51
|
Verstrepen BE, Boonstra A, Koopman G. Immune mechanisms of vaccine induced protection against chronic hepatitis C virus infection in chimpanzees. World J Hepatol 2015; 7:53-69. [PMID: 25624997 PMCID: PMC4295194 DOI: 10.4254/wjh.v7.i1.53] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/22/2014] [Accepted: 11/07/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infection is characterized by a high propensity for development of life-long viral persistence. An estimated 170 million people suffer from chronic hepatitis caused by HCV. Currently, there is no approved prophylactic HCV vaccine available. With the near disappearance of the most relevant animal model for HCV, the chimpanzee, we review the progression that has been made regarding prophylactic vaccine development against HCV. We describe the results of the individual vaccine evaluation experiments in chimpanzees, in relation to what has been observed in humans. The results of the different studies indicate that partial protection against infection can be achieved, but a clear correlate of protection has thus far not yet been defined.
Collapse
Affiliation(s)
- Babs E Verstrepen
- Babs E Verstrepen, Gerrit Koopman, Department of Virology, Biomedical Primate Research Centre, 2280GH Rijswijk, The Netherlands
| | - André Boonstra
- Babs E Verstrepen, Gerrit Koopman, Department of Virology, Biomedical Primate Research Centre, 2280GH Rijswijk, The Netherlands
| | - Gerrit Koopman
- Babs E Verstrepen, Gerrit Koopman, Department of Virology, Biomedical Primate Research Centre, 2280GH Rijswijk, The Netherlands
| |
Collapse
|
52
|
Vercauteren K, Mesalam AA, Leroux-Roels G, Meuleman P. Impact of lipids and lipoproteins on hepatitis C virus infection and virus neutralization. World J Gastroenterol 2014; 20:15975-91. [PMID: 25473151 PMCID: PMC4239485 DOI: 10.3748/wjg.v20.i43.15975] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/09/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infections represent a major global health problem. End-stage liver disease caused by chronic HCV infection is a major indication for liver transplantation. However, after transplantation the engrafted liver inevitably becomes infected by the circulating virus. Direct acting antivirals are not yet approved for use in liver transplant patients, and limited efficacy and severe side effects hamper the use of pegylated interferon combined with ribavirin in a post-transplant setting. Therefore, alternative therapeutic options need to be explored. Viral entry represents an attractive target for such therapeutic intervention. Understanding the mechanisms of viral entry is essential to define the viral and cellular factors involved. The HCV life cycle is dependent of and associated with lipoprotein physiology and the presence of lipoproteins has been correlated with altered antiviral efficacy of entry inhibitors. In this review, we summarise the current knowledge on how lipoprotein physiology influences the HCV life cycle. We focus especially on the influence of lipoproteins on antibodies that target HCV envelope proteins or antibodies that target the cellular receptors of the virus. This information can be particularly relevant for the prevention of HCV re-infection after liver transplantation.
Collapse
|
53
|
Vercauteren K, de Jong YP, Meuleman P. HCV animal models and liver disease. J Hepatol 2014; 61:S26-33. [PMID: 25443343 DOI: 10.1016/j.jhep.2014.07.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/07/2014] [Accepted: 07/10/2014] [Indexed: 01/08/2023]
Abstract
The development and evaluation of effective therapies and vaccines for the hepatitis C virus (HCV) and the study of its interactions with the mammalian host have been hindered for a long time by the absence of suitable small animal models. Due to the narrow host tropism of HCV, the development of mice that can be robustly engrafted with human hepatocytes was a major breakthrough since they recapitulate the complete HCV life cycle. This model has been useful to investigate many aspects of the HCV life cycle, including antiviral interventions. However, studies of cellular immunity, immunopathogenesis and resulting liver diseases have been hampered by the lack of a small animal model with a functional immune system. In this review, we summarize the evolution of in vivo models for the study of HCV.
Collapse
Affiliation(s)
- Koen Vercauteren
- Center for Vaccinology, Ghent University Hospital, Ghent University, Gent, Belgium
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, USA; Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, USA
| | - Philip Meuleman
- Center for Vaccinology, Ghent University Hospital, Ghent University, Gent, Belgium.
| |
Collapse
|
54
|
Baumert TF, Fauvelle C, Chen DY, Lauer GM. A prophylactic hepatitis C virus vaccine: a distant peak still worth climbing. J Hepatol 2014; 61:S34-44. [PMID: 25443345 DOI: 10.1016/j.jhep.2014.09.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/04/2014] [Accepted: 09/05/2014] [Indexed: 12/23/2022]
Abstract
Hepatitis C virus (HCV) infects an estimated more than 150 million people and is a leading cause of liver disease worldwide. The development of direct-acting antivirals (DAAs) will markedly improve the outcome of antiviral treatment with cure of the majority of treated patients. However, several hurdles remain before HCV infection can be considered a menace of the past: High treatment costs will most likely result in absent or limited access in middle and low resource countries and will lead to selective use even in wealthier countries. The limited efficacy of current HCV screening programs leads to a majority of cases being undiagnosed or diagnosed at a late stage and DAAs will not cure virus-induced end-stage liver disease such as hepatocellular carcinoma. Certain patient subgroups may not respond or not be eligible for DAA-based treatment strategies. Finally, reinfection remains possible, making control of HCV infection in people with ongoing infection risk difficult. The unmet medical needs justify continued efforts to develop an effective vaccine, protecting from chronic HCV infection as a mean to impact the epidemic on a global scale. Recent progress in the understanding of virus-host interactions provides new perspectives for vaccine development, but many critical questions remain unanswered. In this review, we focus on what is known about the immune correlates of HCV control, highlight key mechanisms of viral evasion that pose challenges for vaccine development and suggest areas of further investigation that could enable a rational approach to vaccine design. Within this context we also discuss insights from recent HCV vaccination studies and what they suggest about the best way to go forward.
Collapse
Affiliation(s)
- Thomas F Baumert
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, USA; Inserm Unité 1110, France; Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, France; Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| | - Catherine Fauvelle
- Inserm Unité 1110, France; Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, France
| | - Diana Y Chen
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, USA
| | - Georg M Lauer
- Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, USA.
| |
Collapse
|
55
|
Heidrich B, Pischke S, Helfritz FA, Mederacke I, Kirschner J, Schneider J, Raupach R, Jäckel E, Barg-Hock H, Lehner F, Klempnauer J, von Hahn T, Cornberg M, Manns MP, Ciesek S, Wedemeyer H. Hepatitis C virus core antigen testing in liver and kidney transplant recipients. J Viral Hepat 2014; 21:769-79. [PMID: 24251818 DOI: 10.1111/jvh.12204] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/24/2013] [Indexed: 02/07/2023]
Abstract
HCV RNA levels correlate with the long-term outcome of hepatitis C in liver transplant recipients. Nucleic acid testing (NAT) is usually used to confirm HCV reinfection and to examine viral loads after liver transplantation. HCV core antigen (HCVcoreAg) testing could be an alternative to NAT with some potential advantages including very low intra- and interassay variabilities and lower costs. The performance of HCVcoreAg testing in organ transplant recipients is unknown. We prospectively studied 1011 sera for HCV RNA and HCVcoreAg in a routine real-world setting including 222 samples obtained from patients after liver or kidney transplantation. HCV RNA and HCVcoreAg test results showed a consistency of 98% with a very good correlation in transplanted patients (r > 0.85). The correlation between HCV RNA and HCVcoreAg was higher in sera with high viral loads and in samples from patients with low biochemical disease. Patients treated with tacrolimus showed a better correlation between both parameters than individuals receiving cyclosporine A. HCV RNA/HCVcoreAg ratios did not differ between transplanted and nontransplanted patients, and HCV RNA and HCVcoreAg kinetics were almost identical during the first days after liver transplantation. HCVcoreAg testing can be used to monitor HCV viral loads in patients after organ transplantation. However, the assay is not recommended to monitor antiviral therapies.
Collapse
Affiliation(s)
- B Heidrich
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany; Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Grassi A, Ballardini G. Post-liver transplant hepatitis C virus recurrence: an unresolved thorny problem. World J Gastroenterol 2014; 20:11095-115. [PMID: 25170198 PMCID: PMC4145752 DOI: 10.3748/wjg.v20.i32.11095] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/15/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV)-related cirrhosis represents the leading cause of liver transplantation in developed, Western and Eastern countries. Unfortunately, liver transplantation does not cure recipient HCV infection: reinfection universally occurs and disease progression is faster after liver transplant. In this review we focus on what happens throughout the peri-transplant phase and in the first 6-12 mo after transplantation: during this crucial period a completely new balance between HCV, liver graft, the recipient's immune response and anti-rejection therapy is achieved that will deeply affect subsequent outcomes. Nearly all patients show an early graft reinfection, with HCV viremia reaching and exceeding pre-transplant levels; in this setting, histological assessment is essential to differentiate recurrent hepatitis C from acute or chronic rejection; however, differentiating the two patterns remains difficult. The host immune response (mainly cellular mediated) appears to be crucial both in the control of HCV infection and in the genesis of rejection, and it is also strongly influenced by immunosuppressive treatment. At present no clear immunosuppressive strategy could be strongly recommended in HCV-positive recipients to prevent HCV recurrence, even immunotherapy appears to be ineffective. Nonetheless it seems reasonable that episodes of rejection and over-immunosuppression are more likely to enhance the risk of HCV recurrence through immunological mechanisms. Both complete prevention of rejection and optimization of immunosuppression should represent the main goals towards reducing the rate of graft HCV reinfection. In conclusion, post-transplant HCV recurrence remains an unresolved, thorny problem because many factors remain obscure and need to be better determined.
Collapse
|
57
|
Kwon YC, Ray RB, Ray R. Hepatitis C virus infection: establishment of chronicity and liver disease progression. EXCLI JOURNAL 2014; 13:977-96. [PMID: 26417315 PMCID: PMC4464452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/14/2014] [Indexed: 11/16/2022]
Abstract
Hepatitis C virus (HCV) often causes persistent infection, and is an important factor in the etiology of fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). There are no preventive or therapeutic vaccines available against HCV. Treatment strategies of HCV infection are likely to improve with recently discovered direct antiviral agents (DAAs). However, a proportion of patients still progress to liver failure and/or HCC despite having been cured of the infection. Thus, there is a need for early diagnosis and therapeutic modalities for HCV related end stage liver disease prevention. HCV genome does not integrate into its host genome, and has a predominantly cytoplasmic life cycle. Therefore, HCV mediated liver disease progression appears to involve indirect mechanisms from persistent infection of hepatocytes. Studying the underlying mechanisms of HCV mediated evasion of immune responses and liver disease progression is challenging due to the lack of a naturally susceptible small animal model. We and other investigators have used a number of experimental systems to investigate the mechanisms for establishment of chronic HCV infection and liver disease progression. HCV infection modulates immune systems. Further, HCV infection of primary human hepatocytes promotes growth, induces phenotypic changes, modulates epithelial mesenchymal transition (EMT) related genes, and generates tumor initiating stem-like cells (TISCs). HCV infection also modulates microRNAs (miRNAs), and influences growth by overriding normal death progression of primary human hepatocytes for disease pathogenesis. Understanding these ob-servations at the molecular level should aid in developing strategies for additional effective therapies against HCV mediated liver disease progression.
Collapse
Affiliation(s)
- Young-Chan Kwon
- Department of Internal Medicine, Saint Louis University, Missouri
| | - Ratna B. Ray
- Department of Pathology, Saint Louis University, Missouri
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, Missouri,Department of Molecular Microbiology & Immunology, Saint Louis University, Missouri,*To whom correspondence should be addressed: Ranjit Ray, Division of Infectious Diseases, Allergy & Immunology, Edward A. Doisy Research Center, 1100 S. Grand Blvd, 8th Floor, St. Louis, MO 63104, USA, E-mail:
| |
Collapse
|
58
|
Lefèvre M, Felmlee DJ, Parnot M, Baumert TF, Schuster C. Syndecan 4 is involved in mediating HCV entry through interaction with lipoviral particle-associated apolipoprotein E. PLoS One 2014; 9:e95550. [PMID: 24751902 PMCID: PMC3994096 DOI: 10.1371/journal.pone.0095550] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 03/28/2014] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) is a major cause of liver disease worldwide and HCV infection represents a major health problem. HCV associates with host lipoproteins forming host/viral hybrid complexes termed lipoviral particles. Apolipoprotein E (apoE) is a lipoprotein component that interacts with heparan sulfate proteoglycans (HSPG) to mediate hepatic lipoprotein uptake, and may likewise mediate HCV entry. We sought to define the functional regions of apoE with an aim to identify critical apoE binding partners involved in HCV infection. Using adenoviral vectors and siRNA to modulate apoE expression we show a direct correlation of apoE expression and HCV infectivity, whereas no correlation exists with viral protein expression. Mutating the HSPG binding domain (HSPG-BD) of apoE revealed key residues that are critical for mediating HCV infection. Furthermore, a novel synthetic peptide that mimics apoE’s HSPG-BD directly and competitively inhibits HCV infection. Genetic knockdown of the HSPG proteins syndecan (SDC) 1 and 4 revealed that SDC4 principally mediates HCV entry. Our data demonstrate that HCV uses apoE-SDC4 interactions to enter hepatoma cells and establish infection. Targeting apoE-SDC interactions could be an alternative strategy for blocking HCV entry, a critical step in maintaining chronic HCV infection.
Collapse
Affiliation(s)
- Mathieu Lefèvre
- Inserm, U1110, Research Institute on Viral and Hepatic Disease, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Daniel J. Felmlee
- Inserm, U1110, Research Institute on Viral and Hepatic Disease, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Marie Parnot
- Inserm, U1110, Research Institute on Viral and Hepatic Disease, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U1110, Research Institute on Viral and Hepatic Disease, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Pôle hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Catherine Schuster
- Inserm, U1110, Research Institute on Viral and Hepatic Disease, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- * E-mail:
| |
Collapse
|
59
|
Hartfield M, Bull R, White PA, Lloyd A, Luciani F, Alizon S. Evidence that hepatitis C virus genome partly controls infection outcome. Evol Appl 2014; 7:533-47. [PMID: 24944567 PMCID: PMC4055175 DOI: 10.1111/eva.12151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/07/2014] [Indexed: 12/16/2022] Open
Abstract
Infection by hepatitis C virus (HCV) leads to one of two outcomes; either the infection resolves within approximately 6 months or the virus can persist indefinitely. Host genetics are known to affect the likelihood of clearance or persistence. By contrast, the importance of the virus genotype in determining infection outcome is unknown, as quantifying this effect traditionally requires well-characterized transmission networks, which are rare. Extending phylogenetic approaches previously developed to estimate the virus control over set-point viral load in HIV-1 infections, we simulate inheritance of a binary trait along a phylogenetic tree, use this data to quantify how infection outcomes cluster and ascertain the effect of virus genotype on these. We apply our method to the Hepatitis C Incidence and Transmission Study in prisons (HITS-p) data set from Australia, as this cohort prospectively identified incident cases including viraemic subjects who ultimately clear the virus, thus providing us with a unique collection of sequences from clearing infections. We detect significant correlations between infection outcome and virus distance in the phylogeny for viruses of Genotype 1, with estimates lying at around 67%. No statistically significant estimates were obtained for viruses of Genotype 3a.
Collapse
Affiliation(s)
- Matthew Hartfield
- Laboratoire MIVEGEC (UMR CNRS 5290 IRD 224 UM1 UM2) Montpellier Cedex 5, France
| | - Rowena Bull
- Infection and Inflammation Research Centre, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Australia
| | - Peter A White
- Infection and Inflammation Research Centre, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Australia
| | - Andrew Lloyd
- Infection and Inflammation Research Centre, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Australia
| | - Fabio Luciani
- Infection and Inflammation Research Centre, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Australia
| | - Samuel Alizon
- Laboratoire MIVEGEC (UMR CNRS 5290 IRD 224 UM1 UM2) Montpellier Cedex 5, France
| | | |
Collapse
|
60
|
Upadya MH, Aweya JJ, Tan YJ. Understanding the interaction of hepatitis C virus with host DEAD-box RNA helicases. World J Gastroenterol 2014; 20:2913-2926. [PMID: 24659882 PMCID: PMC3961968 DOI: 10.3748/wjg.v20.i11.2913] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/06/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
The current therapeutic regimen to combat chronic hepatitis C is not optimal due to substantial side effects and the failure of a significant proportion of patients to achieve a sustained virological response. Recently developed direct-acting antivirals targeting hepatitis C virus (HCV) enzymes reportedly increase the virologic response to therapy but may lead to a selection of drug-resistant variants. Besides direct-acting antivirals, another promising class of HCV drugs in development include host targeting agents that are responsible for interfering with the host factors crucial for the viral life cycle. A family of host proteins known as DEAD-box RNA helicases, characterized by nine conserved motifs, is known to play an important role in RNA metabolism. Several members of this family such as DDX3, DDX5 and DDX6 have been shown to play a role in HCV replication and this review will summarize our current knowledge on their interaction with HCV. As chronic hepatitis C is one of the leading causes of hepatocellular carcinoma, the involvement of DEAD-box RNA helicases in the development of HCC will also be highlighted. Continuing research on the interaction of host DEAD-box proteins with HCV and the contribution to viral replication and pathogenesis could be the panacea for the development of novel therapeutics against HCV.
Collapse
|
61
|
Abstract
The high variability and the limited knowledge of the structure of the hepatitis C virus (HCV) envelope glycoproteins (GP) are challenging hurdles for vaccine design. Recently, Kong et al. published a new model of HCV E2 GP structure in Science, revealing a globular structure, starkly contrasting from the extended model of class II fusion proteins from other Flaviviridae viruses.
Collapse
Affiliation(s)
- Catherine Fauvelle
- 1] Inserm Unit 1110, Université de Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France [2] Pôle Hépato-digestif, Université de Strasbourg, Strasbourg, France
| | - Daniel J Felmlee
- 1] Inserm Unit 1110, Université de Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France [2] Pôle Hépato-digestif, Université de Strasbourg, Strasbourg, France
| | - Thomas F Baumert
- 1] Inserm Unit 1110, Université de Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France [2] Pôle Hépato-digestif, Université de Strasbourg, Strasbourg, France [3] Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
62
|
Fofana I, Jilg N, Chung RT, Baumert TF. Entry inhibitors and future treatment of hepatitis C. Antiviral Res 2014; 104:136-42. [PMID: 24525381 DOI: 10.1016/j.antiviral.2014.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/29/2014] [Accepted: 02/02/2014] [Indexed: 12/25/2022]
Abstract
Hepatitis C virus (HCV) is a major cause of liver cirrhosis and hepatocellular carcinoma. Furthermore, HCV-induced liver disease is the leading indication for liver transplantation. The recent introduction of direct-acting antivirals (DAAs) has revolutionized HCV treatment by making possible the cure of the majority of patients. However, their efficacy and safety in difficult-to-treat patients such as patients receiving immunosuppression, those with advanced liver disease, co-morbidity and HIV/HCV-co-infection remain to be determined. Furthermore, prevention of liver graft infection remains a pressing issue. HCV entry inhibitors target the very first step of the HCV life cycle and efficiently inhibit cell-cell transmission - a key prerequisite for viral spread. Because of their unique mechanism of action on cell-cell transmission they may provide a promising and simple perspective for prevention of liver graft infection. A high genetic barrier to resistance and complementary mechanism of action compared to DAAs makes entry inhibitors attractive as a new strategy for treatment of multi-resistant or difficult-to-treat patients. Clinical studies are needed to determine the future role of entry inhibitors in the arsenal of antivirals to combat HCV infection. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication."
Collapse
Affiliation(s)
- Isabel Fofana
- Inserm U1110, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | - Nikolaus Jilg
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Raymond T Chung
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Thomas F Baumert
- Inserm U1110, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
63
|
A neutralization epitope in the hepatitis C virus E2 glycoprotein interacts with host entry factor CD81. PLoS One 2014; 9:e84346. [PMID: 24400084 PMCID: PMC3882236 DOI: 10.1371/journal.pone.0084346] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 11/14/2013] [Indexed: 12/17/2022] Open
Abstract
The identification of a specific immunogenic candidate that will effectively activate the appropriate pathway for neutralizing antibody production is fundamental for vaccine design. By using a monoclonal antibody (1H8) that neutralizes HCV in vitro, we have demonstrated here that 1H8 recognized an epitope mapped between residues A524 and W529 of the E2 protein. We also found that the epitope residues A524, P525, Y527 and W529 were crucial for antibody binding, while the residues T526, Y527 and W529 within the same epitope engaged in the interaction with the host entry factor CD81. Furthermore, we detected "1H8-like" antibodies, defined as those with amino acid-specificity similar to 1H8, in the plasma of patients with chronic HCV infection. The time course study of plasma samples from Patient H, a well-characterized case of post-transfusion hepatitis C, showed that "1H8-like" antibodies could be detected in a sample collected almost two years after the initial infection, thus confirming the immunogenicity of this epitope in vivo. The characterization of this neutralization epitope with a function in host entry factor CD81 interaction should enhance our understanding of antibody-mediated neutralization of HCV infections.
Collapse
|
64
|
Wang W, Guan M, Liu Y, Xu Q, Peng H, Liu X, Tang Z, Zhu Y, Wu D, Ren H, Zhao P, Qi Z. Alanine scanning mutagenesis of hepatitis C virus E2 cysteine residues: Insights into E2 biogenesis and antigenicity. Virology 2013; 448:229-37. [PMID: 24314653 DOI: 10.1016/j.virol.2013.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 07/29/2013] [Accepted: 10/14/2013] [Indexed: 12/15/2022]
Abstract
Envelope glycoprotein 2 (E2) of hepatitis C virus contains 18 conserved cysteine (Cys) residues in its ectodomain. By cysteine-alanine mutagenesis and function analysis, six Cys in H77 E2 (C494, C508, C552, C564, C607 and C644) were found to be indispensable for recognition by conformation-dependent mAb H53. Removal of any of these Cys residues did not affect E2 heterodimerization with E1, but notably reduced E1E2 transmembrane transportation. These Cys together with C429 and C503 were required for conformation-dependent mAb H48 recognition. All of the above Cys except C607 were required for H77 and Con1 E2 binding to CD81. None of individual mutation of above Cys affected the ability of E2 to induce neutralizing antibodies in mice. Mouse antibodies mainly recognize E2 linear epitopes and are unrelated to epitopes recognized by human E2 antibodies. The findings provide new insights for understanding the biogenesis of functional HCV envelope proteins and HCV neutralizing immunity.
Collapse
Affiliation(s)
- Wenbo Wang
- Department of Microbiology, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Analysis of serine codon conservation reveals diverse phenotypic constraints on hepatitis C virus glycoprotein evolution. J Virol 2013; 88:667-78. [PMID: 24173227 DOI: 10.1128/jvi.01745-13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Serine is encoded by two divergent codon types, UCN and AGY, which are not interchangeable by a single nucleotide substitution. Switching between codon types therefore occurs via intermediates (threonine or cysteine) or via simultaneous tandem substitutions. Hepatitis C virus (HCV) chronically infects 2 to 3% of the global population. The highly variable glycoproteins E1 and E2 decorate the surface of the viral envelope, facilitate cellular entry, and are targets for host immunity. Comparative sequence analysis of globally sampled E1E2 genes, coupled with phylogenetic analysis, reveals the signatures of multiple archaic codon-switching events at seven highly conserved serine residues. Limited detection of intermediate phenotypes indicates that associated fitness costs restrict their fixation in divergent HCV lineages. Mutational pathways underlying codon switching were probed via reverse genetics, assessing glycoprotein functionality using multiple in vitro systems. These data demonstrate selection against intermediate phenotypes can act at the structural/functional level, with some intermediates displaying impaired virion assembly and/or decreased capacity for target cell entry. These effects act in residue/isolate-specific manner. Selection against intermediates is also provided by humoral targeting, with some intermediates exhibiting increased epitope exposure and enhanced neutralization sensitivity, despite maintaining a capacity for target cell entry. Thus, purifying selection against intermediates limits their frequencies in globally sampled strains, with divergent functional constraints at the protein level restricting the fixation of deleterious mutations. Overall our study provides an experimental framework for identification of barriers limiting viral substitutional evolution and indicates that serine codon-switching represents a genomic "fossil record" of historical purifying selection against E1E2 intermediate phenotypes.
Collapse
|
66
|
Sequence and functional analysis of the envelope glycoproteins of hepatitis C virus variants selectively transmitted to a new host. J Virol 2013; 87:13609-18. [PMID: 24109215 DOI: 10.1128/jvi.02119-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C virus (HCV) remains a challenging public health problem worldwide. The identification of viral variants establishing de novo infections and definition of the phenotypic requirements for transmission would facilitate the design of preventive strategies. We explored the transmission of HCV variants in three cases of acute hepatitis following needlestick accidents. We used single-genome amplification of glycoprotein E1E2 gene sequences to map the genetic bottleneck upon transmission accurately. We found that infection was likely established by a single variant in two cases and six variants in the third case. Studies of donor samples showed that the transmitted variant E1E2 amino acid sequences were identical or closely related to those of variants from the donor virus populations. The transmitted variants harbored a common signature site at position 394, within hypervariable region 1 of E2, together with additional signature amino acids specific to each transmission pair. Surprisingly, these E1E2 variants conferred no greater capacity for entry than the E1E2 derived from nontransmitted variants in lentiviral pseudoparticle assays. Mutants escaping the antibodies of donor sera did not predominate among the transmitted variants either. The fitness parameters affecting the selective outgrowth of HCV variants after transmission in an immunocompetent host may thus be more complex than those suggested by mouse models. Human antibodies directed against HCV envelope effectively cross-neutralized the lentiviral particles bearing E1E2 derived from transmitted variants. These findings provide insight into the molecular mechanisms underlying HCV transmission and suggest that viral entry is a potential target for the prevention of HCV infection.
Collapse
|
67
|
Abstract
Hepatitis C virus (HCV) is a hepatotropic virus and a major cause of chronic hepatitis and liver disease worldwide. Initial interactions between HCV virions and hepatocytes are required for productive viral infection and initiation of the viral life cycle. Furthermore, HCV entry contributes to the tissue tropism and species specificity of this virus. The elucidation of these interactions is critical, not only to understand the pathogenesis of HCV infection, but also to design efficient antiviral strategies and vaccines. This review summarizes our current knowledge of the host factors required for the HCV-host interactions during HCV binding and entry, our understanding of the molecular mechanisms underlying HCV entry into target cells, and the relevance of HCV entry for the pathogenesis of liver disease, antiviral therapy, and vaccine development.
Collapse
|
68
|
Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells. Proc Natl Acad Sci U S A 2013; 110:13109-13. [PMID: 23878230 DOI: 10.1073/pnas.1221899110] [Citation(s) in RCA: 403] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent evidence indicates there is a role for small membrane vesicles, including exosomes, as vehicles for intercellular communication. Exosomes secreted by most cell types can mediate transfer of proteins, mRNAs, and microRNAs, but their role in the transmission of infectious agents is less established. Recent studies have shown that hepatocyte-derived exosomes containing hepatitis C virus (HCV) RNA can activate innate immune cells, but the role of exosomes in the transmission of HCV between hepatocytes remains unknown. In this study, we investigated whether exosomes transfer HCV in the presence of neutralizing antibodies. Purified exosomes isolated from HCV-infected human hepatoma Huh7.5.1 cells were shown to contain full-length viral RNA, viral protein, and particles, as determined by RT-PCR, mass spectrometry, and transmission electron microscopy. Exosomes from HCV-infected cells were capable of transmitting infection to naive human hepatoma Huh7.5.1 cells and establishing a productive infection. Even with subgenomic replicons, lacking structural viral proteins, exosome-mediated transmission of HCV RNA was observed. Treatment with patient-derived IgGs showed a variable degree of neutralization of exosome-mediated infection compared with free virus. In conclusion, this study showed that hepatic exosomes can transmit productive HCV infection in vitro and are partially resistant to antibody neutralization. This discovery sheds light on neutralizing antibodies resistant to HCV transmission by exosomes as a potential immune evasion mechanism.
Collapse
|
69
|
Functional analysis of claudin-6 and claudin-9 as entry factors for hepatitis C virus infection of human hepatocytes by using monoclonal antibodies. J Virol 2013; 87:10405-10. [PMID: 23864633 DOI: 10.1128/jvi.01691-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The relevance of claudin-6 and claudin-9 in hepatitis C virus (HCV) entry remains elusive. We produced claudin-6- or claudin-9-specific monoclonal antibodies that inhibit HCV entry into nonhepatic cells expressing exogenous claudin-6 or claudin-9. These antibodies had no effect on HCV infection of hepatoma cells or primary hepatocytes. Thus, although claudin-6 and claudin-9 can serve as entry factors in cell lines, HCV infection into human hepatocytes is not dependent on claudin-6 and claudin-9.
Collapse
|
70
|
Chang CC, Crane M, Zhou J, Mina M, Post JJ, Cameron BA, Lloyd AR, Jaworowski A, French MA, Lewin SR. HIV and co-infections. Immunol Rev 2013; 254:114-42. [PMID: 23772618 PMCID: PMC3697435 DOI: 10.1111/imr.12063] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite significant reductions in morbidity and mortality secondary to availability of effective combination anti-retroviral therapy (cART), human immunodeficiency virus (HIV) infection still accounts for 1.5 million deaths annually. The majority of deaths occur in sub-Saharan Africa where rates of opportunistic co-infections are disproportionately high. In this review, we discuss the immunopathogenesis of five common infections that cause significant morbidity in HIV-infected patients globally. These include co-infection with Mycobacterium tuberculosis, Cryptococcus neoformans, hepatitis B virus, hepatitis C virus, and Plasmodium falciparum. Specifically, we review the natural history of each co-infection in the setting of HIV, the specific immune defects induced by HIV, the effects of cART on the immune response to the co-infection, the pathogenesis of immune restoration disease (IRD) associated with each infection, and advances in the areas of prevention of each co-infection via vaccination. Finally, we discuss the opportunities and gaps in knowledge for future research.
Collapse
Affiliation(s)
- Christina C Chang
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Murray JM, Moenne-Loccoz R, Velay A, Habersetzer F, Doffoël M, Gut JP, Fofana I, Zeisel MB, Stoll-Keller F, Baumert TF, Schvoerer E. Genotype 1 hepatitis C virus envelope features that determine antiviral response assessed through optimal covariance networks. PLoS One 2013; 8:e67254. [PMID: 23840641 PMCID: PMC3688619 DOI: 10.1371/journal.pone.0067254] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 05/14/2013] [Indexed: 01/25/2023] Open
Abstract
The poor response to the combined antiviral therapy of pegylated alfa-interferon and ribavarin for hepatitis C virus (HCV) infection may be linked to mutations in the viral envelope gene E1E2 (env), which can result in escape from the immune response and higher efficacy of viral entry. Mutations that result in failure of therapy most likely require compensatory mutations to achieve sufficient change in envelope structure and function. Compensatory mutations were investigated by determining positions in the E1E2 gene where amino acids (aa) covaried across groups of individuals. We assessed networks of covarying positions in E1E2 sequences that differentiated sustained virological response (SVR) from non-response (NR) in 43 genotype 1a (17 SVR), and 49 genotype 1b (25 SVR) chronically HCV-infected individuals. Binary integer programming over covariance networks was used to extract aa combinations that differed between response groups. Genotype 1a E1E2 sequences exhibited higher degrees of covariance and clustered into 3 main groups while 1b sequences exhibited no clustering. Between 5 and 9 aa pairs were required to separate SVR from NR in each genotype. aa in hypervariable region 1 were 6 times more likely than chance to occur in the optimal networks. The pair 531-626 (EI) appeared frequently in the optimal networks and was present in 6 of 9 NR in one of the 1a clusters. The most frequent pairs representing SVR were 431-481 (EE), 500-522 (QA) in 1a, and 407-434 (AQ) in 1b. Optimal networks based on covarying aa pairs in HCV envelope can indicate features that are associated with failure or success to antiviral therapy.
Collapse
Affiliation(s)
- John M Murray
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Hepatitis C virus, cholesterol and lipoproteins--impact for the viral life cycle and pathogenesis of liver disease. Viruses 2013; 5:1292-324. [PMID: 23698400 PMCID: PMC3712309 DOI: 10.3390/v5051292] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/10/2013] [Accepted: 04/27/2013] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) is a leading cause of chronic liver disease, including chronic hepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Hepatitis C infection associates with lipid and lipoprotein metabolism disorders such as hepatic steatosis, hypobetalipoproteinemia, and hypocholesterolemia. Furthermore, virus production is dependent on hepatic very-low-density lipoprotein (VLDL) assembly, and circulating virions are physically associated with lipoproteins in complexes termed lipoviral particles. Evidence has indicated several functional roles for the formation of these complexes, including co-opting of lipoprotein receptors for attachment and entry, concealing epitopes to facilitate immune escape, and hijacking host factors for HCV maturation and secretion. Here, we review the evidence surrounding pathogenesis of the hepatitis C infection regarding lipoprotein engagement, cholesterol and triglyceride regulation, and the molecular mechanisms underlying these effects.
Collapse
|
73
|
Different requirements for scavenger receptor class B type I in hepatitis C virus cell-free versus cell-to-cell transmission. J Virol 2013; 87:8282-93. [PMID: 23698298 DOI: 10.1128/jvi.01102-13] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C virus (HCV) is believed to initially infect the liver through the basolateral side of hepatocytes, where it engages attachment factors and the coreceptors CD81 and scavenger receptor class B type I (SR-BI). Active transport toward the apical side brings the virus in close proximity of additional entry factors, the tight junction molecules claudin-1 and occludin. HCV is also thought to propagate via cell-to-cell spread, which allows highly efficient virion delivery to neighboring cells. In this study, we compared an adapted HCV genome, clone 2, characterized by superior cell-to cell spread, to its parental genome, J6/JFH-1, with the goal of elucidating the molecular mechanisms of HCV cell-to-cell transmission. We show that CD81 levels on the donor cells influence the efficiency of cell-to-cell spread and CD81 transfer between neighboring cells correlates with the capacity of target cells to become infected. Spread of J6/JFH-1 was blocked by anti-SR-BI antibody or in cells knocked down for SR-BI, suggesting a direct role for this receptor in HCV cell-to-cell transmission. In contrast, clone 2 displayed a significantly reduced dependence on SR-BI for lateral spread. Mutations in E1 and E2 responsible for the enhanced cell-to-cell spread phenotype of clone 2 rendered cell-free virus more susceptible to antibody-mediated neutralization. Our results indicate that although HCV can lose SR-BI dependence for cell-to-cell spread, vulnerability to neutralizing antibodies may limit this evolutionary option in vivo. Combination therapies targeting both the HCV glycoproteins and SR-BI may therefore hold promise for effective control of HCV dissemination.
Collapse
|
74
|
Fofana I, Xiao F, Thumann C, Turek M, Zona L, Tawar RG, Grunert F, Thompson J, Zeisel MB, Baumert TF. A novel monoclonal anti-CD81 antibody produced by genetic immunization efficiently inhibits Hepatitis C virus cell-cell transmission. PLoS One 2013; 8:e64221. [PMID: 23704981 PMCID: PMC3660333 DOI: 10.1371/journal.pone.0064221] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/12/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS Hepatitis C virus (HCV) infection is a challenge to prevent and treat because of the rapid development of drug resistance and escape. Viral entry is required for initiation, spread, and maintenance of infection, making it an attractive target for antiviral strategies. METHODS Using genetic immunization, we produced four monoclonal antibodies (mAbs) against the HCV host entry factor CD81. The effects of antibodies on inhibition of HCV infection and dissemination were analyzed in HCV permissive human liver cell lines. RESULTS The anti-CD81 mAbs efficiently inhibited infection by HCV of different genotypes as well as a HCV escape variant selected during liver transplantation and re-infecting the liver graft. Kinetic studies indicated that anti-CD81 mAbs target a post-binding step during HCV entry. In addition to inhibiting cell-free HCV infection, one antibody was also able to block neutralizing antibody-resistant HCV cell-cell transmission and viral dissemination without displaying any detectable toxicity. CONCLUSION A novel anti-CD81 mAb generated by genetic immunization efficiently blocks HCV spread and dissemination. This antibody will be useful to further unravel the role of virus-host interactions during HCV entry and cell-cell transmission. Furthermore, this antibody may be of interest for the development of antivirals for prevention and treatment of HCV infection.
Collapse
Affiliation(s)
- Isabel Fofana
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Fei Xiao
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Christine Thumann
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Marine Turek
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Laetitia Zona
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Rajiv G. Tawar
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | | | | | - Mirjam B. Zeisel
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U1110, Institute of Virology, Strasbourg, France
- University of Strasbourg, Strasbourg, France
- Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- * E-mail:
| |
Collapse
|
75
|
Krey T, Meola A, Keck ZY, Damier-Piolle L, Foung SKH, Rey FA. Structural basis of HCV neutralization by human monoclonal antibodies resistant to viral neutralization escape. PLoS Pathog 2013; 9:e1003364. [PMID: 23696737 PMCID: PMC3656090 DOI: 10.1371/journal.ppat.1003364] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 04/02/2013] [Indexed: 01/27/2023] Open
Abstract
The high mutation rate of hepatitis C virus allows it to rapidly evade the humoral immune response. However, certain epitopes in the envelope glycoproteins cannot vary without compromising virus viability. Antibodies targeting these epitopes are resistant to viral escape from neutralization and understanding their binding-mode is important for vaccine design. Human monoclonal antibodies HC84-1 and HC84-27 target conformational epitopes overlapping the CD81 receptor-binding site, formed by segments aa434-446 and aa610-619 within the major HCV glycoprotein E2. No neutralization escape was yet observed for these antibodies. We report here the crystal structures of their Fab fragments in complex with a synthetic peptide comprising aa434-446. The structures show that the peptide adopts an α-helical conformation with the main contact residues F⁴⁴² and Y⁴⁴³ forming a hydrophobic protrusion. The peptide retained its conformation in both complexes, independently of crystal packing, indicating that it reflects a surface feature of the folded glycoprotein that is exposed similarly on the virion. The same residues of E2 are also involved in interaction with CD81, suggesting that the cellular receptor binds the same surface feature and potential escape mutants critically compromise receptor binding. In summary, our results identify a critical structural motif at the E2 surface, which is essential for virus propagation and therefore represents an ideal candidate for structure-based immunogen design for vaccine development.
Collapse
Affiliation(s)
- Thomas Krey
- Institut Pasteur, Unité de Virologie Structurale, Departement Virologie, Paris, France.
| | | | | | | | | | | |
Collapse
|
76
|
Induction of broadly neutralising HCV antibodies in mice by integration-deficient lentiviral vector-based pseudotyped particles. PLoS One 2013; 8:e62684. [PMID: 23626846 PMCID: PMC3633868 DOI: 10.1371/journal.pone.0062684] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/24/2013] [Indexed: 12/26/2022] Open
Abstract
Introduction Integration-deficient lentiviral vectors (IDLVs) are a promising platform for immunisation to elicit both humoral immunity and cellular mediated immunity (CMI). Here, we compared the specific immunity in mice immunised via different regimens (homologous and cocktail) with IDLV-based HCV pseudoparticles (HCVpps) carrying pseudotyped glycoproteins E1E2 and bearing the HCV NS3 gene. Humoral and cell-mediated immune responses were also evaluated after IDLV-HCVpp immunisation combined with heterologous rAd5-CE1E2 priming protocols. Sera from the mice effectively elicited anti-E1, -E2, and -NS3 antibody responses, and neutralised various HCVpp subtypes (1a, 1b, 2a, 3a and 5a). No significant CMI was detected in the groups immunised with IDLV-based HCVpps. In contrast, the combination of rAd5-CE1E2 priming and IDLV-based HCVpp boosting induced significant CMI against multiple antigens (E1, E2, and NS3). Conclusion IDLV-based HCVpps are a promising vaccination platform and the combination of rAd5-CE1E2 and IDLV-based HCVpp prime-boost strategy should be further explored for the development of a cross-protective HCV vaccine.
Collapse
|
77
|
Chung RT, Gordon FD, Curry MP, Schiano TD, Emre S, Corey K, Markmann J, Hertl M, Pomposelli JJ, Pomfret EA, Florman S, Schilsky M, Broering TJ, Finberg RW, Szabo G, Zamore PD, Khettry U, Babcock GJ, Ambrosino DM, Leav B, Leney M, Smith HL, Molrine DC. Human monoclonal antibody MBL-HCV1 delays HCV viral rebound following liver transplantation: a randomized controlled study. Am J Transplant 2013; 13:1047-1054. [PMID: 23356386 PMCID: PMC3618536 DOI: 10.1111/ajt.12083] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 11/20/2012] [Accepted: 12/04/2012] [Indexed: 01/25/2023]
Abstract
Rapid allograft infection complicates liver transplantation (LT) in patients with hepatitis C virus (HCV). Pegylated interferon-α and ribavirin therapy after LT has significant toxicity and limited efficacy. The effect of a human monoclonal antibody targeting the HCV E2 glycoprotein (MBL-HCV1) on viral clearance was examined in a randomized, double-blind, placebo-controlled pilot study in patients infected with HCV genotype 1a undergoing LT. Subjects received 11 infusions of 50 mg/kg MBL-HCV1 (n=6) or placebo (n=5) intravenously with three infusions on day of transplant, a single infusion on days 1 through 7 and one infusion on day 14 after LT. MBL-HCV1 was well-tolerated and reduced viral load for a period ranging from 7 to 28 days. Median change in viral load (log10 IU/mL) from baseline was significantly greater (p=0.02) for the antibody-treated group (range -3.07 to -3.34) compared to placebo group (range -0.331 to -1.01) on days 3 through 6 posttransplant. MBL-HCV1 treatment significantly delayed median time to viral rebound compared to placebo treatment (18.7 days vs. 2.4 days, p<0.001). As with other HCV monotherapies, antibody-treated subjects had resistance-associated variants at the time of viral rebound. A combination study of MBL-HCV1 with a direct-acting antiviral is underway.
Collapse
Affiliation(s)
- R. T. Chung
- Massachusetts General Hospital, Boston, MA, United States
| | - F. D. Gordon
- Lahey Clinic Medical Center, Burlington, MA, United States
| | - M. P. Curry
- Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - T. D. Schiano
- Mount Sinai Medical Center, New York, NY, United States
| | - S. Emre
- Yale New Haven Hospital, New Haven, CT, United States
| | - K. Corey
- Massachusetts General Hospital, Boston, MA, United States
| | - J. Markmann
- Massachusetts General Hospital, Boston, MA, United States
| | - M. Hertl
- Massachusetts General Hospital, Boston, MA, United States
| | | | - E. A. Pomfret
- Lahey Clinic Medical Center, Burlington, MA, United States
| | - S. Florman
- Mount Sinai Medical Center, New York, NY, United States
| | - M. Schilsky
- Yale New Haven Hospital, New Haven, CT, United States
| | - T. J. Broering
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| | - R. W. Finberg
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - G. Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - P. D. Zamore
- Howard Hughes Medical Institute and Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, United States
| | - U. Khettry
- Lahey Clinic Medical Center, Burlington, MA, United States
| | - G. J. Babcock
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| | - D. M. Ambrosino
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| | - B. Leav
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| | - M. Leney
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| | - H. L. Smith
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| | - D. C. Molrine
- MassBiologics, University of Massachusetts Medical School, Boston, MA, United States
| |
Collapse
|
78
|
Host-targeting agents for prevention and treatment of chronic hepatitis C - perspectives and challenges. J Hepatol 2013; 58:375-84. [PMID: 23041307 DOI: 10.1016/j.jhep.2012.09.022] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 09/26/2012] [Accepted: 09/27/2012] [Indexed: 12/12/2022]
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease and hepatocellular carcinoma worldwide. Furthermore, HCV-induced liver disease is a major indication of liver transplantation. In the past years, direct-acting antivirals (DAAs) targeting HCV enzymes have been developed. DAAs increase the virologic response to anti-HCV therapy but may lead to selection of drug-resistant variants and treatment failure. To date, strategies to prevent HCV infection are still lacking and antiviral therapy in immunocompromised patients, patients with advanced liver disease and HIV/HCV-co-infection remains limited. Alternative or complementary approaches addressing the limitations of current antiviral therapies are to boost the host's innate immunity or interfere with host factors required for pathogenesis. Host-targeting agents (HTAs) provide an interesting perspective for novel antiviral strategies against viral hepatitis since they have (i) a high genetic barrier to resistance, (ii) a pan-genotypic antiviral activity, and (iii) complementary mechanisms of action to DAAs and might therefore act in a synergistic manner with current standard of care or DAAs in clinical development. This review highlights HTAs against HCV infection that have potential as novel antivirals and are in preclinical or clinical development.
Collapse
|
79
|
Schvoerer E, Moenne-Loccoz R, Murray JM, Velay A, Turek M, Fofana I, Fafi-Kremer S, Erba AC, Habersetzer F, Doffoël M, Gut JP, Donlin MJ, Tavis JE, Zeisel MB, Stoll-Keller F, Baumert TF. Hepatitis C virus envelope glycoprotein signatures are associated with treatment failure and modulation of viral entry and neutralization. J Infect Dis 2013; 207:1306-15. [PMID: 23335805 DOI: 10.1093/infdis/jit032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A major challenge for antiviral treatment of hepatitis C virus (HCV) infection is viral resistance, potentially resulting from the high variability of HCV envelope glycoproteins and subsequent selection of strains with enhanced infectivity and/or immune escape. METHODS We used a bioinformatics and functional approach to investigate whether E1/E2 envelope glycoprotein structure and function were associated with treatment failure in 92 patients infected with HCV genotype 1. RESULTS Bioinformatics analysis identified 1 sustain virological response (R)-related residue in E1 (219T) and 2 non-SVR (NR)-related molecular signatures in E2 (431A and 642V) in HCV genotype 1a. Two of these positions also appeared in minimal networks separating NR patients from R patients. HCV pseudoparticles (HCVpp) expressing 431A and 642V resulted in a decrease in antibody-mediated neutralization by pretreatment sera. 431A/HCVpp entry into Huh7.5 cells increased with overexpression of CD81 and SR-BI. Moreover, an association of envelope glycoprotein signatures with treatment failure was confirmed in an independent cohort (Virahep-C). CONCLUSIONS Combined in silico and functional analyses demonstrate that envelope glycoprotein signatures associated with treatment failure result in an alteration of host cell entry factor use and escape from neutralizing antibodies, suggesting that virus-host interactions during viral entry contribute to treatment failure.
Collapse
|
80
|
Neumann-Haefelin C, Thimme R. Adaptive immune responses in hepatitis C virus infection. Curr Top Microbiol Immunol 2013; 369:243-62. [PMID: 23463204 DOI: 10.1007/978-3-642-27340-7_10] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adaptive immune response plays a central role in the outcome of hepatitis C virus (HCV) infection. Indeed, spontaneous viral clearance is associated with an early neutralizing antibody response as well as vigorous and sustained HCV-specific CD4+ and CD8+ T cell responses. In persistent HCV infection, however, all three components of the antiviral adaptive immune response fail due to different viral evasion strategies. In this chapter, we will describe the components of a successful immune response against HCV and summarize the mechanisms of immune failure. We will also highlight characteristics of protective CD8+ T cell responses which is the key factor to the design of an efficacious vaccine.
Collapse
|
81
|
Guan M, Wang W, Liu X, Tong Y, Liu Y, Ren H, Zhu S, Dubuisson J, Baumert TF, Zhu Y, Peng H, Aurelian L, Zhao P, Qi Z. Three different functional microdomains in the hepatitis C virus hypervariable region 1 (HVR1) mediate entry and immune evasion. J Biol Chem 2012; 287:35631-35645. [PMID: 22927442 PMCID: PMC3471721 DOI: 10.1074/jbc.m112.382341] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/22/2012] [Indexed: 12/29/2022] Open
Abstract
High genetic heterogeneity is an important characteristic of hepatitis C virus (HCV) that contributes to its ability to establish persistent infection. The hypervariable region 1 (HVR1) that includes the first 27 amino acid residues of the E2 envelope glycoprotein is the most variable region within the HCV polyprotein. HVR1 plays a major role in both HCV cell entry and immune evasion, but the respective contribution of specific amino acid residues is still unclear. Our mutagenesis analyses of HCV pseudoparticles and cell culture-derived HCV using the H77 isolate indicate that five residues at positions 14, 15, and 25-27 mediate binding of the E2 protein to the scavenger receptor class B, type I receptor, and any residue herein is indispensable for HCV cell entry. The region spanning positions 16-24 contains the sole neutralizing epitope and is dispensable for HCV entry, but it is involved in heparan binding. More importantly, this region is necessary for the enhancement of HCV entry by high density lipoprotein and interferes with virus neutralization by E2-neutralizing antibodies. Residues at positions 1-13 are also dispensable for HCV entry, but they can affect HCV infectivity by modulating binding of the envelope protein to scavenger receptor class B, type I. Mutations occurring at this site may confer resistance to HVR1 antibodies. These findings further our understanding about the mechanisms of HCV cell entry and the significance of HVR1 variation in HCV immune evasion. They have major implications for the development of HCV entry inhibitors and prophylactic vaccines.
Collapse
Affiliation(s)
- Mo Guan
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Wenbo Wang
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Xiaoqing Liu
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Yimin Tong
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Yuan Liu
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Hao Ren
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Shiying Zhu
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Jean Dubuisson
- Inserm U1019, CNRS UMR8204, Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Université Lille Nord de France, F-59021 Lille, France
| | - Thomas F Baumert
- Unité Inserm U.748, Université de Strasbourg, Nouvel Hôpital Civil, 3 Rue Koeberlé, F-67000 Strasbourg, France
| | - Yongzhe Zhu
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Haoran Peng
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China
| | - Laure Aurelian
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Ping Zhao
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China.
| | - Zhongtian Qi
- Department of Microbiology, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
82
|
Fafi-Kremer S, Fauvelle C, Felmlee DJ, Zeisel MB, Lepiller Q, Fofana I, Heydmann L, Stoll-Keller F, Baumert TF. Neutralizing antibodies and pathogenesis of hepatitis C virus infection. Viruses 2012. [PMID: 23202451 PMCID: PMC3497039 DOI: 10.3390/v4102016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease worldwide. The interplay between the virus and host innate and adaptive immune responses determines the outcome of infection. There is increasing evidence that host neutralizing responses play a relevant role in the resulting pathogenesis. Furthermore, viral evasion from host neutralizing antibodies has been revealed to be an important contributor in leading both to viral persistence in acute liver graft infection following liver transplantation, and to chronic viral infection. The development of novel model systems to study HCV entry and neutralization has allowed a detailed understanding of the molecular mechanisms of virus-host interactions during antibody-mediated neutralization. The understanding of these mechanisms will ultimately contribute to the development of novel antiviral preventive strategies for liver graft infection and an urgently needed vaccine. This review summarizes recent concepts of the role of neutralizing antibodies in viral clearance and protection, and highlights consequences of viral escape from neutralizing antibodies in the pathogenesis of HCV infection.
Collapse
Affiliation(s)
- Samira Fafi-Kremer
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Catherine Fauvelle
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
| | - Daniel J. Felmlee
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
| | - Mirjam B. Zeisel
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
| | - Quentin Lepiller
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Isabel Fofana
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
| | - Laura Heydmann
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
| | - Françoise Stoll-Keller
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U748, Strasbourg, France ; (S.F.-K.); (C.F.); (D.J.F.); (M.B.Z.); (Q.L.); (I.F.); (L.H.); (F.S.-K.)
- Université de Strasbourg, Strasbourg, France
- Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Author to whom correspondence should be addressed; ; Tel.: +33 3 68 85 37 03; Fax: +33 3 68 85 37 50
| |
Collapse
|
83
|
Reconstitution of the entire hepatitis C virus life cycle in nonhepatic cells. J Virol 2012; 86:11919-25. [PMID: 22896615 DOI: 10.1128/jvi.01066-12] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hepatitis C virus (HCV) is a human hepatotropic virus, but the relevant host factors restricting HCV infection to hepatocytes are only partially understood. We demonstrate that exogenous expression of defined host factors reconstituted the entire HCV life cycle in human nonhepatic 293T cells. This study shows robust HCV entry, RNA replication, and production of infectious virus in human nonhepatic cells and highlights key host factors required for liver tropism of HCV.
Collapse
|
84
|
Hepatitis C virus envelope glycoprotein fitness defines virus population composition following transmission to a new host. J Virol 2012; 86:11956-66. [PMID: 22855498 DOI: 10.1128/jvi.01079-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Genetic variability is a hallmark of RNA virus populations. However, transmission to a new host often results in a marked decrease in population diversity. This genetic bottlenecking is observed during hepatitis C virus (HCV) transmission and can arise via a selective sweep or through the founder effect. To model HCV transmission, we utilized chimeric SCID/Alb-uPA mice with transplanted human hepatocytes and infected them with a human serum HCV inoculum. E1E2 glycoprotein gene sequences in the donor inoculum and recipient mice were determined following single-genome amplification (SGA). In independent experiments, using mice with liver cells grafted from different sources, an E1E2 variant undetectable in the source inoculum was selected for during transmission. Bayesian coalescent analyses indicated that this variant arose in the inoculum pretransmission. Transmitted variants that established initial infection harbored key substitutions in E1E2 outside HVR1. Notably, all posttransmission E1E2s had lost a potential N-linked glycosylation site (PNGS) in E2. In lentiviral pseudoparticle assays, the major posttransmission E1E2 variant conferred an increased capacity for entry compared to the major variant present in the inoculum. Together, these data demonstrate that increased envelope glycoprotein fitness can drive selective outgrowth of minor variants posttransmission and that loss of a PNGS is integral to this improved phenotype. Mathematical modeling of the dynamics of competing HCV variants indicated that relatively modest differences in glycoprotein fitness can result in marked shifts in virus population composition. Overall, these data provide important insights into the dynamics and selection of HCV populations during transmission.
Collapse
|
85
|
Ip PP, Nijman HW, Wilschut J, Daemen T. Therapeutic vaccination against chronic hepatitis C virus infection. Antiviral Res 2012; 96:36-50. [PMID: 22841700 DOI: 10.1016/j.antiviral.2012.07.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 06/25/2012] [Accepted: 07/13/2012] [Indexed: 12/12/2022]
Abstract
Approximately 170 million people worldwide are chronic carriers of Hepatitis C virus (HCV). To date, there is no prophylactic vaccine available against HCV. The standard-of-care therapy for HCV infection involves a combination of pegylated interferon-α and ribavirin. This therapy, which is commonly associated with side effects, has a curative rate varying from 43% (HCV genotype 1) to 80% (HCV genotype 2). In 2011, two direct-acting antiviral agents, telaprevir and boceprevir, were approved by the US Food and drug Administration and are now being used in combination with standard-of-care therapy in selected patients infected with HCV genotype 1. Although both drugs are promising, resulting in a shortening of therapy, these drugs also induce additional side effects and have reduced efficacy in patients who did not respond to standard-of-care previously. An alternative approach would be to treat HCV by stimulating the immune system with a therapeutic vaccine ideally aimed at (i) the eradication of HCV-infected cells and (ii) neutralization of infectious HCV particles. The challenge is to develop therapeutic vaccination strategies that are either at least as effective as antiviral drugs but with lower side effects, or vaccines that, when combined with antiviral drugs, can circumvent long-term use of these drugs thereby reducing their side effects. In this review, we summarize and discuss recent preclinical developments in the area of therapeutic vaccination against chronic HCV infection. Although neutralizing antibodies have been described to exert protective immunity, clinical studies on the induction of neutralizing antibodies in therapeutic settings are limited. Therefore, we will primarily discuss therapeutic vaccines which aim to induce effective cellular immune response against HCV.
Collapse
Affiliation(s)
- Peng Peng Ip
- Department of Medical Microbiology, Molecular Virology Section, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | | | | |
Collapse
|
86
|
FOFANA ISABEL, FAFI–KREMER SAMIRA, CAROLLA PATRIC, FAUVELLE CATHERINE, ZAHID MUHAMMADNAUMAN, TUREK MARINE, HEYDMANN LAURA, CURY KARINE, HAYER JULIETTE, COMBET CHRISTOPHE, COSSET FRANÇOIS, PIETSCHMANN THOMAS, HIET MARIE, BARTENSCHLAGER RALF, HABERSETZER FRANÇOIS, DOFFOËL MICHEL, KECK ZHEN, FOUNG STEVENKH, ZEISEL MIRJAMB, STOLL–KELLER FRANÇOISE, BAUMERT THOMASF. Mutations that alter use of hepatitis C virus cell entry factors mediate escape from neutralizing antibodies. Gastroenterology 2012; 143:223-233.e9. [PMID: 22503792 PMCID: PMC5295797 DOI: 10.1053/j.gastro.2012.04.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 03/14/2012] [Accepted: 04/06/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The development of vaccines and other strategies to prevent hepatitis C virus (HCV) infection is limited by rapid viral evasion. HCV entry is the first step of infection; this process involves several viral and host factors and is targeted by host-neutralizing responses. Although the roles of host factors in HCV entry have been well characterized, their involvement in evasion of immune responses is poorly understood. We used acute infection of liver graft as a model to investigate the molecular mechanisms of viral evasion. METHODS We studied factors that contribute to evasion of host immune responses using patient-derived antibodies, HCV pseudoparticles, and cell culture-derived HCV that express viral envelopes from patients who have undergone liver transplantation. These viruses were used to infect hepatoma cell lines that express different levels of HCV entry factors. RESULTS By using reverse genetic analyses, we identified altered use of host-cell entry factors as a mechanism by which HCV evades host immune responses. Mutations that alter use of the CD81 receptor also allowed the virus to escape neutralizing antibodies. Kinetic studies showed that these mutations affect virus-antibody interactions during postbinding steps of the HCV entry process. Functional studies with a large panel of patient-derived antibodies showed that this mechanism mediates viral escape, leading to persistent infection in general. CONCLUSIONS We identified a mechanism by which HCV evades host immune responses, in which use of cell entry factors evolves with escape from neutralizing antibodies. These findings advance our understanding of the pathogenesis of HCV infection and might be used to develop antiviral strategies and vaccines.
Collapse
Affiliation(s)
- ISABEL FOFANA
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | - SAMIRA FAFI–KREMER
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
,Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - PATRIC CAROLLA
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | - CATHERINE FAUVELLE
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | | | - MARINE TUREK
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | - LAURA HEYDMANN
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | - KARINE CURY
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | - JULIETTE HAYER
- Bases Moléculaires et Structurales des Systèmes Infectieux, UMR 5086, Centre National de la Recherche Scientifique, Université de Lyon, Institut de Biologie et Chimie des Proteines, Lyon, France
| | - CHRISTOPHE COMBET
- Bases Moléculaires et Structurales des Systèmes Infectieux, UMR 5086, Centre National de la Recherche Scientifique, Université de Lyon, Institut de Biologie et Chimie des Proteines, Lyon, France
| | - FRANÇOIS–LOÏC COSSET
- Université de Lyon, Université Claude Bernard Lyon1, IFR 128, Inserm U758; Ecole Normale Supérieure de Lyon, 69364 Lyon, France
| | - THOMAS PIETSCHMANN
- Division of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - MARIE–SOPHIE HIET
- The Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - RALF BARTENSCHLAGER
- The Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - FRANÇOIS HABERSETZER
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
,Pôle Hepato-Digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - MICHEL DOFFOËL
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
,Pôle Hepato-Digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - ZHEN–YONG KECK
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - STEVEN K. H. FOUNG
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - MIRJAM B. ZEISEL
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
| | - FRANÇOISE STOLL–KELLER
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
,Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - THOMAS F. BAUMERT
- Inserm, U748, Strasbourg, France
,Université de Strasbourg, Strasbourg, France
,Pôle Hepato-Digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
87
|
Lacek K, Vercauteren K, Grzyb K, Naddeo M, Verhoye L, Słowikowski MP, Fafi-Kremer S, Patel AH, Baumert TF, Folgori A, Leroux-Roels G, Cortese R, Meuleman P, Nicosia A. Novel human SR-BI antibodies prevent infection and dissemination of HCV in vitro and in humanized mice. J Hepatol 2012; 57:17-23. [PMID: 22414763 DOI: 10.1016/j.jhep.2012.02.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/09/2012] [Accepted: 02/01/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV)-induced end-stage liver disease is currently the major indication for liver transplantation in the Western world. After transplantation, the donor liver almost inevitably becomes infected by the circulating virus and disease progression is accelerated in immune suppressed transplant patients. The current standard therapy, based on pegylated interferon and ribavirin, induces severe side effects and is often ineffective in this population. Therefore, new strategies to prevent graft re-infection are urgently needed. We have previously shown that monoclonal antibodies (mAbs) against the HCV co-receptor scavenger receptor class B type I (SR-BI/Cla1) inhibit infection by different HCV genotypes in cell culture. METHODS Using phage display libraries, we have generated a large set of novel human mAbs against SR-BI and evaluated their effectiveness in preventing HCV infection and direct cell-to-cell spread in vitro and in vivo using uPA-SCID mice with a humanized liver. RESULTS Eleven human monoclonal antibodies were generated that specifically recognize SR-BI. Two antibodies, mAb8 and mAb151, displayed the highest binding and inhibitory properties and also interfered with direct cell-to-cell spread in vitro. Studies in humanized mice showed that both antibodies were capable of preventing HCV infection and could block intrahepatic spread and virus amplification when administered 3 days after infection. Interestingly, anti-SR-BI therapy was effective against an HCV variant that escaped the control of the adaptive immune response in a liver transplant patient. CONCLUSIONS The anti-SR-BI mAbs generated in this study may represent novel therapeutic tools to prevent HCV re-infection of liver allografts.
Collapse
|
88
|
Abstract
Evolution of RNA viruses occurs through disequilibria of collections of closely related mutant spectra or mutant clouds termed viral quasispecies. Here we review the origin of the quasispecies concept and some biological implications of quasispecies dynamics. Two main aspects are addressed: (i) mutant clouds as reservoirs of phenotypic variants for virus adaptability and (ii) the internal interactions that are established within mutant spectra that render a virus ensemble the unit of selection. The understanding of viruses as quasispecies has led to new antiviral designs, such as lethal mutagenesis, whose aim is to drive viruses toward low fitness values with limited chances of fitness recovery. The impact of quasispecies for three salient human pathogens, human immunodeficiency virus and the hepatitis B and C viruses, is reviewed, with emphasis on antiviral treatment strategies. Finally, extensions of quasispecies to nonviral systems are briefly mentioned to emphasize the broad applicability of quasispecies theory.
Collapse
Affiliation(s)
- Esteban Domingo
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), C/ Nicolás Cabrera, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.
| | | | | |
Collapse
|
89
|
Vercauteren K, Leroux-Roels G, Meuleman P. Blocking HCV entry as potential antiviral therapy. Future Virol 2012. [DOI: 10.2217/fvl.12.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
90
|
Unexpected maintenance of hepatitis C viral diversity following liver transplantation. J Virol 2012; 86:8432-9. [PMID: 22623804 DOI: 10.1128/jvi.00749-12] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection can lead to liver cirrhosis in up to 20% of individuals, often requiring liver transplantation. Although the new liver is known to be rapidly reinfected, the dynamics and source of the reinfecting virus(es) are unclear, resulting in some confusion concerning the relationship between clinical outcome and viral characteristics. To clarify the dynamics of liver reinfection, longitudinal serum viral samples from 10 transplant patients were studied. Part of the E1/E2 region was sequenced, and advanced phylogenetic analysis methods were used in a multiparameter analysis to determine the history and ancestry of reinfecting lineages. Our results demonstrated the complexity of HCV evolutionary dynamics after liver transplantation, in which a large diverse population of viruses is transmitted and maintained for months to years. As many as 30 independent lineages in a single patient were found to reinfect the new liver. Several later posttransplant lineages were more closely related to older pretransplant viruses than to viruses detected immediately after transplantation. Although our data are consistent with a number of interpretations, the persistence of high viral genetic variation over long periods of time requires an active mechanism. We discuss possible scenarios, including frequency-dependent selection or variation in selective pressure among viral subpopulations, i.e., the population structure. The latter hypothesis, if correct, could have relevance to the success of newer direct-acting antiviral therapies.
Collapse
|
91
|
Chinnadurai R, Velazquez V, Grakoui A. Hepatic transplant and HCV: a new playground for an old virus. Am J Transplant 2012; 12:298-305. [PMID: 22044693 DOI: 10.1111/j.1600-6143.2011.03812.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hepatitis C virus (HCV) infection is a major global health problem affecting 170 million people worldwide. The majority of infected individuals fail to resolve their infection, with a significant number developing chronic, progressive HCV-related liver disease. HCV infection is the leading indication for liver transplantation and unfortunately, all patients with detectable viral load before transplantation will have rapid, recurrent infection. What remain to be determined are factors contributing to the severity of HCV recurrence. Such factors are unique to the posttransplant setting and include: viral genetic diversity and composition, immunosuppression, donor/recipient age and sex, genetic factors and the liver microenvironment. Importantly, the possibility that the severity of HCV recurrence might be also influenced by factors related to the primary course of disease (i.e. viral set point, previously acquired adaptations of the virus) must be further evaluated. In this sense, recurrent HCV infection should not be regarded merely as another acute infection, but rather, it should be cautioned that problems first arising during the primary course of disease may be accentuated during recurrence. Development of novel therapeutic approaches will require a thorough understanding of viral and host determinants of infection resolution and how these factors may change in the posttransplant setting.
Collapse
Affiliation(s)
- R Chinnadurai
- Department of Medicine, Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | | | | |
Collapse
|
92
|
Meuleman P, Catanese MT, Verhoye L, Desombere I, Farhoudi A, Jones CT, Sheahan T, Grzyb K, Cortese R, Rice CM, Leroux-Roels G, Nicosia A. A human monoclonal antibody targeting scavenger receptor class B type I precludes hepatitis C virus infection and viral spread in vitro and in vivo. Hepatology 2012; 55:364-72. [PMID: 21953761 PMCID: PMC3262867 DOI: 10.1002/hep.24692] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 09/14/2011] [Indexed: 12/21/2022]
Abstract
UNLABELLED Endstage liver disease caused by chronic hepatitis C virus (HCV) infection is the leading indication for liver transplantation in the Western world. However, immediate reinfection of the grafted donor liver by circulating virus is inevitable and liver disease progresses much faster than the original disease. Standard antiviral therapy is not well tolerated and usually ineffective in liver transplant patients, whereas anti-HCV immunotherapy is hampered by the extreme genetic diversity of the virus and its ability to spread by way of cell-cell contacts. We generated a human monoclonal antibody against scavenger receptor class B type I (SR-BI), monoclonal antibody (mAb)16-71, which can efficiently prevent infection of Huh-7.5 hepatoma cells and primary hepatocytes by cell-culture-derived HCV (HCVcc). Using an Huh7.5 coculture system we demonstrated that mAb16-71 interferes with direct cell-to-cell transmission of HCV. Finally we evaluated the in vivo efficacy of mAb16-71 in "human liver urokinase-type plasminogen activator, severe combined immune deficiency (uPA-SCID) mice" (chimeric mice). A 2-week anti-SR-BI therapy that was initiated 1 day before viral inoculation completely protected all chimeric mice from infection with serum-derived HCV of different genotypes. Moreover, a 9-day postexposure therapy that was initiated 3 days after viral inoculation (when viremia was already observed in the animals) suppressed the rapid viral spread observed in untreated control animals. After cessation of anti-SR-BI-specific antibody therapy, a rise of the viral load was observed. CONCLUSION Using in vitro cell culture and human liver-chimeric mouse models, we show that a human mAb targeting the HCV coreceptor SR-BI completely prevents infection and intrahepatic spread of multiple HCV genotypes. This strategy may be an efficacious way to prevent infection of allografts following liver transplantation in chronic HCV patients, and may even hold promise for the prevention of virus rebound during or following antiviral therapy.
Collapse
Affiliation(s)
- Philip Meuleman
- Center for Vaccinology, Ghent University and Hospital, Gent, Belgium.
| | | | - Lieven Verhoye
- Center for Vaccinology, Ghent University and Hospital, Gent, Belgium
| | | | - Ali Farhoudi
- Center for Vaccinology, Ghent University and Hospital, Gent, Belgium
| | - Christopher T. Jones
- Center for the study of Hepatitis C, The Rockefeller University, New York, NY, USA
| | - Timothy Sheahan
- Center for the study of Hepatitis C, The Rockefeller University, New York, NY, USA
| | | | | | - Charles M Rice
- Center for the study of Hepatitis C, The Rockefeller University, New York, NY, USA
| | | | | |
Collapse
|
93
|
Abstract
Hepatitis C virus (HCV) infection is a major cause for liver transplantation worldwide. Still, HCV re-infection of the graft occurs in almost all cases. Most liver transplant recipients experience episodes of graft hepatitis associated with fibrosis progression and graft failure. Clinical management of graft hepatitis can be challenging as in addition to rejection and HCV-induced hepatitis various other factors might be involved including toxic liver injury, steatohepatitis, ischaemic bile duct lesions or infections with other pathogens. Treatment options are often contradictory for different causes of graft hepatitis, and the role of distinct immunosuppressive drugs has been discussed controversially. Corticosteroids increase the infectivity of HCV by altering expression levels of entry factors and other immunosuppressive agents may have diverse effects on HCV replication and fibrosis progression. Interferon alpha-therapy of hepatitis C shows limited efficacy and tolerability in liver transplant recipients and may also cause rejection. In this review we summarize the current knowledge on mechanisms of liver injury in post-transplant hepatitis C, discuss the pros and cons of immunosuppressive agents in this specific setting and describe potential novel approaches to prevent HCV reinfection.
Collapse
Affiliation(s)
- S Ciesek
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
94
|
Naturally occurring antibodies that recognize linear epitopes in the amino terminus of the hepatitis C virus E2 protein confer noninterfering, additive neutralization. J Virol 2011; 86:2739-49. [PMID: 22171278 DOI: 10.1128/jvi.06492-11] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic hepatitis C virus (HCV) infection can persist even in the presence of a broadly neutralizing antibody response. Various mechanisms that underpin viral persistence have been proposed, and one of the most recently proposed mechanisms is the presence of interfering antibodies that negate neutralizing responses. Specifically, it has been proposed that antibodies targeting broadly neutralizing epitopes located within a region of E2 encompassing residues 412 to 423 can be inhibited by nonneutralizing antibodies binding to a less conserved region encompassing residues 434 to 446. To investigate this phenomenon, we characterized the neutralizing and inhibitory effects of human-derived affinity-purified immunoglobulin fractions and murine monoclonal antibodies and show that antibodies to both regions neutralize HCV pseudoparticle (HCVpp) and cell culture-infectious virus (HCVcc) infection albeit with different breadths and potencies. Epitope mapping revealed the presence of overlapping but distinct epitopes in both regions, which may explain the observed differences in neutralizing phenotypes. Crucially, we failed to demonstrate any inhibition between these two groups of antibodies, suggesting that interference by nonneutralizing antibodies, at least for the region encompassing residues 434 to 446, does not provide a mechanism for HCV persistence in chronically infected individuals.
Collapse
|
95
|
Padmanabhan P, Dixit NM. Mathematical model of viral kinetics in vitro estimates the number of E2-CD81 complexes necessary for hepatitis C virus entry. PLoS Comput Biol 2011; 7:e1002307. [PMID: 22174670 PMCID: PMC3234214 DOI: 10.1371/journal.pcbi.1002307] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Accepted: 10/31/2011] [Indexed: 02/07/2023] Open
Abstract
Interaction between the hepatitis C virus (HCV) envelope protein E2 and the host receptor CD81 is essential for HCV entry into target cells. The number of E2-CD81 complexes necessary for HCV entry has remained difficult to estimate experimentally. Using the recently developed cell culture systems that allow persistent HCV infection in vitro, the dependence of HCV entry and kinetics on CD81 expression has been measured. We reasoned that analysis of the latter experiments using a mathematical model of viral kinetics may yield estimates of the number of E2-CD81 complexes necessary for HCV entry. Here, we constructed a mathematical model of HCV viral kinetics in vitro, in which we accounted explicitly for the dependence of HCV entry on CD81 expression. Model predictions of viral kinetics are in quantitative agreement with experimental observations. Specifically, our model predicts triphasic viral kinetics in vitro, where the first phase is characterized by cell proliferation, the second by the infection of susceptible cells and the third by the growth of cells refractory to infection. By fitting model predictions to the above data, we were able to estimate the threshold number of E2-CD81 complexes necessary for HCV entry into human hepatoma-derived cells. We found that depending on the E2-CD81 binding affinity, between 1 and 13 E2-CD81 complexes are necessary for HCV entry. With this estimate, our model captured data from independent experiments that employed different HCV clones and cells with distinct CD81 expression levels, indicating that the estimate is robust. Our study thus quantifies the molecular requirements of HCV entry and suggests guidelines for intervention strategies that target the E2-CD81 interaction. Further, our model presents a framework for quantitative analyses of cell culture studies now extensively employed to investigate HCV infection. The interaction between the hepatitis C virus (HCV) envelope protein E2 and the host cell surface receptor CD81 is critical for HCV entry into hepatocytes and presents a promising drug and vaccine target. Yet, the number of E2-CD81 complexes that must be formed between a virus and a target cell to enable viral entry remains unknown. Direct observation of the E2-CD81 complexes preceding viral entry has not been possible. We constructed a mathematical model of HCV viral kinetics in vitro and using it to analyze data from recent cell culture studies obtained estimates of the threshold number of E2-CD81 complexes necessary for HCV entry. We found that depending on the E2-CD81 binding affinity, between 1 and 13 complexes are necessary for HCV entry into human hepatoma-derived cells. Our study thus presents new, quantitative insights into the molecular requirements of HCV entry, which may serve as a guideline for intervention strategies targeting the E2-CD81 interaction. Further, our study shows that HCV viral kinetics in vitro can be described using a mathematical model, thus facilitating quantitative analyses of the wealth of data now emanating from cell culture studies of HCV infection.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| | - Narendra M. Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
- Bioinformatics Centre, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
96
|
Wang Y, Keck ZY, Foung SKH. Neutralizing antibody response to hepatitis C virus. Viruses 2011; 3:2127-45. [PMID: 22163337 PMCID: PMC3230844 DOI: 10.3390/v3112127] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/18/2011] [Accepted: 10/22/2011] [Indexed: 12/14/2022] Open
Abstract
A critical first step in a "rational vaccine design" approach for hepatitis C virus (HCV) is to identify the most relevant mechanisms of immune protection. Emerging evidence provides support for a protective role of virus neutralizing antibodies, and the ability of the B cell response to modify the course of acute HCV infection. This has been made possible by the development of in vitro cell culture models, based on HCV retroviral pseudotype particles expressing E1E2 and infectious cell culture-derived HCV virions, and small animal models that are robust tools in studies of antibody-mediated virus neutralization. This review is focused on the immunogenic determinants on the E2 glycoprotein mediating virus neutralization and the pathways in which the virus is able to escape from immune containment. Encouraging findings from recent studies provide support for the existence of broadly neutralization antibodies that are not associated with virus escape. The identification of conserved epitopes mediating virus neutralization that are not associated with virus escape will facilitate the design of a vaccine immunogen capable of eliciting broadly neutralizing antibodies against this highly diverse virus.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
97
|
Edwards VC, Tarr AW, Urbanowicz RA, Ball JK. The role of neutralizing antibodies in hepatitis C virus infection. J Gen Virol 2011; 93:1-19. [PMID: 22049091 DOI: 10.1099/vir.0.035956-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Hepatitis C virus (HCV) is a blood-borne virus estimated to infect around 170 million people worldwide and is, therefore, a major disease burden. In some individuals the virus is spontaneously cleared during the acute phase of infection, whilst in others a persistent infection ensues. Of those persistently infected, severe liver diseases such as cirrhosis and primary liver cancer may develop, although many individuals remain asymptomatic. A range of factors shape the course of HCV infection, not least host genetic polymorphisms and host immunity. A number of studies have shown that neutralizing antibodies (nAb) arise during HCV infection, but that these antibodies differ in their breadth and mechanism of neutralization. Recent studies, using both mAbs and polyclonal sera, have provided an insight into neutralizing determinants and the likely protective role of antibodies during infection. This understanding has helped to shape our knowledge of the overall structure of the HCV envelope glycoproteins--the natural target for nAb. Most nAb identified to date target receptor-binding sites within the envelope glycoprotein E2. However, there is some evidence that other viral epitopes may be targets for antibody neutralization, suggesting the need to broaden the search for neutralization epitopes beyond E2. This review provides a comprehensive overview of our current understanding of the role played by nAb in HCV infection and disease outcome and explores the limitations in the study systems currently used. In addition, we briefly discuss the potential therapeutic benefits of nAb and efforts to develop nAb-based therapies.
Collapse
Affiliation(s)
- Victoria C Edwards
- School of Molecular Medical Sciences and The Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Alexander W Tarr
- School of Molecular Medical Sciences and The Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Richard A Urbanowicz
- School of Molecular Medical Sciences and The Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Jonathan K Ball
- School of Molecular Medical Sciences and The Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
98
|
Zeisel MB, Da Costa D, Baumert TF. Opening the door for hepatitis C virus infection in genetically humanized mice. Hepatology 2011; 54:1873-5. [PMID: 22038788 DOI: 10.1002/hep.24603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- Mirjam B Zeisel
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 748, Strasbourg, France
| | | | | |
Collapse
|
99
|
Meunier JC, Bukh J, Diaz G, Tovo PA, Casadei AM, Quinti I, Iorio R, Emerson S, Purcell RH, Farci P. Neutralizing antibodies to hepatitis C virus in perinatally infected children followed up prospectively. J Infect Dis 2011; 204:1741-5. [PMID: 22006993 DOI: 10.1093/infdis/jir631] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Little is known about the presence and role of neutralizing antibodies (NtAbs) in perinatal hepatitis C virus (HCV) infection. Using HCV pseudoparticles, NtAbs were studied longitudinally in 12 HCV-infected children with or without evidence of acute hepatitis during the first year of life. Broadly reactive NtAbs of maternal origin did not prevent vertical HCV transmission or progression to chronicity. NtAbs against homologous genotype or subtype appeared during the chronic phase and were more abundant and sustained in children with acute hepatitis. Cross-reactive NtAbs were present in both groups of children, but their appearance did not correlate with better control of viremia or HCV clearance.
Collapse
Affiliation(s)
- Jean-Christophe Meunier
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Wang W, Lin J, Tan D, Xu Y, Brunt EM, Fan X, Di Bisceglie AM. Divergent quasispecies evolution in de novo hepatitis C virus infection associated with bone marrow transplantation. Biochem Biophys Res Commun 2011; 414:148-52. [PMID: 21945614 PMCID: PMC3196000 DOI: 10.1016/j.bbrc.2011.09.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 09/08/2011] [Indexed: 01/27/2023]
Abstract
Quasispecies is a remarkable characteristic of hepatitis C virus (HCV) and has profound roles in HCV biology and clinical practice. The understanding of HCV quasispecies behavior, in particular in acute HCV infection, is valuable for vaccine development and therapeutic interference. However, acute HCV infection is seldom encountered in clinic practice due to its silent onset. In the present study, we reported a unique case of de novo HCV infection associated with the transplantation of bone marrow from a HCV-positive donor. HCV quasispecies diversity was determined in both the donor and the recipient over a 4-year follow-up, accompanied with simultaneous measurement of HCV neutralizing antibody. Detailed genetic and phylogenetic analyses revealed a divergent quasispecies evolution, which was not related to dynamic changes of HCV neutralizing antibody. Instead, our data suggested an essential role of the fitness adaptation of founder viral population in driving such an evolutionary pattern.
Collapse
Affiliation(s)
- Weihua Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
- Wuhan Center for Tuberculosis Control, Wuhan 430030, Hubei, China
| | - Jianguo Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - De Tan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Yanjuan Xu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Elizabeth M. Brunt
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Xiaofeng Fan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
- Saint Louis University Liver Center, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | - Adrian M. Di Bisceglie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
- Saint Louis University Liver Center, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| |
Collapse
|