virus infection in chimpanzees after antibody-mediated in vitro neutralization. Proc Natl Acad Sci U S A 1994;91:7792-7796. MEDLINE
 
131. Nascimbeni M, Mizukoshi E, Bosmann M, Major ME, Mihalik K, Rice CM, Feinstone SM, et al. Kinetics of CD4(+) and CD8(+) memory T-cell responses during hepatitis C virus rechallenge of previously recovered chimpanzees. J Virol 2003;77:4781-4793. MEDLINECROSSREF
 
132. Iwarson S, Wahl M, Ruttimann E, Snoy P, Seto B, Gerety RJ. Successful postexposure vaccination against hepatitis B in chimpanzees. J Med Virol 1988;25:433-439. MEDLINE
 
133. Boni C, Bertoletti A, Penna A, Cavalli A, Pilli M, Urbani S, Scognamiglio P, et al. Lamivudine treatment can restore T cell responsiveness in chronic hepatitis B. J Clin Invest 1998;102:968-975. MEDLINE
 
134. Boni C, Penna A, Ogg G, Bertoletti A, Pilli M, Cavalli A, Urbani S, et al. Lamivudine treatment can overcome cytotoxic T cell hyporesponsiveness in chronic hepatitis B: new perspective for immune therapy. HEPATOLOGY 2001;33:963-971. MEDLINECROSSREFABSTRACULL TEXT
 
135. Menne S, Roneker CA, Korba BE, Gerin JL, Tennant BC, Cote PJ. Immunization with surface antigen vaccine alone and after treatment with 1-(2-fluoro-5-methyl-beta-L-arabinofuranosyl)-uracil (L-FMAU) breaks humoral and cell-mediated immune tolerance in chronic woodchuck hepatitis virus infection. J Virol 2002;76:5305-5314. MEDLINE CROSSREF
 
136. Le Guerhier F, Thermet A, Guerret S, Chevallier M, Jamard C, Gibbs CS, Trepo C, et al. Antiviral effect of adefovir in combination with a DNA vaccine in the duck hepatitis B virus infection model. J Hepatol 2003;38:328-334. MEDLINE CROSSREF
 
 
 
 
  icon paper stack View Older Articles   Back to Top   www.natap.org