1
|
Schutti O, Klauer L, Baudrexler T, Burkert F, Schmohl J, Hentrich M, Bojko P, Kraemer D, Rank A, Schmid C, Schmetzer H. Effective and Successful Quantification of Leukemia-Specific Immune Cells in AML Patients' Blood or Culture, Focusing on Intracellular Cytokine and Degranulation Assays. Int J Mol Sci 2024; 25:6983. [PMID: 39000091 PMCID: PMC11241621 DOI: 10.3390/ijms25136983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 07/16/2024] Open
Abstract
Novel (immune) therapies are needed to stabilize remissions or the disease in AML. Leukemia derived dendritic cells (DCleu) can be generated ex vivo from AML patients' blasts in whole blood using approved drugs (GM-CSF and PGE-1 (Kit M)). After T cell enriched, mixed lymphocyte culture (MLC) with Kit M pretreated (vs. untreated WB), anti-leukemically directed immune cells of the adaptive and innate immune systems were already shown to be significantly increased. We evaluated (1) the use of leukemia-specific assays [intracellular cytokine production of INFy, TNFa (INCYT), and degranulation detected by CD107a (DEG)] for a detailed quantification of leukemia-specific cells and (2), in addition, the correlation with functional cytotoxicity and patients' clinical data in Kit M-treated vs. not pretreated settings. We collected whole blood (WB) samples from 26 AML patients at first diagnosis, during persisting disease, or at relapse after allogeneic stem cell transplantation (SCT), and from 18 healthy volunteers. WB samples were treated with or without Kit M to generate DC/DCleu. After MLC with Kit M-treated vs. untreated WB antigen-specific/anti-leukemic effects were assessed through INCYT, DEG, and a cytotoxicity fluorolysis assay. The quantification of cell subtypes was performed via flow cytometry. Our study showed: (1) low frequencies of leukemia-specific cells (subtypes) detectable in AML patients' blood. (2) Significantly higher frequencies of (mature) DCleu generable without induction of blast proliferation in Kit M-treated vs. untreated samples. (3) Significant increase in frequencies of immunoreactive cells (e.g., non-naive T cells, Tprol) as well as in INCYT/DEG ASSAYS leukemia-specific adaptive-(e.g., B, T(memory)) or innate immune cells (e.g., NK, CIK) after MLC with Kit M-treated vs. untreated WB. The results of the intracellular production of INFy and TNFa were comparable. The cytotoxicity fluorolysis assay revealed significantly enhanced blast lysis in Kit M-treated vs. untreated WB. Significant correlations could be shown between induced leukemia-specific cells from several lines and improved blast lysis. We successfully detected and quantified immunoreactive cells at a single-cell level using the functional assays (DEG, INCYT, and CTX). We could quantify leukemia-specific subtypes in uncultured WB as well as after MLC and evaluate the impact of Kit M pretreated (DC/DCleu-containing) WB on the provision of leukemia-specific immune cells. Kit M pretreatment (vs. no pretreatment) was shown to significantly increase leukemia-specific IFNy and TNFa producing, degranulating cells and to improve blast-cytotoxicity after MLC. In vivo treatment of AML patients with Kit M may lead to anti-leukemic effects and contribute to stabilizing the disease or remissions. INCYT and DEG assays qualify to quantify potentially leukemia-specific cells on a single cell level and to predict the clinical course of patients under treatment.
Collapse
Affiliation(s)
- Olga Schutti
- Department for Hematopoetic Cell Transplantation, Med. III, University Hospital of Munich, 81377 Munich, Germany; (O.S.)
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Lara Klauer
- Department for Hematopoetic Cell Transplantation, Med. III, University Hospital of Munich, 81377 Munich, Germany; (O.S.)
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Tobias Baudrexler
- Department for Hematopoetic Cell Transplantation, Med. III, University Hospital of Munich, 81377 Munich, Germany; (O.S.)
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Florian Burkert
- Department for Hematopoetic Cell Transplantation, Med. III, University Hospital of Munich, 81377 Munich, Germany; (O.S.)
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Joerg Schmohl
- Department of Haematology and Oncology, University Hospital of Tuebingen, 72076 Tuebingen, Germany
| | - Marcus Hentrich
- Department of Haematology and Oncology, Red Cross Hospital of Munich, 80634 Munich, Germany
| | - Peter Bojko
- Department of Haematology and Oncology, Red Cross Hospital of Munich, 80634 Munich, Germany
| | - Doris Kraemer
- Department of Heamatology and Oncology, St.-Josefs-Hospital Hagen, 58097 Hagen, Germany
| | - Andreas Rank
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
- Department of Haematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Christoph Schmid
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
- Department of Haematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Helga Schmetzer
- Department for Hematopoetic Cell Transplantation, Med. III, University Hospital of Munich, 81377 Munich, Germany; (O.S.)
- Bavarian Cancer Research Center (BZKF), Comprehensive Cancer Center at University Hospital of Augsburg, 86156 Augsburg, Germany
| |
Collapse
|
2
|
Suresh M, Menne S. Recent Drug Development in the Woodchuck Model of Chronic Hepatitis B. Viruses 2022; 14:v14081711. [PMID: 36016334 PMCID: PMC9416195 DOI: 10.3390/v14081711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/24/2022] Open
Abstract
Infection with hepatitis B virus (HBV) is responsible for the increasing global hepatitis burden, with an estimated 296 million people being carriers and living with the risk of developing chronic liver disease and cancer. While the current treatment options for chronic hepatitis B (CHB), including oral nucleos(t)ide analogs and systemic interferon-alpha, are deemed suboptimal, the path to finding an ultimate cure for this viral disease is rather challenging. The lack of suitable laboratory animal models that support HBV infection and associated liver disease progression is one of the major hurdles in antiviral drug development. For more than four decades, experimental infection of the Eastern woodchuck with woodchuck hepatitis virus has been applied for studying the immunopathogenesis of HBV and developing new antiviral therapeutics against CHB. There are several advantages to this animal model that are beneficial for performing both basic and translational HBV research. Previous review articles have focused on the value of this animal model in regard to HBV replication, pathogenesis, and immune response. In this article, we review studies of drug development and preclinical evaluation of direct-acting antivirals, immunomodulators, therapeutic vaccines, and inhibitors of viral entry, gene expression, and antigen release in the woodchuck model of CHB since 2014 until today and discuss their significance for clinical trials in patients.
Collapse
|
3
|
Leukemia derived dendritic cell (DC leu) mediated immune response goes along with reduced (leukemia-specific) regulatory T-cells. Immunobiology 2022; 227:152237. [PMID: 35749805 DOI: 10.1016/j.imbio.2022.152237] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/18/2022] [Accepted: 06/07/2022] [Indexed: 12/17/2022]
Abstract
The blastmodulatory Kit-M, composed of granulocyte-macrophage colony-stimulating-factor (GM-CSF) and Prostaglandin E1 (PGE1), is known to convert myeloid leukaemic blasts (from AML patients) into leukaemia derived dendritic cells (DCleu), which activate immunoreactive cells to gain antileukemic/leukaemia-specific activity. In this study we had a special focus on the influence of Kit-M treated, DC/DCleu containing patients'whole blood (WB, n = 16) on the provision of immunosuppressive regulatory T-cells. We could confirm that Kit-M significantly increased frequencies of (mature) dendritic cells (DC) and DCleu from leukemic whole blood (WB) without induction of blast proliferation. After mixed lymphocyte culture (MLC) with patients' T-cells we confirmed that DCleu mediated leukemia-specific responses- going along with activated and leukemia-specific T- and NK-cells in an intracellular cytokine staining assay (ICS) and a degranulation assay (Deg)- resulted in an increased anti-leukemic cytotoxicity (Cytotoxicity Fluorolysis Assay = CTX). We could demonstrate that (leukemia-specific) CD4+ and CD8+ regulatory T-cell population (Treg) decreased significantly after MLC compared to controls. We found significant positive correlations of leukemia-specific CD3+CD4+ cells with frequencies of (mature) DCleu. Achieved anti-leukemic cytotoxicity correlated significantly positive with leukemia-specific CD3+CD8+ cells and significantly negatively with (leukemia-specific) Treg. In summary we demonstrate that immunesuppressive (leukemia-specific) regulatory T-cells are significantly downregulated after Kit-M triggered MLC- going along with a (reinstalled) antileukemic reactivity of the immune system (as demonstrated with functional assays ICS, Deg, CTX).
Collapse
|
4
|
Menne S, Wildum S, Steiner G, Suresh M, Korolowicz K, Balarezo M, Yon C, Murreddu M, Hong X, Kallakury BV, Tucker R, Yang S, Young JAT, Javanbakht H. Efficacy of an Inhibitor of Hepatitis B Virus Expression in Combination With Entecavir and Interferon-α in Woodchucks Chronically Infected With Woodchuck Hepatitis Virus. Hepatol Commun 2020; 4:916-931. [PMID: 32490326 PMCID: PMC7262289 DOI: 10.1002/hep4.1502] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/13/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
RG7834 is a small‐molecule inhibitor of hepatitis B virus (HBV) gene expression that significantly reduces the levels of hepatitis B surface antigen (HBsAg) and HBV DNA in a humanized liver HBV mouse model. In the current study, we evaluated the potency of RG7834 in the woodchuck model of chronic HBV infection, alone and in combination with entecavir (ETV) and/or woodchuck interferon‐α (wIFN‐α). RG7834 reduced woodchuck hepatitis virus (WHV) surface antigen (WHsAg) by a mean of 2.57 log10 from baseline and WHV DNA by a mean of 1.71 log10. ETV + wIFN‐α reduced WHsAg and WHV DNA by means of 2.40 log10 and 6.70 log10, respectively. The combination of RG7834, ETV, and wIFN‐α profoundly reduced WHsAg and WHV DNA levels by 5.00 log10 and 7.46 log10, respectively. However, both viral parameters rebounded to baseline after treatment was stopped and no antibody response against WHsAg was observed. Effects on viral RNAs were mainly seen with the triple combination treatment, reducing both pregenomic RNA (pgRNA) and WHsAg RNA, whereas RG7834 mainly reduced WHsAg RNA and ETV mainly affected pgRNA. When WHsAg was reduced by the triple combination, peripheral blood mononuclear cells (PBMCs) proliferated significantly in response to viral antigens, but the cellular response was diminished after WHsAg returned to baseline levels during the off‐treatment period. Consistent with this, Pearson correlation revealed a strong negative correlation between WHsAg levels and PBMC proliferation in response to peptides covering the entire WHsAg and WHV nucleocapsid antigen. Conclusion: A fast and robust reduction of WHsAg by combination therapy reduced WHV‐specific immune dysfunction in the periphery. However, the magnitude and/or duration of the induced cellular response were not sufficient to achieve a sustained antiviral response.
Collapse
Affiliation(s)
- Stephan Menne
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | - Steffen Wildum
- Roche Pharma Research and Early Development Roche Innovation Center Basel Basel Switzerland
| | - Guido Steiner
- Roche Pharma Research and Early Development Roche Innovation Center Basel Basel Switzerland
| | - Manasa Suresh
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | - Kyle Korolowicz
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | - Maria Balarezo
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | - Changsuek Yon
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | - Marta Murreddu
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | - Xupeng Hong
- Department of Microbiology and Immunology Georgetown University Medical Center Washington DC
| | | | - Robin Tucker
- Department of Pharmacology Georgetown University Medical Center Washington DC
| | - Song Yang
- Roche Pharma Research and Early Development Roche Innovation Center Shanghai Shanghai China
| | - John A T Young
- Roche Pharma Research and Early Development Roche Innovation Center Basel Basel Switzerland
| | - Hassan Javanbakht
- Roche Pharma Research and Early Development Roche Innovation Center Basel Basel Switzerland
| |
Collapse
|
5
|
Molecular characterization of woodchuck IFI16 and AIM2 and their expression in woodchucks infected with woodchuck hepatitis virus (WHV). Sci Rep 2016; 6:28776. [PMID: 27354260 PMCID: PMC4926060 DOI: 10.1038/srep28776] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/08/2016] [Indexed: 12/25/2022] Open
Abstract
IFI16 and AIM2 are important DNA sensors in antiviral immunity. To characterize these two molecules in a woodchuck model, which is widely used to study hepatitis B virus (HBV) infection, we cloned and analyzed the complete coding sequences (CDSs) of woodchuck IFI16 and AIM2, and found that AIM2 was highly conserved in mammals, whereas the degree of sequence identity between woodchuck IFI16 and its mammalian orthologues was low. IFI16 and IFN-β were upregulated following VACV ds 70 mer transfection, while AIM2 and IL-1β were upregulated following poly (dA:dT) transfection, both in vitro and in vivo; IFI16-targeted siRNA decreased the transcription of IFI16 and IFN-β stimulated by VACV ds 70 mer, and AIM2 siRNA interference downregulated AIM2 and IL-1β transcripts stimulated by poly (dA:dT), in vitro, suggesting that woodchuck IFI16 and AIM2 may play pivotal roles in the DNA-mediated induction of IFN-β and IL-1β, respectively. IFI16 and AIM2 transcripts were upregulated in the liver and spleen following acute WHV infection, while IFI16 was downregulated in the liver following chronic infection, implying that IFI16 and AIM2 may be involved in WHV infection. These data provide the basis for the study of IFI16- and AIM2-mediated innate immunity using the woodchuck model.
Collapse
|
6
|
Li X, Liu G, Chen M, Yang Y, Xie Y, Kong X. A Novel Hydrodynamic Injection Mouse Model of HBV Genotype C for the Study of HBV Biology and the Anti-Viral Activity of Lamivudine. HEPATITIS MONTHLY 2016; 16:e34420. [PMID: 27195013 PMCID: PMC4867405 DOI: 10.5812/hepatmon.34420] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/06/2016] [Accepted: 01/15/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Absence of an immunocompetent mouse model of persistent hepatitis B virus (HBV) infection has hindered the research of HBV infection and the development of antiviral medications. OBJECTIVES In the present study, we aimed to develop a novel HBV genotype C mouse model by hydrodynamic injection (HI) and then used it to evaluate the antiviral activity of lamivudine. MATERIALS AND METHODS A quantity of 15 μg of HBV plasmid [pcDNA3.1 (+)-HBV1.3C], adeno-associated virus-HBV1.3C (pAAV-HBV1.3C) or pAAV-HBV1.2A) were injected into male C57BL/6 mice, by HI, accounting for a total of 13 mice per group. Then, lamivudine was administered to mice with sustained HBV viremia, for 4 weeks. Real-time polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry methods were used to detect HBsAg, HBeAg, HBsAb, HBcAg and HBV DNA, in serum or liver of the mice, at indicated time points. RESULTS In 60% of the mice injected with pcDNA3.1 (+)-HBV1.3C, HBsAg, HBeAg, HBcAg and HBV DNA persisted for > 20 weeks in liver, post-injection, with no HBsAb appearance. Meanwhile, no significant inflammation was observed in these mice. Compared with pAAV-HBV1.2A and pAAV-HBV1.3C, pcDNA3.1 (+)-HBV1.3C administration led to higher and longer HBV viremia. Furthermore, serum HBV DNA was significantly reduced by lamivudine, after 4 weeks administration, and returned to the original level, after ceasing administration for 1 week, in the mice. CONCLUSIONS In conclusion, our observations indicated that pcDNA3.1 (+)-HBV1.3C was superior to AAV/HBV plasmid for establishment of persistent HBV infection by HI, in vivo, and this mouse model could be useful for studies of hepatitis virology and for the development of innovatory treatments for HBV infections.
Collapse
Affiliation(s)
- Xiumei Li
- Liver Disease Key Lab, 458 Hospital of PLA, Guangzhou, China
| | - Guangze Liu
- Liver Disease Key Lab, 458 Hospital of PLA, Guangzhou, China
| | - Meijuan Chen
- Liver Disease Key Lab, 458 Hospital of PLA, Guangzhou, China
| | - Yang Yang
- Liver Disease Key Lab, 458 Hospital of PLA, Guangzhou, China
| | - Yong Xie
- Liver Disease Key Lab, 458 Hospital of PLA, Guangzhou, China
| | - Xiangping Kong
- Liver Disease Key Lab, 458 Hospital of PLA, Guangzhou, China
- Corresponding Author: Xiangping Kong, Liver Disease Key Lab, 458 Hospital of PLA, 801 Dongfengdong Road, 510600, Guangzhou, Guangdong, China. Tel: +86-2087395343, Fax: +86-2087371180, E-mail:
| |
Collapse
|
7
|
Zhang E, Kosinska A, Lu M, Yan H, Roggendorf M. Current status of immunomodulatory therapy in chronic hepatitis B, fifty years after discovery of the virus: Search for the "magic bullet" to kill cccDNA. Antiviral Res 2015; 123:193-203. [PMID: 26476376 DOI: 10.1016/j.antiviral.2015.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/09/2015] [Accepted: 10/09/2015] [Indexed: 12/12/2022]
Abstract
Chronic hepatitis B (CHB) is currently treated with IFN-α and nucleos(t)ide analogues, which have many clinical benefits, but there is no ultimate cure. The major problem consists in the persistence of cccDNA in infected hepatocytes. Because no antiviral drug has been evaluated which significantly reduces copies of cccDNA, cytolytic and noncytolytic approaches are needed. Effective virus-specific T- and B-cell responses remain crucial in eliminating cccDNA-carrying hepatocytes and for the long-term control of HBV infection. Reduction of viremia by antiviral drugs provides a window for reconstitution of an HBV-specific immune response. Preclinical studies in mice and woodchucks have shown that immunostimulatory strategies, such as prime-boost vaccination and PD-1 blockade, can boost a weak virus-specific T cell response and lead to effective control of HBV infection. Based on data obtained in our preclinical studies, the combination of antiviral drugs and immunomodulators may control HBV viremia during a patient's drug-off period. In this article, we review current immune-modulatory approaches for the treatment of chronic hepatitis B and the elimination of cccDNA in preclinical models. This article forms part of a symposium in Antiviral Research on "An unfinished story: from the discovery of the Australia antigen to the development of new curative therapies for hepatitis".
Collapse
Affiliation(s)
- Ejuan Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Anna Kosinska
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Michael Roggendorf
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany; Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
8
|
Mulrooney-Cousins PM, Michalak TI. Asymptomatic Hepadnaviral Persistence and Its Consequences in the Woodchuck Model of Occult Hepatitis B Virus Infection. J Clin Transl Hepatol 2015; 3:211-9. [PMID: 26623268 PMCID: PMC4663203 DOI: 10.14218/jcth.2015.00020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/20/2015] [Accepted: 07/21/2015] [Indexed: 02/06/2023] Open
Abstract
Woodchuck hepatitis virus (WHV) is molecularly and pathogenically closely related to hepatitis B virus (HBV). Both viruses display tropism towards hepatocytes and cells of the immune system and cause similar liver pathology, where acute hepatitis can progress to chronic hepatitis and to hepatocellular carcinoma (HCC). Two forms of occult hepadnaviral persistence were identified in the woodchuck-WHV model: secondary occult infection (SOI) and primary occult infection (POI). SOI occurs after resolution of a serologically apparent infection with hepatitis or after subclinical serologically evident virus exposure. POI is caused by small amounts of virus and progresses without serological infection markers, but the virus genome and its replication are detectable in the immune system and with time in the liver. SOI can be accompanied by minimal hepatitis, while the hallmark of POI is normal liver morphology. Nonetheless, HCC develops in about 20% of animals with SOI or POI within 3 to 5 years. The virus persists throughout the lifespan in both SOI and POI at serum levels rarely greater than 100 copies/mL, causes hepatitis and HCC when concentrated and administered to virus-naïve woodchucks. SOI is accompanied by virus-specific T and B cell immune responses, while only virus-specific T cells are detected in POI. SOI coincides with protection against reinfection, while POI does not and hepatitis develops after challenge with liver pathogenic doses >1000 virions. Both SOI and POI are associated with virus DNA integration into the liver and the immune system genomes. Overall, SOI and POI are two distinct forms of silent hepadnaviral persistence that share common characteristics. Here, we review findings from the woodchuck model and discuss the relevant observations made in human occult HBV infection (OBI).
Collapse
Affiliation(s)
| | - Tomasz I. Michalak
- Correspondence to: Tomasz I. Michalak, Molecular Virology and Hepatology Research Group, Faculty of Medicine, Health Sciences Centre, Memorial University, St. John’s, NL A1B 3V6, Canada. Tel: +1-709-777-7301, Fax: +1-709-777-8279, E-mail:
| |
Collapse
|
9
|
Zeisel MB, Lucifora J, Mason WS, Sureau C, Beck J, Levrero M, Kann M, Knolle PA, Benkirane M, Durantel D, Michel ML, Autran B, Cosset FL, Strick-Marchand H, Trépo C, Kao JH, Carrat F, Lacombe K, Schinazi RF, Barré-Sinoussi F, Delfraissy JF, Zoulim F. Towards an HBV cure: state-of-the-art and unresolved questions--report of the ANRS workshop on HBV cure. Gut 2015; 64:1314-26. [PMID: 25670809 DOI: 10.1136/gutjnl-2014-308943] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/10/2015] [Indexed: 12/11/2022]
Abstract
HBV infection is a major cause of liver cirrhosis and hepatocellular carcinoma. Although HBV infection can be efficiently prevented by vaccination, and treatments are available, to date there is no reliable cure for the >240 million individuals that are chronically infected worldwide. Current treatments can only achieve viral suppression, and lifelong therapy is needed in the majority of infected persons. In the framework of the French National Agency for Research on AIDS and Viral Hepatitis 'HBV Cure' programme, a scientific workshop was held in Paris in June 2014 to define the state-of-the-art and unanswered questions regarding HBV pathobiology, and to develop a concerted strategy towards an HBV cure. This review summarises our current understanding of HBV host-interactions leading to viral persistence, as well as the roadblocks to be overcome to ultimately address unmet medical needs in the treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Mirjam B Zeisel
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France Université de Strasbourg, Strasbourg, France
| | - Julie Lucifora
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
| | | | - Camille Sureau
- INTS, Laboratoire de Virologie Moléculaire, Paris, France
| | - Jürgen Beck
- Department of Internal Medicine 2/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Massimo Levrero
- Center for Life Nanosciences (CNLS)-IIT/Sapienza, Rome, Italy Laboratory of Gene Expression, Department of Internal Medicine (DMISM), Sapienza University of Rome, Italy
| | - Michael Kann
- Université de Bordeaux, Microbiologie fondamentale et Pathogénicité, UMR 5234, Bordeaux, France CNRS, Microbiologie fondamentale et Pathogénicité, UMR 5234, Bordeaux, France CHU de Bordeaux, Bordeaux, France
| | - Percy A Knolle
- Technische Universität München, Institut für Molekulare Immunologie, München, Germany
| | - Monsef Benkirane
- Institut de Génétique Humaine, Laboratoire de Virologie Moléculaire, CNRS UPR1142, Montpellier, France
| | - David Durantel
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
| | - Marie-Louise Michel
- Laboratoire de Pathogenèse des Virus de l'Hépatite B, Département de Virologie, Institut Pasteur, Paris, France
| | - Brigitte Autran
- Laboratory of Immunity and Infection, Inserm U945, Paris, France Laboratory of Immunity and Infection, UPMC University Paris 06, Unité mixte de recherche-S945, Paris, France Inserm, IFR 113, Immunité-Cancer-Infection, Paris, France
| | - François-Loïc Cosset
- CIRI-International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France. Inserm, U1111, Lyon, France Ecole Normale Supérieure de Lyon, Lyon, France. CNRS, UMR5308, Lyon, France LabEx Ecofect, Université de Lyon, Lyon, France
| | | | - Christian Trépo
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France Department of Hepatology, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| | - Jia-Horng Kao
- Department of Internal Medicine, Department of Medical Research, Graduate Institute of Clinical Medicine, and Hepatitis Research Center, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Fabrice Carrat
- Inserm, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France Sorbonne Universités, UPMC Univ Paris 06, Paris, France Assistance Publique Hôpitaux de Paris, Hôpital Saint Antoine, Paris, France
| | - Karine Lacombe
- Inserm, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France Sorbonne Universités, UPMC Univ Paris 06, Paris, France Assistance Publique Hôpitaux de Paris, Hôpital Saint Antoine, Paris, France
| | - Raymond F Schinazi
- Center for AIDS Research, Emory University School of Medicine and Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Françoise Barré-Sinoussi
- Inserm and Unit of Regulation of Retroviral Infections, Department of Virology, Institut Pasteur, Paris, France
| | | | - Fabien Zoulim
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France Department of Hepatology, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
10
|
Marimani M, Hean J, Bloom K, Ely A, Arbuthnot P. Recent advances in developing nucleic acid-based HBV therapy. Future Microbiol 2014; 8:1489-504. [PMID: 24199806 DOI: 10.2217/fmb.13.87] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Chronic HBV infection remains an important public health problem and currently licensed therapies rarely prevent complications of viral persistence. Silencing HBV gene expression using gene therapy, particularly with exogenous activators of RNAi, holds promise for developing an HBV gene therapy. However, immune stimulation, off-targeting effects and inefficient delivery of RNAi activators remain problematic. Several new approaches have recently been employed to address these issues. Chemical modifications to anti-HBV synthetic siRNAs have been investigated and a variety of vectors are being developed for delivery of RNAi effectors. In this article, we review the potential utility of gene therapy for treating HBV infection.
Collapse
Affiliation(s)
- Musa Marimani
- Antiviral Gene Therapy Research Unit, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | | |
Collapse
|
11
|
Wang B, Zhu Z, Zhu B, Wang J, Song Z, Huang S, Fan W, Tao Y, Wang Z, Wang H, Lu M, Yang D. Nucleoside analogues alone or combined with vaccination prevent hepadnavirus viremia and induce protective immunity: alternative strategy for hepatitis B virus post-exposure prophylaxis. Antiviral Res 2014; 105:118-25. [PMID: 24583157 DOI: 10.1016/j.antiviral.2014.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 01/08/2023]
Abstract
OBJECTIVES The current strategies for hepatitis B virus (HBV) post-exposure prophylaxis (PEP) are not generally available in remote and rural areas of developing countries and/or carry potential risks for infection with blood-borne transmitted pathogens. Nucleotide analogues (NAs) are successfully used for human immunodeficiency virus PEP, and maybe effective for HBV PEP. In this study, we tested the NA-based strategies for HBV PEP using the Chinese woodchuck model. METHODS Chinese woodchucks were inoculated intravenously with different doses of woodchuck hepatitis virus (WHV). A deoxyguanosine analogue entacavir (ETV), a DNA vaccine pWHcIm, or ETV plus pWHcIm were applied to the infected animals 24h later. Twenty weeks later, the animals were re-challenged with WHV to test for the presence of immunity against WHV. RESULTS Inoculation with different WHV doses had a strong influence on the course of WHV infection; NA alone or in combination with a DNA vaccine completely prevented viremia after a high dose of WHV inoculation in Chinese woodchucks and induced partial or complete protective immunity, respectively. CONCLUSIONS NA-based PEP strategies (NA alone or in combination with vaccine) may be an alternative of HBV PEP, especially in those living in the remote and rural areas of the developing countries and the non-responders to the current vaccine, and may be valuable in the PEP of HBV and HIV co-infection after occupational and non-occupational exposure. Further clinical studies are warranted to confirm the valuable of NA-based strategies in HBV PEP.
Collapse
Affiliation(s)
- Baoju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenni Zhu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bin Zhu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junzhong Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhitao Song
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shunmei Huang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Fan
- Qinghai Institute for Endemic Disease Prevention and Control, Xining 811602, China
| | - Yuanqing Tao
- Qinghai Institute for Endemic Disease Prevention and Control, Xining 811602, China
| | - Zhongdong Wang
- Qinghai Institute for Endemic Disease Prevention and Control, Xining 811602, China
| | - Hu Wang
- Qinghai Institute for Endemic Disease Prevention and Control, Xining 811602, China
| | - Mengji Lu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institut für Virologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen 45122, Germany.
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
12
|
Locarnini S, Littlejohn M, Aziz MN, Yuen L. Possible origins and evolution of the hepatitis B virus (HBV). Semin Cancer Biol 2013; 23:561-75. [PMID: 24013024 DOI: 10.1016/j.semcancer.2013.08.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/27/2013] [Indexed: 02/06/2023]
Abstract
All members of the family Hepadnaviridae are primarily viruses which contain double-stranded DNA genomes that are replicated via reverse transcription of a pregenomic RNA template. There are two subgroups within this family: mammalian and avian. The avian member's include the duck hepatitis B virus (DHBV), heron hepatitis B virus, Ross goose hepatitis B virus, stork hepatitis B virus and the recently identified parrot hepatitis B virus. More recently, the detection of endogenous avian hepadnavirus DNA integrated into the genomes of zebra finches has revealed a deep evolutionary origin of hepadnaviruses that was not previously recognised, dating back over 40 million years ago. The non-primate mammalian members of the Hepadnaviridae include the woodchuck hepatitis virus (WHV), the ground squirrel hepatitis virus and arctic squirrel virus, as well as the recently described bat hepatitis virus. The identification of hepatitis B virus (HBV) in higher primates such as chimpanzee, gorilla, orangutan, and gibbons that cluster with the human genotypes further implies a more complex origin of this virus. By studying the molecular epidemiology of HBV in indigenous and relict populations in Asia-Pacific we propose a model for the origin and evolution of HBV that involves multiple cross-species transmissions and subsequent recombination events on a background of genotype C HBV infection.
Collapse
Affiliation(s)
- Stephen Locarnini
- Research & Molecular Development, Victorian Infectious Diseases Reference Laboratory, Melbourne, Australia.
| | | | | | | |
Collapse
|
13
|
Liu B, Wen X, Huang C, Wei Y. Unraveling the complexity of hepatitis B virus: from molecular understanding to therapeutic strategy in 50 years. Int J Biochem Cell Biol 2013; 45:1987-96. [PMID: 23819994 DOI: 10.1016/j.biocel.2013.06.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 06/18/2013] [Accepted: 06/21/2013] [Indexed: 02/05/2023]
Abstract
Hepatitis B virus (HBV) is a well-known hepadnavirus with a double-stranded circular DNA genome. Although HBV was first described approximately 50 years ago, the precise mechanisms of HBV infection and effective therapeutic strategies remain unclear. Here, we focus on summarizing the complicated mechanisms of HBV replication and infection, as well as genomic factors and epigenetic regulation. Additionally, we discuss in vivo models of HBV, as well as diagnosis, prevention and therapeutic drugs for HBV. Together, the data in this 50-year review may provide new clues to elucidate molecular mechanisms of HBV pathogenesis and shed new light on the future HBV therapies.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | |
Collapse
|
14
|
Saade F, Buronfosse T, Guerret S, Pradat P, Chevallier M, Zoulim F, Jamard C, Cova L. In vivo infectivity of liver extracts after resolution of hepadnaviral infection following therapy associating DNA vaccine and cytokine genes. J Viral Hepat 2013; 20:e56-65. [PMID: 23490390 DOI: 10.1111/jvh.12023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/24/2012] [Indexed: 12/19/2022]
Abstract
DNA-based vaccination appears of promise for chronic hepatitis B immunotherapy, although there is an urgent need to increase its efficacy. In this preclinical study, we evaluated the therapeutic benefit of cytokine (IL-2, IFN-γ) genes co-delivery with DNA vaccine targeting hepadnaviral proteins in the chronic duck hepatitis B virus (DHBV) infection model. Then, we investigated the persistence of replication-competent virus in the livers of apparently resolved animals. DHBV carriers received four injections of plasmids encoding DHBV envelope and core alone or co-delivered with duck IL-2 (DuIL-2) or duck IFN-γ (DuIFN-γ) plasmids. After long-term (8 months) follow-up, viral covalently closed circular (ccc) DNA was analysed in duck necropsy liver samples. Liver homogenates were also tested for in vivo infectivity in neonatal ducklings. Co-delivery of DuIFN-γ resulted in significantly lower mean viremia starting from week 21. Viral cccDNA was undetectable by conventional methods in the livers of 25% and 57% of animals co-immunized with DuIL-2 and DuIFN-γ, respectively. Interestingly, inoculation of liver homogenates from 7 such apparently resolved animals, exhibiting cccDNA undetectable in Southern blotting and DHBV expression undetectable or restricted to few hepatocytes, revealed that three liver homogenates transmitted high-titre viremia (3-5×10(10) vge/mL) to naïve animals. In conclusion, our results indicate that IFN-γ gene co-delivery considerably enhances immunotherapeutic efficacy of DNA vaccine targeting hepadnaviral proteins. Importantly, we also showed that livers exhibiting only minute amounts of hepadnaviral cccDNA could induce extremely high-titre infection, highlighting the caution that should be taken in occult hepatitis B patients to prevent HBV transmission in liver transplantation context.
Collapse
Affiliation(s)
- F Saade
- Université de Lyon, Lyon, Lyon, France
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Molecular characterization of the type I IFN receptor in two woodchuck species and detection of its expression in liver samples from woodchucks infected with woodchuck hepatitis virus (WHV). Cytokine 2012; 60:179-85. [PMID: 22705153 DOI: 10.1016/j.cyto.2012.05.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 05/15/2012] [Accepted: 05/17/2012] [Indexed: 02/06/2023]
Abstract
Type I interferons (IFN-α/β) serve as the first line of defense against viral infection and share the same type I IFN receptor (IFNAR) complex, which is composed of IFNAR1 and -2. The Eastern woodchuck (Marmota monax) and Chinese woodchuck (Marmota himalayana) are suitable for studying hepatitis B virus (HBV) infection. Here, the complete or partial sequences of the IFNARs of both species were obtained and analyzed. Small interference RNAs targeting wIFNAR1 and -2 specifically down-regulated the expression of wIFNAR1 and -2 and the IFN-stimulated gene MxA in a woodchuck cell line, respectively. IFNAR2 was significantly up-regulated in primary woodchuck hepatocytes stimulated with IFN-α or -γ. The expression of woodchuck IFNAR1 and -2 was decreased in woodchucks chronically infected with woodchuck hepatitis virus (WHV). These results are essential for studying type I IFN-related innate immunity and therapy in hepadnaviral infection in the woodchuck model.
Collapse
|
16
|
Kutscher S, Bauer T, Dembek C, Sprinzl M, Protzer U. Design of therapeutic vaccines: hepatitis B as an example. Microb Biotechnol 2012; 5:270-82. [PMID: 21958338 PMCID: PMC3815787 DOI: 10.1111/j.1751-7915.2011.00303.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/22/2011] [Accepted: 08/22/2011] [Indexed: 12/18/2022] Open
Abstract
Therapeutic vaccines are currently developed for chronic viral infections, such as human papillomavirus (HPV), human immunodeficiency virus (HIV), herpesvirus and hepatitis B (HBV) and C (HCV) virus infections. As an alternative to antiviral treatment or to support only partially effective therapy a therapeutic vaccine shall activate the patient's immune system to fight and finally control or ideally even eliminate the virus. Whereas the success of prophylactic vaccination is based on rapid neutralization of the invading pathogen by antibodies, virus control and elimination of infected cells require T cells. Therefore, induction of a multi-specific and multifunctional T-cell response against key viral antigens is a paradigm of therapeutic vaccination--besides activation of a humoral immune response to limit virus spread. In this review, we describe options to develop a therapeutic vaccine for chronic viral infections using HBV as a promising example.
Collapse
Affiliation(s)
| | | | | | | | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstr. 30, 81675 München, Germany
| |
Collapse
|
17
|
Abstract
Chronic HBV infection remains a leading cause of serious liver disease and hepatocellular carcinoma in spite of the existence of an effective preventive vaccine. Although the actual antiviral treatments have greatly improved, they only rarely clear viral infection. In this regard, therapeutic DNA vaccination appears to have great promise to stimulate and restore the impaired immune responses in chronic HBV carriers. This review examines preclinical studies of preventive and therapeutic DNA vaccines in different animal models (mouse, woodchuck and duck) and the first clinical studies in chronically infected patients. We also focused on different approaches aimed at enhancing the effectiveness of DNA vaccines such as combination therapy with antiviral drugs and in vivo DNA electroporation.
Collapse
Affiliation(s)
- Lucyna Cova
- Université Claude Bernard Lyon 1, Inserm U1052, CRCL team 15, 151 cours Albert Thomas, 69003 Lyon, France
| |
Collapse
|
18
|
Yang Y, Wang B, Yang D, Lu M, Xu Y. Prokaryotic expression of woodchuck cytotoxic T lymphocyte antigen 4 (wCTLA-4) and preparation of polyclonal antibody to wCTLA-4. Protein Expr Purif 2011; 81:181-5. [PMID: 22040606 DOI: 10.1016/j.pep.2011.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/14/2011] [Accepted: 10/15/2011] [Indexed: 12/17/2022]
Abstract
Cytotoxic T lymphocyte antigen 4 (CTLA-4) is an inhibitory T cell receptor predominately expressed on activated T cells and plays an important role in regulation of specific T cell responses to viral infection. The woodchuck model is an informative animal model for hepatitis B virus (HBV) infection. In this study, the extracellular region of woodchuck CTLA-4 (wCTLA-4) was cloned and the fusion protein of GST-wCTLA-4 was expressed and purified. Polyclonal antibody against GST-wCTLA-4 (anti-GST-wCTLA-4) was prepared. The full length wCTLA-4 protein expressed in transfected baby hamster kidney cells was detected by anti-GST-wCTLA-4 in western blot analysis and immunofluorescence staining. Anti-GST-wCTLA-4 provides a useful tool to study the role of CTLA-4 in T-cell response in the woodchuck model. Further, the blocking of CTLA-4 with anti-GST-wCTLA-4, as a novel therapy approach for chronic hepatitis B virus infection, could be studied in woodchuck model now.
Collapse
Affiliation(s)
- Yinke Yang
- Department of Microbiology, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, China
| | | | | | | | | |
Collapse
|
19
|
Therapeutic vaccines and immune-based therapies for the treatment of chronic hepatitis B: perspectives and challenges. J Hepatol 2011; 54:1286-96. [PMID: 21238516 DOI: 10.1016/j.jhep.2010.12.031] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 11/23/2010] [Accepted: 12/20/2010] [Indexed: 12/24/2022]
Abstract
The treatment of chronic hepatitis B virus (HBV) infection has greatly improved over the last 10 years, but alternative treatments are still needed. Therapeutic vaccination is a promising new strategy for controlling chronic infection. However, this approach has not been as successful as initially anticipated for chronic hepatitis B. General impairment of the immune responses generated during persistent HBV infection, with exhausted T cells not responding correctly to therapeutic vaccination, is probably responsible for the poor clinical responses observed to date. Intensive research efforts are now focusing on increasing the efficacy of therapeutic vaccination without causing liver disease. Here we describe new approaches to use with therapeutic vaccination, in order to overcome the inhibitory mechanisms impairing immune responses. We also describe innovative strategies for generating functional immune responses and inducing sustained control of this persistent infection.
Collapse
|