Elsevier

Human Immunology

Volume 72, Issue 9, September 2011, Pages 687-698
Human Immunology

Hepatitis B virus (HBV) core antigen-specific regulatory T cells confer sustained remission to anti-HBV therapy in chronic hepatitis B with acute exacerbation

https://doi.org/10.1016/j.humimm.2010.11.001Get rights and content

Abstract

Acute exacerbations (AEs) of chronic hepatitis B (CH-B) are thought to be the result of breakdown of immune tolerance on the natural history of chronic hepatitis B virus (HBV) infection. Immune tolerance to HBV maintained in CH-B patients without hepatitis is under the control of the host's forkhead box p3-expressing regulatory T cells (Tregs). Its breakdown mimics the occurrence of autoimmune diseases. Severe AEs may lead to liver decompensation and mortalities. Consequently, AEs are currently the major therapeutic targets in patient treatment. In this study, we employed the SYFPEITHI scoring system to identify epitopes on HBV core antigen (HBcAg) for the construction of human leukocyte antigen class II tetramers to measure HBcAg-specific Treg frequencies (Tregf). Upregulation of Treg gene profiling accompanied by increased HBcAg-specific Tregf was detected in AE patients with sustained remission (SR) to anti-HBV therapy. Depletion of Tregs from peripheral blood mononuclear cells enhanced proliferation to HBcAg. HBcAg-specific Treg clones inhibited the killing capacity of cytotoxic T lymphocyte clones in an antigen-independent manner. A greater posttherapy increase in HBcAg-specific Tregf correlated with a higher SR rate to anti-HBV therapy. These results suggest that HBcAg-specific Tregs function as suppressor effectors and confer SR to anti-HBV therapy.

Introduction

Hepatitis B virus (HBV) infects more than 350 million people worldwide. All infected subjects are at risk of developing adverse sequelae, such as acute hepatitis and acute fulminant hepatitis, which may lead to liver failure and mortality. Some HBV infections may evolve into chronic persistent infection with a protracted lifelong clinical course that may flare up or develop acute exacerbations (AEs) spontaneously. Severe AE can be associated with massive or submassive hepatic necrosis analogous to acute fulminant hepatitis. Long-term chronic hepatitis B (CH-B) may progress to liver cirrhosis and hepatocellular carcinoma [1], [2], [3], [4], [5], [6]. Since pilot clinical trials of interferon-α (IFN-α) therapy for CH-B patients were reported in 1976 [7], [8], attempts to treat CH-B have exhausted tremendous medical expenditure worldwide. Disappointingly, CH-B patients have so far responded ineffectively, with a low sustained remission (SR) rate to all types of currently available anti-HBV treatment modalities [9], [10], [11], [12], [13]. The need for immediate anti-HBV therapy with nucleoside analogues for patients developing life-threatening AEs, specifically cancer patients receiving chemotherapy and organ transplant recipients treated with immunosuppressants, has reached a consensus [10], [11], [12]. This strategy has achieved great success in rescuing a large proportion of patients from hepatic failure and mortality [10], [11], [12], [13], [14], [15]. Furthermore, this approach seems to prolong survival among decompensated cirrhotic patients [16]. However, all current oral anti-HBV nucleoside analogues can suppress virus replication via the inhibition of HBV DNA polymerase, but are unable to totally remove the covalently closed circular DNA of the HBV genome to achieve complete viral clearance [5], [10], [11], [12], [13], [17], [18]. Consequently, virus breakthroughs, biochemical relapses, and/or HBV e antigen (HBeAg) reversion in serum are frequently encountered when the use of these drugs is stopped. Moreover, long-term use of nucleoside analogues may select for drug-resistant mutant viruses, such as tyrosine-isoleucine-aspartate-aspartate and/or tyrosine-valine-aspartate-aspartate mutations on the tyrosine–methionine–aspartate–aspartate motif of the HBV DNA polymerase and lead to therapeutic failure [5], [10], [11], [12], [13], [19]. Thus, the mechanism by which chronic hepatitis B patients achieve SR or viral clearance after anti-HBV therapy with nucleoside analogues remains unclear [5], [12], [13]. Once the treatment is initiated, lifelong HBV suppression may be necessary [20].

HBV is not directly cytopathic and the immune response of the host appears to mediate the hepatocellular injury and subsequent viral clearance [2], [3], [5], [13], [21]. Accumulating evidence indicates that the immune response of hepatitis B patients may influence the outcome of anti-HBV therapy [22], [23], [24], [25], [26], [27], [28]. We showed that AEs of CH-B are accompanied by increased T-cell responses to hepatitis B core and e antigens (HBcAg/HBeAg) [29] and that HBcAg and/or HBeAg may represent important “targets” for immune-mediated viral clearance [30], [31].

Evidence shows that naturally arising forkhead/winged helix transcription factor box p3 (Foxp3)-expressing CD4+CD25high regulatory T cells (Tregs), namely activated CD4+ T cells with expression of interleukin-2 (IL-2) receptor α chain [32], play an important role in the regulation of autoimmune diseases [33], but whether Tregs play a role in the control of infectious diseases and persistent viral infections remains undetermined [34], [35], specifically in patients with chronic viral hepatitis B [36], [37], [38] and C [39]. An immune tolerance response to these viruses is considered important for disease progression [3], [5], [13]. We have shown that HBcAg-specific Tregs play a crucial role in modulating spontaneous AEs on the natural history of perinatally acquired chronic HBV infection [40]. Whether HBcAg-specific Tregs play a role in influencing the outcome of anti-HBV therapy remains unclear. Because AEs are critical events in the natural history of chronic HBV infection and are major therapeutic targets in the treatment of CH-B [1], [2], [3], [5], [10], [11], [12], [13], [14], [15], in the present study we aim to investigate the role of HBcAg-specific Tregs during anti-HBV therapy for CH-B patients with AE.

Section snippets

Patients

In the second quarter of 2002 when oral nucleoside analogues and IFN-α injection for the treatment of CH-B patients were included in the health insurance coverage of Taiwan, a nationwide therapeutic trial of anti-HBV therapy for CH-B patients was initiated. This trial included the following regimens: regimen I, 18-month oral lamivudine (3-TC) therapy (100 mg/day) for CH-B patients with AE; regimen II, 18-month oral entecavir (ETV) therapy (0.5 mg/day) for CH-B patients with AE; regimen III,

Overlapping of markers between Tregs and Th17 cells

CD4+ CD25+ contained Foxp3+ CD4+ CD25high T cells as verified by flow cytometry analysis (Supplemental Fig. 1.) Not all CD4+CD25high T cells were 100% positive for Foxp3+ staining using a PE-antihuman FOXP3 flow kit (Biolegend), suggesting that they comprised a mixed population of CD4+ T cells.

Upregulation of Foxp3+ Treg gene profiling on PBMCs is accompanied by increased HBcAg-specific Tregf during and/or after anti-HBV therapy

Treg gene profiling of 1 CH-B patient carrying HLA-A2 with AE achieving SR to ETV therapy is shown in Fig. 2. Before therapy, Foxp3+ Treg gene profiling including cAMP response element binding proteins (

Discussion

Recent studies have revealed that the CD4+CD25+ regulatory T (Tr) cells constitute a rather homogenous population, derive from the thymus, and are naturally nonproliferative (i.e., anergic) to stimulation via the TCR but require activation via their T-cell receptor (TCR) to become suppressive and to inhibit the proliferation of CD4+ or CD8+ T cells. However, once activated, their regulatory/suppressor function was completely antigen nonspecific and cytokine independent, yet cell contact

Acknowledgments

This work was supported by grants from the Chi Mei Foundation (CMFHT 9102, 9401, and 9701 and CMFHR9557), Tainan, Taiwan. The authors are grateful to Miss Chin-Li Lu, statistician, Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, for statistical analysis of the data.

References (62)

  • R.N. Chien et al.

    Determinants for sustained HBeAg response to lamivudine therapy

    Hepatology

    (2003)
  • C.T. Weaver et al.

    17: an effector CD4 T cell lineage with regulatory T cell ties

    Immunity

    (2006)
  • S.L. Tsai et al.

    Hepatitis C virus variants circumventing cytotoxic T lymphocyte activity as a mechanism of chronicity

    Gastroenterology

    (1998)
  • Y.H. Ni et al.

    Viremia profiles in children with chronic hepatitis B virus infection and spontaneous e antigen seroconversion

    Gastroenterology

    (2007)
  • D.S. Chen

    From hepatitis to hepatoma: lessons from type B viral hepatitis

    Science

    (1993)
  • W.M. Lee

    Hepatitis B virus infection

    N Engl J Med

    (1997)
  • A.S.F. Lok

    Chronic hepatitis B

    N Engl J Med

    (2002)
  • D.M. Morens et al.

    The challenge of emerging and re-emerging infectious diseases

    Nature

    (2004)
  • D. Ganem et al.

    Hepatitis B virus infection—natural history and clinical consequences

    N Engl J Med

    (2004)
  • H.B. Greenberg et al.

    Effect of human leukocyte interferon on hepatitis B virus infection in patients with chronic active hepatitis

    N Engl J Med

    (1976)
  • J.H. Hoofnagle et al.

    The treatment of chronic viral hepatitis

    N Engl J Med

    (1997)
  • M. Sherman et al.

    Management of chronic hepatitis B: Consensus guidelines

    Can J Gastroenterol

    (2007)
  • J.L. Dienstag

    Hepatitis B virus infection

    N Engl J Med

    (2008)
  • Y.F. Liaw

    Hepatitis flares and hepatitis B e antigen seroconversion: implication in anti-hepatitis B virus therapy

    J Gastroenterol Hepatol

    (2003)
  • M.Y. Mashhour et al.

    Efficacy of lamivudine therapy on decompensated liver cirrhosis due to chronic hepatitis B

    Hep Mon

    (2007)
  • O. Yokosuka et al.

    Hepatitis B virus RNA transcripts and DNA in chronic liver disease

    N Engl J Med

    (1986)
  • W. Chotiyaputta et al.

    Hepatitis B virus variants

    Nat Rev Gastroenterol Hepatol

    (2009)
  • M. Omata

    Treatment of chronic hepatitis B infection

    N Engl J Med

    (1998)
  • C. Boni et al.

    Lamivudine treatment can restore T cell responsiveness in chronic hepatitis B

    J Clin Invest

    (1998)
  • C. Hultgren et al.

    Cell-mediated immune responses and loss of hepatitis B e-antigen (HBeAg) during successful lamivudine and famciclovir combination therapy for chronic replicating hepatitis B virus infection

    Clin Infect Dis

    (1999)
  • S.L. Tsai et al.

    Activation of Th1 immunity is a common immune mechanism for the successful treatment of hepatitis B and C: tetramer assay and therapeutic implications

    J Biomed Sci

    (2003)
  • Cited by (12)

    • INASL Guidelines on Management of Hepatitis B Virus Infection in Patients receiving Chemotherapy, Biologicals, Immunosupressants, or Corticosteroids

      2018, Journal of Clinical and Experimental Hepatology
      Citation Excerpt :

      There is rise in HBcAg/HBeAg-specific T cell proliferation before and during the hepatitis flares; an increased production of interferon gamma, Th1 phenotypic cytokine IL-2, IFN-α and IL-8 during the flare.28,29 The Treg cells decline during the flare, implicating exaggerated T cell mediated immune response30. This leads to immune mediated cytolysis of hepatocytes expressing HBV antigens and its downstream apoptotic mechanisms.

    • Hepatitis B flares in chronic hepatitis B: Pathogenesis, natural course, and management

      2014, Journal of Hepatology
      Citation Excerpt :

      It was demonstrated that an increase in circulating and intrahepatic IL-17-producing CD4+ T cells correlated well with ALT level and liver injury [27]. Longitudinal immunologic studies showed a decline of HBcAg-specific regulatory T cell (Treg) frequencies, associated with an increase in HBcAg-specific cytotoxic T lymphocyte (CTL) frequencies prior to the peak of the hepatitis flare [28–30], IL-10-producing regulatory B cell frequencies and serum IL-10 level peaked with the increase in viral load and decreased at the same time or shortly after the peak of ALT [31], large fluctuations in serum IFN-α and IL-8 concentrations with peak levels coinciding with a sharp increase in viral load preceding the onset of the hepatitis flare, and the increases in serum IFN-α and IL-8 promoted a pathway for the natural killer (NK)-cell mediated liver damage [32]. It was also shown that hepatitis flares were temporarily associated with high serum levels of INF-γ inducible chemokines CXCL-9 and CXCL-10 [33], and that increase, peak and decline in the levels of the programmed cell death protein 1 (PD-1) and its ligand PD-L1 went parallel with the ascend, peak and decline of HBV-specific T cells and serum ALT levels [34].

    • Immunotherapeutic interventions in chronic hepatitis B virus infection: A review

      2014, Journal of Immunological Methods
      Citation Excerpt :

      Immune tolerance to HBV in CHB patients without hepatitis is under the control of the host forkhead box p3-expressing Tregs. The CD8+ T cell activity increased significantly after depletion of circulating Tregs (Koay et al., 2011). Tregs also reduced the response to treatment and non-responders to INF-α had higher level of Tregs.

    View all citing articles on Scopus

    None of the authors has any potential financial conflict of interest related to the manuscript.

    View full text