Accepted Article

1

1

Article type: Review Article

2

Received date: 08/08/2014

3

Revised date: 10/13/2014

4

Accepted date: 10/14/2014

5

For: Hepatology Research - Review

6

7

The Immunobiology of Hepatitis B Virus

8

Infection1

9

Running Title: The Immunobiology of Hepatitis B Virus Infection

10 11

Masanori Isogawa, and Yasuhito Tanaka

12 13

Department of Virology & Liver Unit

14

Nagoya City University Graduate School of Medical Science

15 16 17

Corresponding Author: This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1111/hepr.12439 This article is protected by copyright. All rights reserved.

Accepted Article

2

1

Yasuhito Tanaka, M.D. PhD.

2

Department of Virology & Liver Unit

3

Nagoya City University Graduate School of Medical Science

4

Kawasumi 1, Mizuho-ku,

5

Nagoya 467-8601, Japan

6

Phone: +81-52-853-8191

7

Fax:

8

email: [email protected]

+81-52-842-0021

9

10

This article is protected by copyright. All rights reserved.

Accepted Article

3

1

Abstract.

2

The adaptive immune response, particularly the virus-specific CD8+ T cell response, is

3

largely responsible for viral clearance and disease pathogenesis during hepatitis B virus

4

(HBV) infection. The HBV-specific CD8+ T cell response is vigorous, polyclonal and

5

multispecific in acutely infected patients who successfully clear the virus and relatively

6

weak and narrowly focused in chronically infected patients. The immunological basis for

7

this dichotomy is unclear. A recent study using HBV transgenic mice and HBV-specific

8

T cell receptor (TCR) transgenic mice suggests that intrahepatic antigen presentation by

9

HBV positive hepatocytes suppresses HBV-specific CD8+ T cell responses through a

10

coinhibitory molecule, programmed cell death 1 (PD-1). In contrast, antigen presentation

11

by activated professional antigen presenting cells (pAPCs) induces functional

12

differentiation of HBV-specific CD8+ T cells. These findings suggest that the outcome of

13

T cell priming is largely dependent on the nature of antigen presenting cells. Another

14

study suggests that the timing of HBV-specific CD4+ T cell priming regulates the

15

magnitude of the HBV-specific CD8+ T cell response. Other factors that could regulate

16

HBV-specific cellular immune responses are high viral loads, mutational epitope

17

inactivation, T cell receptor antagonism and infection of immunologically privileged

18

tissues. However, these pathways become apparent only in the setting of an ineffective

19

cellular immune response, which is therefore the fundamental underlying cause.

20

Understanding the cellular and molecular mechanisms by which HBV evades host

21

immune responses will eventually help develop new immunotherapeutic strategies

22

designed to terminate chronic HBV infection.

This article is protected by copyright. All rights reserved.

Accepted Article

4

1

Key words: Hepatitis B virus, Immune response, Immunological priming,

2

Immunotherapy, T cells

3

Introduction

4

Hepatitis B virus (HBV) is a partially double stranded DNA virus that causes

5

necroinflammatory liver disease of variable severity.1 More than 240 million people

6

worldwide are persistently infected with HBV. Persistent infection by HBV is often

7

associated with chronic liver disease that can lead to the development of cirrhosis and

8

hepatocellular carcinoma (HCC). HBV is thought to be largely non-cytopathic for the

9

infected hepatocyte under normal circumstances,2,3 but could be cytopathic in

10

immunodeficient chimeric mice carrying human hepatocytes,4 suggesting that

11

unmitigated HBV replication and gene expression trigger cellular stresses and cell death.

12

The immune response mediates the clearance of HBV and disease pathogenesis.2

13

Interestingly, HBV appears to be a poor inducer of innate immune responses, acting as a

14

stealth virus in this regard by evading recognition by innate immune sensor molecules.5

15

Despite the weak innate immune responses, acute HBV infection of adults is usually self-

16

limited and cleared by functional HBV-specific CD8+ T cell responses. The induction of

17

functional HBV-specific CD8+ T cell responses is dependent on early CD4+ T cell

18

priming prior to HBV spread,6 which might explain why patients infected with human

19

immunodeficiency virus (HIV) become persistently infected following horizontal

20

transmission of HBV.7,8 Once chronic HBV infection is established, HBV-specific CD8+

21

T cells are subjected to powerful immunoregulatory mechanisms in the liver. Ample

22

evidence suggests that HBV actively suppresses adoptive immune responses, particularly

This article is protected by copyright. All rights reserved.

Accepted Article

5

1

HBV-specific CD8+ T cell responses. Reduction of viral antigen expression by antiviral

2

treatment can overcome CD8+ T cell hyporesponsiveness in patients with chronic HBV

3

infection, suggesting that T cells are present in these subjects but suppressed by high viral

4

loads.9 In addition, recent evidence suggests that antigen recognition in the liver

5

suppresses HBV-specific CD8+ T cell effector functions by signaling through a

6

coinhibitory molecule, programmed cell death 1 (PD-1).10,11 Furthermore, several viral

7

proteins have been shown to regulate the adaptive immune response to HBV,12-14 which

8

might explain the varying chronicity rate between genotypes that express different levels

9

of viral antigens.15,16 We herein review our current knowledge of the host immune

10

responses to HBV as well as the cellular and molecular mechanisms by which HBV

11

evades these immune responses. We also discuss how the accumulating knowledge of

12

HBV immunobiology will help us to develop more inventive therapeutic strategies that

13

are designed to reinvigorate HBV-specific immune responses, thereby terminating

14

chronic infection with this deadly virus.

15 16

1. The Innate Immune Response

17

1-1. Absence of an Innate Response by Infected Cells during Acute HBV Infection.

18

Virus infection usually triggers a robust innate immune response that is heralded by rapid

19

induction of IFNα/β by the infected cell.17 Production of IFNα/β induces the

20

transcriptional expression of a large number of interferon inducible genes (ISGs), which

21

in turn exert a variety of intracellular antiviral mechanisms.17,18 Surprisingly, intrahepatic

This article is protected by copyright. All rights reserved.

Accepted Article

6

1

gene expression profiling in acutely HBV infected chimpanzees revealed that HBV acts

2

like a stealth virus early after infection because it does not induce any cellular gene

3

expression, including ISGs, as it spreads through the liver.5 This contrasts strikingly to

4

the induction of many ISGs during the spread of hepatitis C virus (HCV) that is highly

5

visible to the innate immune system.19 The relative invisibility of HBV to the innate

6

sensing machinery of the cells probably reflects its replication strategy. HBV retains its

7

transcriptional template, namely covalently closed circular DNA (cccDNA), in the

8

nucleus20 and the HBV mRNA is capped and polyadenylated, resembling normal cellular

9

transcripts.21,22 In addition, HBV replicates within viral capsid particles in the

10

cytoplasm20 and is therefore shielded from innate sensing machinery. Thus, the typical

11

widespread expansion of HBV in the liver may reflect the absence of IFNα/β production

12

to which the virus is exquisitely sensitive, as shown in HBV transgenic mice.23,24

13

1-2. The Natural Killer and Natural Killer T Cell Responses during HBV Infection

14

The role of natural killer (NK) cells in acute HBV infection remains largely unknown.

15

Studies that analyzed NK cell functions during acute HBV infection have provided

16

conflicting evidence. A recent study demonstrated that the activation and cytokine

17

producing ability of NK cells were impaired in acute HBV patients,25 while others

18

showed enhanced IFN-γ production by, and cytotoxicity of, NK cells.26,27 The cause of

19

this discrepancy is unclear. On the other hand, NK cells in chronic HBV patients seem

20

generally cytolytic but their cytokine producing ability is selectively impaired.28,29 A

21

recent report demonstrated a strong correlation between liver disease, IFN-α production

22

and NK cell expression of tumor necrosis factor related apoptosis-inducing ligand

This article is protected by copyright. All rights reserved.

Accepted Article

7

1

(TRAIL) during chronic HBV infection,30 suggesting a pathogenic role of NK cells

2

(Figure 1, grey area). Interestingly, NK cells seem highly immunoregulatory as well,

3

because they can apparently delete HBV-specific, but not cytomegalovirus (CMV)-

4

specific, CD8+ T cells through TRAIL-TRAIL receptor 2 (TRAIL-R2) interaction.31

5

Taken together, NK cells appear to exert a detrimental effect on the host during chronic

6

HBV infection. The precise role of NK cells during chronic HBV infection must be fully

7

elucidated to develop new NK cell based therapeutic approaches against chronic HBV

8

infections. The role of natural killer T (NKT) cells during natural HBV infection is even

9

less clear. Therapeutic potential of NKT cell activation against HBV infection was 32

10

initially indicated in HBV transgenic mice model

11

and woodchuck hepatitis virus (WHV) suggested that NKT cells were activated soon

12

after infection.33,34 However, the degree of NKT cell activation was relatively modest in

13

both cases, and the physiological importance of such activation has not been addressed in

14

these studies. Studies with several mouse models provided more convincing evidence that

15

CD1d-restricted NKT cells are important for the control of HBV replication and gene

16

expression.35,36 It is well known, however, that hepatic NKT cell repertoires are

17

phenotypically and functionally very different between humans and mice.37 Further

18

studies are required to confirm the role of NKT cell during HBV infections.

19

2. The Adoptive Immune Response

20

2-1. The Antibody Response

21

Antibodies to the HBV envelope antigen (HBsAg) neutralize HBV infection presumably

22

by preventing virus attachment to hepatocytes. They also facilitate viral clearance by This article is protected by copyright. All rights reserved.

, and subsequent studies with HBV

Accepted Article

8

1

complexing with free viral particles and removing them from the circulation. The

2

induction of anti-HBs by immunization is a T cell-dependent process and requires CD4+

3

T cell help.38 While anti-HBs neutralizing antibodies are essential for providing

4

protection after vaccination, their importance in resolving natural HBV infection is

5

somewhat underappreciated, mostly because the appearance of anti-HBs occurs relatively

6

late after HBV exposure39 (Figure 1, blue lines). It is generally believed that anti-HBs

7

neutralizing antibodies prevent viral spread from the rare cells that remain infected, even

8

after resolution of HBV infection. Supporting this notion, HBV is frequently re-activated

9

long after resolution of infection in patients who receive conventional chemotherapy or a

10

newly developed monoclonal antibody (Rituximab) therapy that targets the CD20 antigen

11

expressed on the surface of normal and malignant B lymphocytes.40 More detailed

12

analysis of HBV-specific B cell responses might reveal a heretofore-unappreciated role

13

of humoral responses in the control of HBV infection.

14

2-2. The CD4+ T Cell Response

15

Ample evidence suggests a clear relationship between the CD4+ T cell response to HBV

16

and the outcome of HBV infection. For example, the peripheral blood CD4+ T cell

17

response to HBV is vigorous and multispecific in patients with acute hepatitis who

18

ultimately clear the virus, while it is relatively weak in chronic HBV patients39,41,42

19

(Figure 1, red areas). Despite the association between a strong CD4+ T cell response and

20

viral clearance, CD4+ T cell depletion at the peak of HBV infection had no effect on viral

21

replication and liver disease in infected chimpanzees.43 In contrast, CD4+ T cell depletion

22

before HBV infection resulted in quantitatively and qualitatively reduced HBV-specific

This article is protected by copyright. All rights reserved.

Accepted Article

9

1

CD8+ T cell responses, leading to persistent HBV infection.6 These results suggest that

2

CD4+ T cells probably contribute indirectly to the control of HBV infection by

3

facilitating the induction and maintenance of the virus-specific CD8+ T cell responses. It

4

is likely that CD4+ T cells help induce functional HBV-specific CD8+ T cell responses by

5

producing IL-2 that is essential for T cell proliferation and by activating professional

6

antigen presenting cells (pAPCs) that are capable of providing co-stimulation to T cells.44

7

In contrast, in the absence of early CD4+ T cell responses, CD8+ T cell priming could

8

occur in the liver, resulting in T cell inactivation, tolerance or apoptosis,11,45,46 as

9

summarized later in this review. This requirement of CD4+ T cells for the induction of

10

HBV-specific CD8+ T cells is one of the reasons patients infected with HIV (low CD4

11

count) become persistently infected after horizontal transmission of HBV7,8 and why

12

genetic variants in the HLA-DP locus are associated with the risk of developing chronic

13

HBV infection.47 Interestingly, early CD4+ T cell priming does not occur when the size of

14

inoculum is small (i.e. 1 to 10 copies) and the appearance of HBV DNA is delayed after

15

infection,6 suggesting that slow spread of HBV might facilitate chronicity, as often

16

observed with genotype A infections.15,16 Interestingly, recent studies indicated that the

17

frequency of IL-17 producing CD4 T cells (Th-17 cells) is increased in chronic HBV

18

patients,

19

liver disease.49 These results suggest that CD4 T cells not only help induce antiviral

20

cellular immune responses during acute HBV infections, but also mediate inflammatory

21

responses during chronic HBV infection.

22

2-3. The CD8+ T Cell Response

48

and IL-17 and another Th-17 related cytokine IL-22 are shown to exacerbate

This article is protected by copyright. All rights reserved.

Accepted Article

10

1

The HBV-specific CD8+ T cell response plays a fundamental role in viral clearance and

2

liver disease. A vigorous polyclonal CD8+ T cell response is readily detectable in the

3

peripheral blood of patients with acute hepatitis B who ultimately clear the virus. In

4

contrast, the peripheral blood T cell response in chronically infected patients is weak and

5

narrowly focused2,50 (Figure 1, green areas). The livers of these patients contain virus-

6

specific T cells that likely contribute to disease pathogenesis but, for functional and/or

7

quantitative reasons, are unable to clear the infection. Interestingly, a recent study that

8

examined the relationship between the number of intrahepatic HBV-specific CD8+ T

9

cells, extent of liver disease, and levels of HBV replication in chronically infected

10

patients indicated that inhibition of virus replication could be independent of liver

11

damage and that the functionality of HBV-specific CD8+ T cells was more important than

12

the number of T cells in controlling HBV replication.51 Experiments in chimpanzees have

13

shown that the viral clearance and the onset of liver disease coincide with the

14

accumulation of virus-specific CD8+ T cells and the induction of IFN-γ and IFN-

15

inducible genes in the liver.52 Importantly, depletion of CD8+ T cells at the peak of

16

viremia delays viral clearance and the onset of viral hepatitis until the T cells return,

17

providing the most definitive evidence that viral clearance and liver disease are mediated

18

by virus specific CD8+ T cells.43

19

3. Non-cytolytic and cytolytic T cell effector functions emerge alternately during

20

HBV clearance

21

It is widely believed that virus specific CD8+ T cells clear the viral infection by killing

22

infected cells. However, CD8+ T cell killing is an inefficient process, requiring direct

This article is protected by copyright. All rights reserved.

Accepted Article

11

1

physical contact between the CD8+ T cells and the infected cells. Therefore, it may not be

2

possible for HBV-specific CD8+ T cells to kill all the HBV infected cells if the CD8+ T

3

cells are greatly outnumbered, as occurs during HBV infection in which as many as 1011

4

hepatocytes can be infected.53 Hence, although the liver disease in HBV infection is

5

clearly due to the cytopathic activity of the CD8+ T cells, viral clearance may require

6

more efficient CD8+ T cell functions than killing. Important insights into the pathogenic

7

and noncytopathic antiviral functions of the CD8+ T cell response have come from

8

studies of HBV transgenic mice that develop an acute necroinflammatory liver disease

9

after adoptive transfer of HBV-specific effector or effector memory CD8+ T cells that

10

were generated by immunizing non-transgenic mice.10,54 In that model, the HBV-specific

11

CD8+ T cells rapidly enter the liver and recognize viral antigen and secrete interferon

12

gamma (IFN-γ), which non-cytopathically inhibits HBV replication in the remaining

13

hepatocytes54 by preventing the assembly of HBV RNA-containing capsids in the

14

cytoplasm23,55 in a proteasome-56 and kinase-dependent57 process. During this remarkable

15

process, the viral nucleocapsids disappear from the cytoplasm of the hepatocytes, yet the

16

hepatocytes remain perfectly healthy.54 Antibody blocking and knockout experiments in

17

the HBV transgenic mouse model further demonstrated that the cytopathic and antiviral

18

functions of CD8+ T cells are completely independent of each other.58,59 Following the

19

secretion of IFN-γ and the suppression of HBV replication, HBV-specific CD8+ T cells

20

expand vigorously in situ. Surprisingly, however, the ability of HBV-specific CD8+ T

21

cells to produce IFN-γ is quickly down-regulated as they expand. Despite the suppression

22

of IFN-γ secretion, the cytotoxic capabilities of the HBV-specific CD8+ T cells increase

23

over time, causing necroinflammatory liver disease, and complete inhibition of HBV

This article is protected by copyright. All rights reserved.

Accepted Article

12

1

mRNA expression.10 Taken together, these results suggest that, in the context of a

2

vigorous immune response during acute HBV infection, the sequential and altering

3

patterns of non-cytolytic and cytolytic effector function, coupled with T cell expansion

4

and contraction, cooperate to minimize the extent of tissue injury required for viral

5

clearance. Conversely, we suggest that diminution of the CD8+ T cell response or poor

6

coordination of these responses could lead to viral persistence and varying degrees of

7

chronic liver disease.

8 9

4. Where, how, and what kind of HBV-specific CD8+ T cells are generated?

10

The results described above were generated by adoptively transferring splenocytes from

11

HBV-immunized mice to HBV transgenic mice. While these results suggest that

12

intrahepatic antigen recognition has a profound impact on the distribution, expansion, and

13

effector functions of primed effector-memory CD8+ T cells, they may not reflect the

14

events that occur when a naïve individual becomes infected with HBV, because

15

immunologically naïve T cells are known to behave very differently than memory T

16

cells.60-62 Accordingly, we generated transgenic mice whose CD8+ T cells express T cell

17

receptors (TCRs) specific for HBV, to study where and how immunologically naïve T

18

cells are primed in response to intrahepatic HBV.

19

When HBV-specific naïve CD8+ T cells were adoptively transferred into HBV transgenic

20

mice, they were predominantly activated rapidly in the liver, independent of T cell

21

homing to lymphoid organs, suggesting that HBV-specific native T cells are primed in

22

the liver.11 While this intrahepatic priming of HBV-specific CD8+ T cells challenges the

This article is protected by copyright. All rights reserved.

Accepted Article

13

1

current T cell priming model postulating that naïve T cells are primed by activated

2

professional antigen presenting cells (pAPCs) in lymphoid organs,63,64 several studies

3

previously indicated that the liver is an exception to this rule.45,46,65 This rather unusual

4

ability of the liver to prime naïve T cells presumably reflects the unique architecture of

5

the hepatic sinusoid, which is characterized by a discontinuous endothelium, the absence

6

of a basement membrane, and a very slow flow rate,66 allowing circulating T cells to

7

make prolonged direct contact with resident liver cells, including hepatocytes.

8

Furthermore, the liver is replete with diverse and unique antigen presenting cell

9

populations, including liver sinusoidal endothelial cells (LSECs), hepatic stellate cells

10

(HSCs), Kupffer cells, and conventional and plasmacytoid dendritic cells, all of which

11

are capable of priming and/or tolerizing naïve T cells.67 In our model, HBV-specific

12

CD8+ T cells are presumably primed by HBV-expressing hepatocytes, because HBV-

13

specific CD8+ T cells were activated when co-cultured with hepatocytes isolated from

14

HBV transgenic mice but not with LSECs, liver residential dendritic cells (DCs) or

15

Kupffer cells. Importantly, the intrahepatically primed HBV-specific CD8+ T cells

16

expand vigorously in the liver but are functionally impaired because they do not express

17

IFN-γ or the cytolytic granule Granzyme B, suggesting that intrahepatic priming by

18

HBV-expressing hepatocytes expands functionally defective CD8+ T cells. Consequently,

19

such intrahepatically primed T cells are incapable of suppressing HBV gene expression

20

or inducing liver disease.11 Interestingly, functionally defective HBV-specific CD8+ T

21

cells expressed a co-inhibitory molecule, PD-1, after antigen recognition in the liver.11

22

Furthermore, a significant fraction of PD-1 deficient HBV-specific naïve T cells

23

expanded more vigorously than PD-1 positive controls, and differentiated into effector T

This article is protected by copyright. All rights reserved.

Accepted Article

14

1

cells capable of producing IFN-γ, as well as Granzyme B, which in turn suppressed HBV

2

gene expression and caused liver disease.11 Collectively, these results suggest that PD-1

3

signaling impairs the effector functions of HBV-specific CD8+ T cells. Importantly, PD-1

4

expression was also observed on HBV-specific CD8+ T cells in chronic HBV patients,68

5

indicating that PD-1 mediated functional suppression is active during natural HBV

6

infection. Taken together, these results suggest that intrahepatic priming induces T cell

7

tolerance and such tolerance is at least partially mediated by PD-1 signaling. This PD-1

8

meditated intrahepatic suppression might be the underlying cause of the weak T cell

9

responsiveness during chronic HBV infection.

10 11

5. Other Mechanisms of HBV Persistence

12

The potential mechanisms by which HBV evades the immune responses are listed on

13

Table 1.

14

following mother-infant transmission.69,70 The underlying cause of adult onset chronic

15

HBV infection is probably multifaceted. Potential contributing factors include mutational

16

escape leading to inactivation of B-cell and T-cell epitopes and specific inhibition of the

17

adaptive immune response by viral proteins.71 For example, HBeAg has been shown to

18

suppress the antibody and T cell response to HBcAg in T cell receptor transgenic

19

mice.12,13 The immunosuppressive potential of HBeAg might explain the clinical

20

observation that viral mutations precluding the production of HBeAg are often associated

21

with exacerbations of liver disease and, sometimes, even with viral clearance from

22

chronically infected patients.72,73 HBsAg might also prevent immune elimination of

Neonatal tolerance to HBV is probably responsible for viral persistence

This article is protected by copyright. All rights reserved.

Accepted Article

15

1

infected cells by functioning as a high dose tolerogen, because extremely high serum

2

HBsAg titers in the mg/ml range are often seen in chronically infected patients74,75 and

3

chronically infected patients have an absence or subnormal levels of HBsAg-specific

4

CD8+ T cells.75 In addition, HBV X protein, a transcriptional transactivator that is

5

required for the initiation of infection,76,77 can inhibit cellular proteasome activity when it

6

is overexpressed78 and might interfere with antigen processing and presentation. Finally,

7

genotypic variation clearly influences the outcome of HBV infection. Up to 10% of adult

8

onset infections with genotype A become persistent, while less than 5% of infections with

9

other genotypes do so.15 The precise mechanism for the difference is unclear, but the high

10

levels of HBsAg expression and low replication activity of genotype A infections might

11

facilitate immune evasion.16

12

In both neonate and adult onset of chronic HBV infections, weak CD4 and CD8 T cell

13

responses are clearly responsible for the viral persistence. The mechanism that suppresses

14

HBV-specific immune responses during chronic HBV infection is likely multifactorial. In

15

addition to PD-1, other negative signaling molecules, such as cytotoxic T lymphocyte

16

antigen-4 (CTLA-4) and T-cell immunoglobulin and mucin 3 (Tim-3), are expressed on

17

functionally compromised HBV-specific CD8 T cells in chronic HBV patients.

18

example, a fraction of CD8 T cells in patients with CHB express the coinhibitory

19

molecule CTLA-4, and its expression level is correlated with serum HBV DNA titer and

20

proapoptotic protein Bim expression. Blockade of CTLA-4 increases the expansion of

21

interferon gamma (IFN-γ) producing HBV-specific CD8 T cells in vitro. Similarly, Tim-

22

3 is also highly expressed on functionally impaired HBV-specific CD8 T cells in chronic

23

HBV patients, and the blockade of Tim-3 in vitro restored effector functions of the CD8 This article is protected by copyright. All rights reserved.

79

For

Accepted Article

16

79

1

T cells,

suggesting that multiple layers of negative co-regulation contribute to T cell

2

exhaustion in chronic HBV infection. Interestingly, Tim-3 expressing CD8 T cells appear

3

to have a immunosuppressive function similar to regulatory T cells (Tregs), limiting

4

proliferation of antigen-specific effector T cells in vitro and in vivo.80 Extrinsic factors

5

could also contribute to the reduced T cell responses during chronic HBV infection. The

6

livers of chronic HBV patients have been shown to express elevated levels of IL-10,81

7

TGF-β,82 and arginase,83 all of which are known to suppress cellular immune responses.

8

These immunosuppressive soluble factors might be produced by regulatory cells, as the

9

frequency of regulatory T cells (Tregs) is increased in chronic HBV patients.84

10

Interestingly, recently identified myeloid-derived suppressor cells (MDSCs) are shown to

11

induce T cell tolerances in mouse models of HBV infection.85,86 In addition, several

12

studies suggest that the number and functionality of DCs are reduced in chronic HBV

13

patients compared with healthy donors, and the reduction of HBV DNA titer by a

14

nucleotide analogue adefovir dipivoxil significantly restored the number and

15

functionality of DCs. Caution should be exercised, however, to interpret the physiological

16

relevance of the antigen nonspecific immunosuppression during chronic HBV infections,

17

because chronic HBV patients are no more susceptible than healthy people to other

18

infectious agents such as influenza virus, etc.

19 20

6. New immunotherapeutic strategies to cure chronic HBV infections

21

The management of chronic HBV has improved significantly in the last decade, mainly

22

because of the development of very effective and safe nucleoside analogues (NAs) that This article is protected by copyright. All rights reserved.

Accepted Article

17

1

primarily inhibit reverse transcription and DNA replication.87 Although NAs strongly

2

inhibit HBV replication and have much fewer side effects than IFN-based therapies, they

3

usually do not achieve sustained viral suppression. Failure to sustain viral suppression by

4

NAs presumably reflects the persistence of cccDNA, which is untouched by reverse

5

transcriptase inhibitors. Because the elimination of cccDNA usually requires the turnover

6

of infected hepatocytes, the induction of functional HBV-specific CD8+ T cells that can

7

specifically kill virus infected hepatocytes has been deemed the most promising approach

8

to cure chronic HBV infections. Immunization of chronically infected patients is the most

9

straightforward approach to boost HBV-specific CD8+ T cell responses. Several

10

strategies have been tested in clinical trials with disappointing results,88 indicating that

11

more inventive approaches are required to induce functional HBV-specific CD8+ T cell

12

responses. There are three approaches that could effectively reinvigorate HBV-specific

13

CD8+ T cell responses (Table 2).

14

The first approach is to block signaling pathways that negatively regulate HBV-specific

15

CD8+ T cell responses. As described in section 4, we showed that blockade of PD-1

16

signaling induced functional differentiation of intrahepatically primed, HBV-specific

17

CD8 T cells in an HBV transgenic mouse model, indicating that PD-1 could be a new

18

therapeutic target to treat chronic HBV infection. HBV-specific CD8 T cell responses

19

might be restored by blocking other negative signaling molecules, such as CTLA-4, Tim-

20

3, and arginase, as well as by depleting Tregs. More evidence is needed to support this

21

approach.

22

The second approach is to provide extra stimulus to override negative signaling that

23

suppresses effector functions of HBV-specific CD8 T cells. One of the promising

This article is protected by copyright. All rights reserved.

Accepted Article

18

1

approaches is to activate professional antigen presenting cells (pAPCs), particularly

2

dendritic cells (DCs). Activated DCs are capable of providing the second and third

3

signals necessary for the functional differentiation of HBV-specific CD8+ T cells. Indeed,

4

we showed that activation of myeloid DCs (mDC) with an agonistic anti-CD40 antibody

5

(αCD40) restored HBV-specific CD8+ T cell responses that are otherwise suppressed by

6

PD-1 signaling.11 Interestingly, PD-1 expression on the functional HBV-specific CD8+ T

7

cells is strongly down-regulated in αCD40 treated HBV transgenic mice. These results

8

suggest that activation of myeloid dendritic cells directly or indirectly suppresses PD-1

9

expression, thereby rescuing HBV-specific CD8+ T cells from PD-1 mediated functional

10

suppression. The data also illustrate the therapeutic potential of mDC activation by

11

αCD40 for the treatment of chronic HBV infection. Other signaling pathways that

12

stimulate pAPCs, such as toll-like receptor (TLR) signaling pathways, should be tested

13

for their ability to restore HBV-specific CD8+ T cell responses. In addition, cytokines

14

required for T cell expansion and differentiation, particularly common gamma-chain

15

cytokines such as IL-2, IL-4, IL-7, IL-15, and IL-21, could serve as effective adjuvants

16

for therapeutic vaccine against chronic HBV infections.

17

The third approach is to genetically engineer peripheral blood T cells from chronic HBV

18

patients to express T cell receptors (TCRs) specific for HBV-derived epitopes. The first

19

and second strategies described above require the presence of HBV-specific CD8+ T cells

20

that can efficiently recognize HBV-derived T cell epitopes presented on infected

21

hepatocytes. However, such T cells are often deleted in chronic HBV patients after years

22

of exposure to HBV antigens. To circumvent this problem, genes encoding TCRs that

23

efficiently recognize HBV derived epitopes can be introduced into the patient’s

This article is protected by copyright. All rights reserved.

Accepted Article

19

1

peripheral blood T cells using retroviral or lentiviral vectors. These TCR redirected

2

autologous T cells could be subsequently transferred into the chronic HBV patient. Such

3

approaches are employed in treating cancer patients89,90 and the results so far have been

4

very encouraging. In addition, T cells with redirected specificity toward the HBV

5

envelope protein have been shown to recognize and lyse HCC cell lines with natural

6

HBV DNA integration in in vitro and animal models.91 Safety concerns, cost

7

effectiveness, and ethical issues should be fully addressed before this promising approach

8

is adapted to chronic HBV patients.

9

10

Summary and Conclusions

11 12

In summary, HBV acts like a stealth virus early in infection, remaining undetected and

13

spreading until the onset of the adaptive immune response several weeks later. The

14

relative invisibility of HBV to the innate sensing machinery of the cells probably reflects

15

its replication strategy, with the replicating viral genome being sheltered within viral

16

capsid particles in the cytoplasm. On the other hand, HBV can be controlled when

17

properly activated HBV-specific CD8+ T cells enter the liver, recognize antigen, kill

18

infected cells, and secrete IFN-γ, which triggers a broad-based cascade that amplifies the

19

inflammatory process and has noncytopathic antiviral activity against HBV. However,

20

HBV-specific CD8+ T cell responses are subjected to a powerful immunoregulatory

21

mechanism in the liver and the induction of an effective HBV-specific CD8+ T cell

22

response is dependent on activation of pAPCs. Failure to activate pAPCs induces

23

functionally impaired CD8+ T cell responses due to intrahepatic priming by HBVThis article is protected by copyright. All rights reserved.

Accepted Article

20

1

expressing hepatocytes, leading to persistent infection (Figure 2). The precise mechanism

2

by which pAPCs are activated during HBV infection remains to be elucidated but HBV-

3

specific CD4+ T cells probably play a critical role. The functional impairment is

4

mediated, at least partially, by PD-1 signaling but other co-inhibitory molecules are likely

5

to be involved as well. Thus, activating pAPCs and suppressing PD-1 and other negative

6

signaling pathways might have therapeutic potential to treat chronic HBV patients.

7

Adoptive transfer of genetically engineered T cells with redirected specificity towards

8

HBV might represent a powerful immune strategy to cure chronic HBV infection, but

9

safety, cost, and ethical issues should be fully addressed before clinical trials.

10 11

Acknowledgements

12

This work was supported in part by a grant-in-aid from the Ministry of Health, Labour,

13

and Welfare of Japan and the Ministry of Education, Culture, Sports, Science and

14

Technology, Japan.

15

This article is protected by copyright. All rights reserved.

Accepted Article

21

1 2 3

References:

4

1.

5 6

consequences. N Engl J Med 350, 1118–1129 (2004).

2.

3.

9

10

Chisari, F. V. Rous-Whipple Award Lecture. Viruses, immunity, and cancer:

lessons from hepatitis B. Am J Pathol 156, 1–16 (2000).

7 8

Ganem, D. & Prince, A. M. Hepatitis B virus infection--natural history and clinical

Guidotti, L. G., Matzke, B., Schaller, H. & Chisari, F. V. High-level hepatitis B

virus replication in transgenic mice. J Virol 69, 6158–6169 (1995).

4.

Sugiyama, M. et al. Direct Cytopathic Effects of Particular Hepatitis B Virus

11

Genotypes in Severe Combined Immunodeficiency Transgenic With Urokinase-

12

Type Plasminogen Activator Mouse With Human Hepatocytes. Gastroenterology

13

136, 652–662.e3 (2009).

14

5.

Wieland, S., Thimme, R., Purcell, R. H. & Chisari, F. V. Genomic analysis of the

15

host response to hepatitis B virus infection. Proc Natl Acad Sci U S A 101, 6669–

16

6674 (2004).

17

6.

18 19

Asabe, S. et al. The size of the viral inoculum contributes to the outcome of

hepatitis B virus infection. J Virol 83, 9652–9662 (2009).

7.

Hadler, S. C. et al. Outcome of hepatitis B virus infection in homosexual men and

20

its relation to prior human immunodeficiency virus infection. J. Infect. Dis. 163,

21

454–459 (1991).

22 23

8.

Puoti, M., Torti, C., Bruno, R., Filice, G. & Carosi, G. Natural history of chronic

hepatitis B in co-infected patients. J Hepatol 44, S65–70 (2006).

This article is protected by copyright. All rights reserved.

Accepted Article

22

1

9.

Boni, C. et al. Lamivudine treatment can overcome cytotoxic T-cell

2

hyporesponsiveness in chronic hepatitis B: new perspectives for immune therapy.

3

Hepatology 33, 963–971 (2001).

4

10.

5 6

Isogawa, M., Furuichi, Y. & Chisari, F. V. Oscillating CD8(+) T cell effector

functions after antigen recognition in the liver. Immunity 23, 53–63 (2005).

11.

Isogawa, M., Chung, J., Murata, Y., Kakimi, K. & Chisari, F. V. CD40 Activation

7

Rescues Antiviral CD8+ T Cells from PD-1-Mediated Exhaustion. PLOS

8

Pathogens 9, e1003490 (2013).

9

12.

10 11

Chen, M. et al. Immune tolerance split between hepatitis B virus precore and core

proteins. J Virol 79, 3016–3027 (2005).

13.

Chen, M. et al. A function of the hepatitis B virus precore protein is to regulate the

12

immune response to the core antigen. Proc Natl Acad Sci U S A 101, 14913–14918

13

(2004).

14

14.

Kakimi, K., Isogawa, M., Chung, J., Sette, A. & Chisari, F. V. Immunogenicity

15

and tolerogenicity of hepatitis B virus structural and nonstructural proteins:

16

implications for immunotherapy of persistent viral infections. J Virol 76, 8609–

17

8620 (2002).

18

15.

Ozasa, A. et al. Influence of genotypes and precore mutations on fulminant or

19

chronic outcome of acute hepatitis B virus infection. Hepatology 44, 326–334

20

(2006).

21

16.

Sugiyama, M. et al. Influence of hepatitis B virus genotypes on the intra- and

22

extracellular expression of viral DNA and antigens. Hepatology 44, 915–924

23

(2006).

This article is protected by copyright. All rights reserved.

Accepted Article

23

1

17.

2 3

table of contents (2001).

18.

4 5

19.

20.

Summers, J. & Mason, W. Replication of the genome of a hepatitis B--like virus

by reverse transcription of an RNA intermediate. Cell 29, 403–415 (1982).

21.

10 11

Su, A. et al. Genomic analysis of the host response to hepatitis C virus infection.

Proc Natl Acad Sci U S A 99, 15669–15674 (2002).

8 9

Stefan F Wieland, F. V. C. Stealth and Cunning: Hepatitis B and Hepatitis C

Viruses. Journal of Virology 79, 9369–9380 (2005).

6 7

Samuel, C. E. Antiviral actions of interferons. Clin. Microbiol. Rev. 14, 778–809–

Seeger, C. & Mason, W. S. Hepatitis B virus biology. Microbiol. Mol. Biol. Rev.

64, 51–68 (2000).

22.

Galibert, F., Mandart, E., Fitoussi, F., Tiollais, P. & Charnay, P. Nucleotide

12

sequence of the hepatitis B virus genome (subtype ayw) cloned in E. coli. Nature

13

281, 646–650 (1979).

14

23.

Wieland, S. F., Guidotti, L. G. & Chisari, F. V. Intrahepatic induction of

15

alpha/beta interferon eliminates viral RNA-containing capsids in hepatitis B virus

16

transgenic mice. J Virol 74, 4165–4173 (2000).

17

24.

Isogawa, M., Robek, M. D., Furuichi, Y. & Chisari, F. V. Toll-like receptor

18

signaling inhibits hepatitis B virus replication in vivo. J Virol 79, 7269–7272

19

(2005).

20

25.

21 22 23

Dunn, C. et al. Temporal analysis of early immune responses in patients with acute

hepatitis B virus infection. Gastroenterology 137, 1289–1300 (2009).

26.

Zhao, J. et al. Natural killer cells are characterized by the concomitantly increased

interferon-γ and cytotoxicity in acute resolved hepatitis B patients. PLoS ONE 7,

This article is protected by copyright. All rights reserved.

Accepted Article

24

1 2

e49135 (2012).

27.

3 4

Lunemann, S. et al. Compromised function of natural killer cells in acute and

chronic viral hepatitis. J. Infect. Dis. 209, 1362–1373 (2014).

28.

Tjwa, E. T. T. L., van Oord, G. W., Hegmans, J. P., Janssen, H. L. A. & Woltman,

5

A. M. Viral load reduction improves activation and function of natural killer cells

6

in patients with chronic hepatitis B. J Hepatol 54, 209–218 (2011).

7

29.

Oliviero, B. et al. Natural Killer Cell Functional Dichotomy in Chronic Hepatitis B

8

and Chronic Hepatitis C Virus Infections. Gastroenterology 137, 1151–1160.e7

9

(2009).

10

30.

Dunn, C. et al. Cytokines induced during chronic hepatitis B virus infection

11

promote a pathway for NK cell-mediated liver damage. J Exp Med 204, 667–680

12

(2007).

13

31.

Peppa, D. et al. Up-regulation of a death receptor renders antiviral T cells

14

susceptible to NK cell-mediated deletion. Journal of Experimental Medicine 210,

15

99–114 (2013).

16

32.

Kakimi, K., Guidotti, L. G., Koezuka, Y. & Chisari, F. V. Natural killer T cell

17

activation inhibits hepatitis B virus replication in vivo. J Exp Med 192, 921–930

18

(2000).

19

33.

20 21

Fisicaro, P. et al. Early kinetics of innate and adaptive immune responses during

hepatitis B virus infection. Gut 58, 974–982 (2009).

34.

Guy, C. S., Mulrooney-Cousins, P. M., Churchill, N. D. & Michalak, T. I.

22

Intrahepatic Expression of Genes Affiliated with Innate and Adaptive Immune

23

Responses Immediately after Invasion and during Acute Infection with

This article is protected by copyright. All rights reserved.

Accepted Article

25

1 2

Woodchuck Hepadnavirus. Journal of Virology 82, 8579–8591 (2008).

35.

3 4

mouse model of hepatitis B virus infection. Immunity 16, 583–594 (2002).

36.

5 6

Baron, J. L. et al. Activation of a nonclassical NKT cell subset in a transgenic

Zeissig, S. et al. Hepatitis B virus–induced lipid alterations contribute to natural

killer T cell–dependent protective immunity. Nat Med 18, 1060–1068 (2012).

37.

Kenna, T. et al. NKT Cells from Normal and Tumor-Bearing Human Livers Are

7

Phenotypically and Functionally Distinct from Murine NKT Cells. The Journal of

8

Immunology 171, 1775–1779 (2003).

9

38.

10 11

independent and a T-cell-dependent antigen. Science 234, 1398–1401 (1986).

39.

12 13

Milich, D. & McLachlan, A. The nucleocapsid of hepatitis B virus is both a T-cell-

Chisari, F. V. & Ferrari, C. Hepatitis B virus immunopathogenesis. Annu Rev

Immunol 13, 29–60 (1995).

40.

Kusumoto, S., Tanaka, Y., Ueda, R. & Mizokami, M. Reactivation of hepatitis B

14

virus following rituximab-plus-steroid combination chemotherapy. J.

15

Gastroenterol. 46, 9–16 (2011).

16

41.

17 18

acute and chronic hepatitis B virus infection. J Immunol 145, 3442–3449 (1990).

42.

19 20

23

Ferrari, C. et al. Identification of immunodominant T cell epitopes of the hepatitis

B virus nucleocapsid antigen. J Clin Invest 88, 214–222 (1991).

43.

21 22

Ferrari, C. et al. Cellular immune response to hepatitis B virus-encoded antigens in

Thimme, R. et al. CD8(+) T cells mediate viral clearance and disease pathogenesis

during acute hepatitis B virus infection. J Virol 77, 68–76 (2003).

44.

Castellino, F. & Germain, R. Cooperation between CD4+ and CD8+ T cells: when,

where, and how. Annu Rev Immunol 24, 519–540 (2006).

This article is protected by copyright. All rights reserved.

Accepted Article

26

1

45.

Bertolino, P., Bowen, D. G., McCaughan, G. W. & Fazekas de St Groth, B.

2

Antigen-specific primary activation of CD8+ T cells within the liver. J Immunol

3

166, 5430–5438 (2001).

4

46.

Limmer, A. et al. Efficient presentation of exogenous antigen by liver endothelial

5

cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat Med 6, 1348–

6

1354 (2000).

7

47.

Kamatani, Y. et al. A genome-wide association study identifies variants in the

8

HLA-DP locus associated with chronic hepatitis B in Asians. Nat. Genet. 41, 591–

9

595 (2009).

10

48.

11 12

liver damage in patients with chronic hepatitis B. Hepatology 51, 81–91 (2009).

49.

13 14

Zhang, J.-Y. et al. Interleukin-17-producing CD4+ T cells increase with severity of

Zhang, Y. et al. A Proinflammatory Role for Interleukin-22 in the Immune

Response to Hepatitis B Virus. Gastroenterology 141, 1897–1906 (2011).

50.

Rehermann, B., Lau, D., Hoofnagle, J. H. & Chisari, F. V. Cytotoxic T

15

lymphocyte responsiveness after resolution of chronic hepatitis B virus infection. J

16

Clin Invest 97, 1655–1665 (1996).

17

51.

Maini, M. K. et al. The role of virus-specific CD8(+) cells in liver damage and

18

viral control during persistent hepatitis B virus infection. J Exp Med 191, 1269–

19

1280 (2000).

20

52.

21 22 23

Guidotti, L. G. et al. Viral clearance without destruction of infected cells during

acute HBV infection. Science 284, 825–829 (1999).

53.

Yoo, J. Y., Howard, R., Waggoner, J. G. & Hoofnagle, J. H. Peroxidase-anti-

peroxidase detection of hepatitis B surface and core antigen in liver biopsy

This article is protected by copyright. All rights reserved.

Accepted Article

27

1

specimens from patients with chronic type B hepatitis. J. Med. Virol. 23, 273–281

2

(1987).

3

54.

4 5

Guidotti, L. G. et al. Intracellular inactivation of the hepatitis B virus by cytotoxic

T lymphocytes. Immunity 4, 25–36 (1996).

55.

Wieland, S. F., Eustaquio, A., Whitten-Bauer, C., Boyd, B. & Chisari, F. V.

6

Interferon prevents formation of replication-competent hepatitis B virus RNA-

7

containing nucleocapsids. Proc Natl Acad Sci U S A 102, 9913–9917 (2005).

8

56.

9

replication by interferon requires proteasome activity. J Virol 76, 3570–3574

10 11

Robek, M. D., Wieland, S. F. & Chisari, F. V. Inhibition of hepatitis B virus

(2002).

57.

Robek, M. D., Boyd, B. S., Wieland, S. F. & Chisari, F. V. Signal transduction

12

pathways that inhibit hepatitis B virus replication. Proc Natl Acad Sci U S A 101,

13

1743–1747 (2004).

14

58.

15 16

Guidotti, L. G. & Chisari, F. V. Immunobiology and pathogenesis of viral

hepatitis. Annu Rev Pathol 1, 23–61 (2006).

59.

Nakamoto, Y., Guidotti, L., Pasquetto, V., Schreiber, R. & Chisari, F. Differential

17

target cell sensitivity to CTL-activated death pathways in hepatitis B virus

18

transgenic mice. J Immunol 158, 5692–5697 (1997).

19

60.

20 21

Lanzavecchia, A., Lezzi, G. & Viola, A. From TCR engagement to T cell

activation: a kinetic view of T cell behavior. Cell 96, 1–4 (1999).

61.

Valitutti, S., Muller, S., Dessing, M. & Lanzavecchia, A. Different responses are

22

elicited in cytotoxic T lymphocytes by different levels of T cell receptor

23

occupancy. J Exp Med 183, 1917–1921 (1996).

This article is protected by copyright. All rights reserved.

Accepted Article

28

1

62.

Zimmermann, C., Prevost-Blondel, A., Blaser, C. & Pircher, H. Kinetics of the

2

response of naive and memory CD8 T cells to antigen: similarities and differences.

3

Eur J Immunol 29, 284–290 (1999).

4

63.

Sprent, J. & Surh, C. D. T CELL MEMORY.

5

http://dx.doi.org/10.1146/annurev.immunol.20.100101.151926 20, 551–579

6

(2002).

7

64.

8 9

Banchereau, J. & Steinman, R. M. Dendritic cells and the control of immunity.

Nature 392, 245–252 (1998).

65.

10

Ando, K., Guidotti, L. G., Cerny, A., Ishikawa, T. & Chisari, F. V. CTL access to

tissue antigen is restricted in vivo. J Immunol 153, 482–488 (1994).

11

66.

Crispe, I. Hepatic T cells and liver tolerance. Nat Rev Immunol 3, 51–62 (2003).

12

67.

Thomson, A. W. & Knolle, P. A. Antigen-presenting cell function in the

13 14

tolerogenic liver environment. Nat Rev Immunol 10, 753–766 (2010).

68.

Boettler, T. et al. Expression of the interleukin-7 receptor alpha chain (CD127) on

15

virus-specific CD8+ T cells identifies functionally and phenotypically defined

16

memory T cells during acute resolving hepatitis B virus infection. J Virol 80,

17

3532–3540 (2006).

18

69.

19 20

Milich, D. R. et al. Is a function of the secreted hepatitis B e antigen to induce

immunologic tolerance in utero? Proc Natl Acad Sci U S A 87, 6599–6603 (1990).

70.

Alexander, G. J. & Eddleston, A. L. Does maternal antibody to core antigen

21

prevent recognition of transplacental transmission of hepatitis-B-virus infection?

22

Lancet 1, 296–297 (1986).

23

71.

Chisari, F. V. et al. A transgenic mouse model of the chronic hepatitis B surface

This article is protected by copyright. All rights reserved.

Accepted Article

29

1 2

antigen carrier state. Science 230, 1157–1160 (1985).

72.

3 4

hepatitis B virus. Gut 37, 568–573 (1995).

73.

5 6

Tillmann, H. et al. Clinical relevance of mutations in the precore genome of the

Ehata, T. et al. Mutations in core nucleotide sequence of hepatitis B virus correlate

with fulminant and severe hepatitis. J Clin Invest 91, 1206–1213 (1993).

74.

Webster, G. J. M. et al. Longitudinal Analysis of CD8+ T Cells Specific for

7

Structural and Nonstructural Hepatitis B Virus Proteins in Patients with Chronic

8

Hepatitis B: Implications for Immunotherapy. Journal of Virology 78, 5707–5719

9

(2004).

10

75.

Reignat, S. et al. Escaping high viral load exhaustion: CD8 cells with altered

11

tetramer binding in chronic hepatitis B virus infection. J Exp Med 195, 1089–1101

12

(2002).

13

76.

14 15

Zoulim, F., Saputelli, J. & Seeger, C. Woodchuck hepatitis virus X protein is

required for viral infection in vivo. Journal of Virology 68, 2026–2030 (1994).

77.

Zhang, Z., Torii, N., Hu, Z., Jacob, J. & Liang, T. J. X-deficient woodchuck

16

hepatitis virus mutants behave like attenuated viruses and induce protective

17

immunity in vivo. J Clin Invest 108, 1523–1531 (2001).

18

78.

Zhang, Z., Protzer, U., Hu, Z., Jacob, J. & Liang, T. J. Inhibition of Cellular

19

Proteasome Activities Enhances Hepadnavirus Replication in an HBX-Dependent

20

Manner. Journal of Virology 78, 4566–4572 (2004).

21

79.

22 23

Nebbia, G. et al. Upregulation of the Tim-3/galectin-9 pathway of T cell

exhaustion in chronic hepatitis B virus infection. PLoS ONE 7, e47648 (2012).

80.

Dolina, J. S., Braciale, T. J. & Hahn, Y. S. Liver-primed CD8 +T cells suppress

This article is protected by copyright. All rights reserved.

Accepted Article

30

1

antiviral adaptive immunity through galectin-9-independent T-Cell

2

immunoglobulin and mucin 3 engagement of high-mobility group box 1 in mice.

3

Hepatology 59, 1351–1365 (2014).

4

81.

5 6

Maini, M. K. & Schurich, A. The molecular basis of the failed immune response in

chronic HBV: therapeutic implications. J Hepatol 52, 616–619 (2010).

82.

Sun, C. et al. TGF-β1 down-regulation of NKG2D/DAP10 and 2B4/SAP

7

expression on human NK cells contributes to HBV persistence. PLOS Pathogens

8

8, e1002594 (2012).

9

83.

Das, A. et al. Functional skewing of the global CD8 T cell population in chronic

10

hepatitis B virus infection. Journal of Experimental Medicine 205, 2111–2124

11

(2008).

12

84.

Xu, D. et al. Circulating and liver resident CD4+CD25+ regulatory T cells actively

13

influence the antiviral immune response and disease progression in patients with

14

hepatitis B. J Immunol 177, 739–747 (2006).

15

85.

Chen, S., Akbar, S. M. F., Abe, M., Hiasa, Y. & Onji, M. Immunosuppressive

16

functions of hepatic myeloid-derived suppressor cells of normal mice and in a

17

murine model of chronic hepatitis B virus. Clinical & Experimental Immunology

18

166, 134–142 (2011).

19

86.

Kong, X., Sun, R., Chen, Y., Wei, H. & Tian, Z. T Cells Drive Myeloid-Derived

20

Suppressor Cell-Mediated CD8+ T Cell Exhaustion in Hepatitis B Virus-Induced

21

Immunotolerance. The Journal of Immunology 193, 1645–1653 (2014).

22 23

87.

Gish, R., Jia, J.-D., Locarnini, S. & Zoulim, F. Selection of chronic hepatitis B

therapy with high barrier to resistance. Lancet Infect Dis 12, 341–353 (2012).

This article is protected by copyright. All rights reserved.

Accepted Article

31

1

88.

Michel, M.-L., Deng, Q. & Mancini-Bourgine, M. Therapeutic vaccines and

2

immune-based therapies for the treatment of chronic hepatitis B: perspectives and

3

challenges. J Hepatol 54, 1286–1296 (2011).

4

89.

Rosenberg, S. A., Restifo, N. P., Yang, J. C., Morgan, R. A. & Dudley, M. E.

5

Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev

6

Cancer 8, 299–308 (2008).

7

90.

8 9

Park, T. S., Rosenberg, S. A. & Morgan, R. A. Treating cancer with genetically

engineered T cells. Trends in Biotechnology 29, 550–557 (2011).

91.

Koh, S. et al. A Practical Approach to Immunotherapy of Hepatocellular

10

Carcinoma Using T Cells Redirected Against Hepatitis B Virus. Mol Ther Nucleic

11

Acids 2, e114 (2013).

12

This article is protected by copyright. All rights reserved.

Accepted Article

32

1

Figure legends

2 3

Figure 1. Schematic representation of immune responses during acute and chronic

4

HBV infections.

5

Figures (a) and (b) schematically represent the time course of serum HBV titer, and

6

immune responses during acute and chronic HBV infections. The upper panel in each

7

figure represents the timing of HBV infection as an arrow, the serum HBV DNA as a

8

dashed black line, and the NK cell response as gray area. The lower panel in each figure

9

represents the timing if CD4 T cell priming to HBV as an arrow, the CD4 T cell response

10

as red area, the CD8 T cell response as green area, the blue line as the antibody response.

11 12

Figure 2. The outcome of T cell priming is determined by the antigen presenting cell

13

population.

14

(a) HBV-specific naive CD8 T cells that are primed by HBV positive hepatocytes receive

15

coinhibitory signals rather than costimulatory signals, resulting in the expansion of

16

functionally impaired CD8 T cells. (b) T cell priming by professional antigen presenting

17

cells, such as DCs, that can deliver costimualtiory signals is presumably required to

18

induce functional HBV-specific CD8 T cell responses.

19 20 21

This article is protected by copyright. All rights reserved.

Accepted Article

33

1 2

Potential mechanisms of HBV persistence            

Neonatal tolerance Peripheral deletion/exhaustion (due to high viral load and negative signaling) Anergy Ignorance Weak CD4 T cell response (associated with low viral titer and HIV infection) Reduced dendritic cell numbers and functions Negative signaling (PD-1/CTLA-4/Tim-3) Immunosuppressive microenvironment (IL-10, TGF-β) Regulatory T cells/Myeloid derived suppressor cells Escape mutations in T cell and B cell epitopes Inhibit antigen processing or presentation Inhibit cytokine signaling

This article is protected by copyright. All rights reserved.

3 4 5 6

Tabl e1

Accepted Article

34

1

2

Strategies to reinvigorate T cell responses

3 1. Block negative signaling pathways (PD-1, CTLA-4, Tim-3, etc.) 4 2. Stimulate pAPCs (anti-CD40, TLR, etc.) and cytokine signaling 5 3. Redirect peripheral T cells with high avidity TCRs

This article is protected by copyright. All rights reserved.

Table 2

Accepted Article

35

1 2

hepr_12439_f1

3

This article is protected by copyright. All rights reserved.

Accepted Article

36

1 2

hepr_12439_f2

This article is protected by copyright. All rights reserved.

Immunobiology of hepatitis B virus infection.

The adaptive immune response, particularly the virus-specific CD8(+) T-cell response, is largely responsible for viral clearance and disease pathogene...
980KB Sizes 1 Downloads 12 Views