1. Introduction
Hepatitis B virus (HBV) is a global health problem affecting more than 250 million people who are chronically infected [
1]. Despite the availability of effective prophylactic vaccines, chronic hepatitis B (CHB) infection still resulted in more than 800,000 deaths a year due to HBV-associated liver failure or hepatocellular carcinoma [
2,
3]. It has been well documented that approximately 90% of the infected adult patients are able to mount a robust immune response that is sufficient to control HBV infection; in addition, each year about 2–3% of CHB patients can spontaneously seroconvert to anti-HBs positive subjects and control their infection [
3]. Current therapies against CHB infection include potent direct-acting antivirals and pegylated interferon (IFN) alpha. However, there is the risk of viral resistance, drug toxicity, and side effects. Furthermore, these therapies rarely achieve functional cure as defined by hepatitis B surface antigen (HBsAg) loss and anti-HBs seroconversion [
4]. These facts encourage the development of immunotherapies for the treatment of CHB. Key elements for controlling HBV infection are the induction of neutralizing antibodies against HBV, further leading to its clearance, and the induction of a broad and robust antigen-specific T cell response. However, the ongoing HBV replication together with the high concentrations of circulating HBsAg in CHB patients causes a general hypo-responsiveness of the immune system to the persisting virus [
5]. In these patients, HBV-specific T cells are deleted or functionally exhausted most likely due to the repeated exposure to a high antigen load. Likewise, it has been shown that therapeutic vaccine-induced antibody responses are inhibited due to the presence of persistently high HBsAg levels [
6]. Therefore, the major objective of a therapeutic vaccination against CHB is to restore the functionality of antiviral B-cell and T-cell responses.
The HBsAg gene includes preS1, preS2, and S domains. Its translation may result in formation of the large (LHBs), the medium (MHBs) HBsAg proteins, containing preS1 + preS2 + S domains or preS2 + S, respectively, or the small (SHBs) surface protein only containing the S domain [
1].
Therapeutic vaccination for CHB had not yet demonstrated clear efficacy in clinical trials, which could be attributed to suboptimal vaccine design based mainly on SHBs [
7]. The main obstacle in CHB is the large quantities of HBsAg forming subviral particles (SVPs) mostly composed of SHBs, which has been proposed to cause antigen-specific immune tolerance. Alternative HBV vaccine candidates other than exclusively SHBs-based are eventually promising therapeutic vaccines [
8]. The preS1 domain serves as a potential target for HBV vaccination because of its key role in hepatocyte entry via the sodium taurocholate co-transporting polypeptide (NTCP) receptor [
9] and in the assembly and release of HBV virions [
10]. More importantly, preS1 exists primarily in mature infectious HBV virions, and the antigen availability is much lower than that of HBsAg [
11], thus having the potential to overcome immune tolerance to HBV. Inhibition of the preS-NTCP interaction is likely to efficiently prevent re-infection and block de novo infections of naïve hepatocytes or those that arise during hepatocyte turnover. Therefore, a blockade of the preS-NTCP interaction may reduce the number of infected cells while accelerating the process of virus elimination.
When used in allergy vaccines as an immunological carrier, preS in combination with allergen-derived peptides was shown to be highly immunogenic [
12]. One of the recombinant peptide carrier vaccines, VVX001, is composed of the preS (preS1 + preS2) of the LHBs fused to peptide sequences from the grass pollen allergen Phl p 5 at both ends [
13]. We have previously shown that by vaccination with VVX001, a potent IgG response to preS could be elicited even in non-responders to HBsAg-based prophylactic vaccines [
14]. The antibody response is strongly focused on the sequence motif in the preS1 domain responsible for HBV entry in hepatocytes [
15]; moreover, sera of subjects immunized with VVX001 efficiently inhibited HBV infection in vitro [
12]. Furthermore, escape mutations seem to be less problematic for preS as compared to the HBsAg [
5,
16,
17].
These findings encouraged us to study the potential of the preS-based vaccine VVX001 in reversing immune tolerance in CHB in an animal model. Chimpanzees are the only nonhuman immune-competent animals that are naturally susceptible to chronic HBV infection, but this model is difficult to access, and there are ethical considerations. Mice have well-characterized immune systems and are widely used in research, but they are naturally not susceptible to HBV. Therefore, various strategies have been exploited to introduce the HBV genome into the hepatocytes of mice. One of the delivery methods is to introduce a replication-competent HBV genome into the mouse liver by hydrodynamic injection through the tail vein; even though HBV replicates in the mouse liver, the virus is cleared rapidly by the immune responses against HBV proteins [
18]. An alternative method uses an adenoviral or adeno-associated viral vector to transfer copies of the HBV genome into immunocompetent mice. Depending on the dose of the vector injected, acute or chronic HBV infection can be induced [
19,
20]. Here we used the chronic HBV infection murine model established by transducing the liver with an adeno-associated virus (AAV) serotype 2/8 carrying a replication-competent HBV-DNA genome which resulted in a long-term chronic HBV-carrier state [
21]. The aim of this study was to evaluate if immunization with VVX001 or preS can induce an HBV-specific immune response in the AAV-HBV mouse model of CHB.
2. Materials and Methods
2.1. Animal Experiments
C57BL/6 female mice were purchased from Janvier Labs (Le Genest-Saint-Isle, France). Mice were housed under pathogen-free conditions and used at 6–8 weeks of age. The mice were housed in groups and provided with appropriate bedding. They were subjected to standardized light–dark cycles and given ad libitum access to food and water. The scheme of the experiment is depicted in
Figure 1. After a one-week acclimatization period, the livers of mice were transduced with either an AAV serotype 2/8 carrying a replication-competent HBV DNA genome (3 groups of 6 mice in each) or an AAV-Empty vector (3 groups of 6 mice in each). The sample size of six mice per group was determined based on the results of our previous study in order to achieve statistically significant differences between the groups, particularly in the case of partial control [
12]. The HBV genome was introduced by i.v. injection with 5 × 10
10 viral genome equivalents (vg) of AAV2/8-HBV in the tail vein as described [
21]. Serum samples were obtained from mice at baseline, shortly before the first immunization, on the days of the second to fifth injection and two weeks after the last injection (
Figure 1). Five immunizations with three week intervals were performed by subcutaneous (s.c.) injections of 20 μg of VVX001 or preS adsorbed onto aluminum hydroxide under the scruff of the neck without anesthesia. Control mice received an identical volume of aluminum hydroxide (vehicle, 200 μL). Following the AAV injections and vaccination, animals were monitored at least three times per week for any signs of discomfort. Mice exhibiting any of the following humane endpoints were excluded from the analysis: progressive loss of activity; abnormal posture; signs of infection; or weight loss of more than 15%. All protocols have been reviewed and approved by the institutional animal care committee of Institute Pasteur for compliance with the French and European regulations on animal welfare and with public health service recommendations (authorization number No. 02651.02).
2.2. Vectors and Immunogens
An AAV serotype 2/8, either carrying a replication-competent HBV DNA genome (AAV-HBV) or not (AAV-Empty), were used. The AAV-HBV (batch GVPN #6565) and AAV-Empty (batch GVPN #5198) vectors were stored at −80 °C at a concentration of 1.2 × 10
13 and 1.0 × 10
13 viral genomes (vg)/mL, respectively. Before use, AAV vectors were thawed at room temperature and then diluted in sterile phosphate-buffered saline (PBS) to reach a titer of 5 × 10
11 vg/mL. Mice were injected intravenously (i.v.) with 100 μL of this solution (dose/mouse: 5 × 10
10 vg) (
Figure 1).
VVX001 is a recombinant fusion protein composed of the preS (preS1 + preS2, GenBank: AAT28735) domain of the large surface antigen (LHBs) of HBV genotype A2 and peptides derived from the grass pollen allergen Phl p 5 at both the C- and N-terminus. VVX001 is one of the components of the grass pollen allergy vaccine BM32, namely BM325; a detailed description of the structure and peptide sequences has been reported [
13]. As an immunogen of comparison, preS (preS + preS2, genotype A2, GenBank: AAT28735) alone containing a C-terminal hexahistidine tag was expressed in
E. coli and purified as described [
12].
2.3. Preparation of Cells
Spleen and liver mononuclear cells as well as blood samples from each mouse were collected 2 weeks after the last immunization and depleted of red blood cells using Lysing Buffer (BD Biosciences, Franklin Lakes, NJ, USA, Cat. 555899). The liver mononuclear cells underwent a specific preparation as described below and according to a method previously published by Tupin et al. [
22] with minor modifications. After mouse euthanasia, the liver was perfused with 10 mL of sterile PBS via the hepatic portal vein using a syringe with a G25 needle until the organ was becoming pale, then it was harvested in Hank’s Balanced Salt Solution (HBSS) (Gibco, Thermo Fisher Scientific, Waltham, MA, USA, Cat. 24020) +5% decomplemented fetal calf serum (FCS). Afterward, the liver was gently pressed through a 100 μm cell strainer (BD Falcon, Franklin Lakes, NJ, USA, Cat. 352360), and cells were suspended in 30 mL of HBSS + 5% FCS. Cell suspension was centrifuged at 50×
g for 5 min, and then the supernatants were centrifuged at 289×
g for 10 min at 4 °C. After centrifugation, supernatants were discarded and pellets were re-suspended in 15 mL of 35% isotonic Percoll solution (GE Healthcare, Chicago, IL, USA, Cat. #17-0891-01) diluted into RPMI 1640 (Gibco, Thermo Fisher Scientific, Cat. 31870) at room temperature and transferred in 15 mL tubes. Cells were further centrifuged at 1360×
g for 25 min at room temperature. Afterward, the supernatant was discarded by aspiration and the pellet containing mononuclear cells was washed twice with HBSS + 5% FCS. Cells were then resuspended in complete medium (α-minimal essential medium (Gibco, Thermo Fisher Scientific, Cat. 22571) supplemented with 10% FCS (Hyclone, Thermo Fisher Scientific, Cat. SH30066), 100 U/mL penicillin + 100 μg/mL streptomycin + 0.3 mg/mL L-glutamine (Gibco, Thermo Fisher Scientific, Cat. 10378), 1X non-essential amino acids (Gibco, Thermo Fisher Scientific, Cat. 11140), 10 mM Hepes (Gibco, Thermo Fisher Scientific, Cat. 15630), 1 mM sodium pyruvate (Gibco, Thermo Fisher Scientific, Cat. 11360) and 50 μM β-mercaptoethanol (LKB, Piscataway, NJ, USA, Cat. 1830).
2.4. B Cell Enzyme-Linked Immunospot (ELISPOT) Assays
Antibody-producing B cells amongst splenocytes were quantified by ELISPOT assay after antigen stimulation, as previously described. To determine the number of antibody-secreting cells (ASC), sterile MSIP 96-well plates (Millipore, Bedford, MA, USA) were pre-wetted for 1 min with 15 μL 35% ethanol, washed with water, coated with MHBs (preS2 + S, genotype D), HBcAg (genotype A) or preS, and incubated overnight at 4 °C. Wells of ELISPOT plates were also coated with an anti-mouse IgG coating antibody that binds to the antibodies released by the antibody-secreting plasma cells as positive controls, and keyhole limpet hemocyanin (KLH) was used as a negative control to determine background levels. Wells were post-coated with a blocking agent for 2 h at 37 °C and subsequently incubated with cell preparations (5 × 105 cells/well). After an overnight incubation, cells were washed away, and the antibodies captured by the immobilized antigen or antibody were detected with biotinylated anti-mouse IgG antibodies followed by incubation with a phosphatase alkaline-labeled detection antibody. Revelation was performed by adding the BCIP/NBT substrate. The response to the stimulant was considered positive if the median number of antibody-secreting cells (ASC) in triplicate wells was at least twice as much as observed in control wells stimulated by KLH and when at least 10 antibody-secreting cells per million splenocytes were detected after subtraction of the background.
2.5. Cell Labeling and Intracellular Cytokine Staining (ICS)
Due to the limited number of lymphocytes collected from the liver, all cells were used for the experiment and divided according to the number of tested parameters. As a consequence, the number of cells/well in the same experiment is identical for the same mouse but variable among different mice.
The detection of cell populations was performed by surface labeling of purified liver mononuclear cells: the cells were seeded in U-bottom 96-well plates and washed with PBS FACS (PBS containing 1% bovine serum albumin and 0.01% sodium azide). Cells were then incubated with 5 μL of PBS FACS containing a rat anti-mouse CD16/CD32 antibody and a viability marker (LD fixable yellow, Thermo Fisher Scientific, Cat. L34959) for 10 min in the dark at 4 °C. Then, cells were stained for 20 min in the dark at 4 °C with 25 μL of PBS FACS containing a mix of monoclonal antibodies (Mab). The mix was composed of hamster Mab anti-mouse CD3-PEVio770 (Miltenyi, Bergisch Gladbach, Germany, Cat. 130-102-359), CD8 (rat Mab anti-mouse CD8-APC-H7, BD Biosciences, Cat. 560182), rat Mab anti-mouse CD4-AF647 (BD Biosciences, Cat. 557681), rat Mab anti-mouse PE-Cy5.5 (eBioscience, Thermo Fisher Scientific, Cat. 35-0193-80), rat Mab anti-mouse NK P46 (BioLegend, San Diego, CA, USA, Cat. 137612) and rat Mab anti-mouse F4/80 FITC (BioLegend, Cat. 123108). After washes, cells were fixed in PBS FACS containing 1% formaldehyde, washed, and resuspended in PBS FACS. Afterward, analysis by flow cytometry using an Attune (ThermoFisher, Waltham, MA, USA) analyzer was performed.
ICS assays were conducted on both splenocytes and liver mononuclear cells. Cells were seeded in U-bottom 96-well plates which were then incubated overnight at 37 °C either in complete medium alone as a negative control or in 3 pools of peptides from the HBc, MHBs, and preS proteins at a concentration of 2 µg/mL. After one hour of incubation, Brefeldin A at 2 µg/mL (B6542, Sigma-Aldrich, Burlington, MA, USA) was added to the cells. Cells were then cultured overnight, then washed with PBS FACS and incubated with 5 µL of PBS FACS containing rat anti-mouse CD16/CD32 antibody and a viability marker (Zombie Violet Fixable Viability Kit, BioLegend, BLE423114) for 10 min in the dark at 4 °C. Afterward, cells were stained for 20 min in the dark at 4 °C with 25 µL of PBS FACS containing a mix of Mab. The mix was composed of hamster Mab anti-mouse CD3-PerCP-Vio700 (Miltenyi, 130-119-656), rat Mab anti-mouse CD8-APCH7 (BD Biosciences, 560182), rat Mab anti-mouse CD4-PE-Cy7 (BD Biosciences, 552775). After several washes, cells were fixed and permeabilized for 20 min in the dark at room temperature with Cytofix/Cytoperm and washed with Perm/Wash solution (BD Biosciences, 554714) at 4 °C. ICS with antibodies against IFNγ (rat Mab anti-mouse IFN-APC, clone XMG1.2, BD Biosciences, 554413), interleukin (IL)-2 (rat Mab anti-mouse IL2-PE, clone JES6-5H4, BD Biosciences, 554428), and tumor necrosis factor alpha (TNF-alpha) (rat Mab anti-mouse TNFa-FITC, clone MP6-XT22; 1/250 (BD Biosciences 554418) was performed for 30 min in the dark at 4 °C. Cells were washed with Perm/Wash and resuspended in PBS FACS and then analyzed by flow cytometry using the MACSQuant Analyzer. Live CD3+CD8+CD4− and CD3+CD8−CD4+ cells were gated and presented on a dot-plot. Two regions were defined to gate for positive cells for each cytokine; numbers of events found in these gates were divided by the total number of events in the parental population to yield percentages of responding T cells. The percentage obtained in medium alone was considered as background and subtracted from the percentage obtained with peptide stimulations. The threshold of positivity was defined according to experiment background, i.e., the mean percentage of stained cells obtained for each group in medium-alone condition plus two standard deviations (SD). Only the percentage of cytokine representing at least 50 events was considered as positive.
2.6. ELISA
Eight synthetic peptides representing HBV genotypes A–H were synthesized [
15]; their sequences are provided in
Table S1. Peptides A, B, and A + B contained additional cysteine residues at the N-terminus for coupling. Each peptide was dissolved in sterile ddH
2O at a concentration of 1 mg/mL and was tested at a final concentration of 2 μg/mL with enzyme-linked immunosorbent assay (ELISA).
E. coli-expressed preS was used in ELISA at a final concentration of 2 μg/mL.
Sera were collected at different time points and stored at −20 °C. HBsAg and HBeAg levels in mouse sera were measured with commercial ELISA kits, respectively (Bio-Rad, Hercules, CA, USA and Diasorin SA, Antony, France). HBsAg concentrations were calculated in international units per milliliter (IU/mL) by reference to a standard curve established with known concentrations of HBsAg (Bio-Rad). Serum HBeAg levels were determined in hundred-fold diluted sera. Concentrations were calculated by reference to a standard curve established with the Paul-Ehrlich-Institut standard and are expressed in PEI U/mL.
For determination of specific IgG1 and IgG2a, ELISA plates (Nunc MaxiSorp 96-well flat bottom, Thermo Fisher Scientific, Cat. 442404) were coated overnight at 4 °C with preS or preS-derived peptides (2 µg/mL in 100 mM sodium bicarbonate buffer, pH 9.6) and washed two times with PBS 0.05% Tween 20 (PBST). Residual binding sites were blocked for 5 h at room temperature with 2% BSA/PBST, then the plates were incubated overnight at 4 °C with sera diluted 1:100, 1:500, 1:1000, 1:1500, 1:2000, 1:4000, 1:6000, 1:8000, and 1:10,000 for IgG1 and 1:50, 1:250, 1:500, 1:1000, 1:1500, 1:2000, 1:4000, 1:6000, and 1:8000 for IgG2a measurements in 0.5% BSA/PBST. After five times washing with PBST, rat anti-mouse IgG1 (BD, Cat. 553440) and IgG2a (BD, Cat. 553387) were diluted 1:1000 in 0.5% BSA/PBST and applied to the plate; after 2 h incubation at room temperature and five times washing, plates were incubated for 1 h at room temperature with horseradish-peroxidase (HRP)-linked goat anti-rat IgG (GE Healthcare, Cat. NA931V) at a dilution of 1:2000. After five times washing, the reaction was developed by incubation with substrate solution: 1 mg/mL 2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) di-ammonium salt (ABTS) (Cat. A1888, Sigma-Aldrich, Burlington, MA, USA) in 70 mM citrate-phosphate buffer containing 0.003% H2O2 (Sigma-Aldrich, Cat.H1009). Absorbance values were measured at wavelength 405 nm (reference wavelength 492 nm) on a Tecan Infinite F50 spectrophotometer (Tecan Trading AG, Männedorf, Switzerland). The endpoint titration cut-off was determined as 3 × (mean + SD) values in the sera of the AAV-Empty + Vehicle group at a dilution of 1:100 for IgG1 and 1:50 for IgG2a.
2.7. ALAT/ASAT Measurement
Alanine aminotransferase (ALAT) and aspartate aminotransferase (ASAT) activities in sera of AAV-HBV- and AAV-Empty-injected mice were determined by the Laboratoire de Biologie Vétérinaire Vebiotel (Paris, France). Results are expressed in IU/liter.
2.8. HBV DNA Titration
Virions were purified from mouse sera by using a DNA purification kit (QIAamp Blood Mini kit, Qiagen, Hilden, Germany) and quantified by quantitative PCR as previously described [
23]. Serial dilutions of the payw1.2 plasmid containing 1.2 copies of the HBV genome were used as quantification standards. The threshold of detection was 1020 IU/mL.
2.9. Statistical Analysis
Data were expressed as means ± standard errors of the means (SEM). Data was assumed to have a non-Gaussian distribution. When comparing groups, a Kruskal–Wallis test followed by a Dunn post hoc test was performed. The Mann–Whitney U test was used for all comparisons of two data sets. Statistical analysis was carried out using GraphPad Prism 8 software (Graphpad, San Diego, CA, USA).
4. Discussion
We have previously shown that the recombinant grass pollen allergy vaccine BM32, which is composed of four recombinant fusion proteins consisting of HBV-derived preS as a carrier and peptides from the four major grass pollen allergens, Phl p 1, Phl p 2, Phl p 5, and Phl p 6, induced preS-specific IgG antibodies in grass pollen allergic patients [
12,
15]. We found that the preS-specific IgG antibodies induced by vaccination with BM32 were able to neutralize in vitro HBV infection because the BM32-induced antibodies were directed against the NTCP-binding site of preS and thus blocked HBV infection [
12]. Recently, we could show that a single component of BM32, i.e., BM325, termed VVX001, was able to induce such preS-specific HBV-neutralizing antibodies in an individual who was non-responsive to HBsAg-based HBV vaccines [
14].
We hypothesized that VVX001 may not only be useful for inducing HBV-neutralizing antibodies in non-responders to HBsAg-based vaccines but might also be a candidate vaccine for therapeutic vaccination in CHB. This hypothesis is based on the idea that VVX001 can induce preS-specific antibodies which may interrupt recurrent infection of liver cells in the course of CHB and additionally generate “mild” preS-specific CD4+ and CD8+ T-cell responses as was observed in grass pollen allergic patients vaccinated with BM32 [
12]. Such a “mild T cell response” may in the long term contribute to the elimination of infected liver cells without causing severe liver damage. It was a major goal of this study to investigate if VVX001 can induce a preS-specific antibody response in an in vivo model of CHB because we had observed that patients with chronic HBV infections mounted no relevant preS-specific antibody responses [
12]. This lack of preS-specific antibody response in CHB may be due to low immunogenicity of preS because it is expressed/present on circulating HBV or on HBV-derived subviral particles in low amounts and/or due to immunological tolerance in the host. We therefore immunized AAV-HBV mice, which are transduced with an AAV carrying a replication-competent HBV-DNA genome, allowing HBV to replicate in the liver [
21]. Like patients with CHB, these mice did not spontaneously develop relevant levels of preS-specific antibodies, as can be observed in
Figure 2 for the AAV-HBV + vehicle-immunized mice through the complete observation period and for AAV-HBV mice before vaccination with preS-containing vaccines five weeks after transduction. However, a strong increase in preS-specific IgG antibodies was observed for AAV-HBV as well as for AAV-Empty mice, demonstrating that the presence of HBV did not prevent the development of a robust preS-specific IgG response. Although the difference was statistically not significant for IgG
1, it is noteworthy that IgG
2a antibody levels in HBV-carrier mice immunized with VVX001 were significantly higher than in mice immunized with preS alone (
Table S2) and mock-transduced mice. Furthermore, preS-specific IgG
2a titers were higher in VVX001-immunized mice than in only preS-immunized mice (
Figure 2,
Table S3).
Importantly, preS-specific IgG antibodies induced in HBV-carrier mice were directed against the epitope represented by the preS-derived peptides A and B, which contain the NTCP binding site of preS. One can therefore bona fide assume that these antibodies will have HBV neutralizing activity. However, due to lack of serum, we could not study the in vitro HBV-neutralizing effects of the mouse sera. It is another limitation of our study that the therapeutic effects of the preS-specific antibodies could not be investigated in the AAV-HBV mouse model because in this model the HBV-DNA and HBsAg levels are not a result of chronic infection but are due to AAV transduction of liver cells producing constantly HBV-DNA and HBV antigens [
24,
25]. Furthermore, the AAV-HBV mice lack human NTCP on their liver cells so that re-infection via this receptor cannot occur [
26]. Therefore, therapeutic vaccination with VVX001 showed no significant effect on the murine model of chronic HBV. However, we were able to demonstrate that preS-specific antibodies induced with VVX001 containing preS from genotype A cross-reacted with the corresponding NTCP-binding site-containing peptides from genotypes B, C, D, E, F, G, and H in AAV-HBV mice. One may therefore hope that vaccination VVX001 may convey cross-protection for other HBV genotypes. The development of preS-specific antibodies was accompanied by the formation of B cells/plasmablasts secreting preS-specific antibodies, as demonstrated by ELISPOT analysis, indicating that a corresponding secondary preS-specific B-cell response was generated by immunization with VVX001. Furthermore, like in three other studies performed in the AAV-HBV model or in similar models with vaccines intended for therapeutic vaccination, we found evidence for the development of vaccine-specific T-cell responses [
27,
28,
29]. In fact, HBV-carrier mice vaccinated with VVX001 or preS showed increased intrahepatic lymphocytes, suggesting that cells activated by the vaccine could be recruited or attracted into the liver by the HBV-expressing hepatocytes. Vaccination with VVX001 or preS was safe because no adverse effects were observed and ALAT and ASAT levels remained in the normal range, indicating that the vaccines are well tolerated.
Moderate favorable effects on HBV DNA and HBsAg levels like for other therapeutic vaccines tested in the AAV-HBV or in comparable murine models were observed [
27,
28,
29]. However, as indicated above, the major mechanism of VVX001, i.e., the blocking of recurrent HBV liver infection could not be tested in the AAV model because the mice do not express human NTCP on their liver cells. Nevertheless, our study has clearly demonstrated that immunization with VVX001 can induce preS-specific antibodies in a murine model of CHB. Our vaccine is a simple subunit vaccine formulated with aluminum hydroxide, an adjuvant which has been used in numerous vaccines with high safety profile. Subunit vaccine like VVX001 have the advantage, that they can be well dosed and induce distinct but mild T-cell responses avoiding excessive and eventually cytotoxic T-cell responses which may be important for the vaccination of patients with CHB who may be at risk for side effects. Our study therefore encourages further evaluation of VVX001 for therapeutic vaccination against CHB.