1
|
Ringelhan M, Schuehle S, van de Klundert M, Kotsiliti E, Plissonnier ML, Faure-Dupuy S, Riedl T, Lange S, Wisskirchen K, Thiele F, Cheng CC, Yuan D, Leone V, Schmidt R, Hünergard J, Geisler F, Unger K, Algül H, Schmid RM, Rad R, Wedemeyer H, Levrero M, Protzer U, Heikenwalder M. HBV-related HCC development in mice is STAT3 dependent and indicates an oncogenic effect of HBx. JHEP Rep 2024; 6:101128. [PMID: 39290403 PMCID: PMC11406364 DOI: 10.1016/j.jhepr.2024.101128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 09/19/2024] Open
Abstract
Background & Aims Although most hepatocellular carcinoma (HCC) cases are driven by hepatitis and cirrhosis, a subset of patients with chronic hepatitis B develop HCC in the absence of advanced liver disease, indicating the oncogenic potential of hepatitis B virus (HBV). We investigated the role of HBV transcripts and proteins on HCC development in the absence of inflammation in HBV-transgenic mice. Methods HBV-transgenic mice replicating HBV and expressing all HBV proteins from a single integrated 1.3-fold HBV genome in the presence or absence of wild-type HBx (HBV1.3/HBVxfs) were analyzed. Flow cytometry, molecular, histological and in vitro analyses using human cell lines were performed. Hepatocyte-specific Stat3- and Socs3-knockout was analyzed in HBV1.3 mice. Results Approximately 38% of HBV1.3 mice developed liver tumors. Protein expression patterns, histology, and mutational landscape analyses indicated that tumors resembled human HCC. HBV1.3 mice showed no signs of active hepatitis, except STAT3 activation, up to the time point of HCC development. HBV-RNAs covering HBx sequence, 3.5-kb HBV RNA and HBx-protein were detected in HCC tissue. Interestingly, HBVxfs mice expressing all HBV proteins except a C-terminally truncated HBx (without the ability to bind DNA damage binding protein 1) showed reduced signs of DNA damage response and had a significantly reduced HCC incidence. Importantly, intercrossing HBV1.3 mice with a hepatocyte-specific STAT3-knockout abrogated HCC development. Conclusions Expression of HBV-proteins is sufficient to cause HCC in the absence of detectable inflammation. This indicates the oncogenic potential of HBV and in particular HBx. In our model, HBV-driven HCC was STAT3 dependent. Our study highlights the immediate oncogenic potential of HBV, challenging the idea of a benign highly replicative phase of HBV infection and indicating the necessity for an HBV 'cure'. Impact and implications Although most HCC cases in patients with chronic HBV infection occur after a sequence of liver damage and fibrosis, a subset of patients develops HCC without any signs of advanced liver damage. We demonstrate that the expression of all viral transcripts in HBV-transgenic mice suffices to induce HCC development independent of inflammation and fibrosis. These data indicate the direct oncogenic effects of HBV and emphasize the idea of early antiviral treatment in the 'immune-tolerant' phase (HBeAg-positive chronic HBV infection).
Collapse
Affiliation(s)
- Marc Ringelhan
- Second Medical Department, University Hospital Rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
- German Centre for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Svenja Schuehle
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Maarten van de Klundert
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Elena Kotsiliti
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | | | - Tobias Riedl
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Lange
- Second Medical Department, University Hospital Rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Wisskirchen
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Frank Thiele
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Detian Yuan
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Valentina Leone
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Research Unit for Radiation Cytogenetics, Helmholtz Munich, Neuherberg, Germany
| | - Ronny Schmidt
- Sciomics GmbH, Karl-Landsteiner-Straβe 6, 69151 Neckargemünd, Germany
| | - Juliana Hünergard
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Fabian Geisler
- Second Medical Department, University Hospital Rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Kristian Unger
- Research Unit for Radiation Cytogenetics, Helmholtz Munich, Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Hana Algül
- Second Medical Department, University Hospital Rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Comprehensive Cancer Center TUM (CCCMTUM), University Hospital rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Roland M Schmid
- Second Medical Department, University Hospital Rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Roland Rad
- Second Medical Department, University Hospital Rechts der Isar, Technical University of Munich, School of Medicine & Health, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Massimo Levrero
- INSERM Unit 1052, Cancer Research Center of Lyon, Lyon, France
- Hepatology Department, Hospices Civils de Lyon, Lyon, France
- Department of Internal Medicine - DMISM, Sapienza University, Rome, Italy
- Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Ulrike Protzer
- German Centre for Infection Research (DZIF), Munich Partner Site, Munich, Germany
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Virology, Technical University of Munich, School of Medicine & Health/Helmholtz Munich, Munich, Germany
- The M3 Research Center, Medical Faculty, University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
2
|
Tang J, Zhang J, Zhang G, Peng W, Ling N, Zhou Y, Xu H, Ren H, Chen M. Stat3 activation-triggered transcriptional networks govern the early stage of HBV-induced hepatic inflammation. mBio 2024; 15:e0306823. [PMID: 38440978 PMCID: PMC11005361 DOI: 10.1128/mbio.03068-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/01/2024] [Indexed: 03/06/2024] Open
Abstract
The chronic carrier state of the hepatitis B virus (HBV) often leads to the development of liver inflammation as carriers age. However, the exact mechanisms that trigger this hepatic inflammation remain poorly defined. We analyzed the sequential processes during the onset of liver inflammation based on time-course transcriptome and transcriptional regulatory networks in an HBV transgenic (HBV-Tg) mice model and chronic HBV-infected (CHB) patients (data from GSE83148). The key transcriptional factor (TF) responsible for hepatic inflammation occurrence was identified and then validated both in HBV-Tg mice and liver specimens from young CHB patients. By time-course analysis, an early stage of hepatic inflammation was demonstrated in 3-month-old HBV-Tg mice: a marked upregulation of genes related to inflammation (Saa1/2, S100a8/9/11, or Il1β), innate immunity (Tlr2, Tlr7, or Tlr8), and cells chemotaxis (Ccr2, Cxcl1, Cxcl13, or Cxcl14). Within CHB samples, a unique early stage of inflammation activation was discriminated from immune tolerance and immune activation groups based on distinct gene expression patterns. Enhanced activation of TF Stat3 was strongly associated with increased inflammatory gene expression in this early stage of inflammation. Expression of phosphorylated Stat3 was higher in liver specimens from young CHB patients with relatively higher alanine aminotransferase levels. Specific inhibition of Stat3 activation significantly attenuated the degree of liver inflammation, the expression of inflammation-related genes, and the inflammatory monocytes and macrophages in 3-month-old HBV-Tg mice. Stat3 activation is essential for hepatic inflammation occurrence and is a novel indicator of early-stage immune activation in chronic HBV carriers. IMPORTANCE Until now, it remains a mystery that chronic hepatitis B virus (HBV)-infected patients in the "immune tolerance phase" will transition to the "immune activation phase" as they age. In this study, we reveal that Stat3 activation-triggered hepatic transcriptional alterations are distinctive characteristics of the early stage of immune/inflammation activation in chronic HBV infection. For the first time, we discover a mechanism that might trigger the transition from immune tolerance to immune activation in chronic HBV carriers.
Collapse
Affiliation(s)
- Jinglin Tang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Transfusion Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiaxuan Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Laboratory Medicine, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Gaoli Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenhui Peng
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Ling
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingzhi Zhou
- Department of Infection, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei Xu
- Department of Infection, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Chen
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Michler T, Kosinska AD, Festag J, Bunse T, Su J, Ringelhan M, Imhof H, Grimm D, Steiger K, Mogler C, Heikenwalder M, Michel ML, Guzman CA, Milstein S, Sepp-Lorenzino L, Knolle P, Protzer U. Knockdown of Virus Antigen Expression Increases Therapeutic Vaccine Efficacy in High-Titer Hepatitis B Virus Carrier Mice. Gastroenterology 2020; 158:1762-1775.e9. [PMID: 32001321 DOI: 10.1053/j.gastro.2020.01.032] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/28/2019] [Accepted: 01/20/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Hepatitis B virus (HBV) infection persists because the virus-specific immune response is dysfunctional. Therapeutic vaccines might be used to end immune tolerance to the virus in patients with chronic infection, but these have not been effective in patients so far. In patients with chronic HBV infection, high levels of virus antigens might prevent induction of HBV-specific immune responses. We investigated whether knocking down expression levels of HBV antigens in liver might increase the efficacy of HBV vaccines in mice. METHODS We performed studies with male C57BL/6 mice that persistently replicate HBV (genotype D, serotype ayw)-either from a transgene or after infection with an adeno-associated virus that transferred an overlength HBV genome-and expressed HB surface antigen at levels relevant to patients. Small hairpin or small interfering (si)RNAs against the common 3'-end of all HBV transcripts were used to knock down antigen expression in mouse hepatocytes. siRNAs were chemically stabilized and conjugated to N-acetylgalactosamine to increase liver uptake. Control mice were given either entecavir or non-HBV-specific siRNAs and vaccine components. Eight to 12 weeks later, mice were immunized twice with a mixture of adjuvanted HBV S and core antigen, followed by a modified Vaccinia virus Ankara vector to induce HBV-specific B- and T-cell responses. Serum and liver samples were collected and analyzed for HBV-specific immune responses, liver damage, and viral parameters. RESULTS In both models of HBV infection, mice that express hepatocyte-specific small hairpin RNAs or that were given subcutaneous injections of siRNAs had reduced levels of HBV antigens, HBV replication, and viremia (1-3 log10 reduction) compared to mice given control RNAs. Vaccination induced production of HBV-neutralizing antibodies and increased numbers and functionality of HBV-specific, CD8+ T cells in mice with low, but not in mice with high, levels of HBV antigen. Mice with initially high titers of HBV and knockdown of HBV antigen expression, but not mice with reduced viremia after administration of entecavir, developed polyfunctional, HBV-specific CD8+ T cells, and HBV was eliminated. CONCLUSIONS In mice with high levels of HBV replication, knockdown of HBV antigen expression along with a therapeutic vaccination strategy, but not knockdown alone, increased numbers of effector T cells and eliminated the virus. These findings indicate that high titers of virus antigens reduce the efficacy of therapeutic vaccination. Anti-HBV siRNAs and therapeutic vaccines are each being tested in clinical trials-their combination might cure chronic HBV infection.
Collapse
Affiliation(s)
- Thomas Michler
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany; German Center for Infection Research, Münich, Heidelberg, Germany
| | - Anna D Kosinska
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany; German Center for Infection Research, Münich, Heidelberg, Germany
| | - Julia Festag
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany
| | - Till Bunse
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany; German Center for Infection Research, Münich, Heidelberg, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany
| | - Marc Ringelhan
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany; Department of Internal Medicine II, University Hospital rechts der Isar, Technical University of Munich, Münich, Germany
| | - Hortenzia Imhof
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany
| | - Dirk Grimm
- German Center for Infection Research, Münich, Heidelberg, Germany; Department of Infectious Diseases/Virology, Heidelberg University Hospital, BioQuant, Heidelberg, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Münich, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Münich, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | | | - Carlos A Guzman
- German Center for Infection Research, Münich, Heidelberg, Germany; Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Percy Knolle
- German Center for Infection Research, Münich, Heidelberg, Germany; Institute of Molecular Immunology, University Hospital rechts der Isar, Technical University of Munich, Münich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Münich, Helmholtz Zentrum München, Münich, Germany; German Center for Infection Research, Münich, Heidelberg, Germany.
| |
Collapse
|
4
|
Bloom K, Maepa MB, Ely A, Arbuthnot P. Gene Therapy for Chronic HBV-Can We Eliminate cccDNA? Genes (Basel) 2018; 9:E207. [PMID: 29649127 PMCID: PMC5924549 DOI: 10.3390/genes9040207] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic infection with the hepatitis B virus (HBV) is a global health concern and accounts for approximately 1 million deaths annually. Amongst other limitations of current anti-HBV treatment, failure to eliminate the viral covalently closed circular DNA (cccDNA) and emergence of resistance remain the most worrisome. Viral rebound from latent episomal cccDNA reservoirs occurs following cessation of therapy, patient non-compliance, or the development of escape mutants. Simultaneous viral co-infections, such as by HIV-1, further complicate therapeutic interventions. These challenges have prompted development of novel targeted hepatitis B therapies. Given the ease with which highly specific and potent nucleic acid therapeutics can be rationally designed, gene therapy has generated interest for antiviral application. Gene therapy strategies developed for HBV include gene silencing by harnessing RNA interference, transcriptional inhibition through epigenetic modification of target DNA, genome editing by designer nucleases, and immune modulation with cytokines. DNA-binding domains and effectors based on the zinc finger (ZF), transcription activator-like effector (TALE), and clustered regularly interspaced short palindromic repeat (CRISPR) systems are remarkably well suited to targeting episomal cccDNA. This review discusses recent developments and challenges facing the field of anti-HBV gene therapy, its potential curative significance and the progress towards clinical application.
Collapse
Affiliation(s)
- Kristie Bloom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, Johannesburg, WITS 2050, South Africa.
| | - Mohube Betty Maepa
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, Johannesburg, WITS 2050, South Africa.
| | - Abdullah Ely
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, Johannesburg, WITS 2050, South Africa.
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, Johannesburg, WITS 2050, South Africa.
| |
Collapse
|
5
|
Kruse RL, Shum T, Legras X, Barzi M, Pankowicz FP, Gottschalk S, Bissig KD. In Situ Liver Expression of HBsAg/CD3-Bispecific Antibodies for HBV Immunotherapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:32-41. [PMID: 29018834 PMCID: PMC5626922 DOI: 10.1016/j.omtm.2017.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023]
Abstract
Current therapies against hepatitis B virus (HBV) do not reliably cure chronic infection, necessitating new therapeutic approaches. The T cell response can clear HBV during acute infection, and the adoptive transfer of antiviral T cells during bone marrow transplantation can cure patients of chronic HBV infection. To redirect T cells to HBV-infected hepatocytes, we delivered plasmids encoding bispecific antibodies directed against the viral surface antigen (HBsAg) and CD3, expressed on almost all T cells, directly into the liver using hydrodynamic tail vein injection. We found a significant reduction in HBV-driven reporter gene expression (184-fold) in a mouse model of acute infection, which was 30-fold lower than an antibody only recognizing HBsAg. While bispecific antibodies triggered, in part, antigen-independent T cell activation, antibody production within hepatocytes was non-cytotoxic. We next tested the bispecific antibodies in a different HBV mouse model, which closely mimics the transcriptional template for HBV, covalently closed circular DNA (cccDNA). We found that the antiviral effect was noncytopathic, mediating a 495-fold reduction in HBsAg levels at day 4. At day 33, bispecific antibody-treated mice exhibited 35-fold higher host HBsAg immunoglobulin G (IgG) antibody production versus untreated groups. Thus, gene therapy with HBsAg/CD3-bispecific antibodies represents a promising therapeutic strategy for patients with HBV.
Collapse
Affiliation(s)
- Robert L Kruse
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA.,Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas Shum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xavier Legras
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mercedes Barzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank P Pankowicz
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Karl-Dimiter Bissig
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA.,Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Abstract
Hepatitis B virus (HBV) infection is a worldwide health problem, with approximately one third of populations have been infected, among which 3-5% of adults and more than 90% of children developed to chronic HBV infection. Host immune factors play essential roles in the outcome of HBV infection. Thus, ineffective immune response against HBV may result in persistent virus replications and liver necroinflammations, then lead to chronic HBV infection, liver cirrhosis, and even hepatocellular carcinoma. Cytokine balance was shown to be an important immune characteristic in the development and progression of hepatitis B, as well as in an effective antiviral immunity. Large numbers of cytokines are not only involved in the initiation and regulation of immune responses but also contributing directly or indirectly to the inhibition of virus replication. Besides, cytokines initiate downstream signaling pathway activities by binding to specific receptors expressed on the target cells and play important roles in the responses against viral infections and, therefore, might affect susceptibility to HBV and/or the natural course of the infection. Since cytokines are the primary causes of inflammation and mediates liver injury after HBV infection, we have discussed recent advances on the roles of various cytokines [including T helper type 1 cells (Th1), Th2, Th17, regulatory T cells (Treg)-related cytokines] in different phases of HBV infection and cytokine-related mechanisms for impaired viral control and liver damage during HBV infection. We then focus on experimental therapeutic applications of cytokines to gain a better understanding of this newly emerging aspect of disease pathogenesis.
Collapse
|
7
|
Hösel M, Lucifora J, Michler T, Holz G, Gruffaz M, Stahnke S, Zoulim F, Durantel D, Heikenwalder M, Nierhoff D, Millet R, Salvetti A, Protzer U, Büning H. Hepatitis B virus infection enhances susceptibility toward adeno-associated viral vector transduction in vitro and in vivo. Hepatology 2014; 59:2110-20. [PMID: 24425003 DOI: 10.1002/hep.26990] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/23/2013] [Indexed: 12/20/2022]
Abstract
UNLABELLED Gene therapy has become an accepted concept for the treatment of a variety of different diseases. In contrast to preclinical models, subjects enrolled in clinical trials, including gene therapy, possess a history of infection with microbes that may influence its safety and efficacy. Especially, viruses that establish chronic infections in the liver, one of the main targets for in vivo gene therapy, raise important concerns. Among them is the hepatitis B virus (HBV), which has chronically infected more than 350 million people worldwide. Here, we investigated the effect of HBV on adeno-associated viral (AAV) vectors, the most frequently applied gene transfer vehicles for in vivo gene therapy. Unexpectedly, we found that HBV greatly improved AAV transduction in cells replicating HBV and identified HBV protein x (HBx) as a key factor. Whereas HBV-positive and -negative cells were indistinguishable with respect to cell-entry efficiency, significantly higher numbers of AAV vector genomes were successfully delivered to the nucleus in the presence of HBV. The HBV-promoting effect was abolished by inhibitors of phosphatidylinositol 3-kinase (PI3K). PI3K was required for efficient trafficking of AAV to the nucleus and was enhanced in HBV-replicating cells and upon HBx expression. Enhancement of AAV transduction was confirmed in vivo using HBV transgenic mice and could successfully be applied to inhibit HBV progeny release. CONCLUSION Our results demonstrate that acute, as well as chronic, infections with unrelated viruses change the intracellular milieu, thereby likely influencing gene therapy outcomes. In the case of HBV, HBx-mediated enhancement of AAV transduction is an advantage that could be exploited for development of novel treatments of HBV infection.
Collapse
Affiliation(s)
- Marianna Hösel
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; German Center for Infection Research, Partner sites Bonn-Cologne and Munich, Germany; Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Ando M, Takahashi Y, Nishikawa M, Takakura Y. [Control of spatiotemporal distribution of interferon γ by genetically fusing functional peptides]. YAKUGAKU ZASSHI 2014. [PMID: 23208047 DOI: 10.1248/yakushi.12-00235-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Type II interferon (IFNγ) is a representative Th1 cytokine and it possesses a variety of functions, including immune regulation, antiviral and antitumor activity. Because of its multifunctional nature, IFNγ is expected to be applied to the treatment of autoimmune diseases, cancer and viral infection. Although IFNγ has therapeutic potential for such diseases, the clinical use of IFNγ has been limited due to its short in vivo half-life and serious adverse effects. In contrast, gene delivery of IFNγ is an alternative approach to increasing the retention time of IFNγ. To extend transgene expression after plasmid DNA (pDNA) gene transfer, we designed and developed pDNA with varying numbers of CpG motifs. CpG-reduced pDNA resulted in more durable transgene expression than its CpG replete counterpart in mice. Comparison of the effect of promoter/enhancer elements on transgene expression showed that ROSA26 promoter-mediated IFNγ expression was safe because of the lack of an initial surge after hydrodynamic gene transfer. We also designed an IFNγ-mouse serum albumin (MSA) fusion protein, IFNγ-MSA. Gene transfer of this fusion protein resulted in a sustained concentration of IFNγ fusion protein in mouse serum, and inhibited tumor metastasis in mice. These results provide experimental evidence that IFNγ gene therapy can be a useful treatment for a variety of diseases.
Collapse
Affiliation(s)
- Mitsuru Ando
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
9
|
Krebs K, Böttinger N, Huang LR, Chmielewski M, Arzberger S, Gasteiger G, Jäger C, Schmitt E, Bohne F, Aichler M, Uckert W, Abken H, Heikenwalder M, Knolle P, Protzer U. T cells expressing a chimeric antigen receptor that binds hepatitis B virus envelope proteins control virus replication in mice. Gastroenterology 2013; 145:456-65. [PMID: 23639914 DOI: 10.1053/j.gastro.2013.04.047] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 03/20/2013] [Accepted: 04/17/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Antiviral agents suppress hepatitis B virus (HBV) replication but do not clear the infection. A strong effector T-cell response is required to eradicate HBV, but this does not occur in patients with chronic infection. T cells might be directed toward virus-infected cells by expressing HBV-specific receptors and thereby clear HBV and help to prevent development of liver cancer. In mice, we studied whether redirected T cells can engraft after adoptive transfer, without prior T-cell depletion, and whether the large amounts of circulating viral antigens inactivate the transferred T cells or lead to uncontrolled immune-mediated damage. METHODS CD8(+) T cells were isolated from mice and stimulated using an optimized protocol. Chimeric antigen receptors (CARs) that bind HBV envelope proteins (S-CAR) and activate T cells were expressed on the surface of cells using retroviral vectors. S-CAR-expressing CD8(+) T cells, which carried the marker CD45.1, were injected into CD45.2(+) HBV transgenic mice. We compared these mice with mice that received CD8(+) T cells induced by vaccination, cells that express a CAR without a proper signaling domain, or cells that express a CAR that does not bind HBV proteins (controls). RESULTS CD8(+) T cells that expressed HBV-specific CARs recognized different HBV subtypes and were able to engraft and expand in immune-competent HBV transgenic mice. After adoptive transfer, the S-CAR-expressing T cells localized to and functioned in the liver and rapidly and efficiently controlled HBV replication compared with controls, causing only transient liver damage. The large amount of circulating viral antigen did not impair or overactivate the S-CAR-grafted T cells. CONCLUSIONS T cells with a CAR specific for HBV envelope proteins localize to the liver in mice to reduce HBV replication, causing only transient liver damage. This immune cell therapy might be developed for patients with chronic hepatitis B, regardless of their HLA type.
Collapse
Affiliation(s)
- Karin Krebs
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, München, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Ando M, Takahashi Y, Nishikawa M, Watanabe Y, Takakura Y. Constant and steady transgene expression of interferon-γ by optimization of plasmid construct for safe and effective interferon-γ gene therapy. J Gene Med 2013; 14:288-95. [PMID: 22359360 DOI: 10.1002/jgm.2616] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hydrodynamic injection of pmCMV(enh) -hEF-1(prom) -muIFN-γ, a plasmid DNA (pDNA) expressing murine interferon (IFN)-γ with a murine cytomegalovirus (mCMV) enhancer and a human elongation factor (EF)-1 promoter, has been proven effective for the treatment of cancer and atopic dermatitis in mice. However, the initial peak of IFN-γ soon after injection was quite high compared to the steady level for subsequent periods, which could cause unwanted adverse effects. Therefore, in the present study, aiming to optimize the efficacy/side-effect ratio of IFN-γ gene transfer, we have developed plasmid vectors encoding murine IFN-γ under the control of different combinations of promoter and enhancer sequences. METHODS The promoter and enhancer sequence of pmCMV(enh) -hEF-1(prom) -huIFN-γ, a prototype plasmid expressing human IFN-γ, was replaced or deleted to obtain various pDNAs. To assess the transgene expression profile, each pDNA was delivered to mice by hydrodynamic injection and the serum IFN-γ concentration was measured periodically. On the basis of the results obtained, murine IFN-γ expressing pDNAs were constructed and the body weight change was monitored as an indicator of adverse effects. RESULTS The prototype pmCMV(enh) -hEF-1(prom) -huIFN-γ showed a high but declining concentration of IFN-γ. Those containing hROSA26 promoter expressed the transgene in a more constant manner with no initial high concentrations and scarcely reduced the body weight. CONCLUSIONS These results indicate that hROSA26 promoter, irrespective of the presence and type of enhancers, is suitable for achieving constant and steady level of transgene expression and effective in avoiding the body weight loss caused by high concentrations of IFN-γ.
Collapse
Affiliation(s)
- Mitsuru Ando
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Science, Kyoto University, Kyoto, Japan
| | | | | | | | | |
Collapse
|
11
|
Huang LR, Gäbel YA, Graf S, Arzberger S, Kurts C, Heikenwalder M, Knolle PA, Protzer U. Transfer of HBV genomes using low doses of adenovirus vectors leads to persistent infection in immune competent mice. Gastroenterology 2012; 142:1447-50.e3. [PMID: 22426294 DOI: 10.1053/j.gastro.2012.03.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 02/29/2012] [Accepted: 03/03/2012] [Indexed: 12/23/2022]
Abstract
Studies of mechanisms responsible for the persistence of hepatitis B virus (HBV) infection have been hindered by a lack of appropriate animal models. HBV genomes can be delivered to livers of mice using hydrodynamic injection or high doses of an adenoviral vector; these lead to clearance of HBV. We found that infection of immunocompetent mice with low doses of an adenoviral vector resulted in persistent HBV infection; the mice neither underwent seroconversion to production of antibodies against HBV nor developed a strong HBV-specific effector T-cell response. As in patients with chronic HBV infection, DNA vaccination failed to generate T cells that cleared infection. This model of persistent HBV infection could be used to study the pathogenesis of chronic HBV infection and develop new therapeutic strategies.
Collapse
Affiliation(s)
- Li-Rung Huang
- Institutes of Molecular Medicine and Experimental Immunology, Universität Bonn, Bonn, Germany
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Specific expression of human interferon-gamma controls hepatitis B virus replication in vitro in secreting hepatitis B surface antigen hepatocytes. J Virol Methods 2012; 180:84-90. [DOI: 10.1016/j.jviromet.2011.12.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 11/04/2011] [Accepted: 12/30/2011] [Indexed: 12/31/2022]
|
13
|
Gene therapy in interventional pulmonology: Interferon gene delivery with focus on thoracic malignancies. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s13665-011-0008-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Ebert G, Poeck H, Lucifora J, Baschuk N, Esser K, Esposito I, Hartmann G, Protzer U. 5' Triphosphorylated small interfering RNAs control replication of hepatitis B virus and induce an interferon response in human liver cells and mice. Gastroenterology 2011; 141:696-706, 706.e1-3. [PMID: 21684282 DOI: 10.1053/j.gastro.2011.05.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 04/26/2011] [Accepted: 05/02/2011] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Approved therapies for chronic hepatitis B include systemic administration of interferon (IFN)-alfa and inhibitors of hepatitis B virus (HBV) reverse-transcription. Systemic application of IFN-alfa is limited by side effects. Reverse-transcriptase inhibitors effectively control HBV replication, but rarely eliminate the virus and can select drug-resistant variants. We aimed to develop an alternative therapeutic approach that combines gene silencing with induction of IFN in the liver. METHODS To stimulate an immune response while inhibiting HBV activity, we designed 3 small interfering (si)RNAs that target highly conserved sequences and multiple HBV transcripts of all genotypes. A 5'-triphosphate (3p) was added to the siRNAs, turning them into a ligand for the cytosolic helicase retinoic acid-inducible protein I, which becomes activated and induces expression of type-I IFNs. Antiviral activity was investigated in cell lines that replicate HBV, in HBV-infected primary human hepatocytes, and in HBV transgenic mice. RESULTS 3p-double-stranded RNA (3p-RNA) activated retinoic acid-inducible protein I, induced a strong type I IFN response (expression of IFN-β) in liver cells and showed transient but strong antiviral activity. Bifunctional, HBV-specific, 3p-siRNAs controlled replication of HBV more efficiently and for longer periods of time than 3p-RNAs without silencing capacity or siRNAs that targeted identical sequences but did not contain 3p. CONCLUSIONS HBV-specific 3p-siRNAs are bifunctional antiviral molecules that induce production of type I IFNs in the liver and target HBV RNAs to inhibit viral replication.
Collapse
Affiliation(s)
- Gregor Ebert
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
15
|
von Freyend MJ, Untergasser A, Arzberger S, Oberwinkler H, Drebber U, Schirmacher P, Protzer U. Sequential control of hepatitis B virus in a mouse model of acute, self-resolving hepatitis B. J Viral Hepat 2011; 18:216-26. [PMID: 20367794 DOI: 10.1111/j.1365-2893.2010.01302.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The determinants of an immune response to the human hepatitis B virus (HBV) are poorly understood. As studies in man and chimpanzees are limited, we aimed at developing a model of self-limiting hepatitis B in mice that helps to dissect the control of HBV by humoral and cellular immune responses. Adenoviral vectors containing 1.3-fold HBV genomes allowed an efficient and reproducible transfer of HBV genomes into mouse livers and initiated HBV replication in mice. HBV transcripts were detected in mouse livers for more than 3 months. HBsAg and HBeAg peaked around day 6 and slowly declined thereafter. A two-phase mild to moderate liver inflammation with elevated serum alanine transaminase activities was observed around day 7 and around day 70 when the vast majority of HBV-specific T cells were detected in the liver. HBV was initially controlled when specific and nonspecific T cells infiltrated the liver and intrahepatic interferon γ levels peaked around day 7, but replicated again from day 10 to day 24 and persisted at low levels thereafter despite the presence of HBV-specific T cells. Finally, HBV replication was terminated after a sufficient B-cell response had been mounted- indicated by anti-HBs seroconversion around day 35. HBV-specific T cells infiltrated the liver a second time around day 70 postinfection. This demonstrates that the established mouse model allows studying the onset and termination of HBV infection and will help to dissect the determinants of HBV control and clearance by the immune response.
Collapse
Affiliation(s)
- M John von Freyend
- Institute for Medical Microbiology, Immunology and Hygiene and Center for Molecular Medicine Cologne, University of Cologne Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
16
|
Brunelle MN, Saboulard D, Massinet H, Lamant C, Soussan P, Brezillon N, Kremsdorf D. Inhibition of hepatitis B virus DNA replication by a thermostable interferon-γ variant. Antivir Ther 2010; 15:861-9. [PMID: 20834098 DOI: 10.3851/imp1639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Treatment of HBV chronic carriers using interferon (IFN)-α or nucleoside/nucleotide analogues fails to suppress viral infection. Type-II IFN-γ has been shown to inhibit HBV replication. The goal of the present work was to evaluate the antiviral efficacy against HBV of a thermostable IFN-γ variant isolated using Massive Mutagenesis and thermoresistant selection (THR) technologies. METHODS The thermostability of wild-type (wt) and S63C IFN-γ was determined in vitro and in vivo. Activation of the IFN-γ responsive element by wt and S63C IFN-γ was tested using a luciferase assay. HepG2.2.15 cells constitutively expressing HBV were used to analyse the antiviral activity of wt and S63C IFN-γ against HBV replication. Intracellular HBV DNA was detected by Southern blot and quantified by real-time PCR analyses. RESULTS S63C IFN-γ was shown to be more thermostable and had a longer half-life than wt IFN-γ. Both wt and S63C IFN-γ displayed a similar capacity to activate the IFN pathway. The treatment of HepG2.2.15 cells with wt or S63C IFN-γ induced the inhibition of HBV viral replication. After heating, S63C IFN-γ displayed better conservation of its antiviral activity against HBV when compared with wt IFN-γ. CONCLUSIONS These results confirm that the THR method can be used to isolate mutants with enhanced thermostability and demonstrate that a thermostable IFN-γ variant presents antiviral properties against HBV replication. This molecule could provide a new strategy to treat patients who do not respond to antiviral therapy.
Collapse
|
17
|
Protzer U, Abken H. Can engineered "designer" T cells outsmart chronic hepatitis B? HEPATITIS RESEARCH AND TREATMENT 2010; 2010:901216. [PMID: 21188203 PMCID: PMC3004001 DOI: 10.1155/2010/901216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 07/14/2010] [Accepted: 07/15/2010] [Indexed: 01/02/2023]
Abstract
More than 350 million people worldwide are persistently infected with human heptatitis B virus (HBV) and at risk to develop liver cirrhosis and hepatocellular carcinoma making long-term treatment necessary. While a vaccine is available and new antiviral drugs are being developed, elimination of persistently infected cells is still a major issue. Recent efforts in adoptive cell therapy are experimentally exploring immunotherapeutic elimination of HBV-infected cells by means of a biological attack with genetically engineered "designer" T cells.
Collapse
Affiliation(s)
- U. Protzer
- Institut für Virologie, Technische Universität München/Helmholtz Zentrum München, 81675 München, Germany
| | - H. Abken
- Zentrum für Molekulare Medizin Köln und Klinik I für Innere Medizin Köln, Universität zu Köln, Robert-Koch-Str. 21, 50931 Köln, Germany
| |
Collapse
|
18
|
Hösel M, Quasdorff M, Wiegmann K, Webb D, Zedler U, Broxtermann M, Tedjokusumo R, Esser K, Arzberger S, Kirschning CJ, Langenkamp A, Falk C, Büning H, Rose-John S, Protzer U. Not interferon, but interleukin-6 controls early gene expression in hepatitis B virus infection. Hepatology 2009; 50:1773-82. [PMID: 19937696 DOI: 10.1002/hep.23226] [Citation(s) in RCA: 286] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED With about 350 million virus carriers, hepatitis B virus (HBV) infection remains a major health problem. HBV is a noncytopathic virus causing persistent infection, but it is still unknown whether host recognition of HBV may activate an innate immune response. We describe that upon infection of primary human liver cells, HBV is recognized by nonparenchymal cells of the liver, mainly by liver macrophages (Kupffer cells), although they are not infected. Within 3 hours, this recognition leads to the activation of nuclear factor kappa B (NF-kappaB) and subsequently to the release of interleukin-6 (IL-6) and other proinflammatory cytokines (IL-8, TNF-alpha, IL-1beta), but does not induce an interferon response. The activation of proinflammatory cytokines, however, is transient, and even inhibits responsiveness toward a subsequent challenge. IL-6 released by Kupffer cells after activation of NF-kappaB controls HBV gene expression and replication in hepatocytes at the level of transcription shortly after infection. Upon binding to its receptor complex, IL-6 activates the mitogen-activated protein kinases exogenous signal-regulated kinase 1/2, and c-jun N-terminal kinase, which inhibit expression of hepatocyte nuclear factor (HNF) 1alpha and HNF 4alpha, two transcription factors essential for HBV gene expression and replication. CONCLUSION Our results demonstrate recognition of HBV patterns by nonparenchymal liver cells, which results in IL-6-mediated control of HBV infection at the transcriptional level. Thus, IL-6 ensures early control of the virus, limiting activation of the adaptive immune response and preventing death of the HBV-infected hepatocyte. This pattern recognition may be essential for a virus, which infects a new host with only a few virions. Our data also indicate that therapeutic neutralization of IL-6 for treatment of certain diseases may represent a risk if the patient is HBV-infected.
Collapse
Affiliation(s)
- Marianna Hösel
- Center for Molecular Medicine Cologne (ZMMK), University Hospital Cologne, Köln, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Stieger K, Belbellaa B, Le Guiner C, Moullier P, Rolling F. In vivo gene regulation using tetracycline-regulatable systems. Adv Drug Deliv Rev 2009; 61:527-41. [PMID: 19394373 PMCID: PMC7103297 DOI: 10.1016/j.addr.2008.12.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 12/15/2008] [Indexed: 10/26/2022]
Abstract
Numerous preclinical studies have demonstrated the efficacy of viral gene delivery vectors, and recent clinical trials have shown promising results. However, the tight control of transgene expression is likely to be required for therapeutic applications and in some instances, for safety reasons. For this purpose, several ligand-dependent transcription regulatory systems have been developed. Among these, the tetracycline-regulatable system is by far the most frequently used and the most advanced towards gene therapy trials. This review will focus on this system and will describe the most recent progress in the regulation of transgene expression in various organs, including the muscle, the retina and the brain. Since the development of an immune response to the transactivator was observed following gene transfer in the muscle of nonhuman primate, focus will be therefore, given on the immune response to transgene products of the tetracycline inducible promoter.
Collapse
Affiliation(s)
- Knut Stieger
- INSERM UMR U649, CHU-Hotel Dieu, Nantes, France
- Department of Ophthalmology, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | |
Collapse
|
20
|
Riedl P, Wieland A, Lamberth K, Buus S, Lemonnier F, Reifenberg K, Reimann J, Schirmbeck R. Elimination of Immunodominant Epitopes from Multispecific DNA-Based Vaccines Allows Induction of CD8 T Cells That Have a Striking Antiviral Potential. THE JOURNAL OF IMMUNOLOGY 2009; 183:370-80. [DOI: 10.4049/jimmunol.0900505] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
21
|
Abstract
This chapter describes the major gene therapeutic approaches for viral infections. The vast majority of published approaches target severe chronic viral infections such as hepatitis B or C and HIV infection. Two basic gene therapy strategies are introduced here. The first involves the expression of a protein or an RNA that inhibits viral replication by targeting crucial steps of the viral life cycle or by interfering with a cellular factor required for virus replication. The major limitation of this approach is that primary levels of gene modification have generally not been sufficient to reduce the availability of target cells permissive for virus replication to a level that significantly decreases overall viral load. Thus, investigators have banked on the expectation that gene-protected cells have a sufficient selective advantage to accumulate and gain prevalence over time, a prediction that so far could not be confirmed in clinical trials. In vivo levels of gene modification can be improved, however, by introducing an additional selectable marker. In addition, a secreted antiviral gene product that exerts a bystander effect could significantly reduce overall virus replication despite relatively low levels of gene modification. In addition to these direct antiviral approaches, several strategies have been developed that employ or aim to enhance host immune responses. The innate immune response has been enhanced, for example, by the in vivo expression of interferons. Alternatively, T cells can be grafted with recombinant receptors to boost adaptive virus-specific immunity. These approaches are especially promising for chronic virus infection, where natural immune responses are evidently not sufficient to effectively control virus replication.
Collapse
|
22
|
Turelli P, Liagre-Quazzola A, Mangeat B, Verp S, Jost S, Trono D. APOBEC3-independent interferon-induced viral clearance in hepatitis B virus transgenic mice. J Virol 2008; 82:6585-90. [PMID: 18434399 PMCID: PMC2447049 DOI: 10.1128/jvi.00216-08] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 04/09/2008] [Indexed: 12/15/2022] Open
Abstract
Interferon (IFN) has been part of the standard treatment of chronic hepatitis B infection for more than 2 decades, yet the mechanism of action of this antiviral remains poorly understood. It was recently observed that members of the human APOBEC family of cytidine deaminases endowed with anti-hepatitis B virus (HBV) activity are upregulated by type I and II IFNs. However, we demonstrated that, in tissue culture, these cellular enzymes are not essential effectors of the anti-HBV action of these cytokines. Here, we show that murine APOBEC3 (muA3) can also block HBV replication. While expressed at low levels in the mouse liver at baseline, muA3 is upregulated upon IFN induction. However, in HBV-transgenic muA3 knockout mice, IFN induction blocked HBV DNA production as efficiently as in control HBV-transgenic muA3-competent animals. We conclude that APOBEC3 is not an essential mediator of the IFN-mediated inhibition of HBV in vivo.
Collapse
Affiliation(s)
- Priscilla Turelli
- Global Health Institute, School of Life Sciences and Frontiers in Genetics National Center for Competence in Research, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
23
|
Bohne F, Chmielewski M, Ebert G, Wiegmann K, Kürschner T, Schulze A, Urban S, Krönke M, Abken H, Protzer U. T cells redirected against hepatitis B virus surface proteins eliminate infected hepatocytes. Gastroenterology 2008; 134:239-47. [PMID: 18166356 DOI: 10.1053/j.gastro.2007.11.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 10/11/2007] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The final goal in hepatitis B therapy is eradication of the hepatitis B virus (HBV) replication template, the so-called covalently closed circular DNA (cccDNA). Current antiviral treatment of chronic hepatitis B depends on interferon alpha or nucleoside analogues inhibiting the viral reverse transcriptase. Despite treatment, cccDNA mostly persists in the host cell nucleus, continues to produce hepatitis B surface antigen (HBsAg), and causes relapsing disease. We therefore aimed at eliminating persistently infected hepatocytes carrying HBV cccDNA by redirecting cytolytic T cells toward HBsAg-producing cells. METHODS We designed chimeric T-cell receptors directed against HBV surface proteins present on HBV-infected cells and used them to graft primary human T cells with antibody-like specificity. The receptors were composed of a single chain antibody fragment directed against HBV S or L protein fused to intracellular signalling domains of CD3xi and the costimulatory CD28 molecule. RESULTS Our results show that these chimeric receptors, when retrovirally delivered and expressed on the cell surface, enable primary human T cells to recognize HBsAg-positive hepatocytes, release interferon gamma and interleukin 2, and, most importantly, lyse HBV replicating cells. When coincubated with HBV-infected primary human hepatocytes, these engineered, antigen-specific T cells selectively eliminated HBV-infected and thus cccDNA-positive target cells. CONCLUSIONS Elimination of HBV cccDNA-positive hepatocytes following antiviral therapy is a major therapeutic goal in chronic hepatitis B, and adoptive transfer of grafted T cells provides a promising novel therapeutic approach. However, T-cell therapy may also cause liver damage and therefore needs further preclinical evaluation.
Collapse
Affiliation(s)
- Felix Bohne
- Molecular Infectiology, University Hospital Cologne, Koeln, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Protzer U, Seyfried S, Quasdorff M, Sass G, Svorcova M, Webb D, Bohne F, Hösel M, Schirmacher P, Tiegs G. Antiviral activity and hepatoprotection by heme oxygenase-1 in hepatitis B virus infection. Gastroenterology 2007; 133:1156-65. [PMID: 17919491 DOI: 10.1053/j.gastro.2007.07.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 07/12/2007] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Induction of heme oxygenase-1 (HO-1) has been shown to be beneficial in immune-mediated liver damage. We now investigate the effects of HO-1 induction in models of human hepatitis B virus (HBV) infection. METHODS Adenoviral transfer of an HBV 1.3 genome into wild-type mice was used as a model for acute hepatitis B. HBV transgenic animals were used as a model for chronic HBV infection. HBV replication was assessed by HBV viremia, antigenemia, and Southern blotting, liver damage was assessed by serum alanine aminotransferase activities and histopathology of liver sections. To investigate HO-1 effects on HBV replication at a molecular level, stably HBV-transfected hepatoma cells were used. HBV gene expression, protein stability, transcription, and replication were determined. HO-1 was induced by either cobalt-protoporphyrin-IX or over expressed by adenoviral gene transfer. RESULTS In the acute hepatitis B model, liver injury was reduced significantly after HO-1 induction. In addition, HO-1 showed a pronounced antiviral effect, which was confirmed in stably HBV-transfected hepatoma cells and in persistently HBV replicating transgenic mice. We showed that HO-1 induction repressed HBV replication directly in hepatocytes at a posttranscriptional step by reducing stability of HBV core protein and thus blocking refill of nuclear HBV covalently closed circular (ccc)DNA. Small interfering RNA directed against HO-1 proved that this effect depended on the expression level of HO-1. CONCLUSIONS Besides its hepatoprotective effect, HO-1 showed a pronounced antiviral activity in HBV infection. Therefore, induction of HO-1 might be a novel therapeutic option for inflammatory flares of hepatitis B.
Collapse
Affiliation(s)
- Ulrike Protzer
- Molecular Infectiology at Center for Molecular Medicine Cologne, Institute for Medical Microbiology, Immunology, and Hygiene, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jost S, Turelli P, Mangeat B, Protzer U, Trono D. Induction of antiviral cytidine deaminases does not explain the inhibition of hepatitis B virus replication by interferons. J Virol 2007; 81:10588-96. [PMID: 17652382 PMCID: PMC2045472 DOI: 10.1128/jvi.02489-06] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interferons (IFNs) play a major role in the control of hepatitis B virus (HBV), whether as endogenous cytokines limiting the spread of the virus during the acute phase of the infection or as drugs for the treatment of its chronic phase. However, the mechanism by which IFNs inhibit HBV replication has so far remained elusive. Here, we show that type I and II IFN treatment of human hepatocytes induces the production of APOBEC3G (A3G) and, to a lesser extent, that of APOBEC3F (A3F) and APOBEC3B (A3B) but not that of two other cytidine deaminases also endowed with anti-HBV activity, activation-induced cytidine deaminase (AID), and APOBEC1. Most importantly, we reveal that blocking A3B, A3F, and A3G by combining RNA interference and the virion infectivity factor (Vif) protein of human immunodeficiency virus does not abrogate the inhibitory effect of IFNs on HBV. We conclude that these cytidine deaminases are not essential effectors of IFN in its action against this pathogen.
Collapse
Affiliation(s)
- Stéphanie Jost
- School of Life Sciences and Frontiers in Genetics National Center for Competence in Research, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
26
|
Winkeler A, Sena-Esteves M, Paulis LE, Li H, Waerzeggers Y, Rückriem B, Himmelreich U, Klein M, Monfared P, Rueger MA, Heneka M, Vollmar S, Hoehn M, Fraefel C, Graf R, Wienhard K, Heiss WD, Jacobs AH. Switching on the lights for gene therapy. PLoS One 2007; 2:e528. [PMID: 17565381 PMCID: PMC1885827 DOI: 10.1371/journal.pone.0000528] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 04/30/2007] [Indexed: 11/19/2022] Open
Abstract
Strategies for non-invasive and quantitative imaging of gene expression in vivo have been developed over the past decade. Non-invasive assessment of the dynamics of gene regulation is of interest for the detection of endogenous disease-specific biological alterations (e.g., signal transduction) and for monitoring the induction and regulation of therapeutic genes (e.g., gene therapy). To demonstrate that non-invasive imaging of regulated expression of any type of gene after in vivo transduction by versatile vectors is feasible, we generated regulatable herpes simplex virus type 1 (HSV-1) amplicon vectors carrying hormone (mifepristone) or antibiotic (tetracycline) regulated promoters driving the proportional co-expression of two marker genes. Regulated gene expression was monitored by fluorescence microscopy in culture and by positron emission tomography (PET) or bioluminescence (BLI) in vivo. The induction levels evaluated in glioma models varied depending on the dose of inductor. With fluorescence microscopy and BLI being the tools for assessing gene expression in culture and animal models, and with PET being the technology for possible application in humans, the generated vectors may serve to non-invasively monitor the dynamics of any gene of interest which is proportionally co-expressed with the respective imaging marker gene in research applications aiming towards translation into clinical application.
Collapse
Affiliation(s)
- Alexandra Winkeler
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Miguel Sena-Esteves
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Leonie E.M. Paulis
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Hongfeng Li
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Yannic Waerzeggers
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Benedikt Rückriem
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Uwe Himmelreich
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Markus Klein
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Parisa Monfared
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Maria A. Rueger
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Michael Heneka
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Stefan Vollmar
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Mathias Hoehn
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Cornel Fraefel
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Rudolf Graf
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Klaus Wienhard
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Wolf D. Heiss
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
| | - Andreas H. Jacobs
- Laboratory for Gene Therapy and Molecular Imaging at the Max Planck-Institute for Neurological Research, Center for Molecular Medicine (CMMC) and Departments of Neurology and Radiology at the University of Cologne, Cologne, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
27
|
Lu M, Menne S, Yang D, Xu Y, Roggendorf M. Immunomodulation as an option for the treatment of chronic hepatitis B virus infection: preclinical studies in the woodchuck model. Expert Opin Investig Drugs 2007; 16:787-801. [PMID: 17501692 DOI: 10.1517/13543784.16.6.787] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
New therapeutic approaches for chronic hepatitis B virus infection based on immunomodulation are now under investigation. The woodchuck model for hepatitis B virus infection has emerged as a useful animal model for the evaluation of such approaches, after developing necessary assays and reagents for immunologic studies in this model. Conventional and novel vaccines such as DNA vaccines were tested in woodchucks for their ability to induce protective immune responses against challenge infection with the woodchuck hepatitis virus (WHV). Furthermore, immunotherapeutic approaches for the control of chronic hepadnaviral infection were evaluated in woodchucks. Immunizations with WHV proteins and DNA vaccines led to the development of antibodies to the WHV surface antigen and to a significant decrease of viral load in chronically WHV-infected woodchucks. Viral vector-mediated gene transfer was explored for the delivery of antiviral cytokines IFN-alpha in woodchucks and resulted in the decrease of viral replication. It is now generally accepted that a combination of antiviral treatment and immunization will be necessary to achieve successful immunomodulation with a long-term control of chronic hepatitis B virus infection.
Collapse
Affiliation(s)
- Mengji Lu
- Institut für Virologie, Universitätsklinikum Essen, Essen, Germany.
| | | | | | | | | |
Collapse
|
28
|
Gonzalez-Aseguinolaza G, Crettaz J, Ochoa L, Otano I, Aldabe R, Paneda A. Gene therapy for viral hepatitis. Expert Opin Biol Ther 2006; 6:1263-78. [PMID: 17223736 DOI: 10.1517/14712598.6.12.1263] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatitis B and C infections are two of the most prevalent viral diseases in the world. Existing therapies against chronic viral hepatitis are far from satisfactory due to low response rates, undesirable side effects and selection of resistant viral strains. Therefore, new therapeutic approaches are urgently needed. This review, after briefly summarising the in vitro and in vivo systems for the study of both diseases and the genetic vehicles commonly used for liver gene transfer, examines the existing status of gene therapy-based antiviral strategies that have been employed to prevent, eliminate or reduce viral infection. In particular, the authors focus on the results obtained in clinical trials and experimental clinically relevant animal models.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Genetic Therapy/methods
- Genetic Therapy/trends
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/prevention & control
- Hepatitis C, Chronic/genetics
- Hepatitis C, Chronic/prevention & control
- Hepatitis, Viral, Animal/genetics
- Hepatitis, Viral, Animal/prevention & control
- Hepatitis, Viral, Human/genetics
- Hepatitis, Viral, Human/prevention & control
- Humans
Collapse
Affiliation(s)
- Gloria Gonzalez-Aseguinolaza
- University of Navarra, Division of Gene Therapy and Hepatology, Center for Applied Medical Research (CIMA), 31008 Pamplona, Spain.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Hepatocellular carcinoma (HCC) is a frequent malignancy worldwide with a high rate of metastasis. The hepatitis B and C viruses are considered major etiological factors associated with the development of HCC, particularly as a result of their induction of chronic inflammation. There is increasing evidence that the inflammatory process is inherently associated with many different cancer types, including HCC. Specifically, this review aims to cover evidence for the potential roles of cytokines, an important component of the immune system, in promoting HCC carcinogenesis and progression. A global summary of cytokine levels, functions, polymorphisms, and therapies with regard to HCC is presented. In particular, the role of proinflammatory Th1 and anti-inflammatory Th2 cytokine imbalances in the microenvironment of HCC patients with metastasis and the possible clinical significance of these findings are addressed. Overall, multiple studies, spanning many decades, have begun to elucidate the important role of cytokines in HCC.
Collapse
Affiliation(s)
- Anuradha Budhu
- National Cancer Institute, 37 Convent Dr., Bldg. 37, Rm. 3044A, Bethesda, MD 20892, USA
| | | |
Collapse
|
30
|
Abstract
Pharmacologic transgene-expression dosing is considered essential for future gene therapy scenarios. Genetic interventions require precise transcription or translation fine-tuning of therapeutic transgenes to enable their titration into the therapeutic window, to adapt them to daily changing dosing regimes of the patient, to integrate them seamlessly into the patient's transcriptome orchestra, and to terminate their expression after successful therapy. In recent years, decisive progress has been achieved in designing high-precision trigger-inducible mammalian transgene control modalities responsive to clinically licensed and inert heterologous molecules or to endogenous physiologic signals. Availability of a portfolio of compatible transcription control systems has enabled assembly of higher-order control circuitries providing simultaneous or independent control of several transgenes and the design of (semi-)synthetic gene networks, which emulate digital expression switches, regulatory transcription cascades, epigenetic expression imprinting, and cellular transcription memories. This review provides an overview of cutting-edge developments in transgene control systems, of the design of synthetic gene networks, and of the delivery of such systems for the prototype treatment of prominent human disease phenotypes.
Collapse
Affiliation(s)
- Wilfried Weber
- Institute for Chemical and Bio-Engineering, Swiss Federal Institute of Technology Zurich-ETH Zurich, ETH Hoenggerberg HCI F 115, Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
| | | |
Collapse
|
31
|
Shin EC, Protzer U, Untergasser A, Feinstone SM, Rice CM, Hasselschwert D, Rehermann B. Liver-directed gamma interferon gene delivery in chronic hepatitis C. J Virol 2005; 79:13412-20. [PMID: 16227262 PMCID: PMC1262601 DOI: 10.1128/jvi.79.21.13412-13420.2005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gamma interferon (IFN-gamma) has been shown to inhibit replication of subgenomic and genomic hepatitis C virus (HCV) RNAs in vitro and to noncytolytically suppress hepatitis B virus (HBV) replication in vivo. IFN-gamma is also known for its immunomodulatory effects and as a marker of a successful cellular immune response to HCV. Therapeutic expression of IFN-gamma in the liver may therefore facilitate resolution of chronic hepatitis C, an infection that is rarely resolved spontaneously. To analyze immunomodulatory and antiviral effects of liver-specific IFN-gamma expression in vivo, we intravenously injected two persistently HCV-infected chimpanzees twice with a recombinant, replication-deficient HBV vector and subsequently with a recombinant adenoviral vector. These vectors expressed human IFN-gamma under control of HBV- and liver-specific promoters, respectively. Gene transfer resulted in a transient increase of intrahepatic IFN-gamma mRNA, without increase in serum alanine aminotransferase levels. Ex vivo analysis of peripheral blood lymphocytes demonstrated enhanced CD16 expression on T cells and upregulation of the liver-homing marker CXCR3. Moreover, an increased frequency of HCV-specific T cells was detected ex vivo in the peripheral blood and in vitro in liver biopsy-derived, antigen-nonspecifically expanded T-cell lines. None of these immunologic effects were observed in the third chimpanzee injected with an HBV control vector. Despite these immunologic effects of the experimental vector, however, IFN-gamma gene transfer did not result in a significant and long-lasting decrease of HCV titers. In conclusion, liver-directed IFN-gamma gene delivery resulted in HCV-specific and nonspecific activation of cellular immune responses but did not result in effective control of HCV replication.
Collapse
Affiliation(s)
- Eui-Cheol Shin
- Liver Diseases Branch, NIDDK, National Institutes of Health, DHHS, 10 Center Drive, Room 9B16, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|