Editorial Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. May 28, 2012; 18(20): 2443-2451
Published online May 28, 2012. doi: 10.3748/wjg.v18.i20.2443
T cell immunopathogenesis and immunotherapeutic strategies for chronic hepatitis B virus infection
Yukihiro Shimizu, Gastroenterology Unit, Takaoka City Hospital, 4-1 Takara-machi, Takaoka city, Toyama 933-8550, Japan
Author contributions: Shimizu Y designed and wrote the manuscript.
Correspondence to: Yukihiro Shimizu, MD, PhD, Gastoenterology Unit, Takaoka City Hospital, 4-1 Takara-machi, Takaoka city, Toyama 933-8550, Japan. rsf14240@nifty.com
Telephone: +81-766-230204 Fax: +81-766-262882
Received: October 17, 2011
Revised: February 8, 2012
Accepted: February 26, 2012
Published online: May 28, 2012

Abstract

Hepatitis B is caused by the host immune response and T cells play a major role in the immunopathogenesis. More importantly, T cells not only destroy hepatocytes infected by hepatitis B virus (HBV), but also control HBV replication or eradicate HBV in a noncytolytic manner. Therefore, analysis of T cell immune response during acute and chronic HBV infection is important to develop a strategy for successful viral control, which could lead to immunotherapy for terminating persistent HBV infection. There have been many attempts at immunotherapy for chronic HBV infection, and some have shown promising results. High viral load has been shown to suppress antiviral immune responses and immunoinhibitory signals have been recently elucidated, therefore, viral suppression by nucleos(t)ide analogs, stimulation of antiviral immune response, and suppression of the immunoinhibitory signals must be combined to achieve desirable antiviral effects.

Key Words: T cells, Immunopathogenesis, Immunotherapy, Hepatitis B virus infection



INTRODUCTION

Hepatitis B virus (HBV) is not cytopathic, and hepatitis B is caused by the host immune response, mainly T-cell-mediated, against virus-related peptides expressed on hepatocytes in conjunction with human leukocyte antigens (HLAs). In acute self-limiting hepatitis, a broad T-cell immune response occurs that is strong enough to eradicate the virus or suppress viral replication[1]. However, there are many mechanisms that hamper the antiviral immune response, leading to persistent infection. To develop an optimal strategy to stimulate antiviral immune response with therapeutic potential, extensive analyses of immune mechanisms for successful viral eradication and immunosuppressive mechanisms induced by viral infection during persistent infection are required. In this review, I focus on T cell immune response during HBV infection, and summarize attempted immunotherapeutic approaches against persistent HBV infection.

T CELL RESPONSE IN ACUTE HBV INFECTION

Immunological analysis has been extensively performed in transgenic and chimpanzee models of acute HBV infection. In one model, transgenic mice, in which infectious HBV virions replicate in the liver with expression of all HBV-related antigens, were injected with hepatitis B surface antigen (HBsAg)-specific cytotoxic T lymphocytes (CTLs) that had been induced in nontransgenic mice. The injected CTLs produced interferon (IFN)-γ and tumor necrosis factor (TNF)-α, which purged viral RNA and DNA without destroying infected hepatocytes[2-4]. Importantly, this noncytolytic clearance of intracellular HBV is more efficient at controlling HBV replication than the killing of infected hepatocytes. In this sense, hepatitis is not only a harmful event but also represents an effective mechanism by which CTLs suppress HBV. Noncytolytic viral eradication can account for recovery from acute HBV infection in that most HBV is cleared from hepatocytes with only a fraction of the hepatocytes being destroyed. This was confirmed in a chimpanzee infection model; HBV DNA level was markedly decreased in the liver and blood of acutely infected chimpanzees before peak serum alanine aminotransferase (ALT) concentrations were reached[5], suggesting that this noncytopathic T cell effector mechanism results in early viral inhibition or eradication, whereas a cytopathic T cell effector mechanism is required to eliminate the remaining virus by destroying infected hepatocytes.

In humans, the HBV-specific T cell response during incubation phase of acute hepatitis B has been analyzed extensively using HLA class I tetramer and cytokine staining[6]. The data showed that maximal reduction in HBV DNA in the serum occurred before the peak of ALT elevation; again indicating that suppression of HBV replication occurs without hepatocyte injury. Moreover, infiltration of HBV-specific CD8+ T cells into the liver has been observed several weeks before the peak of liver injury, suggesting that HBV-specific T cell infiltration occurs at an early stage of infection, resulting in suppression of HBV replication. Thereafter, recruitment of mostly nonspecific cells induced by cytokines or chemokines produced by HBV-specific T cells contributes to significant liver damage. Interestingly, in the HBV transgenic mouse model of acute hepatitis, administration of antibodies against the chemokines, IFN-inducible protein (IP-10) and monokine induced by interferon-(Mig), reduced the recruitment of mostly antigen-nonspecific mononuclear cells into the liver that had been induced by cytokines and chemokines produced by injected CTLs, leading to a reduction in the severity of hepatitis without affecting the antiviral activity of the CTLs[7]. These observations have important therapeutic implications, because suppression of antigen-nonspecific mononuclear cell recruitment may suppress hepatitis, while retaining the antiviral function of the CTLs.

The overall data from studies in chimpanzees and humans are essentially the same, and indicate that a sufficient T cell response to HBV at an early phase of infection is important for eradication of virus infection, and that an insufficient T cell response may lead to persistent viral infection.

The contributions of CD4+ and CD8+ T cells to the control of viral infection have been analyzed in a chimpanzee model of acute hepatitis B by depleting either T cell population with monoclonal antibodies. The data show that CD8+ T cells are the main effector cells responsible for virus elimination[8].

Antigen specificity of T cell response in acute HBV infection

The antigen specificity of the T cell response to HBV in acute hepatitis has been analyzed, and it is clear that acute viral hepatitis involves a vigorous CTL response to multiple epitopes in the viral nucleocapsid, envelope, and polymerase proteins, whereas these are not seen in patients with chronic hepatitis[1]. Although multi-specificity of the CTL response is characteristic in acute hepatitis, there is known to be a hierarchy of epitope-specific CD8+ T cell responses determined by cytokine production after peptide stimulation. In acute hepatitis B, CD8+ T cell response to HBc18-27 (HLA-A2 restricted epitope) is dominant followed by the response to polymerase epitope (455-463), whereas envelope epitopes are always subdominant[9]. The hierarchy is clearly distinct from that observed in chronic hepatitis, in which the CD8+ T cell response to envelope epitope (183-191) is always dominant. Interestingly, chronic hepatitis patients with lower HBV DNA levels in the serum show greater responses to HBc18-27 than those with high HBV DNA. These findings imply that the T cell response to hepatitis B core antigen (HBcAg) is important for viral control, which is important for designing peptide vaccines for the treatment of chronic HBV infection.

Long-lasting T cell immune response after resolution of acute hepatitis B

In humans, most HBV is cleared after resolution of acute hepatitis. However, it has been shown that trace amounts of HBV DNA can be detected for several years after resolution of acute hepatitis, and the long-lasting memory T cell response is maintained by persistent replication of HBV[10], indicating that low levels of HBV replication could continue in most patients even in the convalescent phase of acute hepatitis in balance with immunological pressure.

T CELL RESPONSE IN CHRONIC HBV INFECTION

In peripheral blood, HBV-specific helper T lymphocytes and CTLs are barely detectable in patients with chronic hepatitis B (CHB)[11], possibly due to exhaustion by high viral load or tolerance to HBV.

In contrast, several studies have characterized intrahepatic CD4+ and CD8+ T lymphocytes in CHB. Intrahepatic CD4+ T lymphocytes in patients with CHB have been found to contain T helper (Th)0 cells, which produce not only IFN-γ, but also interleukin (IL)-4 and IL-5, thus differing from cells in the livers of patients with chronic hepatitis C, which are mostly Th1 cells[12]. CD4+ T lymphocytes that produce IL-17 infiltrate into the livers of patients with CHB and are involved in liver inflammation[13].

Livers of patients with low HBV replication contain intralobular CD8+ T lymphocytes[14], and the percentages of virus-specific T lymphocytes in the liver have been clarified by immunohistochemical staining with peptide-MHC tetramer. The proportion of CD8+ T lymphocytes in the livers of patients with chronic HBV specific for HBc18-27, a major HBV epitope, has been found to range from 0.18% to 1.28%[15]. Maini et al[16] have reported that the number of HBc18-27-specific CD8+ T cells, detected using tetramers, was the same in livers with low HBV DNA/ALT as in those with high HBV DNA/ALT. Hence, HBV-specific T cells recognize HBV antigens and carry out immune surveillance in the liver. Thus, they have an important role in controlling HBV replication in the liver without causing hepatic necroinflammation in low DNA/ALT anti-HBe+ HBV carriers. It remains unknown why HBV-specific T cells fail to control effectively HBV replication in the liver with chronic hepatitis. However, recent advances in immunology have given some insight into the mechanism as described below.

IMMUNOSUPPRESSIVE MECHANISM RESPOSIBLE FOR PERSISTENT HBV INFECTION
Regulatory T cells

Regulatory T (Treg) cells expressing the forkhead family transcription factor, FoxP3, are specialized cells that exert negative control on a variety of physiological and pathological immune responses, resulting in maintenance of immunological self-tolerance[17]. They show diverse phenotypes, occurring in both CD4+ and CD8+ T cell subsets, and express CD25 (IL-2 receptor chain) and/or cytotoxic T-lymphocyte antigen 4 (CTLA-4) in addition to Foxp3.

In HBV infection, hepatitis B e antigen (HBeAg)-positive patients with high HBV DNA levels in the serum show elevated numbers of CD4+CD25+ Treg cells in the blood compared to patients with acute and chronic hepatitis C virus (HCV) infection[18]. Significant accumulation of CD4+CD25+FoxP3+ Treg cells in the liver is found in patients with chronic HBV infection. Moreover, patients with high viral load have a higher proportion of Treg cells in the liver[19], suggesting that intrahepatic Treg cells suppress antiviral immune responses in the liver in chronic HBV infection.

Th cells that produce IL-17 (Th17 cells) have recently been identified as the third subset of effector T cells[20], which produce IL-17A, IL-17F, IL-22 and IL-21[21]. Recently, IL-6 has been shown to induce the generation of Th17 cells from naïve T cells together with transforming growth factor (TGF)-β and inhibits TGF-induced Treg cell differentiation[22]. Importantly, there is a reciprocal relationship between Th17 and Treg cells; not only in development, but also in their effector function, indicating that the Treg/Th17 balance may determine the quality and magnitude of immune responses in the liver[20]. Unexpectedly, the increases in circulating and intrahepatic Th17 cells are positively correlated with HBV DNA in the serum, serum ALT levels, and histological activity index of the livers with CHB, suggesting that activation of Th17 cells does not exert antiviral function in CHB[23].

Programmed death-1

Programmed death-1 (PD-1) is a surface receptor critical for the regulation of T cell function[24,25]. Binding to PD-1 by its ligands PD-L1 and PD-L2 results in the antigen-specific inhibition of T cell proliferation, cytokine production, and cytolytic function, leading to exhaustion of T cells. In the liver, PD-1 is expressed on lymphocytes; PD-L1 is expressed on lymphocytes, hepatocytes and sinusoidal endothelial cells; and PD-L2 is expressed on Kupffer cells and dendritic cells (DCs)[26]. HBeAg-positive patients with high HBV DNA levels in the serum show increased PD-1 and CTLA-4 expression on HBV-specific CD8+ T cells[27]. Moreover, PD-1 expression on CD4+ T cells is correlated positively with serum HBV DNA load in CHB patients[28]. Intrahepatic HBV-specific CD8+ T cells express higher levels of PD-1, and upregulation of intrahepatic PD-1/PD-L1 is associated with liver inflammation and ALT elevation[29]. Although the mechanism underlying the upregulation of PD-1 on CD8+ T cells in the inflamed liver is unknown, signals from PD-1 inhibit HBV-specific T cells, resulting in insufficient antiviral responses, leading to failure of viral control and persistent liver inflammation. Importantly, PD-1/PD-L1 blockade increased CD8+ T cell proliferation and enhanced IFN-γ and IL-2 production by intrahepatic lymphocytes[29]. These findings suggest that inhibition of PD-1/PD-L1 may have therapeutic potential for the control of hepatitis B.

IL-10

IL-10 is an important cytokine with anti-inflammatory properties, and is produced by activated monocytes/macrophages and T cell subsets, including Treg and Th1 cells[30]. Immunosuppression by IL-10 is associated with functional exhaustion of memory T cells in chronic lymphocytic choriomeningitis virus (LCMV) infection, and blockade of IL-10 receptors could terminate chronic LCMV infection[31]. In chronic HBV infection, HBcAg stimulates the production of IL-10, which negatively regulates HBcAg-specific Th17 cell responses in CHB patients[32].

T cell immunoglobulin- and mucin-domain-containing molecule-3

It has been reported that not all exhausted T cells show upregulation of PD-1 and downregulation of CD127 (IL-7 receptor), and blockade of the PD-1/PD-L1 signaling pathway does not always restore proliferation and cytokine production[33]. Recently, another inhibitory molecule, T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3), has been reported. A high frequency of Tim3-expressing CD4+ and CD8+ T cells are found in chronic HBV infection, and the frequency of Tim-3+ T cells is positively correlated with the severity of liver inflammation, and negatively correlated with plasma IFN-γ levels[34].

Dysfunction of DCs

DCs are specialized antigen-presenting cells that orchestrate immune responses. They stimulate innate and acquired immune responses, but also act as tolerogenic cells for immune responses in a variety of situations. In viral hepatitis, dysfunction of DCs from peripheral blood has been reported. In patients with CHB, maturation of DCs from peripheral blood of patients after incubation with cytokines is lower than that of normal subjects with lower expression of HLA-DR and co-stimulatory molecules in the former population[35], leading to low allostimulatory function of DCs from CHB patients. The mechanism of impairment of DC function in patients with CHB is unclear, but both HBV particles and purified HBsAg may have immunomodulatory capacity and may directly contribute to the dysfunction of myeloid DCs[36]. Interestingly, impaired function of monocyte-derived DCs from patients with CHB could be reversed by inhibiting viral replication with nucleos(t)ide analogs such as lamivudine[37]. Type 2 precursor plasmacytoid dendritic cells (pDCs), which are the most important cells in antiviral innate immunity, are also reported to have quantitative and qualitative impairment in patients with chronic HBV infection[38]. Recently, HBV itself was shown to inhibit the functions of pDCs[39]. These data indicate that DCs in patients with CHB have impaired function leading to insufficient T cell response to HBV, which could be the mechanism responsible for persistent viral infection.

IMMUNOTHERAPY FOR VIRAL HEPATITIS

In chronic HBV infection, strong long-term viral suppression can now be achieved with various nucleoside or nucleotide analogs. However, there are some problems that must be solved in the near future. One of the problems with treatment with nucleos(t)ide analogs is a low rate of HBe seroconversion even after long-term administration in HBeAg+ patients. Moreover, reactivation rate of HBV replication is high in both HBeAg+ and HBeAg- patients after cessation of treatment, although drug-free viral controls would be better than long-term administration of the drugs in terms of control of medical costs and avoidance of adverse effects of these agents. It could be possible to achieve long-term viral eradication even after cessation of nucleos(t)ide analogs, if viral suppression with nucleos(t)ide analogs could be combined with efficient immunotherapies.

Previous animal studies and human trials in HBV infection are listed in Tables 1 and 2, respectively.

Table 1 Immunotherapeutic approaches for animal models of hepatitis B virus infection.
Animal modelImmunotherapyResultsRef.
Peptide vaccination
HBV transgenic miceA synthesized fusion peptide, consisting HBcAg18-27 and HIV Tat49-57Decrease in serum HBV DNA levels and the expression levels of HBsAg and HBcAg in the liver[40]
Protein vaccination
HBV transgenic miceHBsAg vaccineMost of the mice showed reduction of HBV DNA levels and disappearance of HBeAg and HBsAg[41]
Woodchuck hepatitis virus infectionCombination of vaccine of HBV large surface protein and clevudineRestoration of T-cell response to Pre-S and S region[42]
DNA immunization
Acute DHBV infectionDNA vaccine expressing DHBc and Pre-S/S and entecavir Boosted with fowl poxvirus vectors expressing DHBc and Pre-S/SClearance of DHBV infection at a rate of 100%[43]
Chronic DHBV infectionDNA vaccine encoding the HBV large envelope and/or core protein with or without lamivudineReduction of viremia and liver DHBV cccDNA in 33% of ducks Seroconversion to anti-pre S in 67% of ducks showing cccDNA clearance[44]
DC immunization
HBV transgenic miceActivated bone marrow-derived DCsBreak CTL tolerance to HBsAg[45]
HBV transgenic miceAnti-CD40 agonistic monoclonal AbInduction of noncytopathic inhibition of HBV replication mediated by antiviral cytokines (IL-12 and TNF-α) produced by activated intrahepatic APCs[46]
HBV transgenic miceHBV-specific peptide-pulsed DCsReductions in the serum HBsAg and HBV DNA[47]
Cytokines and adjuvants
HBV transgenic miceRecombinant IL-12Marked inhibition of HBV replication in the liver[48]
HBV transgenic miceα-galactosylceramide that can activate NK T cellsComplete inhibition of HBV replication[49]
HBV transgenic miceRecombinant IL-18Inhibition of HBV replication noncytopathically, mediated by activation of resident intrahepatic NK cells and NK T cells[50]
Gene therapy
HBsAg transgenic miceLentivectors expressing HBsAg and IgFc fusion AgInduction of seroconversion to anti-HBs[51]
Table 2 Immunotherapeutic trials for chronic hepatitis B virus infection in humans.
ImmunotherapyResultsRef.
Peptide vaccination
A vaccine with HBc18-27 peptide comprised of a T-helper cell epitope and two palmitic acid residuesLow levels of CTL activity were induced but no significant changes in liver biochemistry or viral serology were observed[52]
Protein vaccination
PreS2/S (GenHevac B) or S (Recombivax)HBe/anti-HBe seroconversion in 13% and HBV DNA negativity in 16% of the treated patients[53]
Intradermal HBsAg vaccine and laimvudine in combination with IL-2Induction of significant HBV DNA loss in the serum in two of five the treated patients[54]
Oral administration of HBV envelope proteins (HBsAg + preS1 + preS2)Induction of histological improvement in 30%, HBeAg negativity in 26.3% and HBsAg-specific T cell proliferation in 78% of the treated patients[55]
IFN-α-2b monotherapy (9 mo) or IFN-α-2b plus pre-S2/S vaccineGreater reduction in HBV DNA in patients with combination HBV therapy than those who received IFN-α-2b monotherapy[56]
The combination with lamivudune and HBsAg vaccine in HBeAg+ casesNo improvement of HBe seroconversion rate in comparison with lamivudine therapy alone[57]
Combination of lamivudine and HBsAg vaccineInduction of sustained negativity of HBV DNA in 1/4 of patients[58]
Combination of lamivudine and HBsAg vaccineHBV DNA became undetectable in 64% of the patients, and was decreased in the remaining patients[59]
DNA immunization
DNA vaccine encoding HBV envelope proteinInduction of an increase in HBV-specific IFN-γ-secreting T cells in nonresponders to conventional therapies, and HBV DNA levels were transiently decreased in 50% of vaccinated patients[60]
DNA vaccine encoding PreS and S in patients with lamivudine breakthroughDevelopment of IFN-γ-producing T cells specific for preS or S antigen; Two of 10 patients showed seroconversion to anti-HBe[61]
DC immunization
Peripheral blood-derived DCs, activated with GM-CSF and IL-4 pulsed with HBsAgBoth patients with normal and elevated ALT responded equally to DC vaccine and 53% of the patients showed induction of HBeAg negativity[62]
Activated DCs from PBL with GM-CSF and IL-4, pulsed with two peptides, HBc18-27 and PreS2 44-53Undetectable HBV DNA was achieved in 46.3% and 3.1% of HBeAg- and HBeAg+ patients, respectively. ALT normalization was observed in 69% and 30.5% of HBeAg- and HBeAg+ patients, respectively[63]
Cytokines
GM-CSFSafe and tolerable up to 1.0 μg/kg body weight, and induced HBV DNA negativity in 4/8 patients[64]
Combination therapy with GM-CSF and HBsAg vaccine in HBV carrier childrenSignificant reduction of serum HBV DNA[65]
High dose of IL-12 (0.5 μg/kg)HBV DNA clearance was observed in 25% of the patients[66]
Combination of IL-12 and lamivudineStimulation of T cell response to HBV with IFN-γ production. However, IL-12 was unable to suppress re-elevation of HBV DNA after cessation of lamivudine[67]
Combination of IL-12 and IL-18Stimulation of IFN-γ production by CD4+ T cells isolated from peripheral blood in response to HBcAg, and the effect was greater than those observed with either cytokine alone[68]
α-galactosylceramidePoorly tolerated and showed no clear suppressive effect on serum HBV DNA or ALT levels[69]
Tα1
Combination of Tα1 and IFN-αNo statistically significant differences as compared with IFN-α monotherapy with respect to HBeAg seroconversion, changes in histology, normalization of ALT or loss of HBV DNA[70]
Tα1 aloneAt 12 mo after cessation of therapy, 36.4% of patients treated with 1.6 mg of Tα1 achieved ALT normalization, 15% achieved HBV DNA clearance by transcription-mediated amplification, and 22.8% achieved clearance of HBeAg[71]
Comparative effect of Tα1 and IFN-αTα1 treatment was more effective in achieving ALT normalization and HBV DNA negativity at the end of the follow-up period than IFN-α[72]
Combination of Tα1 and lamivudineNo any additional antiviral effect compared with lamivudine monotherapy as determined by HBe seroconversion and the emergence of viral breakthrough[73]
Combination therapy with lamivudine and Tα1Induction of significantly higher rates of ALT normalization, virological response, and HBeAg seroconversion than lamivudine monotherapy[74]
IMMUNOTHERAPEUTIC APPROACHES FOR HBV INFECTION

Immunotherapeutic strategies for CHB include suppression of viral replication, stimulation of T cell immune response to hepatitis virus, activation of nonspecific cells, and administration of cytokines with antiviral activity (Tables 1 and 2).

Suppression of viral replication

High viral load has been shown to suppress CD4+ and CD8+ T cells in addition to induction of Treg cells, which could be reversed by antiviral therapy in CHB[75]. Therefore, immunotherapy followed by restoration of virus-specific T cell response with antiviral therapy could be more efficient in CHB.

Stimulation of immune response to HBV

Peptide immunization: A peptide vaccine containing highly immunogenic HBc18-27 has been developed and administered to CHB patients[52], but the results were disappointing because there was no induction of a significant antiviral T cell response.

Protein immunization: In a model of HBV in transgenic mice, vaccine on the base of surface antigen in complete Freund’s adjuvant once monthly for 1 year induced reduction in HBV DNA, and the disappearance of HBeAg and HBsAg in most mice treated[41]. Moreover, it is important to note that some mice developed anti-HBs in the sera. However, several human trials with HBsAg vaccine showed limited efficacy if used as monotherapy.

Recently, hepatitis B vaccine containing not only S protein but also preS has been used with increased immunogenicity[53,55], or has been combined with lamivudine or IFN-α[56], leading to potential improvement of clinical efficacy. However, analysis of the T cell epitope hierarchy has indicated that the most important epitope for viral control is HBc18-27, and not the HBsAg epitope in HLA-A2 patients[9], suggesting the necessity to reconsider antigen selection for vaccination that could lead to better viral control.

DNA immunization: Injection of plasmid DNA has been shown to elicit strongly both cellular and humoral immune responses, and is now known to be safe and well-tolerated both in mice and humans. In a model of duck HBV infection, DNA vaccine encoding HBV large envelope and/or core protein was shown to induce reduction in not only viremia but also covalently closed circular DNA (cccDNA) in the liver in one thirds of ducks receiving DNA monotherapy or combination treatment with lamivudine[44]. This finding is encouraging because clearance of cccDNA from the liver is the goal of treatment for HBV infection, but is difficult to achieve using IFN-α or nucleos(t)ide analogs. Clinical trials have also been performed in HBV infection with some encouraging results, which remain to be confirmed by future randomized large-scale trials.

DC immunization: DCs are specialized antigen-presenting cells that can induce strong immune responses in T and B cells. We have previously shown that activated bone-marrow-derived DCs can break CTL tolerance to HBsAg in HBV transgenic mice[45]. Thereafter, several immunotherapies with activated DCs have been applied in both animals and humans. In a recent study performed in HBV transgenic mice, peptide-pulsed DCs were shown to reduce significantly the concentrations of serum HBsAg and HBV DNA[47], indicating therapeutic potential in chronic HBV infection. Recently, DCs treated with peptide inhibitors of IL-10 have been shown to induce strong anti-HCV T cell responses in HCV transgenic mice[76], suggesting a strategy to augment the immunogenic function of DCs. Moreover, when intrahepatic antigen-presenting cells, including DCs, are activated by injection of an anti-CD40 agonistic antibody, HBV replication is inhibited by a noncytopathic mechanism, possibly through production of antiviral cytokines such as TNF-α and IL-12[46]. Although no CTL response against HBV antigens was reported in this study, the in vivo activation of DCs could be an alternative way for inducing antiviral immune responses, including possible activation of CTLs against HBV. In humans, injection of activated DCs loaded with HBV peptide or protein has achieved a reduction in HBV DNA level in some patients[62,63]. HBeAg negativity was achieved in more than half of the treated patients in one study[62]. Although preparation of activated and mature DCs incurs financial costs and requires experienced researchers, immunotherapy with DCs is a promising method.

Natural killer T cells: A single injection of α-galacto-sylceramide abolished HBV replication by activating natural killer (NK) T cells in the liver in HBV transgenic mice[49]. However, α-galactosylceramide was poorly tolerated in humans and showed no clear antiviral effect[69], possibly due to smaller numbers of NKT cells in the human liver than in the mouse liver.

Cytokines and thymosin-1: Cytokines such as IL-12[48] and IL-18[50] have been shown to inhibit HBV replication noncytopathically in HBV transgenic mice. In humans, granulocyte–macrophage colony-stimulating factor[64,65] and IL-12[66,67] have been used for treatment with some antiviral effects. They have been used as monotherapy or in combination with hepatitis B vaccine or lamivudine.

Thymosin (T)α1, a synthetic 28-amino acid peptide, is able to enhance the Thl immune response and also exerts a direct antiviral mechanism of action. It has been used for the treatment of chronic HBV infection in humans[70-73], and has shown some antiviral efficacy. Although antiviral effect by the addition of Tα1 to lamivudine or IFN-α therapy was controversial, a meta-analysis has demonstrated that combination therapy with lamivudine and Tα1 shows significantly higher rates of ALT normalization, virological response, and HBeAg seroconversion as compared with lamivudine monotherapy[74]. It is of note that HBeAg seroconversion rate was 45% in the combination group, which was significantly higher than that with lamivudine monotherapy (15%).

Blockade of immunoinhibitory signals

Recently, there have been several basic attempts to improve the efficacy of immunotherapy. Among these reports, augmentation or restoration of T cell response by blocking the inhibitory signals has been extensively analyzed in vitro. It has been demonstrated that exhausted T cells express not only PD-1, but also CTLA-4[77], CD244[78] or Tim-3[33], and blocking of these molecules in combination could be better than blocking any single molecule to achieve full activation of the exhausted T cells.

CONCLUSION

There have been several attempts to apply immunotherapy for the control of chronic HBV infection, and some of the data are promising. Viral suppression, stimulation of antiviral immune response with cytokines or immunization with peptide, protein, DNA or DCs, and suppression of the immunoinhibitory signals must be combined to achieve desirable antiviral effects, although further studies are required to explore the best protocols and their most efficient combinations.

Footnotes

Peer reviewer: Dr. Huiping Zhou, Department of Microbiology and Immunology, Virginia Commonwealth University, 1217 East Marshall Street MB#533, Richmond, VA 23298, United States

S- Editor Gou SX L- Editor Kerr C E- Editor Li JY

References
1.  Rehermann B. Immunopathogenesis of viral hepatitis. Baillieres Clin Gastroenterol. 1996;10:483-500.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity. 1996;4:25-36.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Chisari FV. Cytotoxic T cells and viral hepatitis. J Clin Invest. 1997;99:1472-1477.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Guidotti LG, Chisari FV. Noncytolytic control of viral infections by the innate and adaptive immune response. Annu Rev Immunol. 2001;19:65-91.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Webster GJ, Reignat S, Maini MK, Whalley SA, Ogg GS, King A, Brown D, Amlot PL, Williams R, Vergani D. Incubation phase of acute hepatitis B in man: dynamic of cellular immune mechanisms. Hepatology. 2000;32:1117-1124.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Webster G, Bertoletti A. Quantity and quality of virus-specific CD8 cell response: relevance to the design of a therapeutic vaccine for chronic HBV infection. Mol Immunol. 2001;38:467-473.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Penna A, Artini M, Cavalli A, Levrero M, Bertoletti A, Pilli M, Chisari FV, Rehermann B, Del Prete G, Fiaccadori F. Long-lasting memory T cell responses following self-limited acute hepatitis B. J Clin Invest. 1996;98:1185-1194.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Ferrari C, Penna A, Bertoletti A, Valli A, Antoni AD, Giuberti T, Cavalli A, Petit MA, Fiaccadori F. Cellular immune response to hepatitis B virus-encoded antigens in acute and chronic hepatitis B virus infection. J Immunol. 1990;145:3442-3449.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Bertoletti A, D'Elios MM, Boni C, De Carli M, Zignego AL, Durazzo M, Missale G, Penna A, Fiaccadori F, Del Prete G. Different cytokine profiles of intraphepatic T cells in chronic hepatitis B and hepatitis C virus infections. Gastroenterology. 1997;112:193-199.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Zhang JY, Zhang Z, Lin F, Zou ZS, Xu RN, Jin L, Fu JL, Shi F, Shi M, Wang HF. Interleukin-17-producing CD4(+) T cells increase with severity of liver damage in patients with chronic hepatitis B. Hepatology. 2010;51:81-91.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Tang TJ, Kwekkeboom J, Laman JD, Niesters HG, Zondervan PE, de Man RA, Schalm SW, Janssen HL. The role of intrahepatic immune effector cells in inflammatory liver injury and viral control during chronic hepatitis B infection. J Viral Hepat. 2003;10:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Shimada N, Yamamoto K, Kuroda MJ, Terada R, Hakoda T, Shimomura H, Hata H, Nakayama E, Shiratori Y. HBcAg-specific CD8 T cells play an important role in virus suppression, and acute flare-up is associated with the expansion of activated memory T cells. J Clin Immunol. 2003;23:223-232.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Maini MK, Boni C, Lee CK, Larrubia JR, Reignat S, Ogg GS, King AS, Herberg J, Gilson R, Alisa A. The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J Exp Med. 2000;191:1269-1280.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Miyara M, Sakaguchi S. Human FoxP3(+)CD4(+) regulatory T cells: their knowns and unknowns. Immunol Cell Biol. 2011;89:346-351.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Xu D, Fu J, Jin L, Zhang H, Zhou C, Zou Z, Zhao JM, Zhang B, Shi M, Ding X. Circulating and liver resident CD4+CD25+ regulatory T cells actively influence the antiviral immune response and disease progression in patients with hepatitis B. J Immunol. 2006;177:739-747.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Stoop JN, Claassen MA, Woltman AM, Binda RS, Kuipers EJ, Janssen HL, van der Molen RG, Boonstra A. Intrahepatic regulatory T cells are phenotypically distinct from their peripheral counterparts in chronic HBV patients. Clin Immunol. 2008;129:419-427.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Zhao L, Qiu de K, Ma X. Th17 cells: the emerging reciprocal partner of regulatory T cells in the liver. J Dig Dis. 2010;11:126-133.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Hu Y, Shen F, Crellin NK, Ouyang W. The IL-17 pathway as a major therapeutic target in autoimmune diseases. Ann N Y Acad Sci. 2011;1217:60-76.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Kitani A, Xu L. Regulatory T cells and the induction of IL-17. Mucosal Immunol. 2008;1 Suppl 1:S43-S46.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010;236:219-242.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Fife BT, Pauken KE. The role of the PD-1 pathway in autoimmunity and peripheral tolerance. Ann N Y Acad Sci. 2011;1217:45-59.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Chen J, Wang XM, Wu XJ, Wang Y, Zhao H, Shen B, Wang GQ. Intrahepatic levels of PD-1/PD-L correlate with liver inflammation in chronic hepatitis B. Inflamm Res. 2011;60:47-53.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Peng G, Luo B, Li J, Zhao D, Wu W, Chen F, Chen Z. Hepatitis B e-antigen persistency is associated with the properties of HBV-specific CD8 T cells in CHB patients. J Clin Immunol. 2011;31:195-204.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Nan XP, Zhang Y, Yu HT, Li Y, Sun RL, Wang JP, Bai XF. Circulating CD4+CD25high regulatory T cells and expression of PD-1 and BTLA on CD4+ T cells in patients with chronic hepatitis B virus infection. Viral Immunol. 2010;23:63-70.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Fisicaro P, Valdatta C, Massari M, Loggi E, Biasini E, Sacchelli L, Cavallo MC, Silini EM, Andreone P, Missale G. Antiviral intrahepatic T-cell responses can be restored by blocking programmed death-1 pathway in chronic hepatitis B. Gastroenterology. 2010;138:682-693, 693.e1-4.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Sabat R, Grütz G, Warszawska K, Kirsch S, Witte E, Wolk K, Geginat J. Biology of interleukin-10. Cytokine Growth Factor Rev. 2010;21:331-344.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM, Crotty S, von Herrath MG. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J Exp Med. 2006;203:2461-2472.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Li J, Wu W, Peng G, Chen F, Bai M, Zheng M, Chen Z. HBcAg induces interleukin-10 production, inhibiting HBcAg-specific Th17 responses in chronic hepatitis B patients. Immunol Cell Biol. 2010;88:834-841.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, Castelblanco N, Kuchroo V, Gretch DR, Rosen HR. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol. 2009;83:9122-9130.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Ju Y, Hou N, Zhang XN, Zhao D, Liu Y, Wang JJ, Luan F, Shi W, Zhu FL, Sun WS. Blockade of Tim-3 pathway ameliorates interferon-gamma production from hepatic CD8+ T cells in a mouse model of hepatitis B virus infection. Cell Mol Immunol. 2009;6:35-43.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Op den Brouw ML, Binda RS, van Roosmalen MH, Protzer U, Janssen HL, van der Molen RG, Woltman AM. Hepatitis B virus surface antigen impairs myeloid dendritic cell function: a possible immune escape mechanism of hepatitis B virus. Immunology. 2009;126:280-289.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Beckebaum S, Cicinnati VR, Zhang X, Ferencik S, Frilling A, Grosse-Wilde H, Broelsch CE, Gerken G. Hepatitis B virus-induced defect of monocyte-derived dendritic cells leads to impaired T helper type 1 response in vitro: mechanisms for viral immune escape. Immunology. 2003;109:487-495.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Duan XZ, Wang M, Li HW, Zhuang H, Xu D, Wang FS. Decreased frequency and function of circulating plasmocytoid dendritic cells (pDC) in hepatitis B virus infected humans. J Clin Immunol. 2004;24:637-646.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Woltman AM, Ter Borg MJ, Binda RS, Sprengers D, von Blomberg BM, Scheper RJ, Hayashi K, Nishi N, Boonstra A, van der Molen R. Alpha-galactosylceramide in chronic hepatitis B infection: results from a randomized placebo-controlled Phase I/II trial. Antivir Ther. 2009;14:809-818.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Wang S, Han Q, Zhang N, Chen J, Liu Z, Zhang G, Li Z. HBcAg18-27 epitope fused to HIV-Tat 49-57 adjuvanted with CpG ODN induces immunotherapeutic effects in transgenic mice. Immunol Lett. 2010;127:143-149.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Akbar SM, Kajino K, Tanimoto K, Kurose K, Masumoto T, Michitaka K, Horiike N, Onji M. Placebo-controlled trial of vaccination with hepatitis B virus surface antigen in hepatitis B virus transgenic mice. J Hepatol. 1997;26:131-137.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Menne S, Tennant BC, Gerin JL, Cote PJ. Chemoimmunotherapy of chronic hepatitis B virus infection in the woodchuck model overcomes immunologic tolerance and restores T-cell responses to pre-S and S regions of the viral envelope protein. J Virol. 2007;81:10614-10624.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Miller DS, Boyle D, Feng F, Reaiche GY, Kotlarski I, Colonno R, Jilbert AR. Antiviral therapy with entecavir combined with post-exposure "prime-boost" vaccination eliminates duck hepatitis B virus-infected hepatocytes and prevents the development of persistent infection. Virology. 2008;373:329-341.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Thermet A, Buronfosse T, Werle-Lapostolle B, Chevallier M, Pradat P, Trepo C, Zoulim F, Cova L. DNA vaccination in combination or not with lamivudine treatment breaks humoral immune tolerance and enhances cccDNA clearance in the duck model of chronic hepatitis B virus infection. J Gen Virol. 2008;89:1192-1201.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Shimizu Y, Guidotti LG, Fowler P, Chisari FV. Dendritic cell immunization breaks cytotoxic T lymphocyte tolerance in hepatitis B virus transgenic mice. J Immunol. 1998;161:4520-4529.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Kimura K, Kakimi K, Wieland S, Guidotti LG, Chisari FV. Activated intrahepatic antigen-presenting cells inhibit hepatitis B virus replication in the liver of transgenic mice. J Immunol. 2002;169:5188-5195.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Jiang WZ, Fan Y, Liu X, Zhang YL, Wen JJ, Hao WL, Qian M. Therapeutic potential of dendritic cell-based immunization against HBV in transgenic mice. Antiviral Res. 2008;77:50-55.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Cavanaugh VJ, Guidotti LG, Chisari FV. Interleukin-12 inhibits hepatitis B virus replication in transgenic mice. J Virol. 1997;71:3236-3243.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Kimura K, Kakimi K, Wieland S, Guidotti LG, Chisari FV. Interleukin-18 inhibits hepatitis B virus replication in the livers of transgenic mice. J Virol. 2002;76:10702-10707.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Hong Y, Peng Y, Mi M, Xiao H, Munn DH, Wang GQ, He Y. Lentivector expressing HBsAg and immunoglobulin Fc fusion antigen induces potent immune responses and results in seroconversion in HBsAg transgenic mice. Vaccine. 2011;29:3909-3916.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Heathcote J, McHutchison J, Lee S, Tong M, Benner K, Minuk G, Wright T, Fikes J, Livingston B, Sette A. A pilot study of the CY-1899 T-cell vaccine in subjects chronically infected with hepatitis B virus. The CY1899 T Cell Vaccine Study Group. Hepatology. 1999;30:531-536.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Pol S, Nalpas B, Driss F, Michel ML, Tiollais P, Denis J, Brécho C. Efficacy and limitations of a specific immunotherapy in chronic hepatitis B. J Hepatol. 2001;34:917-921.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Dahmen A, Herzog-Hauff S, Böcher WO, Galle PR, Löhr HF. Clinical and immunological efficacy of intradermal vaccine plus lamivudine with or without interleukin-2 in patients with chronic hepatitis B. J Med Virol. 2002;66:452-460.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Safadi R, Israeli E, Papo O, Shibolet O, Melhem A, Bloch A, Rowe M, Alper R, Klein A, Hemed N. Treatment of chronic hepatitis B virus infection via oral immune regulation toward hepatitis B virus proteins. Am J Gastroenterol. 2003;98:2505-2515.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Helvaci M, Kizilgunesler A, Kasirga E, Ozbal E, Kuzu M, Sozen G. Efficacy of hepatitis B vaccination and interferon-alpha-2b combination therapy versus interferon-alpha-2b monotherapy in children with chronic hepatitis B. J Gastroenterol Hepatol. 2004;19:785-791.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Vandepapelière P, Lau GK, Leroux-Roels G, Horsmans Y, Gane E, Tawandee T, Merican MI, Win KM, Trepo C, Cooksley G. 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.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Senturk H, Tabak F, Ozaras R, Erdem L, Canbakan B, Mert A, Yurdakul I. Efficacy of pre-S-containing HBV vaccine combined with lamivudine in the treatment of chronic HBV infection. Dig Dis Sci. 2009;54:2026-2030.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Al-Mahtab M, Rahman S, Akbar SM, Khan SI, Uddin H, Karim F, Ahmed F. Combination therapy with antiviral drugs and hepatitis B vaccine in incidentally-detected and asymptomatic chronic hepatitis virus B carriers at Bangladesh. Viral Immunol. 2010;23:335-338.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Mancini-Bourgine M, Fontaine H, Scott-Algara D, Pol S, Bréchot C, Michel ML. Induction or expansion of T-cell responses by a hepatitis B DNA vaccine administered to chronic HBV carriers. Hepatology. 2004;40:874-882.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Mancini-Bourgine M, Fontaine H, Bréchot C, Pol S, Michel ML. Immunogenicity of a hepatitis B DNA vaccine administered to chronic HBV carriers. Vaccine. 2006;24:4482-4489.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Chen M, Li YG, Zhang DZ, Wang ZY, Zeng WQ, Shi XF, Guo Y, Guo SH, Ren H. Therapeutic effect of autologous dendritic cell vaccine on patients with chronic hepatitis B: a clinical study. World J Gastroenterol. 2005;11:1806-1808.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Luo J, Li J, Chen RL, Nie L, Huang J, Liu ZW, Luo L, Yan XJ. Autologus dendritic cell vaccine for chronic hepatitis B carriers: a pilot, open label, clinical trial in human volunteers. Vaccine. 2010;28:2497-2504.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Martin J, Bosch O, Moraleda G, Bartolome J, Quiroga JA, Carreño V. Pilot study of recombinant human granulocyte-macrophage colony-stimulating factor in the treatment of chronic hepatitis B. Hepatology. 1993;18:775-780.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Wang J, Zhu Q, Zhang T, Yu H. A pilot study on the combined therapy of granulocyte-macrophage colony-stimulating factor and hepatitis B vaccine on chronic hepatitis B virus carrier children. Chin Med J (Engl). 2002;115:1824-1828.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Zeuzem S, Carreño V. Interleukin-12 in the treatment of chronic hepatitis B and C. Antiviral Res. 2001;52:181-188.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Rigopoulou EI, Suri D, Chokshi S, Mullerova I, Rice S, Tedder RS, Williams R, Naoumov NV. Lamivudine plus interleukin-12 combination therapy in chronic hepatitis B: antiviral and immunological activity. Hepatology. 2005;42:1028-1036.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Szkaradkiewicz A, Jopek A, Wysocki J. Effects of IL-12 and IL-18 on HBcAg-specific cytokine production by CD4 T lymphocytes of children with chronic hepatitis B infection. Antiviral Res. 2005;66:23-27.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Woltman AM, Ter Borg MJ, Binda RS, Sprengers D, von Blomberg BM, Scheper RJ, Hayashi K, Nishi N, Boonstra A, van der Molen R. Alpha-galactosylceramide in chronic hepatitis B infection: results from a randomized placebo-controlled Phase I/II trial. Antivir Ther. 2009;14:809-818.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Arase Y, Tsubota A, Suzuki Y, Suzuki F, Kobayashi M, Someya T, Akuta N, Hosaka T, Saitoh S, Ikeda K. A pilot study of thymosin alpha1 therapy for chronic hepatitis B patients. Intern Med. 2003;42:941-946.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Iino S, Toyota J, Kumada H, Kiyosawa K, Kakumu S, Sata M, Suzuki H, Martins EB. The efficacy and safety of thymosin alpha-1 in Japanese patients with chronic hepatitis B; results from a randomized clinical trial. J Viral Hepat. 2005;12:300-306.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  You J, Zhuang L, Cheng HY, Yan SM, Yu L, Huang JH, Tang BZ, Huang ML, Ma YL, Chongsuvivatwong V. Efficacy of thymosin alpha-1 and interferon alpha in treatment of chronic viral hepatitis B: a randomized controlled study. World J Gastroenterol. 2006;12:6715-6721.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Lee HW, Lee JI, Um SH, Ahn SH, Chang HY, Park YK, Hong SP, Moon YM, Han KH. Combination therapy of thymosin alpha-1 and lamivudine for HBeAg positive chronic hepatitis B: A prospective randomized, comparative pilot study. J Gastroenterol Hepatol. 2008;23:729-735.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Zhang YY, Chen EQ, Yang J, Duan YR, Tang H. Treatment with lamivudine versus lamivudine and thymosin alpha-1 for e antigen-positive chronic hepatitis B patients: a meta-analysis. Virol J. 2009;6:63.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Boni C, Penna A, Ogg GS, Bertoletti A, Pilli M, Cavallo C, Cavalli A, Urbani S, Boehme R, Panebianco R. Lamivudine treatment can overcome cytotoxic T-cell hyporesponsiveness in chronic hepatitis B: new perspectives for immune therapy. Hepatology. 2001;33:963-971.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Díaz-Valdés N, Manterola L, Belsúe V, Riezu-Boj JI, Larrea E, Echeverria I, Llópiz D, López-Sagaseta J, Lerat H, Pawlotsky JM. Improved dendritic cell-based immunization against hepatitis C virus using peptide inhibitors of interleukin 10. Hepatology. 2011;53:23-31.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Nakamoto N, Cho H, Shaked A, Olthoff K, Valiga ME, Kaminski M, Gostick E, Price DA, Freeman GJ, Wherry EJ. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathog. 2009;5:e1000313.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Raziorrouh B, Schraut W, Gerlach T, Nowack D, Grüner NH, Ulsenheimer A, Zachoval R, Wächtler M, Spannagl M, Haas J. The immunoregulatory role of CD244 in chronic hepatitis B infection and its inhibitory potential on virus-specific CD8+ T-cell function. Hepatology. 2010;52:1934-1947.  [PubMed]  [DOI]  [Cited in This Article: ]