101
|
Synergistic anti-HCV broadly neutralizing human monoclonal antibodies with independent mechanisms. Proc Natl Acad Sci U S A 2017; 115:E82-E91. [PMID: 29255018 DOI: 10.1073/pnas.1718441115] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
There is an urgent need for a vaccine to combat the hepatitis C virus (HCV) pandemic, and induction of broadly neutralizing monoclonal antibodies (bNAbs) against HCV is a major goal of vaccine development. Even within HCV genotype 1, no single bNAb effectively neutralizes all viral strains, so induction of multiple neutralizing monoclonal antibodies (NAbs) targeting distinct epitopes may be necessary for protective immunity. Therefore, identification of optimal NAb combinations and characterization of NAb interactions can guide vaccine development. We analyzed neutralization profiles of 12 human NAbs across diverse HCV strains, assigning the NAbs to two functionally distinct clusters. We then measured neutralizing breadth of 35 NAb combinations against genotype 1 isolates, with each combination including one NAb from each neutralization cluster. Many NAbs displayed complementary neutralizing breadth, forming combinations with greater neutralization across diverse strains than any individual bNAb. Remarkably, one of the most broadly neutralizing combinations of two NAbs, designated HEPC74/HEPC98, also displayed enhanced potency, with interactions matching the Bliss independence model, suggesting that these NAbs inhibit HCV infection through independent mechanisms. Subsequent experiments showed that HEPC74 primarily blocks HCV envelope protein binding to CD81, while HEPC98 primarily blocks binding to scavenger receptor B1 and heparan sulfate. Together, these data identify a critical vulnerability resulting from the reliance of HCV on multiple cell surface receptors, suggesting that vaccine induction of multiple NAbs with distinct neutralization profiles is likely to enhance the breadth and potency of the humoral immune response against HCV.
Collapse
|
102
|
Shawa IT, Felmlee DJ, Hegazy D, Sheridan DA, Cramp ME. Exploration of potential mechanisms of hepatitis C virus resistance in exposed uninfected intravenous drug users. J Viral Hepat 2017; 24:1082-1088. [PMID: 28475247 DOI: 10.1111/jvh.12720] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022]
Abstract
A rare outcome following exposure to hepatitis C virus (HCV) is a lack of observable infection as clinically measured by HCV RNA- or HCV-recognizing antibodies. The population who exhibit this trait is termed exposed uninfected (EU). Increasing evidence has refined characterization of these individuals, distinct from those who become infected but spontaneously clear HCV. Study of the EU population is highly pertinent for the discovery of antiviral mechanisms of resistance that can reveal antiviral therapeutic strategies. This review provides an overview of similarities and differences of the EU population relative to spontaneous resolvers and the majority whom develop chronic HCV infection, and focusses on possible mechanisms of resistance including innate and adaptive immunity, genetics and lipid interactions.
Collapse
Affiliation(s)
- I T Shawa
- Hepatology Research Group, Institute of Translational and Stratified Medicine, , Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - D J Felmlee
- Hepatology Research Group, Institute of Translational and Stratified Medicine, , Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - D Hegazy
- Hepatology Research Group, Institute of Translational and Stratified Medicine, , Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - D A Sheridan
- Hepatology Research Group, Institute of Translational and Stratified Medicine, , Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - M E Cramp
- Hepatology Research Group, Institute of Translational and Stratified Medicine, , Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| |
Collapse
|
103
|
Torresi J. The Rationale for a Preventative HCV Virus-Like Particle (VLP) Vaccine. Front Microbiol 2017; 8:2163. [PMID: 29163442 PMCID: PMC5674006 DOI: 10.3389/fmicb.2017.02163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022] Open
Abstract
HCV represents a global health problem with ~200 million individuals currently infected, worldwide. With the high cost of antiviral therapies, the global burden of chronic hepatitis C infection (CHCV) infection will be substantially reduced by the development of an effective vaccine for HCV. The field of HCV vaccines is generally divided into proponents of strategies to induce neutralizing antibodies (NAb) and those who propose to elicit cell mediated immunity (CMI). However, for a hepatitis C virus (HCV) vaccine to be effective in preventing infection, it must be capable of generating cross-reactive CD4+, CD8+ T cell, and NAb responses that will cover the major viral genotypes. Simulation models of hepatitis C have predicted that a vaccine of even modest efficacy and coverage will significantly reduce the incidence of hepatitis C. A HCV virus like particle (VLP) based vaccine would fulfill the requirement of delivering critical conformational neutralizing epitopes in addition to providing HCV specific CD4+ and CD8+ epitopes. Several approaches have been reported including insect cell-derived genotype 1b HCV VLPs; a human liver-derived quadrivalent genotype 1a, 1b, 2, and 3a vaccine; a genotype 1a HCV E1 and E2 glycoprotein/MLV Gag pseudotype VLP vaccine; and chimeric HBs-HCV VLP vaccines. All to result in the production of cross-NAb and/or T cell responses against HCV. This paper summarizes the evidence supporting the development of a HCV VLP based vaccine.
Collapse
Affiliation(s)
- Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
104
|
Mendoza JL, Schneider WM, Hoffmann HH, Vercauteren K, Jude KM, Xiong A, Moraga I, Horton TM, Glenn JS, de Jong YP, Rice CM, Garcia KC. The IFN-λ-IFN-λR1-IL-10Rβ Complex Reveals Structural Features Underlying Type III IFN Functional Plasticity. Immunity 2017; 46:379-392. [PMID: 28329704 DOI: 10.1016/j.immuni.2017.02.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/08/2017] [Accepted: 01/26/2017] [Indexed: 12/31/2022]
Abstract
Type III interferons (IFN-λs) signal through a heterodimeric receptor complex composed of the IFN-λR1 subunit, specific for IFN-λs, and interleukin-10Rβ (IL-10Rβ), which is shared by multiple cytokines in the IL-10 superfamily. Low affinity of IL-10Rβ for cytokines has impeded efforts aimed at crystallizing cytokine-receptor complexes. We used yeast surface display to engineer a higher-affinity IFN-λ variant, H11, which enabled crystallization of the ternary complex. The structure revealed that IL-10Rβ uses a network of tyrosine residues as hydrophobic anchor points to engage IL-10 family cytokines that present complementary hydrophobic binding patches, explaining its role as both a cross-reactive but cytokine-specific receptor. H11 elicited increased anti-proliferative and antiviral activities in vitro and in vivo. In contrast, engineered higher-affinity type I IFNs did not increase antiviral potency over wild-type type I IFNs. Our findings provide insight into cytokine recognition by the IL-10R family and highlight the plasticity of type III interferon signaling and its therapeutic potential.
Collapse
Affiliation(s)
- Juan L Mendoza
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William M Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Hans-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Koen Vercauteren
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Kevin M Jude
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anming Xiong
- Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ignacio Moraga
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tim M Horton
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey S Glenn
- Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ype P de Jong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Center for the Study of Hepatitis C, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
105
|
Colpitts CC, Chung RT, Baumert TF. Entry Inhibitors: A Perspective for Prevention of Hepatitis C Virus Infection in Organ Transplantation. ACS Infect Dis 2017; 3:620-623. [PMID: 28812869 DOI: 10.1021/acsinfecdis.7b00091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Entry inhibitors are emerging as an attractive class of therapeutics for hepatitis C virus (HCV) infection. Entry inhibitors target either virion-associated factors or cellular factors necessary for infection. By blocking entry into cells, entry inhibitors prevent both the establishment of persistent reservoirs and the emergence of resistant variants during viral replication. Furthermore, entry inhibitors protect naïve cells from virus-induced alterations. Combining entry inhibitors with direct-acting antivirals (DAAs) may therefore improve treatment outcomes, particularly in the context of organ transplantation. The role of DAAs in transplantation, while still under clinical investigation, carries the risk of recipient infection and HCV-induced disease, since DAAs act only after infection is established. Thus, entry inhibitors provide a perspective to improve patient outcomes during organ transplantation. Applying this approach for transplant of organs from HCV-positive donors to HCV-negative recipients may also contribute to alleviate the medical burden of organ shortage.
Collapse
Affiliation(s)
- Che C. Colpitts
- Inserm, U1110, Institut de Recherche
sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000 Strasbourg, France
- Université de Strasbourg, 67000 Strasbourg, France
- Division of Infection
and Immunity, University College London, WC1E 6BT London, United Kingdom
| | - Raymond T. Chung
- Liver
Center and Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche
sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000 Strasbourg, France
- Université de Strasbourg, 67000 Strasbourg, France
- Institut
Hospitalo-Universitaire, Pôle Hépato-digestif, Hopitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
106
|
Li D, Wang X, von Schaewen M, Tao W, Zhang Y, Heller B, Hrebikova G, Deng Q, Sun Q, Ploss A, Zhong J, Huang Z. Immunization With a Subunit Hepatitis C Virus Vaccine Elicits Pan-Genotypic Neutralizing Antibodies and Intrahepatic T-Cell Responses in Nonhuman Primates. J Infect Dis 2017; 215:1824-1831. [PMID: 28398489 DOI: 10.1093/infdis/jix180] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/07/2017] [Indexed: 12/22/2022] Open
Abstract
Background The global control of hepatitis C virus (HCV) infection remains a great burden, owing to the high prices and potential drug resistance of the new direct-acting antivirals (DAAs), as well as the risk of reinfection in DAA-cured patients. Thus, a prophylactic vaccine for HCV is of great importance. We previously reported that a single recombinant soluble E2 (sE2) vaccine produced in insect cells was able to induce broadly neutralizing antibodies (NAbs) and prevent HCV infection in mice. Here the sE2 vaccine was evaluated in non-human primates. Methods Rhesus macaques were immunized with sE2 vaccine in combination with different adjuvants. Vaccine-induced NAbs in antisera were tested for neutralization activities against a panel of cell culture-derived HCV (HCVcc), while T-cell responses were evaluated in splenocytes, peripheral blood mononuclear cells, and hepatic lymphocytes. Results sE2 is able to elicit NAbs against HCVcc harboring structural proteins from multiple HCV genotypes in rhesus macaques. Moreover, sE2-immunized macaques developed systemic and intrahepatic memory T cells specific for E2. A significant correlation between the sE2-specific immunoglobulin G titers and neutralization spectrum was observed, highlighting the essential role of sE2 immunogenicity on achieving broad NAbs. Conclusions sE2 is a promising HCV vaccine candidate that warrants further preclinical and clinical development.
Collapse
Affiliation(s)
- Dapeng Li
- Unit of Vaccinology and Antiviral Strategies.,Unit of Viral Hepatitis, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai
| | - Xuesong Wang
- Unit of Vaccinology and Antiviral Strategies.,Unit of Viral Hepatitis, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai
| | | | - Wanyin Tao
- Unit of Viral Hepatitis, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai
| | | | - Brigitte Heller
- Department of Molecular Biology, Princeton University, New Jersey
| | | | - Qiang Deng
- Unit of Vaccinology and Antiviral Strategies
| | - Qiang Sun
- Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai.,Suzhou Nonhuman Primate Facility, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Suzhou, China
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, New Jersey
| | - Jin Zhong
- Unit of Viral Hepatitis, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai
| | - Zhong Huang
- Unit of Vaccinology and Antiviral Strategies
| |
Collapse
|
107
|
Ortega-Prieto AM, Dorner M. Immune Evasion Strategies during Chronic Hepatitis B and C Virus Infection. Vaccines (Basel) 2017; 5:E24. [PMID: 28862649 PMCID: PMC5620555 DOI: 10.3390/vaccines5030024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 12/15/2022] Open
Abstract
Both hepatitis B virus (HBV) and hepatitis C virus (HCV) infections are a major global healthcare problem with more than 240 million and 70 million infected, respectively. Both viruses persist within the liver and result in progressive liver disease, resulting in liver fibrosis, cirrhosis and hepatocellular carcinoma. Strikingly, this pathogenesis is largely driven by immune responses, unable to clear an established infection, rather than by the viral pathogens themselves. Even though disease progression is very similar in both infections, HBV and HCV have evolved distinct mechanisms, by which they ensure persistence within the host. Whereas HCV utilizes a cloak-and-dagger approach, disguising itself as a lipid-like particle and immediately crippling essential pattern-recognition pathways, HBV has long been considered a "stealth" virus, due to the complete absence of innate immune responses during infection. Recent developments and access to improved model systems, however, revealed that even though it is among the smallest human-tropic viruses, HBV may, in addition to evading host responses, employ subtle immune evasion mechanisms directed at ensuring viral persistence in the absence of host responses. In this review, we compare the different strategies of both viruses to ensure viral persistence by actively interfering with viral recognition and innate immune responses.
Collapse
Affiliation(s)
| | - Marcus Dorner
- Section of Virology, Department of Medicine, Imperial College London, London W2 1PG, UK.
| |
Collapse
|
108
|
Ortega-Prieto AM, Dorner M. Novel methods to prevent HCV transmission. Future Virol 2017. [DOI: 10.2217/fvl-2017-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
| | - Marcus Dorner
- Section of Virology, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
109
|
Andreo U, de Jong YP, Scull MA, Xiao JW, Vercauteren K, Quirk C, Mommersteeg MC, Bergaya S, Menon A, Fisher EA, Rice CM. Analysis of Hepatitis C Virus Particle Heterogeneity in Immunodeficient Human Liver Chimeric fah-/- Mice. Cell Mol Gastroenterol Hepatol 2017; 4:405-417. [PMID: 28936471 PMCID: PMC5602752 DOI: 10.1016/j.jcmgh.2017.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 07/10/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV) is a leading cause of chronic liver diseases and the most common indication for liver transplantation in the United States. HCV particles in the blood of infected patients are characterized by heterogeneous buoyant densities, likely owing to HCV association with lipoproteins. However, clinical isolates are not infectious in vitro and the relative infectivity of the particles with respect to their buoyant density therefore cannot be determined, pointing to the need for better in vivo model systems. METHODS To analyze the evolution of the buoyant density of in vivo-derived infectious HCV particles over time, we infected immunodeficient human liver chimeric fumaryl acetoacetate hydrolase-/- mice with J6/JFH1 and performed ultracentrifugation of infectious mouse sera on isopicnic iodixanol gradients. We also evaluated the impact of a high sucrose diet, which has been shown to increase very-low-density lipoprotein secretion by the liver in rodents, on lipoprotein and HCV particle characteristics. RESULTS Similar to the severe combined immunodeficiency disease/Albumin-urokinase plasminogen activator human liver chimeric mouse model, density fractionation of infectious mouse serum showed higher infectivity in the low-density fractions early after infection. However, over the course of the infection, viral particle heterogeneity increased and the overall in vitro infectivity diminished without loss of the human liver graft over time. In mice provided with a sucrose-rich diet we observed a minor shift in HCV infectivity toward lower density that correlated with a redistribution of triglycerides and cholesterol among lipoproteins. CONCLUSIONS Our work indicates that the heterogeneity in buoyant density of infectious HCV particles evolves over the course of infection and can be influenced by diet.
Collapse
Key Words
- Alb-uPA, Albumin-urokinase plasminogen activator
- CETP, cholesterol ester transfer protein
- FAH, fumaryl acetoacetate hydrolase
- FNRG, absence of fumaryl acetoacetate hydrolase on a immunodeficient NOD Rag gamma IL2 deficient mouse background
- FPLC, fast-performance liquid chromatography
- HCV
- HCV, hepatitis C virus
- HCVcc, cell culture–derived hepatitis C virus
- HDL, high-density lipoprotein
- Human Liver Chimeric Mice
- LVP, lipoviroparticle
- Lipoprotein
- Mouse Model
- NRG, nod rag γ
- NTBC, nitisinone
- PBS, phosphate-buffered saline
- SCID, severe combined immunodeficiency disease
- VLDL, very low density lipoprotein
- apo, apolipoprotein
Collapse
Affiliation(s)
- Ursula Andreo
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
- Correspondence Address correspondence to: Ursula Andreo, PhD, Center for the Study of Hepatitis C, The Rockefeller University, 1230 York Avenue, Box 64, New York, New York 10065. fax: (212) 327-7048.Center for the Study of Hepatitis CThe Rockefeller University1230 York AvenueBox 64New YorkNew York 10065
| | - Ype P. de Jong
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
- Division of Gastroenterology and Hepatology, Center for the Study of Hepatitis C, Weill Cornell Medical College, New York, New York
| | - Margaret A. Scull
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| | - Jing W. Xiao
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| | - Koen Vercauteren
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| | - Corrine Quirk
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| | | | - Sonia Bergaya
- Division of Cardiology, Department of Medicine, New York University Langone Medical Center, New York, New York
| | - Arjun Menon
- Division of Cardiology, Department of Medicine, New York University Langone Medical Center, New York, New York
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University Langone Medical Center, New York, New York
| | - Charles M. Rice
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| |
Collapse
|
110
|
Fujiwara S. Humanized mice: A brief overview on their diverse applications in biomedical research. J Cell Physiol 2017; 233:2889-2901. [PMID: 28543438 DOI: 10.1002/jcp.26022] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 02/06/2023]
Abstract
Model animals naturally differ from humans in various respects and results from the former are not directly translatable to the latter. One approach to address this issue is humanized mice that are defined as mice engrafted with functional human cells or tissues. In humanized mice, we can investigate the development and function of human cells or tissues (including their products encoded by human genes) in the in vivo context of a small animal. As such, humanized mouse models have played important roles that cannot be substituted by other animal models in various areas of biomedical research. Although there are obvious limitations in humanized mice and we may need some caution in interpreting the results obtained from them, it is reasonably expected that they will be utilized in increasingly diverse areas of biomedical research, as the technology for preparing humanized mice are rapidly improved. In this review, I will describe the methodology for generating humanized mice and overview their recent applications in various disciplines including immunology, infectious diseases, drug metabolism, and neuroscience.
Collapse
Affiliation(s)
- Shigeyoshi Fujiwara
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo, Japan.,Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| |
Collapse
|
111
|
Hollevoet K, Declerck PJ. State of play and clinical prospects of antibody gene transfer. J Transl Med 2017; 15:131. [PMID: 28592330 PMCID: PMC5463339 DOI: 10.1186/s12967-017-1234-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/31/2017] [Indexed: 12/31/2022] Open
Abstract
Recombinant monoclonal antibodies (mAbs) are one of today's most successful therapeutic classes in inflammatory diseases and oncology. A wider accessibility and implementation, however, is hampered by the high product cost and prolonged need for frequent administration. The surge in more effective mAb combination therapies further adds to the costs and risk of toxicity. To address these issues, antibody gene transfer seeks to administer to patients the mAb-encoding nucleotide sequence, rather than the mAb protein. This allows the body to produce its own medicine in a cost- and labor-effective manner, for a prolonged period of time. Expressed mAbs can be secreted systemically or locally, depending on the production site. The current review outlines the state of play and clinical prospects of antibody gene transfer, thereby highlighting recent innovations, opportunities and remaining hurdles. Different expression platforms and a multitude of administration sites have been pursued. Viral vector-mediated mAb expression thereby made the most significant strides. Therapeutic proof of concept has been demonstrated in mice and non-human primates, and intramuscular vectored mAb therapy is under clinical evaluation. However, viral vectors face limitations, particularly in terms of immunogenicity. In recent years, naked DNA has gained ground as an alternative. Attained serum mAb titers in mice, however, remain far below those obtained with viral vectors, and robust pharmacokinetic data in larger animals is limited. The broad translatability of DNA-based antibody therapy remains uncertain, despite ongoing evaluation in patients. RNA presents another emerging platform for antibody gene transfer. Early reports in mice show that mRNA may be able to rival with viral vectors in terms of generated serum mAb titers, although expression appears more short-lived. Overall, substantial progress has been made in the clinical translation of antibody gene transfer. While challenges persist, clinical prospects are amplified by ongoing innovations and the versatility of antibody gene transfer. Clinical introduction can be expedited by selecting the platform approach currently best suited for the mAb or disease of interest. Innovations in expression platform, administration and antibody technology are expected to further improve overall safety and efficacy, and unlock the vast clinical potential of antibody gene transfer.
Collapse
Affiliation(s)
- Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
112
|
Designing an HCV vaccine: a unique convergence of prevention and therapy? Curr Opin Virol 2017; 23:113-119. [PMID: 28550816 DOI: 10.1016/j.coviro.2017.03.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/27/2017] [Indexed: 12/23/2022]
Abstract
Direct acting antivirals can cure chronic hepatitis C virus (HCV) infection but whether they will reduce global liver disease burden is uncertain. Most chronic infections are undiagnosed and transmission has increased in recent years. The first trial of a preventive vaccine is now underway in humans at risk for HCV infection. It will test the novel hypothesis that T cell-mediated immunity alone can prevent persistent HCV infection. Another vaccine that elicits neutralizing antibodies is at an advanced stage of development. Attention is turning to the understudied question of whether direct acting antiviral (DAA) cure of chronic infection restores HCV immunity. If not, it will be important to determine if preventive vaccines can also act therapeutically to reverse immune dysfunction and protect from re-infection.
Collapse
|
113
|
Chaipan C, Pryszlak A, Dean H, Poignard P, Benes V, Griffiths AD, Merten CA. Single-Virus Droplet Microfluidics for High-Throughput Screening of Neutralizing Epitopes on HIV Particles. Cell Chem Biol 2017; 24:751-757.e3. [PMID: 28552581 DOI: 10.1016/j.chembiol.2017.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/07/2017] [Accepted: 05/03/2017] [Indexed: 11/18/2022]
Abstract
Analyzing surface epitopes of single HIV particles holds great potential for the development of vaccine candidates. However, existing technologies do not allow corresponding screens at high throughput. We present here a single-virus droplet-based microfluidics platform enabling sorting of millions of HIV-1 particles with >99% efficiency, based on the expression of epitopes recognized by broadly neutralizing antibodies. We show that virus particles displaying these epitopes can be identified, sorted, and analyzed by next-generation sequencing: an approximately 1,900-fold enrichment of viral particles displaying neutralizing epitopes could be obtained in a single sort, thus opening the way for screening diverse virus libraries with optimal antigenic features for HIV vaccine candidates.
Collapse
Affiliation(s)
- Chawaree Chaipan
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Anna Pryszlak
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Hansi Dean
- International AIDS Vaccine Initiative, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Pascal Poignard
- International AIDS Vaccine Initiative, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vladimir Benes
- European Molecular Biology Laboratory (EMBL), Genomics Core Facility, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Andrew D Griffiths
- Ecole Supérieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), CNRS UMR 8231, 75231 Paris, France
| | - Christoph A Merten
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
114
|
Reply to Padmanabhan and Dixit: Hepatitis C virus entry inhibitors for optimally boosting direct-acting antiviral-based treatments. Proc Natl Acad Sci U S A 2017; 114:E4527-E4529. [PMID: 28512226 DOI: 10.1073/pnas.1705234114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
115
|
Bailey JR, Flyak AI, Cohen VJ, Li H, Wasilewski LN, Snider AE, Wang S, Learn GH, Kose N, Loerinc L, Lampley R, Cox AL, Pfaff JM, Doranz BJ, Shaw GM, Ray SC, Crowe JE. Broadly neutralizing antibodies with few somatic mutations and hepatitis C virus clearance. JCI Insight 2017; 2:92872. [PMID: 28469084 PMCID: PMC5414559 DOI: 10.1172/jci.insight.92872] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/21/2017] [Indexed: 01/15/2023] Open
Abstract
Here, we report the isolation of broadly neutralizing mAbs (bNAbs) from persons with broadly neutralizing serum who spontaneously cleared hepatitis C virus (HCV) infection. We found that bNAbs from two donors bound the same epitope and were encoded by the same germline heavy chain variable gene segment. Remarkably, these bNAbs were encoded by antibody variable genes with sparse somatic mutations. For one of the most potent bNAbs, these somatic mutations were critical for antibody neutralizing breadth and for binding to autologous envelope variants circulating late in infection. However, somatic mutations were not necessary for binding of the bNAb unmutated ancestor to envelope proteins of early autologous transmitted/founder viruses. This study identifies a public B cell clonotype favoring early recognition of a conserved HCV epitope, proving that anti-HCV bNAbs can achieve substantial neutralizing breadth with relatively few somatic mutations, and identifies HCV envelope variants that favored selection and maturation of an anti-HCV bNAb in vivo. These data provide insight into the molecular mechanisms of immune-mediated clearance of HCV infection and present a roadmap to guide development of a vaccine capable of stimulating anti-HCV bNAbs with a physiologic number of somatic mutations characteristic of vaccine responses.
Collapse
Affiliation(s)
- Justin R. Bailey
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Andrew I. Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Valerie J. Cohen
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hui Li
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lisa N. Wasilewski
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Anna E. Snider
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shuyi Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gerald H. Learn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Leah Loerinc
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Rebecca Lampley
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Andrea L. Cox
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - George M. Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stuart C. Ray
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
116
|
Xiong H, Rong X, Wang M, Xu R, Huang K, Liao Q, Huang J, Chen J, Li C, Tang X, Shan Z, Zhang M, Nelson K, Fu Y. HBV/HCV co-infection is associated with a high level of HCV spontaneous clearance among drug users and blood donors in China. J Viral Hepat 2017; 24:312-319. [PMID: 27943542 DOI: 10.1111/jvh.12644] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/20/2016] [Indexed: 12/11/2022]
Abstract
Understanding the biology of spontaneous clearance of hepatitis C virus (HCV) infection could lead to improved strategies to prevent the sequelae associated with chronic HCV infection. Chronic infections with hepatitis virus are very common in China, but the factors associated with spontaneous clearance of HCV have not been adequately studied. We evaluated the spontaneous clearance of HCV among 1918 drug users and 1526 HCV-seropositive blood donors in Guangzhou, China. Among participants who were co-infected with hepatitis B virus (HBV), 41.38% of drug users and 39.47% of blood donors had cleared their HCV infection without antiviral therapy compared to 9.41% of drug users and 16.73% of blood donors who were mono-infected with a single virus (P<.01). The proportion of subjects who had cleared their HCV infection was significantly greater in the co-infected subjects whose serum HBV DNA was greater than 2000IU/mL than those with lower levels. A multiple logistic regression analysis found female gender, IL28B rs8099917 TT genotype, HBV co-infection and blood donors (vs drug users) associated with increased spontaneous clearance of HCV infection. Although acute HCV infections are common in China, the incidence of chronic HCV may be reduced among the high prevalence of chronic HBV and IL28B genotypes associated with spontaneous clearance of HCV in Chinese populations.
Collapse
Affiliation(s)
- H Xiong
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - X Rong
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - M Wang
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - R Xu
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - K Huang
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - Q Liao
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - J Huang
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - J Chen
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - C Li
- Department of Transfusion Medicine, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - X Tang
- Department of Transfusion Medicine, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Z Shan
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| | - M Zhang
- Faculty of Infectious Diseases, Department of Epidemiology and Biostatistics, University of Georgia, Athens, GA, USA
| | - K Nelson
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Y Fu
- Guangzhou Blood Center, Guangzhou, Guangdong, China
- The Key Medical Disciplines and Specialties Program of Guangzhou, Guangdong, China
| |
Collapse
|
117
|
Vietheer PT, Boo I, Gu J, McCaffrey K, Edwards S, Owczarek C, Hardy MP, Fabri L, Center RJ, Poumbourios P, Drummer HE. The core domain of hepatitis C virus glycoprotein E2 generates potent cross-neutralizing antibodies in guinea pigs. Hepatology 2017; 65:1117-1131. [PMID: 27997681 PMCID: PMC5408392 DOI: 10.1002/hep.28989] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED A vaccine that prevents hepatitis C virus (HCV) infection is urgently needed to support an emerging global elimination program. However, vaccine development has been confounded because of HCV's high degree of antigenic variability and the preferential induction of type-specific immune responses with limited potency against heterologous viral strains and genotypes. We showed previously that deletion of the three variable regions from the E2 receptor-binding domain (Δ123) increases the ability of human broadly neutralizing antibodies (bNAbs) to inhibit E2-CD81 receptor interactions, suggesting improved bNAb epitope exposure. In this study, the immunogenicity of Δ123 was examined. We show that high-molecular-weight forms of Δ123 elicit distinct antibody specificities with potent and broad neutralizing activity against all seven HCV genotypes. Antibody competition studies revealed that immune sera raised to high-molecular-weight Δ123 was poly specific, given that it inhibited the binding of human bNAbs directed to three major neutralization epitopes on E2. By contrast, the immune sera raised to monomeric Δ123 predominantly blocked the binding of a non-neutralizing antibody to Δ123, while having reduced ability to block bNAb binding to E2, and neutralization was largely toward the homologous genotype. This increased ability of oligomeric Δ123 to generate bNAbs correlates with occlusion of the non-neutralizing face of E2 in this glycoprotein form. CONCLUSION The results from this study reveal new information on the antigenic and immunogenic potential of E2-based immunogens and provide a pathway for the development of a simple, recombinant protein-based prophylactic vaccine for HCV with potential for universal protection. (Hepatology 2017;65:1117-1131).
Collapse
Affiliation(s)
- Patricia T. Vietheer
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
- Department of MicrobiologyMonash UniversityClaytonAustralia
| | - Irene Boo
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
| | - Jun Gu
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
- Department of MicrobiologyMonash UniversityClaytonAustralia
| | - Kathleen McCaffrey
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneParkvilleAustralia
| | | | | | | | | | - Rob J. Center
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneParkvilleAustralia
| | - Pantelis Poumbourios
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
- Department of MicrobiologyMonash UniversityClaytonAustralia
| | - Heidi E. Drummer
- Centre for Biomedical ResearchBurnet InstituteMelbourneAustralia
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and ImmunityUniversity of MelbourneParkvilleAustralia
- Department of MicrobiologyMonash UniversityClaytonAustralia
| |
Collapse
|
118
|
Song MY, Nguyen D, Hong SW, Kim BC. Broadly reactive aptamers targeting bacteria belonging to different genera using a sequential toggle cell-SELEX. Sci Rep 2017; 7:43641. [PMID: 28272554 PMCID: PMC5341558 DOI: 10.1038/srep43641] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/25/2017] [Indexed: 12/23/2022] Open
Abstract
Conventional cell-SELEX aims to isolate aptamers to a single unique target bacteria species. We propose a method to isolate single-stranded DNA aptamers that have broad reactivity to multiple bacterial targets belonging to different genera. The key of the proposed method is that targets of interest are changed sequentially at each SELEX round. The general scheme was examined using six bacteria from different genera, Escherichia coli, Enterobacter aerogenes, Klebsiella pneumoniae, Citrobacter freundii, Bacillus subtilis, and Staphylococcus epidermidis (four gram-negative and two gram-positive bacteria). In the first round of SELEX, the DNA library was incubated with E. coli and amplicons bound to E. coli were separated. The amplicons were sequentially separated by incubation with E. aerogenes, K. pneumoniae, C. freundii, B. subtilis, and S. epidermidis at each SELEX. The amplicons obtained using the last bacterial species were incubated again with the first bacterial species and this loop was repeated two more times. We refer to this method as sequential toggle cell-SELEX (STC-SELEX). The isolated aptamers had dissociation constants of 9.22–38.5 nM and had no affinity to other bacteria that were not included in STC-SELEX. These results demonstrate the potential to isolate aptamers with broad affinity to bacterial taxa in different genera.
Collapse
Affiliation(s)
- Min Young Song
- Center for Environment, Health and Welfare Research, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Dung Nguyen
- Department of Energy and Environmental Engineering, University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Seok Won Hong
- Department of Energy and Environmental Engineering, University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.,Center for Water Resources Cycle Research, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Byoung Chan Kim
- Center for Environment, Health and Welfare Research, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Department of Energy and Environmental Engineering, University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
119
|
El-Diwany R, Cohen VJ, Mankowski MC, Wasilewski LN, Brady JK, Snider AE, Osburn WO, Murrell B, Ray SC, Bailey JR. Extra-epitopic hepatitis C virus polymorphisms confer resistance to broadly neutralizing antibodies by modulating binding to scavenger receptor B1. PLoS Pathog 2017; 13:e1006235. [PMID: 28235087 PMCID: PMC5342271 DOI: 10.1371/journal.ppat.1006235] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 03/08/2017] [Accepted: 02/11/2017] [Indexed: 12/15/2022] Open
Abstract
Broadly-neutralizing monoclonal antibodies (bNAbs) may guide vaccine development for highly variable viruses including hepatitis C virus (HCV), since they target conserved viral epitopes that could serve as vaccine antigens. However, HCV resistance to bNAbs could reduce the efficacy of a vaccine. HC33.4 and AR4A are two of the most potent anti-HCV human bNAbs characterized to date, binding to highly conserved epitopes near the amino- and carboxy-terminus of HCV envelope (E2) protein, respectively. Given their distinct epitopes, it was surprising that these bNAbs showed similar neutralization profiles across a panel of natural HCV isolates, suggesting that some viral polymorphisms may confer resistance to both bNAbs. To investigate this resistance, we developed a large, diverse panel of natural HCV envelope variants and a novel computational method to identify bNAb resistance polymorphisms in envelope proteins (E1 and E2). By measuring neutralization of a panel of HCV pseudoparticles by 10 μg/mL of each bNAb, we identified E1E2 variants with resistance to one or both bNAbs, despite 100% conservation of the AR4A binding epitope across the panel. We discovered polymorphisms outside of either binding epitope that modulate resistance to both bNAbs by altering E2 binding to the HCV co-receptor, scavenger receptor B1 (SR-B1). This study is focused on a mode of neutralization escape not addressed by conventional analysis of epitope conservation, highlighting the contribution of extra-epitopic polymorphisms to bNAb resistance and presenting a novel mechanism by which HCV might persist even in the face of an antibody response targeting multiple conserved epitopes. Generation of an immune response that is protective against a wide variety of naturally occurring isolates is necessary for vaccines against highly variable viruses like hepatitis C virus (HCV). Two broadly neutralizing human monoclonal antibodies, HC33.4 and AR4A, neutralize multiple highly divergent HCV isolates, raising hope that a vaccine against HCV is possible. Previous reports have defined the distinct, highly conserved sites on the viral envelope proteins where these antibodies bind. However, little is known about naturally occurring variation in sensitivity of different HCV isolates to these antibodies. We developed a high throughput assay and computational algorithm to evaluate over 100 naturally occurring HCV variants for their sensitivity to these two antibodies, identifying several resistance polymorphisms to each antibody which do not fall within their mapped binding sites. Furthermore, two of these polymorphisms modulate resistance to both antibodies by enhancing or reducing envelope protein binding to HCV co-receptor scavenger receptor B1 (SR-B1). By developing this broadly applicable platform, we have shown the important neutralization resistance conferred by changes distant from antibody binding sites, presenting a potential mechanism by which HCV might persist even in the face of an antibody response targeting multiple conserved sites.
Collapse
Affiliation(s)
- Ramy El-Diwany
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Valerie J. Cohen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Madeleine C. Mankowski
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Lisa N. Wasilewski
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jillian K. Brady
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Anna E. Snider
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William O. Osburn
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ben Murrell
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Stuart C. Ray
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Justin R. Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
120
|
Velázquez-Moctezuma R, Law M, Bukh J, Prentoe J. Applying antibody-sensitive hypervariable region 1-deleted hepatitis C virus to the study of escape pathways of neutralizing human monoclonal antibody AR5A. PLoS Pathog 2017; 13:e1006214. [PMID: 28231271 PMCID: PMC5358973 DOI: 10.1371/journal.ppat.1006214] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 03/20/2017] [Accepted: 02/02/2017] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) is a major cause of end-stage liver diseases. With 3–4 million new HCV infections yearly, a vaccine is urgently needed. A better understanding of virus escape from neutralizing antibodies and their corresponding epitopes are important for this effort. However, for viral isolates with high antibody resistance, or antibodies with moderate potency, it remains challenging to induce escape mutations in vitro. Here, as proof-of-concept, we used antibody-sensitive HVR1-deleted (ΔHVR1) viruses to generate escape mutants for a human monoclonal antibody, AR5A, targeting a rare cross-genotype conserved epitope. By analyzing the genotype 1a envelope proteins (E1/E2) of recovered Core-NS2 recombinant H77/JFH1ΔHVR1 and performing reverse genetic studies we found that resistance to AR5A was caused by substitution L665W, also conferring resistance to the parental H77/JFH1. The mutation did not induce viral fitness loss, but abrogated AR5A binding to HCV particles and intracellular E1/E2 complexes. Culturing J6/JFH1ΔHVR1 (genotype 2a), for which fitness was decreased by L665W, with AR5A generated AR5A-resistant viruses with the substitutions I345V, L665S, and S680T, which we introduced into J6/JFH1 and J6/JFH1ΔHVR1. I345V increased fitness but had no effect on AR5A resistance. L665S impaired fitness and decreased AR5A sensitivity, while S680T combined with L665S compensated for fitness loss and decreased AR5A sensitivity even further. Interestingly, S680T alone had no fitness effect but sensitized the virus to AR5A. Of note, H77/JFH1L665S was non-viable. The resistance mutations did not affect cell-to-cell spread or E1/E2 interactions. Finally, introducing L665W, identified in genotype 1, into genotypes 2–6 parental and HVR1-deleted variants (not available for genotype 4a) we observed diverse effects on viral fitness and a universally pronounced reduction in AR5A sensitivity. Thus, we were able to take advantage of the neutralization-sensitive HVR1-deleted viruses to rapidly generate escape viruses aiding our understanding of the divergent escape pathways used by HCV to evade AR5A. Worldwide hepatitis C virus (HCV) is one of the leading causes of chronic liver diseases, including cirrhosis and cancer. Treatment accessibility is limited and development of a preventive vaccine has proven difficult, partly due to the high mutation rate of the virus. Recent studies of HCV antibody neutralization resistance have revealed important information about escape pathways and barriers to escape for several clinically promising human monoclonal antibodies. However, due to the varying levels of antibody shielding between HCV isolates these studies have been mostly limited to a few neutralization-sensitive HCV isolates. Here, we took advantage of the fact that deletion of the hypervariable region 1 (HVR1) increased antibody sensitivity of HCV isolates by increasing the exposure of important epitopes, thus facilitating studies of antibody escape for neutralization resistant isolates. We identified escape mutations in the envelope glycoprotein E2, at amino acid position L665, which conferred antibody resistance in parental HCV viruses from genotypes 1–6. We found that antibody escape was associated with loss of binding to HCV particles and intracellular envelope protein complexes. We also identified escape substitutions at L665 that were isolate-specific. Thus, our data sheds new light on antibody resistance mechanisms across diverse HCV isolates.
Collapse
Affiliation(s)
- Rodrigo Velázquez-Moctezuma
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Mansun Law
- Department of Immunology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- * E-mail: (JP); (JB)
| | - Jannick Prentoe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- * E-mail: (JP); (JB)
| |
Collapse
|
121
|
Sayed IM, Foquet L, Verhoye L, Abravanel F, Farhoudi A, Leroux-Roels G, Izopet J, Meuleman P. Transmission of hepatitis E virus infection to human-liver chimeric FRG mice using patient plasma. Antiviral Res 2017; 141:150-154. [PMID: 28232247 DOI: 10.1016/j.antiviral.2017.02.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 02/07/2023]
Abstract
Hepatitis E virus (HEV) is considered as an important pathogen in developing countries but there is growing evidence of its increasing significance and prevalence in the Western world. Although most acute HEV infections resolve spontaneously, chronicity has been observed in immunocompromised patients. The study of HEV has been hampered by the absence of practical animal models. Because the in vivo study of HEV was essentially limited to primates and pigs we recently established the human-liver chimeric uPA-SCID mouse model as a useful tool to study HEV infection. Because the humanized FRG mouse model, another type of mouse with humanized liver, is more easily accessible to the scientific community, we investigated its susceptibility to HEV infection. FRG mice were transplanted with human hepatocytes and challenged with different HEV genotypes using different routes of exposure. Our data clearly shows that the humanized FRG mouse is an alternative animal model for the study HEV infection. As observed in the uPA-SCID model, controlled oral inoculation did not lead to active infection. However, intrasplenic injection of genotype 3-infected patient plasma did result into persistent infection. Although the efficiency of transmission was low, this observation corroborates previously published case reports of blood transfusion-associated HEV transmission.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Dept. Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Microbiology and Immunology Department, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Lander Foquet
- Dept. Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| | - Lieven Verhoye
- Dept. Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| | - Florence Abravanel
- INSERM U1043, IFR-BMT, CHU Purpan, Toulouse, France; Université Paul-Sabatier, Toulouse, France; Laboratory of Virology, CHU Purpan, Toulouse, France.
| | - Ali Farhoudi
- Dept. Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| | - Geert Leroux-Roels
- Dept. Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| | - Jacques Izopet
- INSERM U1043, IFR-BMT, CHU Purpan, Toulouse, France; Université Paul-Sabatier, Toulouse, France; Laboratory of Virology, CHU Purpan, Toulouse, France.
| | - Philip Meuleman
- Dept. Clinical Chemistry, Microbiology and Immunology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
122
|
Sanders JW, Ponzio TA. Vectored immunoprophylaxis: an emerging adjunct to traditional vaccination. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2017; 3:3. [PMID: 28883973 PMCID: PMC5531025 DOI: 10.1186/s40794-017-0046-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/16/2017] [Indexed: 01/08/2023]
Abstract
The successful development of effective vaccines has been elusive for many of the world's most important infectious diseases. Additionally, much of the population, such as the aged or immunocompromised, are unable to mount an effective immunologic response for existing vaccines. Vectored Immunoprophylaxis (VIP) is a novel approach designed to address these challenges. Rather than utilizing an antigen to trigger a response from the host's immune system as is normally done with traditional vaccines, VIP genetically engineers the production of tailored antibodies from non-hematopoietic cells, bypassing the humoral immune system. Direct administration of genes encoding for neutralizing antibodies has proven to be effective in both preventing and treating several infectious diseases in animal models. While, a significant amount of work has focused on HIV, including an ongoing clinical trial, the approach has also been shown to be effective for malaria, dengue, hepatitis C, influenza, and more. In addition to presenting itself as a potentially efficient approach to solving long-standing vaccine challenges, the approach may be the best, if not only, method to vaccinate immunocompromised individuals. Many issues still need to be addressed, including which tissue(s) makes the most suitable platform, which vector(s) are most efficient at transducing the platform tissue used to secrete the antibodies, and what are the long-term effects of such a treatment. Here we provide a brief overview of this approach, and its potential application in treating some of the world's most intractable infectious diseases.
Collapse
Affiliation(s)
- John W Sanders
- Wake Forest University School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA.,Salisbury Veterans Affairs Medical Center, Salisbury, NC USA
| | - Todd A Ponzio
- Naval Medical Research Center, 503 Robert Grant Ave, Silver Spring, MD 20910 USA.,Section on Infectious Diseases and Department of Bio-Engineering, Wake Forest University School of Medicine, Winston-Salem, USA
| |
Collapse
|
123
|
Mice Expressing Minimally Humanized CD81 and Occludin Genes Support Hepatitis C Virus Uptake In Vivo. J Virol 2017; 91:JVI.01799-16. [PMID: 27928007 DOI: 10.1128/jvi.01799-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) causes chronic infections in at least 150 million individuals worldwide. HCV has a narrow host range and robustly infects only humans and chimpanzees. The underlying mechanisms for this narrow host range are incompletely understood. At the level of entry, differences in the amino acid sequences between the human and mouse orthologues of two essential host factors, the tetraspanin CD81 and the tight junction protein occludin (OCLN), explain, at least in part, HCV's limited ability to enter mouse hepatocytes. We have previously shown that adenoviral or transgenic overexpression of human CD81 and OCLN facilitates HCV uptake into mouse hepatocytes in vitro and in vivo In efforts to refine these models, we constructed knock-in mice in which the second extracellular loops of CD81 and OCLN were replaced with the respective human sequences, which contain the determinants that are critical for HCV uptake. We demonstrate that the humanized CD81 and OCLN were expressed at physiological levels in a tissue-appropriate fashion. Mice bearing the humanized alleles formed normal tight junctions and did not exhibit any immunologic abnormalities, indicating that interactions with their physiological ligands were intact. HCV entry factor knock-in mice take up HCV with an efficiency similar to that in mice expressing HCV entry factors transgenically or adenovirally, demonstrating the utility of this model for studying HCV infection in vivo IMPORTANCE: At least 150 million individuals are chronically infected with hepatitis C virus (HCV). Chronic hepatitis C can result in progressive liver disease and liver cancer. New antiviral treatments can cure HCV in the majority of patients, but a vaccine remains elusive. To gain a better understanding of the processes culminating in liver failure and cancer and to prioritize vaccine candidates more efficiently, small-animal models are needed. Here, we describe the characterization of a new mouse model in which the parts of two host factors that are essential for HCV uptake, CD81 and occludin (OCLN), which differ between mice and humans, were humanized. We demonstrate that such minimally humanized mice develop normally, express the modified genes at physiological levels, and support HCV uptake. This model is of considerable utility for studying viral entry in the three-dimensional context of the liver and to test approaches aimed at preventing HCV entry.
Collapse
|
124
|
Pietschmann T. Clinically Approved Ion Channel Inhibitors Close Gates for Hepatitis C Virus and Open Doors for Drug Repurposing in Infectious Viral Diseases. J Virol 2017; 91:e01914-16. [PMID: 27807238 PMCID: PMC5215334 DOI: 10.1128/jvi.01914-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection causes severe liver disease and affects ca. 146 million individuals. Novel directly acting antivirals targeting HCV have revolutionized treatment. However, high costs limit access to therapy. Recently, several related drugs used in humans to treat allergies or as neuroleptics emerged as potent HCV cell entry inhibitors. Insights into their antiviral modes of action may increase opportunities for drug repurposing in hepatitis C and possibly other important human viral infections.
Collapse
Affiliation(s)
- Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany, and German Centre for Infection Research, Hannover-Braunschweig, Germany
| |
Collapse
|
125
|
Winer BY, Huang T, Low BE, Avery C, Pais MA, Hrebikova G, Siu E, Chiriboga L, Wiles MV, Ploss A. Recapitulation of treatment response patterns in a novel humanized mouse model for chronic hepatitis B virus infection. Virology 2016; 502:63-72. [PMID: 28006671 DOI: 10.1016/j.virol.2016.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 02/08/2023]
Abstract
There are ~350 million chronic carriers of hepatitis B (HBV). While a prophylactic vaccine and drug regimens to suppress viremia are available, chronic HBV infection is rarely cured. HBV's limited host tropism leads to a scarcity of susceptible small animal models and is a hurdle to developing curative therapies. Mice that support engraftment with human hepatoctyes have traditionally been generated through crosses of murine liver injury models to immunodeficient backgrounds. Here, we describe the disruption of fumarylacetoacetate hydrolase directly in the NOD Rag1-/- IL2RγNULL (NRG) background using zinc finger nucleases. The resultant human liver chimeric mice sustain persistent HBV viremia for >90 days. When treated with standard of care therapy, HBV DNA levels decrease below detection but rebound when drug suppression is released, mimicking treatment response observed in patients. Our study highlights the utility of directed gene targeting approaches in zygotes to create new humanized mouse models for human diseases.
Collapse
Affiliation(s)
- Benjamin Y Winer
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Tiffany Huang
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Benjamin E Low
- Department of Technology Evaluation and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609-1500 USA
| | - Cindy Avery
- Department of Technology Evaluation and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609-1500 USA
| | - Mihai-Alexandru Pais
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Gabriela Hrebikova
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Evelyn Siu
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Luis Chiriboga
- Department of Pathology, New York University Medical Center, New York, NY 10016, USA
| | - Michael V Wiles
- Department of Technology Evaluation and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609-1500 USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA.
| |
Collapse
|
126
|
Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, Shultz LD. Humanized Mouse Models of Clinical Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:187-215. [PMID: 27959627 DOI: 10.1146/annurev-pathol-052016-100332] [Citation(s) in RCA: 399] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunodeficient mice engrafted with functional human cells and tissues, that is, humanized mice, have become increasingly important as small, preclinical animal models for the study of human diseases. Since the description of immunodeficient mice bearing mutations in the IL2 receptor common gamma chain (IL2rgnull) in the early 2000s, investigators have been able to engraft murine recipients with human hematopoietic stem cells that develop into functional human immune systems. These mice can also be engrafted with human tissues such as islets, liver, skin, and most solid and hematologic cancers. Humanized mice are permitting significant progress in studies of human infectious disease, cancer, regenerative medicine, graft-versus-host disease, allergies, and immunity. Ultimately, use of humanized mice may lead to the implementation of truly personalized medicine in the clinic. This review discusses recent progress in the development and use of humanized mice and highlights their utility for the study of human diseases.
Collapse
Affiliation(s)
- Nicole C Walsh
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Sonal Jangalwe
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Ken-Edwin Aryee
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Michael A Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | | |
Collapse
|
127
|
Vercauteren K, Brown RJP, Mesalam AA, Doerrbecker J, Bhuju S, Geffers R, Van Den Eede N, McClure CP, Troise F, Verhoye L, Baumert T, Farhoudi A, Cortese R, Ball JK, Leroux-Roels G, Pietschmann T, Nicosia A, Meuleman P. Targeting a host-cell entry factor barricades antiviral-resistant HCV variants from on-therapy breakthrough in human-liver mice. Gut 2016; 65:2029-2034. [PMID: 26306759 DOI: 10.1136/gutjnl-2014-309045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Direct-acting antivirals (DAAs) inhibit hepatitis C virus (HCV) infection by targeting viral proteins that play essential roles in the replication process. However, selection of resistance-associated variants (RAVs) during DAA therapy has been a cause of therapeutic failure. In this study, we wished to address whether such RAVs could be controlled by the co-administration of host-targeting entry inhibitors that prevent intrahepatic viral spread. DESIGN We investigated the effect of adding an entry inhibitor (the anti-scavenger receptor class B type I mAb1671) to a DAA monotherapy (the protease inhibitor ciluprevir) in human-liver mice chronically infected with HCV of genotype 1b. Clinically relevant non-laboratory strains were used to achieve viraemia consisting of a cloud of related viral variants (quasispecies) and the emergence of RAVs was monitored at high resolution using next-generation sequencing. RESULTS HCV-infected human-liver mice receiving DAA monotherapy rapidly experienced on-therapy viral breakthrough. Deep sequencing of the HCV protease domain confirmed the manifestation of drug-resistant mutants upon viral rebound. In contrast, none of the mice treated with a combination of the DAA and the entry inhibitor experienced on-therapy viral breakthrough, despite detection of RAV emergence in some animals. CONCLUSIONS This study provides preclinical in vivo evidence that addition of an entry inhibitor to an anti-HCV DAA regimen restricts the breakthrough of DAA-resistant viruses. Our approach is an excellent strategy to prevent therapeutic failure caused by on-therapy rebound of DAA-RAVs. Inclusion of an entry inhibitor to the newest DAA combination therapies may further increase response rates, especially in difficult-to-treat patient populations.
Collapse
Affiliation(s)
- Koen Vercauteren
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| | - Richard J P Brown
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Ahmed Atef Mesalam
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| | - Juliane Doerrbecker
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Sabin Bhuju
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Naomi Van Den Eede
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| | - C Patrick McClure
- School of Life Sciences and the NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | | | - Lieven Verhoye
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| | - Thomas Baumert
- Institut National de la Santé et de la Recherche Médicale, U1110, Strasbourg, France.,Université de Strasbourg, Strasbourg et Pole Hépato-digestif, Hopitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ali Farhoudi
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| | | | - Jonathan K Ball
- School of Life Sciences and the NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Geert Leroux-Roels
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
| | - Alfredo Nicosia
- CEINGE, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Philip Meuleman
- Department Clinical Chemistry, Microbiology and Immunology, Center for Vaccinology, Ghent University, Ghent, Belgium
| |
Collapse
|
128
|
Bankwitz D, Pietschmann T. Hepatitis C virus plays hide and seek with neutralizing antibodies. Hepatology 2016; 64:1840-1842. [PMID: 27515101 DOI: 10.1002/hep.28760] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Dorothea Bankwitz
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany.,German Centre for Infection Research, Hannover-Braunschweig, Germany
| |
Collapse
|
129
|
Altered Glycosylation Patterns Increase Immunogenicity of a Subunit Hepatitis C Virus Vaccine, Inducing Neutralizing Antibodies Which Confer Protection in Mice. J Virol 2016; 90:10486-10498. [PMID: 27630242 DOI: 10.1128/jvi.01462-16] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/06/2016] [Indexed: 12/18/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a global health problem for which no vaccine is available. HCV has a highly heterogeneous RNA genome and can be classified into seven genotypes. Due to the high genetic and resultant antigenic variation among the genotypes, inducing antibodies capable of neutralizing most of the HCV genotypes by experimental vaccination has been challenging. Previous efforts focused on priming humoral immune responses with recombinant HCV envelope E2 protein produced in mammalian cells. Here, we report that a soluble form of HCV E2 (sE2) produced in insect cells possesses different glycosylation patterns and is more immunogenic, as evidenced by the induction of higher titers of broadly neutralizing antibodies (bNAbs) against cell culture-derived HCV (HCVcc) harboring structural proteins from a diverse array of HCV genotypes. We affirm that continuous and discontinuous epitopes of well-characterized bNAbs are conserved, suggesting that sE2 produced in insect cells is properly folded. In a genetically humanized mouse model, active immunization with sE2 efficiently protected against challenge with a heterologous HCV genotype. These data not only demonstrate that sE2 is a promising HCV vaccine candidate, but also highlight the importance of glycosylation patterns in developing subunit viral vaccines. IMPORTANCE A prophylactic vaccine with high efficacy and low cost is urgently needed for global control of HCV infection. Induction of broadly neutralizing antibodies against most HCV genotypes has been challenging due to the antigenic diversity of the HCV genome. Here, we refined a high-yield subunit HCV vaccine that elicited broadly neutralizing antibody responses in preclinical trials. We found that soluble HCV E2 protein (sE2) produced in insect cells is distinctly glycosylated and is more immunogenic than sE2 produced in mammalian cells, suggesting that glycosylation patterns should be taken into consideration in efforts to generate antibody-based recombinant vaccines against HCV. We further showed that sE2 vaccination confers protection against HCV infection in a genetically humanized mouse model. Thus, our work identified a promising broadly protective HCV vaccine candidate that should be considered for further preclinical and clinical development.
Collapse
|
130
|
Freedman H, Logan MR, Law JLM, Houghton M. Structure and Function of the Hepatitis C Virus Envelope Glycoproteins E1 and E2: Antiviral and Vaccine Targets. ACS Infect Dis 2016; 2:749-762. [PMID: 27933781 DOI: 10.1021/acsinfecdis.6b00110] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The hepatitis C virus (HCV) envelope glycoproteins E1 and E2 are critical in viral attachment and cell fusion, and studies of these proteins may provide valuable insights into their potential uses in vaccines and antiviral strategies. Progress has included elucidating the crystal structures of portions of their ectodomains, as well as many other studies of hypervariable regions, stem regions, glycosylation sites, and the participation of E1/E2 in viral fusion with the endosomal membrane. The available structural data have shed light on the binding sites of cross-neutralizing antibodies. A large amount of information has been discovered concerning heterodimerization, including the roles of transmembrane domains, disulfide bonding, and heptad repeat regions. The possible organization of higher order oligomers within the HCV virion has also been evaluated on the basis of experimental data. In this review, E1/E2 structure and function is discussed, and some important issues requiring further study are highlighted.
Collapse
Affiliation(s)
- Holly Freedman
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael R. Logan
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - John Lok Man Law
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Houghton
- Li Ka Shing Institute of Virology, Department of Medical Microbiology
and Immunology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
131
|
Swann RE, Mandalou P, Robinson MW, Ow MM, Foung SKH, McLauchlan J, Patel AH, Cramp ME. Anti-envelope antibody responses in individuals at high risk of hepatitis C virus who resist infection. J Viral Hepat 2016; 23:873-880. [PMID: 27405885 PMCID: PMC5244678 DOI: 10.1111/jvh.12568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 02/05/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022]
Abstract
Injection drug users uninfected by hepatitis C virus (HCV) despite likely repeated exposure through high-risk behaviour are well documented. Factors preventing infection in these individuals are incompletely understood. Here, we looked for anti-HCV-envelope antibody responses in a cohort of repeatedly exposed but uninfected subjects. Forty-two hepatitis C diagnostic antibody- and RNA-negative injection drug users at high risk of exposure were studied and findings compared to healthy controls and cases with chronic HCV infection. Purified IgGs from sera were tested by ELISA for binding to genotype 1a and 3a envelope glycoproteins E1E2 with further testing for IgG and IgM reactivity against soluble E2. Virus-neutralizing activity was assessed using an HCV pseudoparticle system. Uninfected subjects demonstrated significantly greater IgG and IgM reactivities to envelope glycoproteins than healthy controls with IgG from 6 individuals additionally showing significant neutralization. This study is the first to describe humoral immunological responses targeting the HCV envelope, important for viral neutralization, in exposed uninfected individuals. A subset of these cases also had evidence of viral neutralization via anti-envelope antibodies. In addition to confirming viral exposure, the presence of specific anti-envelope antibodies may be a factor that helps these individuals resist HCV infection.
Collapse
Affiliation(s)
- R. E. Swann
- MRC – University of Glasgow Centre for Virus ResearchUniversity of GlasgowGlasgowUK
| | - P. Mandalou
- Hepatology Research GroupPlymouth University Peninsula Schools of Medicine and DentistryPlymouthUK,South West Liver UnitDerriford HospitalPlymouthUK
| | - M. W. Robinson
- MRC – University of Glasgow Centre for Virus ResearchUniversity of GlasgowGlasgowUK,School of Biochemistry and ImmunologyTrinity College DublinDublinIreland
| | - M. M. Ow
- Hepatology Research GroupPlymouth University Peninsula Schools of Medicine and DentistryPlymouthUK,South West Liver UnitDerriford HospitalPlymouthUK
| | - S. K. H. Foung
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - J. McLauchlan
- MRC – University of Glasgow Centre for Virus ResearchUniversity of GlasgowGlasgowUK
| | - A. H. Patel
- MRC – University of Glasgow Centre for Virus ResearchUniversity of GlasgowGlasgowUK
| | - M. E. Cramp
- Hepatology Research GroupPlymouth University Peninsula Schools of Medicine and DentistryPlymouthUK,South West Liver UnitDerriford HospitalPlymouthUK
| |
Collapse
|
132
|
Merat SJ, Molenkamp R, Wagner K, Koekkoek SM, van de Berg D, Yasuda E, Böhne M, Claassen YB, Grady BP, Prins M, Bakker AQ, de Jong MD, Spits H, Schinkel J, Beaumont T. Hepatitis C virus Broadly Neutralizing Monoclonal Antibodies Isolated 25 Years after Spontaneous Clearance. PLoS One 2016; 11:e0165047. [PMID: 27776169 PMCID: PMC5077102 DOI: 10.1371/journal.pone.0165047] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/24/2016] [Indexed: 01/18/2023] Open
Abstract
Hepatitis C virus (HCV) is world-wide a major cause of liver related morbidity and mortality. No vaccine is available to prevent HCV infection. To design an effective vaccine, understanding immunity against HCV is necessary. The memory B cell repertoire was characterized from an intravenous drug user who spontaneously cleared HCV infection 25 years ago. CD27+IgG+ memory B cells were immortalized using BCL6 and Bcl-xL. These immortalized B cells were used to study antibody-mediated immunity against the HCV E1E2 glycoproteins. Five E1E2 broadly reactive antibodies were isolated: 3 antibodies showed potent neutralization of genotype 1 to 4 using HCV pseudotyped particles, whereas the other 2 antibodies neutralized genotype 1, 2 and 3 or 1 and 2 only. All antibodies recognized non-linear epitopes on E2. Finally, except for antibody AT12-011, which recognized an epitope consisting of antigenic domain C /AR2 and AR5, all other four antibodies recognized epitope II and domain B. These data show that a subject, who spontaneously cleared HCV infection 25 years ago, still has circulating memory B cells that are able to secrete broadly neutralizing antibodies. Presence of such memory B cells strengthens the argument for undertaking the development of an HCV vaccine.
Collapse
Affiliation(s)
| | - Richard Molenkamp
- Department of Medical Microbiology, Section of Clinical Virology, Academic Medical Center, Amsterdam, the Netherlands
| | - Koen Wagner
- AIMM Therapeutics, Amsterdam, the Netherlands
| | - Sylvie M. Koekkoek
- Department of Medical Microbiology, Section of Clinical Virology, Academic Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | - Bart P. Grady
- Department of Infectious Diseases Research and Prevention, Cluster of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | - Maria Prins
- Department of Infectious Diseases Research and Prevention, Cluster of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Department of infectious diseases, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Menno D. de Jong
- Department of Medical Microbiology, Section of Clinical Virology, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Janke Schinkel
- Department of Medical Microbiology, Section of Clinical Virology, Academic Medical Center, Amsterdam, the Netherlands
| | - Tim Beaumont
- AIMM Therapeutics, Amsterdam, the Netherlands
- * E-mail:
| |
Collapse
|
133
|
Bukh J. The history of hepatitis C virus (HCV): Basic research reveals unique features in phylogeny, evolution and the viral life cycle with new perspectives for epidemic control. J Hepatol 2016; 65:S2-S21. [PMID: 27641985 DOI: 10.1016/j.jhep.2016.07.035] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 12/11/2022]
Abstract
The discovery of hepatitis C virus (HCV) in 1989 permitted basic research to unravel critical components of a complex life cycle for this important human pathogen. HCV is a highly divergent group of viruses classified in 7 major genotypes and a great number of subtypes, and circulating in infected individuals as a continuously evolving quasispecies destined to escape host immune responses and applied antivirals. Despite the inability to culture patient viruses directly in the laboratory, efforts to define the infectious genome of HCV resulted in development of experimental recombinant in vivo and in vitro systems, including replicons and infectious cultures in human hepatoma cell lines. And HCV has become a model virus defining new paradigms in virology, immunology and biology. For example, HCV research discovered that a virus could be completely dependent on microRNA for its replication since microRNA-122 is critical for the HCV life cycle. A number of other host molecules critical for HCV entry and replication have been identified. Thus, basic HCV research revealed important molecules for development of host targeting agents (HTA). The identification and characterization of HCV encoded proteins and their functional units contributed to the development of highly effective direct acting antivirals (DAA) against the NS3 protease, NS5A and the NS5B polymerase. In combination, these inhibitors have since 2014 permitted interferon-free therapy with cure rates above 90% among patients with chronic HCV infection; however, viral resistance represents a challenge. Worldwide control of HCV will most likely require the development of a prophylactic vaccine, and numerous candidates have been pursued. Research characterizing features critical for antibody-based virus neutralization and T cell based virus elimination from infected cells is essential for this effort. If the world community promotes an ambitious approach by applying current DAA broadly, continues to develop alternative viral- and host- targeted antivirals to combat resistant variants, and invests in the development of a vaccine, it would be possible to eradicate HCV. This would prevent about 500 thousand deaths annually. However, given the nature of HCV, the millions of new infections annually, a high chronicity rate, and with over 150 million individuals with chronic infection (which are frequently unidentified), this effort remains a major challenge for basic researchers, clinicians and communities.
Collapse
Affiliation(s)
- Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
134
|
Moradpour D, Grakoui A, Manns MP. Future landscape of hepatitis C research - Basic, translational and clinical perspectives. J Hepatol 2016; 65:S143-S155. [PMID: 27641984 DOI: 10.1016/j.jhep.2016.07.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 12/14/2022]
Abstract
With the latest all-oral interferon- and ribavirin-free regimens based on direct acting antivirals against the hepatitis C virus (HCV), sustained virological response rates of >90% are achieved, which is equivalent to cure. This has become possible for all genotypes and all subgroups of patients, including many of the most difficult-to-treat populations so far. Since a prophylactic HCV vaccine is not yet available, control of HCV infection will for the time being have to rely on the use of effective and safe antiviral treatments as well as their accessibility and affordability. Different approaches may apply to different parts of the world, eradication of HCV representing a major long-term goal. Whether hepatitis C becomes the first chronic viral infection to be eradicated without a prophylactic vaccine remains to be shown. Here, we briefly summarize advances in the molecular virology of hepatitis C, highlight lessons of biological relevance that were learned through the study of HCV, and its translational and clinical implications. We have also listed selected unsolved challenges, emphasizing that HCV is a unique model and that advances in this direction may yield knowledge of broad biological significance, novel technologies and insights into related important human pathogens.
Collapse
Affiliation(s)
- Darius Moradpour
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Switzerland.
| | - Arash Grakoui
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine and Yerkes National Primate Research Center, Emory Vaccine Center, Atlanta, GA, USA.
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany.
| |
Collapse
|
135
|
Dustin LB, Bartolini B, Capobianchi MR, Pistello M. Hepatitis C virus: life cycle in cells, infection and host response, and analysis of molecular markers influencing the outcome of infection and response to therapy. Clin Microbiol Infect 2016; 22:826-832. [PMID: 27592089 PMCID: PMC5627509 DOI: 10.1016/j.cmi.2016.08.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 08/16/2016] [Accepted: 08/25/2016] [Indexed: 12/13/2022]
Abstract
Hepatitis C virus (HCV) is a major global health burden accounting for around 170 million chronic infections worldwide. Since its discovery, which dates back to about 30 years ago, many details of the viral genome organization and the astonishing genetic diversity have been unveiled but, owing to the difficulty of culturing HCV in vitro and obtaining fully susceptible yet immunocompetent in vivo models, we are still a long way from the full comprehension of viral life cycle, host cell pathways facilitating or counteracting infection, pathogenetic mechanisms in vivo, and host defences. Here, we illustrate the viral life cycle into cells, describe the interplay between immune and genetic host factors shaping the course of infection, and provide details of the molecular approaches currently used to genotype, monitor replication in vivo, and study the emergence of drug-resistant viral variants.
Collapse
Affiliation(s)
- L B Dustin
- Kennedy Institute for Rheumatology and Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - B Bartolini
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - M R Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - M Pistello
- Virology Unit, Pisa University Hospital, and Virology Section and Retrovirus Centre, Department of Translational Research, University of Pisa, Pisa, Italy.
| |
Collapse
|
136
|
Viral evasion and challenges of hepatitis C virus vaccine development. Curr Opin Virol 2016; 20:55-63. [PMID: 27657659 DOI: 10.1016/j.coviro.2016.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
Hepatitis C virus (HCV) is a major global disease burden, often leading to chronic liver diseases, cirrhosis, cancer, and death in those infected. Despite the recent approval of antiviral therapeutics, a preventative vaccine is recognized as the most effective means to control HCV globally, particularly in at-risk and developing country populations. Here we describe the efforts and challenges related to the development of an HCV vaccine, which after decades of research have not been successful. Viral sequence variability poses a major challenge, yet recent research has provided unprecedented views of the atomic structure of HCV epitopes and immune recognition by antibodies and T cell receptors. This, coupled with insights from deep sequencing, robust neutralization assays, and other technological advances, is spurring research toward rationally HCV designed vaccines that preferentially elicit responses toward conserved epitopes of interest that are associated with viral neutralization and clearance.
Collapse
|
137
|
Identification of a New Benzimidazole Derivative as an Antiviral against Hepatitis C Virus. J Virol 2016; 90:8422-34. [PMID: 27412600 DOI: 10.1128/jvi.00404-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/05/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Aminoquinolines and piperazines, linked or not, have been used successfully to treat malaria, and some molecules of this family also exhibit antiviral properties. Here we tested several derivatives of 4-aminoquinolines and piperazines for their activity against hepatitis C virus (HCV). We screened 11 molecules from three different families of compounds, and we identified anti-HCV activity in cell culture for six of them. Of these, we selected a compound (B5) that is currently ending clinical phase I evaluation for neurodegenerative diseases. In hepatoma cells, B5 inhibited HCV infection in a pangenotypic and dose-dependent manner, and its antiviral activity was confirmed in primary hepatocytes. B5 also inhibited infection by pseudoparticles expressing HCV envelope glycoproteins E1 and E2, and we demonstrated that it affects a postattachment stage of the entry step. Virus with resistance to B5 was selected by sequential passage in the presence of the drug, and reverse genetics experiments indicated that resistance was conferred mainly by a single mutation in the putative fusion peptide of E1 envelope glycoprotein (F291I). Furthermore, analyses of the effects of other closely related compounds on the B5-resistant mutant suggest that B5 shares a mode of action with other 4-aminoquinoline-based molecules. Finally, mice with humanized liver that were treated with B5 showed a delay in the kinetics of the viral infection. In conclusion, B5 is a novel interesting anti-HCV molecule that could be used to decipher the early steps of the HCV life cycle. IMPORTANCE In the last 4 years, HCV therapy has been profoundly improved with the approval of direct-acting antivirals in clinical practice. Nevertheless, the high costs of these drugs limit access to therapy in most countries. The present study reports the identification and characterization of a compound (B5) that inhibits HCV propagation in cell culture and is currently ending clinical phase I evaluation for neurodegenerative diseases. This molecule inhibits the HCV life cycle by blocking virus entry. Interestingly, after selection of drug-resistant virus, a resistance mutation in the putative fusion peptide of E1 envelope glycoprotein was identified, indicating that B5 could be used to further investigate the fusion mechanism. Furthermore, mice with humanized liver treated with B5 showed a delay in the kinetics of the viral infection. In conclusion, B5 is a novel interesting anti-HCV molecule that could be used to decipher the early steps of the HCV life cycle.
Collapse
|
138
|
Ernst W. Humanized mice in infectious diseases. Comp Immunol Microbiol Infect Dis 2016; 49:29-38. [PMID: 27865261 DOI: 10.1016/j.cimid.2016.08.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 08/12/2016] [Accepted: 08/12/2016] [Indexed: 02/06/2023]
Abstract
The pathogenesis of infectious agents with human tropism can only be properly studied in an in vivo model featuring human cells or tissue. Humanized mice represent a small animal model featuring human cells or tissue that can be infected by human-specific viruses, bacteria, and parasites and also providing a functional human immune system. This makes the analysis of a human immune response to infection possible and allows for preclinical testing of new vaccines and therapeutic agents. Results of various studies using humanized mice to investigate pathogens with human tropism are presented in this review. In addition, the limitations of humanized mice and methods to improve this valuable animal model are discussed.
Collapse
Affiliation(s)
- W Ernst
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Bavaria, Germany.
| |
Collapse
|
139
|
Converting monoclonal antibody-based immunotherapies from passive to active: bringing immune complexes into play. Emerg Microbes Infect 2016; 5:e92. [PMID: 27530750 PMCID: PMC5034104 DOI: 10.1038/emi.2016.97] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/13/2022]
Abstract
Monoclonal antibodies (mAbs), which currently constitute the main class of biotherapeutics, are now recognized as major medical tools that are increasingly being considered to fight severe viral infections. Indeed, the number of antiviral mAbs developed in recent years has grown exponentially. Although their direct effects on viral blunting have been studied in detail, their potential immunomodulatory actions have been overlooked until recently. The ability of antiviral mAbs to modulate antiviral immune responses in infected organisms has recently been revealed. More specifically, upon recognition of their cognate antigens, mAbs form immune complexes (ICs) that can be recognized by the Fc receptors expressed on different immune cells of infected individuals. This binding may be followed by the modulation of the host immune responses. Harnessing this immunomodulatory property may facilitate improvements in the therapeutic potential of antiviral mAbs. This review focuses on the role of ICs formed with different viral determinants and mAbs in the induction of antiviral immune responses in the context of both passive immunotherapies and vaccination strategies. Potential deleterious effects of ICs on the host immune response are also discussed.
Collapse
|
140
|
Hepatocarcinogenesis associated with hepatitis B, delta and C viruses. Curr Opin Virol 2016; 20:1-10. [PMID: 27504999 DOI: 10.1016/j.coviro.2016.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 12/13/2022]
Abstract
Globally, over half a billion people are persistently infected with hepatitis B (HBV) and/or hepatitis C viruses. Chronic HBV and HCV infection frequently lead to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Co-infections with hepatitis delta virus (HDV), a subviral satellite requiring HBV for its propagation, accelerates the progression of liver disease toward HCC. The mechanisms by which these viruses cause malignant transformation, culminating in HCC, remain incompletely understood, partially due to the lack of adequate experimental models for dissecting these complex disease processes in vivo.
Collapse
|
141
|
Billerbeck E, Mommersteeg MC, Shlomai A, Xiao JW, Andrus L, Bhatta A, Vercauteren K, Michailidis E, Dorner M, Krishnan A, Charlton MR, Chiriboga L, Rice CM, de Jong YP. Humanized mice efficiently engrafted with fetal hepatoblasts and syngeneic immune cells develop human monocytes and NK cells. J Hepatol 2016; 65:334-43. [PMID: 27151182 PMCID: PMC4955758 DOI: 10.1016/j.jhep.2016.04.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Human liver chimeric mice are useful models of human hepatitis virus infection, including hepatitis B and C virus infections. Independently, immunodeficient mice reconstituted with CD34(+) hematopoietic stem cells (HSC) derived from fetal liver reliably develop human T and B lymphocytes. Combining these systems has long been hampered by inefficient liver reconstitution of human fetal hepatoblasts. Our study aimed to enhance hepatoblast engraftment in order to create a mouse model with syngeneic human liver and immune cells. METHODS The effects of human oncostatin-M administration on fetal hepatoblast engraftment into immunodeficient fah(-/-) mice was tested. Mice were then transplanted with syngeneic human hepatoblasts and HSC after which human leukocyte chimerism and functionality were analyzed by flow cytometry, and mice were challenged with HBV. RESULTS Addition of human oncostatin-M enhanced human hepatoblast engraftment in immunodeficient fah(-/-) mice by 5-100 fold. In contrast to mice singly engrafted with HSC, which predominantly developed human T and B lymphocytes, mice co-transplanted with syngeneic hepatoblasts also contained physiological levels of human monocytes and natural killer cells. Upon infection with HBV, these mice displayed rapid and sustained viremia. CONCLUSIONS Our study provides a new mouse model with improved human fetal hepatoblast engraftment and an expanded human immune cell repertoire. With further improvements, this model may become useful for studying human immunity against viral hepatitis. LAY SUMMARY Important human pathogens such as hepatitis B virus, hepatitis C virus and human immunodeficiency virus only infect human cells which complicates the development of mouse models for the study of these pathogens. One way to make mice permissive for human pathogens is the transplantation of human cells into immune-compromised mice. For instance, the transplantation of human liver cells will allow the infection of these so-called "liver chimeric mice" with hepatitis B virus and hepatitis C virus. The co-transplantation of human immune cells into liver chimeric mice will further allow the study of human immune responses to hepatitis B virus or hepatitis C virus. However, for immunological studies it will be crucial that the transplanted human liver and immune cells are derived from the same human donor. In our study we describe the efficient engraftment of human fetal liver cells and immune cells derived from the same donor into mice. We show that liver co-engraftment resulted in an expanded human immune cell repertoire, including monocytes and natural killer cells in the liver. We further demonstrate that these mice could be infected with hepatitis B virus, which lead to an expansion of natural killer cells. In conclusion we have developed a new mouse model that could be useful to study human immune responses to human liver pathogens.
Collapse
Affiliation(s)
- Eva Billerbeck
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Michiel C. Mommersteeg
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Amir Shlomai
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Jing W. Xiao
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Linda Andrus
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Ankit Bhatta
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Koen Vercauteren
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Eleftherios Michailidis
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Marcus Dorner
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Anuradha Krishnan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Michael R. Charlton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Luis Chiriboga
- Department of Pathology, New York University Medical Center, New York, NY, USA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA,Corresponding authors. Address: The Rockefeller University, Laboratory of Virology and Infectious Disease, 1230 York Avenue, Box 64, New York, NY 10065, USA. Tel.: +1 212 327 7009; fax: +1 212 327 7048. (C.M. Rice), (Y.P. de Jong)
| | - Ype P. de Jong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA,Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA,Corresponding authors. Address: The Rockefeller University, Laboratory of Virology and Infectious Disease, 1230 York Avenue, Box 64, New York, NY 10065, USA. Tel.: +1 212 327 7009; fax: +1 212 327 7048. (C.M. Rice), (Y.P. de Jong)
| |
Collapse
|
142
|
Abstract
Hepatitis A virus (HAV), hepatitis B virus (HBV) and hepatitis C virus (HCV) are responsible for most cases of viral hepatitis. Infection by each type of virus results in a different typical natural disease course and clinical outcome that are determined by virological and immunological factors. HCV tends to establish a chronic persistent infection, whereas HAV does not. HBV is effectively controlled in adults, although it persists for a lifetime after neonatal infection. In this Review, we discuss the similarities and differences in immune responses to and immunopathogenesis of HAV, HBV and HCV infections, which may explain the distinct courses and outcomes of each hepatitis virus infection.
Collapse
|
143
|
Affiliation(s)
- Zachary T. Freeman
- Division of Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Andrea L. Cox
- Division of Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
144
|
Large scale production of a mammalian cell derived quadrivalent hepatitis C virus like particle vaccine. J Virol Methods 2016; 236:87-92. [PMID: 27373602 DOI: 10.1016/j.jviromet.2016.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022]
Abstract
A method for the large-scale production of a quadrivalent mammalian cell derived hepatitis C virus-like particles (HCV VLPs) is described. The HCV core E1 and E2 coding sequences of genotype 1a, 1b, 2a or 3a were co-expressed in Huh7 cell factories using a recombinant adenoviral expression system. The structural proteins self-assembled into VLPs that were purified from Huh7 cell lysates by iodixanol ultracentrifugation and Stirred cell ultrafiltration. Electron microscopy, revealed VLPs of the different genotypes that are morphologically similar. Our results show that it is possible to produce large quantities of individual HCV genotype VLPs with relative ease thus making this approach an alternative for the manufacture of a quadrivalent mammalian cell derived HCV VLP vaccine.
Collapse
|
145
|
Bivalent vaccine platform based on Japanese encephalitis virus (JEV) elicits neutralizing antibodies against JEV and hepatitis C virus. Sci Rep 2016; 6:28688. [PMID: 27345289 PMCID: PMC4922013 DOI: 10.1038/srep28688] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/08/2016] [Indexed: 12/20/2022] Open
Abstract
Directly acting antivirals recently have become available for the treatment of hepatitis C virus (HCV) infection, but there is no prophylactic vaccine for HCV. In the present study, we took advantage of the properties of Japanese encephalitis virus (JEV) to develop antigens for use in a HCV vaccine. Notably, the surface-exposed JEV envelope protein is tolerant of inserted foreign epitopes, permitting display of novel antigens. We identified 3 positions that permitted insertion of the HCV E2 neutralization epitope recognized by HCV1 antibody. JEV subviral particles (SVP) containing HCV-neutralization epitope (SVP-E2) were purified from culture supernatant by gel chromatography. Sera from mice immunized with SVP-E2 inhibited infection by JEV and by trans-complemented HCV particles (HCVtcp) derived from multi-genotypic viruses, whereas sera from mice immunized with synthetic E2 peptides did not show any neutralizing activity. Furthermore, sera from mice immunized with SVP-E2 neutralized HCVtcp with N415K escape mutation in E2. As with the SVP-E2 epitope-displaying particles, JEV SVPs with HCV E1 epitope also elicited neutralizing antibodies against HCV. Thus, this novel platform harboring foreign epitopes on the surface of the particle may facilitate the development of a bivalent vaccine against JEV and other pathogens.
Collapse
|
146
|
Felmlee DJ, Coilly A, Chung RT, Samuel D, Baumert TF. New perspectives for preventing hepatitis C virus liver graft infection. THE LANCET. INFECTIOUS DISEASES 2016; 16:735-745. [PMID: 27301929 PMCID: PMC4911897 DOI: 10.1016/s1473-3099(16)00120-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 01/29/2016] [Accepted: 02/15/2016] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) infection is a leading cause of end-stage liver disease that necessitates liver transplantation. The incidence of virus-induced cirrhosis and hepatocellular carcinoma continues to increase, making liver transplantation increasingly common. Infection of the engrafted liver is universal and accelerates progression to advanced liver disease, with 20-30% of patients having cirrhosis within 5 years of transplantation. Treatments of chronic HCV infection have improved dramatically, albeit with remaining challenges of failure and access, and therapeutic options to prevent graft infection during liver transplantation are emerging. Developments in directed use of new direct-acting antiviral agents (DAAs) to eliminate circulating HCV before or after transplantation in the past 5 years provide renewed hope for prevention and treatment of liver graft infection. Identification of the ideal regimen and use of DAAs reveals new ways to treat this specific population of patients. Complementing DAAs, viral entry inhibitors have been shown to prevent liver graft infection in animal models and delay graft infection in clinical trials, which shows their potential for use concomitant to transplantation. We review the challenges and pathology associated with HCV liver graft infection, highlight current and future strategies of DAA treatment timing, and discuss the potential role of entry inhibitors that might be used synergistically with DAAs to prevent or treat graft infection.
Collapse
Affiliation(s)
- Daniel J Felmlee
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Hepatology Research Group, Peninsula School of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Audrey Coilly
- AP-HP Hôpital Paul Brousse, Centre Hépato-Biliaire, Villejuif, France; University Paris-Sud, UMR-S 1193, Villejuif, France; Inserm Unit 1193, Villejuif F-94800, France
| | - Raymond T Chung
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Didier Samuel
- AP-HP Hôpital Paul Brousse, Centre Hépato-Biliaire, Villejuif, France; University Paris-Sud, UMR-S 1193, Villejuif, France; Inserm Unit 1193, Villejuif F-94800, France.
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
147
|
Earnest-Silveira L, Chua B, Chin R, Christiansen D, Johnson D, Herrmann S, Ralph SA, Vercauteren K, Mesalam A, Meuleman P, Das S, Boo I, Drummer H, Bock CT, Gowans EJ, Jackson DC, Torresi J. Characterization of a hepatitis C virus-like particle vaccine produced in a human hepatocyte-derived cell line. J Gen Virol 2016; 97:1865-1876. [PMID: 27147296 DOI: 10.1099/jgv.0.000493] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
An effective immune response against hepatitis C virus (HCV) requires the early development of multi-specific class 1 CD8+ and class II CD4+ T-cells together with broad neutralizing antibody responses. We have produced mammalian-cell-derived HCV virus-like particles (VLPs) incorporating core, E1 and E2 of HCV genotype 1a to produce such immune responses. Here we describe the biochemical and morphological characterization of the HCV VLPs and study HCV core-specific T-cell responses to the particles. The E1 and E2 glycoproteins in HCV VLPs formed non-covalent heterodimers and together with core protein assembled into VLPs with a buoyant density of 1.22 to 1.28 g cm-3. The HCV VLPs could be immunoprecipited with anti-ApoE and anti-ApoC. On electron microscopy, the VLPs had a heterogeneous morphology and ranged in size from 40 to 80 nm. The HCV VLPs demonstrated dose-dependent binding to murine-derived dendritic cells and the entry of HCV VLPs into Huh7 cells was blocked by anti-CD81 antibody. Vaccination of BALB/c mice with HCV VLPs purified from iodixanol gradients resulted in the production of neutralizing antibody responses while vaccination of humanized MHC class I transgenic mice resulted in the prodution of HCV core-specific CD8+ T-cell responses. Furthermore, IgG purified from the sera of patients chronically infected with HCV genotypes 1a and 3a blocked the binding and entry of the HCV VLPs into Huh7 cells. These results show that our mammalian-cell-derived HCV VLPs induce humoral and HCV-specific CD8+ T-cell responses and will have important implications for the development of a preventative vaccine for HCV.
Collapse
Affiliation(s)
- L Earnest-Silveira
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - B Chua
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - R Chin
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - D Christiansen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Surgery, Austin Hospital, University of Melbourne, Australia
| | - D Johnson
- Department of Infectious Diseases, Austin Hospital, Heidelberg, Victoria 3084, Australia
| | - S Herrmann
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Australia
| | - S A Ralph
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Australia
| | - K Vercauteren
- Center for Vaccinology, Ghent University and Hospital, De Pintelaan 185 9000, Ghent, Belgium
| | - A Mesalam
- Center for Vaccinology, Ghent University and Hospital, De Pintelaan 185 9000, Ghent, Belgium
| | - P Meuleman
- Center for Vaccinology, Ghent University and Hospital, De Pintelaan 185 9000, Ghent, Belgium
| | - S Das
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - I Boo
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia
| | - H Drummer
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia.,Centre for Biomedical Research, Burnet Institute, Melbourne, Australia.,Department of Microbiology, Monash University, Clayton, Australia
| | - C-T Bock
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - E J Gowans
- The Basil Hetzel Institute and Queen Elizabeth Hospital, University of Adelaide, Australia
| | - D C Jackson
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
148
|
Broad Anti-Hepatitis C Virus (HCV) Antibody Responses Are Associated with Improved Clinical Disease Parameters in Chronic HCV Infection. J Virol 2016; 90:4530-4543. [PMID: 26912610 PMCID: PMC4836347 DOI: 10.1128/jvi.02669-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/15/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED During hepatitis C virus (HCV) infection, broadly neutralizing antibody (bNAb) responses targeting E1E2 envelope glycoproteins are generated in many individuals. It is unclear if these antibodies play a protective or a pathogenic role during chronic infection. In this study, we investigated whether bNAb responses in individuals with chronic infection were associated with differences in clinical presentation. Patient-derived purified serum IgG was used to assess the breadth of HCV E1E2 binding and the neutralization activity of HCV pseudoparticles. The binding and neutralization activity results for two panels bearing viral envelope proteins representing either an intergenotype or an intragenotype 1 group were compared. We found that the HCV load was negatively associated with strong cross-genotypic E1E2 binding (P= 0.03). Overall, we observed only a modest correlation between total E1E2 binding and neutralization ability. The breadth of intergenotype neutralization did not correlate with any clinical parameters; however, analysis of individuals with genotype 1 (gt1) HCV infection (n= 20), using an intragenotype pseudoparticle panel, found a strong association between neutralization breadth and reduced liver fibrosis (P= 0.006). A broad bNAb response in our cohort with chronic infection was associated with a single nucleotide polymorphism (SNP) in theHLA-DQB1 gene (P= 0.038), as previously reported in a cohort with acute disease. Furthermore, the bNAbs in these individuals targeted more than one region of E2-neutralizing epitopes, as assessed through cross-competition of patient bNAbs with well-characterized E2 antibodies. We conclude that the bNAb responses in patients with chronic gt1 infection are associated with lower rates of fibrosis and host genetics may play a role in the ability to raise such responses. IMPORTANCE Globally, there are 130 million to 150 million people with chronic HCV infection. Typically, the disease is progressive and is a major cause of severe liver cirrhosis and hepatocellular carcinoma. While it is known that neutralizing antibodies have a role in spontaneous clearance during acute infection, little is known about their role in chronic infection. In the present work, we investigated the antibody response in a cohort of chronically infected individuals and found that a broadly neutralizing antibody response is protective and is associated with reduced levels of liver fibrosis and cirrhosis. We also found an association between SNPs in class II HLA genes and the presence of a broadly neutralizing response, indicating that antigen presentation may be important for the production of HCV-neutralizing antibodies.
Collapse
|
149
|
Pelegrin M, Naranjo-Gomez M, Piechaczyk M. Antiviral Monoclonal Antibodies: Can They Be More Than Simple Neutralizing Agents? Trends Microbiol 2016; 23:653-665. [PMID: 26433697 PMCID: PMC7127033 DOI: 10.1016/j.tim.2015.07.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/06/2015] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) are increasingly being considered as agents to fight severe viral diseases. So far, they have essentially been selected and used on the basis of their virus-neutralizing activity and/or cell-killing activity to blunt viral propagation via direct mechanisms. There is, however, accumulating evidence that they can also induce long-lasting protective antiviral immunity by recruiting the endogenous immune system of infected individuals during the period of immunotherapy. Exploiting this property may revolutionize antiviral mAb-based immunotherapies, with benefits for both patients and healthcare systems. Antiviral monoclonal antibodies (mAbs) are promising, high-added-value biotherapeutics. During recent years, the number of antiviral mAbs developed against both acute and chronic viruses has grown exponentially, some of them being currently tested in clinical trials. Antiviral mAbs can be used to blunt viral propagation through direct effects. They can also engage the host's immune system, leading to the induction of long-lasting protective vaccine-like effects. The assessment of mechanisms at play in the induction of vaccine-like effects by antiviral mAbs will help in improving antiviral treatments. Exploiting this effect will translate into therapeutic benefit for patients. The benefit will also help healthcare systems through the reduction of treatment costs.
Collapse
Affiliation(s)
- Mireia Pelegrin
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France.
| | - Mar Naranjo-Gomez
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Marc Piechaczyk
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| |
Collapse
|
150
|
Winer BY, Ding Q, Gaska JM, Ploss A. In vivo models of hepatitis B and C virus infection. FEBS Lett 2016; 590:1987-99. [PMID: 27009462 DOI: 10.1002/1873-3468.12157] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 12/17/2022]
Abstract
Globally, more than 500 million individuals are chronically infected with hepatitis B (HBV), delta (HDV), and/or C (HCV) viruses, which can result in severe liver disease. Mechanistic studies of viral persistence and pathogenesis have been hampered by the scarcity of animal models. The limited species and cellular host range of HBV, HDV, and HCV, which robustly infect only humans and chimpanzees, have posed challenges for creating such animal models. In this review, we will discuss the barriers to interspecies transmission and the progress that has been made in our understanding of the HBV, HDV, and HCV life cycles. Additionally, we will highlight a variety of approaches that overcome these barriers and thus facilitate in vivo studies of these hepatotropic viruses.
Collapse
Affiliation(s)
| | - Qiang Ding
- Department of Molecular Biology, Princeton University, NJ, USA
| | - Jenna M Gaska
- Department of Molecular Biology, Princeton University, NJ, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, NJ, USA
| |
Collapse
|