1
|
Tscherne A, Kalodimou G, Kupke A, Rohde C, Freudenstein A, Jany S, Kumar S, Sutter G, Krähling V, Becker S, Volz A. Rapid Development of Modified Vaccinia Virus Ankara (MVA)-Based Vaccine Candidates Against Marburg Virus Suitable for Clinical Use in Humans. Vaccines (Basel) 2024; 12:1316. [PMID: 39771978 PMCID: PMC11680136 DOI: 10.3390/vaccines12121316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Marburg virus (MARV) is the etiological agent of Marburg Virus Disease (MVD), a rare but severe hemorrhagic fever disease with high case fatality rates in humans. Smaller outbreaks have frequently been reported in countries in Africa over the last few years, and confirmed human cases outside Africa are, so far, exclusively imported by returning travelers. Over the previous years, MARV has also spread to non-endemic African countries, demonstrating its potential to cause epidemics. Although MARV-specific vaccines are evaluated in preclinical and clinical research, none have been approved for human use. Modified Vaccinia virus Ankara (MVA), a well-established viral vector used to generate vaccines against emerging pathogens, can deliver multiple antigens and has a remarkable clinical safety and immunogenicity record, further supporting its evaluation as a vaccine against MARV. The rapid availability of safe and effective MVA-MARV vaccine candidates would expand the possibilities of multi-factored intervention strategies in endemic countries. METHODS We have used an optimized methodology to rapidly generate and characterize recombinant MVA candidate vaccines that meet the quality requirements to proceed to human clinical trials. As a proof-of-concept for the optimized methodology, we generated two recombinant MVAs that deliver either the MARV glycoprotein (MVA-MARV-GP) or the MARV nucleoprotein (MVA-MARV-NP). RESULTS Infections of human cell cultures with recombinant MVA-MARV-GP and MVA-MARV-NP confirmed the efficient synthesis of MARV-GP and MARV-NP proteins in mammalian cells, which are non-permissive for MVA replication. Prime-boost immunizations in C57BL/6J mice readily induced circulating serum antibodies binding to recombinant MARV-GP and MARV-NP proteins. Moreover, the MVA-MARV-candidate vaccines elicited MARV-specific T-cell responses in C57BL/6J mice. CONCLUSIONS We confirmed the suitability of our two backbone viruses MVA-mCherry and MVA-GFP in a proof-of-concept study to rapidly generate candidate vaccines against MARV. However, further studies are warranted to characterize the protective efficacy of these recombinant MVA-MARV vaccines in other preclinical models and to evaluate them as vaccine candidates in humans.
Collapse
Affiliation(s)
- Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany
| | - Alexandra Kupke
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Cornelius Rohde
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
| | - Sylvia Jany
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
| | - Satendra Kumar
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- German Center for Infection Research, Partner Site Hannover-Braunschweig, 30559 Hannover, Germany
| |
Collapse
|
2
|
Vijayasimha K, Leestemaker-Palmer AL, Gibbs JS, Yewdell JW, Dolan BP. MLN4924 Inhibits Defective Ribosomal Product Antigen Presentation Independently of Direct NEDDylation of Protein Antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2273-2282. [PMID: 35428693 PMCID: PMC9288214 DOI: 10.4049/jimmunol.2100584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 03/01/2022] [Indexed: 05/17/2023]
Abstract
Successful direct MHC class I Ag presentation is dependent on the protein degradation machinery of the cell to generate antigenic peptides that can be loaded onto MHC class I molecules for surveillance by CD8+ T cells of the immune system. Most often this process involves the ubiquitin (Ub)-proteasome system; however, other Ub-like proteins have also been implicated in protein degradation and direct Ag presentation. In this article, we examine the role of neuronal precursor cell-expressed developmentally downregulated protein 8 (NEDD8) in direct Ag presentation in mouse cells. NEDD8 is the Ub-like protein with highest similarity to Ub, and fusion of NEDD8 to the N terminus of a target protein can lead to the degradation of target proteins. We find that appending NEDD8 to the N terminus of the model Ag OVA resulted in degradation by both the proteasome and the autophagy protein degradation pathways, but only proteasomal degradation, involving the proteasomal subunit NEDD8 ultimate buster 1, resulted in peptide presentation. When directly compared with Ub, NEDD8 fusion was less efficient at generating peptides. However, inactivation of the NEDD8-conugation machinery by treating cells with MLN4924 inhibited the presentation of peptides from the defective ribosomal product-derived form of a model Ag. These results demonstrate that NEDD8 activity in the cell is important for direct Ag presentation, but not by directly targeting proteins for degradation.
Collapse
Affiliation(s)
- Kartikeya Vijayasimha
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR; and
| | - Amy L Leestemaker-Palmer
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR; and
| | - James S Gibbs
- Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, Bethesda, MD
| | - Jonathan W Yewdell
- Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, Bethesda, MD
| | - Brian P Dolan
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR; and
| |
Collapse
|
3
|
Marín-López A, Utrilla-Trigo S, Jiménez-Cabello L, Ortego J. Recombinant Modified Vaccinia Virus Ankara Development to Express VP2, NS1, and VP7 Proteins of Bluetongue Virus. Methods Mol Biol 2022; 2465:177-193. [PMID: 35118622 DOI: 10.1007/978-1-0716-2168-4_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Modified vaccinia virus Ankara (MVA) is employed widely as an experimental vaccine vector for its abortive replication in mammalian cells and high expression level of foreign/heterologous genes. Recombinant MVAs (rMVAs) are used as platforms for protein production as well as vectors to generate vaccines against a wide range of infectious diseases and other pathologies. The portrait of the virus combines desirable elements such as high-level biological safety, the ability to activate appropriate innate immune mediators upon vaccination , and the capacity to deliver substantial amounts of heterologous antigens. rMVAs encoding proteins of Bluetongue virus (BTV), an orbivirus that infects domestic and wild ruminants through transmission by biting midges of the Culicoides species, are excellent vaccine candidates against this virus. In this chapter, we describe the methods for the generation of rMVAs encoding VP2, NS1, and VP7 proteins of BTV . The included protocols cover the cloning of VP2, NS1, and VP7 BTV-4 genes in a transfer plasmid, the construction of rMVAs, the titration of virus working stocks, and the protein expression analysis by immunofluorescence and radiolabeling of rMVA infected cells as well as virus purification procedure.
Collapse
Affiliation(s)
- Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Javier Ortego
- Centro de Investigación en Sanidad Animal (CISA), INIA-CSIC, Madrid, Spain.
| |
Collapse
|
4
|
Shenderov E, Kandasamy M, Gileadi U, Chen J, Shepherd D, Gibbs J, Prota G, Silk JD, Yewdell JW, Cerundolo V. Generation and characterization of HLA-A2 transgenic mice expressing the human TCR 1G4 specific for the HLA-A2 restricted NY-ESO-1 157-165 tumor-specific peptide. J Immunother Cancer 2021; 9:jitc-2021-002544. [PMID: 34088742 PMCID: PMC8183295 DOI: 10.1136/jitc-2021-002544] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2021] [Indexed: 01/07/2023] Open
Abstract
Background NY-ESO-1 is a tumor-specific, highly immunogenic, human germ cell antigen of the MAGE-1 family that is a promising vaccine and cell therapy candidate in clinical trial development. The mouse genome does not encode an NY-ESO-1 homolog thereby not subjecting transgenic T-cells to thymic tolerance mechanisms that might impair in-vivo studies. We hypothesized that an NY-ESO-1 T cell receptor (TCR) transgenic mouse would provide the unique opportunity to study avidity of TCR response against NY-ESO-1 for tumor vaccine and cellular therapy development against this clinically relevant and physiological human antigen. Methods To study in vitro and in vivo the requirements for shaping an effective T cell response against the clinically relevant NY-ESO-1, we generated a C57BL/6 HLA-A*0201 background TCR transgenic mouse encoding the 1G4 TCR specific for the human HLA-A2 restricted, NY-ESO-1157-165 SLLMWITQC (9C), initially identified in an NY-ESO-1 positive melanoma patient. Results The HLA-A*0201 restricted TCR was positively selected on both CD4+ and CD8+ cells. Mouse 1G4 T cells were not activated by endogenous autoimmune targets or a large library of non-cognate viral antigens. In contrast, their activation by HLA-A2 NY-ESO-1157-165 complexes was evident by proliferation, CD69 upregulation, interferon-γ production, and interleukin-2 production, and could be tuned using a twofold higher affinity altered peptide ligand, NY-ESO-1157-165V. NY-ESO-1157-165V recombinant vaccination of syngeneic mice adoptively transferred with m1G4 CD8+ T cells controlled tumor growth in vivo. 1G4 transgenic mice suppressed growth of syngeneic methylcholanthrene (MCA) induced HHD tumor cells expressing the full-length human NY-ESO-1 protein but not MCA HHD tumor cells lacking NY-ESO-1. Conclusions The 1G4 TCR mouse model for the physiological human TCR against the clinically relevant antigen, NY-ESO-1, is a valuable tool with the potential to accelerate clinical development of NY-ESO-1-targeted T-cell and vaccine therapies.
Collapse
Affiliation(s)
- Eugene Shenderov
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK .,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Matheswaran Kandasamy
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Jili Chen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Dawn Shepherd
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - James Gibbs
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Jonathan D Silk
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK.,Next Generation Research, Adaptimmune, Abingdon, UK
| | - Jonathan W Yewdell
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, UK
| |
Collapse
|
5
|
Matía A, Lorenzo MM, Blasco R. Tools for the targeted genetic modification of poxvirus genomes. Curr Opin Virol 2020; 44:183-190. [PMID: 33242829 DOI: 10.1016/j.coviro.2020.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
The potential of viruses as biotechnology platforms is becoming more appealing due to technological advances in synthetic biology techniques and to the increasing accessibility of means to manipulate virus genomes. Among viral systems, poxviruses, and their prototype member Vaccinia Virus, are one of the outstanding choices for different biotechnological and medical applications based on heterologous gene expression, recombinant vaccines or oncolytic viruses. The refinement of genetic engineering methods on Vaccinia Virus over the last decades have contributed to facilitate the manipulation of the genomes of poxviruses, and may aid in the improvement of virus variants designed for different goals through reverse genetic approaches. Targeted genetic changes are usually performed by homologous recombination with the viral genome. In addition to the classic approach, recent methodological advances that may assist new strategies for the mutation or edition of poxvirus genomes are reviewed.
Collapse
Affiliation(s)
- Alejandro Matía
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain
| | - María M Lorenzo
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain
| | - Rafael Blasco
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain.
| |
Collapse
|
6
|
Rapid poxvirus engineering using CRISPR/Cas9 as a selection tool. Commun Biol 2020; 3:643. [PMID: 33144673 PMCID: PMC7641209 DOI: 10.1038/s42003-020-01374-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/01/2020] [Indexed: 02/08/2023] Open
Abstract
In standard uses of CRISPR/Cas9 technology, the cutting of genomes and their efficient repair are considered to go hand-in-hand to achieve desired genetic changes. This includes the current approach for engineering genomes of large dsDNA viruses. However, for poxviruses we show that Cas9-guide RNA complexes cut viral genomes soon after their entry into cells, but repair of these breaks is inefficient. As a result, Cas9 targeting makes only modest, if any, improvements to basal rates of homologous recombination between repair constructs and poxvirus genomes. Instead, Cas9 cleavage leads to inhibition of poxvirus DNA replication thereby suppressing virus spread in culture. This unexpected outcome allows Cas9 to be used as a powerful tool for selecting conventionally generated poxvirus recombinants, which are otherwise impossible to separate from a large background of parental virus without the use of marker genes. This application of CRISPR/Cas9 greatly speeds up the generation of poxvirus-based vaccines, making this platform considerably more attractive in the context of personalised cancer vaccines and emerging disease outbreaks. Gowripalan, Smith et al. use CRISPR/Cas9 technology to rapidly select recombinant poxviruses without using selectable marker genes. They find that Cas9 cleavage inhibits poxvirus DNA replication, suppressing virus spread in culture. This application makes poxviruses more attractive vector platforms for fighting cancer and emerging disease outbreaks.
Collapse
|
7
|
Moss B. Investigating Viruses During the Transformation of Molecular Biology: Part II. Annu Rev Virol 2020; 7:15-36. [PMID: 32392458 DOI: 10.1146/annurev-virology-021020-100558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
My scientific career started at an extraordinary time, shortly after the discoveries of the helical structure of DNA, the central dogma of DNA to RNA to protein, and the genetic code. Part I of this series emphasizes my education and early studies highlighted by the isolation and characterization of numerous vaccinia virus enzymes, determination of the cap structure of messenger RNA, and development of poxviruses as gene expression vectors for use as recombinant vaccines. Here I describe a shift in my research focus to combine molecular biology and genetics for a comprehensive understanding of poxvirus biology. The dominant paradigm during the early years was to select a function, isolate the responsible proteins, and locate the corresponding gene, whereas later the common paradigm was to select a gene, make a mutation, and determine the altered function. Motivations, behind-the-scenes insights, importance of new technologies, and the vital roles of trainees and coworkers are emphasized.
Collapse
Affiliation(s)
- Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
8
|
Raman SC, Mejías-Pérez E, Gomez CE, García-Arriaza J, Perdiguero B, Vijayan A, Pérez-Ruiz M, Cuervo A, Santiago C, Sorzano COS, Sánchez-Corzo C, Moog C, Burger JA, Schorcht A, Sanders RW, Carrascosa JL, Esteban M. The Envelope-Based Fusion Antigen GP120C14K Forming Hexamer-Like Structures Triggers T Cell and Neutralizing Antibody Responses Against HIV-1. Front Immunol 2019; 10:2793. [PMID: 31867001 PMCID: PMC6904342 DOI: 10.3389/fimmu.2019.02793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/14/2019] [Indexed: 11/20/2022] Open
Abstract
There is an urgent need for the development of potent vaccination regimens that are able to induce specific T and B cell responses against human immunodeficiency virus type 1 (HIV-1). Here, we describe the generation and characterization of a fusion antigen comprised of the HIV-1 envelope GP120 glycoprotein from clade C (GP120C) fused at its C-terminus, with the modified vaccinia virus (VACV) 14K protein (A27L gene) (termed GP120C14K). The design is directed toward improving the immunogenicity of the GP120C protein through its oligomerization facilitated by the fused VACV 14K protein that results in hexamer-like structures. Two different immunogens were generated: a recombinant GP120C14K fusion protein (purified from a stable CHO-K1 cell line) and a recombinant modified vaccinia virus Ankara (MVA) poxvirus vector expressing the GP120C14K fusion protein (termed MVA-GP120C14K). The GP120C14K fusion protein is recognized by broadly neutralizing antibodies (bNAbs) against HIV-1. In a murine model, a heterologous prime/boost immunization regimen with MVA-GP120C14K prime followed by adjuvanted GP120C14K protein boost generated stronger and polyfunctional HIV-1 Env-specific CD8 T cell responses when compared with the delivery of the monomeric GP120C form. Furthermore, the immunization protocol MVA-GP120C14K/GP120C14K elicited higher HIV-1 Env-specific T follicular helper cells, germinal center B cells and antibody responses than monomeric GP120. In addition, a similar MVA-GP120C14K prime/GP120C14K protein boost regimen performed in rabbits triggered high HIV-1-Env-specific IgG binding antibody titers that were capable of neutralizing HIV-1 pseudoviruses. The extent of HIV-1 neutralization was comparable to that elicited by the current standard GP140 SOSIP trimers from clades B and C when immunized as MVA-SOSIP prime/SOSIP protein boost regimen. Overall, the novel fusion antigen and the corresponding immunization scheme provided in this report can therefore be considered as potential vaccine strategies against HIV-1.
Collapse
Affiliation(s)
- Suresh C Raman
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Carmen E Gomez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Aneesh Vijayan
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Mar Pérez-Ruiz
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Ana Cuervo
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - César Santiago
- X-ray Crystallization Unit, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Carlos Oscar S Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Cristina Sánchez-Corzo
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Christiane Moog
- INSERM U1109, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Judith A Burger
- Department of Medical Microbiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Anna Schorcht
- Department of Medical Microbiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Rogier W Sanders
- Department of Medical Microbiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, United States
| | - José L Carrascosa
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
9
|
Modified Vaccinia Virus Ankara Can Induce Optimal CD8 + T Cell Responses to Directly Primed Antigens Depending on Vaccine Design. J Virol 2019; 93:JVI.01154-19. [PMID: 31375596 PMCID: PMC6803277 DOI: 10.1128/jvi.01154-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/29/2019] [Indexed: 01/19/2023] Open
Abstract
A variety of strains of vaccinia virus (VACV) have been used as recombinant vaccine vectors with the aim of inducing robust CD8+ T cell immunity. While much of the pioneering work was done with virulent strains, such as Western Reserve (WR), attenuated strains such as modified vaccinia virus Ankara (MVA) are more realistic vectors for clinical use. To unify this literature, side-by-side comparisons of virus strains are required. Here, we compare the form of antigen that supports optimal CD8+ T cell responses for VACV strains WR and MVA using equivalent constructs. We found that for multiple antigens, minimal antigenic constructs (epitope minigenes) that prime CD8+ T cells via the direct presentation pathway elicited optimal responses from both vectors, which was surprising because this finding contradicts the prevailing view in the literature for MVA. We then went on to explore the discrepancy between current and published data for MVA, finding evidence that the expression locus and in some cases the presence of the viral thymidine kinase may influence the ability of this strain to prime optimal responses from antigens that require direct presentation. This extends our knowledge of the design parameters for VACV vectored vaccines, especially those based on MVA.IMPORTANCE Recombinant vaccines based on vaccinia virus and particularly attenuated strains such as MVA are in human clinical trials, but due to the complexity of these large vectors much remains to be understood about the design parameters that alter their immunogenicity. Previous work had found that MVA vectors should be designed to express stable protein in order to induce robust immunity by CD8+ (cytotoxic) T cells. Here, we found that the primacy of stable antigen is not generalizable to all designs of MVA and may depend where a foreign antigen is inserted into the MVA genome. This unexpected finding suggests that there is an interaction between genome location and the best form of antigen for optimal T cell priming in MVA and thus possibly other vaccine vectors. It also highlights that our understanding of antigen presentation by even the best studied of vaccine vectors remains incomplete.
Collapse
|
10
|
Gutjahr A, Papagno L, Nicoli F, Kanuma T, Kuse N, Cabral-Piccin MP, Rochereau N, Gostick E, Lioux T, Perouzel E, Price DA, Takiguchi M, Verrier B, Yamamoto T, Paul S, Appay V. The STING ligand cGAMP potentiates the efficacy of vaccine-induced CD8+ T cells. JCI Insight 2019; 4:125107. [PMID: 30944257 DOI: 10.1172/jci.insight.125107] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/14/2019] [Indexed: 11/17/2022] Open
Abstract
Pathogen recognition receptor (PRR) agonists are currently being developed and tested as adjuvants in various formulations to optimize the immunogenicity and efficacy of vaccines. Using an original in vitro approach to prime naive precursors from unfractionated human peripheral blood mononuclear cells, we assessed the influence of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), a ligand for the stimulator of interferon genes (STING), on the induction of antigen-specific CD8+ T cells. We found that 2'3'-cGAMP and 3'3'-cGAMP were especially potent adjuvants in this system, driving the expansion and maturation of functionally replete antigen-specific CD8+ T cells via the induction of type I IFNs. The biological relevance of these findings was confirmed in vivo using two mouse models, in which 2'3'-cGAMP-adjuvanted vaccination elicited protective antitumor or antiviral CD8+ T cell responses. These results identify particular isoforms of cGAMP as effective adjuvants that may find utility in the development of novel immunotherapies and vaccines.
Collapse
Affiliation(s)
- Alice Gutjahr
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM, Centre d'Investigation Clinique en Vaccinologie 1408, Faculté de Médecine de Saint-Etienne, Saint-Etienne, France.,Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Unité Mixte de Recherche 5305, Université Lyon 1, CNRS, Lyon, France.,InvivoGen, Toulouse, France
| | - Laura Papagno
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Francesco Nicoli
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Tomohiro Kanuma
- Laboratory of Immunosenescence, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Nozomi Kuse
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | | | | | - Emma Gostick
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | | | | | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | | | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Unité Mixte de Recherche 5305, Université Lyon 1, CNRS, Lyon, France
| | - Takuya Yamamoto
- Laboratory of Immunosenescence, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM, Centre d'Investigation Clinique en Vaccinologie 1408, Faculté de Médecine de Saint-Etienne, Saint-Etienne, France
| | - Victor Appay
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
11
|
Marzook NB, Newsome TP. Construction and Isolation of Recombinant Vaccinia Virus Expressing Fluorescent Proteins. Methods Mol Biol 2019; 2023:73-92. [PMID: 31240671 DOI: 10.1007/978-1-4939-9593-6_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vaccinia virus recombinants that express fluorescent proteins have a variety of applications such as the identification of infected cells, efficient screening for genetically modified strains, and molecular characterization of virus replication and spread. The detection of fluorescent proteins and viral-fluorescent fusion proteins by fluorescence microscopy is noninvasive and can be used to describe protein localization in live cells and track the intracellular movement of virus particles. This chapter describes a number of approaches for the construction of plasmids and subsequent generation and isolation of fluorescent recombinant viruses.
Collapse
Affiliation(s)
- N Bishara Marzook
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW, Australia
| | - Timothy P Newsome
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW, Australia.
| |
Collapse
|
12
|
Batf3-Dependent Dendritic Cells Promote Optimal CD8 T Cell Responses against Respiratory Poxvirus Infection. J Virol 2018; 92:JVI.00495-18. [PMID: 29875235 DOI: 10.1128/jvi.00495-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Respiratory infection with vaccinia virus (VacV) elicits robust CD8+ T cell responses that play an important role in host resistance. In the lung, VacV encounters multiple tissue-resident antigen-presenting cell (APC) populations, but which cell plays a dominant role in priming of virus-specific CD8+ effector T cell responses remains poorly defined. We used Batf3-/- mice to investigate the impact of CD103+ and CD8α+ dendritic cell (DC) deficiency on anti-VacV CD8+ T cell responses. We found that Batf3-/- mice were more susceptible to VacV infection, exhibiting profound weight loss, which correlated with impaired accumulation of gamma interferon (IFN-γ)-producing CD8+ T cells in the lungs. This was largely due to defective priming since early in the response, antigen-specific CD8+ T cells in the draining lymph nodes of Batf3-/- mice expressed significantly reduced levels of Ki67, CD25, and T-bet. These results underscore a specific role for Batf3-dependent DCs in regulating priming and expansion of effector CD8+ T cells necessary for host resistance against acute respiratory VacV infection.IMPORTANCE During respiratory infection with vaccinia virus (VacV), a member of Poxviridae family, CD8+ T cells play important role in resolving the primary infection. Effector CD8+ T cells clear the virus by accumulating in the infected lungs in large numbers and secreting molecules such as IFN-γ that kill virally infected cells. However, precise cell types that regulate the generation of effector CD8+ T cells in the lungs are not well defined. Dendritic cells (DCs) are a heterogeneous population of immune cells that are recognized as key initiators and regulators of T-cell-mediated immunity. In this study, we reveal that a specific subset of DCs that are dependent on the transcription factor Batf3 for their development regulate the magnitude of CD8+ T cell effector responses in the lungs, thereby providing protection during pulmonary VacV infection.
Collapse
|
13
|
Abstract
The vaccinia virus protein F13, encoded by the F13L gene, is conserved across the subfamily Chordopoxvirinae and is critical among orthopoxviruses to produce the wrapped form of virus that is required for cell-to-cell spread. F13 is the major envelope protein on the membrane of extracellular forms of virus; however, it is not known if F13 is required in steps postwrapping. In this report, we utilize two temperature-sensitive vaccinia virus mutants from the Condit collection of temperature-sensitive viruses whose small plaque phenotypes have been mapped to the F13L gene. Despite the drastic reduction in plaque size, the temperature-sensitive viruses were found to produce levels of extracellular virions similar to those of the parental strain, Western Reserve (WR), at the permissive and nonpermissive temperatures, suggesting that they are not defective in extracellular virion formation. Analyses of extracellular virions produced by one temperature-sensitive mutant found that those produced at the nonpermissive temperature had undetectable levels of F13 and bound cells with efficiency similar to that of WR but displayed delayed cell entry kinetics. Additionally, low-pH treatment of cells bound by extracellular virions produced at the nonpermissive temperature by the temperature-sensitive reporter virus was unable to overcome a block in infection by bafilomycin A1, suggesting that these virions display increased resistance to dissolution of the extracellular virion envelope. Taken together, our results suggest that F13 plays a role both in the formation of extracellular virions and in the promotion of their rapid entry into cells by enhancing the sensitivity of the membrane to acid-induced dissolution.IMPORTANCE Vaccinia virus (VACV) is an orthopoxvirus and produces two infectious forms, mature virions (MV) and extracellular virions (EV). EV are derived from MV and contain an additional membrane that must first be removed prior to cell entry. F13 is critical for the formation of EV, but a postenvelopment role has not been described. Here, two temperature-sensitive VACV mutants whose deficiencies were previously mapped to the F13L locus are characterized. Both viruses produced EV at the nonpermissive temperature at levels similar to those of a virus that has F13L, yet they had a small plaque phenotype and rate of spread similar to that of an F13L deletion virus. F13 was undetectable on the EV membrane at the nonpermissive temperature, and these EV exhibited delayed cell entry kinetics compared to EV containing F13. This study is the first to conclusively demonstrate a novel role for F13 in cell entry of the EV form of the virus.
Collapse
|
14
|
Manning NM, Bachanek-Bankowska K, Mertens PPC, Castillo-Olivares J. Vaccination with recombinant Modified Vaccinia Ankara (MVA) viruses expressing single African horse sickness virus VP2 antigens induced cross-reactive virus neutralising antibodies (VNAb) in horses when administered in combination. Vaccine 2017; 35:6024-6029. [DOI: 10.1016/j.vaccine.2017.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/27/2017] [Accepted: 04/04/2017] [Indexed: 12/11/2022]
|
15
|
Guo ZS, Liu Z, Sathaiah M, Wang J, Ravindranathan R, Kim E, Huang S, Kenniston TW, Bell JC, Zeh HJ, Butterfield LH, Gambotto A, Bartlett DL. Rapid Generation of Multiple Loci-Engineered Marker-free Poxvirus and Characterization of a Clinical-Grade Oncolytic Vaccinia Virus. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:112-122. [PMID: 29085848 PMCID: PMC5651493 DOI: 10.1016/j.omtm.2017.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
Recombinant poxviruses, utilized as vaccine vectors and oncolytic viruses, often require manipulation at multiple genetic loci in the viral genome. It is essential for viral vectors to possess no adventitious mutations and no (antibiotic) selection marker in the final product for human patients in order to comply with the guidance from the regulatory agencies. Rintoul et al. have previously developed a selectable and excisable marker (SEM) system for the rapid generation of recombinant vaccinia virus. In the current study, we describe an improved methodology for rapid creation and selection of recombinant poxviruses with multiple genetic manipulations solely based on expression of a fluorescent protein and with no requirement for drug selection that can lead to cellular stress and the risk of adventitious mutations throughout the viral genome. Using this improved procedure combined with the SEM system, we have constructed multiple marker-free oncolytic poxviruses expressing different cytokines and other therapeutic genes. The high fidelity of inserted DNA sequences validates the utility of this improved procedure for generation of therapeutic viruses for human patients. We have created an oncolytic poxvirus expressing human chemokine CCL5, designated as vvDD-A34R-hCCL5, with manipulations at two genetic loci in a single virus. Finally, we have produced and purified this virus in clinical grade for its use in a phase I clinical trial and presented data on initial in vitro characterization of the virus.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Corresponding author: Zong Sheng Guo, PhD, UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zuqiang Liu
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Magesh Sathaiah
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiahu Wang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Roshni Ravindranathan
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eun Kim
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shaohua Huang
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thomas W. Kenniston
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John C. Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Herbert J. Zeh
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa H. Butterfield
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Departments of Medicine and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrea Gambotto
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L. Bartlett
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Earl PL, Moss B, Wyatt LS. Generation of Recombinant Vaccinia Viruses. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2017; 89:5.13.1-5.13.18. [PMID: 28762491 PMCID: PMC5765993 DOI: 10.1002/cpps.33] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This unit describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector or PCR fragment to generate a recombinant virus. Selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses are described. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Patricia L. Earl
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, MD 20892-3210
| | - Bernard Moss
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, MD 20892-3210
| | - Linda S. Wyatt
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, MD 20892-3210
| |
Collapse
|
17
|
Novel Nonreplicating Vaccinia Virus Vector Enhances Expression of Heterologous Genes and Suppresses Synthesis of Endogenous Viral Proteins. mBio 2017; 8:mBio.00790-17. [PMID: 28588133 PMCID: PMC5461411 DOI: 10.1128/mbio.00790-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Viruses are used as expression vectors for protein synthesis, immunology research, vaccines, and therapeutics. Advantages of poxvirus vectors include the accommodation of large amounts of heterologous DNA, the presence of a cytoplasmic site of transcription, and high expression levels. On the other hand, competition of approximately 200 viral genes with the target gene for expression and immune recognition may be disadvantageous. We describe a vaccinia virus (VACV) vector that uses an early promoter to express the bacteriophage T7 RNA polymerase; has the A23R intermediate transcription factor gene deleted, thereby restricting virus replication to complementing cells; and has a heterologous gene regulated by a T7 promoter. In noncomplementing cells, viral early gene expression and DNA replication occurred normally but synthesis of intermediate and late proteins was prevented. Nevertheless, the progeny viral DNA provided templates for abundant expression of heterologous genes regulated by a T7 promoter. Selective expression of the Escherichia coli lac repressor gene from an intermediate promoter reduced transcription of the heterologous gene specifically in complementing cells, where large amounts might adversely impact VACV replication. Expression of heterologous proteins mediated by the A23R deletion vector equaled that of a replicating VACV, was higher than that of a nonreplicating modified vaccinia virus Ankara (MVA) vector used for candidate vaccines in vitro and in vivo, and was similarly immunogenic in mice. Unlike the MVA vector, the A23R deletion vector still expresses numerous early genes that can restrict immunogenicity as demonstrated here by the failure of the prototype vector to induce interferon alpha. By deleting immunomodulatory genes, we anticipate further improvements in the system. Vaccines provide an efficient and effective way of preventing infectious diseases. Nevertheless, new and better vaccines are needed. Vaccinia virus, which was used successfully as a live vaccine to eradicate smallpox, has been further attenuated and adapted as a recombinant vector for immunization against other pathogens. However, since the initial description of this vector system, only incremental improvements largely related to safety have been implemented. Here we described novel modifications of the platform that increased expression of the heterologous target gene and decreased expression of endogenous vaccinia virus genes while providing safety by preventing replication of the candidate vaccine except in complementing cells used for vector propagation.
Collapse
|
18
|
Wyatt LS, Earl PL, Moss B. Generation of Recombinant Vaccinia Viruses. ACTA ACUST UNITED AC 2017; 117:16.17.1-16.17.18. [PMID: 28060405 DOI: 10.1002/cpmb.32] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This unit describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector or PCR fragment to generate a recombinant virus. Selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses are described. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Linda S Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
19
|
Volz A, Sutter G. Modified Vaccinia Virus Ankara: History, Value in Basic Research, and Current Perspectives for Vaccine Development. Adv Virus Res 2016; 97:187-243. [PMID: 28057259 PMCID: PMC7112317 DOI: 10.1016/bs.aivir.2016.07.001] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development of new candidate vaccines against infectious diseases and cancer. Historically, MVA was developed by serial tissue culture passage in primary chicken cells of vaccinia virus strain Ankara, and clinically used to avoid the undesirable side effects of conventional smallpox vaccination. Adapted to growth in avian cells MVA lost the ability to replicate in mammalian hosts and lacks many of the genes orthopoxviruses use to conquer their host (cell) environment. As a biologically well-characterized mutant virus, MVA facilitates fundamental research to elucidate the functions of poxvirus host-interaction factors. As extremely safe viral vectors MVA vaccines have been found immunogenic and protective in various preclinical infection models. Multiple recombinant MVA currently undergo clinical testing for vaccination against human immunodeficiency viruses, Mycobacterium tuberculosis or Plasmodium falciparum. The versatility of the MVA vector vaccine platform is readily demonstrated by the swift development of experimental vaccines for immunization against emerging infections such as the Middle East Respiratory Syndrome. Recent advances include promising results from the clinical testing of recombinant MVA-producing antigens of highly pathogenic avian influenza virus H5N1 or Ebola virus. This review summarizes our current knowledge about MVA as a unique strain of vaccinia virus, and discusses the prospects of exploiting this virus as research tool in poxvirus biology or as safe viral vector vaccine to challenge existing and future bottlenecks in vaccinology.
Collapse
Affiliation(s)
- A Volz
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany
| | - G Sutter
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany.
| |
Collapse
|
20
|
Marín-López A, Ortego J. Generation of Recombinant Modified Vaccinia Virus Ankara Encoding VP2, NS1, and VP7 Proteins of Bluetongue Virus. Methods Mol Biol 2016; 1349:137-50. [PMID: 26458834 DOI: 10.1007/978-1-4939-3008-1_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Modified Vaccinia Virus Ankara (MVA) is employed widely as an experimental vaccine vector for its lack of replication in mammalian cells and high expression level of foreign/heterologous genes. Recombinant MVAs (rMVAs) are used as platforms for protein production as well as vectors to generate vaccines against a high number of infectious diseases and other pathologies. The portrait of the virus combines desirable elements such as high-level biological safety, the ability to activate appropriate innate immune mediators upon vaccination, and the capacity to deliver substantial amounts of heterologous antigens. Recombinant MVAs encoding proteins of bluetongue virus (BTV), an Orbivirus that infects domestic and wild ruminants transmitted by biting midges of the Culicoides species, are excellent vaccine candidates against this virus. In this chapter we describe the methods for the generation of rMVAs encoding VP2, NS1, and VP7 proteins of bluetongue virus as a model example for orbiviruses. The protocols included cover the cloning of VP2, NS1, and VP7 BTV-4 genes in a transfer plasmid, the construction of recombinant MVAs, the titration of virus working stocks and the protein expression analysis by immunofluorescence and radiolabeling of rMVA infected cells as well as virus purification.
Collapse
Affiliation(s)
- Alejandro Marín-López
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Almentaria (INIA), Ctra de Valdeolmos-El Casar s/n, Valdeolmos, 28130, Madrid, Spain
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Almentaria (INIA), Ctra de Valdeolmos-El Casar s/n, Valdeolmos, 28130, Madrid, Spain.
| |
Collapse
|
21
|
Use of Reporter Genes in the Generation of Vaccinia Virus-Derived Vectors. Viruses 2016; 8:v8050134. [PMID: 27213433 PMCID: PMC4885089 DOI: 10.3390/v8050134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
Vaccinia virus (VACV) is one of the most extensively-studied viruses of the Poxviridae family. It is easy to genetically modify, so it has become a key tool for many applications. In this context, reporter genes facilitate the study of the role of foreign genes introduced into the genome of VACV. In this review, we describe the type of reporter genes that have been used to generate reporter-expressing VACV and the applications of the recombinant viruses obtained. Reporter-expressing VACV are currently employed in basic and immunology research, in the development of vaccines and cancer treatment.
Collapse
|
22
|
Influenza Neuraminidase Subtype N1: Immunobiological Properties and Functional Assays for Specific Antibody Response. PLoS One 2016; 11:e0153183. [PMID: 27054879 PMCID: PMC4824357 DOI: 10.1371/journal.pone.0153183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/24/2016] [Indexed: 01/08/2023] Open
Abstract
Influenza neuraminidase (NA) proteins expressed in TK− cells infected with recombinant vaccinia virus carrying NA gene of highly pathogenic avian influenza H5N1 virus or 2009 pandemic H1N1 (H1N1pdm) virus were characterized for their biological properties, i.e., cell localization, molecular weight (MW), glycosylation and sialidase activity. Immune sera collected from BALB/c mice immunized with these recombinant viruses were assayed for binding and functional activities of anti-NA antibodies. Recombinant NA proteins were found localized in cytoplasm and cytoplasmic membrane of the infected cells. H1N1pdm NA protein had MW at about 75 kDa while it was 55 kDa for H5N1 NA protein. Hyperglycosylation was more pronounced in H1N1pdm NA compared to H5N1 NA according to N-glycosidase F treatment. Three dimensional structures also predicted that H1N1 NA globular head contained 4 and that of H5N1 contained 2 potential glycosylation sites. H5N1 NA protein had higher sialidase activity than H1N1pdm NA protein as measured by both MUNANA-based assay and fetuin-based enzyme-linked lectin assay (ELLA). Plaque reduction assay demonstrated that anti-NA antibody could reduce number of plaques and plaque size through inhibiting virus release, not virus entry. Assay for neuraminidase-inhibition (NI) antibody by ELLA showed specific and cross reactivity between H5N1 NA and H1N1pdm NA protein derived from reverse genetic viruses or wild type viruses. In contrast, replication-inhibition assay in MDCK cells showed that anti-H1N1 NA antibody moderately inhibited viruses with homologous NA gene only, while anti-H5N1 NA antibody modestly inhibited the replication of viruses containing homologous NA gene and NA gene derived from H1N1pdm virus. Anti-H1N1 NA antibody showed higher titers of inhibiting virus replication than anti-H5N1 NA antibody, which are consistent with the results on reduction in plaque numbers and sizes as well as in inhibiting NA enzymatic activity. No assay showed cross reactivity with reassorted PR8 (H1N1) virus and H3N2 wild type viruses.
Collapse
|
23
|
Wyatt LS, Earl PL, Moss B. Generation of Recombinant Vaccinia Viruses. CURRENT PROTOCOLS IN MICROBIOLOGY 2015; 39:14A.4.1-14A.4.18. [PMID: 26528782 PMCID: PMC5123791 DOI: 10.1002/9780471729259.mc14a04s39] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This unit describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector or PCR fragment to generate a recombinant virus. Selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses are described. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented.
Collapse
Affiliation(s)
- Linda S Wyatt
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia L Earl
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bernard Moss
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
24
|
A marker-free system for highly efficient construction of vaccinia virus vectors using CRISPR Cas9. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15035. [PMID: 26417609 PMCID: PMC4571730 DOI: 10.1038/mtm.2015.35] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/05/2015] [Indexed: 02/07/2023]
Abstract
The current method for creation of vaccinia virus (VACV) vectors involves using a selection and purification marker, however inclusion of a gene without therapeutic value in the resulting vector is not desirable for clinical use. The Cre-LoxP system has been used to make marker-free Poxviruses, but the efficiency was very low. To obtain a marker-free VACV vector, we developed marker gene excision systems to modify the thymidine kinase (TK) region and N1L regions using Cre-Loxp and Flp-FRET systems respectively. CRISPR-Cas9 system significantly resulted in a high efficiency (~90%) in generation of marker gene-positive TK-mutant VACV vector. The marker gene (RFP) could be excised from the recombinant virus using Cre recombinase. To make a marker-free VV vector with double gene deletions targeting the TK and N1L gene, we constructed a donor repair vector targeting the N1L gene, which can carry a therapeutic gene and the marker (RFP) that could be excised from the recombinant virus using Flp recombinase. The marker-free system developed here can be used to efficiently construct VACV vectors armed with any therapeutic genes in the TK region or N1L region without marker genes. Our marker-free system platform has significant potential for development of new marker-free VACV vectors for clinical application.
Collapse
|
25
|
Ondondo B, Faulkner L, Williams NA, Morgan AJ, Morgan DJ. The B subunit of Escherichia coli enterotoxin helps control the in vivo growth of solid tumors expressing the Epstein-Barr virus latent membrane protein 2A. Cancer Med 2015; 4:457-71. [PMID: 25641882 PMCID: PMC4380971 DOI: 10.1002/cam4.380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/19/2014] [Accepted: 10/09/2014] [Indexed: 01/23/2023] Open
Abstract
Latent membrane protein 2A (LMP2A) is expressed on almost all Epstein–Barr virus (EBV)-associated tumors and is a potential target for immunotherapeutic intervention and vaccination. However, LMP2A is not efficiently processed and presented on major histocompatibility antigens class I molecules to generate potent cytotoxic T-lymphocytes (CTL) responses capable of killing these tumors. The B subunit of Escherichia coli enterotoxin (EtxB), causes rapid internalization and processing of membrane-bound LMP2A on EBV-infected B cells, and facilitates loading of processed-LMP2A peptides onto MHC class I. This re-directed trafficking/delivery of LMP2A to the MHC class I machinery enhances recognition and killing by LMP2A-specific CTL in vitro. To test the potential of EtxB to enhance immune targeting of LMP2A expressed in solid tumors, we generated a murine tumor model (Renca-LMP2A), in which LMP2A is expressed as a transgenic neoantigen on a renal carcinoma (Renca) cell line and forms solid tumors when injected subcutaneously into BALB/c mice. The data show that in BALB/c mice which have only low levels of peripheral Kd-LMP2A-specific CD8+ T cells, merely a transient inhibition of tumor growth is achieved compared with naïve mice; suggesting that there is suboptimal LMP2A-specifc CTL recognition and poorly targeted tumor killing. However, importantly, treatment of these mice with EtxB led to a significant delay in the onset of tumor growth and significantly lower tumor volumes compared with similar mice that did not receive EtxB. Moreover, this remarkable effect of EtxB was achieved despite progressive reduction in tumor expression of LMP2A and MHC class I molecules. These data clearly demonstrate the potential efficacy of EtxB as a novel therapeutic agent that could render EBV-associated tumors susceptible to immune control.
Collapse
Affiliation(s)
- Beatrice Ondondo
- The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, Oxfordshire OX3 7DQ, United Kingdom
| | | | | | | | | |
Collapse
|
26
|
Stellberger T, Haase M, Guertler P, Stockmar I, Busch U, Baiker A. Characterization of Recombinant Vaccinia Viruses by MLPA Technology. APPLIED BIOSAFETY 2014. [DOI: 10.1177/153567601401900304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | - Maren Haase
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Patrick Guertler
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Iris Stockmar
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Ulrich Busch
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Armin Baiker
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| |
Collapse
|
27
|
Alberca B, Bachanek-Bankowska K, Cabana M, Calvo-Pinilla E, Viaplana E, Frost L, Gubbins S, Urniza A, Mertens P, Castillo-Olivares J. Vaccination of horses with a recombinant modified vaccinia Ankara virus (MVA) expressing African horse sickness (AHS) virus major capsid protein VP2 provides complete clinical protection against challenge. Vaccine 2014; 32:3670-4. [PMID: 24837765 PMCID: PMC4061461 DOI: 10.1016/j.vaccine.2014.04.036] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/01/2014] [Accepted: 04/14/2014] [Indexed: 11/15/2022]
Abstract
A recombinant modified Vaccinia Ankara virus expressing VP2 of African horse sickness virus serotype 9 was generated. Four horses were vaccinated on days 0 and 20. Three unvaccinated controls were used. Vaccinated and control horses were challenged intravenously with 107.4TCID50 of AHSV-9 on day 34 of the study. At challenge, vaccinates had virus neutralising antibodies but were negative for antibodies to AHSV-VP7. All vaccinates were completely protected against clinical signs of African horse sickness.
African horse sickness virus (AHSV) is an arthropod-borne pathogen that infects all species of equidae and causes high mortality in horses. Previously, a recombinant modified vaccinia Ankara (MVA) virus expressing the protein VP2 of AHSV serotype 4 was shown to induce virus neutralising antibodies in horses and protected interferon alpha receptor gene knock-out mice (IFNAR −/−) against virulent AHSV challenge. This study builds on the previous work, examining the protective efficacy of MVA-VP2 vaccination in the natural host of AHSV infection. A study group of 4 horses was vaccinated twice with a recombinant MVA virus expressing the major capsid protein (VP2) of AHSV serotype 9. Vaccinated animals and a control group of unvaccinated horses were then challenged with a virulent strain of AHSV-9. The vaccinated animals were completely protected against clinical disease and also against viraemia as measured by standard end-point dilution assays. In contrast, all control horses presented viraemia after challenge and succumbed to the infection. These results demonstrate the potential of recombinant MVA viruses expressing the outer capsid VP2 of AHSV as a protective vaccine against AHSV infection in the field.
Collapse
Affiliation(s)
- Berta Alberca
- Zoetis-Spain, Ctra de Comprodon, Finca La Riba, 17813 Vall de Bianya, Girona, Spain
| | | | - Marta Cabana
- Zoetis-Spain, Ctra de Comprodon, Finca La Riba, 17813 Vall de Bianya, Girona, Spain
| | - Eva Calvo-Pinilla
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, Surrey, UK
| | - Elisenda Viaplana
- Zoetis-Spain, Ctra de Comprodon, Finca La Riba, 17813 Vall de Bianya, Girona, Spain
| | - Lorraine Frost
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, Surrey, UK
| | - Simon Gubbins
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, Surrey, UK
| | - Alicia Urniza
- Zoetis-Spain, Ctra de Comprodon, Finca La Riba, 17813 Vall de Bianya, Girona, Spain
| | - Peter Mertens
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, Surrey, UK
| | | |
Collapse
|
28
|
Keil GM, Giesow K, Portugal R. A novel bromodeoxyuridine-resistant wild boar lung cell line facilitates generation of African swine fever virus recombinants. Arch Virol 2014; 159:2421-8. [DOI: 10.1007/s00705-014-2095-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/19/2014] [Indexed: 10/25/2022]
|
29
|
Robinson MJ, Tan CS, Fenwick F, Chambers CJ, Routledge EG, Toms GL. Generation and epitope mapping of a sub-group cross-reactive anti-respiratory syncytial virus G glycoprotein monoclonal antibody which is protective in vivo. J Med Virol 2014; 86:1267-77. [PMID: 24415460 DOI: 10.1002/jmv.23881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2013] [Indexed: 11/07/2022]
Abstract
Passively administered antibodies to conserved epitopes on the attachment (G) glycoprotein of human respiratory syncytial virus (hRSV) have potential in the immunoprophylaxis of human infections. This study set out to generate monoclonal antibodies (MAbs) recognizing all prevalent lineages of HRSV and capable of immunoprophylaxis in mice. Two murine MAbs of broad specificity for prevalent virus strains were generated by immunization of mice with hRSV of sub-group A followed by selection of hybridomas on recombinant G glycoprotein from a sub-group B virus. The anti-G hybridomas generated secreted antibody of high affinity but negligible neutralizing capacity one of which was tested in mice and found to be protective against live virus challenge. Western blotting and partial epitope mapping on transiently expressed G-glycoprotein fragments indicate that these antibodies recognize a complex epitope on the protein backbone of the molecule involving residues both C'- and N-terminal to the central conserved motif.
Collapse
Affiliation(s)
- Mark J Robinson
- Institute of Cellular Medicine, The Medical School, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | |
Collapse
|
30
|
Lin LCW, Flesch IEA, Tscharke DC. Immunodomination during peripheral vaccinia virus infection. PLoS Pathog 2013; 9:e1003329. [PMID: 23633956 PMCID: PMC3635974 DOI: 10.1371/journal.ppat.1003329] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 03/14/2013] [Indexed: 11/20/2022] Open
Abstract
Immunodominance is a fundamental property of CD8(+) T cell responses to viruses and vaccines. It had been observed that route of administration alters immunodominance after vaccinia virus (VACV) infection, but only a few epitopes were examined and no mechanism was provided. We re-visited this issue, examining a panel of 15 VACV epitopes and four routes, namely intradermal (i.d.), subcutaneous (s.c.), intraperitoneal (i.p.) and intravenous (i.v.) injection. We found that immunodominance is sharpened following peripheral routes of infection (i.d. and s.c.) compared with those that allow systemic virus dissemination (i.p. and i.v.). This increased immunodominance was demonstrated with native epitopes of VACV and with herpes simplex virus glycoprotein B when expressed from VACV. Responses to some subdominant epitopes were altered by as much as fourfold. Tracking of virus, examination of priming sites, and experiments restricting virus spread showed that priming of CD8(+) T cells in the spleen was necessary, but not sufficient to broaden responses. Further, we directly demonstrated that immunodomination occurs more readily when priming is mainly in lymph nodes. Finally, we were able to reduce immunodominance after i.d., but not i.p. infection, using a VACV expressing the costimulators CD80 (B7-1) and CD86 (B7-2), which is notable because VACV-based vaccines incorporating these molecules are in clinical trials. Taken together, our data indicate that resources for CD8(+) T cell priming are limiting in local draining lymph nodes, leading to greater immunodomination. Further, we provide evidence that costimulation can be a limiting factor that contributes to immunodomination. These results shed light on a possible mechanism of immunodomination and highlight the need to consider multiple epitopes across the spectrum of immunogenicities in studies aimed at understanding CD8(+) T cell immunity to viruses.
Collapse
Affiliation(s)
- Leon C. W. Lin
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Inge E. A. Flesch
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - David C. Tscharke
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
31
|
Guha R, Das S, Ghosh J, Naskar K, Mandala A, Sundar S, Dujardin JC, Roy S. Heterologous priming-boosting with DNA and vaccinia virus expressing kinetoplastid membrane protein-11 induces potent cellular immune response and confers protection against infection with antimony resistant and sensitive strains of Leishmania (Leishmania) donovani. Vaccine 2013; 31:1905-15. [PMID: 23499564 DOI: 10.1016/j.vaccine.2013.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 02/08/2013] [Accepted: 02/11/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND Emergence of resistance against commonly available drugs poses a major threat in the treatment of visceral leishmaniasis (VL), particularly in the Indian subcontinent. Absence of any licensed vaccine against VL emphasizes the urgent need to develop an effective alternative vaccination strategy. METHODOLOGY We developed a novel heterologous prime boost immunization strategy using kinetoplastid membrane protein-11 (KMP-11) DNA priming followed by boosting with recombinant vaccinia virus (rVV) expressing the same antigen. The efficacy of this vaccination regimen in a murine and hamster model of visceral leishmaniasis caused by both antimony resistant (Sb-R) and sensitive (Sb-S) Leishmania (L.) donovani is examined. RESULT Heterologous prime-boost (KMP-11 DNA/rVV) vaccination was able to protect mice and hamsters from experimental VL induced by both Sb-S and Sb-R-L. (L.) donovani isolates. Parasite burden is kept significantly low in the vaccinated groups even after 60 days post-infection in hamsters, which are extremely susceptible to VL. Protection in mice is correlated with strong cellular and humoral immune responses. Generation of polyfunctional CD8(+) T cell was observed in vaccinated groups, which is one of the most important prerequisite for successful vaccination against VL. Protection was accompanied with generation of antigen specific CD4(+) and CD8(+) cells that produced effector cytokines such as IFN-γ, IL-2 and TNF-α. KMP-11-DNA/rVV vaccination also developed strong cytotoxic response and reversed T-cell impairment to induce antigen specific T cell proliferation. CONCLUSION KMP-11 is a unique antigen with high epitope density. Heterologous prime boost vaccination activates CD4(+) and CD8(+) T-cell mediated immunity to confer resistance to VL. This immunization method also produces high quality T-cells secreting multiple effector cytokines thus enhancing durability of the immune response. Thus the vaccination regime as described in the present study could provide a potent strategy for future anti-leishmanial vaccine development.
Collapse
Affiliation(s)
- Rajan Guha
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Santos-Fernandes É, Beltrame CO, Byrd CM, Cardwell KB, Schnellrath LC, Medaglia MLG, Hruby DE, Jordan R, Damaso CR. Increased susceptibility of Cantagalo virus to the antiviral effect of ST-246®. Antiviral Res 2012; 97:301-11. [PMID: 23257396 DOI: 10.1016/j.antiviral.2012.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/26/2012] [Accepted: 11/28/2012] [Indexed: 11/16/2022]
Abstract
Cantagalo virus (CTGV) is the etiologic agent of a pustular disease in dairy cows and dairy workers in Brazil with important economical and occupational impacts. Nevertheless, no antiviral therapy is currently available. ST-246 is a potent inhibitor of orthopoxvirus egress from cells and has proved its efficacy in cell culture and in animal models. In this work, we evaluated the effect of ST-246 on CTGV replication. Plaque reduction assays indicated that CTGV is 6-38 times more susceptible to the drug than VACV-WR and cowpox virus, respectively, with an EC50 of 0.0086μM and a selective index of >11,600. The analysis of β-gal activity expressed by recombinant viruses in the presence of ST-246 confirmed these results. In addition, ST-246 had a greater effect on the reduction of CTGV spread in comet tail assays and on the production of extracellular virus relative to VACV-WR. Infection of mice with CTGV by tail scarification generated primary lesions at the site of scarification that appeared less severe than those induced by VACV-WR. Animals infected with CTGV and treated with ST-246 at 100mg/kg for 5days did not develop primary lesions and virus yields were inhibited by nearly 98%. In contrast, primary lesions induced by VACV-WR were not affected by ST-246. The analysis of F13 (p37) protein from CTGV revealed a unique substitution in residue 217 (D217N) not found in other orthopoxviruses. Construction of recombinant VACV-WR containing the D217N polymorphism did not lead to an increase in the susceptibility to ST-246. Therefore, it is still unknown why CTGV is more susceptible to the antiviral effects of ST-246 compared to VACV-WR. Nonetheless, our data demonstrates that ST-246 is a potent inhibitor of CTGV replication that should be further evaluated as a promising anti-CTGV therapy.
Collapse
Affiliation(s)
- Élida Santos-Fernandes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rintoul JL, Lemay CG, Tai LH, Stanford MM, Falls TJ, de Souza CT, Bridle BW, Daneshmand M, Ohashi PS, Wan Y, Lichty BD, Mercer AA, Auer RC, Atkins HL, Bell JC. ORFV: a novel oncolytic and immune stimulating parapoxvirus therapeutic. Mol Ther 2012; 20:1148-57. [PMID: 22273579 PMCID: PMC3369287 DOI: 10.1038/mt.2011.301] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/18/2011] [Indexed: 12/27/2022] Open
Abstract
Replicating viruses for the treatment of cancer have a number of advantages over traditional therapeutic modalities. They are highly targeted, self-amplifying, and have the added potential to act as both gene-therapy delivery vehicles and oncolytic agents. Parapoxvirus ovis or Orf virus (ORFV) is the prototypic species of the Parapoxvirus genus, causing a benign disease in its natural ungulate host. ORFV possesses a number of unique properties that make it an ideal viral backbone for the development of a cancer therapeutic: it is safe in humans, has the ability to cause repeat infections even in the presence of antibody, and it induces a potent T(h)-1-dominated immune response. Here, we show that live replicating ORFV induces an antitumor immune response in multiple syngeneic mouse models of cancer that is mediated largely by the potent activation of both cytokine-secreting, and tumoricidal natural killer (NK) cells. We have also highlighted the clinical potential of the virus by demonstration of human cancer cell oncolysis including efficacy in an A549 xenograft model of cancer.
Collapse
Affiliation(s)
- Julia L Rintoul
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Chantal G Lemay
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Lee-Hwa Tai
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Marianne M Stanford
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Theresa J Falls
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Christiano T de Souza
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Byram W Bridle
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Manijeh Daneshmand
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Pamela S Ohashi
- The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute/Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Brian D Lichty
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Andrew A Mercer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Rebecca C Auer
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - Harold L Atkins
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| | - John C Bell
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, Ontario, Canada
| |
Collapse
|
34
|
Ricci PS, Schäfer B, Kreil TR, Falkner FG, Holzer GW. Selection of recombinant MVA by rescue of the essential D4R gene. Virol J 2011; 8:529. [PMID: 22152060 PMCID: PMC3293099 DOI: 10.1186/1743-422x-8-529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 12/12/2011] [Indexed: 11/25/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) has become a promising vaccine vector due to its immunogenicity and its proven safety in humans. As a general approach for stringent and rapid selection of recombinant MVA, we assessed marker rescue of the essential viral D4R gene in an engineered deletion mutant that is fully replication defective in wild-type cells. Recombinant, replicating virus was obtained by re-introduction of the deleted viral gene as a dominant selection marker into the deletion mutant.
Collapse
Affiliation(s)
- Patricia S Ricci
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| | - Birgit Schäfer
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| | | | - Falko G Falkner
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| | - Georg W Holzer
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| |
Collapse
|
35
|
George JA, Eo SK. Distinct Humoral and Cellular Immunity Induced by Alternating Prime-boost Vaccination Using Plasmid DNA and Live Viral Vector Vaccines Expressing the E Protein of Dengue Virus Type 2. Immune Netw 2011; 11:268-80. [PMID: 22194710 PMCID: PMC3243001 DOI: 10.4110/in.2011.11.5.268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 09/28/2011] [Accepted: 10/04/2011] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Dengue virus, which belongs to the Flavivirus genus of the Flaviviridae family, causes fatal dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) with infection risk of 2.5 billion people worldwide. However, approved vaccines are still not available. Here, we explored the immune responses induced by alternating prime-boost vaccination using DNA vaccine, adenovirus, and vaccinia virus expressing E protein of dengue virus type 2 (DenV2). METHODS Following immunization with DNA vaccine (pDE), adenovirus (rAd-E), and/or vaccinia virus (VV-E) expressing E protein, E protein-specific IgG and its isotypes were determined by conventional ELISA. Intracellular CD154 and cytokine staining was used for enumerating CD4+ T cells specific for E protein. E protein-specific CD8+ T cell responses were evaluated by in vivo CTL killing activity and intracellular IFN-γ staining. RESULTS Among three constructs, VV-E induced the most potent IgG responses, Th1-type cytokine production by stimulated CD4+ T cells, and the CD8+ T cell response. Furthermore, when the three constructs were used for alternating prime-boost vaccination, the results revealed a different pattern of CD4+ and CD8+ T cell responses. i) Priming with VV-E induced higher E-specific IgG level but it was decreased rapidly. ii) Strong CD8+ T cell responses specific for E protein were induced when VV-E was used for the priming step, and such CD8+ T cell responses were significantly boosted with pDE. iii) Priming with rAd-E induced stronger CD4+ T cell responses which subsequently boosted with pDE to a greater extent than VV-E and rAd-E. CONCLUSION These results indicate that priming with live viral vector vaccines could induce different patterns of E protein- specific CD4+ and CD8+ T cell responses which were significantly enhanced by booster vaccination with the DNA vaccine. Therefore, our observation will provide valuable information for the establishment of optimal prime-boost vaccination against DenV.
Collapse
Affiliation(s)
- Junu A George
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| | | |
Collapse
|
36
|
Cohen M, Braun E, Tsalenchuck Y, Panet A, Steiner I. Restrictions that control herpes simplex virus type 1 infection in mouse brain ex vivo. J Gen Virol 2011; 92:2383-2393. [DOI: 10.1099/vir.0.031013-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Elucidating the cellular and molecular factors governing herpes simplex virus type 1 (HSV-1) neurotropism is a prerequisite for understanding HSV-1 encephalitis and for targeting HSV-1-derived vectors for gene transfer to the brain. Earlier we had described an ex vivo system of mouse brain slices and demonstrated a selective and unique infection pattern, mostly around the ventricles. Here, we examined tissue factors controlling HSV-1 infection of brain slices. We demonstrated that heparan sulphate, while an important factor, does not determine the infection pattern. Hyaluronic acid, but not collagen, appears to enhance HSV-1 brain infection. To investigate whether tissue distribution of viral receptors determines the infection pattern, we examined transcription of herpes virus entry mediator and nectin-1 receptor genes in infected and uninfected brain regions. Both the infected and the uninfected regions express the receptors. We also explored the influence of intra-cellular factors. HSV-1 does not preferentially infect proliferating cells in the brain slices, despite its predilection to the ventricular zones. To delineate the step at which the HSV-1 infection cascade is restricted, mRNA was isolated following tissue infection, and transcription of the immediate-early and late viral genes was evaluated. The results indicated that HSV-1 genes are not expressed in regions that do not express a viral reporter gene. Therefore, we conclude that tissue resistance to infection is associated with a block at or prior to the immediate-early mRNA synthesis. Taken together, using the ex vivo system of organotypic culture we describe here extra-cellular and intra-cellular restriction levels of HSV-1 brain infection.
Collapse
Affiliation(s)
- Meytal Cohen
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Efrat Braun
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yael Tsalenchuck
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Amos Panet
- Department of Biochemistry, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Israel Steiner
- Department of Neurology, Rabin Medical Center, Campus Beilinson, Petach Tikva, Israel
- Laboratory of Neurovirology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
37
|
Rintoul JL, Wang J, Gammon DB, van Buuren NJ, Garson K, Jardine K, Barry M, Evans DH, Bell JC. A selectable and excisable marker system for the rapid creation of recombinant poxviruses. PLoS One 2011; 6:e24643. [PMID: 21931792 PMCID: PMC3169633 DOI: 10.1371/journal.pone.0024643] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 08/16/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Genetic manipulation of poxvirus genomes through attenuation, or insertion of therapeutic genes has led to a number of vector candidates for the treatment of a variety of human diseases. The development of recombinant poxviruses often involves the genomic insertion of a selectable marker for purification and selection purposes. The use of marker genes however inevitably results in a vector that contains unwanted genetic information of no therapeutic value. METHODOLOGY/PRINCIPAL FINDINGS Here we describe an improved strategy that allows for the creation of marker-free recombinant poxviruses of any species. The Selectable and Excisable Marker (SEM) system incorporates a unique fusion marker gene for the efficient selection of poxvirus recombinants and the Cre/loxP system to facilitate the subsequent removal of the marker. We have defined and characterized this new methodological tool by insertion of a foreign gene into vaccinia virus, with the subsequent removal of the selectable marker. We then analyzed the importance of loxP orientation during Cre recombination, and show that the SEM system can be used to introduce site-specific deletions or inversions into the viral genome. Finally, we demonstrate that the SEM strategy is amenable to other poxviruses, as demonstrated here with the creation of an ectromelia virus recombinant lacking the EVM002 gene. CONCLUSION/SIGNIFICANCE The system described here thus provides a faster, simpler and more efficient means to create clinic-ready recombinant poxviruses for therapeutic gene therapy applications.
Collapse
Affiliation(s)
- Julia L. Rintoul
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Jiahu Wang
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Don B. Gammon
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Nicholas J. van Buuren
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Kenneth Garson
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Karen Jardine
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Michele Barry
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - David H. Evans
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - John C. Bell
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
- * E-mail:
| |
Collapse
|
38
|
Bridge SH, Sharpe SA, Dennis MJ, Dowall SD, Getty B, Anson DS, Skinner MA, Stewart JP, Blanchard TJ. Heterologous prime-boost-boost immunisation of Chinese cynomolgus macaques using DNA and recombinant poxvirus vectors expressing HIV-1 virus-like particles. Virol J 2011; 8:429. [PMID: 21899739 PMCID: PMC3177910 DOI: 10.1186/1743-422x-8-429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 09/07/2011] [Indexed: 01/13/2023] Open
Abstract
Background There is renewed interest in the development of poxvirus vector-based HIV vaccines due to the protective effect observed with repeated recombinant canarypox priming with gp120 boosting in the recent Thai placebo-controlled trial. This study sought to investigate whether a heterologous prime-boost-boost vaccine regimen in Chinese cynomolgus macaques with a DNA vaccine and recombinant poxviral vectors expressing HIV virus-like particles bearing envelopes derived from the most prevalent clades circulating in sub-Saharan Africa, focused the antibody response to shared neutralising epitopes. Methods Three Chinese cynomolgus macaques were immunised via intramuscular injections using a regimen composed of a prime with two DNA vaccines expressing clade A Env/clade B Gag followed by boosting with recombinant fowlpox virus expressing HIV-1 clade D Gag, Env and cholera toxin B subunit followed by the final boost with recombinant modified vaccinia virus Ankara expressing HIV-1 clade C Env, Gag and human complement protein C3d. We measured the macaque serum antibody responses by ELISA, enumerated T cell responses by IFN-γ ELISpot and assessed seroneutralisation of HIV-1 using the TZM-bl β-galactosidase assay with primary isolates of HIV-1. Results This study shows that large and complex synthetic DNA sequences can be successfully cloned in a single step into two poxvirus vectors: MVA and FPV and the recombinant poxviruses could be grown to high titres. The vaccine candidates showed appropriate expression of recombinant proteins with the formation of authentic HIV virus-like particles seen on transmission electron microscopy. In addition the b12 epitope was shown to be held in common by the vaccine candidates using confocal immunofluorescent microscopy. The vaccine candidates were safely administered to Chinese cynomolgus macaques which elicited modest T cell responses at the end of the study but only one out of the three macaques elicited an HIV-specific antibody response. However, the antibodies did not neutralise primary isolates of HIV-1 or the V3-sensitive isolate SF162 using the TZM-bl β-galactosidase assay. Conclusions MVA and FP9 are ideal replication-deficient viral vectors for HIV-1 vaccines due to their excellent safety profile for use in humans. This study shows this novel prime-boost-boost regimen was poorly immunogenic in Chinese cynomolgus macaques.
Collapse
Affiliation(s)
- Simon H Bridge
- Clinical Research Group, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Embry A, Meng X, Cantwell A, Dube PH, Xiang Y. Enhancement of immune response to an antigen delivered by vaccinia virus by displaying the antigen on the surface of intracellular mature virion. Vaccine 2011; 29:5331-9. [PMID: 21664218 PMCID: PMC3139018 DOI: 10.1016/j.vaccine.2011.05.088] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 05/23/2011] [Accepted: 05/25/2011] [Indexed: 12/01/2022]
Abstract
Vaccinia virus (VACV) is the vaccine for smallpox and a widely used vaccine vector for infectious diseases and cancers. The majority of the antibodies elicited by live VACV vaccination respond to virion structural proteins, including many integral membrane proteins on the intracellular mature virion (MV). Here, we showed that antibody response to an exogenous antigen delivered by VACV was greatly enhanced by incorporating the antigen as an integral membrane protein of MV. We constructed recombinant VACV expressing a Yersinia pestis protective antigen, LcrV, unmodified or fused with either a signal peptide or with the transmembrane domain of VACV D8 protein (LcrV-TM). Electron microscopy showed that LcrV-TM was displayed on the surface of MV. Importantly, VACV expressing LcrV-TM elicited a significantly higher titer of anti-LcrV antibody in mice than viruses expressing other forms of LcrV. Only mice immunized with LcrV-TM-expressing VACV were protected from lethal Y. pestis and VACV WR challenges. Antigen engineering through fusion with D8 transmembrane domain may be broadly applicable for enhancing the immune response to antigens delivered by a VACV vector. The recombinant virus described here could also serve as the basis for developing a vaccine against both smallpox and plague.
Collapse
Affiliation(s)
- Addie Embry
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xiangzhi Meng
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Angelene Cantwell
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Peter H. Dube
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Yan Xiang
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX
| |
Collapse
|
40
|
Wong YC, Lin LCW, Melo-Silva CR, Smith SA, Tscharke DC. Engineering recombinant poxviruses using a compact GFP-blasticidin resistance fusion gene for selection. J Virol Methods 2010; 171:295-8. [PMID: 21073901 DOI: 10.1016/j.jviromet.2010.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 10/27/2010] [Accepted: 11/01/2010] [Indexed: 11/29/2022]
Abstract
Recombinant poxviruses are important tools for research and some are candidate vaccines. To make these viruses a simple, small vector that can be used to engineer multiple strains of vaccinia virus and other model poxviruses, including ectromelia virus is of value. Here a set of plasmids and methods for making these viruses that uses an enhanced green fluorescent protein-blasticidin resistance (GFP-bsd) fusion gene as a transient selectable marker are described. This gene is smaller than any of the bi-functional selection markers used previously. The versatility of the method across different poxviruses is demonstrated by engineering changes into multiple loci of the WR and Modified Vaccinia Ankara (MVA) strains of vaccinia virus and also ectromelia virus. Finally, a set of vaccinia virus sequences for directing homologous recombination that are very highly conserved was designed and tested. These sequences allow a single plasmid to be used to insert a transgene into multiple strains of the virus.
Collapse
Affiliation(s)
- Yik Chun Wong
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | | | | | | | | |
Collapse
|
41
|
Chen W, Hu S, Qu L, Hu Q, Zhang Q, Zhi H, Huang K, Bu Z. A goat poxvirus-vectored peste-des-petits-ruminants vaccine induces long-lasting neutralization antibody to high levels in goats and sheep. Vaccine 2010; 28:4742-50. [DOI: 10.1016/j.vaccine.2010.04.102] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 04/23/2010] [Accepted: 04/29/2010] [Indexed: 10/19/2022]
|
42
|
Liu B, Tan W, Barsoum A, Gu X, Chen K, Huang W, Ramsay A, Kolls JK, Schwarzenberger P. IL-17 is a potent synergistic factor with GM-CSF in mice in stimulating myelopoiesis, dendritic cell expansion, proliferation, and functional enhancement. Exp Hematol 2010; 38:877-884.e1. [PMID: 20600582 DOI: 10.1016/j.exphem.2010.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 05/31/2010] [Accepted: 06/10/2010] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Interleukin (IL)-17, which now defines the Th(17) immune response, is a critical cytokine expressed and required for stress granulopoiesis during microbial invasion. Dendritic cells (DC) can instigate this response by inducing IL-17 expression in CD4(+) T cells. Besides IL-17, microbial invasion also stimulates production of the DC growth factor granulocyte-macrophage colony-stimulating factor (GM-CSF). The objective was the in vitro and in vivo investigation of IL-17 on DC proliferation and function in mice. MATERIALS AND METHODS Murine IL-17 (mIL-7) or murine GM-CSF (mGM-CSF), or both, was expressed in C57BL6 mice using adenoviral technology to assess hematopoietic and DC changes. The E-22 tymoma tumor cell line using a previously described vaccinia virus ovalbumin/LacZ murine tumor model was employed to study effects on tumor rejection. RESULTS The combination of mIL-17 and mGM-CSF increased peripheral neutrophila by 28-fold and splenic colonies by 11- and 14-fold over each individual factor in mice, respectively. The effect of mIL-17 by itself on murine DCs in vitro and in vivo was minimal; however, the combination greatly enhanced the stimulating effects of mGM-CSF, increasing the total numbers of CD14b/c(+) spleen DC by fourfold, as well as their function measured by enhanced endocytosis. Mixed lymphocyte reactions using mIL-17/mGM-CSF cultured DCs stimulator cells enhanced lymphocyte responses by twofold over mGM-CSF alone. Vaccination against LacZ in the C57BL6 E22 syngenic thymoma tumor model effectively delayed tumor growth in animals pretreated with the mIL-17/mGM-CSF combination prior to vaccination. CONCLUSIONS mIL-17 effectively synergizes with mGM-CSF in stimulating granulopoiesis and DC expansion, as well as in functional enhancement of DCs.
Collapse
Affiliation(s)
- Bainan Liu
- Quantumimmunologics, Tampa, FL 33623-0727, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Moser TS, Jones RG, Thompson CB, Coyne CB, Cherry S. A kinome RNAi screen identified AMPK as promoting poxvirus entry through the control of actin dynamics. PLoS Pathog 2010; 6:e1000954. [PMID: 20585561 PMCID: PMC2887478 DOI: 10.1371/journal.ppat.1000954] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 05/18/2010] [Indexed: 01/05/2023] Open
Abstract
Poxviruses include medically important human pathogens, yet little is known about the specific cellular factors essential for their replication. To identify genes essential for poxvirus infection, we used high-throughput RNA interference to screen the Drosophila kinome for factors required for vaccinia infection. We identified seven genes including the three subunits of AMPK as promoting vaccinia infection. AMPK not only facilitated infection in insect cells, but also in mammalian cells. Moreover, we found that AMPK is required for macropinocytosis, a major endocytic entry pathway for vaccinia. Furthermore, we show that AMPK contributes to other virus-independent actin-dependent processes including lamellipodia formation and wound healing, independent of the known AMPK activators LKB1 and CaMKK. Therefore, AMPK plays a highly conserved role in poxvirus infection and actin dynamics independent of its role as an energy regulator.
Collapse
Affiliation(s)
- Theresa S. Moser
- Department of Microbiology, Penn Genome Frontiers Institute, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Russell G. Jones
- Goodman Cancer Center and Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Craig B. Thompson
- Department of Cancer Biology, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Carolyn B. Coyne
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sara Cherry
- Department of Microbiology, Penn Genome Frontiers Institute, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
44
|
Manuel ER, Wang Z, Li Z, La Rosa C, Zhou W, Diamond DJ. Intergenic region 3 of modified vaccinia ankara is a functional site for insert gene expression and allows for potent antigen-specific immune responses. Virology 2010; 403:155-62. [PMID: 20471051 DOI: 10.1016/j.virol.2010.04.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/21/2009] [Accepted: 04/15/2010] [Indexed: 11/16/2022]
Abstract
Integration of exogenous DNA into modified vaccinia Ankara (MVA) is often accomplished using mapped deletion sites in the viral genome. Since MVA has a large capacity (> or =30kb) for foreign gene inserts and a limited number of unique integration sites, development of additional integration sites is needed to take full advantage of the extraordinary capacity for foreign gene insertion. In this report, we evaluate an alternative insertion site known as intergenic region 3 (IGR3). Recombinant MVA carrying the cytomegalovirus pp65 gene in IGR3 (rMVA-pp65-IGR3) demonstrated expression and genetic stability of the insert gene upon passage. Immunization of transgenic HLA-A2 mice with rMVA-pp65-IGR3 induced robust antigen-specific immune responses. Moreover, rMVA-pp65-IGR3-infected human EBV-transformed B cell lines were able to stimulate high levels of pp65-specific memory T cell responses in human PBMCs. These data support the usage of IGR3 for the development of highly immunogenic rMVA vaccines for clinical or veterinary use.
Collapse
Affiliation(s)
- Edwin R Manuel
- Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | |
Collapse
|
45
|
Immunogenicity and protective efficacy of mycobacterial DNA vaccines incorporating plasmid-encoded cytokines against Mycobacterium bovis. Immunol Cell Biol 2010; 88:651-7. [DOI: 10.1038/icb.2010.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
Calvo-Pinilla E, Rodríguez-Calvo T, Sevilla N, Ortego J. Heterologous prime boost vaccination with DNA and recombinant modified vaccinia virus Ankara protects IFNAR(−/−) mice against lethal bluetongue infection. Vaccine 2009; 28:437-45. [DOI: 10.1016/j.vaccine.2009.10.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 09/29/2009] [Accepted: 10/07/2009] [Indexed: 11/28/2022]
|
47
|
Backes S, Sperling KM, Zwilling J, Gasteiger G, Ludwig H, Kremmer E, Schwantes A, Staib C, Sutter G. Viral host-range factor C7 or K1 is essential for modified vaccinia virus Ankara late gene expression in human and murine cells, irrespective of their capacity to inhibit protein kinase R-mediated phosphorylation of eukaryotic translation initiation factor 2alpha. J Gen Virol 2009; 91:470-82. [PMID: 19846675 DOI: 10.1099/vir.0.015347-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Vaccinia virus (VACV) infection induces phosphorylation of eukaryotic translation initiation factor 2alpha (eIF2alpha), which inhibits cellular and viral protein synthesis. In turn, VACV has evolved the capacity to antagonize this antiviral response by expressing the viral host-range proteins K3 and E3. This study revealed that the host-range genes K1L and C7L also prevent eIF2alpha phosphorylation in modified VACV Ankara (MVA) infection of several human and murine cell lines. Moreover, C7L-deleted MVA (MVA-DeltaC7L) lacked late gene expression, which could be rescued by the function of host-range factor K1 or C7. It was demonstrated that viral gene expression was blocked after viral DNA replication and that it was independent of apoptosis induction. Furthermore, it was found that eIF2alpha phosphorylation in MVA-DeltaC7L-infected cells is mediated by protein kinase R (PKR) as shown in murine embryonic fibroblasts lacking PKR function, and it was shown that this was not due to reduced E3L gene expression. The block of eIF2alpha phosphorylation by C7 could be complemented by K1 in cells infected with MVA-DeltaC7L encoding a reinserted K1L gene (MVA-DeltaC7L-K1L). Importantly, these data illustrated that eIF2alpha phosphorylation by PKR is not responsible for the block of late viral gene expression. This suggests that other mechanisms targeted by C7 and K1 are essential for completing the MVA gene expression cycle and probably also for VACV replication in a diverse set of cell types.
Collapse
Affiliation(s)
- Simone Backes
- Institute of Virology, Technical University of Munich, and Helmholtz Center Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Risks associated with vaccinia virus in the laboratory. Virology 2009; 385:1-4. [DOI: 10.1016/j.virol.2008.11.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 11/23/2008] [Accepted: 11/26/2008] [Indexed: 11/23/2022]
|
49
|
Li J, Srivastava T, Rawal R, Manuel E, Isbell D, Tsark W, La Rosa C, Wang Z, Li Z, Barry PA, Hagen KD, Longmate J, Diamond DJ. Mamu-A01/K(b) transgenic and MHC Class I knockout mice as a tool for HIV vaccine development. Virology 2009; 387:16-28. [PMID: 19249807 DOI: 10.1016/j.virol.2009.01.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 09/23/2008] [Accepted: 01/26/2009] [Indexed: 01/09/2023]
Abstract
We have developed a murine model expressing the rhesus macaque (RM) Mamu-A01 MHC allele to characterize immune responses and vaccines based on antigens of importance to human disease processes. Towards that goal, transgenic (Tg) mice expressing chimeric RM (alpha1 and alpha2 Mamu-A01 domains) and murine (alpha3, transmembrane, and cytoplasmic H-2K(b) domains) MHC Class I molecules were derived by transgenesis of the H-2K(b)D(b) double MHC Class I knockout strain. After immunization of Mamu-A01/K(b) Tg mice with rVV-SIVGag-Pol, the mice generated CD8(+) T-cell IFN-gamma responses to several known Mamu-A01 restricted epitopes from the SIV Gag and Pol antigen sequence. Fusion peptides of highly recognized CTL epitopes from SIV Pol and Gag and a strong T-help epitope were shown to be immunogenic and capable of limiting an rVV-SIVGag-Pol challenge. Mamu-A01/K(b) Tg mice provide a model system to study the Mamu-A01 restricted T-cell response for various infectious diseases which are applicable to a study in RM.
Collapse
Affiliation(s)
- Jinliang Li
- Division of Translational Vaccine Research, Fox South, 1000B, Beckman Research Institute of the City of Hope, 1500 E. Duarte Rd., Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhao HP, Sun JF, Li N, Sun Y, Xia ZH, Wang Y, Cheng D, Qi QF, Jin ML, Qiu HJ. Assessment of the cell-mediated immunity induced by alphavirus replicon-vectored DNA vaccines against classical swine fever in a mouse model. Vet Immunol Immunopathol 2008; 129:57-65. [PMID: 19150136 DOI: 10.1016/j.vetimm.2008.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2008] [Revised: 12/01/2008] [Accepted: 12/04/2008] [Indexed: 11/30/2022]
Abstract
We have previously shown that an alphavirus replicon-vectored DNA vaccine (pSFV1CS-E2) encoding the E2 glycoprotein of classical swine fever virus (CSFV) completely protected the immunized pigs from lethal challenge. These animals developed only low or moderate level viral-specific antibody titers before challenge, implying that cell-mediated immunity (CMI) probably played an important role in the protective immunity against CSFV conferred by the DNA vaccine. In this study, the CMI induced by pSFV1CS-E2 and its derivative pSFV1CS-E2-UL49 encoding a fusion protein of CSFV E2 and pseudorabies virus (PRV) VP22 was evaluated in a mouse model by lymphoproliferation assays based on CFSE or WST-8, intracellular cytokine staining, and cytokine ELISA. The results showed that both vaccines induced CSFV-specific lymphoproliferative responses and cytokine production, and pSFV1CS-E2-UL49 induced stronger lymphoproliferative responses and higher cytokine levels than pSFV1CS-E2. These findings suggest that the alphavirus replicon-delivered DNA vaccines are capable of inducing CMI, and PRV VP22 is able to enhance the immunogenicity of the co-delivered antigen.
Collapse
Affiliation(s)
- He-Ping Zhao
- Division of Swine Infectious Diseases, National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|