Abstract
Hepatitis B virus (HBV) and hepatitis C virus (HCV) cause a large proportion of acute and chronic liver disease worldwide. Over the past decades many immunological studies defined host immune responses that mediate spontaneous clearance of acute HBV and HCV infection. However, host immune responses are also relevant in the context of treatment-induced clearance of chronic HBV and HCV infection. First, the pretreatment level of interferon-stimulated genes as well as genetic determinants of innate immune responses, such as single nucleotide polymorphisms near the IFNL3 gene, are strong predictors of the response to interferon-alpha (IFN-α)-based therapy. Second, IFN-α, which has been a mainstay of HBV and HCV therapy over decades, and ribavirin, which has also been included in interferon-free direct antiviral therapy for HCV, modulate host immune responses. Third, both IFN-α-based and IFN-α-free treatment regimens of HBV and HCV infection alter the short-term and long-term adaptive immune response against these viruses. Finally, treatment studies have not just improved the clinical outcomes, but also provided opportunities to study virus-host interaction. This review summarizes our current knowledge on how a patient's immune response affects the treatment outcome of HBV and HCV infection and how innate and adaptive immune responses themselves are altered by the different treatment regimens.
Spontaneous hepatitis B virus (HBV) and hepatitis C virus (HCV) clearance occurs most frequently in the acute phase of infection and is mediated by vigorous adaptive immune responses. In contrast, spontaneous viral clearance rarely occurs in the chronic phase of HBV infection and almost never in the chronic phase of HCV infection when virus-specific T-cell responses are exhausted. While host immune responses that result in spontaneous HBV and HCV clearance have been extensively characterized, it has recently become clear that they also play a role in the context of antiviral therapy in chronic infection (Table1).
Table 1.
HBV | HCV | |
---|---|---|
Chronic infections | 350 million people infected | 170 million people infected |
Cause of chronic infection | Mostly vertical/perinatal transmission: mother-to-neonate transmission most common worldwide, followed by childhood infection | Mostly horizontal transmission: injection drug use, parenteral, sexual, nosocomial |
Virology | ||
Virus | 42 nm; enveloped nucleocapsid; partially double-stranded DNA genome | 50 nm; enveloped nucleocapsid; positive stranded RNA genome |
Family | Hepadnaviridae | Flaviviridae; Hepacivirus genus |
Genotypes | 8 genotypes | 6 major genotypes; more than 50 subtypes; quasispecies in each infected patient |
Mutation rate | Low | High |
Virus half-life | 2-3 days | 3 hours |
Virus production | 1010 -1012 virions/day | 1012 virions/day |
Natural immunity | ||
Innate response | Minimal IFN and ISG response | Strong IFN and ISG response |
Protective response | ||
- primary infection | T cells, neutralizing antibodies | T cells; transient strain-specific neutralizing antibodies? |
- secondary infection | T cells, neutralizing antibodies | T cells |
Virus clearance | No; cccDNA (transcriptional template) may persist; disease reactivation possible | Yes |
Therapy of chronic infection | ||
Targets | HBV replication HBV transcription (cccDNA) | HCV replication |
IFN-α based therapy | - Cure (serconversion) possible but rare; | Cure (SVR) possible |
- Induces epigenetic changes in cccDNA; | ||
- Slow decrease in viral titer; | - Fast decrease in viremia; | |
- ISG and NK cell activation | - ISG and NK cell activation in inverse correlation to pretreatment levels (genetically determined); | |
- Suppression of HBV-spec. T cells | - Suppression of HCV-spec. T cells | |
IFN-free antiviral therapy | - Cure (seroconversion) very unlikely; | - High incidence of cure (SVR) |
- Fast reduction in viremia | - Decreased ISG expression | |
- Partial reversal of T-cell exhaustion | - Fast reduction in viremia | |
- Partial reversal of T-cell exhaustion |
Abbreviations: IFN, interferon; ISG, interferon-stimulated gene; NK cell, natural killer cell; SVR, sustained virological response.
Immunological Aspects of HBV Antiviral Therapy
When assessing the prospects of successful antiviral therapy of chronic HBV infection one has to consider that even spontaneously recovered patients do not completely clear HBV and may reactivate the infection upon immunosuppression, e.g., during cancer chemotherapy or organ transplantation.1 Spontaneous recovery with seroconversion to hepatitis B virus surface antigen (HBsAg)-negative/anti-HBs-positive is observed in >95% of acute HBV infections and <1%/year of chronic HBV infection. Although spontaneous recovery results in lifelong protective immunity, trace amounts of HBV DNA appear sporadically in the circulation.2 These trace amounts of HBV can be infectious and stimulate HBV-specific antibody and T-cell responses, which in turn control viremia. Thus, natural immunity is considered protective rather than sterilizing. This is because the transcriptional template of the virus, the covalently closed circular DNA (cccDNA), persists in the form of a minichromosome in host cells.3 Immunosuppression, e.g., as part of cancer chemotherapy or organ transplantation, carries the risk of HBV reactivation.1 An ideal antiviral therapy should therefore not just decrease HBV replication but also eliminate or at least control cccDNA. Antiviral therapy may need to synergize with and ideally boost adaptive immune responses to achieve this.
Interferon (IFN)-α-Based Therapy
The rationale for using IFN-α to treat chronic HBV infection is identical to its use in chronic HCV infection: IFN-α exerts direct antiviral effects and induces complex immune alterations that ideally result in the clearance of the respective viruses.
In a small percentage of patients with chronic HBV infection IFN-α-based therapy results in HBeAg and HBsAg loss, seroconversion to anti-HBe and anti-HBs status, reduction of viral replication, and alanine aminotransferase (ALT) normalization.4 High doses of IFN-α have been shown to induce the degradation of HBV cccDNA in cell culture models by up-regulating the expression of the deaminase APOBEC3A and to deaminate cccDNA,5 presumably when it is transiently rendered single-stranded by RNA polymerase II before transcription initiation. Cytidine deamination results in apurinic/apyrimidinic site formation and in degradation of cccDNA degradation by endonucleases.5 IFN-α also induces epigenetic changes in cccDNA-bound histones, which results in inhibition of HBV replication and decreases the transcription of pregenomic RNA and subgenomic RNA from cccDNA.6,7 Finally, IFN-α accelerates decay of nucelocapsids that contain pregenomic HBV RNA.8,9 The direct antiviral effect of IFN-α is sufficient to induce a reduction in viral load and HBV antigen levels as shown in HBV-infected humanized uPA/SCID mice that lack immune cells.10
Successful antiviral treatment also appears to synergize with the natural trend to viral control in some patient populations, such as HBeAg+ patients with increased aminotransferase levels. These patients are not only more likely to establish partial immune control of HBV replication with seroconversion to HBe-Ag-negative/antiHBeAg-positive status and a decrease in disease activity, but are also more likely to respond to antiviral therapy.
Unfortunately, both the induction of endogenous IFN-α during the natural course of HBV infection and the antiviral function of exogenously administered IFN-α are not fully effective in most patients. Patients with acute HBV infection or HBV reactivation do not have much evidence of IFN-α production,11 and IFN-stimulated genes (ISGs) are neither significantly induced in the livers of chimpanzees with acute HBV infection12 nor in the livers of woodchucks chronically infected with woodchuck hepatitis virus (WHV), which is closely related to HBV.13 The reasons for HBV's ability to avoid a robust IFN-α-mediated innate response are controversial and have been reviewed in detail elsewhere.14 In addition to hiding from intracellular innate immune mechanism of pathogen detection,15 HBV may prevent Jak STAT signaling.16 To explain the overall low antiviral effect of IFN-α on HBV-infected hepatocytes, a recent report has suggested that its antiviral activity is mediated by exosomes that are secreted by noninfected nonparenchymal cells.17
The limited antiviral effects of IFN-α in HBV infection are reflected by the relatively small percentage of patients with chronic HBV infection who respond to IFN-α-based therapy with HBeAg and HBsAg loss, seroconversion to anti-HBe and anti-HBs status, reduction of viral replication, and ALT normalization.4 A decrease in HBV viremia is observed about 3-4 weeks after the start of IFN-α-based therapy18,19 and takes several months to reach its maximum,20 whereas a sharp decrease in HCV viremia occurs within the first 48 hours of IFN-α-based therapy.21
This delayed kinetic may reflect a progressive recovery of immune-mediated endogenous antiviral mechanisms, which, however, have scarcely been analyzed. A recent study characterized innate and adaptive immune responses of HBeAg-negative HBV-infected patients during 48 weeks of pegylated (Peg)IFN-α therapy.20 The decrease in HBV viremia coincided with an increase in the frequency of circulating CD56bright natural killer (NK) cells, increased expression of the activatory receptor NKp46, and the cytotoxic receptor TRAIL by NK cells and recovery of their IFN-γ production.20 The improved function of CD56bright NK cells paralleled an increase in IL-15 levels, which may have stimulated the NK cells.20 These observations were subsequently extended to HBeAg-positive patients.22 Interrogation of the transcriptome of peripheral blood mononuclear cells (PBMC) showed ISG induction within 6 hours after the start of therapy. This study also confirmed the ability of IFN-α treatment to boost IL-15 (in addition to IL-6 and CXCL-10) levels and to trigger NK cell proliferation. However, the robust activation of innate immune parameters was not associated with a decrease in HBV titer within the first 2 weeks of therapy, suggesting that the modulation of innate immune responses was a direct effect of the administered IFN-α and not secondary to its antiviral effect.
In contrast to innate immune responses, HBV-specific T-cell responses did not recover during IFN-based therapy.20,23 This is unexpected because IFN-α is known to increase T-cell survival,24 expression of antigen-processing enzymes,25 cross-presentation of T-cell antigens,26 and production of interleukin (IL)-12,27 a cytokine that has been shown to rescue the function of exhausted HBV-specific T cells in vitro.28 While the frequency of activated T cells increases within the first 2-3 days of IFN-α-based therapy, a numeric reduction in CD8 T cells, in particular late effector CD8 T cells, is observed during long-term treatment.20 Moreover, the HBV-specific CD8 T-cell subset maintains expression of inhibitory cell surface markers such as PD-1, Lag-3, and CTLA-4,22 and thus remains nonfunctional throughout therapy.20 Similar negative effects of IFN-α on virus-specific adaptive immunity have recently been emphasized in two studies of mice that were chronically infected with lymphocytic choriomengitis virus. In these studies, T-cell responses were restored by inhibiting the effects of virus-induced IFN-α rather than by treating the infection with IFN-α.29,30 Although this scenario is not fully applicable to HBV-infected patients, where IFN-α signaling is suppressed,13,31 the strong and continued activation of ISGs during IFN-α-based therapy22 may negatively impact the function of HBV-specific T cells. Accordingly, flares in liver disease activity are often observed after cessation of IFN-based therapy. Seroconversion to anti-HBe may follow a flare and is specifically associated with an increase in IL-12, IFN-γ, and IL-2 serum levels.32 While a recovery of CD8 T-cell function is not observed during IFN-based therapy,22 cytotoxic CD8 T-cell lines can be more readily expanded from the blood of patients who have responded to IFN-based therapy with anti-HBe or with anti-HBe/anti-HBs seroconversion than from nonresponders33
Thus, IFN-α-based therapy stimulates predominantly innate immune responses. A note of caution is warranted because all published data are based on interrogation of immune responses in the blood. Although reduced expression of ISGs in the liver of chronic HBV patients mirrors that in PBMCs,31 the intrahepatic microenvironment poses additional limitations to the effector arm of antiviral immunity. For example, intrahepatic levels of suppression of cytokine signaling 3 (SOCS3), a negative regulator of cytokine signaling, are increased in patients with chronic HBV infection and in woodchucks with chronic WHV infection.13,31 SOCS3 has not only been associated with HBV's or WHV's ability to evade innate immunity but it is a predictor of poor IFN-α responses in HCV-infected patients34 and as such may also impact the therapeutic efficacy of IFN-α in patients with chronic HBV infection. Because the intrahepatic environment in chronic HBV infection is also rich in IL-10,35 transforming growth factor beta (TGF-β),36 and arginase,37 factors that impair both T and NK cell function, it may not be surprising that the activation of innate immune cells within the first hours of IFN-α treatment is not accompanied by a reduction in HBV titer. A decrease in intrahepatic inflammation, as observed in chronic HBV patients who are treated with NUCs, may be required to restore immune responses in the liver.
Therapy With Nucleos(t)ide Analogs (NUC)
HBV-DNA titers decline rapidly in blood and liver during treatment with NUCs, and inflammatory liver disease improves. The potent antiviral effect of NUCs is based on an inhibition of the reverse transcriptase function of the HBV DNA polymerase, the enzyme responsible for HBV replication.38 The rapid reduction of viral replication should theoretically blunt any potential immunomodulatory effect of HBV and restore IFN-α signaling in infected hepatocytes.39 This hypothesis is supported by the recent description of a progressive expression of ISGs in PBMCs of HBeAg+ HBV-infected patients as early as day 8 of treatment with tenofovir along with a 100-fold decrease in HBV titer.22
The impact of NUC treatment on innate immune cells is controversial. While a recovery of the function of myeloid dendritic cells has been observed in adefovir-treated patients,40 the effect of NUCs on NK cells is less clear. An improvement of NK cell IFN-γ production has been described in a study of entecavir-treated patients,41 but other studies of adefovir and lamivudine-treated patients did not confirm these results and instead showed decreased expression of the NK cell effector molecule TRAIL.35 Since TRAIL-mediated hepatocyte lysis is thought to contribute to liver injury and inflammation in chronic HBV infection, the latter finding would be consistent with the decrease in liver inflammation that is characteristic for treatment with NUCs.
The reported effects of NUC treatment on adaptive immune cells are more consistent, because an increased frequency and function of HBV-specific CD4 and CD8 T cells is reported in multiple studies.42–49 Telbivudine and lamivudine both reduce the expression of the inhibitory molecule PD-1 on total T cells and their HBV-specific subset,46,47 and telbuvidine has also been associated with improved production of IL-21 and recovery of follicular helper T cells, which provide functional help to both T and B cells.50 However, this recovery of T-cell function remains incomplete and quantitatively not comparable to adaptive immune responses of patients who spontaneously control HBV infection. It is mainly detectable with assays that are based on in vitro expansion of T cells and not readily observed with ex vivo assays.51 Furthermore, restoration of HBV-specific T-cell responses can be transient, which may contribute to the frequent failure of HBV control when treatment is discontinued.43,52 Likewise, NUC-mediated HBV suppression is not sufficient to restore responsiveness to HBsAg vaccination53 and lamivudine/IL-12 combination therapy is equally ineffective in boosting HBV-specific T-cell responses.54
While a consensus exists on the ability of NUC therapy to partially recover HBV-specific T-cell function, the underlying mechanisms are controversial. It has been suggested that a substantial decline of HBV antigen levels in the blood is required for recovery of T-cell function, because high HBV antigen levels are regarded as the cause of HBV-specific T-cell deletion and exhaustion in natural infection.14 Unfortunately, with the exception of very few patients who clear HBsAg during treatment,49 NUCs have little impact on HBV antigen levels,55 particularly in the first year of therapy when HBV-specific T-cell recovery is more pronounced. As an alternative hypothesis it has been proposed that decreased HBV replication by limiting HSP60 release from infected hepatocytes, results in a secondary decrease in the frequency of CD4 T cells with regulatory/suppressor capacity (Tregs).56 According to that hypothesis, NUC-mediated inhibition of HBV replication and reduction in Treg frequency would enhance the HBV-specific T-cell response.57 However, even though a reduced frequency of circulating Treg cells was observed in entecavir-, adefovir-, and tenofovir-treated patients,56,58,59 it is not clear whether this is the result of reduced HBV replication or reduced liver inflammation.
In this context, it is surprising that a direct immunomodulatory effect of NUCs has not been contemplated in HBV infection. Tenofovir and adefovir have been shown to increase the production of TNF-α, IL-6, IL-10, CCL5 (Rantes), and CCR5 (MIP-1α) in human monocytes.60 Tenofovir has also been shown to modulate Toll-like receptor (TLR)-mediated responses and to reduce the threshold of T-cell activation.61 A better evaluation of these direct immune effects is attractive because further attempts to boost HBV-specific immune responses will be performed in patients in whom HBV replication is suppressed by some of the potent NUCs. Along this line, it has been shown that entecavir enhances the effects of therapeutic vaccination in the woodchuck model of chronic hepatitis.62
IFN-α/NUC Combination Therapy
The limited efficacy of IFN-α or NUC monotherapy in achieving HBsAg seroconversion and/or sustained HBV control was the rationale for studies that combine the two classes of drugs. Because NUCs potently inhibit HBV replication and partially restore HBV-specific T-cell responses, and because IFN-α targets HBV cccDNA and stimulates NK cell responses, the combination of IFN-α and NUCs may generate a better antiviral response and immune recovery in patients with chronic HBV infection. Combination NUC/PegIFN-α therapy does indeed have a slightly better on-treatment antiviral effect in HBeAg+ and in HBeAg- patients than IFN-α or NUC monotherapy, but the rate of sustained off-treatment responses is not higher than with PegIFN-α monotherapy.63–68 A longitudinal study of lamivudine and PegIFN-α-treated children with infancy-acquired HBV infection, a patient group that is considered immunotolerant, demonstrated a recovery of HBV-specific T-cell responses in those who responded with HBsAg loss.69 The magnitude of the recovered T-cell response was, however, weak and a comparison with children treated with lamivudine monotherapy was not performed. Based on recent data, an extended period of viral suppression prior to NUC/PegIFN-α combination therapy may increase the HBs seroconversion rate, as evidenced by HBsAg loss in 60% of patients who were treated with PegIFN and entecavir after more than 3 years of complete viral suppression by entecavir monotherapy.70
Immunological Aspects of HCV Antiviral Therapy
In contrast to HBV, HCV can be completely cleared by an individual's immune response. Key mediators of spontaneous HCV clearance are virus-specific T cells (reviewed in71), which remain readily detectable in the circulation for decades after clearance.72 Studies in chimpanzees were important to demonstrate that these T cells protect upon reinfection because depletion of either CD4 or CD8 T cells after recovery from a primary challenge abrogates protective immunity upon HCV rechallenge.73,74 Based on the fact that acute HCV infection can be cleared spontaneously, effective antiviral therapy of chronic infection should be able to completely eliminate HCV without the risk of relapse. This is supported by the experience that a sustained virological response (SVR) after IFN-based therapy is durable in more than 97% of patients.75
Over the past three decades, treatment regimens have evolved from IFN-α to PegIFN-α/ribavirin combination therapy,76 to inclusion of the first protease inhibitors and, most recently, to potent interferon-free combinations of two and three direct-acting antivirals.77,78 In addition, the use of IFN-α has stimulated virological and immunological studies on host responses to endogenous and therapeutic IFN-α. This line of research has, for example, led to the discovery of genetic determinants of IFN-responsiveness79–82 and to the identification and characterization of key molecules in antiviral defense, such as the signaling molecule MAVS83 and a broad spectrum of antiviral IFN-stimulated genes.84
IFN-α-Based Therapy
IFN-α was used to treat patients with chronic HCV infection, then known as non-A non-B hepatitis, years prior to the identification of the virus itself.85 Treatment outcomes improved significantly after the nucleoside analog ribavirin was added to prevent relapse86 and again, after IFN-α's pharmacokinetics was optimized by pegylation (PegIFN-α).87 Treatment responders typically exhibit a 1-2 log10 decline in viremia within 1-2 days of treatment initiation, a response that is, as discussed, much faster than that observed in HBV-infected patients. This first-phase response is thought to be due to inhibition of HCV replication and is followed by a slower second-phase response.21 The overall response to therapy is determined by viral factors such as viral titer and genotype and by host factors, which include age, race, gender, cirrhosis, comorbidities, and the baseline activation level of the endogenous IFN-α response.
IFN-α-based therapy of acute HCV infection is highly successful, with >90% of patients responding.88 This may be due to the fact that a type II IFN signature prevails at the time of maximal intrahepatic T-cell responses in acute infection (due to IFN-γ production by HCV-specific T cells) and that type I IFN-induced negative feedback and resistance mechanisms are not yet full induced.89 In contrast, IFN-α-based therapy has much lower response rates in chronic HCV infection. High pretreatment levels of type I and type III interferon induced genes and the presence of the unfavorable rs12979860-T allele in the IFNL3 gene locus79–81 are associated with treatment nonresponse. Presence of the ΔG allele of the ss469415590 variant (IFNL4-ΔG) adds negative predictive value in African Americans.82 While the molecular mechanisms that underlie these associations require further investigation, it has become clear that high baseline ISG levels limit further induction of these genes during IFN-α-based therapy. Induction of negative feedback mechanisms, such as SOCS likely contributes to this refractoriness.90
A patient's responsiveness to IFN-α-based therapy can also be assessed by the activation of innate immune cells. Similar to what is observed during IFN-α-based therapy of chronic hepatitis B,20 NK cells are strongly activated within hours of IFN-α injection with increased cytotoxicity and decreased IFN-γ production.91 This response pattern is due to IFN-α-mediated induction of STAT1 in NK cells, which displaces STAT4 at the IFN-α/β receptor.92 Continued IFN-α stimulation then results in preferential STAT1 over STAT4 phosphorylation, with an increase in pSTAT1-dependent NK cell cytotoxicity and a decrease in pSTAT4-dependent IFN-γ production. Accordingly, NK cells from patients with a rapid first phase HCV RNA decline exhibit maximal responsiveness to type I IFN as evidenced by a high induction of pSTAT1 in NK cells compared to patients with slow HCV RNA decline.93 This is consistent with greater NK cell cytotoxicity in treatment responders than nonresponders.91
The negative correlation between pretreatment and on-treatment activation of IFN-α-responsive immune cells such as NK cells recapitulates what has been reported for intracellular ISGs in hepatocytes. For example, the expression levels of the activating NK cell receptors NKp30, NKp46, and DNAM-1 correlate inversely with the increase in these parameters during IFN-α-based treatment and the subsequent virological treatment response.94 Similar to chronic activation of the innate immune response, chronic activation of the adaptive immune response also predicts treatment failure. This is evidenced by increased pretreatment expression of the programmed death 1 (PD-1) molecule on lymphocytes of treatment nonresponders.95 Expression of PD-1 reflects T-cell activation by way of the T-cell receptor,96 and PD-1 blockade by itself, is not sufficient to induce HCV clearance in the chimpanzee model.97 Longitudinal analysis of HCV-specific T-cell reactivity during IFN-based therapy has shown some differences between those with a fast and slow early response.98 Overall, however, IFN-α-based therapy suppresses rather than stimulates HCV-specific T cells. As in chronic HBV infection, the frequency of HCV-specific T cells and their capacity to produce IFN-γ decreases in HCV-infected patients who undergo IFN-α-based therapy.99 An exception is treatment during the acute phase of HCV infection. Although the bulk HCV-specific T-cell response as measured by IFN-γ production in response to overlapping HCV peptides also decreases when patients with acute HCV infection are treated,100,101 a very small population of HCV-specific memory T cells can be rescued.102
Ribavirin
The guanosine analogue ribavirin is a component not only of interferon-based therapies for HCV but also of new therapies with direct-acting antivirals (DAAs). While ribavirin itself only has a modest effect on HCV RNA levels and does not induce viral clearance,103 it is associated with a faster second-phase virological response when administered together with PegIFN-α,104 and it decreases on-treatment virological breakthrough and posttreatment virological relapse105 and/or DAAs.106 Several direct effects on viral replication have been proposed as ribavirin's mechanism of action. These include inhibition of the HCV NS5B-encoded RNA polymerase, depletion of guanosine triphosphates by interference with a host enzyme, and promotion of viral error catastrophe (reviewed in107). While each of these mechanisms is supported by in vitro data, several in vivo observations argue against a primarily antiviral mode of action. For example, ribavirin monotherapy results only in a modest, less than a 1 log10 decline in viral levels,104 does not increase the rate of nucleotide substitutions,108 and does not improve the first-phase virological decline in combination therapy with PegIFN-α.109 An immunomodulatory effect has been proposed because ribavirin induces a set of ISGs that differs from those induced by IFN-α110,111 and may improve the ability of the HCV-infected liver to response to IFN-α-based therapy.112 Consistent with this proposed synergistic action, ribavirin accelerates the second-phase virological response in patients with a suboptimal, but not absent, interferon response.113 In addition, ribavirin counteracts IFN-α's negative effect on NK cell IFN-γ production to some extent114 and improved IFN-γ production by NK cells correlates with the second-phase virological response.114 Whether it also contributes to better immunosurveillance and prevention of virological relapse is an interesting question for further studies. Finally, ribavirin was shown to enhance T-cell function and to inhibit production of the immunosuppressive cytokine IL-10 in vitro115 but this has not been confirmed with T cells from treated patients.
DAAs
The development of interferon-free regimens that combine two or three DAAs and achieve high response rates for genotypes 1, 2, and 3 infections77,78 is exciting not only from a clinical, but also from an immunological perspective. A key question is whether effective inhibition of HCV replication restores defective innate and adaptive immune responses. Inhibition of HCV replication by interferon-free DAA regimens may decrease the virus-induced ISG signature and restore IFN-responsiveness. It would also be of interest to investigate whether these regimens recover NK cell IFN-γ production, which is suppressed in chronic HCV infection.116,117 Finally, it is important to investigate whether exhausted and dysfunctional T-cell responses recover. A recent study demonstrates that both the frequency of HCV-specific CD8 T cells in the blood and their proliferative in vitro response increases in DAA responders but not in nonresponders.118 Of note, DAA-mediated viral clearance preferentially increased the frequency of T cells that targeted conserved HCV epitopes—most of these T cells were thought to be exhausted due to chronic antigenic stimulation. While it is not possible to distinguish between the outgrowth of a small number of functional T cells and reversion of T-cell exhaustion, and while it is not yet clear whether increased proliferation is associated with increased effector function, it is important that a functional T-cell response may contribute to prevention of viral relapse and establishment of long-term immunity against reinfection. Future studies may clarify the extent and mechanisms of improved T-cell function. A reversion of T-cell exhaustion may also be relevant for other diseases, such as HIV infection and cancer, where T-cell exhaustion has been described.
Future Directions
For chronic hepatitis B, the current treatment regimens rarely result in complete eradication of the virus and its cccDNA. New regimens are needed that complement antiviral therapy with strategies to induce better immune control. This may be achieved by enhancing immune responses directly (TLR-agonists, vaccine therapy, adoptive T-cell therapy) or indirectly (through inhibition of HBsAg production). For chronic HCV infection, the prospect is different because antiviral therapies can completely eradicate the virus. IFN-α-free regimens that are effective against all HCV genotypes and that can be administered orally with minimal side effects appear within reach. However, even with a reduction in treatment duration and costs, these regimens will likely not be affordable in countries where the prevalence of HCV and the incidence of new infections are highest. The development of a prophylactic vaccine that induces protective immunity is therefore still worth pursuing.
Glossary
Abbreviations:
- cccDNA
covalently closed circular DNA
- HBV
hepatitis B virus
- HCV
hepatitis C virus
- IFN-α
interferon-alpha
- ISG
IFN-stimulated gene
- PBMC
peripheral blood mononuclear cells
- SOCS3
suppression of cytokine signaling 3
- WHV
woodchuck hepatitis virus
References
- Hoofnagle JH. Reactivation of hepatitis B. Hepatology. 2009;49:S156–165. doi: 10.1002/hep.22945. [DOI] [PubMed] [Google Scholar]
- 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. doi: 10.1038/nm1096-1104. [DOI] [PubMed] [Google Scholar]
- Levrero M, Pollicino T, Petersen J, Belloni L, Raimondo G, Dandri M. Control of cccDNA function in hepatitis B virus infection. J Hepatol. 2009;51:581–592. doi: 10.1016/j.jhep.2009.05.022. [DOI] [PubMed] [Google Scholar]
- Kwon H, Lok AS. Hepatitis B therapy. Nat Rev Gastroenterol Hepatol. 2011;8:275–284. doi: 10.1038/nrgastro.2011.33. [DOI] [PubMed] [Google Scholar]
- Lucifora J, Xia Y, Reisinger F, Zhang K, Stadler D, Cheng X, et al. Specific and nonhepatotoxic degradation of nuclear hepatitis B virus cccDNA. Science. 2014;343:1221–1228. doi: 10.1126/science.1243462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu F, Campagna M, Qi Y, Zhao X, Guo F, Xu C, et al. Alpha-interferon suppresses hepadnavirus transcription by altering epigenetic modification of cccDNA minichromosomes. PLoS Pathog. 2013;9:e1003613. doi: 10.1371/journal.ppat.1003613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belloni L, Allweiss L, Guerrieri F, Pediconi N, Volz T, Pollicino T, et al. IFN-alpha inhibits HBV transcription and replication in cell culture and in humanized mice by targeting the epigenetic regulation of the nuclear cccDNA minichromosome. J Clin Invest. 2012;122:529–537. doi: 10.1172/JCI58847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pasquetto V, Wieland SF, Uprichard SL, Tripodi M, Chisari FV. Cytokine-sensitive replication of hepatitis B virus in immortalized mouse hepatocyte cultures. J Virol. 2002;76:5646–5653. doi: 10.1128/JVI.76.11.5646-5653.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu C, Guo H, Pan XB, Mao R, Yu W, Xu X, et al. Interferons accelerate decay of replication-competent nucleocapsids of hepatitis B virus. J Virol. 2010;84:9332–9340. doi: 10.1128/JVI.00918-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allweiss L, Volz T, Lutgehetmann M, Giersch K, Bornscheuer T, Lohse AW, et al. Immune cell responses are not required to induce substantial hepatitis B virus antigen decline during pegylated interferon-alpha administration. J Hepatol. 2014;60:500–507. doi: 10.1016/j.jhep.2013.10.021. [DOI] [PubMed] [Google Scholar]
- Dunn C, Peppa D, Khanna P, Nebbia G, Jones M, Brendish N, et al. Temporal analysis of early immune responses in patients with acute hepatitis B virus infection. Gastroenterology. 2009;137:1289–1300. doi: 10.1053/j.gastro.2009.06.054. [DOI] [PubMed] [Google Scholar]
- Wieland S, Thimme R, Purcell RH, Chisari FV. Genomic analysis of the host response to hepatitis B virus infection. Proc Natl Acad Sci U S A. 2004;101:6669–6674. doi: 10.1073/pnas.0401771101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fletcher SP, Chin DJ, Ji Y, Iniguez AL, Taillon B, Swinney DC, et al. Transcriptomic analysis of the woodchuck model of chronic hepatitis B. Hepatology. 2012;56:820–830. doi: 10.1002/hep.25730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertoletti A, Ferrari C. Innate and adaptive immune responses in chronic hepatitis B virus infections: towards restoration of immune control of viral infection. Gut. 2012;61:1754–1764. doi: 10.1136/gutjnl-2011-301073. [DOI] [PubMed] [Google Scholar]
- Wieland SF, Chisari FV. Stealth and cunning: hepatitis B and hepatitis C viruses. J Virol. 2005;79:9369–9380. doi: 10.1128/JVI.79.15.9369-9380.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christen V, Duong F, Bernsmeier C, Sun D, Nassal M, Heim MH. Inhibition of alpha interferon signaling by hepatitis B virus. J Virol. 2007;81:159–165. doi: 10.1128/JVI.01292-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Liu K, Liu Y, Xu Y, Zhang F, Yang H, et al. Exosomes mediate the cell-to-cell transmission of IFN-alpha-induced antiviral activity. Nat Immunol. 2013;14:793–803. doi: 10.1038/ni.2647. [DOI] [PubMed] [Google Scholar]
- Lau GK, Piratvisuth T, Luo KX, Marcellin P, Thongsawat S, Cooksley G, et al. Peginterferon Alfa-2a, lamivudine, and the combination for HBeAg-positive chronic hepatitis B. N Engl J Med. 2005;352:2682–2695. doi: 10.1056/NEJMoa043470. [DOI] [PubMed] [Google Scholar]
- Marcellin P, Lau GK, Bonino F, Farci P, Hadziyannis S, Jin R, et al. Peginterferon alfa-2a alone, lamivudine alone, and the two in combination in patients with HBeAg-negative chronic hepatitis B. N Engl J Med. 2004;351:1206–1217. doi: 10.1056/NEJMoa040431. [DOI] [PubMed] [Google Scholar]
- Micco L, Peppa D, Loggi E, Schurich A, Jefferson L, Cursaro C, et al. Differential boosting of innate and adaptive antiviral responses during pegylated-interferon-alpha therapy of chronic hepatitis B. J Hepatol. 2013;58:225–233. doi: 10.1016/j.jhep.2012.09.029. [DOI] [PubMed] [Google Scholar]
- Neumann AU, Lam NP, Dahari H, Gretch DR, Wiley TE, Layden TJ, et al. Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy. Science. 1998;282:103–107. doi: 10.1126/science.282.5386.103. [DOI] [PubMed] [Google Scholar]
- Tan AT, Hoang LT, Chin D, Rasmussen E, Lopatin U, Hart S, et al. Reduction of HBV replication prolongs the early immunological response to IFNalpha therapy. J Hepatol. 2014;60:54–61. doi: 10.1016/j.jhep.2013.08.020. [DOI] [PubMed] [Google Scholar]
- Penna A, Laccabue D, Libri I, Giuberti T, Schivazappa S, Alfieri A, et al. Peginterferon-α does not improve early peripheral blood HBV-specific T cell response in HbeAg-negative chronic hepatitis. J Hepatol. 2012;56:1239–1246. doi: 10.1016/j.jhep.2011.12.032. [DOI] [PubMed] [Google Scholar]
- Marrack P, Kappler J, Mitchell T. Type I interferons keep activated T cells alive. J Exp Med. 1999;189:521–530. doi: 10.1084/jem.189.3.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shin EC, Seifert U, Kato T, Rice CM, Feinstone SM, Kloetzel PM, et al. Virus-induced type I IFN stimulates generation of immunoproteasomes at the site of infection. J Clin Invest. 2006;116:3006–3014. doi: 10.1172/JCI29832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spadaro F, Lapenta C, Donati S, Abalsamo L, Barnaba V, Belardelli F, et al. IFN-alpha enhances cross-presentation in human dendritic cells by modulating antigen survival, endocytic routing, and processing. Blood. 2012;119:1407–1417. doi: 10.1182/blood-2011-06-363564. [DOI] [PubMed] [Google Scholar]
- Trinchieri G. Type I interferon: friend or foe? J Exp Med. 2010;207:2053–2063. doi: 10.1084/jem.20101664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schurich A, Pallett LJ, Lubowiecki M, Singh HD, Gill US, Kennedy PT, et al. The third signal cytokine IL-12 rescues the anti-viral function of exhausted HBV-specific CD8 T cells. PLoS Pathog. 2013;9:e1003208. doi: 10.1371/journal.ppat.1003208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teijaro JR, Ng C, Lee AM, Sullivan BM, Sheehan KC, Welch M, et al. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science. 2013;340:207–211. doi: 10.1126/science.1235214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson EB, Yamada DH, Elsaesser H, Herskovitz J, Deng J, Cheng G, et al. Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science. 2013;340:202–207. doi: 10.1126/science.1235208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patzwahl R, Meier V, Ramadori G, Mihm S. Enhanced expression of interferon-regulated genes in the liver of patients with chronic hepatitis C virus infection: detection by suppression-subtractive hybridization. J Virol. 2001;75:1332–1338. doi: 10.1128/JVI.75.3.1332-1338.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossol S, Marinos G, Carucci P, Singer MV, Williams R, Naoumov NV. Interleukin-12 induction of Th1 cytokines is important for viral clearance in chronic hepatitis B. J Clin Invest. 1997;99:3025–3033. doi: 10.1172/JCI119498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rehermann B, Lau D, Hoofnagle JH, Chisari FV. Cytotoxic T lymphocyte responsiveness after resolution of chronic hepatitis B virus infection. J Clin Invest. 1996;97:1655–1665. doi: 10.1172/JCI118592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walsh MJ, Jonsson JR, Richardson MM, Lipka GM, Purdie DM, Clouston AD, et al. Non-response to antiviral therapy is associated with obesity and increased hepatic expression of suppressor of cytokine signalling 3 (SOCS-3) in patients with chronic hepatitis C, viral genotype 1. Gut. 2006;55:529–535. doi: 10.1136/gut.2005.069674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peppa D, Micco L, Javaid A, Kennedy PT, Schurich A, Dunn C, et al. Blockade of immunosuppressive cytokines restores NK cell antiviral function in chronic hepatitis B virus infection. PLoS Pathog. 2010;6:e1001227. doi: 10.1371/journal.ppat.1001227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun C, Fu B, Gao Y, Liao X, Sun R, Tian Z, et al. TGF-beta1 down-regulation of NKG2D/DAP10 and 2B4/SAP expression on human NK cells contributes to HBV persistence. PLoS Pathog. 2012;8:e1002594. doi: 10.1371/journal.ppat.1002594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Das A, Maini MK. Innate and adaptive immune responses in hepatitis B virus infection. Dig Dis. 2010;28:126–132. doi: 10.1159/000282075. [DOI] [PubMed] [Google Scholar]
- Zoulim F, Locarnini S. Hepatitis B virus resistance to nucleos(t)ide analogues. Gastroenterology. 2009;137:1593–1608. doi: 10.1053/j.gastro.2009.08.063. [DOI] [PubMed] [Google Scholar]
- Lutgehetmann M, Bornscheuer T, Volz T, Allweiss L, Bockmann JH, Pollok JM, et al. Hepatitis B virus limits response of human hepatocytes to interferon-alpha in chimeric mice. Gastroenterology. 2011;140:2074–2083. doi: 10.1053/j.gastro.2011.02.057. [DOI] [PubMed] [Google Scholar]
- van der Molen RG, Sprengers D, Biesta PJ, Kusters JG, Janssen HL. Favorable effect of adefovir on the number and functionality of myeloid dendritic cells of patients with chronic HBV. Hepatology. 2006;44:907–914. doi: 10.1002/hep.21340. [DOI] [PubMed] [Google Scholar]
- Tjwa ET, van Oord GW, Hegmans JP, Janssen HL, Woltman AM. Viral load reduction improves activation and function of natural killer cells in patients with chronic hepatitis B. J Hepatol. 2011;54:209–218. doi: 10.1016/j.jhep.2010.07.009. [DOI] [PubMed] [Google Scholar]
- Boni C, Bertoletti A, Penna A, Cavalli A, Pilli M, Urbani S, et al. Lamivudine treatment can restore T cell responsiveness in chronic hepatitis B. J Clin Invest. 1998;102:968–975. doi: 10.1172/JCI3731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boni C, Penna A, Bertoletti A, Lamonaca V, Rapti I, Missale G, et al. Transient restoration of anti-viral T cell responses induced by lamivudine therapy in chronic hepatitis B. J Hepatol. 2003;39:595–605. doi: 10.1016/s0168-8278(03)00292-7. [DOI] [PubMed] [Google Scholar]
- Boni C, Penna A, Ogg GS, Bertoletti A, Pilli M, Cavallo C, et al. Lamivudine treatment can overcome cytotoxic T-cell hyporesponsiveness in chronic hepatitis B: new perspectives for immune therapy. Hepatology. 2001;33:963–971. doi: 10.1053/jhep.2001.23045. [DOI] [PubMed] [Google Scholar]
- Cooksley H, Chokshi S, Maayan Y, Wedemeyer H, Andreone P, Gilson R, et al. Hepatitis B virus e antigen loss during adefovir dipivoxil therapy is associated with enhanced virus-specific CD4+ T-cell reactivity. Antimicrob Agents Chemother. 2008;52:312–320. doi: 10.1128/AAC.00467-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans A, Riva A, Cooksley H, Phillips S, Puranik S, Nathwani A, et al. Programmed death 1 expression during antiviral treatment of chronic hepatitis B: impact of hepatitis B e-antigen seroconversion. Hepatology. 2008;48:759–769. doi: 10.1002/hep.22419. [DOI] [PubMed] [Google Scholar]
- Chen Y, Li X, Ye B, Yang X, Wu W, Chen B, et al. Effect of telbivudine therapy on the cellular immune response in chronic hepatitis B. Antiviral Res. 2011;91:23–31. doi: 10.1016/j.antiviral.2011.04.008. [DOI] [PubMed] [Google Scholar]
- TrehanPati N, Kotillil S, Hissar SS, Shrivastava S, Khanam A, Sukriti S, et al. Circulating Tregs correlate with viral load reduction in chronic HBV-treated patients with tenofovir disoproxil fumarate. J Clin Immunol. 2011;31:509–520. doi: 10.1007/s10875-011-9509-7. [DOI] [PubMed] [Google Scholar]
- Wursthorn K, Jung M, Riva A, Goodman ZD, Lopez P, Bao W, et al. Kinetics of hepatitis B surface antigen decline during 3 years of telbivudine treatment in hepatitis B e antigen-positive patients. Hepatology. 2010;52:1611–1620. doi: 10.1002/hep.23905. [DOI] [PubMed] [Google Scholar]
- Li Y, Ma S, Tang L, Wang W, Huang X, Lai Q, et al. Circulating chemokine (C-X-C Motif) receptor 5(+) CD4(+) T cells benefit hepatitis B e antigen seroconversion through IL-21 in patients with chronic hepatitis B virus infection. Hepatology. 2013;58:1277–1286. doi: 10.1002/hep.26489. [DOI] [PubMed] [Google Scholar]
- Boni C, Laccabue D, Lampertico P, Giuberti T, Vigano M, Schivazappa S, et al. Restored function of HBV-specific T cells after long-term effective therapy with nucleos(t)ide analogues. Gastroenterology. 2012;143:963–973. doi: 10.1053/j.gastro.2012.07.014. [DOI] [PubMed] [Google Scholar]
- Tan AT, Koh S, Goh W, Zhe HY, Gehring AJ, Lim SG, et al. A longitudinal analysis of innate and adaptive immune profile during hepatic flares in chronic hepatitis B. J Hepatol. 2010;52:330–339. doi: 10.1016/j.jhep.2009.12.015. [DOI] [PubMed] [Google Scholar]
- Vandepapeliere P, Lau GK, Leroux-Roels G, Horsmans Y, Gane E, Tawandee T, et al. Therapeutic vaccination of chronic hepatitis B patients with virus suppression by antiviral therapy: a randomized, controlled study of co-administration of HBsAg/AS02 candidate vaccine and lamivudine. Vaccine. 2007;25:8585–8597. doi: 10.1016/j.vaccine.2007.09.072. [DOI] [PubMed] [Google Scholar]
- Rigopoulou EI, Suri D, Chokshi S, Mullerova I, Rice S, Tedder RS, et al. Lamivudine plus interleukin-12 combination therapy in chronic hepatitis B: antiviral and immunological activity. Hepatology. 2005;42:1028–1036. doi: 10.1002/hep.20888. [DOI] [PubMed] [Google Scholar]
- Wong DK, Seto WK, Fung J, Ip P, Huang FY, Lai CL, et al. Reduction of hepatitis B surface antigen and covalently closed circular DNA by nucleos(t)ide analogues of different potency. Clin Gastroenterol Hepatol. 2013;11:1004–1010. doi: 10.1016/j.cgh.2013.01.026. [DOI] [PubMed] [Google Scholar]
- Kondo Y, Ueno Y, Kobayashi K, Kakazu E, Shiina M, Inoue J, et al. Hepatitis B virus replication could enhance regulatory T cell activity by producing soluble heat shock protein 60 from hepatocytes. J Infect Dis. 2010;202:202–213. doi: 10.1086/653496. [DOI] [PubMed] [Google Scholar]
- Franzese O, Kennedy PT, Gehring AJ, Gotto J, Williams R, Maini MK, et al. Modulation of the CD8+-T-cell response by CD4+ CD25+ regulatory T cells in patients with hepatitis B virus infection. J Virol. 2005;79:3322–3328. doi: 10.1128/JVI.79.6.3322-3328.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang JY, Song CH, Shi F, Zhang Z, Fu JL, Wang FS. Decreased ratio of Treg cells to Th17 cells correlates with HBV DNA suppression in chronic hepatitis B patients undergoing entecavir treatment. PLoS One. 2010;5:e13869. doi: 10.1371/journal.pone.0013869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stoop JN, van der Molen RG, Kuipers EJ, Kusters JG, Janssen HL. Inhibition of viral replication reduces regulatory T cells and enhances the antiviral immune response in chronic hepatitis B. Virology. 2007;361:141–148. doi: 10.1016/j.virol.2006.11.018. [DOI] [PubMed] [Google Scholar]
- Zidek Z, Frankova D, Holy A. Activation by 9-(R)-[2-(phosphonomethoxy)propyl]adenine of chemokine (RANTES, macrophage inflammatory protein 1alpha) and cytokine (tumor necrosis factor alpha, interleukin-10 [IL-10], IL-1beta) production. Antimicrob Agents Chemother. 2001;45:3381–3386. doi: 10.1128/AAC.45.12.3381-3386.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melchjorsen J, Risor MW, Sogaard OS, O'Loughlin KL, Chow S, Paludan SR, et al. Tenofovir selectively regulates production of inflammatory cytokines and shifts the IL-12/IL-10 balance in human primary cells. J Acquir Immune Defic Syndr. 2011;57:265–275. doi: 10.1097/QAI.0b013e3182185276. [DOI] [PubMed] [Google Scholar]
- Kosinska AD, Zhang E, Johrden L, Liu J, Seiz PL, Zhang X, et al. Combination of DNA prime—adenovirus boost immunization with entecavir elicits sustained control of chronic hepatitis B in the woodchuck model. PLoS Pathog. 2013;9:e1003391. doi: 10.1371/journal.ppat.1003391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janssen HL, van Zonneveld M, Senturk H, Zeuzem S, Akarca US, Cakaloglu Y, et al. Pegylated interferon alfa-2b alone or in combination with lamivudine for HBeAg-positive chronic hepatitis B: a randomised trial. Lancet. 2005;365:123–129. doi: 10.1016/S0140-6736(05)17701-0. [DOI] [PubMed] [Google Scholar]
- Lau GK, Piratvisuth T, Luo KX, Marcellin P, Thongsawat S, Cooksley G, et al. Peginterferon Alfa-2a, lamivudine, and the combination for HBeAg-positive chronic hepatitis B. N Engl J Med. 2005;352:2682–2695. doi: 10.1056/NEJMoa043470. [DOI] [PubMed] [Google Scholar]
- Marcellin P, Lau GK, Bonino F, Farci P, Hadziyannis S, Jin R, et al. Peginterferon alfa-2a alone, lamivudine alone, and the two in combination in patients with HBeAg-negative chronic hepatitis B. N Engl J Med. 2004;351:1206–1217. doi: 10.1056/NEJMoa040431. [DOI] [PubMed] [Google Scholar]
- Piccolo P, Lenci I, Demelia L, Bandiera F, Piras MR, Antonucci G, et al. A randomized controlled trial of pegylated interferon-alpha2a plus adefovir dipivoxil for hepatitis B e antigen-negative chronic hepatitis B. Antivir Ther. 2009;14:1165–1174. doi: 10.3851/IMP1466. [DOI] [PubMed] [Google Scholar]
- Wursthorn K, Lutgehetmann M, Dandri M, Volz T, Buggisch P, Zollner B, et al. Peginterferon alpha-2b plus adefovir induce strong cccDNA decline and HBsAg reduction in patients with chronic hepatitis B. Hepatology. 2006;44:675–684. doi: 10.1002/hep.21282. [DOI] [PubMed] [Google Scholar]
- Takkenberg B, Terpstra V, Zaaijer H, Weegink C, Dijkgraaf M, Jansen P, et al. Intrahepatic response markers in chronic hepatitis B patients treated with peginterferon alpha-2a and adefovir. J Gastroenterol Hepatol. 2011;26:1527–1535. doi: 10.1111/j.1440-1746.2011.06766.x. [DOI] [PubMed] [Google Scholar]
- Carey I, D'Antiga L, Bansal S, Longhi MS, Ma Y, Mesa IR, et al. Immune and viral profile from tolerance to hepatitis B surface antigen clearance: a longitudinal study of vertically hepatitis B virus-infected children on combined therapy. J Virol. 2011;85:2416–2428. doi: 10.1128/JVI.01449-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ouzan D, Penaranda G, Joly H, Khiri H, Pironti A, Halfon P. Add-on peg-interferon leads to loss of HBsAg in patients with HBeAg-negative chronic hepatitis and HBV DNA fully suppressed by long-term nucleotide analogs. J Clin Virol. 2013;58:713–717. doi: 10.1016/j.jcv.2013.09.020. [DOI] [PubMed] [Google Scholar]
- SH Park, B Rehermann. Immune responses to HCV and other hepatitis viruses. Immunity. 2014;40:13–24. doi: 10.1016/j.immuni.2013.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takaki A, Wiese M, Maertens G, Depla E, Seifert U, Liebetrau A, 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;6:578–582. doi: 10.1038/75063. [DOI] [PubMed] [Google Scholar]
- Grakoui A, Shoukry NH, Woollard DJ, Han JH, Hanson HL, Ghrayeb J, et al. HCV persistence and immune evasion in the absence of memory T cell help. Science. 2003;302:659–662. doi: 10.1126/science.1088774. [DOI] [PubMed] [Google Scholar]
- Shoukry NH, Grakoui A, Houghton M, Chien DY, Ghrayeb J, Reimann KA, et al. Memory CD8+ T cells are required for protection from persistent hepatitis C virus infection. J Exp Med. 2003;197:1645–1655. doi: 10.1084/jem.20030239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Welker MW, Zeuzem S. Occult hepatitis C: how convincing are the current data? Hepatology. 2009;49:665–675. doi: 10.1002/hep.22706. [DOI] [PubMed] [Google Scholar]
- Heim MH. 25 years of interferon-based treatment of chronic hepatitis C: an epoch coming to an end. Nat Rev Immunol. 2013;13:535–542. doi: 10.1038/nri3463. [DOI] [PubMed] [Google Scholar]
- Liang TJ, Ghany MG. Current and future therapies for hepatitis C virus infection. N Engl J Med. 2013;368:1907–1917. doi: 10.1056/NEJMra1213651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang TJ, Ghany MG. Therapy of hepatitis C—back to the future. N Engl J Med. 2014;370:2043–2047. doi: 10.1056/NEJMe1403619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, et al. Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance. Nature. 2009;461:399–401. doi: 10.1038/nature08309. [DOI] [PubMed] [Google Scholar]
- Suppiah V, Moldovan M, Ahlenstiel G, Berg T, Weltman M, Abate ML, et al. IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy. Nat Genet. 2009;41:1100–1104. doi: 10.1038/ng.447. [DOI] [PubMed] [Google Scholar]
- Tanaka Y, Nishida N, Sugiyama M, Kurosaki M, Matsuura K, Sakamoto N, et al. Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C. Nat Genet. 2009;41:1105–1109. doi: 10.1038/ng.449. [DOI] [PubMed] [Google Scholar]
- Prokunina-Olsson L, Muchmore B, Tang W, Pfeiffer RM, Park H, Dickensheets H, et al. A variant upstream of IFNL3 (IL28B) creating a new interferon gene IFNL4 is associated with impaired clearance of hepatitis C virus. Nat Genet. 2013;45:164–171. doi: 10.1038/ng.2521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foy E, Li K, Wang C, Sumpter R, Jr, Ikeda M, Lemon SM, et al. Regulation of interferon regulatory factor-3 by the hepatitis C virus serine protease. Science. 2003;300:1145–1148. doi: 10.1126/science.1082604. [DOI] [PubMed] [Google Scholar]
- Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, et al. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472:481–485. doi: 10.1038/nature09907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoofnagle JH, Mullen KD, Jones DB, Rustgi V, Di Bisceglie A, Peters M, et al. Treatment of chronic non-A,non-B hepatitis with recombinant human alpha interferon. A preliminary report. N Engl J Med. 1986;315:1575–1578. doi: 10.1056/NEJM198612183152503. [DOI] [PubMed] [Google Scholar]
- Reichard O, Norkrans G, Fryden A, Braconier JH, Sonnerborg A, Weiland O. Randomised, double-blind, placebo-controlled trial of interferon alpha-2b with and without ribavirin for chronic hepatitis C. The Swedish Study Group. Lancet. 1998;351:83–87. doi: 10.1016/s0140-6736(97)06088-1. [DOI] [PubMed] [Google Scholar]
- Zeuzem S, Feinman SV, Rasenack J, Heathcote EJ, Lai MY, Gane E, et al. Peginterferon alfa-2a in patients with chronic hepatitis C. N Engl J Med. 2000;343:1666–1672. doi: 10.1056/NEJM200012073432301. [DOI] [PubMed] [Google Scholar]
- Jaeckel E, Cornberg M, Wedemeyer H, Santantonio T, Mayer J, Zankel M, et al. Treatment of acute hepatitis C with interferon alfa-2b. N Engl J Med. 2001;345:1452–1457. doi: 10.1056/NEJMoa011232. [DOI] [PubMed] [Google Scholar]
- Dill MT, Makowska Z, Duong FH, Merkofer F, Filipowicz M, Baumert TF, et al. Interferon-gamma-stimulated genes, but not USP18, are expressed in livers of patients with acute hepatitis C. Gastroenterology. 2012;143:777–786. doi: 10.1053/j.gastro.2012.05.044. e771-776. [DOI] [PubMed] [Google Scholar]
- Sarasin-Filipowicz M, Wang X, Yan M, Duong FH, Poli V, Hilton DJ, et al. Alpha interferon induces long-lasting refractoriness of JAK-STAT signaling in the mouse liver through induction of USP18/UBP43. Mol Cell Biol. 2009;29:4841–4851. doi: 10.1128/MCB.00224-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahlenstiel G, Edlich B, Hogdal LJ, Rotman Y, Noureddin M, Feld JJ, et al. Early changes in natural killer cell function indicate virologic response to interferon therapy for hepatitis C. Gastroenterology. 2011;141:1231–1239. doi: 10.1053/j.gastro.2011.06.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyagi T, Gil MP, Wang X, Louten J, Chu WM, Biron CA. High basal STAT4 balanced by STAT1 induction to control type 1 interferon effects in natural killer cells. J Exp Med. 2007;204:2383–2396. doi: 10.1084/jem.20070401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edlich B, Ahlenstiel G, Azpiroz AZ, Stoltzfus J, Noureddin M, Serti E, et al. Early changes in interferon signaling define natural killer cell response and refractoriness to interferon-based therapy of hepatitis C patients. Hepatology. 2012;55:39–48. doi: 10.1002/hep.24628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Maria A, Fogli M, Mazza S, Basso M, Picciotto A, Costa P, et al. Increased natural cytotoxicity receptor expression and relevant IL-10 production in NK cells from chronically infected viremic HCV patients. Eur J Immunol. 2007;37:445–455. doi: 10.1002/eji.200635989. [DOI] [PubMed] [Google Scholar]
- Golden-Mason L, Klarquist J, Wahed AS, Rosen HR. Cutting edge: programmed death-1 expression is increased on immunocytes in chronic hepatitis C virus and predicts failure of response to antiviral therapy: race-dependent differences. J Immunol. 2008;180:3637–3641. doi: 10.4049/jimmunol.180.6.3637. [DOI] [PubMed] [Google Scholar]
- Rutebemberwa A, Ray SC, Astemborski J, Levine J, Liu L, Dowd KA, et al. High-programmed death-1 levels on hepatitis C virus-specific T cells during acute infection are associated with viral persistence and require preservation of cognate antigen during chronic infection. J Immunol. 2008;181:8215–8225. doi: 10.4049/jimmunol.181.12.8215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuller MJ, Callendret B, Zhu B, Freeman GJ, Hasselschwert DL, Satterfield W, et al. Immunotherapy of chronic hepatitis C virus infection with antibodies against programmed cell death-1 (PD-1) Proc Natl Acad Sci U S A. 2013;110:15001–15006. doi: 10.1073/pnas.1312772110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang KH, Herrmann E, Cooksley H, Tatman N, Chokshi S, Williams R, et al. Relationship between early HCV kinetics and T-cell reactivity in chronic hepatitis C genotype 1 during peginterferon and ribavirin therapy. J Hepatol. 2005;43:776–782. doi: 10.1016/j.jhep.2005.05.024. [DOI] [PubMed] [Google Scholar]
- Pilli M, Zerbini A, Penna A, Orlandini A, Lukasiewicz E, Pawlotsky J, et al. Role of the HCV- specific T cell response in relation to viral kinetics and treatment outcome in PEG-Interferon-alpha-2a and Ribavirin therapy (DITTO-HCV project) for chronic hepatitis C. Gastroenterology. 2007;133:1132–1143. doi: 10.1053/j.gastro.2007.06.059. [DOI] [PubMed] [Google Scholar]
- Rahman F, Heller T, Sobao Y, Mizukoshi E, Nascimbeni M, Alter H, et al. Effects of antiviral therapy on the cellular immune response in acute hepatitis C. Hepatology. 2004;40:87–97. doi: 10.1002/hep.20253. [DOI] [PubMed] [Google Scholar]
- Lauer GM, Lucas M, Timm J, Ouchi K, Kim AY, Day CL, et al. Full-breadth analysis of CD8+ T-cell responses in acute hepatitis C virus infection and early therapy. J Virol. 2005;79:12979–12988. doi: 10.1128/JVI.79.20.12979-12988.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badr G, Bedard N, Abdel-Hakeem MS, Trautmann L, Willems B, Villeneuve JP, et al. Early interferon therapy for hepatitis C virus infection rescues polyfunctional, long-lived CD8+ memory T cells. J Virol. 2008;82:10017–10031. doi: 10.1128/JVI.01083-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawlotsky JM, Dahari H, Neumann AU, Hezode C, Germanidis G, Lonjon I, et al. Antiviral action of ribavirin in chronic hepatitis C. Gastroenterology. 2004;126:703–714. doi: 10.1053/j.gastro.2003.12.002. [DOI] [PubMed] [Google Scholar]
- Rotman Y, Noureddin M, Feld JJ, Guedj J, Witthaus M, Han H, et al. Effect of ribavirin on viral kinetics and liver gene expression in chronic hepatitis C. Gut. 2014;63:161–169. doi: 10.1136/gutjnl-2012-303852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Goncales FL, Jr, et al. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med. 2002;347:975–982. doi: 10.1056/NEJMoa020047. [DOI] [PubMed] [Google Scholar]
- Hezode C, Forestier N, Dusheiko G, Ferenci P, Pol S, Goeser T, et al. Telaprevir and peginterferon with or without ribavirin for chronic HCV infection. N Engl J Med. 2009;360:1839–1850. doi: 10.1056/NEJMoa0807650. [DOI] [PubMed] [Google Scholar]
- Feld JJ. Is there a role for ribavirin in the era of hepatitis C virus direct-acting antivirals? Gastroenterology. 2012;142:1356–1359. doi: 10.1053/j.gastro.2011.12.064. [DOI] [PubMed] [Google Scholar]
- Dietz J, Schelhorn SE, Fitting D, Mihm U, Susser S, Welker MW, et al. Deep sequencing reveals mutagenic effects of ribavirin during monotherapy of hepatitis C virus genotype 1-infected patients. J Virol. 2013;87:6172–6181. doi: 10.1128/JVI.02778-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrmann E, Lee JH, Marinos G, Modi M, Zeuzem S. Effect of ribavirin on hepatitis C viral kinetics in patients treated with pegylated interferon. Hepatology. 2003;37:1351–1358. doi: 10.1053/jhep.2003.50218. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Jamaluddin M, Wang S, Tian B, Garofalo RP, Casola A, et al. Ribavirin treatment up-regulates antiviral gene expression via the interferon-stimulated response element in respiratory syncytial virus-infected epithelial cells. J Virol. 2003;77:5933–5947. doi: 10.1128/JVI.77.10.5933-5947.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas E, Feld JJ, Li Q, Hu Z, Fried MW, Liang TJ. Ribavirin potentiates interferon action by augmenting interferon-stimulated gene induction in hepatitis C virus cell culture models. Hepatology. 2011;53:32–41. doi: 10.1002/hep.23985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feld JJ, Nanda S, Huang Y, Chen W, Cam M, Pusek SN, et al. Hepatic gene expression during treatment with peginterferon and ribavirin: identifying molecular pathways for treatment response. Hepatology. 2007;46:1548–1563. doi: 10.1002/hep.21853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feld JJ, Lutchman GA, Heller T, Hara K, Pfeiffer JK, Leff RD, et al. Ribavirin improves early responses to peginterferon through improved interferon signaling. Gastroenterology. 2010;139:154–162. doi: 10.1053/j.gastro.2010.03.037. e154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Werner JM, Serti E, Chepa-Lotrea X, Stoltzfus JD, Ahlenstiel G, Feld JJ, et al. Ribavirin resets the STAT4-dependent IFN-γ responsivesness of natural killer cells in hepatitis C. Hepatology. 2013;58:1106A. doi: 10.1002/hep.27092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnes E, Harcourt G, Brown D, Lucas M, Phillips R, Dusheiko G, et al. The dynamics of T-lymphocyte responses during combination therapy for chronic hepatitis C virus infection. Hepatology. 2002;36:743–754. doi: 10.1053/jhep.2002.35344. [DOI] [PubMed] [Google Scholar]
- Ahlenstiel G, Titerence RH, Koh C, Edlich B, Feld JJ, Rotman Y, et al. Natural killer cells are polarized toward cytotoxicity in chronic hepatitis C in an interferon-alfa-dependent manner. Gastroenterology. 2010;138:325–335. doi: 10.1053/j.gastro.2009.08.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliviero B, Varchetta S, Paudice E, Michelone G, Zaramella M, Mavilio D, et al. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology. 2009;137:1151–1160. doi: 10.1053/j.gastro.2009.05.047. 1160 e1151-1157. [DOI] [PubMed] [Google Scholar]
- Martin M, Hennecke N, Lohmann V, Kayser A, Neumann-Haefelin C, Kukolj G, et al. Restoration of HCV-specific CD8 T cell function by interferon-free therapy. J Hepatol. 2014 doi: 10.1016/j.jhep.2014.05.043. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]