I Anna Cone 2021

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

REVIEWS

Immunobiology and pathogenesis


of hepatitis B virus infection
Matteo Iannacone 1,2,3 ✉ and Luca G. Guidotti 1,2 ✉

Abstract | Hepatitis B virus (HBV) is a non-​cytopathic, hepatotropic virus with the potential to cause
a persistent infection, ultimately leading to cirrhosis and hepatocellular carcinoma. Over the past
four decades, the basic principles of HBV gene expression and replication as well as the viral and
host determinants governing infection outcome have been largely uncovered. Whereas HBV
appears to induce little or no innate immune activation, the adaptive immune response mediates
both viral clearance as well as liver disease. Here, we review our current knowledge on the
immunobiology and pathogenesis of HBV infection, focusing in particular on the role of CD8+
T cells and on several recent breakthroughs that challenge current dogmas. For example, we now
trust that HBV integration into the host genome often serves as a relevant source of hepatitis B
surface antigen (HBsAg) expression during chronic infection, possibly triggering dysfunctional
T cell responses and favouring detrimental immunopathology. Further, the unique haemodynamics
and anatomy of the liver — and the changes they frequently endure during disease progression
to liver fibrosis and cirrhosis — profoundly influence T cell priming, differentiation and function.
We also discuss why therapeutic approaches that limit the intrahepatic inflammatory processes
triggered by HBV-​specific T cells might be surprisingly beneficial for patients with chronic infection.

Cirrhosis
Hepatitis B virus (HBV), the prototypical member of the paramount importance for HBV control2–4. Unfortunately,
The final stage of fibrosis Hepadnaviridae family, is a non-​cytopathic DNA virus the vaccine is ineffective in patients who are already
in which fibrous septa that is transmitted by contacts with infected blood and chronically infected, and current therapy for these
surrounding nodules of body fluids and triggers immune-​mediated liver diseases individuals is limited to either immunomodulatory
regenerating hepatocytes
induce profound architectural
of varying severity and duration1. The infectious virion agents — such as conventional or pegylated type I inter-
distortion of the liver and is an enveloped nucleocapsid that selectively enters the ferons — or a few direct-​acting antivirals (DAAs) such
functional insufficiency. hepatocyte and delivers an incomplete circular DNA as entecavir, tenofovir and tenofovir alafenamide2–4.
genome, thereby initiating a multifaceted process of viral These DAAs, referred to as third-​generation nucleos(t)
replication (Box 1; Figs 1,2). ide analogues (NUCs), represent the backbone of treat-
According to WHO (World Health Organization) ment in most countries and — as orally administered
data, almost one-​third of the world’s population has been and well tolerated drugs that potently curtail the activity
infected by HBV at some point in their lives2–4. The vast of the HBV polymerase with extremely low resistance
majority of these people encountered HBV in adulthood, rates — safely and efficiently maintain the viraemia of
1
Division of Immunology, developed a self-​limited infection (that is, acute hepa- patients with CHB below the detection limit of commer-
Transplantation and
titis B (AHB)) and successfully controlled the virus2–4. cial assays2–4. Similar to anti-​HIV drugs, however, NUCs
Infectious Diseases, IRCCS
San Raffaele Scientific
Although fewer than 5% of individuals who encountered rarely achieve complete viral elimination (that is, HBV
Institute, Milan, Italy. HBV as immunocompetent adults develop a persistent eradication from the liver), and most patients therefore
2
Vita-​Salute San Raffaele infection (that is, chronic hepatitis B (CHB)), most of require lifelong treatment2–4.
University, Milan, Italy. the infections acquired in infancy or early childhood Based on the above, and coupled with the fact that
3
Experimental Imaging become chronic2–4. CHB affects more than 250 million NUC treatment does not completely eliminate the risk of
Center, IRCCS San Raffaele individuals worldwide and almost 1 million die annually developing HCC5, it is not surprising that considerable
Scientific Institute, from complications of persistent infection, liver cirrhosis resources are directed at developing ‘curative’ treatments
Milan, Italy.
and hepatocellular carcinoma (HCC)2–4. CHB is highly for CHB. The scientific community has long recognized
✉e-​mail:
endemic in Africa and Asia, as well as parts of Central that a ‘sterilizing’ cure — the permanent elimination of
iannacone.matteo@hsr.it;
guidotti.luca@hsr.it and Eastern Europe2–4. all HBV forms, including the HBV transcriptional tem-
https://doi.org/10.1038/ Preventive measures, including a prophylactic vac- plate (an episomal ‘minichromosome’ termed covalently
s41577-021-00549-4 cine already adopted in all developed nations, remain of closed circular DNA (cccDNA)) (Box 1), from all infected

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

Box 1 | HBV tropism and life cycle


It has been long known that hepatitis B virus (HBV) infects a
only humans, chimpanzees and, to a lesser extent, tree
shrews (Tupaia belangeri) and that the parenchymal cell of LSEC
the liver (the hepatocyte) is the sole site of productive HBV
infection137,138. Through the engagement of specific loops in HSPG
the envelope proteins, circulating virions initially attach to
heparan sulfate proteoglycans (HSPGs)139,140 that extend from
the space of Disse — which separates hepatocytes from liver Space of Disse
sinusoidal endothelial cells (LSECs) — into the lumen of the
sinusoids141 (see the figure). The following step of viral entry
involves the interaction of a specific domain of the HBV L
envelope protein with a high-​affinity receptor on the surface
of the hepatocytes. The nature of this receptor remained ssDNA rcDNA MVB
NTCP
elusive until the laboratory of Wenhui Li in 2012 elegantly or HBV
Capsid virion
identified the sodium taurocholate co-​transporting assembly
polypeptide (NTCP) as the surface molecule allowing HBV
Core Pol dsIDNA
entry into the hepatocyte142. NTCP is a hepatocyte-specific
transporter of bile acids that is predominantly localized in Capsid
disassembly pgRNA Env
the basolateral membrane that faces the sinusoidal lumen143. Filament Sphere
ER SVP SVP
Following viral entry (a process involving still unclear
phases that include membrane fusion), HBV nucleocapsids
containing a relaxed circular DNA (rcDNA) are transported to mRNA
the nucleus, where host enzymes then repair the viral genome cccDNA RNA Precore
into an episomal transcriptional template, the covalently HBeAg
closed circular DNA (cccDNA) . This circular template
10
Nucleus
encodes six capped, polyadenylated and overlapping RNAs Hepatocyte
(Fig. 2) that leave the nucleus unspliced and produce the viral
structural and non-structural proteins10. Part a of the figure b
shows how one of these transcripts — termed pre-​genomic
RNA (pgRNA) — is selectively encapsidated together with the
viral polymerase (Pol) in the hepatocellular cytoplasm10. HBV
replication takes place within these nucleocapsids by pgRNA
reverse transcription, which ultimately produces either
rcDNA or double-​stranded linear DNA (dslDNA) replicative
intermediates, depending on whether a specific event of
RNA primer translocation occurs or not144. Nucleocapsids
containing canonical (rcDNA) or non-​canonical (dslDNA)
genomes traffic back to the nucleus to amplify the pool
of cccDNA molecules or proceed to multivesicular bodies NTCP
(MVBs), where they engage the viral envelope proteins, Capsid
disassembly
bud into the lumen and exit the hepatocyte as infectious pgRNA
virions circulating in the bloodstream10,145. Env Filament Sphere
Part b of the figure shows how HBV nucleocapsids SVP SVP
containing non-​canonical dslDNA replicative intermediates,
once delivered intranuclearly, can serve as substrates for
host genome integration occurring at double-​stranded DNA mRNA
breaks. Integrated dslDNA cannot support viral replication ER
Integration
but allows the expression of specific gene products,
particularly the envelope proteins (Env) that differently
decorate filamentous and spherical subviral particles (SVPs).
SVP morphogenesis occurs at the level of the endoplasmic reticulum (ER), likely starting from the self-​assembly of the S protein into filaments; most
filaments are thought to be converted into spherical SVPs before secretion. HBeAg, HBV precore protein; ssDNA, single-​stranded DNA.

hepatocytes — will be difficult to attain in the foresee- over virions and reach exceptional quantities of up to
able future6,7, particularly given that complete cccDNA >300 μg ml–1 (ref.9). The reason why the virus displays
elimination apparently does not even occur following this extraordinary level of biosynthetic effort is not well
self-​limited AHB8. In its place, the concept of ‘functional’ understood, but a likely explanation is that it allows
cure — the loss of detectable serum hepatitis B surface for the absorption of circulating anti-​HBsAg antibod-
antigen (HBsAg) with or without seroconversion to ies that would otherwise limit the spread of infectious
anti-​HBs antibody — has taken hold. It is notable that virions within the liver. Similar to what is observed
each HBV envelope protein (L, M and S) possesses the in patients with resolved AHB, the serum HBsAg
HBsAg determinant (Fig. 1) and that HBsAg circulates loss associated with the concept of a ‘functional’ cure
in the blood of patients infected with HBV mostly in should serve as a surrogate marker for immune control
the form of non-​infectious subviral spheres and fila- and long-​term suppression of HBV replication. This
ments, which are present in a 102-​fold to 105-​fold excess may represent a more achievable target than complete

www.nature.com/nri

0123456789();:
Reviews

HBV elimination and could allow for cessation of More specifically, experimental evidence demonstrated
NUC treatment6,7. that during self-​limited AHB the highly non-​cytopathic
All immunomodulators and DAAs that have received HBV induces little or no innate immune responses and
approval thus far stemmed from the work of many dif- is controlled (without being completely eliminated) by
ferent laboratories over the last four decades. Indeed, HBV-​specific CD4+ and CD8+ T cell responses as well as
through the study of patients, animal models and cell neutralizing antibodies11,12. Additional work indicated
cultures, the basic principles of HBV gene expression that viral persistence and chronic liver injury mostly
and replication have been unveiled, infectious viral reflect the dysregulation of these adaptive immune
genomes and derivative gene products have been responses11,12, which are also affected by the unique
cloned, sequenced and characterized10 and the main anatomy and haemodynamics of the liver (Box 2).
viral or host determinants governing the outcome of Here, we discuss the main findings of the work car-
infection (AHB versus CHB) have been uncovered11. ried out over the last 40 years and focus on numerous
recent breakthroughs that reveal that some virologic and
immunologic assumptions from the past are likely too
Polymerase
simplistic. For example, it was recently shown that defec-
Core HBcAg tive and replication-​incompetent HBV linear fragments
integrate into the host genome early upon infection13–15
Large envelope
and may serve as the relevant source of HBsAg in the
Middle envelope HBsAg context of chronic infection16 (Box 3). The extent to
which this event triggers dysfunctional T cell responses,
Capsid Capsid section Small envelope
favours detrimental immunopathology and/or influ-
ences therapeutic strategies, will be discussed. Along
42 nm these lines, a present dogma in HBV immunobiology
asserts that high levels of circulating HBsAg negatively
impact both HBV-​specific B and T cells, and, therefore,
strategies that reduce serum HBsAg and lead to serocon-
version could result in the ‘awakening’ of these otherwise
dysfunctional responses. New experimental evidence
that challenges the validity of this dogma will be also dis-
cussed. Further, we review recent work that reveals how
anatomic changes associated with advanced liver fibro-
sis and cirrhosis may affect the priming, differentiation
and effector functions of naive or antigen-​experienced
Hepatitis B virion
(Dane particle) T cells. Finally, we discuss the provocative hypothesis
that patients with CHB might benefit from therapeutic
approaches that limit the pathogenic potential of
HBV-​specific T cells. One example of such an approach
is anti-​platelet therapy (for example, with low-​dose aspi-
rin), which seems to reduce the risk of HCC and related
mortality17 by mechanisms that may include the capacity
of platelets to support the homing of pathogenic T cells
into the liver18,19.

The early phase of infection


It has long been known that, upon entering the circula-
tory system, HBV and related hepadnaviruses retain an
~90% ~10% ~1% extraordinary capacity to reach and infect parenchymal
liver cells (Box 1). Indeed, intravenous inoculation with
just one duck HBV or HBV virion in ducks or chimpan-
zees can result in a productive infection20,21. Anatomic
rcDNA dsIDNA RNA and haemodynamic characteristics of the liver micro-
circulation, such as the protrusion of heparin sulfate
Fig. 1 | HBV particle. Circulating hepatitis B virions (Dane particles) are ~42 nm in proteoglycans (HSPGs) in the lumen of sinusoids and
diameter and are equipped with an envelope accommodating three in-​frame viral gene the slow blood flow that typifies this vascular bed22,23,
products termed large (L), middle (M) and small (S) envelope proteins, each containing are thought to increase the likelihood of HBV eventually
the hepatitis B surface antigen (HBsAg) determinant. The hepatitis B virus (HBV) envelope engaging its receptor on the hepatocellular membrane
encloses an inner nucleocapsid particle (~28 nm in diameter) that is composed, in the vast
and initiating the processes of viral entry and replication
majority of virions, of 120 core protein (also known as HBcAg) homodimers assembled
(Box 1). HBV starts replicating and spreading throughout
into an icosahedral structure with a T = 4 symmetry. In turn, the nucleocapsid particles
contain a copy of a relaxed circular partially double-​stranded DNA genome of ~3.2 kb, the liver quite rapidly, with an estimated doubling time
and a copy of the viral polymerase. In place of relaxed circular DNA (rcDNA), ~10% of the of 2–4 days21,24. Notably, the main features of the early
nucleocapsid particles are thought to contain double-​stranded linear DNA (dslDNA) and phase of infection have been best studied in biopsies col-
~1% of the particles appear to contain mostly unspliced forms of HBV RNA. lected on a weekly basis from young adult chimpanzees

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

PreS2 (that is, the cccDNA molecule), the use of capped and
polyadenylated transcripts resembling host-​derived
1 S mRNAs and the confinement of replicating RNA/DNA
eS
Pr
genomes within cytoplasmic nucleocapsid particles10.
b
Interestingly, more recent work on human liver biop-
2.1 k sies indicated that HBV seems not to elicit detectable
innate immune responses in the CHB setting either29.
e
as

Along with the observation that different aspects of the


er

kb
ym

HBV life cycle are highly sensitive to antiviral molecules


2.4
Pol

produced by experimentally activated innate immune


cells such as intrahepatic antigen-​presenting cells, natu-
ral killer cells and natural killer T cells30–38, these results
suggest that — instead of actively counteracting the
effect of such molecules as many other viruses do39 —
HBV has evolved a hiding strategy to avoid recognition
by the innate immune system, which allows it to spread
and replicate proficiently within the liver.
Regarding the studies in chimpanzees, it is note-
Core

worthy that at the same time points at which the


AA AA
AA
AA AA

hepatic HBV titres and the percentage of infected


AA AAA A

kb
A AA

0.7
3.5 hepatocytes reach their peak, viraemia can be as high as
A

kb kb
3.9 1011 genomes ml–1 and, in keeping with the lack of innate
immune cell-​derived pro-​inflammatory mediators, this
occurs in animals showing no clinical, biochemical or
histological evidence of liver disease21,26–28. Note that sim-
Pr
eC ilar levels of viraemia have been documented in patients
during the incubation phase of infection, at least 4 weeks
before the onset of symptoms40. It therefore appears that
at times of maximum viral production, the capacity of
X this non-​cytopathic virus to transmit to others might be
very high (a nanolitre of blood from a clinically ‘healthy’
Fig. 2 | genomic structure of HBV. The inner helixes represent the partially double-​ individual may contain up to 105 genomes).
stranded, relaxed circular hepatitis B virus (HBV) DNA genome. The outer thin lines
represent the six capped, polyadenylated and overlapping RNAs. Of note, the depicted Priming of adaptive immune responses in immuno­
3.5-​kb RNA represents two transcripts 3.5 kb in length that differ from one another by competent adults infected with HBV. Human and animal
only a few nucleotides. The slightly longer transcript encodes for the precore protein studies over the last 30 years have indicated that the out-
(PreC); the slightly shorter one encodes for both the core protein and the polymerase come of most adult infections is strongly determined
protein. The 3.9-​kb RNA is thought to represent a long form of a HBV xRNA; note that
by the kinetics, breadth, vigour, trafficking and effec-
the canonical HBV xRNA is 0.7 kb in length. The 2.4-​kb RNA encodes for the PreS1 (or L)
envelope protein. The 2.1-​kb RNA encodes for both the PreS2 and S envelope proteins tor functions of HBV-​specific adaptive immune cells1.
(also called M and S). The outermost colour lines indicate the translated HBV proteins. Through their antiviral functions, such as the killing of
infected hepatocytes and local production of antiviral
cytokines, HBV-​specific CD8+ T cells are widely con-
that were inoculated intravenously with transgenic sidered the ultimate effectors of viral clearance during
mouse serum containing varying amounts of the same AHB, whereas HBV-​specific CD4+ T cells are viewed as
infectious HBV DNA (HBV genotype D subtype ayw)25. essential facilitators of the induction and maintenance of
These studies revealed that, depending on the inoculum both CD8+ T cell and antibody responses1. Accordingly,
dose, HBV easily manages to infect 100% of the hepat- the depletion of either CD8+ or CD4+ T cells during the
ocytes (~1011 cells in the chimp or human liver)23 within initial phase of infection in acutely infected chimpan-
a period of 7–10 weeks and this occurs without any evi- zees prevented both viral clearance and the onset of
dence of hepatocellular damage21,26–28. Interestingly, the liver disease21,41. It is of note that, in chimpanzees, anti-
outcome of infection was found not to strictly depend on bodies with neutralizing potential appear only after the
the inoculum dose — meaning that a chronic infection virus has been eliminated from most hepatocytes21,26–28,
with associated immunopathology can also result from suggesting that their main function is to prevent the
the inoculation of a very low HBV dose — but, rather, re-​emergence of HBV following clinical recovery. This
on the relationship between the kinetics of viral spread stands in contrast to the HBcAg-​specific IgM (anti-​HBc)
and the priming of adaptive immunity, particularly that observed early in infection, which can coexist with
of HBV-​specific CD4+ T cells21. These studies in chim- high levels of HBV replication42. The function of these
panzees also uncovered that, during the initial spreading antibodies in HBV pathogenesis is currently unclear43.
phase, HBV is highly efficient at avoiding recognition We still know very little about the timing and the
by the innate immune system26–28, possibly owing to its location of T cell priming in both AHB and CHB. In
unique replication strategy that involves, for instance, the setting of adult infection, and in keeping with data
the nuclear sequestration of the transcriptional template derived from experiments in chimpanzees, HBV-​specific

www.nature.com/nri

0123456789();:
Reviews

Cross-​priming CD8+ T cells have been detected during the initial of HBV-​specific naive CD8+ T cells in the liver does not
A functional outcome of asymptomatic phase of AHB, typically no sooner than promote viral clearance but, rather, contributes to the
cross-​presentation (the 9–11 weeks after infection21,26,40. The presence of these establishment of persistent infection.
presentation of extracellular cells is often kinetically associated with non-​cytolytic, Recent experiments in mouse models of HBV infec-
antigens on MHC class I
molecules), whereby
cytokine-​dependent antiviral events that were first tion have indeed demonstrated that hepatocellular prim-
antigen-​specific naive CD8+ described in HBV transgenic mice 44,45. Why the ing of naive HBV-​specific CD8+ T cells leads to their
T cells are activated by HBV-​specific effector CD8+ T cells cannot be detected local activation and proliferation, but they do not dif-
antigen-​presenting cells at much earlier time points (for instance, 2 or 3 weeks ferentiate into effector cells47,48 (Fig. 3). These studies also
to become effector cells.
after viral exposure when HBV could have infected only revealed that the CD8+ T cell dysfunction induced by
a minority of the hepatocytes) remains to be determined. hepatocellular priming can be counteracted in vivo
The immunological dogma would dictate that naive by treatment with IL-2 (ref.48) (Fig. 3). IL-2 was previously
CD8+ T cells initially encounter their cognate antigen in shown to rescue CD8+ T cells from death induced by
secondary lymphoid organs (SLOs), where cross-​priming in vitro priming by hepatocytes49 and different formula-
by professional antigen-​presenting cells of circulating tions of IL-2 have been clinically used as therapeutics to
virions and subviral particles promotes the differentia- increase T cell responses against viruses or tumours50–52.
tion into effector cells endowed with liver homing poten- The cellular and molecular mechanisms whereby
tial. Yet there is no direct experimental evidence that IL-2 reinvigorates intrahepatically primed T cells and
SLOs are the priming site for CD8+ T cells in humans whether IL-2 plays a role during natural HBV infection
or chimpanzees infected with HBV, and it is assumed remain elusive. However, based on the aforementioned
that intrahepatic priming of naive CD8+ T cells (espe- results48, one could envisage that, during AHB, high local
cially in the presence of high antigen load) promotes the IL-2 concentrations might render the liver fully capable
functional impairment of these cells46. Based on these of supporting efficient HBV-​specific CD8+ T cell prim-
considerations, it is generally believed that the priming ing. A possible source of IL-2 in this setting is effector

Box 2 | The organ: how the liver influences HBV immunobiology and pathogenesis
The liver is composed of functional units (that is, lobules) where plates of hepatocytes one cell thick are in close contact with the
fenestrated and basement membraneless liver sinusoidal endothelial cells (LSECs) that form the organ microcirculation146,147
(see the figure). Hepatocytes and LSECs are separated by the space of Disse where about half of the lymph in the body is
formed148. The liver microenvironment also includes epithelial cells of the bile ducts termed cholangiocytes, intrasinusoidal
macrophages termed Kupffer cells, perisinusoidal myofibroblasts termed hepatic stellate cells and selective populations
of resident cells of lymphoid or myeloid origin that constantly patrol liver sinusoids or reside within periportal areas147,149.
Under both physiologic and pathologic conditions, the liver is biased towards inducing a state of immune unresponsiveness
or hypo-​responsiveness. This involves a complex array of diverse and coordinated cellular and molecular events that,
ultimately, hinder the effector functions of lymphocytes147,149,150. For instance, the unique anatomy and haemodynamics of
the liver sinusoids — through which about one-​third of all blood cells transit slowly every minute23,90 — allow circulating T cells
to sense tolerogenic MHC–Ag complexes and other surface ligands displayed by non-​professional antigen-​presenting cells
such as the hepatocytes without the need for extravasating89,147,149. Such engagement promotes the induction and maintenance
of immune tolerance, explaining, for example, the acceptance of liver allografts across complete MHC mismatch barriers151
or the propensity of hepatitis B virus (HBV) to establish lifelong persistent infections1.

Bile duct Cholangiocyte Hepatocyte Bile canaliculi

Hepatic artery

LSEC Fenestrations

Sinusoid Kupffer cell Central vein

Lymphatic
vessel Space of Disse

Hepatic
stellate cell

Portal vein

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

Box 3 | HBV integration into the host genome Priming and other immune correlates of neonatal and
perinatal infections. As mentioned above, the vast
Hepatitis B virus (HBV) and other closely related hepadnaviruses (such as duck HBV majority of neonatal and perinatal HBV infections
and woodchuck hepatitis virus) tend to extensively integrate into the host genome10 become persistent1. This outcome is thought to be due
(Box 1). The mechanisms governing this phenomenon are poorly defined but it is clear
to immune inhibitory mechanisms that — depending
that non-​canonical double-​stranded linear DNA (dslDNA) replicative intermediates,
on the relative functional avidity of HBV-​specific T cell
once delivered intranuclearly, can serve as substrates for host genome integration
occurring at double-​stranded DNA breaks through non‑homologous end joining or clones for tolerogenic epitopes — either delete these
microhomology-​mediated end joining10. HBV integration can start as early as <3 days cells in the thymus or render them dysfunctional in the
after infection in cell cultures13 and rapid integration events have long been known periphery53. However, direct evidence of thymic deletion
to happen in vivo as well14,15,152. The process of HBV integration has the potential to of T cell clones following viral exposure is lacking.
endlessly continue during persistent infection, but cannot support viral replication. Moreover, circulating HBV-​specific T cells are detecta-
This is because even intact forms of integrated 3.2-​kb dslDNA cannot generate the ble in adolescents and young adults infected neonatally
3.5-​kb pre-​genomic RNA (pgRNA) from which the replication of HBV actually starts. and perinatally (albeit at levels that are both quantita-
Indeed, pgRNA synthesis requires a 3.2-​kb circular genome on which the viral polymerase tively and qualitatively deficient when compared with
recognizes the unique polyadenylation signal not at the first but at the second passage10
those characterizing self-​limited infections acquired in
(Fig. 2), and the redundant sequences that the polymerase uses to produce pgRNA are
adulthood)54, and HBV-​specific T cell clones, although
simply missing in dslDNA forms. Nonetheless, forms of integrated dslDNA can support
the expression of specific gene products. The virologic and tumorigenic implications of tolerized, are still present in fully grown transgenic mice
this phenomenon — which include the production of amino-​terminal truncated versions that were exposed to HBV antigens at birth55,56. These
of the HBV X protein (a transactivator that regulates viral transcription from covalently observations seem to argue against a predominant role
closed circular DNA (cccDNA)) or the generation of HBV–cellular fusion transcripts that of thymic deletion of HBV-​specific T cells in the estab-
yield chimeric proteins with oncogenic potential — have been expertly reviewed lishment of viral persistence. Considering that naive
elsewhere119,153,154. Notably, the open reading frames (ORFs) encoding the viral polymerase, CD8+ T cells emerging from the thymus should easily
the precore protein (a non-​structural gene product whose mature form is secreted in the engage hepatocellular antigens in the poorly inflamed
bloodstream as HBV precore protein (HBeAg)) and the core protein (the structural and liver, and that HBV robustly replicates during the first
non-​secretable component of HBV nucleocapsids also referred to as HBcAg) detach from
20–30 years of life in patients with CHB exposed neo­
their promoters in dslDNA genomes153. As such, HBV integrants may produce HBeAg or
natally/perinatally54, it is plausible that tolerogenic
HBcAg only if properly inserted near active cellular enhancers/promoters and, to our
knowledge, definitive information about this possibility or its related frequency during events that result from the initial antigen encounter in
HBV infection is lacking. A different scenario pertains to the ORFs encoding the L, M and the liver may impact the outcome of infection.
S envelope proteins, which can remain properly positioned under their native promoters As mentioned above, recent experimental evidence in
upon dslDNA integration153 (Box 1). mouse models of HBV infection showed that hepatocel-
It has long been suggested that integration leads to hepatic hepatitis B surface lular priming of naive HBV-​specific CD8+ T cells leads
antigen (HBsAg) expression in patients with chronic hepatitis B (CHB)155 and that to local activation and proliferation of these cells but a
such expression can be significantly dysregulated in vitro or in vivo — for instance, lack of differentiation into effector cells47,48. These dys-
with abnormal production and accumulation of the L envelope protein and/or specific functional cells form clusters that coalesce around portal
L or M mutants in the endoplasmic reticulum and the formation of ground glass
tracts48 (Fig. 3), similar to what is observed in patients
hepatocytes156–158. Ground glass hepatocytes are indeed historical hallmarks of CHB159
with CHB57. This periportal accumulation happens even
and may represent preneoplastic foci of HBV-​related hepatocellular carcinoma
(HCC)158. Taking advantage of patients and chimpanzees with chronic infection though the initial antigen recognition by naive CD8+
undergoing RNA interference (RNAi)-based anti-​HBV treatment, Wooddell et al. T cells occurs homogeneously throughout the liver
recently made a challenging observation: HBV integrants, rather than cccDNA, seem lobule48. Hepatocellular HBV antigen recognition by
to support most of the hepatic HBsAg biosynthesis, particularly in individuals with naive CD8+ T cells initiates a differentiation programme
long-term infection who become serum HBeAg-​negative16. This observation, coupled characterized by a progressive accumulation of tran-
with the evidence that HBV integration can start rapidly after infection and that large scriptional and chromatin changes that, ultimately, result
hepatocyte clones holding integrated HBV DNA can be detected during all stages of in a dysregulated T cell phenotype48. Genes involved in
CHB160, may force us not only to revise the way in which spontaneous or therapy-​induced tissue development, remodelling, cell differentiation
changes in serum HBsAg (or the lack thereof) have been interpreted thus far but also to
and wound healing are upregulated48, suggesting that
reflect on the immunological and pathological consequences of having hepatocellular
this programme might favour tissue protection over
HBsAg continuously produced and presented to T cells or B cells independently of
viral replication. antiviral activity. The acquisition of this dysfunctional
phenotype appears to not be affected by the relative anti-
gen load in the hepatocyte (even very small amounts of
CD4+ T cells; in support of this hypothesis are the obser- HBV antigens can trigger severe T cell dysfunction)48.
vations that CD4+ T cell depletion in chimpanzees prior Interestingly, the transcriptional signature of CD8+
to HBV infection prevents effective CD8+ T cell prim- T cells primed by hepatocytes does not overlap signif-
ing and leads to persistent infection with negligible icantly with that of exhausted T cells described in other
immunopathology21 and that detection of CD8+ T cells viral infections and in cancer48. Consistent with this,
in the liver of chimpanzees infected with HBV coincides hepatocellularly primed CD8+ T cells do not respond
with the detection of CD4+ T cells in the liver26. Without to anti-​PDL1 checkpoint inhibitor therapy48. Whereas
the creation of a manipulable animal model of infection priming by hepatocytes in a non-​inflamed liver (a likely
Checkpoint inhibitor (such as an immunocompetent mouse engineered to scenario following neonatal HBV infection) triggers
therapy support HBV entry, cccDNA formation and productive this unique dysfunctional gene programme, antigen
A form of cancer
immunotherapy targeting
viral replication in the liver), it will be difficult to provide persistence may gradually activate an exhaustion-​like
immune checkpoints (for definitive insights into when or where HBV priming gene signature48,58. These considerations should guide
example, PD1, CTLA4). occurs following the initial exposure. the design and interpretation of ongoing clinical trials

www.nature.com/nri

0123456789();:
Reviews

aimed at evaluating the therapeutic potential of immune T cells60,61 and, more recently, that maternally derived
checkpoint inhibitors in patients with CHB. HBeAg appears to condition the hepatic macrophages
Unfortunately, very few immunological studies have of the offspring to repress HBV-​specific CD8+ T cell
been performed in experimentally infected chimpan- responses by yet undetermined mechanisms 62,63.
zees that eventually developed CHB59, and none of them In keeping with a role for HBeAg in blunting T cell
investigated whether T cell priming can occur in the responses, it has long been known that neonatal infec-
liver. However, one study indicated that HBV-​specific tions with HBeAg-​negative variants often result in
CD4+ T cell responses are not detectable in these ani- viral clearance and that adult infections with these
mals before the peak of viraemia or before the virus variants are usually more clinically severe than those
has infected most hepatocytes21. As the hepatic appear- with HBeAg-​c ompetent genomes 64. In addition to
ance of HBV-​specific CD8+ T cells is also significantly HBeAg, HBsAg — which is abundantly produced by
delayed in relation to those time points, it seems — as either cccDNA or integrated HBV forms — could serve
mentioned earlier — that the late priming of CD4+ T cell as a high-​dose tolerogen during CHB. The fact that
responses fails to support the development of quanti- envelope-​specific T cell responses are usually very dif-
tively and qualitatively effective CD8+ T cell responses, ficult to detect in patients with CHB — as compared,
thus favouring viral persistence21. for instance, with those specific for the core or the poly-
Whatever the relative contribution of central or merase proteins65,66 — fits with this hypothesis. As stated
peripheral tolerance to viral persistence, it is noteworthy above, it is generally acknowledged that the relatively
that viral factors such as the HBV precore protein high levels of circulating HBsAg that characterize most
(HBeAg) — a viral protein that has potential to cross the CHB infections negatively influence both virus-​specific
placenta and is not required for viral assembly, replica- B and T cell responses and that the clearance of serum
tion or infection10 — have long been suggested to pro- HBsAg might restore dysfunctional responses. However,
mote these tolerogenic events. Indeed, mouse models of recent data obtained in transgenic mice and from
HBV infection have demonstrated that HBeAg can act patients challenge this assumption67,68. In mice, for
as a tolerogenic protein for HBcAg and HBeAg-​specific instance, serum HBsAg loss does not alter naive CD8+

HBV-specific
naive CD8+ T cell

Kupffer cell priming Hepatocellular priming Hepatocellular priming + IL-2

IL-2

CD8 Kupffer cell


TCR
MHC class I

• Bona fide effector cells • Genes of tissue remodelling • Rescuing of effector genes
• Parenchymal clusters • Periportal clusters • Parenchymal clusters
• Effector functions • Dysfunctional phenotype • Effector functions

Fig. 3 | Spatiotemporal dynamics and genomic landscape of CD8+ T cells undergoing intrahepatic priming. Priming
of naive CD8+ T cells by Kupffer cells — which are not natural targets of hepatitis B virus (HBV) — in mouse models leads to
the generation of bona fide CD8+ effector T cells that accumulate in clusters scattered throughout the liver parenchyma
(left). Priming of naive CD8+ T cells by hepatocytes induces their local activation and proliferation but fails to induce the
differentiation into effector cells. The resulting dysfunctional cells show little transcriptional overlap with exhausted
T cells, upregulate genes associated with wound healing/tissue remodelling and accumulate in clusters around portal
tracts (middle). The CD8+ T cell dysfunction induced by hepatocellular priming can be overcome by in vivo IL-2 administration,
which appears to rescue the expression of effector genes48 (right).

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

T cell fate upon intrahepatic priming and does not alter Effector phase during self-​limited AHB
their response to an IL-2-​based immunotherapy67. This Regardless of where priming occurs, effector CD8 +
suggests that enhanced virus-​specific T cell responses in T cells, which could facilitate viral clearance, need to
patients with CHB might actually cause (rather than be a traffic within the liver and recognize hepatocellular
consequence of) HBsAg clearance by killing or ‘curing’ antigens in order to exert their antiviral functions85–88.
HBsAg-​producing hepatocytes. Monoclonal antibody Recent studies in transgenic mice revealed that effec-
therapy targeting HBsAg is currently being evaluated in tor CD8+ T cells arrest in liver sinusoids, rather than in
patients with CHB69–72. However, these results suggest in postcapillary venules. This process does not involve
that antibody therapy alone, although it might reduce classical leukocyte adhesion molecules (such as selec-
the cccDNA pool possibly maintained by the infection tins, chemokines and integrins) or antigen recognition89;
of new hepatocytes7, is unlikely to improve HBV-​specific rather, CD8+ T cells bind to small platelet aggregates that
CD8+ T cell responses. form when platelet-​expressed CD44 interacts with sinu-
In addition to specific viral proteins, viral mutations soidal hyaluronic acid89. Upon this initial arrest, effector
may also contribute to T cell impairment. Although CD8+ T cells crawl at 10 μm min–1 (ref.89) (~1,000-​fold
definitive evidence that HBV can mutate, and thereby slower than the sinusoidal blood flow90) and recognize
escape pathogenic T cell responses, is yet to be pro- hepatocellular antigens while still intravascular89. This
duced, mutations in epitopes that ultimately antagonize occurs via cytoplasmic protrusions that extend through
T cell recognition or induce anergy in T cells have been sinusoidal endothelial fenestration89 (Fig. 4). It is only after
described in patients with CHB73–75. Moreover, several deploying effector functions (including the production
other non-​viral physiologic or disease-​driven tolero- of IFNγ or the killing of HBV-​expressing hepatocytes)
genic mechanisms that affect not only the priming but that effector CD8+ T cells extravasate89, a process that
also the expansion and effector function of HBV-​specific might have evolved to limit excessive immunopathology
T cells have been proposed. These include the produc- (note that hepatocytes express MHC class I mostly on
tion of high levels of soluble mediators (such as argin- the membrane side that faces the sinusoidal lumen)91.
ase, indoleamine 2,3-​d ioxygenase and suppressive As mentioned above, hepatocellular recognition by
cytokines), the upregulation of inhibitory checkpoint HBV-​specific effector CD8+ T cells eventually leads to
receptor/ligand pairs and the expansion of regulatory/ antiviral cytokine production and hepatocyte killing92–94.
suppressive cells and natural killer cells53,76,77. Indeed, The latter is an intrinsically inefficient process, demand-
multiple primary and secondary immune mechanisms ing physical contact between the infected hepatocyte
of HBV hypo-​responsiveness are likely to simultaneously and the T cell. It could therefore be difficult for effector
operate in the liver and/or SLOs of individuals infected CD8+ T cells to reach and kill each infected hepatocyte,
neonatally and perinatally (and, possibly, the liver and/or particularly if one considers that all 1011 hepatocytes
SLOs of the few individuals that acquire CHB in adult- are usually infected during self-​limited AHB in chim-
hood). These considerations call into question whether panzees and relatively few virus-​specific effector CD8+
simple immunotherapeutic approaches — tackling, for T cells are present in the blood and liver of these ani-
instance, one tolerogenic mechanism at a time, as cur- mals. Further, should the rapid clearance of HBV merely
rently pursued in some current clinical studies — will require the killing of most hepatocytes, one would expect
have curative potential for CHB. these acutely infected chimpanzees to show a much
In contrast to T cells, very little is known about B cell higher degree of liver disease severity than is actually
priming in the context of neonatal and perinatal infec- observed26. Along with these considerations, a large body
tions. In stark contrast to what is observed in individ- of experimental evidence in HBV replication-​competent
uals with resolved AHB, most patients who eventually transgenic mice demonstrated that the inhibition
develop CHB do not show detectable levels of anti-​HBs of viral replication by effector CD8+ T cells is mainly
antibodies43. The reason for this might be the sequestra- dependent on non-​cytolytic mechanisms that involve
tion of antibodies by large amounts of soluble HBsAg (we the local production of IFNγ and TNF by these cells26,45.
still lack a widespread clinical assay for the detection of As already mentioned, the initial phase of viral clearance
HBsAg–HBsAb complexes), the lack of HBsAg-​specific in chimpanzees kinetically coincides with the intra­
B cells or their functional impairment43. Recent work hepatic appearance of IFNγ and TNF26. It is of note that
showed that, when compared with HBV-​vaccinated con- these cytokines, possibly through their ability to induce
trols, patients with CHB with persistently high HBsAg nitric oxide in the liver95, appear to prevent the assem-
SSB/La-​dependent
levels have similar frequencies of HBsAg-​specific B cells bly of replication-​competent HBV RNA-​containing
mechanism but show impaired production of anti-​HBs antibodies78–81. capsids in the hepatocyte96 in a proteasome-​dependent
T cell-​induced cytokines HBsAg-​specific B cells in patients with CHB were found manner97. During this process, the HBV nucleocapsids
such as IFNγ and TNF have to be enriched in cells with an atypical memory pheno- vanish from the cytoplasm of the hepatocytes25 and viral
been shown to induce
type — to the detriment of conventional, classic memory RNAs are destabilized in the hepatocellular nucleus by
the post-​transcriptional
downregulation of hepatitis B B cells78,79. When compared with HBsAg-​specific B cells, an SSB/La-​dependent mechanism30–32. IFNγ and TNF also
virus (HBV) RNAs in vivo. This HBcAg-​specific B cells are present at higher frequen- appear to affect the stability of cccDNA in the nucleus
process appears to rely on the cies in patients with CHB, and these can mature into of non-​dividing hepatocytes and, again, this occurs dur-
degradation of the full-​length anti-​HBc-​producing cells in vitro82,83. This difference ing the initial phase of viral clearance in acutely infected
SSB/La protein, which normally
functions as a HBV RNA
might be due to the lower quantity of HBcAg available chimpanzees26. More recent in vitro experiments with
stabilizer in the nucleus of the (compared with HBsAg) or on the ability of HBcAg primary and immortalized human hepatocytes that had
hepatocyte. to activate B cells in a T cell-​independent fashion84. previously been infected with HBV have confirmed

www.nature.com/nri

0123456789();:
Reviews

Effector
CD8+ T cell
Platelets
Docking on
adherent platelets Antigen recognition
and killing
Crawling

Hyaluronan
LSEC

Apoptosis

Hepatocyte HBV

Fig. 4 | Immune surveillance of the liver by effector CD8+ T cells. Effector CD8+ T cells circulating through the mouse
liver initially arrest within sinusoids, not postcapillary venules. They do so independently of antigen recognition and of
numerous molecules variably implicated in leukocyte trafficking to other organs, such as selectins, Gαi-​coupled receptors,
β2 and α4 integrins, platelet endothelial cell adhesion molecule 1 (PECAM1) and vascular adhesion protein 1 (VAP1).
Rather, the predominant mechanism for arrest of circulating effector CD8+ T cells within liver sinusoids is by docking onto
platelets that have previously adhered to hyaluronan on liver sinusoidal endothelial cells (LSECs) via the surface receptor
CD44. After the initial platelet-​dependent arrest, effector CD8+ T cells actively crawl along liver sinusoids. Binding to
hepatitis B virus (HBV) antigens presented by infected hepatocytes leads to antiviral cytokine production, and hepatocyte
killing occurs in a diapedesis-​independent manner. These processes are mediated via the extension of cellular protrusions
through sinusoidal endothelial fenestrae by effector CD8+ T cells, producing contact sites with the sub-​sinusoidal
hepatocellular membrane.

these findings98. Indeed, in these systems, IFNγ and It is unknown how the few HBV-​replicating hepatocytes
TNF were shown to non-​cytopathically destabilize the manage to avoid T cell recognition and persist over time.
nuclear pool of cccDNA via the activation of nuclear A potential explanation may be that the virus or the
APOBEC3 deaminases98. Together, these results suggest process of viral integration induces the downregulation
that under specific circumstances (such as when exposed of molecules that are necessary to process and present
to the local production of antiviral cytokines by activated viral antigens.
effector CD8+ T cells) this form of HBV DNA, which is HBV-​ s pecific CD8 + T cells may promote liver
generally acknowledged to have a long half-​life, might be immunopathology, either by directly killing infected
eliminated from the liver rather quickly and even in the hepatocytes or by indirectly recruiting pathogenic
absence of hepatocellular division. In acutely infected inflammatory cells into the liver. Studies in mouse mod-
chimpanzees, this phase of non-​cytopathic clearance els of HBV pathogenesis show that CD8+ T cells first kill
of HBV replicative intermediates and cccDNA usually HBV-​expressing hepatocytes by rapidly inducing hepato­
precedes a phase of disease-​associated cccDNA loss26, cellular apoptosis via both the perforin-​dependent and
where both the processes of hepatocellular death and Fas-​dependent pathways101,102. Effector CD8+ T cells
compensatory hepatocellular division could contrib- initially trigger the apoptotic death of a relatively
ute to further cccDNA elimination10. According to small number of hepatocytes101,102 that are then rapidly
this scenario, the liver disease that HBV-​specific CD8+ removed by Kupffer cells, which helps to contain liver
T cells eventually trigger may be viewed as a necessary inflammation103. As time progresses, however, apop-
step to complete the elimination of HBV replicative totic hepatocytes that are not promptly removed by
intermediates and cccDNA from most hepatocytes. As Kupffer cells become secondarily necrotic and release
stated above, clearance of HBV from the liver follow- damage-​associated molecular pattern molecules that
ing AHB is rarely complete, and very small numbers actively recruit neutrophils into necro-​inflammatory
of HBV-​replicating hepatocytes are thought to persist foci103,104. Within these initial necro-​inflammatory foci,
indefinitely in the organ8,99. Along with neutralizing anti- which are scattered throughout the liver parenchyma,
bodies that are likely to prevent the re-​emergence of pro- the neutrophils become activated and release a number
ductive infections from these reservoirs, it is tempting to of inflammatory mediators as well as matrix metallo-
speculate that the long-​term presence of traces of viral proteinases (MMPs)105,106. MMPs can efficiently degrade
antigens may actually help to maintain T cell responses. matrix components (including collagen, laminin and
On this account, recent work showed that a population fibronectin) that are deposited de novo (possibly by acti-
of tissue-​resident memory CD8+ T cells survives — vated hepatic stellate cells) during the early process of
possibly due to cell-​autonomous IL-2 production — in liver repair106. In turn, these neutrophil-​derived MMPs —
the liver of individuals with resolved AHB infection (and in conjunction with chemokines that are locally pro-
in the liver of patients with CHB with low viral loads)100. duced by parenchymal and non-​parenchymal liver cells

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

in response to T cell-​derived cytokines107 — promote anergized in the periphery end up attacking liver cells
the arrival of numerous mononuclear cells (including in the attempt to eliminate HBV from the liver. Small
HBV-​non-​specific B and T cells, as well as natural killer numbers of HBV-​specific CD8+ T cells that are incapable
cells, natural killer T cells, dendritic cells and mono- of producing antiviral cytokines may therefore sustain
cytes)105,106. In the end, the intrahepatic recruitment of long-​term immunopathological responses without ever
these inflammatory cells, which ultimately exceed the achieving viral clearance. The extent to which some of
number of HBV-​specific effector CD8+ T cells within these responses — in particular, those of T cells specific
the lesions by more than an order of magnitude, signifi- for HBsAg — are affected by HBV integration into the
cantly exacerbate the severity of liver disease107. Of note, hepatocyte genome and the resulting antigen presenta-
work in patients with HBV indicates that the degree of tion by hepatocytes that are either replicating HBV or
liver damage correlates better with the extent of hepatic not is still poorly understood. Notably, a recent study
infiltration of inflammatory cells than with the frequency utilized antibodies specific for HLA/HBsAg peptide
of intrahepatic HBV-​specific CD8+ cells108. It should be complexes to probe liver biopsies and various cell cul-
also noted that we know very little about the pathogenic ture systems for antigen presentation115. The findings
mechanisms whereby the abundantly recruited inflam- indicate that envelope epitopes that can be recognized
matory cells induce organ damage. It is conceivable that by CD8+ T cells are present, albeit not uniformly dis-
the local production of pro-​inflammatory and cytotoxic tributed, in the liver of patients with CHB and that
mediators and/or the formation of thromboembolic HBsAg derived from either cccDNA or HBV integrated
reactions in the liver microcirculation underlie this into the genome can be properly processed and pre-
damage, and this should be addressed experimentally. sented to CD8+ T cells115. Accordingly, mouse models
Overall, preclinical data105–107 suggest that up to 50% of of immune-​mediated pathology of CHB showed that
the sALT values that are associated with immune-​active the persistent recognition of HBsAg produced by an
phases of HBV infection could reflect the capacity of integrated transgene renders envelope-​specific CD8+
inflammatory cells to kill both infected and uninfected T cells fully capable to sustain prolonged, albeit mod-
hepatocytes. erate, liver cell injury18,113,116. Although suggesting that
HBV integration may indeed have a significant impact
Effector phase during CHB on the immunopathology of CHB, the latter experiments
Similar to AHB, the basic principle of HBV-​specific also revealed that moderately severe but persistent CD8+
CD8+ T cells initially eliciting pathogenic events, with T cell-​dependent liver disease underlies the continuous
secondary involvement of inflammatory cells, also episodes of hepatocellular necrosis and hepatocellular
holds true for CHB. The fact that, when detectable, regeneration that, ultimately, lead to the complications
HBV-​specific CD8+ T cells are only present at very low of CHB: liver fibrosis, liver cirrhosis and HCC18,113,116.
frequencies in the blood and liver of patients with CHB, It should be noted that these complications in humans
and that these peripheral frequencies are usually much usually take place after decades of low-​level liver disease
lower than those of patients with AHB65,66,109–112, helps to with associated hepatocellular regeneration (that is, cel-
explain why individuals with CHB tend to develop much lular DNA synthesis) and inflammation (that is, the pro-
less severe peaks of liver disease11,12. The probability of duction of mutagens)1,117. Based on this preclinical and
detecting peripheral HBV-​specific CD8+ T cells is higher clinical evidence, it has therefore been postulated that
in patients with CHB with low viraemia, and, as men- the spatiotemporal coexistence of hepatocellular necro-
tioned earlier, varies with the specificity of the T cells, in sis, hepatocellular regeneration and liver inflammation
that core-​specific and polymerase-​specific CD8+ T cells may trigger abnormal repair functions and random
are usually more easily detectable than envelope-​specific genetic damage, ultimately leading to liver fibrosis, liver
CD8+ T cells65,66,109–112. Whether these findings reflect dif- cirrhosis and HCC1,117. As the interval between HBV
ferences in the intrahepatic load of the viral antigens — exposure and the complications of persistent infection
with high loads promoting the deletion and/or liver is typically several decades, HBV is thought to be nei-
sequestration of cognate CD8+ T cells — is unknown. ther directly nor acutely oncogenic. Nonetheless, viral
Additional ex vivo and in vitro analyses of peripheral factors, including the deregulation of procarcinogenic
T cells from patients with CHB indicate that core-​specific, genes due to HBV integration into the genome and/or
polymerase-​specific or envelope-​specific CD8+ T cells the expression of HBV-​derived procarcinogenic poly­
are also differently impaired in their capacity to peptides, have been proposed to promote hepatocarcino­
proliferate or kill target cells, although their capacity genesis during CHB, and we refer to other reviews for
to produce antiviral cytokines (for example, IFNγ and the in-​depth discussion of this topic118–120.
sALT values TNF) appears to be uniformly compromised66,109,111,112. As mentioned above, a relevant aspect of the immune-
The serum concentrations of Similarly, mouse models of HBV infection revealed that mediated effector phase during CHB is the development
the liver enzyme alanine
sustained antigen stimulation in vivo, as occurs when of liver fibrosis and cirrhosis121. Of note, these pathol-
aminotransferase. Commonly
measured clinically as a HBV is not rapidly cleared from the liver, creates a set- ogies are characterized by an imbalance between
biomarker for liver damage. ting in which HBV-​specific CD8+ T cells lose the capa­ fibrogenesis and fibrolysis, resulting in an intrahepatic
city to secrete IFNγ and TNF but keep some cytotoxic deposition of extracellular matrix that is qualitatively
Space of Disse activity18,113,114. different in its organization and composition from that
(Also referred to as
perisinusoidal space). The
According to the data discussed above, CHB could be of normal liver repair122. Extracellular matrix depo-
space that lies between the viewed as a disease in which HBV-​specific CD8+ T cells sition in the space of Disse and reduced liver porosity
hepatocytes and the sinusoids. that escape central tolerance and are not completely (that is, the decrease in the number and size of sinusoidal

www.nature.com/nri

0123456789();:
Reviews

endothelial cell fenestrae) are pathological hallmarks of intrasinusoidal small aggregates that function as dock-
liver fibrosis and cirrhosis123. These events (referred to ing sites for circulating virus-​specific CD8+ T cells89.
as sinusoidal ‘capillarization’ and ‘defenestration’, respec- Of note, the antitumour effect of anti-​platelet therapy
tively) not only alter the normal exchange of soluble fac- could have also resulted from additional inhibition of
tors between blood and hepatocytes123 but also appear to platelet-​derived oncogenic products such as serotonin
negatively impact T cell function in the liver89. Indeed, (stimulating hepatocellular proliferation)126 and numer-
recent experiments in mouse models of HBV infection ous growth factors (including epidermal growth factor,
demonstrated that both liver capillarization and/or insulin-​like growth factor, platelet-​derived growth factor
defenestration inhibit hepatocellular antigen recogni- and vascular endothelial growth factor). However, the
tion by effector CD8+ T cells89. This suggests that the experimental evidence that platelet inhibition did not
processes of liver fibrosis and cirrhosis might reduce prevent chemical hepatocarcinogenesis18 suggests that
CD8+ T cell-​mediated immune surveillance towards anti-​platelet therapy inhibits HCC development during
infected or transformed hepatocytes. In addition to lim- CHB mainly by limiting the intrahepatic accumulation
iting the potential of virus-​specific or tumour-​specific of HBV-​specific T cells.
CD8+ T cells for eliminating hepatocytes that are either It is of note that, following these preclinical studies,
HBV-​infected or transformed, these anatomical and a large number of meta-​analyses similarly reported that
functional perturbations of the liver microenvironment patients with CHB who undergo low-​dose aspirin treat-
need to be considered when selecting patients with ment have a reduced incidence of HCC and reduced
CHB for potential future immunotherapeutic inter- mortality, with no significant side effects17,127–134. These
ventions. In fact, it is plausible that virus-​specific or findings should motivate a constructive discussion by
tumour-​specific CD8+ T cells — either activated with the international scientific community on whether the
therapeutic vaccines or other immune stimulators, or preclinical and clinical results collected thus far repre-
exogenously infused (as in the case of CAR T cells sent sufficient background evidence to warrant the use
or TCR-​redirected T cells) — may not be fully functional of low-​dose aspirin in patients with CHB or whether
in fibrotic/cirrhotic livers. they should prompt prospective randomized clinical
trials with careful patient selection and monitoring135.
Targeting immunopathology
Morbidity and mortality in patients with CHB increase Concluding remarks
as liver fibrosis, cirrhosis and, especially, HCC develop4. Even though much has been learned about the molecular
Hence, therapeutic approaches aimed at delaying or biology of HBV and the disease correlates of AHB and
preventing the onset of these complications may be CHB, several immunologic and pathogenic aspects that
beneficial. One option would be to limit T cell-​induced potentially influence the outcome of infection remain
immunopathology without inducing generalized poorly understood. Novel DAAs are being continuously
immune suppression. The discovery that platelets sup- developed for the treatment of CHB, and these include mol-
port the homing of pathogenic effector CD8+ T cells into ecules that target the biogenesis and persistence of cccDNA
the liver124, and that this process is hindered by specific in the hepatocyte136. Whether these DAAs, alone or in com-
platelet inhibitors including aspirin19, prompted the bination with existing or forthcoming antivirals, will be able
preclinical evaluation of platelet inhibitors in CHB. In to achieve a long‐lasting functional cure is unclear. It is
mouse models of immune-​mediated CHB, treatment also unclear whether strategies aimed at boosting immune
with aspirin and/or other anti-​platelet drugs reduced the responses to HBV or dampening immunopathology will
accumulation of virus-​specific T cells within the liver, have a positive impact in this scenario. Undoubtedly, the
as well as liver damage18. As a consequence, fibrosis and development of curative strategies for CHB will greatly
compensatory hepatocellular proliferation were reduced, benefit from a deeper understanding of the mechanisms
HCC development was prevented and overall survival that govern HBV immunobiology and pathogenesis, which
improved18. Mechanistically, low-​dose aspirin — which may guide the design of novel, evidence-​based clinical stud-
inhibits the enzyme COX1 and has almost exclusively ies aimed at terminating persistent HBV infection and its
platelet-​specific effects, with little or no interference life-​threatening complications.
with COX2 expression and/or activity125 — specif-
ically hampers the capacity of platelets to form the Published online xx xx xxxx

1. Guidotti, L. G. & Chisari, F. V. Immunobiology and 6. Levrero, M., Testoni, B. & Zoulim, F. HBV cure: 10. Seeger, C. & Mason, W. S. Molecular biology of
pathogenesis of viral hepatitis. Annu. Rev. Pathol. why, how, when? Curr. Opin. Virol. 18, 135–143 hepatitis B virus infection. Virology 479, 672–686
Mech. Dis. 1, 23–61 (2006). (2016). (2015).
2. Locarnini, S., Hatzakis, A., Chen, D.-S. & Lok, A. 7. Fanning, G. C., Zoulim, F., Hou, J. & Bertoletti, A. 11. Bertoletti, A. & Ferrari, C. Adaptive immunity in HBV
Strategies to control hepatitis B: public policy, Therapeutic strategies for hepatitis B virus infection: infection. J. Hepatol. 64, S71–S83 (2016).
epidemiology, vaccine and drugs. J. Hepatol. 62, towards a cure. Nat. Rev. Drug Discov. 18, 827–844 12. Guidotti, L. G., Isogawa, M. & Chisari, F. V. Host–virus
S76–S86 (2015). (2019). interactions in hepatitis B virus infection. Curr. Opin.
3. Yuen, M.-F. et al. Hepatitis B virus infection. Nat. Rev. 8. Rehermann, B., Ferrari, C., Pasquinelli, C. Immunol. 36, 61–66 (2015).
Dis. Primers 4, 18035 (2018). & Chisari, F. V. The hepatitis B virus persists for 13. Tu, T. et al. Integration occurs early in the viral life
4. Revill, P. A. et al. A global scientific strategy to cure decades after patients’ recovery from acute viral cycle in an in vitro infection model via sodium
hepatitis B. Lancet Gastroenterol. Hepatol. 4, hepatitis despite active maintenance of a cytotoxic taurocholate cotransporting polypeptide-​dependent
545–558 (2019). T-lymphocyte response. Nat. Med. 2, 1104–1108 uptake of enveloped virus particles. J. Virol. 92,
5. Udompap, P. & Kim, W. R. Development of (1996). e02007–e02017 (2018).
hepatocellular carcinoma in patients with suppressed 9. Kim, C. Y. & Tilles, J. G. Purification and biophysical This study shows that HBV DNA integration
viral replication: changes in risk over time. Clin. Liver characterization of hepatitis B antigen. J. Clin. Invest. occurs early upon infection in an in vitro
Dis. 15, 85–90 (2020). 52, 1176–1186 (1973). infection model.

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

14. Summers, J. et al. Hepatocyte turnover during cells inhibit hepatitis B virus replication in the liver of the core antigen. Proc. Natl Acad. Sci. USA 101,
resolution of a transient hepadnaviral infection. Proc. transgenic mice. J. Immunol. 169, 5188–5195 (2002). 14913–14918 (2004).
Natl Acad. Sci. USA 100, 11652–11659 (2003). 36. Vilarinho, S., Ogasawara, K., Nishimura, S., Lanier, L. L. 61. Chen, M. et al. Immune tolerance split between
15. Yang, W. & Summers, J. Integration of hepadnavirus & Baron, J. L. Blockade of NKG2D on NKT cells hepatitis B virus precore and core proteins. J. Virol.
DNA in infected liver: evidence for a linear precursor. prevents hepatitis and the acute immune response 79, 3016–3027 (2005).
J. Virol. 73, 9710–9717 (1999). to hepatitis B virus. Proc. Natl Acad. Sci. USA 104, 62. Tian, Y., Kuo, C., Akbari, O. & Ou, J. J. Maternal-​
16. Wooddell, C. I. et al. RNAi-​based treatment of 18187–18192 (2007). derived hepatitis B virus e antigen alters macrophage
chronically infected patients and chimpanzees reveals 37. Isogawa, M., Robek, M. D., Furuichi, Y. & Chisari, F. V. function in offspring to drive viral persistence after
that integrated hepatitis B virus DNA is a source Toll-​like receptor signaling inhibits hepatitis B virus vertical transmission. Immunity 44, 1204–1214
of HBsAg. Sci. Transl Med. 9, eaan0241 (2017). replication in vivo. J. Virol. 79, 7269–7272 (2005). (2016).
This paper reveals integrated HBV DNA as a relevant 38. Suslov, A., Wieland, S. & Menne, S. Modulators of 63. Publicover, J. et al. Age-​dependent hepatic lymphoid
source of HBsAg in patients and chimpanzees with innate immunity as novel therapeutics for treatment organization directs successful immunity to hepatitis B.
chronic infection. of chronic hepatitis B. Curr. Opin. Virol. 30, 9–17 J. Clin. Invest. 123, 3728–3739 (2013).
17. Simon, T. G. et al. Association of aspirin with hepato­ (2018). 64. Brunetto, M. R. et al. Wild-​type and e antigen-​minus
cellular carcinoma and liver-​related mortality. N. Engl. 39. Iwasaki, A. A virological view of innate immune hepatitis B viruses and course of chronic hepatitis.
J. Med. 382, 1018–1028 (2020). recognition. Annu. Rev. Microbiol. 66, 177–196 Proc. Natl Acad. Sci. USA 88, 4186–4190 (1991).
This manuscript represents one of a large number (2012). 65. Rivino, L. et al. Hepatitis B virus-​specific T cells
of meta-​analyses describing an association between 40. Webster, G. J. M. et al. Incubation phase of acute associate with viral control upon nucleos(t)ide-​
low-​dose aspirin treatment and reduced HCC hepatitis B in man: dynamic of cellular immune analogue therapy discontinuation. J. Clin. Invest. 128,
incidence. mechanisms. Hepatology 32, 1117–1124 (2000). 668–681 (2018).
18. Sitia, G. et al. Antiplatelet therapy prevents 41. Thimme, R. et al. CD8+ T cells mediate viral clearance 66. Schuch, A. et al. Phenotypic and functional differences
hepatocellular carcinoma and improves survival in a and disease pathogenesis during acute hepatitis B of HBV core-​specific versus HBV polymerase-​specific
mouse model of chronic hepatitis B. Proc. Natl Acad. virus infection. J. Virol. 77, 68–76 (2003). CD8+ T cells in chronically HBV-​infected patients with
Sci. USA 109, E2165–E2172 (2012). 42. Hoofnagle, J. H., Gerety, R. J. & Barker, L. F. low viral load. Gut 68, 905–915 (2019).
This preclinical study shows that anti-​platelet Antibody to hepatitis-​B-virus core in man. Lancet 302, 67. Fumagalli, V. et al. Serum HBsAg clearance has minimal
therapy reduces liver fibrosis and prevents HCC 869–873 (1973). impact on CD8+ T cell responses in mouse models of
in mouse models of CHB. 43. Maini, M. K. & Burton, A. R. Restoring, releasing or HBV infection. J. Exp. Med. 217, e20200298 (2020).
19. Iannacone, M., Sitia, G., Narvaiza, I., Ruggeri, Z. M. replacing adaptive immunity in chronic hepatitis B. This study shows that circulating HBsAg clearance
& Guidotti, L. G. Antiplatelet drug therapy moderates Nat. Rev. Gastroenterol. Hepatol. 16, 662–675 does not improve HBV-​specific CD8+ T cell responses.
immune-​mediated liver disease and inhibits viral (2019). 68. Bert, N. L. et al. Effects of hepatitis B surface antigen
clearance in mice infected with a replication-​deficient 44. Guidotti, L. G. et al. Cytotoxic T lymphocytes inhibit on virus-​specific and global T cells in patients with
adenovirus. Clin. Vaccine Immunol. 14, 1532–1535 hepatitis B virus gene expression by a noncytolytic chronic hepatitis B virus infection. Gastroenterology
(2007). mechanism in transgenic mice. Proc. Natl Acad. Sci. 159, 652–664 (2020).
20. Jilbert, A. R., Miller, D. S., Scougall, C. A., Turnbull, H. USA 91, 3764–3768 (1994). 69. Li et al. A potent human neutralizing antibody
& Burrell, C. J. Kinetics of duck hepatitis B virus 45. Guidotti, L. G. et al. Intracellular inactivation of the Fc-dependently reduces established HBV infections.
infection following low dose virus inoculation: one virus hepatitis B virus by cytotoxic T lymphocytes. Immunity eLife 6, e26738 (2017).
DNA genome is infectious in neonatal ducks. Virology 4, 25–36 (1996). 70. Zhang, T.-Y. et al. Prolonged suppression of HBV in
226, 338–345 (1996). 46. Wong, Y. C., Tay, S. S., McCaughan, G. W., Bowen, D. G. mice by a novel antibody that targets a unique epitope
21. Asabe, S. et al. The size of the viral inoculum & Bertolino, P. Immune outcomes in the liver: is CD8 on hepatitis B surface antigen. Gut 65, 658 (2015).
contributes to the outcome of hepatitis B virus T cell fate determined by the environment? J. Hepatol. 71. Neumann et al. Novel mechanism of antibodies to
infection. J. Virol. 83, 9652–9662 (2009). 63, 1005–1014 (2015). hepatitis B virus in blocking viral particle release from
22. Wisse, E., Jacobs, F., Topal, B., Frederik, P. 47. Isogawa et al. CD40 activation rescues antiviral CD8+ cells. Hepatology 52, 875–885 (2010).
& Geest, B. D. The size of endothelial fenestrae in T cells from PD-1-mediated exhaustion. PLoS Pathog. 72. Galun, E. et al. Clinical evaluation (phase I) of a
human liver sinusoids: implications for hepatocyte-​ 9, e1003490 (2013). combination of two human monoclonal antibodies to
directed gene transfer. Gene Ther. 15, 1193–1199 48. Bénéchet, A. P. et al. Dynamics and genomic HBV: safety and antiviral properties. Hepatology 35,
(2008). landscape of CD8+ T cells undergoing hepatic priming. 673–679 (2002).
23. Vollmar, B. & Menger, M. D. The hepatic Nature 574, 200–205 (2019). 73. Bertoletti, A. et al. Cytotoxic T lymphocyte response
microcirculation: mechanistic contributions and This paper reveals that hepatocellular priming to a wild type hepatitis B virus epitope in patients
therapeutic targets in liver injury and repair. Physiol. leads to a T cell dysfunction that is refractory to chronically infected by variant viruses carrying
Rev. 89, 1269–1339 (2009). checkpoint inhibition but responds to IL-2. substitutions within the epitope. J. Exp. Med. 180,
24. Whalley, S. A. et al. Kinetics of acute hepatitis B virus 49. Bertolino, P. et al. Death by neglect as a deletional 933–943 (1994).
infection in humans. J. Exp. Med. 193, 847–854 mechanism of peripheral tolerance. Int. Immunol. 11, 74. Bertoletti, A. et al. Natural variants of cytotoxic
(2001). 1225–1238 (1999). epitopes are T-​cell receptor antagonists for antiviral
25. Guidotti, L. G., Matzke, B., Schaller, H. & Chisari, F. V. 50. Pol et al. Effects of interleukin-2 in immunostimulation cytotoxic T cells. Nature 369, 407–410 (1994).
High-​level hepatitis B virus replication in transgenic and immunosuppression. J. Exp. Med. 217, 2261 75. Maini, M. K. et al. T cell receptor usage of virus-​
mice. J. Virol. 69, 6158–6169 (1995). (2020). specific CD8 cells and recognition of viral mutations
26. Guidotti, L. G. et al. Viral clearance without destruction 51. Blattman, J. N. et al. Therapeutic use of IL-2 to during acute and persistent hepatitis B virus infection.
of infected cells during acute HBV infection. Science enhance antiviral T-​cell responses in vivo. Nat. Med. 9, Eur. J. Immunol. 30, 3067–3078 (2000).
284, 825–829 (1999). 540–547 (2003). 76. Bertoletti, A. & Kennedy, P. T. The immune tolerant
27. Wieland, S. F., Spangenberg, H. C., Thimme, R., 52. West, E. E. et al. PD-​L1 blockade synergizes with IL-2 phase of chronic HBV infection: new perspectives on
Purcell, R. H. & Chisari, F. V. Expansion and contraction therapy in reinvigorating exhausted T cells. J. Clin. an old concept. Cell Mol. Immunol. 12, 258–263
of the hepatitis B virus transcriptional template in Invest. 123, 2604–2615 (2013). (2015).
infected chimpanzees. Proc. Natl Acad. Sci. USA 101, 53. Kuipery, A., Gehring, A. J. & Isogawa, M. Mechanisms 77. Fisicaro, P. et al. Pathogenetic mechanisms of T cell
2129–2134 (2004). of HBV immune evasion. Antivir. Res. 179, 104816 dysfunction in chronic HBV infection and related
28. Wieland, S., Thimme, R., Purcell, R. H. & Chisari, F. V. (2020). therapeutic approaches. Front. Immunol. 11, 849
Genomic analysis of the host response to hepatitis B 54. Kennedy, P. T. F. et al. Preserved T-​cell function in (2020).
virus infection. Proc. Natl Acad. Sci. USA 101, children and young adults with immune-​tolerant 78. Burton, A. R. et al. Circulating and intrahepatic
6669–6674 (2004). chronic hepatitis B. Gastroenterology 143, 637–645 antiviral B cells are defective in hepatitis B. J. Clin.
29. Suslov, A. et al. Virus does not interfere with innate (2012). Invest. 128, 4588–4603 (2018).
immune responses in the human liver. Gastroenterology 55. Shimizu, Y., Guidotti, L. G., Fowler, P. & Chisari, F. V. 79. Salimzadeh, L. et al. PD-1 blockade partially recovers
154, 1778–1790 (2018). Dendritic cell immunization breaks cytotoxic dysfunctional virus-​specific B cells in chronic hepatitis
30. Tsui, L. V., Guidotti, L. G., Ishikawa, T. & Chisari, F. V. T lymphocyte tolerance in hepatitis B virus transgenic B infection. J. Clin. Invest. 128, 4573–4587 (2018).
Posttranscriptional clearance of hepatitis B virus mice. J. Immunol. 161, 4520–4529 (1998). Together with Burton et al. (2018), this paper
RNA by cytotoxic T lymphocyte-​activated hepatocytes. 56. Kakimi, K., Isogawa, M., Chung, J., Sette, A. detects and characterizes dysfunctional HBsAg-​
Proc. Natl Acad. Sci. USA 92, 12398–12402 (1995). & Chisari, F. V. Immunogenicity and tolerogenicity of specific B cell responses in patients with chronic
31. Heise, T., Guidotti, L. G., Cavanaugh, V. J. & Chisari, F. V. hepatitis B virus structural and nonstructural proteins: HBV infection.
Hepatitis B virus RNA-​binding proteins associated with implications for immunotherapy of persistent viral 80. Tian, C. et al. Use of ELISpot assay to study
cytokine-​induced clearance of viral RNA from the liver infections. J. Virol. 76, 8609–8620 (2002). HBs-specific B cell responses in vaccinated and
of transgenic mice. J. Virol. 73, 474–481 (1999). 57. Ishak, K. et al. Histological grading and staging of HBV infected humans. Emerg. Microbes Infec 7, 16
32. Heise, T., Guidotti, L. G. & Chisari, F. V. La autoantigen chronic hepatitis. J. Hepatol. 22, 696–699 (1995). (2018).
specifically recognizes a predicted stem-​loop in 58. Fisicaro, P. et al. Targeting mitochondrial dysfunction 81. Xu, X. et al. Reversal of B-​cell hyperactivation and
hepatitis B virus RNA. J. Virol. 73, 5767–5776 (1999). can restore antiviral activity of exhausted HBV-​specific functional impairment is associated with HBsAg
33. McClary, H., Koch, R., Chisari, F. V. & Guidotti, L. G. CD8 T cells in chronic hepatitis B. Nat. Med. 23, seroconversion in chronic hepatitis B patients.
Relative sensitivity of hepatitis B virus and other 327–336 (2017). Cell Mol. Immunol. 12, 309–316 (2015).
hepatotropic viruses to the antiviral effects of cytokines. This article suggests a central role for reactive 82. Bert, N. L. et al. Comparative characterization
J. Virol. 74, 2255–2264 (2000). oxygen species in T cell exhaustion during CHB, of B cells specific for HBV nucleocapsid and envelope
34. Wieland, S. F., Guidotti, L. G. & Chisari, F. V. thus providing novel potential therapeutic targets. proteins in patients with chronic hepatitis B. J. Hepatol.
Intrahepatic induction of α/β interferon eliminates viral 59. Wieland, S. F. The chimpanzee model for hepatitis B 72, 34–44 (2019).
RNA-​containing capsids in hepatitis B virus transgenic virus infection. CSH Perspect. Med. 5, a021469 83. Vanwolleghem, T. et al. Hepatitis B core-​specific
mice. J. Virol. 74, 4165–4173 (2000). (2015). memory B cell responses associate with clinical
35. Kimura, K., Kakimi, K., Wieland, S., Guidotti, L. G. & 60. Chen, M. T. et al. A function of the hepatitis B virus parameters in patients with chronic HBV. J. Hepatol.
Chisari, F. V. Activated intrahepatic antigen-​presenting precore protein is to regulate the immune response to 73, 52–61 (2020).

www.nature.com/nri

0123456789();:
Reviews

84. Milich, D. & McLachlan, A. The nucleocapsid of 108. Maini, M. K. et al. The role of virus-​specific CD8+ 134. Hayashi, T. et al. Antiplatelet therapy improves the
hepatitis B virus is both a T-​cell-independent and a cells in liver damage and viral control during prognosis of patients with hepatocellular carcinoma.
T-cell-dependent antigen. Science 234, 1398–1401 persistent hepatitis B virus infection. J. Exp. Med. Cancers 12, 3215 (2020).
(1986). 191, 1269–1280 (2000). 135. Guidotti, L. G., Vecchia, C. L. & Colombo, M. Is it
85. Guidotti, L. G. & Iannacone, M. Effector CD8 T cell 109. Reignat, S. et al. Escaping high viral load exhaustion time to recommend low-​dose aspirin treatment
trafficking within the liver. Mol. Immunol. 55, 94–99 CD8 cells with altered tetramer binding in chronic for the prevention of hepatocellular carcinoma?
(2013). hepatitis B virus infection. J. Exp. Med. 195, Gastroenterology 159, 1988–1990 (2020).
86. Iannacone, M. Hepatic effector CD8+ T-​cell dynamics. 1089–1101 (2002). 136. Martinez, M. G., Villeret, F., Testoni, B. & Zoulim, F.
Cell Mol. Immunol. 12, 269–272 (2015). 110. Webster, G. J. M. et al. Longitudinal analysis of CD8+ Can we cure hepatitis B virus with novel direct-​acting
87. Inverso, D. & Iannacone, M. Spatiotemporal dynamics T cells specific for structural and nonstructural antivirals? Liver Int. 40, 27–34 (2020).
of effector CD8+ T cell responses within the liver. hepatitis B virus proteins in patients with chronic 137. Hillis, W. D. Viral hepatitis associated with sub-​human
J. Leukoc. Biol. 99, 51–55 (2016). hepatitis B: implications for immunotherapy. J. Virol. primates. Transfusion 3, 445–454 (1963).
88. Benechet, A. P. & Iannacone, M. Determinants of 78, 5707–5719 (2004). 138. Walter, E., Keist, R., Niederöst, B., Pult, I. & Blum, H. E.
hepatic effector CD8+ T cell dynamics. J. Hepatol. 66, 111. Boni, C. et al. Characterization of hepatitis B virus Hepatitis B virus infection of tupaia hepatocytes in vitro
228–233 (2017). (HBV)-specific T-​cell dysfunction in chronic HBV and in vivo. Hepatology 24, 1–5 (1996).
89. Guidotti, L. G. et al. Immunosurveillance of the liver by infection. J. Virol. 81, 4215–4225 (2007). 139. Schulze, A., Gripon, P. & Urban, S. Hepatitis B virus
intravascular effector CD8+ T cells. Cell 161, 486–500 112. Hoogeveen, R. C. et al. Phenotype and function of infection initiates with a large surface protein-​dependent
(2015). HBV-​specific T cells is determined by the targeted binding to heparan sulfate proteoglycans. Hepatology
This manuscript reports that effector CD8+ T cells epitope in addition to the stage of infection. Gut 68, 46, 1759–1768 (2007).
can recognize and kill antigen-​expressing 893–904 (2018). 140. Sureau, C. & Salisse, J. A conformational heparan
hepatocytes without extravasating by extending 113. Nakamoto, Y., Guidotti, L. G., Kuhlen, C. V., Fowler, P. sulfate binding site essential to infectivity overlaps
cytoplasmic protrusions through endothelial & Chisari, F. V. Immune pathogenesis of hepatocellular with the conserved hepatitis B virus A-​determinant.
fenestration. carcinoma. J. Exp. Med. 188, 341–350 (1998). Hepatology 57, 985–994 (2013).
90. Sironi, L. et al. In vivo flow mapping in complex vessel 114. Isogawa, M., Furuichi, Y. & Chisari, F. V. Oscillating 141. Roskams, T. et al. Heparan sulfate proteoglycan
networks by single image correlation. Sci. Rep. 4, CD8+ T cell effector functions after antigen recognition expression in normal human liver. Hepatology 21,
7341 (2014). in the liver. Immunity 23, 53–63 (2005). 950–958 (1995).
91. Warren, A. et al. T lymphocytes interact with 115. Khakpoor, A. et al. Spatiotemporal differences in 142. Yan, H. et al. Sodium taurocholate cotransporting
hepatocytes through fenestrations in murine presentation of CD8 T cell epitopes during hepatitis B polypeptide is a functional receptor for human
liver sinusoidal endothelial cells. Hepatology 44, virus infection. J. Virol. 93, e01457-18 (2018). hepatitis B and D virus. eLife 1, e00049 (2012).
1182–1190 (2006). 116. Nakamoto, Y., Suda, T., Momoi, T. & Kaneko, S. 143. Döring, B., Lütteke, T., Geyer, J. & Petzinger, E. The
92. Guidotti, L. G. The role of cytotoxic T cells and Different procarcinogenic potentials of lymphocyte SLC10 carrier family: transport functions and molecular
cytokines in the control of hepatitis B virus infection. subsets in a transgenic mouse model of chronic structure. Curr. Top. Membr. 70, 105–168 (2012).
Vaccine 20, A80–A82 (2002). hepatitis B. Cancer Res. 64, 3326–3333 (2004). 144. Hu, J. & Liu, K. Complete and incomplete hepatitis B
93. Fioravanti, J. et al. Effector CD8+ T cell-​derived 117. Tang, L. S. Y., Covert, E., Wilson, E. & Kottilil, S. virus particles: formation, function, and application.
interleukin-10 enhances acute liver immunopathology. Chronic hepatitis B infection: a review. JAMA 319, Viruses 9, 56 (2017).
J. Hepatol. 67, 543–548 (2017). 1802–1813 (2018). 145. Seitz, S., Habjanič, J., Schütz, A. K. & Bartenschlager, R.
94. Iannacone, M. & Guidotti, L. G. Mouse models of 118. Buendia, M.-A. & Neuveut, C. Hepatocellular carcinoma. The hepatitis B virus envelope proteins: molecular
hepatitis B virus pathogenesis. CSH Perspect. Med. 5, CSH Perspect. Med. 5, a021444 (2015). gymnastics throughout the viral life cycle. Ann. Rev.
a021477 (2015). 119. Levrero, M. & Zucman-​Rossi, J. Mechanisms of Virol. 7, 1–26 (2020).
95. Guidotti, L. G., McClary, H., Loudis, J. M. & Chisari, F. V. HBV-induced hepatocellular carcinoma. J. Hepatol. 146. Wisse, E., de Zanger, R. B., Charels, K.,
Nitric oxide inhibits hepatitis b virus replication in 64, S84–S101 (2016). Van Der Smissen, P. & McCuskey, R. S. The liver sieve:
the livers of transgenic mice. J. Exp. Med. 191, 120. Bisceglie, A. M. D. Hepatitis B and hepatocellular considerations concerning the structure and function
1247–1252 (2000). carcinoma. Hepatology 49, S56–S60 (2009). of endothelial fenestrae, the sinusoidal wall and the
96. Wieland, S. F., Eustaquio, A., Whitten-​Bauer, C., 121. Schuppan, D. & Afdhal, N. H. Liver cirrhosis. Lancet space of disse. Hepatology 5, 683–692 (1985).
Boyd, B. & Chisari, F. V. Interferon prevents formation 371, 838–851 (2008). 147. Ficht, X. & Iannacone, M. Immune surveillance
of replication-​competent hepatitis B virus RNA-​ 122. Bataller, R. & Brenner, D. A. Liver fibrosis. J. Clin. Invest. of the liver by T cells. Sci. Immunol. 5, eaba2351
containing nucleocapsids. Proc. Natl Acad. Sci. USA 115, 209–218 (2005). (2020).
102, 9913–9917 (2005). 123. Friedman, S. L. Mechanisms of disease: mechanisms 148. Iwakiri, Y. The lymphatic system: a new frontier in
97. Robek, M. D., Wieland, S. F. & Chisari, F. V. Inhibition of hepatic fibrosis and therapeutic implications. hepatology. Hepatology 64, 706–707 (2016).
of hepatitis B virus replication by interferon requires Nat. Clin. Pract. Gastr 1, 98–105 (2004). 149. Jenne, C. N. & Kubes, P. Immune surveillance by the
proteasome activity. J. Virol. 76, 3570–3574 (2002). 124. Iannacone, M. et al. Platelets mediate cytotoxic liver. Nat. Immunol. 14, 996–1006 (2013).
98. Xia, Y. et al. Interferon-​γ and tumor necrosis factor-​α T lymphocyte-​induced liver damage. Nat. Med. 11, 150. Horst, A. K., Neumann, K., Diehl, L. & Tiegs, G.
produced by T cells reduce the HBV persistence form, 1167–1169 (2005). Modulation of liver tolerance by conventional
cccDNA, without cytolysis. Gastroenterology 150, This study establishes platelets as critical mediators and nonconventional antigen-​presenting cells and
194–205 (2016). of liver damage through their capacity to promote regulatory immune cells. Cell Mol. Immunol. 13,
99. Michalak, T. I., Pasquinelli, C., Guilhot, S. & Chisari, F. V. liver homing of effector CD8+ T cells. 277–292 (2016).
Hepatitis B virus persistence after recovery from acute 125. Ornelas, A. et al. Beyond COX-1: the effects of aspirin 151. Wong, Y. C., McCaughan, G. W., Bowen, D. G. &
viral hepatitis. J. Clin. Invest. 93, 230–239 (1994). on platelet biology and potential mechanisms of Bertolino, P. The CD8 T-​cell response during tolerance
100. Pallett, L. J. et al. IL-2high tissue-​resident T cells in chemoprevention. Cancer Metast Rev. 36, 289–303 induction in liver transplantation. Clin. Transl Immunol.
the human liver: sentinels for hepatotropic infection. (2017). 5, e102 (2016).
J. Exp. Med. 214, 1567–1580 (2017). 126. Haemmerle, M., Stone, R. L., Menter, D. G., 152. Mason, W. S. et al. HBV DNA integration and clonal
This paper characterizes tissue-​resident memory Afshar-Kharghan, V. & Sood, A. K. The platelet lifeline hepatocyte expansion in chronic hepatitis B patients
T cells in the liver of patients chronically infected to cancer: challenges and opportunities. Cancer Cell considered immune tolerant. Gastroenterology 151,
by HBV. 33, 965–983 (2018). 986–998.e4 (2016).
101. Ando, K. et al. Class I-​restricted cytotoxic T 127. Lee, P.-C. et al. Antiplatelet therapy is associated with 153. Tu, T., Budzinska, M. A., Shackel, N. A. & Urban, S.
lymphocytes are directly cytopathic for their target a better prognosis for patients with hepatitis B virus-​ HBV DNA integration: molecular mechanisms and
cells in vivo. J. Immunol. 152, 3245–3253 (1994). related hepatocellular carcinoma after liver resection. clinical implications. Viruses 9, 75 (2017).
102. Nakamoto, Y., Guidotti, L. G., Pasquetto, V., Ann. Surg. Oncol. 23, 874–883 (2016). 154. Budzinska, M. A., Shackel, N. A., Urban, S. & Tu, T.
Schreiber, R. D. & Chisari, F. V. Differential target 128. Hwang, I. C., Chang, J., Kim, K. & Park, S. M. Aspirin Cellular genomic sites of hepatitis B virus DNA
cell sensitivity to CTL-​activated death pathways in use and risk of hepatocellular carcinoma in a national integration. Genes 9, 365 (2018).
hepatitis B virus transgenic mice. J. Immunol. 158, cohort study of Korean adults. Sci. Rep. 8, 4968 155. Huang, Z. M. & Yen, T. S. Dysregulated surface gene
5692–5697 (1997). (2018). expression from disrupted hepatitis B virus genomes.
103. Sitia, G. et al. Kupffer cells hasten resolution of liver 129. Simon, T. G. et al. Association between aspirin use and J. Virol. 67, 7032–7040 (1993).
immunopathology in mouse models of viral hepatitis. risk of hepatocellular carcinoma. JAMA Oncol. 4, 156. Dienes, H. P. et al. Hepatic expression patterns
PLoS Pathog. 7, e1002061 (2011). 1683 (2018). of the large and middle hepatitis B virus surface
104. Sitia et al. Treatment with HMGB1 inhibitors diminishes 130. Lee, T.-Y. et al. Association of daily aspirin therapy with proteins in viremic and nonviremic chronic hepatitis B.
CTL-​induced liver disease in HBV transgenic mice. risk of hepatocellular carcinoma in patients with chronic Gastroenterology 98, 1017–1023 (1990).
J. Leukoc. Biol. 81, 100–107 (2007). hepatitis B. JAMA Intern. Med. 179, 633–640 (2019). 157. Chisari, F. V. et al. Molecular pathogenesis of
105. Sitia, G. et al. Depletion of neutrophils blocks the 131. Wang, S. et al. Association of aspirin therapy with hepatocellular carcinoma in hepatitis B virus
recruitment of antigen-​nonspecific cells into the liver risk of hepatocellular carcinoma: a systematic review transgenic mice. Cell 59, 1145–1156 (1989).
without affecting the antiviral activity of hepatitis B and dose–response analysis of cohort studies with 158. Su, I., Wang, H., Wu, H. & Huang, W. Ground glass
virus-​specific cytotoxic T lymphocytes. Proc. Natl 2.5 million participants. Pharmacol. Res. 151, hepatocytes contain pre-​S mutants and represent
Acad. Sci. USA 99, 13717–13722 (2002). 104585 (2019). preneoplastic lesions in chronic hepatitis B virus
106. Sitia, G. et al. MMPs are required for recruitment 132. Liao, Y.-H. et al. Aspirin decreases hepatocellular infection. J. Gastroen Hepatol. 23, 1169–1174 (2008).
of antigen-​nonspecific mononuclear cells into the liver carcinoma risk in hepatitis C virus carriers: a 159. Hadziyannis, S., Gerber, M. A., Vissoulis, C.
by CTLs. J. Clin. Invest. 113, 1158–1167 (2004). nationwide cohort study. BMC Gastroenterol. 20, 6 & Popper, H. Cytoplasmic hepatitis B antigen in
107. Kakimi, K. et al. Blocking chemokine responsive to (2020). “ground-​glass” hepatocytes of carriers. Arch. Pathol.
γ-2/interferon (IFN)-γ inducible protein and monokine 133. Bosetti, C., Santucci, C., Gallus, S., Martinetti, M. 96, 327–330 (1973).
induced by IFN-​γ activity in vivo reduces the & Vecchia, C. L. Aspirin and the risk of colorectal 160. Tu, T. et al. Clonal expansion of hepatocytes with
pathogenetic but not the antiviral potential of and other digestive tract cancers: an updated meta-​ a selective advantage occurs during all stages of
hepatitis B virus-​specific cytotoxic T lymphocytes. analysis through 2019. Ann. Oncol. 31, 558–568 chronic hepatitis B virus infection. J. Viral Hepat. 22,
J. Exp. Med. 194, 1755–1766 (2001). (2020). 737–753 (2015).

NATuRE REVIEWS | Immunology

0123456789();:
Reviews

Acknowledgements of Education, University and Research, and Funded Research on technology related to work discussed in this manuscript
The authors thank M. Silva for secretarial assistance, Agreements from Gilead Sciences, Avalia Therapeutics and ( WO 2 0 2 0 / 01 6 4 3 4 , WO 2 0 2 0 / 01 6 4 2 7 , WO 2 0 2 0 /
F. Andreata for help with figure preparation and the members of CNCCS SCARL. 030781, WO2020/234483, EU patent applications n.
the Iannacone and Guidotti laboratories for helpful discussions. 19211249.8 and n. 20156716.1, and UK patent application n.
They apologize to all authors whose work they could not cite due Author contributions 1907493.9).
to space constraints. M.I. is supported by the European M.I. and L.G.G. contributed equally to this work.
Research Council (ERC) Consolidator Grant 725038, ERC Proof Peer review information
of Concept Grant 957502, Italian Association for Cancer Competing interests Nature Reviews Immunology thanks Anna Lok, Antonio
Research (AIRC) Grants 19891 and 22737, Italian Ministry of M.I. participates in advisory boards/consultancies for Gilead Bertoletti and Mala Maini for their contribution to the peer
Health (MoH) Grants RF-2018-12365801 and COVID-2020- Sciences, Roche, Third Rock Ventures, Amgen, Asher Bio and review of this work.
12371617, Lombardy Foundation for Biomedical Research Allovir. L.G.G is a member of the board of directors at
(FRRB) Grant 2015-0010, the European Molecular Biology Genenta Science and Epsilon Bio and participates in advisory Publisher’s note
Organization Young Investigator Program and a Funded boards/consultancies for Gilead Sciences, Roche and Arbutus Springer Nature remains neutral with regard to jurisdictional
Research Agreement from Gilead Sciences. L.G.G. is supported Biopharma. M.I. and L.G.G. are inventors on patents filed, claims in published maps and institutional affiliations.
by the AIRC Grant 22737, Lombardy Open Innovation Grant owned and managed by San Raffaele Scientific Institute,
229452, PRIN Grant 2017MPCWPY from the Italian Ministry Vita-​Salute San Raffaele University and Telethon Foundation © Springer Nature Limited 2021

www.nature.com/nri

0123456789();:

You might also like