Introduction
Despite the availability of safe and efficacious hepatitis B virus (HBV) vaccines, worldwide approximately 300 million people are currently chronic carrier of hepatitis B surface antigen (HBsAg)
[1]- Razavi-Shearer D.G.I.
- Nguyen M.H.
- et al.
Global prevalence, treatment, and prevention of hepatitis B virus infection in 2016: a modelling study.
. Chronic HBV infection may eventually lead to liver cirrhosis and hepatocellular carcinoma (HCC) development. HBV is responsible for a heavy disease burden and an estimated 820,000 liver-related deaths annually
[2]Hepatitis B Treatment: What We Know Now and What Remains to Be Researched.
,. Moreover, at least 12 million and up to 60 million chronic HBV patients are estimated to suffer from a concomitant hepatitis D virus (HDV) infection
4- Stockdale A.J.
- Kreuels B.
- Henrion M.Y.R.
- Giorgi E.
- Kyomuhangi I.
- de Martel C.
- et al.
The global prevalence of hepatitis D virus infection: Systematic review and meta-analysis.
, 5- Hughes S.A.
- Wedemeyer H.
- Harrison P.M.
Hepatitis delta virus.
, 6- Miao Z.
- Zhang S.
- Ou X.
- Li S.
- Ma Z.
- Wang W.
- et al.
Estimating the global prevalence, disease progression and clinical outcome of hepatitis delta virus infection.
, 7- Chen H.Y.
- Shen D.T.
- Ji D.Z.
- Han P.C.
- Zhang W.M.
- Ma J.F.
- et al.
Prevalence and burden of hepatitis D virus infection in the global population: a systematic review and meta-analysis.
. Since HDV infection exacerbates disease progression tremendously by very prompt induction of cirrhosis, liver dysfunction and HCC, it is considered as the most severe type of all viral
hepatitides [5]- Hughes S.A.
- Wedemeyer H.
- Harrison P.M.
Hepatitis delta virus.
. Patients co-infected with HBV and HDV usually recover spontaneously through immune-mediated viral elimination. However, chronic HBV patients that become superinfected with HDV progress in 80% of the cases to a chronic disease state, resulting in rapid deterioration of the pre-existing HBV-related liver damage and very high fatality rates
[8]Hepatitis D virus coinfection and superinfection.
.
Current lifelong HBV therapy with nucleos(t)ide analogues (NAs) suppresses viral replication, but only about 10% of all treated HBeAg-positive patients (and 1% in HBeAg-negative patients) may achieve a functional cure, i.e. complete HBsAg loss with/without seroconversion after a 5-year follow up
9- Locarnini S.
- Hatzakis A.
- Chen D.S.
- Lok A.
Strategies to control hepatitis B: Public policy, epidemiology, vaccine and drugs.
, 10HBV cccDNA: viral persistence reservoir and key obstacle for a cure of chronic hepatitis B.
, 11EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection.
. Therefore, NAs will not eliminate the risk of cancer development. Regarding HDV therapy, this virus does not code for polymerases or proteases that could be therapeutically targeted as is the case for HBV or hepatitis C virus, but relies on both the host replicative machinery and on the helper function of HBV to complete its life cycle
[12]- Caviglia G.P.
- Rizzetto M.
Treatment of hepatitis D, an unmet medical need.
. Therefore, the only recommended clinical regimen remains pegylated-IFNα, a therapy showing poor efficacy and infrequent long-term responses
[13]- Abbas Z.
- Memon M.S.
- Mithani H.
- Jafri W.
- Hamid S.
Treatment of chronic hepatitis D patients with pegylated interferon: a real-world experience.
. Specifically, clearance of serum HDV 6 months after cessation of a 48-week therapy was only achieved in 28% of subjects and 50% of the responders experienced late HDV RNA relapse during longer follow-up
14- Wedemeyer H.
- Yurdaydin C.
- Hardtke S.
- Caruntu F.A.
- Curescu M.G.
- Yalcin K.
- et al.
Peginterferon alfa-2a plus tenofovir disoproxil fumarate for hepatitis D (HIDIT-II): a randomised, placebo controlled, phase 2 trial.
, 15- Wedemeyer H.
- Yurdaydin C.
- Dalekos G.N.
- Erhardt A.
- Cakaloglu Y.
- Degertekin H.
- et al.
Peginterferon plus adefovir versus either drug alone for hepatitis delta.
, 16- Heidrich B.
- Yurdaydin C.
- Kabacam G.
- Ratsch B.A.
- Zachou K.
- Bremer B.
- et al.
Late HDV RNA relapse after peginterferon alpha-based therapy of chronic hepatitis delta.
. While phase III results are still pending, in 2020 the European Medicines Agency has conditionally approved the entry-inhibitor Myrcludex B (bulevirtide), a preS1-derived peptide able to block the HBV/HDV host receptor natrium taurocholate co-transporting polypeptide (NTCP), for the treatment of patients with compensated chronic HDV infection
17- Wedemeyer H.
- Schöneweis K.
- Bogomolov P.O.
- Chulanov V.
- Stepanova T.
- Viacheslav M.
- et al.
48 weeks of high dose (10 mg) bulevirtide as monotherapy or with peginterferon alfa-2a in patients with chronic HBV/HDV co-infection.
, 18- Wedemeyer H.
- Schöneweis K.
- Bogomolov P.O.
- Voronkova N.
- Chulanov V.
- Stepanova T.
Interim results of a multicentre, open-label phase 2 clinical trial (MYR203) to assess safety and efficacy of Myrcludex B in combination with Peg-Interferon alpha 2a in patients with chronic HBV/HDV co-infection.
, 19- Wedemeyer H.
- Bogomolov P.
- Blank A.
- Allweiss L.
- Dandri-Petersen M.
- Bremer B.
- et al.
Final results of a multicenter, open-label phase 2b clinical trial to assess safety and efficacy of Myrcludex B in combination with Tenofovir in patients with chronic HBV/HDV co-infection.
, 20- Degasperi E.
- Anolli M.P.
- Lampertico P.
Bulevirtide for patients with compensated chronic hepatitis delta: A review.
. Several other HDV treatment options are currently assessed in clinical trials
[21]New Treatments for Chronic Hepatitis B Virus/Hepatitis D Virus Infection.
. It is however widely accepted that a combination of antiviral compounds targeting different steps in the viral life cycle is required to eliminate both viruses.
In this study, we evaluated the capacity of a previously generated HBsAg-specific human monoclonal antibody (hu-mAb) both
in vitro and
in vivo to prevent and treat HBV and HDV infection
[22]- Depraetere S.
- Verhoye L.
- Leclercq G.
- Leroux-Roels G.
Human B cell growth and differentiation in the spleen of immunodeficient mice.
. Targeting the entry-step in combination with diminishing circulating HBsAg would simultaneously block viral spread and potentially provide a window of opportunity for other antivirals to break immune tolerance in a chronic setting.
Material and methods
More details can be found in Supplemental Materials online.
Human monoclonal anti-HBsAg production and purification
The human monoclonal anti-HBsAg specific antibody was produced using classical hybridoma technology, more specifically by fusion of SCID-engrafted human peripheral blood lymphocytes isolated from a vaccinated individual (6,981 mIU/ml anti-HBsAg at time of blood donation) with K6H6/B5 heteromyeloma cells as previously described
[22]- Depraetere S.
- Verhoye L.
- Leclercq G.
- Leroux-Roels G.
Human B cell growth and differentiation in the spleen of immunodeficient mice.
. Fused cells were then seeded in culture medium supplemented with hybridoma growth factors and selection drugs hypoxanthine-aminopterin-thymidine and ouabain. Anti-HBs-positive cultures were sequentially cloned and several monoclonal hybridoma lines were isolated and confirmed via commercial anti-HBs ELISA (DiaSorin, Italy). The hu-mAb was then purified from collected supernatant using conventional Protein-G columns (Hitrap Protein G HP, Sigma Aldrich, USA). After evaluation by serum protein electrophoresis (SAS-MX Serum Protein-10, Sysmex, Japan), the antibody was concentrated (Amicon Ultra-15 50k, Sigma Aldrich, USA) and its concentration was determined using spectrophotometry at 280 nm.
In vitro prevention of HBV and HDV infection
For in vitro prevention of HBV infection, anti-HBsAg was applied to HepG2.hNTCP cells in duplicate at 5-fold serial dilutions ranging from 10 μg/ml to 0.128 ng/ml. Two hours later, cells were infected with HBV (4,990 IU/cell; genotype D) and after one week, infection was assessed using immunofluorescent (IF) staining of HBV-Core positive cells. Images were captured by automated spinning disk microscopy using a 40X objective (CSU-X1, Nikon). Per condition, a 20 x 10 field was captured (in duplicate) and positive cells were automatically counted using ImageJ software v1.53c. In vitro prevention of HDV infection (genotype 1) was examined in the same manner, but in 3 different cell lines (HepG2.hNTCP, Huh7.5.hNTCP and NEB2.7). Imaging was performed using the Leica TCS-SPE microscope with a 20X objective. Per condition, 3 random pictures were taken (in duplicate) and HDV-positive cells were automatically counted using ImageJ software v1.53c. More details are provided in Supplemental Materials.
Mice
All mice were bred under sterile conditions and all experiments were approved by the Animal Ethics Committee of the Faculty of Medicine and Health Sciences of Ghent University. Human-liver chimeric mice were generated by transplanting 10
6 primary human hepatocytes (donor C342, C399 and HH223 from Corning, the Netherlands; and donor L191501 from Lonza, Switzerland) into homozygous uPA
+/+-SCID mice as previously described
[23]- Meuleman P.
- Libbrecht L.
- De Vos R.
- de Hemptinne B.
- Gevaert K.
- Vandekerckhove J.
- et al.
Morphological and biochemical characterization of a human liver in a uPA-SCID mouse chimera.
,[24]- Meuleman P.
- Vanlandschoot P.
- Leroux-Roels G.
A simple and rapid method to determine the zygosity of uPA-transgenic SCID mice.
. Human albumin was quantified in mouse plasma to evaluate successful humanization of the mouse liver using conventional ELISA (Bethyl Laboratories, USA). Mice with albumin levels ranging between 2-10 mg/ml were selected for this study and groups were randomized taking into consideration human albumin levels, infection levels and general condition (e.g. based on body weight). Mice were 8 to 9 weeks old at the start of infection.
In vivo prevention of HBV, HBV/HDV and HDV superinfection
For in vivo prevention of HBV, human-liver chimeric mice (n=6) repopulated with human hepatocytes from two different donors (n=3 for C342 and n=3 for HH223) were passively immunized through intraperitoneal (IP) administration of 1 mg hu-mAb 3 days before challenge with HBV (patient serum, 106 IU/mouse). As controls, 6 additional mice were infected without prior passive immunization. For prevention of HBV/HDV co-infection, humanized mice (n=4) engrafted with hepatocytes from donor C342 were passively immunized (1 mg hu-mAb, IP) 3 days prior to HBV/HDV co-infection with cell culture-derived virus (5 x 106 IU HBV DNA/mouse and 2.3 x 106 IU HDV RNA/mouse). As controls, 3 additional mice were HBV/HDV co-infected without prior passive immunization. Mice (co-)infected with HBV or HBV/HDV were monitored until week 12-13 post-inoculation. Finally, for prevention of HDV superinfection, humanized mice were first infected with either patient-derived HBV (106 IU/mouse, n=7, mice engrafted with donor L191501 hepatocytes) or cell-culture derived HBV (5 x 106 IU per mouse, n=14, i.e. 5 donor C342 and 9 C399 engrafted mice). Respectively 6 and 8 weeks later, mice were superinfected with HDV from either patient or cell culture origin (both 2.55 x 105 IU/mouse). Three days prior to superinfection, respectively 5 out of 7 and 9 out of 14 mice were passively immunized with 1 mg of hu-mAb (IP). Mice superinfected with HDV were monitored for 9-10 weeks following HDV inoculation. Blood plasma was collected every 1-2 weeks (depending on the condition of the mouse and the timing post-infection) and HBV DNA and HDV RNA levels were quantified via commercial HBV/HDV RealStar® (RT-)qPCR (Altona Diagnostics, Germany) or the RealTime m2000 HBV assay (Abbot, USA). Anti-HBsAg ELISA was performed using a commercially available quantitative kit (Beijing Wantai Biological, China).
Treatment of HBV and HBV/HDV (co-)infected mice
Human-liver chimeric mice (n=7) transplanted with donor C342 hepatocytes were chronically infected with cell-culture derived HBV (5 x 106 IU/mouse). Four out of 7 mice received 1 mg hu-mAb (IP) twice a week for 4 consecutive weeks (8 doses in total), starting at w11 post viral inoculation. The remaining 3 infected mice were left untreated as controls. In a second treatment experiment, 13 human-liver chimeric mice, transplanted with donor L191501 hepatocytes, were first infected with cell-culture derived HBV (5 x 106 IU/mouse) and 6 weeks later superinfected with cell-culture derived HDV (2.55 x 105 IU/mouse). At week 10, 7 out of 13 mice received 1 mg hu-mAb (IP) twice a week for 4 consecutive weeks (9 doses in total). The 6 remaining co-infected mice did not receive any treatment (control). Blood plasma was collected every 1 to 2 weeks (depending on the condition of the mouse and the timing post-infection) and the levels of HBV DNA, HDV RNA and anti-HBsAg mAb levels were determined as described above. Plasma samples were analyzed by SDS-PAGE and Western blot to assess HBsAg.
SDS-PAGE and Western blot analysis
Exactly 0.5μl of mouse plasma was used for sample preparation and denaturation (70°C, 10 minutes), and subjected to SDS-polyacrylamide gel (12%) electrophoresis and proteins were transferred to a PVDF membrane. Membranes were blocked (5% skim milk in TBS-T) and the expressed HBsAg was detected by a primary goat anti-HBsAg antibody (70-HG15; Fitzgerald industries; 1/1,000), followed by a HRP-conjugated rabbit anti-goat antibody (31402; Thermo Fisher Scientific; 1/20,000). Immunoblots were developed using the SuperSignal™ West Femto Maximum Sensitivity Substrate kit (Thermo Fisher Scientific, USA) and exposed to the ImageQuant LAS4000 chemiluminescent imaging system (GE Healthcare, Diegem, Belgium). More details are provided in Supplemental Materials.
Statistics
GraphPad Prism v9.3.1 was used for graph visualization and statistical analyses. IC50 values were calculated using non-linear regression (curve fit). Normality tests and Mann-Whitney U-tests enabled comparisons between normalized log mean virus levels of treated versus non-treated mice. Data were considered statistically significant if p < 0.05.
Discussion
The excessive HBsAg release in chronic HBV patients tolerizes antibody- and cell-mediated immune responses, and represent a major hurdle for viral eradication by current treatments
[25]- Kuipery A.
- Gehring A.J.
- Isogawa M.
Mechanisms of HBV immune evasion.
,[26]Innate and adaptive immune responses in chronic hepatitis B virus infections: towards restoration of immune control of viral infection.
. Consequently, it is of utmost importance to find ways to break immune tolerance, enabling the host to mount effective immune responses to clear the virus and to prevent HDV superinfection. Here, we demonstrate the prophylactic and therapeutic efficacy of a hu-mAb targeting the HBV envelope in the context of HBV and HDV infection
in vitro and in human-liver chimeric mice.
The hu-mAb neutralized HBV and HDV
in vitro with respective IC
50 values of 3.53 ng/ml (∼0.23 nM) and 3.58 ng/ml (∼0.24 nM) and can be considered as highly potent, since Hehle
et al. [27]- Hehle V.
- Beretta M.
- Bourgine M.
- Ait-Goughoulte M.
- Planchais C.
- Morisse S.
- et al.
Potent human broadly neutralizing antibodies to hepatitis B virus from natural controllers.
evaluated 170 different hu-mAbs isolated from natural HBV controllers or vaccinated individuals and showed that only 35% of antibodies harbored IC
50 values <50 ng/ml.
Next, we demonstrated efficient prevention of HBV mono- and HBV/HDV co-infection in human-liver chimeric mice. Conceivably, once a chronic HBV infection is fully established, preventing HDV superinfection may be even more challenging due to massive HBsAg presence. However, depending on the hepatocyte donor, we showed either near complete prevention (donor C342 and C399) or at least delayed HDV RNA levels (donor L191501). Noteworthy, transplantation of mice with donor L191501 human hepatocytes rendered the animals more prone to hepatitis infection compared to other donors tested (
data not shown), which may explain for the viral breakthrough. Overall, our HDV superinfection prevention data is quite impressive since only a single injection of 1mg hu-mAb was sufficient to overcome the high load of circulating HBsAg and to (partially) neutralize the high-titer HDV inoculum. Furthermore, since HDV can silently persist and replicate in human hepatocytes for up to 6 weeks, the hu-mAb effectively prohibits entry of HDV virions into hepatocytes
[28]- Giersch K.
- Helbig M.
- Volz T.
- Allweiss L.
- Mancke L.V.
- Lohse A.W.
- et al.
Persistent hepatitis D virus mono-infection in humanized mice is efficiently converted by hepatitis B virus to a productive co-infection.
,[29]- Giersch K.
- Hermanussen L.
- Volz T.
- Kah J.
- Allweiss L.
- Casey J.
- et al.
Murine hepatocytes do not support persistence of Hepatitis D virus mono-infection in vivo.
.
Notably, antibody half-life varies throughout our experiments, i.e. the antibody is still present in circulation 3-4 weeks or longer after a 1 mg injection. This may potentially be explained by variability in body weight (and hence distribution volume) since we injected 1 mg of antibody irrespective of mouse body weight. Furthermore, antibody clearance might be greatly accelerated by high replication of the virus (high circulating viral load). The source of viral inoculum also appears to influence the success of prevention and lack of viral rebound. For example, in the prophylactic HBV/HDV co-infection experiment, cell-culture derived virus was used and no viral rebound was observed, while in the prophylactic HBV mono-infection experiment, patient-derived virus was applied while we did observe HBV DNA rebound. Also more animals were protected from HDV superinfection when infected with cell-culture derived viruses. Therefore, it seems that primary patient-derived virus preparations are intrinsically less sensitive to antibody neutralization, or may contain variants that are.
The therapeutic potential of the hu-mAb was furthermore demonstrated by (significantly) reduced mean viral plasma levels at the end of a 4-week therapy in HBV-infected (2.02 x log
10 drop) and HBV/HDV co-infected mice (2.82 x log
10 and 3.50 x log
10 drops for resp. HBV and HDV). Since the repeated injections of antibody result in a clear drop in HBV DNA (and HDV RNA levels) in this therpautic setup, we assume that the single dose regimen of the HDV superinfection prevention experiment was insufficient to show an antiviral effect on the already established HBV infection (i.e. no effect on HBV DNA). Interestingly, our 4-week therapy data are in line with the most potent hu-mAb (namely Bc1.187) tested by Hehle
et al. [27]- Hehle V.
- Beretta M.
- Bourgine M.
- Ait-Goughoulte M.
- Planchais C.
- Morisse S.
- et al.
Potent human broadly neutralizing antibodies to hepatitis B virus from natural controllers.
that treated HUHEP mice (weekly ∼1mg (50 mg/kg) hu-mAb IP for 3 weeks), inducing a 1.76 x log
10 decrease in serum HBV DNA at the end of therapy. Interestingly, circulating HBsAg levels dropped and remained undetectable for 2 weeks following the last injection in 60% of treated animals. Importantly, these mice had initial lower HBV DNA levels (<10
6 IU/ml) compared to mice in our study. Another hu-mAb (HBC34) was evaluated at a very comparable schedule, but at lower dose (a 4-week treatment with 2 IP injections per week of 1mg/kg) in HBV/HDV co-infected USB/USG mice
[30]- Corti D.
- Volz T.
- Giersch K.
- Agatic G.
- Cameroni E.
- Kah J.
- et al.
Prevention of Viral Spread, Viremia Reduction and HBsAg Clearance in Hepatitis B and D Coinfected Humanized Mice by a Human Neutralizing Monoclonal Antibody.
,[31]Lempp FA, volz T, cameroni E, Benigni F, Zhou J, Rosen LE, et al. The potent broadly neutralizing antibody VIR-3434 controls Hepatitis B and D Virus infection and reduces HBsAg in humanized mice.; 2022.
. They revealed a similar reduction in HBV DNA and HDV RNA levels (mean 1 to 2.7 x log
10 and 2.4 to 2.5 x log
10 reduction, respectively, in both models) and a concomitant 2.7 to 2.8 x log
10 reduction of HBsAg levels after 4 weeks. Finally, we also show HBsAg loss on-therapy with rebound after therapy cessation, a phenomenon that is frequently noted upon withdrawal of mAb therapy
[27]- Hehle V.
- Beretta M.
- Bourgine M.
- Ait-Goughoulte M.
- Planchais C.
- Morisse S.
- et al.
Potent human broadly neutralizing antibodies to hepatitis B virus from natural controllers.
,[32]- Galun E.
- Eren R.
- Safadi R.
- Ashour Y.
- Terrault N.
- Keeffe E.B.
- et al.
Clinical evaluation (phase I) of a combination of two human monoclonal antibodies to HBV: safety and antiviral properties.
. Interestingly, the HBsag loss might provide a therapeutic window for viral eradication using NAs, possibly in combination with other antivirals, that could take advantage of a (partially) reconstituted adaptive immune response
[23]- Meuleman P.
- Libbrecht L.
- De Vos R.
- de Hemptinne B.
- Gevaert K.
- Vandekerckhove J.
- et al.
Morphological and biochemical characterization of a human liver in a uPA-SCID mouse chimera.
,[24]- Meuleman P.
- Vanlandschoot P.
- Leroux-Roels G.
A simple and rapid method to determine the zygosity of uPA-transgenic SCID mice.
. Interestingly, although no effect on HBsAg was seen when the recently EU approved bulevirtide was used as mono-therapy, HBsAg decreases could be observed when combined with peg-IFNα
[17]- Wedemeyer H.
- Schöneweis K.
- Bogomolov P.O.
- Chulanov V.
- Stepanova T.
- Viacheslav M.
- et al.
48 weeks of high dose (10 mg) bulevirtide as monotherapy or with peginterferon alfa-2a in patients with chronic HBV/HDV co-infection.
,[20]- Degasperi E.
- Anolli M.P.
- Lampertico P.
Bulevirtide for patients with compensated chronic hepatitis delta: A review.
. Notably, decreasing HBsAg might be a valuable therapeutic strategy, but currently no direct evidence is available to support this hypothesis and therefore, additional studies addressing the relationship between circulating HBsAg levels and HBV-specific immune responses are required to provide further insights on the immunobiology of HBV, and presumably, additional immune activation would be necessary to control the virus
[33]- Kim J.H.
- Ghosh A.
- Ayithan N.
- Romani S.
- Khanam A.
- Park J.J.
- et al.
Circulating serum HBsAg level is a biomarker for HBV-specific T and B cell responses in chronic hepatitis B patients.
. Nowadays, polyclonal antibodies derived from pooled plasma from recovered or vaccinated individuals (HBIG) are successfully and routinely used in clinical practice as post-exposure prophylaxis, to avoid HBV recurrence after liver-transplantation or therapeutic immunosuppression, or to prevent perinatal transmission
34- Cerino A.
- Mantovani S.
- Mele D.
- Oliviero B.
- Varchetta S.
- Mondelli M.U.
Human Monoclonal Antibodies as Adjuvant Treatment of Chronic Hepatitis B Virus Infection.
, 35The option of HBIG-free prophylaxis against recurrent HBV.
, 36Hepatitis B immune globulin to prevent hepatitis B virus graft reinfection following liver transplantation: a concise review.
. However, only a small antibody proportion in HBIG preparations is actually HBV-specific and neutralizing, and blood products always remain subject to bio-safety concerns, especially for immunosuppressed patients
[37]- Cerino A.
- Bremer C.M.
- Glebe D.
- Mondelli M.U.
A Human Monoclonal Antibody against Hepatitis B Surface Antigen with Potent Neutralizing Activity.
,[38]- Wang W.
- Sun L.
- Li T.
- Ma Y.
- Li J.
- Liu Y.
- et al.
A human monoclonal antibody against small envelope protein of hepatitis B virus with potent neutralization effect.
. Consequently, monoclonal antibodies may be an attractive alternative since they represent a stable, reproducible source for prolonged immunotherapy. Hu-mAbs are already successfully explored in many therapeutic fields such as oncology
[39]- Kimiz-Gebologlu I.
- Gulce-Iz S.
- Biray-Avci C.
Monoclonal antibodies in cancer immunotherapy.
, auto-immune diseases
[40]- Barnas J.L.
- Looney R.J.
- Anolik J.H.
B cell targeted therapies in autoimmune disease.
and recently also for infectious diseases such as SARS-CoV-2
[41]- Weinreich D.M.
- Sivapalasingam S.
- Norton T.
- Ali S.
- Gao H.
- Bhore R.
- et al.
REGEN-COV Antibody Combination and Outcomes in Outpatients with Covid-19.
,[42]Efficacy and safety of two neutralising monoclonal antibody therapies, sotrovimab and BRII-196 plus BRII-198, for adults hospitalised with COVID-19 (TICO): a randomised controlled trial.
. Our research group also demonstrated successful hu-mAb use in the context of HCV infection
43- Desombere I.
- Mesalam A.A.
- Urbanowicz R.A.
- Van Houtte F.
- Verhoye L.
- Keck Z.Y.
- et al.
A novel neutralizing human monoclonal antibody broadly abrogates hepatitis C virus infection in vitro and in vivo.
, 44- Mesalam A.A.
- Desombere I.
- Farhoudi A.
- Van Houtte F.
- Verhoye L.
- Ball J.
- et al.
Development and characterization of a human monoclonal antibody targeting the N-terminal region of hepatitis C virus envelope glycoprotein E1.
, 45- Vercauteren K.
- Van Den Eede N.
- Mesalam A.A.
- Belouzard S.
- Catanese M.T.
- Bankwitz D.
- et al.
Successful anti-scavenger receptor class B type I (SR-BI) monoclonal antibody therapy in humanized mice after challenge with HCV variants with in vitro resistance to SR-BI-targeting agents.
. Regarding other HBV-specific hu-mAbs, previous studies similarly have shown efficient prevention, delayed infection, or therapeutic efficacy in cell culture
[27]- Hehle V.
- Beretta M.
- Bourgine M.
- Ait-Goughoulte M.
- Planchais C.
- Morisse S.
- et al.
Potent human broadly neutralizing antibodies to hepatitis B virus from natural controllers.
,46- Jo G.
- Jeong M.S.
- Wi J.
- Kim D.H.
- Kim S.
- Kim D.
- et al.
Generation and Characterization of a Neutralizing Human Monoclonal Antibody to Hepatitis B Virus PreS1 from a Phage-Displayed Human Synthetic Fab Library.
, 47- Zankharia U.S.
- Kudchodkar S.
- Khoshnejad M.
- Perales-Puchalt A.
- Choi H.
- Ho M.
- et al.
Neutralization of hepatitis B virus by a novel DNA-encoded monoclonal antibody.
, 48- Corti D.
- Benigni F.
- Shouval D.
Viral envelope-specific antibodies in chronic hepatitis B virus infection.
, chimpanzees
49- Chi S.W.
- Maeng C.Y.
- Kim S.J.
- Oh M.S.
- Ryu C.J.
- Kim S.J.
- et al.
Broadly neutralizing anti-hepatitis B virus antibody reveals a complementarity determining region H3 lid-opening mechanism.
, 50- Zhang P.
- Yu M.Y.
- Venable R.
- Alter H.J.
- Shih J.W.
Neutralization epitope responsible for the hepatitis B virus subtype-specific protection in chimpanzees.
, 51- Hong H.J.
- Ryu C.J.
- Hur H.
- Kim S.
- Oh H.K.
- Oh M.S.
- et al.
In vivo neutralization of hepatitis B virus infection by an anti-preS1 humanized antibody in chimpanzees.
, 52- Ogata N.
- Ostberg L.
- Ehrlich P.H.
- Wong D.C.
- Miller R.H.
- Purcell R.H.
Markedly prolonged incubation period of hepatitis B in a chimpanzee passively immunized with a human monoclonal antibody to the a determinant of hepatitis B surface antigen.
, 53- Kim S.H.
- Shin Y.W.
- Hong K.W.
- Chang K.H.
- Ryoo K.H.
- Paik S.H.
- et al.
Neutralization of hepatitis B virus (HBV) by human monoclonal antibody against HBV surface antigen (HBsAg) in chimpanzees.
, 54- Eren R.
- Ilan E.
- Nussbaum O.
- Lubin I.
- Terkieltaub D.
- Arazi Y.
- et al.
Preclinical evaluation of two human anti-hepatitis B virus (HBV) monoclonal antibodies in the HBV-trimera mouse model and in HBV chronic carrier chimpanzees.
and various mouse models
[27]- Hehle V.
- Beretta M.
- Bourgine M.
- Ait-Goughoulte M.
- Planchais C.
- Morisse S.
- et al.
Potent human broadly neutralizing antibodies to hepatitis B virus from natural controllers.
,[38]- Wang W.
- Sun L.
- Li T.
- Ma Y.
- Li J.
- Liu Y.
- et al.
A human monoclonal antibody against small envelope protein of hepatitis B virus with potent neutralization effect.
,54- Eren R.
- Ilan E.
- Nussbaum O.
- Lubin I.
- Terkieltaub D.
- Arazi Y.
- et al.
Preclinical evaluation of two human anti-hepatitis B virus (HBV) monoclonal antibodies in the HBV-trimera mouse model and in HBV chronic carrier chimpanzees.
, 55- Li D.
- He W.
- Liu X.
- Zheng S.
- Qi Y.
- Li H.
- et al.
A potent human neutralizing antibody Fc-dependently reduces established HBV infections.
, 56- Zhang T.Y.
- Yuan Q.
- Zhao J.H.
- Zhang Y.L.
- Yuan L.Z.
- Lan Y.
- et al.
Prolonged suppression of HBV in mice by a novel antibody that targets a unique epitope on hepatitis B surface antigen.
, 57- Wang Q.
- Michailidis E.
- Yu Y.
- Wang Z.
- Hurley A.M.
- Oren D.A.
- et al.
A Combination of Human Broadly Neutralizing Antibodies against Hepatitis B Virus HBsAg with Distinct Epitopes Suppresses Escape Mutations.
, 58- Kim J.-H.
- Kim H.
- Kim T.-H.
- Kim W.
- Jung J.
- Lee A.
PS-077-Lenvervimab, a monoclonal antibody against HBsAg, can induce sustained HBsAg loss in a chronic hepatitis B mouse model.
. Some have reached clinical stages as well, such as Lenvervimab
[59]- Lee H.W.
- Park J.Y.
- Hong T.
- Park M.S.
- Ahn S.H.
Efficacy of Lenvervimab, a Recombinant Human Immunoglobulin, in Treatment of Chronic Hepatitis B Virus Infection.
and VIR-3434 , the Fc-engineered version of antibody HBC34, with extended serum half-life; NCT04423393 and NCT04856085
[60]Advances in human monoclonal antibody therapy for HBV infection.
. Hu-mAb studies involving HDV prophylaxis or therapy are currently very limited: Li
et al. [40]- Barnas J.L.
- Looney R.J.
- Anolik J.H.
B cell targeted therapies in autoimmune disease.
demonstrated prevention in a mouse model harboring human NTCP-receptors, thus in absence of relevant HBV infection and moreover in a mouse background, and as mentioned above, antibody HBC34 revealed apotent decrease in HBV/HDV viremia in HBV/HDV co-infected USB/USG mice
[30]- Corti D.
- Volz T.
- Giersch K.
- Agatic G.
- Cameroni E.
- Kah J.
- et al.
Prevention of Viral Spread, Viremia Reduction and HBsAg Clearance in Hepatitis B and D Coinfected Humanized Mice by a Human Neutralizing Monoclonal Antibody.
,[31]Lempp FA, volz T, cameroni E, Benigni F, Zhou J, Rosen LE, et al. The potent broadly neutralizing antibody VIR-3434 controls Hepatitis B and D Virus infection and reduces HBsAg in humanized mice.; 2022.
.
Currently evaluated or approved antibody dosages vary tremendously in human applications, even beyond the field of infectious diseases. For example, omalizumab, a humanized recombinant monoclonal anti-IgE antibody approved for the treatment of allergic asthma is recommended at 75 to 600 mg every two weeks
. Rituximab, a genetically engineered chimeric mouse/human mAb approved for rheumatoid arthritis is recommended at 2 infusions of 1,000 mg IV, with 2 weeks of interval
. Eculizumab, a humanized mAb approved for myasthenia gravis, is administered at 600 mg IV weekly for 4 weeks, followed by 900 mg IV in the 5th week and then, 900 mg every 2 weeks as maintenance therapy
[63]- Xiao H.
- Wu K.
- Liang X.
- Li R.
- Lai K.P.
Clinical Efficacy and Safety of Eculizumab for Treating Myasthenia Gravis.
. Finally, the FDA recently authorized two mAb’s (Casirivimab and Imdevimab) for the treatment of COVID-19 disease at dosages as high as 1,400 mg IV
[64]- Razonable R.R.
- Pawlowski C.
- O'Horo J.C.
- Arndt L.L.
- Arndt R.
- Bierle D.M.
- et al.
Casirivimab-Imdevimab treatment is associated with reduced rates of hospitalization among high-risk patients with mild to moderate coronavirus disease-19.
,[65]Casirivimab. Drugs and Lactation Database (LactMed). Bethesda (MD): National Library of Medicine (US); 2006.
. These examples illustrate the high variability of applied doses in various applications, but also that much higher dosages are achievable and tolerated compared to our regimen. Assuming an average mouse body weight of 15 g, we applied about 66.7 mg/kg per injection. In the treatment regimen, we hence administered a total dose of 533 mg over a 4-week period; clearly in range of what is feasible in humans. However, dosages used in mice cannot simply be translated into human doses. More specifically, the common perception of scaling of dose based on body weight (mg/kg) alone is not entirely correct. This is primarily because the biochemical, functional systems in species vary which in turn alter pharmacokinetics
[66]A simple practice guide for dose conversion between animals and human.
.
The main putative mechanism of action of the HBsAg-targeting antibody in the applied mouse model is functional neutralization of the viral envelope, which directly blocks entry of HBV and HDV, and consequently prevents spread of infection to naïve hepatocytes. Furthermore, the antibody also promotes HBsAg reduction (or clearance) in circulation. It has also been shown that HBsAg-specific antibodies could be internalized into infected cells and therefore partially inhibit HBsAg secretion from these cells, implicating intracellular blocking of virion and protein release as an additional possible mechanism of action
[67]- Schilling R.
- Ijaz S.
- Davidoff M.
- Lee J.Y.
- Locarnini S.
- Williams R.
- et al.
Endocytosis of hepatitis B immune globulin into hepatocytes inhibits the secretion of hepatitis B virus surface antigen and virions.
,[68]- Neumann A.U.
- Phillips S.
- Levine I.
- Ijaz S.
- Dahari H.
- Eren R.
- et al.
Novel mechanism of antibodies to hepatitis B virus in blocking viral particle release from cells.
. Importantly, in the context of infected patients, additional intrinsic effector functions could be induced through the formation of immune complexes. More specifically, clearance of circulating virions, antigens or infected cells could be promoted via antibody-dependent cell-mediated cytotoxicity and/or other Fc-mediated effector responses such as complement lysis or phagocytosis
[55]- Li D.
- He W.
- Liu X.
- Zheng S.
- Qi Y.
- Li H.
- et al.
A potent human neutralizing antibody Fc-dependently reduces established HBV infections.
,[56]- Zhang T.Y.
- Yuan Q.
- Zhao J.H.
- Zhang Y.L.
- Yuan L.Z.
- Lan Y.
- et al.
Prolonged suppression of HBV in mice by a novel antibody that targets a unique epitope on hepatitis B surface antigen.
,69The growth and potential of human antiviral monoclonal antibody therapeutics.
, 70Comparing antibody and small-molecule therapies for cancer.
, 71- Halper-Stromberg A.
- Lu C.L.
- Klein F.
- Horwitz J.A.
- Bournazos S.
- Nogueira L.
- et al.
Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice.
, 72Immunoregulatory functions of immune complexes in vaccine and therapy.
, 73Antibodies to hepatitis B surface antigen potentiate the response of human T lymphocyte clones to the same antigen.
. Accordingly, many therapeutic advantages could be proposed in a clinical setting: 1) reduction of the hepatic accumulation of cccDNA, one of the main causes of HBV persistence
[74]- Allweiss L.
- Volz T.
- Giersch K.
- Kah J.
- Raffa G.
- Petersen J.
- et al.
Proliferation of primary human hepatocytes and prevention of hepatitis B virus reinfection efficiently deplete nuclear cccDNA in vivo.
,[75]- Volz T.
- Allweiss L.
- Ben M.M.
- Warlich M.
- Lohse A.W.
- Pollok J.M.
- et al.
The entry inhibitor Myrcludex-B efficiently blocks intrahepatic virus spreading in humanized mice previously infected with hepatitis B virus.
; 2) mediation of serum HBV/HDV and HBsAg clearance, which may ultimately counteract the challenging T-cell exhaustion mechanisms that are characteristic for patients with chronic HBV infection
[76]- Bachmann M.F.
- Hunziker L.
- Zinkernagel R.M.
- Storni T.
- Kopf M.
Maintenance of memory CTL responses by T helper cells and CD40-CD40 ligand: antibodies provide the key.
; 3) overall risk reduction of viral reactivation (following liver-transplantation)
[77]- Paul S.
- Dickstein A.
- Saxena A.
- Terrin N.
- Viveiros K.
- Balk E.M.
- et al.
Role of surface antibody in hepatitis B reactivation in patients with resolved infection and hematologic malignancy: A meta-analysis.
; and finally 4) potential allowance of safe withdrawal of antiviral drugs
[78]- Whitacre D.C.
- Peters C.J.
- Sureau C.
- Nio K.
- Li F.
- Su L.
- et al.
Designing a therapeutic hepatitis B vaccine to circumvent immune tolerance.
.
In conclusion, we show the added value of an entry-inhibiting hu-mAb in the context of HBV and HDV (super)infection prophylaxis and its potential use as treatment for chronic HBV or HBV/HDV (co-)infection. For the here presented experiments we used either patient-derived virus or cell-culture derived virus (genotype D for HBV and genotype 1 for HDV) in human-liver chimeric mice that were transplanted with hepatocytes from in total 4 different donors. Further confirmation using additional viral genotypes and various hepatocyte donors having distinct genetic backgrounds is warranted. Overall, our data suggests that this hu-mAb is an interesting candidate to complement current therapies or antivirals (under development) to eradicate both HBV and HDV infections. Decreasing or even eliminating the high levels of circulating HBsAg in chronic patients may provide an opportunistic window for other antiviral therapies targeting later steps in the viral life cycle or the immune system to eventually cure chronic infection.