ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 11, pp. 1504-1520 © Pleiades Publishing, Ltd., 2025.
Russian Text © The Author(s), 2025, published in Biokhimiya, 2025, Vol. 90, No. 11, pp. 1601-1620.
1504
REVIEW
In vivo and in vitro Models of Hepatitis B Virus Infection
Yuliya V. Kolyako
1,a
*, Alla S. Zhitkevich
1
, Daria V. Avdoshina
1
, Daria Y. Tanygina
1
,
Vasiliy D. Apolokhov
1
, Tatiana V. Gorodnicheva
1,2
, Dmitry S. Kostyushev
3,4,5
,
Ekaterina O. Bayurova
1
, and Ilya V. Gordeychuk
1,3
1
Chumakov Federal Scientific Center for Research and Development
of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis),
108819 Moscow, Russia
2
Pirogov Russian National Research Medical University, 117997 Moscow, Russia
3
Sechenov First Moscow State Medical University
of the Ministry of Health of the Russian Federation (Sechenov University), 117418 Moscow, Russia
4
Faculty of bioengineering and bioinformatics, Lomonosov Moscow State University, 119192 Moscow, Russia
5
Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
a
e-mail: kolyako_jv@chumakovs.su
Received July 23, 2025
Revised September 24, 2025
Accepted September 25, 2025
AbstractHepatitis B virus (Orthohepadnavirus hominoidei, HBV) is a hepatotropic virus from the Hepad-
naviridae family and the causative agent of both acute and chronic hepatitis B (CHB). The possible outcomes
of CHB include liver cirrhosis and hepatocellular carcinoma (HCC) that pose a significant burden on the
healthcare systems worldwide. In the nuclei of infected hepatocytes of patients with CHB, the HBV genome
persists as a pool of covalently closed circular DNA (cccDNA) molecules. Current therapeutic strategies cannot
directly target cccDNA. Instead, the available treatments focus on long-term suppression of viral replication
and require lifelong administration. Development and evaluation of novel antiviral agents capable of achiev-
ing complete HBV eradication require relevant in vivo and in vitro models of HBV infection. Among the
available animal models, the following categories are distinguished: (i)  animals naturally susceptible to HBV;
(ii)  surrogate models using animal species susceptible to the related hepadnaviruses; (iii)  non-susceptible ani-
mals receiving HBV genome via recombinant viral vectors; (iv)  models utilizing human hepatocyte xenografts.
Among the available in vitro models, primary human and northern treeshrew (Tupaia belangeri) hepato-
cytes fully support the HBV replication cycle, but they rapidly lose susceptibility to the virus in cell culture.
In turn, unmodified human hepatoma cell lines are not susceptible to HBV but can support viral replication
after transfection with the viral genome. This review discusses key characteristics, advantages, limitations,
and areas of application of the currently available invivo and in vitro models of HBV infection.
DOI: 10.1134/S0006297925602230
Keywords: hepatitis B virus, hepatitis B virus infection, chronic hepatitis B, viral infection models, laboratory
animals
* To whom correspondence should be addressed.
INTRODUCTION
According to the WHO estimates, approximately
1.2 million new cases of hepatitis  B virus (HBV) infec-
tion are recorded annually and more than 250 mil-
lion people are living with chronic hepatitis  B (CHB)
(https://www.who.int/news-room/fact-sheets/detail/
hepatitis-b, accessed 10.07.25). The HBV genome is rep-
resented by relaxed circular partially double-stranded
DNA (rcDNA) (~3200 base pairs) that contains four
overlapping open reading frames (ORF): S, pre-C/C,
P and X. ORF S includes pre-S1, pre-S2, and S re-
gions, which encode three isoforms of the surface
protein (HBsAg): small (SHBsAg), middle (MHBsAg)
HEPATITIS B VIRUS INFECTION MODELS 1505
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
and large (LHBsAg), all sharing a common C-termi-
nus but differing in N-terminal extensions and dif-
ferent glycosylation levels. ORF pre-C/C encodes pre-
core protein (HBeAg) and core protein (HBcAg). ORF
P encodes polymerase with reverse transcriptase and
RNase H activities. ORF X encodes a key transcription
activation factor (HBxAg) [1]. HBV hepatotropism is
determined by three factors: (i)  the unique receptor
expressed on hepatocyte surface, (ii)  HBV capsid nu-
clear import, and (iii)  hepatocyte-specific factors for
viral transcription. HBV infection begins with low-af-
finity binding of the virus to heparan sulfates pro-
teoglycans on the cell surface, followed by specific
interaction between the pre-S1 domain of LHBsAg
and its receptor – sodium-taurocholate cotransport-
ing polypeptide (NTCP). Next, the HBV/NTCP complex
is internalized into cells by interacting with the epi-
dermal growth factor receptor (EGFR) via clathrin-
dependent endocytosis. HBV enters the endolysosomal
compartment, where the rcDNA-containing capsid is
released into the cytosol. Import of the viral genome
into the nucleus is ensured by the interaction of the
nuclear localization signal in the terminal region of
HBcAg on the surface of the viral capsid with the
importin α1 [2]. In the nucleus, the HBV genome
is released from a capsid followed by conversion
of rcDNA into covalently closed circular DNA (cccDNA)
in a multistep process mediated by cellular enzymes.
The host RNA polymerase II mediates cccDNA tran-
scription into pregenomic RNA (pgRNA), which serves
as a template for reverse transcription of the(−) strand
of rcDNA [3]. The hepatocyte-specific factors such
as hepatocyte nuclear factors (HNF) HNF1α, HNF3β,
HNF4α, HNF6, and factor C/EBP, play a crucial role in
the HBV replication cycle [4]. The rcDNA is released
from cells within denovo virions or re-imported into
the nucleus, thereby ensuring maintenance of the in-
tranuclear pool of cccDNA [5].
HBV causes both acute and chronic infection,
transmitted from mother to child or via bloodborne
transmission. Incubation period of the infection varies
from 30 to 180 days. In most cases, acute infection in
adults is self-limiting and results in virus elimination.
However, in rare cases, fulminant hepatitis develops
followed by liver failure and patient death. CHB is
characterized by a long-term inflammatory process in
the liver eventually leading to the development of liver
cirrhosis and hepatocellular carcinoma (HCC). The risk
of developing CHB in adults is low (~5%), whereas in
children under one year it reaches 95%. In 2022, 1.1
million deaths related to CHB outcomes, such as liver
cirrhosis and HCC, had been recorded worldwide [6].
Serological markers related to HBV infection in-
clude viral antigens HBsAg and HBeAg and antibod-
ies – anti-HBs, anti-HBe, and anti-HBc (IgM and IgG
classes). These markers allow for the identification of
patients with HBV infection as well as the diagnosis
of CHB and the differentiation of the clinical phases
of the disease, being also important for monitoring of
the effectiveness of antiviral therapy [7].
HBsAg can be detected in the blood of the pa-
tients during both acute infection and CHB. HBsAg is
located at the surface of the virions and circulates in
the blood as a part of subviral particles of various
shapes. Quantitative assessment of HBsAg levels as a
marker of viremia is important for monitoring of the
response to antiviral therapy. Anti-HBsAg antibodies
exert virus-neutralizing activity and provide protec-
tion against HBV infection [8]. HBsAg seroconversion,
defined as the lack of detectable HBsAg (<0.05  IU/ml)
and emergence of anti-HBs antibodies (>10  IU/ml) is
a crucial endpoint of CHB functional cure, implying
(in rare cases) a long-term decline in viral replication
and immunologically controlled infection. However,
in some cases occult HBV infection has also been de-
scribed, when patients with undetectable HBsAg in the
blood serum still have viral replication and inflamma-
tion in the liver, which represents an increased risk
of developing HCC and infection reactivation [9].
HBV DNA detection in the blood is the most rel-
evant and accurate marker of viral replication, which
is also widely used in HBV infection diagnostics.
Serum DNA level in CHB patients correlates with the
risk of liver damage progression. Assessment of se-
rum DNA levels is used to determine the need for
antiviral therapy and for monitoring of treatment ef-
fectiveness [7].
cccDNA is a stable form of HBV DNA that per-
sists as a viral mini-chromosome capable of binding
histones to form a nucleosome [10]. Up to 50 copies
of cccDNA are detected in the nuclei of HBV-infected
human hepatocytes. Due to the presence of the nu-
clear pool of cccDNA and the lack of approved drugs
that can directly block or destroy cccDNA, viral repli-
cation resumes following therapy cessation [11].
Development of new approaches to the treat-
ment of CHB requires adequate in  vitro and in vivo
models reproducing all of the described features of
HBV infection, which, however, is challenging. For
example, HBV exhibits strict species tropism partially
determined by the species specificity of the recep-
tor NTCP [12]. Furthermore, development of produc-
tive HBV infection requires several host intracellular
factors, additionally limiting the range of biological
species suitable for infection modeling [13]. Apart
from searching for the species susceptible and per-
missive to HBV, studies with other Hepadnaviridae,
such as duck hepatitis  B virus (DHBV), woodchuck
hepatitis virus (WHV), and woolly monkey hepati-
tis B virus (WMHBV), are underway. This review is
aimed to describe the current in  vivo and in vitro
models used to study HBV and related viruses as well
KOLYAKO et al.1506
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
as for testing of CHB-specific antiviral therapies. In
addition, we paid close attention to the stages and
features of HBV infection in humans that can be re-
produced by each model, which, we believe, could
help to develop next-generation drugs aiming at a
functional cure of CHB.
In vivo HBV INFECTION MODELS
Naturally susceptible animals. Chimpanzees
(Pan troglodytes) are the only non-human primates
fully susceptible to HBV [14]. They reproduce acute
HBV infection and can develop its chronic and per-
sistent forms, resulting in less pronounced pathologi-
cal changes in the liver as compared to humans [15].
Since 1972, when hepatitis B development in chim-
panzees following inoculation of viral material ob-
tained from humans was first reported [16], this spe-
cies has been used as an animal model to investigate
both HBV pathogenesis and immune response against
HBV. For instance, cross-reactive immune response to
HBsAg derived from the diverse viral subtypes has
been proven using chimpanzees [17]. Delayed-type
hypersensitivity was demonstrated in chimpanzees
following subcutaneous HBsAg inoculation, thereby
proving HBsAg immunogenicity [18]. Hence, these
studies laid the foundation for the development of
the first HBV vaccines, efficacy and safety of which
were also tested in chimpanzees. First-generation
vaccines were made by purifying HBsAg from the se-
rum of asymptomatic HBV carriers [19]. The resulting
preparation became the first human serum-derived
vaccine licensed in the United States [20]. However,
due to the concerns about the residual presence of
HBV and other viruses in the biomaterial, alternative
vaccine types have been later developed and tested
on chimpanzees, including the pre-S2 region-encoded
synthetic peptide [21], human hepatoma cell line PLC/
PRF/5-derived HBsAg [22], DNA vaccines [23], etc. Fur-
thermore, the currently used vaccine consisting of re-
combinant HBsAg produced in yeast (Saccharomyces
cerevisiae), has been tested in the chimpanzee model
[24]. Studies using chimpanzees revealed the crucial
role of proinflammatory cytokines in the clearance of
HBV during acute infection, achieved without exten-
sive destruction of hepatocytes [25].
Due to the ethical concerns and high cost, labora-
tory use of chimpanzees is limited, which has led to
the search for new models of HBV infection primarily
among other primates. However, the species-specific
differences in the amino acid sequence of the NTCP
orthologs make Old World primates insusceptible to
HBV. To skip the viral entry stage, a plasmid encod-
ing HBV genome dimer was directly injected into the
liver of Barbary macaques (Macaca sylvanus). Two
days after the injection and several weeks onward,
the presence of HBsAg and HBV DNA was detected in
the serum and liver tissue of the animals. Subsequent-
ly, the analysis of animal serum samples revealed the
HBV-like viral particles along with pathological liver
changes resembling those observed during hepatitis B
infection in humans. Hence, the obtained data demon-
strate that the HBV replication could occur in the
Barbary macaque hepatocytes [26].
In 2013, HBV DNA as well as HBsAg and HBcAg
were detected in the blood serum and liver tissue, re-
spectively, of cynomolgus macaques (Macaca fascicu-
laris) from the island of Mauritius, whereas no patho-
logical changes in the liver tissue were observed [27].
However, it was later reported that the virus isolated
during this study was unable to induce HBV infection
in the same macaque species [28].
The northern treeshrew (Tupaia belangeri) is
an HBV-susceptible small rodent-like mammal that
is phylogenetically more closely related to primates
[29]. Adult animals directly infected with HBV ex-
hibited a self-limiting acute infection with increased
HBsAg levels in the serum 2-3 weeks post-infection
along with the rapidly produced anti-HBeAg anti-
bodies [30]. Injection of the virus into neonatal ani-
mals caused chronic infection lasting over 48-weeks.
cccDNA has been detected in the liver of northern
treeshrews with CHB along with pathological changes
in the liver morphology, which developed at a slower
rate than in humans [31]. Nonetheless, chronic HBV
infection in treeshrews could result in HCC develop-
ment [32], which is not typical for some other animal
models of HBV infection. Additionally, immunohisto-
chemical staining revealed the presence of HBcAg in
hepatocytes during both acute and chronic infection,
confirming viral replication process [31]. Studying of
HBV using treeshrews is hampered by the difficulty in
selecting adequate species-specific reagents and pro-
tocols, low viral loads, and the need to infect neona-
tal animals, while the incidence of CHB development
is low (13%) [30].
Genetically modified animals. Susceptibility to
HBV depends on the species-specific host factors, pri-
marily the sequence of NTCP orthologs. Human NTCP
gene (hNTCP) transduction into mouse, rat, and dog
hepatocytes followed by HBV infection ensures virus
entry into the cells but does not result in effective
viral replication, suggesting that species-specific in-
tracellular factors affect the HBV replication cycle.
Hepatocytes derived from cynomolgus macaques, rhe-
sus macaques, and pigs, after transduction of hNTCP,
supported complete HBV replication cycle compara-
ble to that in human hepatocytes, thereby opening
the way to use immunocompetent macaques in stud-
ies of HBV infection, immune response, and viral
pathogenesis [33].
HEPATITIS B VIRUS INFECTION MODELS 1507
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Due to the general similarity between the hu-
man and macaque immune systems, rhesus macaques
(Macaca mulatta) have several advantages over oth-
er HBV infection models [34]. Inoculation of rhesus
macaques with the hNTCP-encoding adenoviral vec-
tor followed by infection with HBV [28] led to mod-
erate viremia. However, no chronic infection devel-
oped in this model. Immunohistochemical staining
of the liver sections revealed the presence of HBcAg
solely inside the nuclei of 0.5-1.0% hepatocytes [28].
In order to achieve chronic infection, immunosup-
pression was additionally induced in infected rhesus
macaques, which resulted in persistent 20-week long
HBV replication identified by the presence of HBsAg
and HBeAg in the blood as well as HBV DNA in liver
biopsies. However, after cessation of immunosuppres-
sion, immune-mediated virus elimination occurred in
most animals with emergence of serum anti-HBs and
anti-HBc IgG antibodies, as well as CD8
+
and CD4
+
T cell activation [35]. Thus, the genetically modified
hNTCP-expressing rhesus macaques could maintain
prolonged HBV infection subsequently resulting in
specific immune-related virus elimination.
Surrogate models. Duck hepatitis B virus. The
studies with other members of Hepadnaviridae fam-
ily were conducted in parallel with the search for
HBV-susceptible animal species. In 1980, a virus later
named duck hepatitis  B virus (DHBV) was discovered
in the blood serum obtained from domestic ducks
(Anas domesticus). Viral DNA was detected mainly
in the liver, suggesting hepatospecificity of the virus.
Electron microscopy analysis of the purified virus re-
vealed particles (40  nm in diameter) similar to those
of other members of the Hepadnaviridae family [36].
Similar to HBV, the DHBV genome is represented by
rcDNA [37]. The DHBV-infected ducks were used as
a model for testing of antiviral drugs such as poly-
merase inhibitors [38] and nucleocapsid assembly
inhibitors [39]. Although ducks offer a relatively con-
venient model for laboratory use, extrapolation of the
research data to humans is complicated because of the
differences in the genome sequence and pathogenesis
between DHBV and HBV. In particular, nucleotide se-
quence of the DHBV genome is only 40% homologous
to that of HBV [40]. Moreover, specific virus receptors
also are different: the entry receptor for DHBV is car-
boxypeptidase D [41], which does not allow to study
the drugs targeting the interaction between HBV and
its receptor. In addition, no correlation was found be-
tween chronic hepatitis and the development of HCC
in various duck species infected with DHBV [42].
Woolly monkey hepatitis B virus. Besides HBV,
Orthohepdnavirus genus also includes woolly monkey
hepatitis  B virus (WMHBV), identified in the blood of
common woolly monkeys (Lagothrix lagotricha). The
WMHBV amino acid sequence is 44% homologous to
that of HBV, however the HBcAg gene which is pre-
sumably conserved among the Hepadnaviridae family,
has 85-86% and 75-77% homology to the HBV HBcAg
in amino acid and nucleotide sequence, respectively.
The WMHBV causes either acute (fulminant) or chron-
ic infection in common woolly monkeys [43]. Other
primates were also experimentally infected with
WMHBV. HBsAg and WMHBV DNA were detected in
black-handed spider monkey (Ateles geoffroyi) blood
serum for 4-6 weeks after infection, while no patho-
logical changes were detected in liver biopsies [43].
In order to establish chronic WMHBV infection, neona-
tal black-handed spider monkeys were infected with a
WMHBV clone, which did not lead to the development
of a chronic disease [44]. As common woolly monkeys
and black-handed spider monkeys are endangered
species, ethical reasons hamper their large-scale re-
search use, which motivated researchers to use a re-
lated primate species, squirrel monkeys (Saimiri sciu-
reus), as an alternative. Infection of squirrel monkeys
with WMHBV-containing biomaterial lead to acute in-
fection with viremia that lasted for 4 weeks. To pro-
long the infection, the animals were inoculated with
the adeno-associated virus (AAV8) encoding infectious
WMHBV genome, which resulted in viremia lasting
32 weeks. In all neonatal squirrel monkeys infected
with WMHBV viremia lasted for 3 months, followed
by virus elimination [45]. Thus, despite the absence
of chronic infection, squirrel monkeys infected with
WMHBV can be used as a surrogate model of HBV
infection.
Woodchuck hepatitis virus (WHV). WHV also be-
longs to the genus Orthohepdnavirus. Along with
general similarity in the genome and virion struc-
ture (WHV genome has 62-70% nucleotide sequence
homology to HBV), WHV and HBV exhibit antigenic
cross- reactivity. Also, these viruses have the same
timeline of viral antigens appearance in the liver and
blood after infection, which indicates the similarity
of the stages of infection course [46]. In adult wood-
chucks, acute WHV infection may result either in
complete virus elimination or progression into chron-
ic form. Inneonatal woodchucks, the infection mainly
becomes chronic [47]. Chronic WHV-infection leads to
exhaustion of the WHV-specific T-cell response as well
as to low interferon production by hepatocytes [48].
WHV was first detected in the tissues of dead
woodchucks (Marmota monax), which showed signs
of HCC development [47]. Similar to HBV, WHV is an
oncogenic virus [49], with HCC developing in 90% of
cases of verified chronic WHV infection [49]. There-
fore, high HCC incidence allows to use woodchucks
as a convenient model in studies of viral oncogenesis.
The ability of the WHV genome to integrate into the
cell genome and cause increased proliferation of he-
patocytes was studied using the marmot model [50].
KOLYAKO et al.1508
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Woodchuck models have been useful for preclinical
testing of antiviral drugs, including nucleoside ana-
logues [51]. In 2011, Himalayan marmots (Marmota
himalayana) were also shown to be susceptible to
WHV, expanding the panel of available surrogate
models of HBV infection [52]. However, susceptibility
to WHV varied between different animals [53]. Appli-
cability of woodchucks as an HBV infection model is
limited due to the difficulties in laboratory housing
and care of these animals and limited availability of
reagents for the immune response studies.
Mouse models. For biomedical research, mice are
the most common and easy-to-use model, with stan-
dardized methods and reagents available for a wide
range of experiments. Because mice are normally nei-
ther susceptible nor permissive to HBV, gene modifi-
cation and human tissue transplantation are used to
generate mouse models of HBV infection.
Transgenic mice. Due to the development of
the embryonic microinjection technology, transgen-
ic mouse models have been created, which carry in
their genome either individual HBV genome regions
encoding HBsAg [54], HBeAg [55], and HBxAg [56] or
full-length HBV genome [57]. The hepatocyte-specif-
ic promoters such as the mouse metallothionein or
albumin promoters were used to control HBV gene
expression in the mouse liver [58].
HBsAg-producing transgenic mice were used for
the modeling of the asymptomatic HBV carriage [54]
as well as for the validation of the indirect oncogenic
effect of HBsAg [59]. Moreover, the oncogenic effect
of HBxAg was demonstrated in transgenic mice, as
production of HBxAg was directly associated with his-
topathological changes in the liver [56]. HBV vertical
transmission was shown in transgenic female mice
bearing complete HBV genome. In addition, it has
been shown that HBeAg could induce the production
of the regulatory peptide PD-L1 by liver macrophages.
Production of PD-L1 leads to the decrease in CD8
+
T
cell proliferation [60], resulting in HBV persistence
in the transgenic mouse offspring [61]. A disadvan-
tage of transgenic mouse models is that the proteins
produced from the transferred genes are not recog-
nized as foreign by the immune system of the ani-
mal, which hinders the assessment of the immune re-
sponse. Furthermore, there is no recirculation of the
assembled viral particles into hepatocytes of transgen-
ic mice bearing complete HBV DNA sequence, as the
mouse NTCP ortholog is unable to facilitate HBV entry
into cells. The transgenic mouse model is not relevant
for testing of therapeutic drugs aimed at blocking or
destroying cccDNA, because this form of HBV genome
is not formed in mice. The virions assemble due to
the expression of linear genomic sequences of the vi-
rus integrated into the mouse genome, which makes
their complete elimination impossible [62].
Mouse models with invivo transfection. To avoid
genetic aberrations caused by the HBV genome inte-
gration into the mouse genome, models of episomal
HBV assembly have been developed. Currently, two
methods are used to deliver HBV genome into the
mouse hepatocytes either as HBV-encoding plasmids
or as recombinant cccDNA (rcccDNA): high-pres-
sure hydrodynamic injection-based HBV transfection
(HDI-HBV) and transduction using adeno-associated
virus (AAV) or adenovirus (Adv) vectors, bearing HBV
genome. The HDI-HBV results in transfection of 10%
hepatocytes, whereas adenoviruses provide transduc-
tion of 90% hepatocytes. The drawbacks of such ap-
proaches are as follows: (i) lack of the infection stage
and full viral replication cycle, as the “surrogate” HBV
cccDNA is formed in the transduced mouse cells, from
which viral transcripts are produced; (ii) non-physi-
ological viremia levels – tens of times higher than
in natural infection; (iii)potential for development of
the robust immune response both against viral com-
ponents due to the high level of virion production
and against adenovirus components; (iv) tissue non-
specificity of HDI-HBV, as along with hepatocytes the
viral genome may potentially enter other cell types
[63]. Since hepatotoxic effects are observed upon us-
ing the plasmids/rcccDNA at high doses in HDI-HBV or
when inoculating >10
9
adenovirus particles encoding
the HBV genome (AdHBV), 10μg DNA in HDI-HBV and
up to 10
8
AdHBV should be used in in  vivo experi-
ments [64, 65].
Genetic background of the mice used in the ex-
periments also affects the course of HBV infection.
In particular, BALB/c mice after HDI-HBV (10  μg of
plasmid in a volume equivalent to 8% of mouse
body weight) are predisposed to HBV elimination
with production of anti-HBsAg antibodies within 14
days post-injection, whereas in C57BL/6 mice HBsAg
was detected for 35 days after plasmid injection [66].
The main drawback of the described mouse models
is the absence of cccDNA. To overcome this obstacle,
a precursor recombinant cccDNA plasmid (prcccDNA)
was synthesized and transduced using Adv into the
C57BL/6 and Alb-Cre Tg mice (C57BL/6-Tg [Alb-cre]
21Mgn/J). Recombination of prcccDNA occurred in
the nuclei of the transduced hepatocytes resulting in
the production of rcccDNA. Viremia in C57BL/6 mice
was not observed following HDI-HBV, whereas HBcAg
was detected at low levels and located primarily in
the hepatocyte nuclei. At the same time, in Alb-Cre
Tg mice rcccDNA was detectable for 62  weeks accom-
panied by the pronounced viremia as evidenced by
the detection of HBcAg and HBsAg in the blood. In-
creased PD-L1 production in the liver also resulted in
the decrease in specific T-cell response against the rc-
ccDNA-positive hepatocytes, which contributed to the
rcccDNA persistence [67].
HEPATITIS B VIRUS INFECTION MODELS 1509
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig. 1. Stages of HBV infection and related pathogenesis reproduced in the in vivo models. The animal species and relevant
viruses as well as recombinant vectors used for each model are presented. Designations: DHBV, Duck Hepatitis B Virus;
WHV, woodchuck hepatitis virus; WMHBV, woolly monkey hepatitis B virus; AAV, adeno-associated virus. Species-specific
descriptions: P.t., Pan troglodytes; T.b., Tupaia belangeri; M.f., Macaca fascicularis; M.m-a, Macaca mulatta; M.m., Mus mus-
culus; A.d., Anas domesticus; M.m-x, Marmota monax; M.h., Marmota himalayana; L.l., Lagothrix lagotricha; A.g., Ateles
geoffroyi; S.s., Saimiri sciureus). cccDNA, covalently closed circular DNA; HCC, hepatocellular carcinoma.
To assess the efficacy of novel HBV treatments
in vivo, an episomal vector based on the adeno-as-
sociated virus AAV9-HBsAg was developed. AAV9-HB-
sAg serves as a surrogate for cccDNA and contains a
partial HBV sequence under the control of the hep-
atospecific promoter. Inoculation of AAV9-HBsAg into
NOD-SCID IL2rg
−/−
mice resulted in HBsAg secretion
lasting for 7 weeks. Subsequently, the AAV8-HBsAg
was also used in the cynomolgus macaque (Macaca
fascicularis) model to test the HBV genome-specific
engineered nuclease (ARCUS-POL) [68].
Thus, a wide range of in vivo HBV infection
models designed in diverse animal species is current-
ly available, each of which (except for chimpanzee
model) only partially reproduces the course of human
HBV infection. The advantages and drawbacks of such
models are shown in Fig. 1.
Xenograft-based mouse models. Transgenic and
transduced mice models do not fully reproduce the
natural course of HBV infection in vivo; moreover,
many antiviral drugs cannot be tested using such
models. To reproduce the complete HBV replication
cycle in mice, the Trimera model was developed. For
this, the immunocompetent CB6F1 mice were exposed
to lethal gamma irradiation followed by inoculation
with bone marrow cells derived from SCID/NOD mice.
Fragments of human HBV-infected liver tissue were
then transplanted under the subrenal capsule or into
the auricle of the animals. HBV DNA was detected in
the serum of the CB6F1 mice bearing human liver
KOLYAKO et al.1510
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
xenografts from day 8 to day 25 post-transplantation
and declined afterwards. A similar infection profile
was achieved using the Trimera model in the immu-
nodeficient BNX mice. cccDNA was detected in the liv-
er grafts indicating viral replication inside the human
hepatocytes. The Trimera model was used to evaluate
the efficacy of antiviral drugs including polyclonal
anti- HBsAg antibodies, reverse transcriptase inhib-
itors, and nucleoside analogues [69]. Another mod-
el was also created using nude (nu/nu) mice, which
were subcutaneously injected with the HepAD38 cells
bearing complete HBV genome under the control of
tetracycline repressor. Viral replication was regulated
by the presence of tetracycline (tet) in drinking wa-
ter. In the absence of tet, the mice developed viremia
lasting 35 days after infection, and cccDNA was de-
tected in the liver. Viral load decreased after adding
tet to drinking water and relapsed after tet removal
[70]. The developed model was used to test combi-
nation therapy against HBV in vivo [71]. Long-term
HBV viremia was achieved using a transplantation
model based on mice with combined RAG-2 deficiency
(RAG-2M). For this, a primary human hepatocyte cul-
ture was immortalized by transduction with the SV40
T-antigen followed by transfection with the full-length
HBV genome and obtaining monoclonal cell line
(IHBV6.7). The latter was transplanted into the mice
via intrasplenic injection. Although the transplanted
IHB6.7 cells comprised as few as 1% of the mouse
hepatocyte mass, viremia lasted for at least 5 months
post-transplantation [72]. The ability to not only in-
tegrate into, but also partially repopulate the mouse
liver with human hepatocytes (up to 15% mouse liv-
er) was first demonstrated in the uPA/RAG-2 model,
using immunodeficient RAG-2 knockout mice crossed
with the uPA transgenic mice. Overexpression of the
urokinase-type plasminogen activator (uPA) gene in
mouse hepatocytes resulted in neonatal hemorrhage
and liver necrosis. Following human hepatocyte xeno-
transplantation, mouse hepatocytes were replaced in
the liver by human hepatocytes. Infection of the uPA/
RAG-2 mice with serum samples derived from HBV
carriers resulted in viremia and detection of HBsAg
for 8 weeks following infection [73]. Another mod-
el was developed based on SCID mice homozygous
for the Alb-uPA transgene [74]. Infection of the xeno-
transplanted SCID Alb-uPA mice with serum samples
derived from HBV carriers resulted in viremia that
was detected at week 2 after infection and persisted
for 12 weeks. It was also shown that HBV particles
derived from HepG2 cells bearing the full-length vi-
ral genome were able to infect SCID Alb-uPA mice.
However, the viral load detected from week 4 was
significantly lower compared to infection with blood
serum samples. The drawback of the Alb-uPA SCID
model was the lack of the control over uPA expres-
sion, the need for transplantation into neonatal mice,
and increased risk of hemorrhage [75]. Therefore, an
alternative Fah
−/−
Rag-2
−/−
Il2rg
−/−
mouse model was
created using immunodeficient animals with deleted
gene encoding fumarylacetoacetate hydrolase (Fah).
Loss of the latter results in accumulation of toxic
tyrosine metabolites in mouse hepatocytes causing
their death, which promotes liver repopulation with
the xenograft hepatocytes. Furthermore, the toxicity
level could be controlled by the orally administered
2-(2-nitro-4-trifluoro-methylbenzoyl)-1,3-cyclohexane-
dione (NTBC). Following human hepatocyte trans-
plantation, the mouse liver comprised as many as
20% of the xenograft hepatocytes, and repopulation
success rate ranged from 45 to 95% [76]. Mice with
high proportion of liver repopulation were infected
with HBV. From the 2nd to the 7th week of follow-up,
cccDNA was detected in liver samples and viremia
was confirmed with the peak of the viral load detect-
ed at the 6th week. Immunocytochemical staining for
HBcAg demonstrated that the infection proceeded ex-
clusively in the transplanted human hepatocytes, with
HBcAg accumulation observed in both the nuclei and
cytoplasm [77]. The model was used to evaluate the
efficacy of novel antiviral drugs [78].
Assessment of the immune response in xenograft
mouse models is complicated, as it requires the use
of immunocompromised animals. To overcome this
limitation, additionally to human hepatocytes, mice
were transplanted with human immune system (HIS)
elements. In particular, the neonatal A2/NSG mice ex-
pressing human HLA-A2 gene were inoculated with
donor-derived CD34
+
hematopoietic stem cells (HSC)
and human hepatocyte progenitor cells (Hep). This
resulted in the development of a functional human
immune system along with liver repopulation. In this
process, HLA-A2 expression increased the selection of
T cells producing only human MHC. To promote liv-
er repopulation with human cells, mouse hepatocyte
death was selectively induced by anti-murine Fas an-
tibodies. It was found that the A2/NSG-hu HSC/Hep
mice developed low viremia over 12 weeks after HBV
infection. The immune response was characterized
by the increased production of IFN-γ, IP10, and IL-6
along with the low levels of IL-10 and IFN-α, as well
as the presence of anti-HBsAg antibodies and HBV-spe-
cific CD8
+
T cells. While examining the liver tissue
in HBV-infected mice, increased collagen deposition
was described, which is typical for HBV-induced tis-
sue fibrosis during CHB [79]. For dual humanization,
the BALB/c Rag-2
−/−
Il2rg
−/−
Sirpa
NOD
Alb-uPA
tg/tg
mice
were also used for transplantation of human hema-
topoietic stem cells. The resulting model (HIS-HUHEP)
demonstrated high human hepatocyte engraftment
rate (about 20-50% of mouse liver was replaced with
human cells) [80]. Infection of HIS-HUHEP mice with
HEPATITIS B VIRUS INFECTION MODELS 1511
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
HBV showed that the viral load and accumulation
of T cells, Kupffer cells and natural killer cells (NK
cells) in the liver correlated with the administered
HBV dose. However, the disease progressed to chron-
ic course without the development of liver fibrosis.
Itwas also shown that the liver PD-L1 protein expres-
sion in infected animals was elevated in comparison
with intact mice. It has been shown previously that
PD-L1 expression leads to T-cell exhaustion, which is
one of the causes of CHB [81]. Another alternative
model is based on Fah
−/−
Rag-2
−/−
IL-2Rγc
−/−
SCID (FRG)
mice transplanted with human bone marrow-derived
stem cells (hBMSCs). After hBMSC-FRG animals were
infected with HBV, viral DNA and antigens were de-
tected in serum at relatively high levels for 56 weeks.
Immunohistochemical staining of the liver sections
for human albumin, HBsAg, and HBcAg revealed
that only human hepatocytes were infected with HBV.
The percentage of infected hepatocytes increased up
to 80% over time. Analysis of mouse liver sections
also showed increased levels of proinflammatory
cytokines, human chemokines, as well as of hCD3,
hCD86, and NK cells, suggesting an active immune
response. Moreover, the humoral response was also
characterized by the presence of specific IgG against
HBsAg and HBcAg, which, however, declined sharply
at the week 32. Pathological liver changes typical to
chronically infected patients were observed through-
out the experiment, which included lobular inflamma-
tion, lymphoid aggregates, and duct damage, as well
as lymphocyte infiltration from the portal vein. At the
later stages, signs specific to the developing cirrhosis
were observed: collagen accumulation, fibrous tissue
hyperplasia, and abnormal liver lobe structure [82].
Xenograft mouse models played an important role
in investigating HBV pathogenesis in vivo as well as
efficacy assessment of antiviral therapy. The double-
humanized mice also allowed to assess the impact
of chronic HBV infection on the development of im-
mune response. However, use of such animal models
is complicated by the labor-intensive process of their
obtaining, high cost, and risk of sudden death. The
advantages and drawbacks of the described models
are summarized in Table S1 (Online Resource 1).
In vitro HBV INFECTION MODELS
Primary human hepatocytes. Primary human
hepatocytes are derived from the livers of organ do-
nors, tissues after liver surgery including resections,
cadaveric material, and liver biopsies [83]. Due to
the natural susceptibility to HBV infection, human
hepatocyte culture serves a reference model for HBV
research. Hepatocytes fully support HBV replication
cycle and, moreover, possess a functional intracellular
innate immunity allowing for studies of the innate
antiviral immune response against HBV [84]. How-
ever, primary hepatocytes undergo rapid phenotypic
changes during in  vitro culturing [85]. Initially, pri-
mary hepatocytes could be maintained in  vitro for up
to one month. Modifications in culturing conditions
allowed to extend hepatocyte viability up to 2 months
without affecting their metabolism [86], which is still
insufficient for CHB modeling. Culture media supple-
mentation with dimethyl sulfoxide (DMSO) has been
shown to promote hepatocyte differentiation, leading
to a significant increase in HBV replication level [87].
HBV viral particle and protein production levels vary
between the primary hepatocyte cultures obtained
from different donors [88]. Thus, although primary
human hepatocytes support full HBV replication cycle,
use of this model is limited due to the low acces-
sibility and heterogeneity of the biomaterial, which
profoundly complicates their large-scale application.
Human hepatoma cultures. HepG2 and Huh-7
are the most widely used human hepatoma cell lines
due to their in  vitro stability and ease of cultivation.
However, due to the lack of surface hNTCP, they are
not susceptible to HBV infection. The hepatoma cells
can only be used as HBV infection models after their
modification by HBV genome transfer or by hNTCP
expression followed by HBV infection.
Huh-7 cells, isolated from hepatocellular carcino-
ma in 1982 [89], were shown to be a useful in  vitro
model for studying of HBV replication cycle. After
Huh-7 cells were transfected with a plasmid encod-
ing HBV DNA genome dimer, HBsAg and HBeAg were
detected in the culture medium for 25 days. More-
over, the presence of viral particles was confirmed
in culture medium at day 20 after transfection [90].
The Huh-7 cell line was used as a model to investi-
gate the molecular mechanisms underlying HBV rep-
lication [91, 92] including virion assembly and mat-
uration [93], impact of the immune factors on viral
replication [94] and efficacy assessment of antiviral
agents [95, 96] including novel therapeutic approach-
es based on TALENs- and CRISPR/Cas9 cccDNA cleav-
age systems [97].
HepG2 cells were isolated from the hepatocellular
carcinoma of a 15-year-old patient [98]. After transfec-
tion of HepG2 cells with a plasmid encoding an HBV
DNA genome dimer, the HepG2.2.15 monoclonal cell
line with genome-integrated HBV DNA was obtained
[99]. Infectivity of the viral particles isolated from
HepG2.2.15 cells was demonstrated in the chimpanzee
model [100]. Additionally, HepG2.2.15 cells allowed to
identify the potential molecular targets for CHB ther-
apy [101]. In 1997, the HepAD38 cell line was derived
from HepG2 cells after transfection with the plasmid
encoding HBV genome under the control of tetracy-
cline-responsive promoter [102]. Later, the modified
KOLYAKO et al.1512
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
cell lines, HepDE19 and HepDES19, were obtained,
in which viral particle production was tetracycline-
depended [103]. These cell lines were used to screen
potential inhibitors of cccDNA formation [104].
HepaRG cells were derived from the hepato-
cellular carcinoma cells of a hepatitis C-infected
patient. After differentiation by adding DMSO and
corticosteroids to the culture medium HepaRG cells
morphologically and functionally resembled primary
hepatocytes and were successfully infected with HBV.
HBsAg, HBcAg, and HBV DNA were detected in the
culture medium after infection [105]. The presence of
cccDNA in the infected cells was later confirmed in
a 50-day experiment. However, susceptibility of the
HepaRG cells is strictly dependent on cell differen-
tiation, which requires extended period of time to
achieve (2 weeks for proliferation and 2 weeks for
differentiation in the culture medium supplemented
with DMSO). Moreover, the HepaRG cell line consists
of differentiated and polarized hepatocyte-like cells
and biliary-like epithelial cells, which results devel-
opment of the infection in islands of differentiated
cells [106]. Additionally, prolonged exposure to DMSO
induces cell death, which also affects the HBV infec-
tion characteristics [107].
Upon discovering hNTCP as a functional receptor
of HBV [12], the research focus was directed towards
the generation of HBV-susceptible cell lines by hNTCP
overexpression in cells. It was experimentally con-
firmed that hNTCP-expressing HepG2 and Huh-7 cell
lines (HepG2-NTCP and Huh7-NTCP, respectively) are
susceptible to HBV purified from serum samples of in-
fected patients, as well as the virus isolated from cul-
ture medium. The HepG2-NTCP and Huh7-NTCP cells
support complete HBV replication cycle, from viral
entry to cccDNA formation. Due to the ease of their
production, these cell lines have been widely used for
testing and development of antiviral drugs [108, 109].
However, HBV infection efficiency in HepG2-NTCP and
Huh7-NTCP cell lines varies. Although hNTCP expres-
sion allows HBV to enter Huh7-NTCP cells, further viral
replication occurs in a smaller proportion of cells in
comparison with the HepG2-NTCP cell line [110]. The
current HepG2-hNTCP models allow for infection of
up to 50-60% of cells (https://www.abmgood.com/ntcp-
stable-hepg2-cell-line.html, accessed 18.04.25). Despite
this, virus particle production by the HepG2-NTCP cell
line is low. In order to create a cell culture with high
virus yield, the monoclonal HepG2-NTCP-sec
+
culture
with low proliferative activity was selected, in which
more than 90% of the cells remained infected for
4weeks after HBV infection without a decrease in the
levels of detectable HBV DNA [111].
Overall, human hepatoma cells represent a useful
and convenient tool for researchers, but their wide-
spread use is profoundly limited by the fact that
HBV infection is transient, without significant spread
among new cells. Moreover, the hepatoma cells differ
from the primary hepatocytes in multiple physiolog-
ical functions and mechanisms, that complicates the
data interpretation. Oncogenic origin of such cell lines
complicates their use in investigating the mechanisms
behind the HBV-induced carcinogenesis.
Animal cell lines. Being susceptible to HBV, the
northern treeshrew played a crucial role not only in
the in  vivo but also in in  vitro studies of HBV infec-
tion. The infection of primary treeshrew hepatocytes
resulted in HBV DNA and RNA production lasting
for 12 days as well as in the detection of HBsAg and
HBeAg in the culture medium for up to 5-6 days [30].
Later, accumulation of cccDNA was also demonstrat-
ed. Furthermore, treeshrew hepatocytes were shown
to be susceptible to WMHBV, with higher efficien-
cy of viral replication compared to HBV [112]. The
HBV-infected treeshrew hepatocytes were used as an
in  vitro model for efficacy assessment of antiviral
agents [113] and in studies of virus-cell interactions
[114]. This model was used to prove that the HBV
pre-S1 domain enables HBV entry, which occurs in a
species-specific manner [115]. Treeshrew hepatocytes
were used to assess the infectivity of viral particles
produced by human hepatoma cell lines [116]. Tree-
shrew primary hepatocytes are also a more readily
available alternative to human primary hepatocytes.
This allowed their use in the development of a xe-
nograft mouse model, which was subsequently suc-
cessfully infected with HBV [117] and used to test a
potential inhibitor of HBV entry [118]. In addition, the
treeshrew xenograft mouse model helped to describe
the emerging hepatocyte proliferation, which resulted
in the decreased population of the cccDNA-containing
cells [119].
Among other studied animals, the common mar-
moset (Callithrix jacchus) is a promising animal mod-
el of HBV infection. Although common marmoset he-
patocytes are not directly susceptible to HBV infection,
it has been shown that the Adv-mediated delivery of
the full-length HBV genome to a primary hepatocyte
culture resulted in viral replication with detection of
cccDNA and its derivatives, which was followed by
HBV particle formation, confirming permissiveness of
the common marmoset cells to HBV [120]. Recently it
has been found that common marmoset hepatocyte
resistance to HBV is associated with amino acid differ-
ences between common marmoset NTCP and hNTCP
at two positions directly involved in HBV binding.
In order to overcome this obstacle, a chimeric HBV/
WMHBV pre-S1 virus was constructed and isolated.
This virus successfully entered and replicated in
common marmoset hepatocytes, as evidenced by the
detection of HBeAg in the culture medium and the
presence of HBV cccDNA in cells [121]. Taken together,
HEPATITIS B VIRUS INFECTION MODELS 1513
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
these results suggest high potential for further use
of common marmoset hepatocytes in HBV research.
However, currently common marmoset hepatocyte
cell lines are not yet available.
Primary hepatocytes from woodchucks and ducks
have also been used for in  vitro studies. When prima-
ry woodchuck hepatocytes were infected with WHV,
cccDNA formation was detected on day 2 post-infec-
tion followed by the detection of DNA fragments for
7-10 days, indicating active WHV replication  [122].
HBV-infected woodchuck hepatocytes served as a mod-
el for efficacy assessment of antiviral drugs, such as
DNA synthesis inhibitors [123]. Due to the relative ease
of culturing, primary duck hepatocytes were more
widely used [124]. The DHBV-infected duck hepato-
cyte culture allowed to explore reverse transcription
activation [125] and cccDNA formation from rcDNA
[126], as well as nucleocapsid assembly [127]. How-
ever, translational value of the obtained data is ham-
pered by the above-mentioned genomic differences
between HBV and DHBV.
Hence, hepatocyte cell cultures of various origins
are widely used in the studies of various stages of
HBV replication cycle, as well as in testing of potential
antiviral drugs. However, similar to the in vivo mod-
els, each type of cell cultures has certain limitations
that must be taken into consideration while making
the decisions regarding their use in experiments. The
hepatocyte culture-based models described here are
summarized in Table S2 (Online Resource 1).
CONCLUSION
Undoubtedly, HBV infection poses a serious global
health threat. Relevant and accessible models of viral
infection are required to investigate the mechanisms
underlying HBV pathogenesis, identify new antiviral
targets, as well as to test novel antiviral vaccines and
therapeutics. Here, we review most of the currently
available models used to study HBV infection.
Among the in vivo models, the chimpanzee has
historically served as a reference research model due
to the natural susceptibility to HBV and phylogenetic
proximity to humans. However, high cost and difficul-
ty in maintenance in laboratory conditions limit the
widespread use of chimpanzees. Among the in  vitro
models, primary hepatocyte cultures serve as a stan-
dard reference model. Both models are virtually inap-
plicable for large-scale studies due to ethical concerns
and limited availability.
Ideally, a universal HBV model should meet the
following parameters: (i)  viral replication level similar
to the one observed in natural infection, with max-
imum infection efficiency; (ii)  recapitulate all stages
of infection development, replication cycle and im-
mune response; (iii)  be widely accessible with regard
to cost-effectiveness and availability of reagents;
(iv)  ensure data reproducibility. Owing to decades of
research, a wide range of diverse HBV infection mod-
els have been developed, but none of them fully meet
these criteria.
In this regard, modifications of the existing
in  vitro models such as HepG2-NTCP-sec
+
cell line
and the search for new susceptible animals could
provide most promising approaches to development
of a universal model. It is worth noting that, despite
the mentioned drawbacks, genetically modified and
humanized mice are most widely used as in  vivo
models, in part due to accessibility and availability
of the necessary reagents, particularly specific anti-
bodies. In this regard, the model using common mar-
moset hepatocytes and chimeric HBV/WMHBV pre-S1
virus undoubtedly has an advantage due to the wide
panel of available methods and reagents.
In conclusion, no universal model of HBV infec-
tion able to recapitulate all the stages of viral repli-
cation cycle and host immune responses is currently
available. Its development might be a crucial step
toward creating therapies for CHB and HBV-induced
pathologies.
Abbreviations
AAV adeno-associated virus
Adv adenovirus vector
cccDNA covalently closed circular DNA
CHB chronic hepatitis B
DHBV duck hepatitis B virus
HBV hepatitis B virus
HBeAg hepatitis B virus E antigen
HBcAg hepatitis B virus core antigen
HBsAg
hepatitis B virus surface antigen iso-
forms
HCC hepatocellular carcinoma
HDI-HBV
high-pressure hydrodynamic injection-
based HBV transfection
NTCP
Na
+
-taurocholate cotransporting poly-
peptide
rcDNA relaxed circular DNA
rcccDNA recombinant cccDNA
tet tetracycline
uPA urokinase-type plasminogen activator
WHV woodchuck hepatitis virus
WMHBV woolly monkey hepatitis B virus
Supplementary information
The online version contains supplementary material
available at https://doi.org/10.1134/S0006297925602230.
Contributions
Y. V. Kolyako – wrote the initial version of the
manuscript, edited the text; A. S. Zhitkevich and
KOLYAKO et al.1514
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
D. V. Avdoshina – edited the text, created the illustra-
tions; D. Y. Tanygina and V. D. Apolokhov – created
the illustrations; T. V. Gorodnicheva – edited the text;
E. O. Bayurova – edited the text, conceptualized the
study; D. S. Kostyushev and I. V. Gordeychuk – edited
the text, conceptualized the study, funding acquisition.
Funding
The study was financially supported by the Russian
Science Foundation (grant no.25-65-00010).
Ethics approval and consent to participate
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest
The authors of this work declare that they have
noconflicts of interest.
REFERENCES
1. Kramvis,A. (2014) Genotypes and genetic variability
of hepatitis B virus, Intervirology, 57, 141-150, https://
doi.org/10.1159/000360947.
2. Yang,R., Ko, Y.-H., Li,F., Lokareddy, R.K., Hou, C.-F.D.,
Kim, C., Klein, S., Antolínez, S., Marín, J. F., Pérez-
Segura,C., Jarrold, M. F., Zlotnick,A., Hadden-Perilla,
J.A., and Cingolani, G. (2024) Structural basis for nu-
clear import of hepatitis B virus (HBV) nucleocapsid
core, Sci. Adv., 10, eadi7606, https://doi.org/10.1126/
sciadv.adi7606.
3. Tsukuda, S., and Watashi, K. (2020) Hepatitis B virus
biology and life cycle, Antiviral Res., 182, 104925,
https://doi.org/10.1016/j.antiviral.2020.104925.
4. Kim, D. H., Kang, H. S., and Kim, K.-H. (2016) Roles
of hepatocyte nuclear factors in hepatitis B virus
infection, World J. Gastroenterol., 22, 7017, https://
doi.org/10.3748/wjg.v22.i31.7017.
5. Ko, C., Chakraborty, A., Chou, W.-M., Hasreiter, J.,
Wettengel, J. M., Stadler, D., Bester, R., Asen, T.,
Zhang,K., Wisskirchen,K., McKeating, J.A., Ryu, W.-S.,
and Protzer,U. (2018) HepatitisB virus genome recy-
cling and de novo secondary infection events main-
tain stable cccDNA levels, J. Hepatol., 69, 1231-1241,
https://doi.org/10.1016/j.jhep.2018.08.012.
6. (2024) Global Hepatitis Report 2024: Action for Ac-
cess in Low- and Middle-Income Countries, 1st ed ed.,
World Health Organization, Geneva.
7. Song, J. E., and Kim, D. Y. (2016) Diagnosis of hep-
atitis B, Ann. Translat. Med., 4, 338, https://doi.org/
10.21037/atm.2016.09.11.
8. Vittal, A., and Ghany, M. G. (2019) WHO Guidelines
for Prevention, Care and Treatment of Individuals In-
fected with HBV, Clinics in Liver Disease, 23, 417-432,
https://doi.org/10.1016/j.cld.2019.04.008.
9. Raimondo,G., Locarnini,S., Pollicino, T., Levrero,M.,
Zoulim,F., Lok, A.S., Allain, J.-P., Berg,T., Bertoletti,A.,
Brunetto, M. R., Bruno, R., Chen, D.-S., Coppola, N.,
Cornberg, M., Craxì, A., Dandri, M., Di Marco, V.,
Ferrari,C., Gaeta, G.B., Glebe,D., etal. (2019) Update
of the statements on biology and clinical impact of
occult hepatitisB virus infection, J.Hepatol., 71, 397-
408, https://doi.org/10.1016/j.jhep.2019.03.034.
10. Bock, C.-T., Schranz, P., Schröder, C. H., and Zent-
graf,H. (1994) HepatitisB virus genome is organized
into nucleosomes in the nucleus of the infected
cell, Virus Genes, 8, 215-229, https://doi.org/10.1007/
BF01703079.
11. Ghosh, S., Chakraborty, A., and Banerjee, S. (2021)
Persistence of hepatitis B virus infection: a multi-
faceted player for hepatocarcinogenesis, Front. Mi-
crobiol., 12, 678537, https://doi.org/10.3389/fmicb.
2021.678537.
12. Yan, H., Zhong, G., Xu, G., He, W., Jing, Z., Gao, Z.,
Huang, Y., Qi, Y., Peng, B., Wang, H., Fu, L., Song, M.,
Chen, P., Gao, W., Ren, B., Sun, Y., Cai, T., Feng, X.,
Sui,J., and Li,W. (2012) Sodium taurocholate cotrans-
porting polypeptide is a functional receptor for hu-
man hepatitisB andD virus, eLife, 1, e00049, https://
doi.org/10.7554/eLife.00049.
13. Schieck, A., Schulze, A., Gähler, C., Müller, T., Haber-
korn, U., Alexandrov, A., Urban, S., and Mier, W.
(2013) Hepatitis B virus hepatotropism is mediated
by specific receptor recognition in the liver and not
restricted to susceptible hosts, Hepatology, 58, 43-53,
https://doi.org/10.1002/hep.26211.
14. Barker, L.F., Maynard, J.E., Purcell, R.H., Hoofnagle,
J. H., Berquist, K. R., and London, W. T. (1975) Viral
hepatitis, typeB, in experimental animals, Am.J. Med.
Sci., 270, 189-195, https://doi.org/10.1097/00000441-
197507000-00026.
15. Shikata,T., Karasawa,T., and Abe, K. (1980) Two dis-
tinct types of hepatitis in experimental hepatitis B
virus infection, Am.J. Pathol., 99, 353-368.
16. Maynard, J. E., Berquist, K. R., Krushak, D. H., and
Purcell, R. H. (1972) Experimental infection of chim-
panzees with the virus of hepatitis B, Nature, 237,
514-515, https://doi.org/10.1038/237514a0.
17. Maynard, J. E., Krushak, D. H., Bradley, D. W., and
Berquist, K.R. (1975) Infectivity studies of hepatitisA
and B in non-human primates, Dev. Biol. Standard.,
30, 229-235.
18. Ibrahim, A. B., Vyas, G. N., and Prince, A. M. (1974)
Studies on delayed hypersensitivity to hepatitisB an-
tigen in chimpanzees, Clin. Exp. Immunol., 17, 311-
318.
19. Buynak, E. B., Roehm, R. R., Tytell, A. A., Bertland,
A. U., Lampson, G. P., and Hilleman, M. R. (1976)
Development and chimpanzee testing of a vaccine
against human hepatitis B, Exp. Biol. Med., 151, 694-
700, https://doi.org/10.3181/00379727-151-39288.
HEPATITIS B VIRUS INFECTION MODELS 1515
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
20. CDC (1982) Recommendation of the Immunization
Practices Advisory Committee (ACIP). Inactivated hep-
atitis B virus vaccine, Morbid. Mortal. Weekly Rep.,
31, 317-322.
21. Itoh,Y., Takai,E., Ohnuma,H., Kitajima,K., Tsuda, F.,
Machida,A., Mishiro,S., Nakamura,T., Miyakawa,Y.,
and Mayumi, M. (1986) A synthetic peptide vaccine
involving the product of the pre-S(2) region of hepa-
titisB virus DNA: protective efficacy in chimpanzees,
Proc. Natl. Acad. Sci. USA, 83, 9174-9178, https://
doi.org/10.1073/pnas.83.23.9174.
22. Tabor, E., Copeland, J. A., Mann, G. F., Howard, C. R.,
Skelly, J., Snoy, P., Zuckerman, A. J., and Gerety, R. J.
(1981) Nondetection of infectious hepatitis B virus
in a human hepatoma cell line producing hepati-
tisB surface antigen, Intervirology, 15, 82-86, https://
doi.org/10.1159/000149217.
23. Davis, H.L., McCluskie, M.J., Gerin, J.L., and Purcell,
R. H. (1996) DNA vaccine for hepatitis B: evidence
for immunogenicity in chimpanzees and comparison
with other vaccines, Proc. Natl. Acad. Sci. USA, 93,
7213-7218, https://doi.org/10.1073/pnas.93.14.7213.
24. McAleer, W.J., Buynak, E.B., Maigetter, R.Z., Wampler,
D.E., Miller, W.J., and Hilleman, M.R. (1984) Human
hepatitis B vaccine from recombinant yeast, Nature,
307, 178-180, https://doi.org/10.1038/307178a0.
25. Guidotti, L. G., Rochford, R., Chung, J., Shapiro, M.,
Purcell, R., and Chisari, F. V. (1999) Viral clearance
without destruction of infected cells during acute
HBV infection, Science, 284, 825-829, https://doi.org/
10.1126/science.284.5415.825.
26. Gheit, T., Sekkat, S., Cova, L., Chevallier, M., Petit,
M. A., Hantz, O., Lesénéchal, M., Benslimane, A.,
Trépo, C., and Chemin, I. (2002) Experimental trans-
fection of Macaca sylvanus with cloned human hep-
atitis B virus, J. Gen. Virol., 83, 1645-1649, https://
doi.org/10.1099/0022-1317-83-7-1645.
27. Dupinay, T., Gheit, T., Roques, P., Cova, L., Chevallier-
Queyron, P., Tasahsu, S.-i., Le Grand, R., Simon, F.,
Cordier, G., Wakrim, L., Benjelloun, S., Trépo, C.,
and Chemin, I. (2013) Discovery of naturally occur-
ring transmissible chronic hepatitisB virus infection
among Macaca fascicularis from mauritius island,
Hepatology, 58, 1610-1620, https://doi.org/10.1002/
hep.26428.
28. Burwitz, B. J., Wettengel, J. M., Mück-Häusl, M. A.,
Ringelhan,M., Ko,C., Festag, M.M., Hammond, K.B.,
Northrup, M., Bimber, B. N., Jacob, T., Reed, J. S.,
Norris,R., Park,B., Moller-Tank,S., Esser, K., Greene,
J.M., Wu, H.L., Abdulhaqq,S., Webb,G., Sutton, W.F.,
etal. (2017) Hepatocytic expression of human sodium-
taurocholate cotransporting polypeptide enables hep-
atitisB virus infection of macaques, Nat. Commun., 8,
2146, https://doi.org/10.1038/s41467-017-01953-y.
29. Kriegs, J. O., Churakov, G., Jurka, J., Brosius, J., and
Schmitz,J. (2007) Evolutionary history of 7SL RNA-de-
rived SINEs in supraprimates, Trends Genet., 23, 158-
161, https://doi.org/10.1016/j.tig.2007.02.002.
30. Walter, E., Keist, R., Niederöst, B., Pult, I., and Blum,
H. E. (1996) Hepatitis B virus infection of tupaia he-
patocytes in vitro and in vivo, Hepatology, 24, 1-5,
https://doi.org/10.1002/hep.510240101.
31. Wang, Q., Schwarzenberger, P., Yang, F., Zhang, J.,
Su,J., Yang,C., Cao,J., Ou,C., Liang,L., Shi,J., Yang,F.,
Wang, D., Wang, J., Wang, X., Ruan, P., and Li, Y.
(2012) Experimental chronic hepatitis B infection
of neonatal tree shrews (Tupaia belangeri chinen-
sis): A model to study molecular causes for suscep-
tibility and disease progression to chronic hepatitis
in humans, Virol. J., 9, 170, https://doi.org/10.1186/
1743-422X-9-170.
32. Yang, C., Ruan, P., Ou, C., Su, J., Cao, J., Luo, C.,
Tang, Y., Wang, Q., Qin, H., Sun, W., and Li, Y. (2015)
Chronic hepatitis B virus infection and occurrence
of hepatocellular carcinoma in tree shrews (Tupaia
belangeri chinensis), Virol. J., 12, 26, https://doi.org/
10.1186/s12985-015-0256-x.
33. Lempp, F.A., Wiedtke,E., Qu,B., Roques,P., Chemin,I.,
Vondran, F. W. R., Le Grand, R., Grimm, D., and
Urban, S. (2017) Sodium taurocholate cotransporting
polypeptide is the limiting host factor of hepatitis B
virus infection in macaque and pig hepatocytes, Hepa-
tology, 66, 703-716, https://doi.org/10.1002/hep.29112.
34. Messaoudi,I., Estep,R., Robinson,B., and Wong, S.W.
(2011) Nonhuman primate models of human immu-
nology, Antioxid. Redox Signal., 14, 261-273, https://
doi.org/10.1089/ars.2010.3241.
35. Biswas, S., Rust, L. N., Wettengel, J. M., Yusova, S.,
Fischer,M., Carson, J.N., Johnson,J., Wei,L., Thode,T.,
Kaadige, M.R., Sharma,S., Agbaria,M., Bimber, B.N.,
Tu,T., Protzer,U., Ploss,A., Smedley, J.V., Golomb,G.,
Sacha, J. B., and Burwitz, B. J. (2022) Long-term hep-
atitis B virus infection of rhesus macaques requires
suppression of host immunity, Nat. Commun., 13,
2995, https://doi.org/10.1038/s41467-022-30593-0.
36. Mason, W. S., Seal, G., and Summers, J. (1980) Virus
of Pekin ducks with structural and biological related-
ness to human hepatitisB virus, J.Virol., 36, 829-836,
https://doi.org/10.1128/jvi.36.3.829-836.1980.
37. Mandart, E., Kay, A., and Galibert, F. (1984) Nucleo-
tide sequence of a cloned duck hepatitis B virus ge-
nome: comparison with woodchuck and human hep-
atitisB virus sequences, J. Virol., 49, 782-792, https://
doi.org/10.1128/jvi.49.3.782-792.1984.
38. Foster, W.K., Miller, D.S., Scougall, C.A., Kotlarski,I.,
Colonno, R. J., and Jilbert, A. R. (2005) Effect of an-
tiviral treatment with entecavir on age- and dose-
related outcomes of duck hepatitis B virus infec-
tion, J. Virol., 79, 5819-5832, https://doi.org/10.1128/
JVI.79.9.5819-5832.2005.
39. Campagna, M. R., Liu, F., Mao, R., Mills, C., Cai, D.,
Guo,F., Zhao,X., Ye,H., Cuconati,A., Guo,H., Chang,J.,
KOLYAKO et al.1516
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Xu, X., Block, T. M., and Guo, J.-T. (2013) Sulfamoyl-
benzamide derivatives inhibit the assembly of hep-
atitis B virus nucleocapsids, J. Virol., 87, 6931-6942,
https://doi.org/10.1128/JVI.00582-13.
40. Schaefer, S. (2007) Hepatitis B virus taxonomy and
hepatitis B virus genotypes, World J. Gastroenterol.,
13, 14, https://doi.org/10.3748/wjg.v13.i1.14.
41. Tong, S., Li, J., and Wands, J. R. (1999) Carboxypep-
tidase D is an avian hepatitis B virus receptor, J. Vi-
rol., 73, 8696-8702, https://doi.org/10.1128/JVI.73.10.
8696-8702.1999.
42. Cova, L., Duflot, A., Prave, M., and Trepo, C. (1993)
Duck hepatitisB virus infection, aflatoxinB1 and liv-
er cancer in ducks, in Research in Chronic Viral Hep-
atitis (Gerlich, W. H., ed.), Springer Vienna, Vienna,
pp.81-87, https://doi.org/10.1007/978-3-7091-9312-9_9.
43. Lanford, R.E., Chavez, D., Brasky, K.M., Burns, R.B.,
and Rico-Hesse, R. (1998) Isolation of a hepadnavi-
rus from the woolly monkey, a New World primate,
Proc. Natl. Acad. Sci. USA, 95, 5757-5761, https://
doi.org/10.1073/pnas.95.10.5757.
44. Lanford, R. E., Chavez, D., Barrera, A., and Brasky,
K. M. (2003) An infectious clone of woolly monkey
hepatitis B virus, J. Virol., 77, 7814-7819, https://
doi.org/10.1128/JVI.77.14.7814-7819.2003.
45. Chen, C.Y., Winer, B.Y., Chavez,D., Guerra,B., Brasky,
K.M., Eng,S., Salas,E., Tam,D., Simmons, J.H., Abee,
C. R., Delaney, W. E., Ploss, A., Lanford, R. E., and
Voitenleitner,C. (2020) Woolly monkey-HBV infection
in squirrel monkeys as a surrogate nonhuman pri-
mate model of HBV infection, Hepatol. Commun., 4,
371-386, https://doi.org/10.1002/hep4.1471.
46. Mulrooney-Cousins, P. M., and Michalak, T. I. (2015)
Asymptomatic hepadnaviral persistence and its con-
sequences in the Woodchuck model of occult hep-
atitis B virus infection, J.Clin. Translat. Hepatol.,
3, 211-219, https://doi.org/10.14218/JCTH.2015.00020.
47. Summers,J., Smolec, J.M., and Snyder,R. (1978) Avi-
rus similar to human hepatitis B virus associated
with hepatitis and hepatoma in woodchucks, Proc.
Natl. Acad. Sci. USA, 75, 4533-4537, https://doi.org/
10.1073/pnas.75.9.4533.
48. Hodgson, P. D., and Michalak, T. I. (2001) Augment-
ed hepatic interferon gamma expression and T-cell
influx characterize acute hepatitis progressing to re-
covery and residual lifelong virus persistence in ex-
perimental adult woodchuck hepatitis virus infection,
Hepatology, 34, 1049-1059, https://doi.org/10.1053/
jhep.2001.29004.
49. Korba, B. E., Wells, F. V., Baldwin, B., Cote, P. J.,
Tennant, B. C., Popper, H., and Gerin, J. L. (1989) He-
patocellular carcinoma in woodchuck hepatitis vi-
rus-infected woodchucks: Presence of viral DNA in
tumor tissue from chronic carriers and animals sero-
logically recovered from acute infections, Hepatology,
9, 461-470, https://doi.org/10.1002/hep.1840090321.
50. Mason, W.S., Jilbert, A.R., and Summers,J. (2005) Clon-
al expansion of hepatocytes during chronic woodchuck
hepatitis virus infection, Proc. Natl. Acad. Sci. USA,
102, 1139-1144, https://doi.org/10.1073/pnas.0409332102.
51. Colonno, Richard J., Genovesi, Eugene V., Medina, I.,
Lamb,L., Durham,StephenK., Huang, M.L., Corey,L.,
Littlejohn,M., Locarnini,S., Tennant,BudC., Rose,B.,
and Junius, C. M. (2001) Long-term entecavir treat-
ment results in sustained antiviral efficacy and pro-
longed life span in the woodchuck model of chron-
ic hepatitis infection, J.Infect. Dis., 184, 1236-1245,
https://doi.org/10.1086/324003.
52. Wang, B.-J., Tian, Y.-J., Meng, Z.-J., Jiang, M., Wei,
B.-Q., Tao, Y.-Q., Fan, W., Li, A.-Y., Bao, J.-J., Li, X.-Y.,
Zhang, Z.-M., Wang, Z.-D., Wang, H., Roggendorf, M.,
Lu, M.-J., and Yang, D.-L. (2011) Establishing a new
animal model for hepadnaviral infection: susceptibil-
ity of Chinese Marmota-species to woodchuck hepati-
tis virus infection, J. Gen. Virol., 92, 681-691, https://
doi.org/10.1099/vir.0.025023-0.
53. Zhu, B., Zhu, Z., Wang, J., Huang, S., Li, F., Wang, L.,
Liu, Y., Yan, Q., Zhou, S., Lu, M., Yang, D., and
Wang, B. (2018) Chinese woodchucks with different
susceptibility to WHV infection differ in their ge-
netic background exemplified by cytochrome B and
MHC-DRB molecules, Virol.J., 15, 101, https://doi.org/
10.1186/s12985-018-1010-y.
54. Chisari, F. V., Pinkert, C. A., Milich, D. R., Filippi, P.,
McLachlan, A., Palmiter, R. D., and Brinster, R. L.
(1985) Atransgenic mouse model of the chronic hep-
atitis B surface antigen carrier state, Science, 230,
1157-1160, https://doi.org/10.1126/science.3865369.
55. Milich, D.R., Jones, J.E., Hughes, J.L., Price,J., Raney,
A. K., and McLachlan, A. (1990) Is a function of the
secreted hepatitis B e antigen to induce immunolog-
ic tolerance in utero? Proc. Natl. Acad. Sci. USA, 87,
6599-6603, https://doi.org/10.1073/pnas.87.17.6599.
56. Kim, C.-M., Koike, K., Saito, I., Miyamura, T., and
Jay, G. (1991) HBx gene of hepatitis B virus induces
liver cancer in transgenic mice, Nature, 351, 317-320,
https://doi.org/10.1038/351317a0.
57. Guidotti, L. G., Matzke, B., Schaller, H., and Chisari,
F. V. (1995) High-level hepatitis B virus replication
in transgenic mice, J. Virol., 69, 6158-6169, https://
doi.org/10.1128/jvi.69.10.6158-6169.1995.
58. Akbar, S. M. F., and Onji, M. (1998) Hepatitis B vi-
rus (HBV)-transgenic mice as an investigative tool to
study immunopathology during HBV infection, Int. J.
Exp. Pathol., 79, 279-291, https://doi.org/10.1046/
j.1365-2613.1998.740406.x.
59. Crawford, D. R., Ostrowski, S., Vakharia, D., Ilic, Z.,
and Sell, S. (2006) Separate origins of hepatitis B
virus surface antigen-negative foci and hepatocellu-
lar carcinomas in transgenic HBsAg (alb/psx) mice,
Am. J. Pathol., 169, 223-232, https://doi.org/10.2353/
ajpath.2006.051284.
HEPATITIS B VIRUS INFECTION MODELS 1517
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
60. Isogawa, M., Chung, J., Murata, Y., Kakimi, K., and
Chisari, F. V. (2013) CD40 activation rescues antiviral
CD8
+
T cells from PD-1-mediated exhaustion, PLoS
Pathog., 9, e1003490, https://doi.org/10.1371/journal.
ppat.1003490.
61. Tian, Y., Kuo, C.-F., Akbari, O., and Ou, J.-H. J. (2016)
Maternal-derived Hepatitis B Virus E antigen alters
macrophage function in offspring to drive viral persis-
tence after vertical transmission, Immunity, 44, 1204-
1214, https://doi.org/10.1016/j.immuni.2016.04.008.
62. Yan, H., Peng, B., He, W., Zhong, G., Qi, Y., Ren, B.,
Gao, Z., Jing, Z., Song, M., Xu, G., Sui, J., and Li, W.
(2013) Molecular determinants of hepatitis B and D
virus entry restriction in mouse sodium taurocholate
cotransporting polypeptide, J. Virol., 87, 7977-7991,
https://doi.org/10.1128/JVI.03540-12.
63. Huang, L. R., Gäbel, Y. A., Graf, S., Arzberger, S.,
Kurts, C., Heikenwalder, M., Knolle, P. A., and
Protzer, U. (2012) Transfer of HBV genomes using
low doses of adenovirus vectors leads to persistent
infection in immune competent mice, Gastroenter-
ology, 142, 1447-1450.e1443, https://doi.org/10.1053/
j.gastro.2012.03.006.
64. Li, L., Li, S., Zhou, Y., Yang, L., Zhou, D., Yang, Y.,
Lu,M., Yang, D., and Song,J. (2017) Thedose of HBV
genome contained plasmid has a great impact on
HBV persistence in hydrodynamic injection mouse
model, Virol.J., 14, 205, https://doi.org/10.1186/s12985-
017-0874-6.
65. John Von Freyend, M., Untergasser, A., Arzberger, S.,
Oberwinkler, H., Drebber, U., Schirmacher, P., and
Protzer, U. (2011) Sequential control of hepatitis B
virus in a mouse model of acute, self-resolving hepa-
titisB: control of HBV infection in mice, J.Viral Hep-
atit., 18, 216-226, https://doi.org/10.1111/j.1365-2893.
2010.01302.x.
66. Huang, L.-R., Wu, H.-L., Chen, P.-J., and Chen, D.-S.
(2006) An immunocompetent mouse model for the
tolerance of human chronic hepatitis B virus infec-
tion, Proc. Natl. Acad. Sci. USA, 103, 17862-17867,
https://doi.org/10.1073/pnas.0608578103.
67. Li,G., Zhu,Y., Shao,D., Chang,H., Zhang,X., Zhou,D.,
Gao, Y., Lan, K., and Deng, Q. (2018) Recombinant
covalently closed circular DNA of hepatitis B virus
induces long-term viral persistence with chronic
hepatitis in a mouse model, Hepatology, 67, 56-70,
https://doi.org/10.1002/hep.29406.
68. Gorsuch, C. L., Nemec, P., Yu, M., Xu, S., Han, D.,
Smith, J., Lape, J., Van Buuren, N., Ramirez, R.,
Muench, R.C., Holdorf, M. M., Feierbach,B., Falls,G.,
Holt, J., Shoop, W., Sevigny, E., Karriker, F., Brown,
R. V., Joshi, A., Goodwin, T., et al. (2022) Targeting
the hepatitis B cccDNA with a sequence-specific
ARCUS nuclease to eliminate hepatitisB virus invivo,
Mol. Ther., 30, 2909-2922, https://doi.org/10.1016/
j.ymthe.2022.05.013.
69. Ilan, E., Burakova, T., Dagan, S., Nussbaum, O.,
Lubin, I., Eren, R., Ben-Moshe, O., Arazi, J., Berr, S.,
Neville,L., Yuen,L., Mansour, T.S., Gillard,J., Eid,A.,
Jurim, O., Shouval, D., Reisner, Y., and Galun, E.
(1999) The hepatitis B virus-trimera mouse: a model
for human Hbv infection and evaluation of anti-Hbv
therapeutic agents, Hepatology, 29, 553-562, https://
doi.org/10.1002/hep.510290228.
70. Feitelson, M.A., Clayton, M.M., Sun,B., and Schinazi,
R. F. (2007) Development of a novel mouse model
to evaluate drug candidates against hepatitis B vi-
rus, Antiviral Chem. Chemother., 18, 213-223, https://
doi.org/10.1177/095632020701800405.
71. Schinazi, R.F., Bassit,L., Clayton, M.M., Sun,B., Kohler,
J.J., Obikhod,A., Arzumanyan,A., and Feitelson, M.A.
(2012) Evaluation of single and combination thera-
pies with tenofovir disoproxil fumarate and emtric-
itabine in vitro and in a robust mouse model sup-
porting high levels of hepatitis B virus replication,
Antimicrob. Agents Chemother., 56, 6186-6191, https://
doi.org/10.1128/AAC.01483-12.
72. Brown, J. J., Parashar, B., Moshage, H., Tanaka, K. E.,
Engelhardt, D., Rabbani, E., Roy-Chowdhury, N., and
Roy-Chowdhury, J. (2000) A long-term hepatitis B vi-
remia model generated by transplanting nontumori-
genic immortalized human hepatocytes in Rag-2-de-
ficient mice, Hepatology, 31, 173-181, https://doi.org/
10.1002/hep.510310126.
73. Dandri, M., Burda, M. R., Török, E., Pollok, J. M.,
Iwanska, A., Sommer, G., Rogiers, X., Rogler, C. E.,
Gupta, S., Will, H., Greten, H., and Petersen, J. (2001)
Repopulation of mouse liver with human hepato-
cytes and in vivo infection with hepatitis B virus,
Hepatology, 33, 981-988, https://doi.org/10.1053/jhep.
2001.23314.
74. Meuleman, P., Libbrecht, L., De Vos, R., De Hempt-
inne,B., Gevaert,K., Vandekerckhove,J., Roskams,T.,
and Leroux-Roels, G. (2005) Morphological and bio-
chemical characterization of a human liver in a
uPA-SCID mouse chimera, Hepatology, 41, 847-856,
https://doi.org/10.1002/hep.20657.
75. Tsuge,M., Hiraga,N., Takaishi,H., Noguchi,C., Oga,H.,
Imamura, M., Takahashi, S., Iwao, E., Fujimoto, Y.,
Ochi, H., Chayama, K., Tateno, C., and Yoshizato, K.
(2005) Infection of human hepatocyte chimeric
mouse with genetically engineered hepatitis B virus,
Hepatology, 42, 1046-1054, https://doi.org/10.1002/
hep.20892.
76. Bissig, K.-D., Le, T. T., Woods, N.-B., and Verma, I. M.
(2007) Repopulation of adult and neonatal mice
with human hepatocytes: a chimeric animal model,
Proc. Natl. Acad. Sci. USA, 104, 20507-20511, https://
doi.org/10.1073/pnas.0710528105.
77. Bissig, K.-D., Wieland, S. F., Tran, P., Isogawa, M., Le,
T. T., Chisari, F. V., and Verma, I. M. (2010) Human
liver chimeric mice provide a model for hepatitis B
KOLYAKO et al.1518
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
and C virus infection and treatment, J.Clin. Invest.,
120, 924-930, https://doi.org/10.1172/JCI40094.
78. Long, K.R., Lomonosova,E., Li,Q., Ponzar, N.L., Villa,
J.A., Touchette,E., Rapp,S., Liley, R.M., Murelli, R.P.,
Grigoryan,A., Buller, R.M., Wilson,L., Bial,J., Sagartz,
J. E., and Tavis, J.E. (2018) Efficacy of hepatitis B vi-
rus ribonucleaseH inhibitors, a new class of replica-
tion antagonists, in FRG human liver chimeric mice,
Antiviral Res., 149, 41-47, https://doi.org/10.1016/
j.antiviral.2017.11.008.
79. Bility, M.T., Cheng,L., Zhang,Z., Luan,Y., Li,F., Chi,L.,
Zhang,L., Tu, Z., Gao, Y., Fu,Y., Niu, J., Wang,F., and
Su, L. (2014) Hepatitis B virus infection and immu-
nopathogenesis in a humanized mouse model: in-
duction of human-specific liver fibrosis and M2-like
macrophages, PLoS Pathog., 10, e1004032, https://
doi.org/10.1371/journal.ppat.1004032.
80. Strick-Marchand,H., Dusséaux,M., Darche,S., Hunting-
ton, N. D., Legrand, N., Masse-Ranson, G., Corcuff, E.,
Ahodantin,J., Weijer, K., Spits,H., Kremsdorf,D., and
Di Santo, J. P. (2015) A novel mouse model for sta-
ble engraftment of a human immune system and
human hepatocytes, PLoS One, 10, e0119820, https://
doi.org/10.1371/journal.pone.0119820.
81. Dusséaux, M., Masse-Ranson, G., Darche, S., Ahodan-
tin, J., Li, Y., Fiquet, O., Beaumont, E., Moreau, P.,
Rivière, L., Neuveut, C., Soussan, P., Roingeard, P.,
Kremsdorf,D., Di Santo, J.P., and Strick-Marchand,H.
(2017) Viral load affects the immune response to
HBV in mice with humanized immune system and
liver, Gastroenterology, 153, 1647-1661.e1649, https://
doi.org/10.1053/j.gastro.2017.08.034.
82. Yuan, L., Jiang, J., Liu, X., Zhang, Y., Zhang, L., Xin,J.,
Wu, K., Li, X., Cao, J., Guo, X., Shi, D., Li, J., Jiang, L.,
Sun,S., Wang,T., Hou,W., Zhang,T., Zhu,H., Zhang,J.,
Yuan, Q., et al. (2019) HBV infection-induced liv-
er cirrhosis development in dual-humanised mice
with human bone mesenchymal stem cell transplan-
tation, Gut, 68, 2044-2056, https://doi.org/10.1136/
gutjnl-2018-316091.
83. Gomez-Lechon,M., Donato,M., Castell,J., and Jover,R.
(2004) Human hepatocytes in primary culture: the
choice to investigate drug metabolism in man, Curr.
Drug Metab., 5, 443-462, https://doi.org/10.2174/
1389200043335414.
84. Yoneda, M., Hyun, J., Jakubski, S., Saito, S., Naka-
jima, A., Schiff, E. R., and Thomas, E. (2016) Hep-
atitis B virus and DNA stimulation trigger a rapid
innate immune response through NF-κB, J. Immu-
nol., 197, 630-643, https://doi.org/10.4049/jimmunol.
1502677.
85. Guillouzo, A. (1998) Liver cell models in in vitro
toxicology, Environ. Health Perspect., 106, 511-532,
https://doi.org/10.1289/ehp.98106511.
86. Xiang,C., Du,Y., Meng,G., Soon Yi,L., Sun,S., Song,N.,
Zhang, X., Xiao, Y., Wang, J., Yi, Z., Liu, Y., Xie, B.,
Wu, M., Shu, J., Sun, D., Jia, J., Liang, Z., Sun, D.,
Huang, Y., Shi, Y., et al. (2019) Long-term functional
maintenance of primary human hepatocytes invitro,
Science, 364, 399-402, https://doi.org/10.1126/science.
aau7307.
87. Gripon, P., Diot, C., Thézé, N., Fourel, I., Loreal, O.,
Brechot, C., and Guguen-Guillouzo, C. (1988) Hep-
atitis B virus infection of adult human hepatocytes
cultured in the presence of dimethyl sulfoxide, J.Vi-
rol., 62, 4136-4143, https://doi.org/10.1128/jvi.62.11.
4136-4143.1988.
88. Galle, P.R., Hagelstein,J., Kommerell,B., Volkmann,M.,
Schranz, P., and Zentgraf, H. (1994) In vitro experi-
mental infection of primary human hepatocytes with
hepatitis B virus, Gastroenterology, 106, 664-673,
https://doi.org/10.1016/0016-5085(94)90700-5.
89. Nakabayashi, H., Taketa, K., Miyano, K., Yamane, T.,
and Sato, J. (1982) Growth of human hepatoma cells
lines with differentiated functions in chemically de-
fined medium, Cancer Res., 42, 3858-3863.
90. Chang, C. M., Jeng, K. S., Hu, C. P., Lo, S. J., Su, T. S.,
Ting, L. P., Chou, C. K., Han, S. H., Pfaff, E., and
Salfeld, J. (1987) Production of hepatitis B virus
in vitro by transient expression of cloned HBV DNA
in a hepatoma cell line, EMBO J., 6, 675-680, https://
doi.org/10.1002/j.1460-2075.1987.tb04807.x.
91. Hoare, J., Henkler, F., Dowling, J. J., Errington, W.,
Goldin, R. D., Fish, D., and McGarvey, M. J. (2001)
Subcellular localisation of the X protein in HBV in-
fected hepatocytes, J.Med. Virol., 64, 419-426, https://
doi.org/10.1002/jmv.1067.
92. Elizalde, M. M., Tadey, L., Mammana, L., Quarleri,
J. F., Campos, R. H., and Flichman, D. M. (2021) Bi-
ological characterization of hepatitis B virus geno-
types: their role in viral replication and antigen ex-
pression, Front. Microbiol., 12, 758613, https://doi.org/
10.3389/fmicb.2021.758613.
93. Hui, E. K.-W., Chen, K.-L., and Lo, S. J. (1999) Hepati-
tisB virus maturation is affected by the incorporation
of core proteins having a C-terminal substitution of
arginine or lysine stretches, J. Gen. Virol., 80, 2661-
2671, https://doi.org/10.1099/0022-1317-80-10-2661.
94. Thompson, A. J., Colledge, D., Rodgers, S., Wilson, R.,
Revill, P., Desmond, P., Mansell, A., Visvanathan, K.,
and Locarnini, S. (2009) Stimulation of the inter-
leukin-1 receptor and Toll-like receptor 2 inhibits
hepatitis B virus replication in hepatoma cell lines
in vitro, Antiviral Ther., 14, 797-808, https://doi.org/
10.3851/IMP1294.
95. Chen, H.-C., Chou, C.-K., Sun, C.-M., and Yeh, S. F.
(1997) Suppressive effects of destruxin B on hepati-
tisB virus surface antigen gene expression in human
hepatoma cells, Antiviral Res., 34, 137-144, https://
doi.org/10.1016/S0166-3542(97)01031-0.
96. Phillips, S., Chokshi, S., Chatterji, U., Riva, A.,
Bobardt,M., Williams,R., Gallay,P., and Naoumov,N.V.
HEPATITIS B VIRUS INFECTION MODELS 1519
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
(2015) Alisporivir inhibition of hepatocyte cyclophil-
ins reduces HBV replication and hepatitis B surface
antigen production, Gastroenterology, 148, 403-414.
e407, https://doi.org/10.1053/j.gastro.2014.10.004.
97. Lin, S.-R., Yang, H.-C., Kuo, Y.-T., Liu, C.-J., Yang, T.-Y.,
Sung, K.-C., Lin, Y.-Y., Wang, H.-Y., Wang, C.-C., Shen,
Y.-C., Wu, F.-Y., Kao, J.-H., Chen, D.-S., and Chen, P.-J.
(2014) The CRISPR/Cas9 system facilitates clear-
ance of the intrahepatic HBV templates in vivo, Mol.
Ther. Nucleic Acids, 3, e186, https://doi.org/10.1038/
mtna.2014.38.
98. Li, F., Wang, Z., Hu, F., and Su, L. (2020) Cell Culture
Models and Animal Models for HBV Study, in Hepa-
titis B Virus Infection (Tang, H. ed.) Springer Singa-
pore, Singapore, pp. 109-135, https://doi.org/10.1007/
978-981-13-9151-4_5.
99. Sells, M.A., Chen, M.L., and Acs,G. (1987) Production
of hepatitis B virus particles in Hep G2 cells trans-
fected with cloned hepatitisB virus DNA, Proc. Natl.
Acad. Sci. USA, 84, 1005-1009, https://doi.org/10.1073/
pnas.84.4.1005.
100. Acs,G., Sells, M. A., Purcell, R. H., Price, P., Engle, R.,
Shapiro, M., and Popper, H. (1987) Hepatitis B virus
produced by transfected Hep G2 cells causes hepa-
titis in chimpanzees, Proc. Natl. Acad. Sci. USA, 84,
4641-4644, https://doi.org/10.1073/pnas.84.13.4641.
101. Ding, X.R., Yang, J., Sun, D.C., Lou, S. K., and Wang,
S. Q. (2008) Whole genome expression profiling of
hepatitis B virus-transfected cell line reveals the po-
tential targets of anti-HBV drugs, Pharmacogenom-
icsJ., 8, 61-70, https://doi.org/10.1038/sj.tpj.6500459.
102. Ladner, S. K., Otto, M. J., Barker, C. S., Zaifert, K.,
Wang, G. H., Guo, J. T., Seeger, C., and King, R. W.
(1997) Inducible expression of human hepatitis B vi-
rus (HBV) in stably transfected hepatoblastoma cells:
a novel system for screening potential inhibitors of
HBV replication, Antimicrob. Agents Chemother., 41,
1715-1720, https://doi.org/10.1128/AAC.41.8.1715.
103. Guo,H., Jiang, D., Zhou, T., Cuconati, A., Block, T. M.,
and Guo, J.-T. (2007) Characterization of the intra-
cellular deproteinized relaxed circular DNA of hep-
atitis B virus: an intermediate of covalently closed
circular DNA formation, J. Virol., 81, 12472-12484,
https://doi.org/10.1128/JVI.01123-07.
104. Cai, D., Mills, C., Yu, W., Yan, R., Aldrich, C. E.,
Saputelli, J. R., Mason, W. S., Xu, X., Guo, J.-T., Block,
T. M., Cuconati, A., and Guo, H. (2012) Identifica-
tion of disubstituted sulfonamide compounds as
specific inhibitors of hepatitis B virus covalently
closed circular DNA formation, Antimicrob. Agents
Chemother., 56, 4277-4288, https://doi.org/10.1128/
AAC.00473-12.
105. Gripon,P., Rumin,S., Urban,S., Le Seyec,J., Glaise,D.,
Cannie, I., Guyomard, C., Lucas, J., Trepo, C., and
Guguen-Guillouzo,C. (2002) Infection of a human hep-
atoma cell line by hepatitisB virus, Proc. Natl. Acad.
Sci. USA, 99, 15655-15660, https://doi.org/10.1073/
pnas.232137699.
106. Schulze,A., Mills,K., Weiss, T.S., and Urban,S. (2012)
Hepatocyte polarization is essential for the produc-
tive entry of the hepatitis B virus, Hepatology, 55,
373-383, https://doi.org/10.1002/hep.24707.
107. Hantz, O., Parent,R., Durantel,D., Gripon, P., Guguen-
Guillouzo,C., and Zoulim,F. (2009) Persistence of the
hepatitis B virus covalently closed circular DNA in
HepaRG human hepatocyte-like cells, J. Gen. Virol.,
90, 127-135, https://doi.org/10.1099/vir.0.004861-0.
108. Urban,S., Bartenschlager,R., Kubitz,R., and Zoulim,F.
(2014) Strategies to inhibit entry of HBV and HDV into
hepatocytes, Gastroenterology, 147, 48-64, https://
doi.org/10.1053/j.gastro.2014.04.030.
109. Veloso Alves Pereira,I., Buchmann,B., Sandmann,L.,
Sprinzl, K., Schlaphoff, V., Döhner, K., Vondran, F.,
Sarrazin, C., Manns, M. P., Pinto Marques Souza De
Oliveira, C., Sodeik, B., Ciesek, S., and Von Hahn, T.
(2015) Primary biliary acids inhibit hepatitis D vi-
rus (HDV) entry into human hepatoma cells ex-
pressing the sodium-taurocholate cotransporting
polypeptide (NTCP), PLoS One, 10, e0117152, https://
doi.org/10.1371/journal.pone.0117152.
110. Ni, Y., Lempp, F. A., Mehrle, S., Nkongolo, S.,
Kaufman,C., Fälth,M., Stindt,J., Königer,C., Nassal,M.,
Kubitz, R., Sültmann, H., and Urban, S. (2014) Hep-
atitis B and D viruses exploit sodium taurocholate
co-transporting polypeptide for species-specific entry
into hepatocytes, Gastroenterology, 146, 1070-1083.
e1076, https://doi.org/10.1053/j.gastro.2013.12.024.
111. König,A., Yang,J., Jo,E., Park, K.H.P., Kim,H., Than,
T. T., Song, X., Qi, X., Dai, X., Park, S., Shum, D., Ryu,
W.-S., Kim, J.-H., Yoon, S. K., Park, J. Y., Ahn, S. H.,
Han, K.-H., Gerlich, W. H., and Windisch, M.P. (2019)
Efficient long-term amplification of hepatitis B virus
isolates after infection of slow proliferating HepG2-
NTCP cells, J. Hepatol., 71, 289-300, https://doi.org/
10.1016/j.jhep.2019.04.010.
112. Köck, J., Nassal, M., MacNelly, S., Baumert, T. F.,
Blum, H. E., and Von Weizsäcker, F. (2001) Efficient
infection of primary tupaia hepatocytes with purified
human and woolly monkey hepatitis B virus, J. Vi-
rol., 75, 5084-5089, https://doi.org/10.1128/JVI.75.11.
5084-5089.2001.
113. Köck,J., Baumert, T.F., Delaney, W.E., Blum, H.E., and
Von Weizsäcker, F. (2003) Inhibitory effect of adefovir
and lamivudine on the initiation of hepatitis B virus
infection in primary tupaia hepatocytes, Hepatology,
38, 1410-1418, https://doi.org/10.1016/j.hep.2003.09.009.
114. Sanada, T., Tsukiyama-Kohara, K., Yamamoto, N.,
Ezzikouri,S., Benjelloun,S., Murakami,S., Tanaka,Y.,
Tateno, C., and Kohara, M. (2016) Property of hepa-
titis B virus replication in Tupaia belangeri hepato-
cytes, Biochem. Biophys. Res. Commun., 469, 229-235,
https://doi.org/10.1016/j.bbrc.2015.11.121.
KOLYAKO et al.1520
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
115. Glebe, D., Urban, S., Knoop, E. V., Çaǧ, N., Krass, P.,
Grün, S., Bulavaite, A., Sasnauskas, K., and Gerlich,
W. H. (2005) Mapping of the hepatitis B virus attach-
ment site by use of infection-inhibiting preS1 lipopep-
tides and tupaia hepatocytes, Gastroenterology, 129,
234-245, https://doi.org/10.1053/j.gastro.2005.03.090.
116. Sun, D., and Nassal, M. (2006) Stable HepG2- and
Huh7-based human hepatoma cell lines for efficient
regulated expression of infectious hepatitis B virus,
J. Hepatol., 45, 636-645, https://doi.org/10.1016/j.jhep.
2006.05.019.
117. Dandri, M., Burda, M. R., Zuckerman, D. M., Wurst-
horn, K., Matschl, U., Pollok, J. M., Rogiers, X.,
Gocht, A., Köck,J., Blum, H. E., Weizsäcker, F. V., and
Petersen, J. (2005) Chronic infection with hepatitis B
viruses and antiviral drug evaluation in uPA mice
after liver repopulation with tupaia hepatocytes,
J. Hepatol., 42, 54-60, https://doi.org/10.1016/j.jhep.
2004.09.021.
118. Petersen, J., Dandri, M., Mier, W., Lütgehetmann, M.,
Volz,T., Von Weizsäcker,F., Haberkorn,U., Fischer,L.,
Pollok, J.-M., Erbes, B., Seitz, S., and Urban, S. (2008)
Prevention of hepatitis B virus infection in vivo by
entry inhibitors derived from the large envelope
protein, Nat. Biotechnol., 26, 335-341, https://doi.org/
10.1038/nbt1389.
119. Lutgehetmann, M., Volz, T., Köpke, A., Broja, T.,
Tigges, E., Lohse, A. W., Fuchs, E., Murray, J. M.,
Petersen, J., and Dandri, M. (2010) In vivo prolifer-
ation of hepadnavirus-infected hepatocytes induces
loss of covalently closed circular DNA in mice, Hepa-
tology, 52, 16-24, https://doi.org/10.1002/hep.23611.
120. Chin,R., Earnest-Silveira,L., Koeberlein,B., Franz,S.,
Zentgraf, H., Dong, X., Gowans, E., Bock, C.-T., and
Torresi, J. (2007) Modulation of MAPK pathways and
cell cycle by replicating hepatitisB virus: factors con-
tributing to hepatocarcinogenesis, J.Hepatol., 47, 325-
337, https://doi.org/10.1016/j.jhep.2007.03.025.
121. Liu,Y., Cafiero, T.R., Park,D., Biswas,A., Winer, B.Y.,
Cho, C. H., Bram, Y., Chandar, V., Connell, A. K. O.,
Gertje, H. P., Crossland, N., Schwartz, R. E., and
Ploss,A. (2023) Targeted viral adaptation generates a
simian-tropic hepatitisB virus that infects marmoset
cells, Nat. Commun., 14, 3582, https://doi.org/10.1038/
s41467-023-39148-3.
122. Aldrich, C. E., Coates, L., Wu, T.-T., Newbold, J.,
Tennant, B. C., Summers, J., Seeger, C., and Mason,
W. S. (1989) In vitro infection of woodchuck hepato-
cytes with woodchuck hepatitis virus and ground
squirrel hepatitis virus, Virology, 172, 247-252,
https://doi.org/10.1016/0042-6822(89)90126-8.
123. Moraleda, G., Saputelli, J., Aldrich, C. E., Averett, D.,
Condreay, L., and Mason, W. S. (1997) Lack of effect
of antiviral therapy in nondividing hepatocyte cul-
tures on the closed circular DNA of woodchuck hep-
atitis virus, J. Virol., 71, 9392-9399, https://doi.org/
10.1128/jvi.71.12.9392-9399.1997.
124. Tuttleman, J.S., Pugh, J.C., and Summers, J.W. (1986)
In vitro experimental infection of primary duck he-
patocyte cultures with duck hepatitisB virus, J.Virol.,
58, 17-25, https://doi.org/10.1128/jvi.58.1.17-25.1986.
125. Tavis, J.E., and Ganem,D. (1996) Evidence for activa-
tion of the hepatitis B virus polymerase by binding
of its RNA template, J. Virol., 70, 5741-5750, https://
doi.org/10.1128/jvi.70.9.5741-5750.1996.
126. Tuttleman, J. S., Pourcel, C., and Summers, J. (1986)
Formation of the pool of covalently closed circular
viral DNA in hepadnavirus-infected cells, Cell, 47, 451-
460, https://doi.org/10.1016/0092-8674(86)90602-1.
127. Junker-Niepmann, M., Bartenschlager, R., and
Schaller, H. (1990) A short cis-acting sequence is re-
quired for hepatitis B virus pregenome encapsida-
tion and sufficient for packaging of foreign RNA,
EMBO J., 9, 3389-3396, https://doi.org/10.1002/j.1460-
2075.1990.tb07540.x.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.