1
|
Link EK, Tscherne A, Sutter G, Smith ER, Gurwith M, Chen RT, Volz A. A Brighton collaboration standardized template with key considerations for a benefit/risk assessment for a viral vector vaccine based on a non-replicating modified vaccinia virus Ankara viral vector. Vaccine 2025; 43:126521. [PMID: 39612556 DOI: 10.1016/j.vaccine.2024.126521] [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] [Received: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript provides an overview of Modified Vaccinia virus Ankara (MVA)-vectored vaccines and reviews molecular and biological key features of this platform. In particular, this review aims to provide fundamental information about the promising candidate vaccine MVA-MERS-S which has been evaluated successfully in different preclinical animal models and has undergone clinical testing including a phase Ib study involving more than 170 participants. Infectious diseases continue to be a major cause of human death worldwide. In this context, emerging zoonotic infectious diseases pose a particular challenge for public health systems. In the last two decades, three different respiratory coronaviruses, including the Middle East respiratory syndrome Coronavirus (MERS-CoV) have emerged. For many years, safe and efficacious vaccines have been a major tool to combat infectious diseases. Here, we report on a promising candidate vaccine (MVA-MERS-S) against MERS-CoV based on MVA. Upon application, MVA-MERS-S has been well tolerated and immunogenic, inducing both, cellular and humoral immune responses in different animal models and humans. We demonstrate that the MVA vector platform, with the example of MVA-MERS-S, is a viable and effective tool for producing safe, immunogenic, and efficient vaccines against emerging infectious diseases.
Collapse
Affiliation(s)
- Ellen K Link
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Alina Tscherne
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Gerd Sutter
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Emily R Smith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA.
| | - Marc Gurwith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T Chen
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Asisa Volz
- University of Veterinary Medicine Hannover, Institute of Virology, Hannover, Germany
| |
Collapse
|
2
|
Gómez-Carballa A, Albericio G, Montoto-Louzao J, Pérez P, Astorgano D, Rivero-Calle I, Martinón-Torres F, Esteban M, Salas A, García-Arriaza J. Lung transcriptomics of K18-hACE2 mice highlights mechanisms and genes involved in the MVA-S vaccine-mediated immune response and protection against SARS-CoV-2 infection. Antiviral Res 2023; 220:105760. [PMID: 37992765 DOI: 10.1016/j.antiviral.2023.105760] [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] [Received: 07/27/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023]
Abstract
Unravelling the molecular mechanism of COVID-19 vaccines through transcriptomic pathways involved in the host response to SARS-CoV-2 infection is key to understand how vaccines work, and for the development of optimized COVID-19 vaccines that can prevent the emergence of SARS-CoV-2 variants of concern (VoCs) and future outbreaks. In this study, we investigated the effects of vaccination with a modified vaccinia virus Ankara (MVA)-based vector expressing the full-length SARS-CoV-2 spike protein (MVA-S) on the lung transcriptome from susceptible K18-hACE2 mice after SARS-CoV-2 infection. One dose of MVA-S regulated genes related to viral infection control, inflammation processes, T-cell response, cytokine production and IFN-γ signalling. Down-regulation of Rhcg and Tnfsf18 genes post-vaccination with one and two doses of MVA-S may represent a mechanism for controlling infection immunity and vaccine-induced protection. One dose of MVA-S provided partial protection with a distinct lung transcriptomic profile to healthy animals, while two doses of MVA-S fully protected against infection with a transcriptomic profile comparable to that of non-vaccinated healthy animals. This suggests that the MVA-S booster generates a robust and rapid antigen-specific immune response preventing virus infection. Notably, down-regulation of Atf3 and Zbtb16 genes in mice vaccinated with two doses of MVA-S may contribute to vaccine control of innate immune system and inflammation processes in the lungs during SARS-CoV-2 infection. This study shows host transcriptomic mechanisms likely involved in the MVA-S vaccine-mediated immune response against SARS-CoV-2 infection, which could help in improving vaccine dose assessment and developing novel, well-optimized SARS-CoV-2 vaccine candidates against prevalent or emerging VoCs.
Collapse
Affiliation(s)
- Alberto Gómez-Carballa
- Genetics, Vaccines and Infectious Diseases Research Group (Genvip), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Julián Montoto-Louzao
- Genetics, Vaccines and Infectious Diseases Research Group (Genvip), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines and Infectious Diseases Research Group (Genvip), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infectious Diseases Research Group (Genvip), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Antonio Salas
- Genetics, Vaccines and Infectious Diseases Research Group (Genvip), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| |
Collapse
|
3
|
Falqui M, Perdiguero B, Coloma R, Albert M, Marcos-Villar L, McGrail JP, Sorzano CÓS, Esteban M, Gómez CE, Guerra S. An MVA-based vector expressing cell-free ISG15 increases IFN-I production and improves HIV-1-specific CD8 T cell immune responses. Front Cell Infect Microbiol 2023; 13:1187193. [PMID: 37313341 PMCID: PMC10258332 DOI: 10.3389/fcimb.2023.1187193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 06/15/2023] Open
Abstract
The human immunodeficiency virus (HIV), responsible of the Acquired Immune Deficiency Syndrome (AIDS), continues to be a major global public health issue with any cure or vaccine available. The Interferon-stimulated gene 15 (ISG15) encodes a ubiquitin-like protein that is induced by interferons and plays a critical role in the immune response. ISG15 is a modifier protein that covalently binds to its targets via a reversible bond, a process known as ISGylation, which is the best-characterized activity of this protein to date. However, ISG15 can also interact with intracellular proteins via non-covalent binding or act as a cytokine in the extracellular space after secretion. In previous studies we proved the adjuvant effect of ISG15 when delivered by a DNA-vector in heterologous prime-boost combination with a Modified Vaccinia virus Ankara (MVA)-based recombinant virus expressing HIV-1 antigens Env/Gag-Pol-Nef (MVA-B). Here we extended these results evaluating the adjuvant effect of ISG15 when expressed by an MVA vector. For this, we generated and characterized two novel MVA recombinants expressing different forms of ISG15, the wild-type ISG15GG (able to perform ISGylation) or the mutated ISG15AA (unable to perform ISGylation). In mice immunized with the heterologous DNA prime/MVA boost regimen, the expression of the mutant ISG15AA from MVA-Δ3-ISG15AA vector in combination with MVA-B induced an increase in the magnitude and quality of HIV-1-specific CD8 T cells as well as in the levels of IFN-I released, providing a better immunostimulatory activity than the wild-type ISG15GG. Our results confirm the importance of ISG15 as an immune adjuvant in the vaccine field and highlights its role as a potential relevant component in HIV-1 immunization protocols.
Collapse
Affiliation(s)
- Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rocio Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Albert
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Joseph Patrick McGrail
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Óscar S. Sorzano
- Biocomputing Unit and Computational Genomics, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
4
|
Pérez P, Astorgano D, Albericio G, Flores S, Sánchez-Cordón PJ, Luczkowiak J, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. Intranasal administration of a single dose of MVA-based vaccine candidates against COVID-19 induced local and systemic immune responses and protects mice from a lethal SARS-CoV-2 infection. Front Immunol 2022; 13:995235. [PMID: 36172368 PMCID: PMC9510595 DOI: 10.3389/fimmu.2022.995235] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Current coronavirus disease-19 (COVID-19) vaccines are administered by the intramuscular route, but this vaccine administration failed to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infection in the upper respiratory tract, mainly due to the absence of virus-specific mucosal immune responses. It is hypothesized that intranasal (IN) vaccination could induce both mucosal and systemic immune responses that blocked SARS-CoV-2 transmission and COVID-19 progression. Here, we evaluated in mice IN administration of three modified vaccinia virus Ankara (MVA)-based vaccine candidates expressing the SARS-CoV-2 spike (S) protein, either the full-length native S or a prefusion-stabilized [S(3P)] protein; SARS-CoV-2-specific immune responses and efficacy were determined after a single IN vaccine application. Results showed that in C57BL/6 mice, MVA-based vaccine candidates elicited S-specific IgG and IgA antibodies in serum and bronchoalveolar lavages, respectively, and neutralizing antibodies against parental and SARS-CoV-2 variants of concern (VoC), with MVA-S(3P) being the most immunogenic vaccine candidate. IN vaccine administration also induced polyfunctional S-specific Th1-skewed CD4+ and cytotoxic CD8+ T-cell immune responses locally (in lungs and bronchoalveolar lymph nodes) or systemically (in spleen). Remarkably, a single IN vaccine dose protected susceptible K18-hACE2 transgenic mice from morbidity and mortality caused by SARS-CoV-2 infection, with MVA-S(3P) being the most effective candidate. Infectious SARS-CoV-2 viruses were undetectable in lungs and nasal washes, correlating with high titers of S-specific IgGs and neutralizing antibodies against parental SARS-CoV-2 and several VoC. Moreover, low histopathological lung lesions and low levels of pro-inflammatory cytokines in lungs and nasal washes were detected in vaccinated animals. These results demonstrated that a single IN inoculation of our MVA-based vaccine candidates induced potent immune responses, either locally or systemically, and protected animal models from COVID-19. These results also identified an effective vaccine administration route to induce mucosal immunity that should prevent SARS-CoV-2 host-to-host transmission.
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sara Flores
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pedro J. Sánchez-Cordón
- Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Joanna Luczkowiak
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Microbiology, Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Department of Microbiology, Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - José M. Casasnovas
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- *Correspondence: Mariano Esteban, ; Juan García-Arriaza,
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- *Correspondence: Mariano Esteban, ; Juan García-Arriaza,
| |
Collapse
|
5
|
Boudewijns R, Pérez P, Lázaro-Frías A, Van Looveren D, Vercruysse T, Thibaut HJ, Weynand B, Coelmont L, Neyts J, Astorgano D, Montenegro D, Puentes E, Rodríguez E, Dallmeier K, Esteban M, García-Arriaza J. MVA-CoV2-S Vaccine Candidate Neutralizes Distinct Variants of Concern and Protects Against SARS-CoV-2 Infection in Hamsters. Front Immunol 2022; 13:845969. [PMID: 35371064 PMCID: PMC8966703 DOI: 10.3389/fimmu.2022.845969] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/17/2022] [Indexed: 12/31/2022] Open
Abstract
To control the coronavirus disease 2019 (COVID-19) pandemic and the emergence of different variants of concern (VoCs), novel vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are needed. In this study, we report the potent immunogenicity and efficacy induced in hamsters by a vaccine candidate based on a modified vaccinia virus Ankara (MVA) vector expressing a human codon optimized full-length SARS-CoV-2 spike (S) protein (MVA-S). Immunization with one or two doses of MVA-S elicited high titers of S- and receptor-binding domain (RBD)-binding IgG antibodies and neutralizing antibodies against parental SARS-CoV-2 and VoC alpha, beta, gamma, delta, and omicron. After SARS-CoV-2 challenge, MVA-S-vaccinated hamsters showed a significantly strong reduction of viral RNA and infectious virus in the lungs compared to the MVA-WT control group. Moreover, a marked reduction in lung histopathology was also observed in MVA-S-vaccinated hamsters. These results favor the use of MVA-S as a potential vaccine candidate for SARS-CoV-2 in clinical trials.
Collapse
Affiliation(s)
- Robbert Boudewijns
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Adrián Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Dominique Van Looveren
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, Leuven, Belgium
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | | | | | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
6
|
Mooij P, García-Arriaza J, Pérez P, Lázaro-Frías A, Verstrepen BE, Böszörményi KP, Mortier D, Fagrouch Z, Kiemenyi-Kayere G, Niphuis H, Acar RF, Meijer L, Stammes MA, Kondova I, Verschoor EJ, GeurtsvanKessel CH, de Bruin E, Sikkema RS, Luczkowiak J, Delgado R, Montenegro D, Puentes E, Rodríguez E, Bogers WMJM, Koopman G, Esteban M. Poxvirus MVA Expressing SARS-CoV-2 S Protein Induces Robust Immunity and Protects Rhesus Macaques From SARS-CoV-2. Front Immunol 2022; 13:845887. [PMID: 35371043 PMCID: PMC8966779 DOI: 10.3389/fimmu.2022.845887] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
Novel safe, immunogenic, and effective vaccines are needed to control the COVID-19 pandemic, caused by SARS-CoV-2. Here, we describe the safety, robust immunogenicity, and potent efficacy elicited in rhesus macaques by a modified vaccinia virus Ankara (MVA) vector expressing a full-length SARS-CoV-2 spike (S) protein (MVA-S). MVA-S vaccination was well tolerated and induced S and receptor-binding domain (RBD)-binding IgG antibodies and neutralizing antibodies against SARS-CoV-2 and several variants of concern. S-specific IFNγ, but not IL-4, -producing cells were also elicited. After SARS-CoV-2 challenge, vaccinated animals showed a significant strong reduction of virus loads in bronchoalveolar lavages (BAL) and decreased levels in throat and nasal mucosa. Remarkably, MVA-S also protected macaques from fever and infection-induced cytokine storm. Computed tomography and histological examination of the lungs showed reduced lung pathology in MVA-S-vaccinated animals. These findings favor the use of MVA-S as a potential vaccine for SARS-CoV-2 in clinical trials.
Collapse
Affiliation(s)
- Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Adrian Lázaro-Frías
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Babs E. Verstrepen
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Kinga P. Böszörményi
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | | | - Henk Niphuis
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Roja Fidel Acar
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Lisette Meijer
- Department of Parasitology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Marieke A. Stammes
- Department of Parasitology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Ivanela Kondova
- Animal Science Department, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Ernst J. Verschoor
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | | | - Erwin de Bruin
- Department of Viroscience, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Reina S. Sikkema
- Department of Viroscience, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense School of Medicine, Madrid, Spain
| | | | | | | | - Willy M. J. M. Bogers
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, Netherlands
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
7
|
Ding Y, Fan J, Deng L, Peng Y, Zhou B, Huang B. Evaluation of Tumor Specificity and Immunity of Thymidine Kinase-Deleted Vaccinia Virus Guang9 Strain. Onco Targets Ther 2020; 13:7683-7697. [PMID: 32801778 PMCID: PMC7415446 DOI: 10.2147/ott.s260288] [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: 04/28/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose Oncolytic viruses are emerging as promising options for clinical cancer treatment due to their inherent ability of tumor tropism and oncolytic property. Aside from tumor lysis, oncolytic viruses can induce host immune responses against tumor cells and may thus be viewed as a form of immunotherapy. Methods The attenuated vaccinia VG9-Luc, which originated from Chinese vaccinia Tian Tan strain, was constructed to express firefly luciferase for bioluminescence imaging and to disrupt the thymidine kinase gene for promoting tumor specificity. An in vivo bioluminescence imaging was performed to observe the virus distribution in live mice. The titers of neutralizing antiviral and antitumor antibodies in plasma were determined by time-resolved fluoroimmunoassay. Results Except BALB/c mice treated with intravenous virus injection, all immunocompromised and immunocompetent mice showed obvious tumor targeting ability of vaccinia VG9-Luc. Besides, host immune response activated by vaccinia VG9-Luc showed the production of antiviral and antitumor antibodies, the process of which was similar between intravenous and intratumoral viral delivery systems. The results indicated that virus infection promoted tumor-specific immunity by increasing the production of antitumor antibodies. Moreover, virus reinjection was performed and a more rapid viral clearance was observed in immunocompetent mice compared with first virus infection. Conclusion The thymidine kinase-deleted vaccinia Guang9 strain, which has the properties of tumor specificity and antitumor immunity, is a promising candidate vector for cancer therapy.
Collapse
Affiliation(s)
- Yuedi Ding
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Jun Fan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Lili Deng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Bin Zhou
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| | - Biao Huang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, People's Republic of China
| |
Collapse
|
8
|
Perdiguero B, Gómez CE, Esteban M. Bioluminescence Imaging as a Tool for Poxvirus Biology. Methods Mol Biol 2019; 2023:269-285. [PMID: 31240684 DOI: 10.1007/978-1-4939-9593-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bioluminescence imaging, with luciferase as a reporter-encoding gene, has been successfully and widely used for studies to follow viral infection in an organism and to measure therapeutic efficacy of antiviral agents in small animal models. Bioluminescence is produced by the reaction of a luciferase enzyme stably inserted into the viral genome with a defined substrate systemically delivered into the animal. The light emitted is captured allowing the detection of viral infection sites and the quantification of viral replication in the context of tissues of a living animal. The goal of this chapter is to provide a technical background for the evaluation of poxvirus infection in cells and animals through bioluminescence imaging technology using luciferase-expressing recombinant poxviruses.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain.
| |
Collapse
|
9
|
Elshina E, Allen ER, Flaxman A, van Diemen PM, Milicic A, Rollier CS, Yamaguchi Y, Wyllie DH. Vaccination with the Staphylococcus aureus secreted proteins EapH1 and EapH2 impacts both S. aureus carriage and invasive disease. Vaccine 2018; 37:502-509. [PMID: 30502067 DOI: 10.1016/j.vaccine.2018.11.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION There is a need for an efficacious vaccine reducing infections due to Staphylococcus aureus, a common cause of community and hospital infection. Infecting organisms originate from S. aureus populations colonising the nares and bowel. Antimicrobials are widely used to transiently reduce S. aureus colonisation prior to surgery, a practice which is selecting for resistant S. aureus isolates. S. aureus secretes multiple proteins, including the protease inhibitors extracellular adhesion protein homologue 1 and 2 (EapH1 and EapH2). METHODS Mice were vaccinated intramuscularly or intranasally with Adenovirus serotype 5 and Modified Vaccinia Ankara viral vectors expressing EapH1 and EapH2 proteins, or with control viruses. Using murine S. aureus colonisation models, we monitored S. aureus colonisation by sequential stool sampling. Monitoring of S. aureus invasive disease after intravenous challenge was performed using bacterial load and abscess numbers in the kidney. RESULTS Intramuscular vaccination with Adenovirus serotype 5 and Modified Vaccinia Ankara viral vectors expressing EapH1 and EapH2 proteins significantly reduces bacterial recovery in the murine renal abscess model of infection, but the magnitude of the effect is small. A single intranasal vaccination with an adenoviral vaccine expressing these proteins reduced S. aureus gastrointestinal (GI) tract colonisation. CONCLUSION Vaccination against EapH1 / EapH2 proteins may offer an antibiotic independent way to reduce S. aureus colonisation, as well as contributing to protection against S. aureus invasive disease.
Collapse
Affiliation(s)
- Elizaveta Elshina
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Elizabeth R Allen
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Amy Flaxman
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Pauline M van Diemen
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Anita Milicic
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, United Kingdom
| | - Yuko Yamaguchi
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - David H Wyllie
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom.
| |
Collapse
|
10
|
Rosenbaum P, Tchitchek N, Joly C, Stimmer L, Hocini H, Dereuddre-Bosquet N, Beignon AS, Chapon C, Levy Y, Le Grand R, Martinon F. Molecular and Cellular Dynamics in the Skin, the Lymph Nodes, and the Blood of the Immune Response to Intradermal Injection of Modified Vaccinia Ankara Vaccine. Front Immunol 2018; 9:870. [PMID: 29922280 PMCID: PMC5996922 DOI: 10.3389/fimmu.2018.00870] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/09/2018] [Indexed: 01/05/2023] Open
Abstract
New vaccine design approaches would be greatly facilitated by a better understanding of the early systemic changes, and those that occur at the site of injection, responsible for the installation of a durable and oriented protective response. We performed a detailed characterization of very early infection and host response events following the intradermal administration of the modified vaccinia virus Ankara as a live attenuated vaccine model in non-human primates. Integrated analysis of the data obtained from in vivo imaging, histology, flow cytometry, multiplex cytokine, and transcriptomic analysis using tools derived from systems biology, such as co-expression networks, showed a strong early local and systemic inflammatory response that peaked at 24 h, which was then progressively replaced by an adaptive response during the installation of the host response to the vaccine. Granulocytes, macrophages, and monocytoid cells were massively recruited during the local innate response in association with local productions of GM-CSF, IL-1β, MIP1α, MIP1β, and TNFα. We also observed a rapid and transient granulocyte recruitment and the release of IL-6 and IL-1RA, followed by a persistent phase involving inflammatory monocytes. This systemic inflammation was confirmed by molecular signatures, such as upregulations of IL-6 and TNF pathways and acute phase response signaling. Such comprehensive approaches improve our understanding of the spatiotemporal orchestration of vaccine-elicited immune response, in a live-attenuated vaccine model, and thus contribute to rational vaccine development.
Collapse
Affiliation(s)
- Pierre Rosenbaum
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France.,Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Nicolas Tchitchek
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France
| | - Candie Joly
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France
| | - Lev Stimmer
- CEA - INSERM, MIRCen, UMS27, Fontenay-aux-Roses, France.,INSERM U1169, Kremlin-Bicêtre, France
| | - Hakim Hocini
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France.,INSERM U955, Henri Mondor Hospital, University of Paris East, Créteil, France
| | - Nathalie Dereuddre-Bosquet
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France.,Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France.,Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Yves Levy
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France.,INSERM U955, Henri Mondor Hospital, University of Paris East, Créteil, France
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France.,Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France.,Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| |
Collapse
|
11
|
Swindle S. Biosafety in Handling Gene Transfer Vectors. ACTA ACUST UNITED AC 2018; 96:12.1.1-12.1.17. [DOI: 10.1002/cphg.54] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Scott Swindle
- Occupational Medicine and Research Safety, The University of Alabama at Birmingham Birmingham Alabama
| |
Collapse
|
12
|
Kadoki M, Patil A, Thaiss CC, Brooks DJ, Pandey S, Deep D, Alvarez D, von Andrian UH, Wagers AJ, Nakai K, Mikkelsen TS, Soumillon M, Chevrier N. Organism-Level Analysis of Vaccination Reveals Networks of Protection across Tissues. Cell 2017; 171:398-413.e21. [PMID: 28942919 DOI: 10.1016/j.cell.2017.08.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/24/2017] [Accepted: 08/14/2017] [Indexed: 02/07/2023]
Abstract
A fundamental challenge in immunology is to decipher the principles governing immune responses at the whole-organism scale. Here, using a comparative infection model, we observe immune signal propagation within and between organs to obtain a dynamic map of immune processes at the organism level. We uncover two inter-organ mechanisms of protective immunity mediated by soluble and cellular factors. First, analyzing ligand-receptor connectivity across tissues reveals that type I IFNs trigger a whole-body antiviral state, protecting the host within hours after skin vaccination. Second, combining parabiosis, single-cell analyses, and gene knockouts, we uncover a multi-organ web of tissue-resident memory T cells that functionally adapt to their environment to stop viral spread across the organism. These results have implications for manipulating tissue-resident memory T cells through vaccination and open up new lines of inquiry for the analysis of immune responses at the organism level.
Collapse
Affiliation(s)
- Motohiko Kadoki
- Faculty of Arts & Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ashwini Patil
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Cornelius C Thaiss
- Faculty of Arts & Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Donald J Brooks
- Faculty of Arts & Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Surya Pandey
- Faculty of Arts & Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Deeksha Deep
- Faculty of Arts & Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - David Alvarez
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Kenta Nakai
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tarjei S Mikkelsen
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Magali Soumillon
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Nicolas Chevrier
- Faculty of Arts & Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
13
|
Townsend DG, Trivedi S, Jackson RJ, Ranasinghe C. Recombinant fowlpox virus vector-based vaccines: expression kinetics, dissemination and safety profile following intranasal delivery. J Gen Virol 2017; 98:496-505. [PMID: 28056224 PMCID: PMC5797952 DOI: 10.1099/jgv.0.000702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/03/2017] [Indexed: 12/23/2022] Open
Abstract
We have previously established that mucosal uptake of recombinant fowlpox virus (rFPV) vaccines is far superior to other vector-based vaccines. Specifically, intranasal priming with rFPV vaccines can recruit unique antigen-presenting cells, which induce excellent mucosal and systemic HIV-specific CD8+ T-cell immunity. In this study, we have for the first time investigated the in vivo dissemination, safety and expression kinetics of rFPV post intranasal delivery using recombinant viruses expressing green fluorescent protein or mCherry. Both confocal microscopy of tissue sections using green fluorescent protein and in vivo Imaging System (IVIS) spectrum live animal and whole organ imaging studies using mCherry revealed that (i) the peak antigen expression occurs 12 to 24 h post vaccination and no active viral gene expression is detected 96 h post vaccination. (ii) The virus only infects the initial vaccination site (lung and nasal cavity) and does not disseminate to distal sites such as the spleen or gut. (iii) More importantly, rFPV does not cross the olfactory receptor neuron pathway. Collectively, our findings indicate that rFPV vector-based vaccines have all the hallmarks of a safe and effective mucosal delivery vector, suitable for clinical evaluation.
Collapse
Affiliation(s)
- David G Townsend
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Shubhanshi Trivedi
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
- Present address: Division of Infectious Diseases, Department of Internal Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Ronald J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| |
Collapse
|
14
|
Del Medico Zajac MP, Zanetti FA, Esusy MS, Federico CR, Zabal O, Valera AR, Calamante G. Induction of Both Local Immune Response in Mice and Protection in a Rabbit Model by Intranasal Immunization with Modified Vaccinia Ankara Virus Expressing a Secreted Form of Bovine Herpesvirus 1 Glycoprotein D. Viral Immunol 2016; 30:70-76. [PMID: 27809679 DOI: 10.1089/vim.2016.0090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In this study, we evaluated the immunogenicity and efficacy of mucosal delivery of a recombinant modified vaccinia Ankara virus (MVA) expressing the secreted version of bovine herpesvirus type 1 (BoHV-1) glycoprotein D (MVA-gDs) without addition of adjuvant in two animal models. First, we demonstrated the capability of MVA-gDs of inducing both local and systemic anti-gD humoral immune response after intranasal immunization of mice. Then, we confirmed that two doses of MVA-gDs administered intranasally to rabbits induced systemic anti-gD antibodies and conferred protection against BoHV-1 challenge. Our results show the potential of using MVA as a vector for the rational design of veterinary vaccines capable of inducing specific and protective immune responses both at local and systemic level.
Collapse
Affiliation(s)
- María Paula Del Medico Zajac
- 1 Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria (CICVyA-INTA) , Hurlingham, Argentina .,2 Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Ciudad Autónoma de Buenos Aires, Argentina
| | - Flavia Adriana Zanetti
- 1 Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria (CICVyA-INTA) , Hurlingham, Argentina .,2 Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Ciudad Autónoma de Buenos Aires, Argentina
| | - María Soledad Esusy
- 1 Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria (CICVyA-INTA) , Hurlingham, Argentina
| | - Carlos Rodolfo Federico
- 1 Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria (CICVyA-INTA) , Hurlingham, Argentina .,2 Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Ciudad Autónoma de Buenos Aires, Argentina
| | - Osvaldo Zabal
- 3 Instituto de Virología, Instituto Nacional de Tecnología Agropecuaria (CICVyA-INTA) , Hurlingham, Argentina
| | - Alejandro Rafael Valera
- 4 Cátedra de Virología, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata , La Plata, Argentina
| | - Gabriela Calamante
- 1 Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria (CICVyA-INTA) , Hurlingham, Argentina
| |
Collapse
|
15
|
Affiliation(s)
- Andrew Mehle
- Medical Microbiology and Immunology, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
16
|
Goossens M, Pauwels K, Willemarck N, Breyer D. Environmental risk assessment of clinical trials involving modified vaccinia virus Ankara (MVA)-based vectors. Curr Gene Ther 2014; 13:413-20. [PMID: 24397528 PMCID: PMC4031919 DOI: 10.2174/156652321306140103221941] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/18/2013] [Accepted: 09/18/2013] [Indexed: 12/05/2022]
Abstract
The modified vaccinia virus Ankara (MVA) strain, which has been developed as a vaccine against smallpox, is
since the nineties widely tested in clinical trials as recombinant vector for vaccination or gene therapy applications. Although
MVA is renowned for its safety, several biosafety aspects need to be considered when performing the risk assessment
of a recombinant MVA (rMVA). This paper presents the biosafety issues and the main lessons learned from the
evaluation of the clinical trials with rMVA performed in Belgium. Factors such as the specific characteristics of the
rMVA, the inserted foreign sequences/transgene, its ability for reconversion, recombination and dissemination in the
population and the environment are the main points of attention. Measures to prevent or manage identified risks are also
discussed.
Collapse
Affiliation(s)
| | | | | | - Didier Breyer
- Scientific Institute of Public Health, Biosafety and Biotechnology Unit, Rue J. Wytsmanstraat 14, B- 1050 Brussels, Belgium.
| |
Collapse
|
17
|
Rapid expansion of CD8+ T cells in wild-type and type I interferon receptor-deficient mice correlates with protection after low-dose emergency immunization with modified vaccinia virus Ankara. J Virol 2014; 88:10946-57. [PMID: 25008931 DOI: 10.1128/jvi.00945-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
UNLABELLED Immunization with modified vaccinia virus Ankara (MVA) can rapidly protect mice against lethal ectromelia virus (ECTV) infection, serving as an experimental model for severe systemic infections. Importantly, this early protective capacity of MVA vaccination completely depends on virus-specific cytotoxic CD8(+) T cell responses. We used MVA vaccination in the mousepox challenge model using ECTV infection to investigate the previously unknown factors required to elicit rapid protective T cell immunity in normal C57BL/6 mice and in mice lacking the interferon alpha/beta receptor (IFNAR(-/-)). We found a minimal dose of 10(5) PFU of MVA vaccine fully sufficient to allow robust protection against lethal mousepox, as assessed by the absence of disease symptoms and failure to detect ECTV in organs from vaccinated animals. Moreover, MVA immunization at low dosage also protected IFNAR(-/-) mice, indicating efficient activation of cellular immunity even in the absence of type I interferon signaling. When monitoring for virus-specific CD8(+) T cell responses in mice vaccinated with the minimal protective dose of MVA, we found significantly enhanced levels of antigen-specific T cells in animals that were MVA vaccinated and ECTV challenged compared to mice that were only vaccinated. The initial priming of naive CD8(+) T cells by MVA immunization appears to be highly efficient and, even at low doses, mediates a rapid in vivo burst of pathogen-specific T cells upon challenge. Our findings define striking requirements for protective emergency immunization against severe systemic infections with orthopoxviruses. IMPORTANCE We demonstrate that single-shot low-dose immunizations with vaccinia virus MVA can rapidly induce T cell-mediated protective immunity against lethal orthopoxvirus infections. Our data provide new evidence for an efficient protective capacity of vaccination with replication-deficient MVA. These data are of important practical relevance for public health, as the effectiveness of a safety-tested, next-generation smallpox vaccine based on MVA is still debated. Furthermore, producing sufficient amounts of vaccine is expected to be a major challenge should an outbreak occur. Moreover, prevention of other infections may require rapidly protective immunization; hence, MVA could be an extremely useful vaccine for delivering heterologous T cell antigens, particularly for infectious diseases that fit a scenario of emergency vaccination.
Collapse
|
18
|
Cáceres A, Perdiguero B, Gómez CE, Cepeda MV, Caelles C, Sorzano CO, Esteban M. Involvement of the cellular phosphatase DUSP1 in vaccinia virus infection. PLoS Pathog 2013; 9:e1003719. [PMID: 24244156 PMCID: PMC3828168 DOI: 10.1371/journal.ppat.1003719] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 09/05/2013] [Indexed: 12/30/2022] Open
Abstract
Poxviruses encode a large variety of proteins that mimic, block or enhance host cell signaling pathways on their own benefit. It has been reported that mitogen-activated protein kinases (MAPKs) are specifically upregulated during vaccinia virus (VACV) infection. Here, we have evaluated the role of the MAPK negative regulator dual specificity phosphatase 1 (DUSP1) in the infection of VACV. We demonstrated that DUSP1 expression is enhanced upon infection with the replicative WR virus and with the attenuated VACV viruses MVA and NYVAC. This upregulation is dependent on early viral gene expression. In the absence of DUSP1 in cultured cells, there is an increased activation of its molecular targets JNK and ERK and an enhanced WR replication. Moreover, DUSP1 knock-out (KO) mice are more susceptible to WR infection as a result of enhanced virus replication in the lungs. Significantly, MVA, which is known to produce non-permissive infections in most mammalian cell lines, is able to grow in DUSP1 KO immortalized murine embryo fibroblasts (MEFs). By confocal and electron microscopy assays, we showed that in the absence of DUSP1 MVA morphogenesis is similar as in permissive cell lines and demonstrated that DUSP1 is involved at the stage of transition between IVN and MV in VACV morphogenesis. In addition, we have observed that the secretion of pro-inflammatory cytokines at early times post-infection in KO mice infected with MVA and NYVAC is increased and that the adaptive immune response is enhanced in comparison with WT-infected mice. Altogether, these findings reveal that DUSP1 is involved in the replication and host range of VACV and in the regulation of host immune responses through the modulation of MAPKs. Thus, in this study we demonstrate that DUSP1 is actively involved in the antiviral host defense mechanism against a poxvirus infection. Phosphorylation is a post-translational modification that is highly conserved throughout the animal kingdom. Viruses have evolved to acquire their own kinases and phosphatases and to be able to modulate host phosphorylation mechanisms on their benefit. DUSP1 is an early induced gene that belongs to the superfamily of Dual-specificity phosphatases and provides an essential negative feedback regulation of MAPKs. DUSP1 is involved in innate and adaptive immune responses against different bacteria and parasites infections. The use of Knock-out technology has allowed us to understand the role of DUSP1 in the context of VACV infection both in cultured cells and in the in vivo mouse model. Here, we have showed that DUSP1 expression is upregulated during VACV infection and that DUSP1 plays an important role in VACV replication. Interestingly, we have demonstrated that the VACV attenuated virus MVA is able to grow in immortalized murine embryo fibroblasts in the absence of DUSP1. In vivo results showed that VACV replication-competent WR pathogenesis is enhanced in the absence of DUSP1. Furthermore, we have demonstrated that DUSP1 is involved in the host innate and adaptive responses against VACV. Altogether, we have presented a novel role for DUSP1 in VACV replication and anti-VACV host immune response.
Collapse
Affiliation(s)
- Ana Cáceres
- Department of Molecular and Cellular Biology, National Centre of Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, National Centre of Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen E. Gómez
- Department of Molecular and Cellular Biology, National Centre of Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Maria Victoria Cepeda
- Department of Molecular and Cellular Biology, National Centre of Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carme Caelles
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Carlos Oscar Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, National Centre of Biotechnology, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
19
|
Gómez CE, Perdiguero B, García-Arriaza J, Esteban M. Clinical applications of attenuated MVA poxvirus strain. Expert Rev Vaccines 2013; 12:1395-416. [PMID: 24168097 DOI: 10.1586/14760584.2013.845531] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The highly attenuated poxvirus strain modified vaccinia virus Ankara (MVA) has reached maturity as a vector delivery system and as a vaccine candidate against a broad spectrum of diseases. This has been largely recognized from research on virus-host cell interactions and immunological studies in pre-clinical and clinical trials. This review addresses the studies of MVA vectors used in phase I/II clinical trials, with the aim to provide the main findings obtained on their behavior when tested against relevant human diseases and cancer and also highlights the strategies currently implemented to improve the MVA immunogenicity. The authors assess that MVA vectors are progressing as strong vaccine candidates either alone or when administered in combination with other vectors.
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | | | | |
Collapse
|
20
|
Attenuated and replication-competent vaccinia virus strains M65 and M101 with distinct biology and immunogenicity as potential vaccine candidates against pathogens. J Virol 2013; 87:6955-74. [PMID: 23596295 DOI: 10.1128/jvi.03013-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Replication-competent poxvirus vectors with an attenuation phenotype and with a high immunogenic capacity of the foreign expressed antigen are being pursued as novel vaccine vectors against different pathogens. In this investigation, we have examined the replication and immunogenic characteristics of two vaccinia virus (VACV) mutants, M65 and M101. These mutants were generated after 65 and 101 serial passages of persistently infected Friend erythroleukemia (FEL) cells. In cultured cells of different origins, the mutants are replication competent and have growth kinetics similar to or slightly reduced in comparison with those of the parental Western Reserve (WR) virus strain. In normal and immune-suppressed infected mice, the mutants showed different levels of attenuation and pathogenicity in comparison with WR and modified vaccinia Ankara (MVA) strains. Wide genome analysis after deep sequencing revealed selected genomic deletions and mutations in a number of viral open reading frames (ORFs). Mice immunized in a DNA prime/mutant boost regimen with viral vectors expressing the LACK (Leishmania homologue for receptors of activated C kinase) antigen of Leishmania infantum showed protection or a delay in the onset of cutaneous leishmaniasis. Protection was similar to that triggered by MVA-LACK. In immunized mice, both polyfunctional CD4(+) and CD8(+) T cells with an effector memory phenotype were activated by the two mutants, but the DNA-LACK/M65-LACK protocol preferentially induced CD4(+) whereas DNA-LACK/M101-LACK preferentially induced CD8(+) T cell responses. Altogether, our findings showed the adaptive changes of the WR genome during long-term virus-host cell interaction and how the replication competency of M65 and M101 mutants confers distinct biological properties and immunogenicity in mice compared to those of the MVA strain. These mutants could have applicability for understanding VACV biology and as potential vaccine vectors against pathogens and tumors.
Collapse
|
21
|
Bethell D, Saunders D, Jongkaewwattana A, Kramyu J, Thitithayanont A, Wiboon-ut S, Yongvanitchit K, Limsalakpetch A, Kum-Arb U, Uthaimongkol N, Garcia JM, Timmermans AE, Peiris M, Thomas S, Engering A, Jarman RG, Mongkolsirichaikul D, Mason C, Khemnu N, Tyner SD, Fukuda MM, Walsh DS, Pichyangkul S. Evaluation of in vitro cross-reactivity to avian H5N1 and pandemic H1N1 2009 influenza following prime boost regimens of seasonal influenza vaccination in healthy human subjects: a randomised trial. PLoS One 2013; 8:e59674. [PMID: 23555741 PMCID: PMC3608534 DOI: 10.1371/journal.pone.0059674] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/16/2013] [Indexed: 11/19/2022] Open
Abstract
Introduction Recent studies have demonstrated that inactivated seasonal influenza vaccines (IIV) may elicit production of heterosubtypic antibodies, which can neutralize avian H5N1 virus in a small proportion of subjects. We hypothesized that prime boost regimens of live and inactivated trivalent seasonal influenza vaccines (LAIV and IIV) would enhance production of heterosubtypic immunity and provide evidence of cross-protection against other influenza viruses. Methods In an open-label study, 26 adult volunteers were randomized to receive one of four vaccine regimens containing two doses of 2009-10 seasonal influenza vaccines administered 8 (±1) weeks apart: 2 doses of LAIV; 2 doses of IIV; LAIV then IIV; IIV then LAIV. Humoral immunity assays for avian H5N1, 2009 pandemic H1N1 (pH1N1), and seasonal vaccine strains were performed on blood collected pre-vaccine and 2 and 4 weeks later. The percentage of cytokine-producing T-cells was compared with baseline 14 days after each dose. Results Subjects receiving IIV had prompt serological responses to vaccine strains. Two subjects receiving heterologous prime boost regimens had enhanced haemagglutination inhibition (HI) and neutralization (NT) titres against pH1N1, and one subject against avian H5N1; all three had pre-existing cross-reactive antibodies detected at baseline. Significantly elevated titres to H5N1 and pH1N1 by neuraminidase inhibition (NI) assay were observed following LAIV-IIV administration. Both vaccines elicited cross-reactive CD4+ T-cell responses to nucleoprotein of avian H5N1 and pH1N1. All regimens were safe and well tolerated. Conclusion Neither homologous nor heterologous prime boost immunization enhanced serum HI and NT titres to 2009 pH1N1 or avian H5N1 compared to single dose vaccine. However heterologous prime-boost vaccination did lead to in vitro evidence of cross-reactivity by NI; the significance of this finding is unclear. These data support the strategy of administering single dose trivalent seasonal influenza vaccine at the outset of an influenza pandemic while a specific vaccine is being developed. Trial Registration ClinicalTrials.gov NCT01044095
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Birds
- Cross Reactions
- Feasibility Studies
- Female
- Health
- Humans
- Immunization, Secondary/adverse effects
- Immunization, Secondary/methods
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/physiology
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Male
- Middle Aged
- Orthomyxoviridae/immunology
- Orthomyxoviridae/physiology
- Pandemics/prevention & control
- Pilot Projects
- Safety
- Seasons
- T-Lymphocytes/immunology
- T-Lymphocytes/virology
- Vaccination/adverse effects
- Vaccination/methods
- Viral Vaccines/adverse effects
- Viral Vaccines/immunology
- Young Adult
Collapse
Affiliation(s)
- Delia Bethell
- Department of Immunology and Medicine, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Verheust C, Goossens M, Pauwels K, Breyer D. Biosafety aspects of modified vaccinia virus Ankara (MVA)-based vectors used for gene therapy or vaccination. Vaccine 2012; 30:2623-32. [DOI: 10.1016/j.vaccine.2012.02.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/01/2012] [Accepted: 02/05/2012] [Indexed: 11/16/2022]
|
23
|
Falivene J, Del Médico Zajac MP, Pascutti MF, Rodríguez AM, Maeto C, Perdiguero B, Gómez CE, Esteban M, Calamante G, Gherardi MM. Improving the MVA vaccine potential by deleting the viral gene coding for the IL-18 binding protein. PLoS One 2012; 7:e32220. [PMID: 22384183 PMCID: PMC3285208 DOI: 10.1371/journal.pone.0032220] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 01/25/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Modified Vaccinia Ankara (MVA) is an attenuated strain of Vaccinia virus (VACV) currently employed in many clinical trials against HIV/AIDS and other diseases. MVA still retains genes involved in host immune response evasion, enabling its optimization by removing some of them. The aim of this study was to evaluate cellular immune responses (CIR) induced by an IL-18 binding protein gene (C12L) deleted vector (MVAΔC12L). METHODOLOGY/PRINCIPAL FINDINGS BALB/c and C57BL/6 mice were immunized with different doses of MVAΔC12L or MVA wild type (MVAwt), then CIR to VACV epitopes in immunogenic proteins were evaluated in spleen and draining lymph nodes at acute and memory phases (7 and 40 days post-immunization respectively). Compared with parental MVAwt, MVAΔC12L immunization induced a significant increase of two to three-fold in CD8(+) and CD4(+) T-cell responses to different VACV epitopes, with increased percentage of anti-VACV cytotoxic CD8(+) T-cells (CD107a/b(+)) during the acute phase of the response. Importantly, the immunogenicity enhancement was also observed after MVAΔC12L inoculation with different viral doses and by distinct routes (systemic and mucosal). Potentiation of MVA's CIR was also observed during the memory phase, in correlation with a higher protection against an intranasal challenge with VACV WR. Of note, we could also show a significant increase in the CIR against HIV antigens such as Env, Gag, Pol and Nef from different subtypes expressed from two recombinants of MVAΔC12L during heterologous DNA prime/MVA boost vaccination regimens. CONCLUSIONS/SIGNIFICANCE This study demonstrates the relevance of IL-18 bp contribution in the immune response evasion during MVA infection. Our findings clearly show that the deletion of the viral IL-18 bp gene is an effective approach to increase MVA vaccine efficacy, as immunogenicity improvements were observed against vector antigens and more importantly to HIV antigens.
Collapse
Affiliation(s)
- Juliana Falivene
- Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - María Fernanda Pascutti
- Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana María Rodríguez
- Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cynthia Maeto
- Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Beatriz Perdiguero
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología, CSIC, Campus Universidad Autónoma, Madrid, Spain
| | - Carmen E. Gómez
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología, CSIC, Campus Universidad Autónoma, Madrid, Spain
| | - Mariano Esteban
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología, CSIC, Campus Universidad Autónoma, Madrid, Spain
| | - Gabriela Calamante
- Instituto de Biotecnología, CICVyA-INTA Castelar, Buenos Aires, Argentina
| | - María Magdalena Gherardi
- Centro Nacional de Referencia para el SIDA, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
24
|
Employing Live Microbes for Vaccine Delivery. DEVELOPMENT OF NOVEL VACCINES 2012. [PMCID: PMC7123214 DOI: 10.1007/978-3-7091-0709-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
25
|
Ferrer MF, Del Médico Zajac MP, Zanetti FA, Valera AR, Zabal O, Calamante G. Recombinant MVA expressing secreted glycoprotein D of BoHV-1 induces systemic and mucosal immunity in animal models. Viral Immunol 2011; 24:331-9. [PMID: 21830904 DOI: 10.1089/vim.2011.0018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Bovine herpesvirus-1 (BoHV-1) infection is distributed worldwide and the development of new tools to fight against this pathogen has become extremely important. In this work a recombinant modified vaccinia virus Ankara (MVA) vector expressing the secreted version of glycoprotein D, MVA-gDs, was obtained and evaluated as a candidate vaccine. First, the correct expression, antigenicity, and N-glycosylation of glycoprotein D were confirmed by molecular techniques. Then MVA-gDs was used as parenteral immunogen in BALB/C mice in which a specific anti-gD humoral immune response was induced and maintained for 7 mo. Two doses of MVA-gDs supplemented with cholera toxin delivered by intranasal immunization induced IgA anti-gD humoral immune responses in nasal and bronchopulmonary washes, as well as IgG anti-gD antibodies in serum samples. In order to evaluate the protection conferred by MVA-gDs immunization, a rabbit BoHV-1 challenge assay was performed. A shorter viral excretion period and a reduction in the number of animals shedding BoHV-1 was observed in the group immunized with recombinant MVA-gDs. In conclusion our data encourage further studies to evaluate MVA-gDs, alone or combined with other immunogens, as a candidate vaccine for BoHV-1.
Collapse
Affiliation(s)
- María Florencia Ferrer
- Consejo Nacional de Investigaciones Científicas y Técnicas, Castilla de Correo 25, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
26
|
Kibler KV, Gomez CE, Perdiguero B, Wong S, Huynh T, Holechek S, Arndt W, Jimenez V, Gonzalez-Sanz R, Denzler K, Haddad EK, Wagner R, Sékaly RP, Tartaglia J, Pantaleo G, Jacobs BL, Esteban M. Improved NYVAC-based vaccine vectors. PLoS One 2011; 6:e25674. [PMID: 22096477 PMCID: PMC3212513 DOI: 10.1371/journal.pone.0025674] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 09/07/2011] [Indexed: 01/13/2023] Open
Abstract
While as yet there is no vaccine against HIV/AIDS, the results of the phase III Thai trial (RV144) have been encouraging and suggest that further improvements of the prime/boost vaccine combination of a poxvirus and protein are needed. With this aim, in this investigation we have generated derivatives of the candidate vaccinia virus vaccine vector NYVAC with potentially improved functions. This has been achieved by the re-incorporation into the virus genome of two host range genes, K1L and C7L, in conjunction with the removal of the immunomodulatory viral molecule B19, an antagonist of type I interferon action. These novel virus vectors, referred to as NYVAC-C-KC and NYVAC-C-KC-ΔB19R, have acquired relevant biological characteristics, giving higher levels of antigen expression in infected cells, replication-competency in human keratinocytes and dermal fibroblasts, activation of selective host cell signal transduction pathways, and limited virus spread in tissues. Importantly, these replication-competent viruses have been demonstrated to maintain a highly attenuated phenotype.
Collapse
Affiliation(s)
- Karen V. Kibler
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
| | - Carmen E. Gomez
- Centro Nacional de Biotecnologia-Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Beatriz Perdiguero
- Centro Nacional de Biotecnologia-Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Shukmei Wong
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
| | - Trung Huynh
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
| | - Susan Holechek
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
| | - William Arndt
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
| | - Victoria Jimenez
- Centro Nacional de Biotecnologia-Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Ruben Gonzalez-Sanz
- Centro Nacional de Biotecnologia-Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Karen Denzler
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
| | - Elias K. Haddad
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
| | - Ralf Wagner
- University of Regensburg, Regensburg, Germany
| | - Rafick P. Sékaly
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
- University of Montreal, Montreal, Canada
| | - James Tartaglia
- Sanofi-Pasteur, Swiftwater, Pennsylvania, United States of America
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Bertram L. Jacobs
- The Biodesign Institute at Arizona State University, Tempe, Arizona, United States of America
- * E-mail: (BLJ); (ME)
| | - Mariano Esteban
- Centro Nacional de Biotecnologia-Consejo Superior de Investigaciones Cientificas, Madrid, Spain
- * E-mail: (BLJ); (ME)
| |
Collapse
|
27
|
Hwa SH, Iams KP, Hall JS, Kingstad BA, Osorio JE. Characterization of recombinant raccoonpox vaccine vectors in chickens. Avian Dis 2011; 54:1157-65. [PMID: 21313834 DOI: 10.1637/9315-032410-reg.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Raccoonpox virus (RCN) has been used as a recombinant vector against several mammalian pathogens but has not been tested in birds. The replication of RCN in chick embryo fibroblasts (CEFs) and chickens was studied with the use of highly pathogenic avian influenza virus H5N1 hemagglutinin (HA) as a model antigen and luciferase (luc) as a reporter gene. Although RCN replicated to low levels in CEFs, it efficiently expressed recombinant proteins and, in vivo, elicited anti-HA immunoglobulin yolk (IgY) antibody responses comparable to inactivated influenza virus. Biophotonic in vivo imaging of 1-wk-old chicks with RCN-luc showed strong expression of the luc reporter gene lasting up to 3 days postinfection. These studies demonstrate the potential of RCN as a vaccine vector for avian influenza and other poultry pathogens.
Collapse
Affiliation(s)
- Shi-Hsia Hwa
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
28
|
Signore A, Mather SJ, Piaggio G, Malviya G, Dierckx RA. Molecular imaging of inflammation/infection: nuclear medicine and optical imaging agents and methods. Chem Rev 2010; 110:3112-45. [PMID: 20415479 DOI: 10.1021/cr900351r] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- A Signore
- Nuclear Medicine Unit, II Faculty of Medicine and Surgery, Sapienza University of Rome, Rome, Italy.
| | | | | | | | | |
Collapse
|
29
|
Guerra S, González JM, Climent N, Reyburn H, López-Fernández LA, Nájera JL, Gómez CE, García F, Gatell JM, Gallart T, Esteban M. Selective induction of host genes by MVA-B, a candidate vaccine against HIV/AIDS. J Virol 2010; 84:8141-52. [PMID: 20534857 PMCID: PMC2916545 DOI: 10.1128/jvi.00749-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 05/28/2010] [Indexed: 01/08/2023] Open
Abstract
The aim of this study was to define the effects on antigen-presenting cells of the expression of HIV antigens from an attenuated poxvirus vector. We have analyzed the transcriptional changes in gene expression following infection of human immature monocyte-derived dendritic cells (DC) with recombinant modified vaccinia virus Ankara (MVA) expressing the genes encoding the gp120 and Gag-Pol-Nef antigens of HIV type 1 clade B (referred to as MVA-B) versus parental MVA infection. Using microarray technology and real-time reverse transcription-PCR, we demonstrated that the HIV proteins induced the expression of cytokines, cytokine receptors, chemokines, chemokine receptors, and molecules involved in antigen uptake and processing, including major histocompatibility complex (MHC) genes. Levels of mRNAs for interleukin-1, beta interferon, CCR8, and SCYA20 were higher after HIV antigen production. MVA-B infection also modulated the expression of antigen processing and presentation genes: the gene for MICA was upregulated, whereas those for HLA-DRA and HSPA5 were downregulated. Indeed, the increased expression of the gene for MICA, a glycoprotein related to major histocompatibility complex class I molecules, was shown to enhance the interaction between MVA-B-infected target cells and cytotoxic lymphocytes. The expression profiles of the genes for protein kinases such as JAK1 and IRAK2 were activated after HIV antigen expression. Several genes included in the JAK-STAT and mitogen-activated protein kinase signaling pathways were regulated after HIV antigen expression. Our findings provide the first gene signatures in DC of a candidate MVA-B vaccine expressing four HIV antigens and identified the biological roles of some of the regulatory genes, like that for MICA, which will help in the design of more effective MVA-derived vaccines.
Collapse
Affiliation(s)
- Susana Guerra
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - José Manuel González
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Núria Climent
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Hugh Reyburn
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Luis A. López-Fernández
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - José L. Nájera
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Carmen E. Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Felipe García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - José M. Gatell
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Teresa Gallart
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, E-28049 Madrid, Spain, Department of Preventive Medicine and Public Health, Universidad Autónoma, E-28029 Madrid, Spain, Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain, Servicio de Enfermedades Infecciosas, Servicio de Inmunología, Hospital Clínic de Barcelona, AIDS Research Group, Instituto de Investigaciones Biomedicas August Pi i Sunyer (IDIBAPS), HIVACAT Program, Universidad de Barcelona, Villaroel 170, 08036 Barcelona, Spain
| |
Collapse
|
30
|
Nájera JL, Gómez CE, García-Arriaza J, Sorzano CO, Esteban M. Insertion of vaccinia virus C7L host range gene into NYVAC-B genome potentiates immune responses against HIV-1 antigens. PLoS One 2010; 5:e11406. [PMID: 20613977 PMCID: PMC2894869 DOI: 10.1371/journal.pone.0011406] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 06/08/2010] [Indexed: 11/18/2022] Open
Abstract
Background The highly attenuated vaccinia virus strain NYVAC expressing HIV-1 components has been evaluated as a vaccine candidate in preclinical and clinical trials with encouraging results. We have previously described that the presence of C7L in the NYVAC genome prevents the induction of apoptosis and renders the vector capable of replication in human and murine cell lines while maintaining an attenuated phenotype in mice. Methodology/Principal Findings In an effort to improve the immunogenicity of NYVAC, we have developed a novel poxvirus vector by inserting the VACV host-range C7L gene into the genome of NYVAC-B, a recombinant virus that expresses four HIV-1 antigens from clade B (Env, Gag, Pol and Nef) (referred as NYVAC-B-C7L). In the present study, we have compared the in vitro and in vivo behavior of NYVAC-B and NYVAC-B-C7L. In cultured cells, NYVAC-B-C7L expresses higher levels of heterologous antigen than NYVAC-B as determined by Western blot and fluorescent-activated cell sorting to score Gag expressing cells. In a DNA prime/poxvirus boost approach with BALB/c mice, both recombinants elicited robust, broad and multifunctional antigen-specific T-cell responses to the HIV-1 immunogens expressed from the vectors. However, the use of NYVAC-B-C7L as booster significantly enhanced the magnitude of the T cell responses, and induced a more balanced cellular immune response to the HIV-1 antigens in comparison to that elicited in animals boosted with NYVAC-B. Conclusions/Significance These findings demonstrate the possibility to enhance the immunogenicity of the highly attenuated NYVAC vector by the insertion of the host-range gene C7L and suggest the use of this modified vector as an improved vaccine candidate against HIV/AIDS.
Collapse
Affiliation(s)
- José Luis Nájera
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Carlos Oscar Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
- * E-mail:
| |
Collapse
|
31
|
Abadie V, Bonduelle O, Duffy D, Parizot C, Verrier B, Combadière B. Original encounter with antigen determines antigen-presenting cell imprinting of the quality of the immune response in mice. PLoS One 2009; 4:e8159. [PMID: 19997562 PMCID: PMC2785484 DOI: 10.1371/journal.pone.0008159] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2009] [Accepted: 11/02/2009] [Indexed: 01/29/2023] Open
Abstract
Background Obtaining a certain multi-functionality of cellular immunity for the control of infectious diseases is a burning question in immunology and in vaccine design. Early events, including antigen shuttling to secondary lymphoid organs and recruitment of innate immune cells for adaptive immune response, determine host responsiveness to antigens. However, the sequence of these events and their impact on the quality of the immune response remain to be elucidated. Here, we chose to study Modified Vaccinia virus Ankara (MVA) which is now replacing live Smallpox vaccines and is proposed as an attenuated vector for vaccination strategies against infectious diseases. Methodology/Principal findings We analyzed in vivo mechanisms triggered following intradermal (i.d.) and intramuscular (i.m.) Modified Vaccinia virus Ankara (MVA) administration. We demonstrated significant differences in the antigen shuttling to lymphoid organs by macrophages (MΦs), myeloid dendritic cells (DCs), and neutrophils (PMNs). MVA i.d. administration resulted in better antigen distribution and more sustained antigen-presenting cells (APCs) recruitment into draining lymph nodes than with i.m. administration. These APCs, which comprise both DCs and MΦs, were differentially involved in T cell priming and shaped remarkably the quality of cytokine-producing virus-specific T cells according to the entry route of MVA. Conclusions/Significance This study improves our understanding of the mechanisms of antigen delivery and their consequences on the quality of immune responses and provides new insights for vaccine development.
Collapse
Affiliation(s)
- Valérie Abadie
- Institut National de la Santé et de la Recherche Médicale (INSERM) U945, Paris, France
- University of Pierre and Marie Curie (UPMC)- Univ Paris 06, Paris, France
- Assistance-Publique/Hopitaux-de-Paris, Immunity and Infection, Paris, France
| | - Olivia Bonduelle
- Institut National de la Santé et de la Recherche Médicale (INSERM) U945, Paris, France
- University of Pierre and Marie Curie (UPMC)- Univ Paris 06, Paris, France
- Assistance-Publique/Hopitaux-de-Paris, Immunity and Infection, Paris, France
| | - Darragh Duffy
- Institut National de la Santé et de la Recherche Médicale (INSERM) U945, Paris, France
- University of Pierre and Marie Curie (UPMC)- Univ Paris 06, Paris, France
- Assistance-Publique/Hopitaux-de-Paris, Immunity and Infection, Paris, France
| | - Christophe Parizot
- Institut National de la Santé et de la Recherche Médicale (INSERM) U945, Paris, France
- University of Pierre and Marie Curie (UPMC)- Univ Paris 06, Paris, France
- Assistance-Publique/Hopitaux-de-Paris, Immunity and Infection, Paris, France
| | - Bernard Verrier
- Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS/UCBL, Lyon, France
| | - Béhazine Combadière
- Institut National de la Santé et de la Recherche Médicale (INSERM) U945, Paris, France
- University of Pierre and Marie Curie (UPMC)- Univ Paris 06, Paris, France
- Assistance-Publique/Hopitaux-de-Paris, Immunity and Infection, Paris, France
- * E-mail:
| |
Collapse
|
32
|
Abstract
Traditional vaccine technologies have resulted in an impressive array of efficacious vaccines against a variety of infectious agents. However, several potentially deadly pathogens, including retroviruses and parasites, have proven less amenable to the application of traditional vaccine platforms, indicating the need for new approaches. Viral vectors represent an attractive way to deliver and present vaccine antigens that may offer advantages over traditional platforms. Due to their ability to induce strong cell-mediated immunity (CMI) in addition to antibodies, viral vectors may be suitable for infectious agents, such as malaria parasites, where potent CMI is required for protection. Poxvirus-vectored malaria vaccines have been the most extensively studied in the clinic, achieving significant reductions in liver-stage parasite burden. More recently, adenovirus-vectored malaria vaccines have entered clinical testing. The most promising approach - heterologous prime-boost regimens, in which different viral vectors are sequentially paired with each other or with DNA or recombinant protein vaccines - is now being explored, and could provide high-grade protection, if findings in animal models are translatable to humans. Significant barriers remain, however, such as pre-existing immunity to the vector particle and an unexplained safety signal observed in one trial suggesting an increased risk of HIV acquisition in volunteers with pre-existing immunity to the vector.
Collapse
Affiliation(s)
- K J Limbach
- U.S. Military Malaria Vaccine Program, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 20910-7500, USA.
| | | |
Collapse
|
33
|
Application of bioluminescence imaging to the prediction of lethality in vaccinia virus-infected mice. J Virol 2009; 83:10437-47. [PMID: 19656894 DOI: 10.1128/jvi.01296-09] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To find an alternative endpoint for the efficacy of antismallpox treatments, bioluminescence was measured in live BALB/c mice following lethal challenge with a recombinant WR vaccinia virus expressing luciferase. Intravenous vaccinia immunoglobulin treatments were used to confer protection on a proportion of animals. Using known lethality outcomes in 200 animals and total fluxes recorded daily in live animals, we performed univariate receiver operating characteristic (ROC) curve analysis to assess whether lethality can be predicted based on bioluminescence. Total fluxes in the spleens on day 3 and in the livers on day 5 generated accurate predictive models; the area under the ROC curve (AUC) was 0.91. Multiple logistic regression analysis utilizing a linear combination of six measurements: total flux in the liver on days 2, 3, and 5; in the spleen on days 1 and 3; and in the nasal cavity on day 4 generated the most accurate predictions (AUC = 0.96). This model predicted lethality in 90% of animals with only 10% of nonsurviving animals incorrectly predicted to survive. Compared with bioluminescence, ROC analysis with 25% and 30% weight loss as thresholds accurately predicted survival on day 5, but lethality predictions were low until day 9. Collectively, our data support the use of bioimaging for lethality prediction following vaccinia virus challenge and for gaining insight into protective mechanisms conferred by vaccines and therapeutics.
Collapse
|
34
|
Delaloye J, Roger T, Steiner-Tardivel QG, Le Roy D, Knaup Reymond M, Akira S, Petrilli V, Gomez CE, Perdiguero B, Tschopp J, Pantaleo G, Esteban M, Calandra T. Innate immune sensing of modified vaccinia virus Ankara (MVA) is mediated by TLR2-TLR6, MDA-5 and the NALP3 inflammasome. PLoS Pathog 2009; 5:e1000480. [PMID: 19543380 PMCID: PMC2691956 DOI: 10.1371/journal.ppat.1000480] [Citation(s) in RCA: 258] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 05/21/2009] [Indexed: 12/21/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is an attenuated double-stranded DNA poxvirus currently developed as a vaccine vector against HIV/AIDS. Profiling of the innate immune responses induced by MVA is essential for the design of vaccine vectors and for anticipating potential adverse interactions between naturally acquired and vaccine-induced immune responses. Here we report on innate immune sensing of MVA and cytokine responses in human THP-1 cells, primary human macrophages and mouse bone marrow-derived macrophages (BMDMs). The innate immune responses elicited by MVA in human macrophages were characterized by a robust chemokine production and a fairly weak pro-inflammatory cytokine response. Analyses of the cytokine production profile of macrophages isolated from knockout mice deficient in Toll-like receptors (TLRs) or in the adapter molecules MyD88 and TRIF revealed a critical role for TLR2, TLR6 and MyD88 in the production of IFNbeta-independent chemokines. MVA induced a marked up-regulation of the expression of RIG-I like receptors (RLR) and the IPS-1 adapter (also known as Cardif, MAVS or VISA). Reduced expression of RIG-I, MDA-5 and IPS-1 by shRNAs indicated that sensing of MVA by RLR and production of IFNbeta and IFNbeta-dependent chemokines was controlled by the MDA-5 and IPS-1 pathway in the macrophage. Crosstalk between TLR2-MyD88 and the NALP3 inflammasome was essential for expression and processing of IL-1beta. Transcription of the Il1b gene was markedly impaired in TLR2(-/-) and MyD88(-/-) BMDM, whereas mature and secreted IL-1beta was massively reduced in NALP3(-/-) BMDMs or in human THP-1 macrophages with reduced expression of NALP3, ASC or caspase-1 by shRNAs. Innate immune sensing of MVA and production of chemokines, IFNbeta and IL-1beta by macrophages is mediated by the TLR2-TLR6-MyD88, MDA-5-IPS-1 and NALP3 inflammasome pathways. Delineation of the host response induced by MVA is critical for improving our understanding of poxvirus antiviral escape mechanisms and for designing new MVA vaccine vectors with improved immunogenicity.
Collapse
Affiliation(s)
- Julie Delaloye
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Quynh-Giao Steiner-Tardivel
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Marlies Knaup Reymond
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Virginie Petrilli
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Carmen E. Gomez
- Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Beatriz Perdiguero
- Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Jürg Tschopp
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Giuseppe Pantaleo
- Laboratory of AIDS Immunopathogenesis, Immunology and Allergology Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Mariano Esteban
- Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, Madrid, Spain
| | - Thierry Calandra
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
35
|
Gómez CE, Nájera JL, Sánchez R, Jiménez V, Esteban M. Multimeric soluble CD40 ligand (sCD40L) efficiently enhances HIV specific cellular immune responses during DNA prime and boost with attenuated poxvirus vectors MVA and NYVAC expressing HIV antigens. Vaccine 2009; 27:3165-74. [PMID: 19446187 DOI: 10.1016/j.vaccine.2009.03.049] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 03/06/2009] [Accepted: 03/19/2009] [Indexed: 02/07/2023]
Abstract
The attenuated poxvirus vectors MVA and NYVAC are now in clinical trials against HIV/AIDS. Due to the vectors restricted replication capacity in human cells, approaches to enhance their immunogenicity are highly desirable. Here, we have analyzed the ability of a soluble form of hexameric CD40L (sCD40L) to stimulate specific immune responses to HIV antigens when inoculated in mice during priming with DNA and in the booster with MVA or NYVAC, expressing the vectors HIV-1 Env, Gag, Pol and Nef antigens from clade B. Our findings revealed that sCD40L in DNA/poxvirus combination enhanced the magnitude about 2-fold (DNA-B/MVA-B) and 4-fold (DNA-B/NYVAC-B), as well as the breath of the HIV antigen specific cellular immune responses. sCD40L was necessary in both prime and boost inoculations triggering a potent polarization of the Th response towards a Th1 type. In DNA-B/NYVAC-B regime the addition of sCD40L significantly enhanced the humoral immune response against HIV gp160, but not in DNA-B/MVA-B combination. These findings provided evidence for the immunostimulatory benefit of sCD40L when DNA and the poxvirus vectors MVA and NYVAC are used as immunogens.
Collapse
Affiliation(s)
- Carmen E Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Ciudad Universitaria Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | |
Collapse
|
36
|
Hu N, Yu R, Shikuma C, Shiramizu B, Ostrwoski MA, Yu Q. Role of cell signaling in poxvirus-mediated foreign gene expression in mammalian cells. Vaccine 2009; 27:2994-3006. [PMID: 19428911 DOI: 10.1016/j.vaccine.2009.02.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 02/15/2009] [Accepted: 02/24/2009] [Indexed: 10/21/2022]
Abstract
Poxviruses have been extensively used as a promising vehicle to efficiently deliver a variety of antigens in mammalian hosts to induce immune responses against infectious diseases and cancer. Using recombinant vaccinia virus (VV) and canarypox virus (ALVAC) expressing enhanced green fluorescent protein (EGFP) or multiple HIV-1 gene products, we studied the role of four cellular signaling pathways, the phosphoinositide-3-OH kinase (PI3K), extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38 MAPK), and c-Jun N-terminal kinase (JNK), in poxvirus-mediated foreign gene expression in mammalian cells. In nonpermissive infection (human monocytes), activation of PI3K, ERK, p38 MAPK, and JNK was observed in both VV and ALVAC and blocking PI3K, p38 MAKP, and JNK pathways with their specific inhibitors significantly reduced viral and vaccine antigen gene expression. Whereas, blocking the ERK pathway had no significant effect. Among these cellular signaling pathways studied, PI3K was the most critical pathway involved in gene expression by VV- or ALVAC-infected monocytes. The important role of PI3K in poxvirus-mediated gene expression was further confirmed in mouse epidermal cells stably transfected with dominant-negative PI3K mutant, as poxvirus-mediated targeted gene expression was significantly decreased in these cells when compared with their parental cells. Signaling pathway activation influenced gene expression at the mRNA level rather than virus binding. In permissive mammalian cells, however, VV DNA copies were also significantly decreased in the absence of normal function of the PI3K pathway. Poxvirus-triggered activation of PI3K pathway could be completely abolished by atazanavir, a new generation of antiretroviral protease inhibitors (PIs). As a consequence, ALVAC-mediated EGFP or HIV-1 gag gene expression in infected primary human monocytes was significantly reduced in the presence of atazanavir. These findings implicate that antiretroviral therapy (ART), also known as highly active antiretroviral therapy (HAART), may negatively impact the efficacy of live poxvirus vector-based vaccines and should be carefully considered when administering such live vaccines to individuals on ART.
Collapse
Affiliation(s)
- Ningjie Hu
- Hawaii AIDS Clinical Research Program, University of Hawaii at Manoa, Leahi Hospital, Honolulu, HI 96816, USA
| | | | | | | | | | | |
Collapse
|
37
|
Paran N, Suezer Y, Lustig S, Israely T, Schwantes A, Melamed S, Katz L, Preuss T, Hanschmann KM, Kalinke U, Erez N, Levin R, Velan B, Löwer J, Shafferman A, Sutter G. Postexposure immunization with modified vaccinia virus Ankara or conventional Lister vaccine provides solid protection in a murine model of human smallpox. J Infect Dis 2009; 199:39-48. [PMID: 19012492 DOI: 10.1086/595565] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Decades after the cessation of smallpox vaccination, the potential of the deliberate release of pathogenic orthopoxviruses has forced a reconsideration of using these extremely efficient human vaccines. Scenarios of sudden biothreats have prompted demand for rapidly protective vaccination. However, the feasibility of short-term vaccination (i.e., vaccination shortly before exposure) with vaccinia virus (VACV) is uncertain. METHODS We tested the rapid protective capacity of vaccines based on VACV strain Lister (VACV-Lister) and on modified VACV Ankara (MVA) in different mouse models, comparing lethal infections with VACV strain Western Reserve (VACV-WR) or ectromelia virus (ECTV). RESULTS In contrast to VACV-WR challenge, we found extended incubation periods after ECTV challenge, allowing successful therapeutic immunization with VACV-Lister and MVA when applied 2-3 days after exposure. Rapid protection from respiratory tract ECTV infection was significantly affected by vaccine dose and was associated with occurrence of poxvirus-specific antibodies. Vaccinations in type I interferon receptor-deficient mice were protective, whereas recombination activating gene 1-deficient mice lacking mature T and B cells failed to mount immunity after short-term vaccination, confirming an essential role of adaptive immune responses. CONCLUSIONS ECTV infection in mice models the course of human smallpox. Our data provide evidence to substantiate historical data on the usefulness of postexposure vaccination with conventional VACV and the new candidate MVA to protect against fatal orthopoxvirus infections.
Collapse
Affiliation(s)
- Nir Paran
- Israel Institute for Biological Research, Ness-Ziona, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mooij P, Balla-Jhagjhoorsingh SS, Koopman G, Beenhakker N, van Haaften P, Baak I, Nieuwenhuis IG, Kondova I, Wagner R, Wolf H, Gómez CE, Nájera JL, Jiménez V, Esteban M, Heeney JL. Differential CD4+ versus CD8+ T-cell responses elicited by different poxvirus-based human immunodeficiency virus type 1 vaccine candidates provide comparable efficacies in primates. J Virol 2008; 82:2975-88. [PMID: 18184713 PMCID: PMC2258966 DOI: 10.1128/jvi.02216-07] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Accepted: 12/13/2007] [Indexed: 12/20/2022] Open
Abstract
Poxvirus vectors have proven to be highly effective for boosting immune responses in diverse vaccine settings. Recent reports reveal marked differences in the gene expression of human dendritic cells infected with two leading poxvirus-based human immunodeficiency virus (HIV) vaccine candidates, New York vaccinia virus (NYVAC) and modified vaccinia virus Ankara (MVA). To understand how complex genomic changes in these two vaccine vectors translate into antigen-specific systemic immune responses, we undertook a head-to-head vaccine immunogenicity and efficacy study in the pathogenic HIV type 1 (HIV-1) model of AIDS in Indian rhesus macaques. Differences in the immune responses in outbred animals were not distinguished by enzyme-linked immunospot assays, but differences were distinguished by multiparameter fluorescence-activated cell sorter analysis, revealing a difference between the number of animals with both CD4(+) and CD8(+) T-cell responses to vaccine inserts (MVA) and those that elicit a dominant CD4(+) T-cell response (NYVAC). Remarkably, vector-induced differences in CD4(+)/CD8(+) T-cell immune responses persisted for more than a year after challenge and even accompanied antigenic modulation throughout the control of chronic infection. Importantly, strong preexposure HIV-1/simian immunodeficiency virus-specific CD4(+) T-cell responses did not prove deleterious with respect to accelerated disease progression. In contrast, in this setting, animals with strong vaccine-induced polyfunctional CD4(+) T-cell responses showed efficacies similar to those with stronger CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), P.O. Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|