1
|
Mazloum A, Karagyaur M, Chernyshev R, van Schalkwyk A, Jun M, Qiang F, Sprygin A. Post-genomic era in agriculture and veterinary science: successful and proposed application of genetic targeting technologies. Front Vet Sci 2023; 10:1180621. [PMID: 37601766 PMCID: PMC10434572 DOI: 10.3389/fvets.2023.1180621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Gene editing tools have become an indispensable part of research into the fundamental aspects of cell biology. With a vast body of literature having been generated based on next generation sequencing technologies, keeping track of this ever-growing body of information remains challenging. This necessitates the translation of genomic data into tangible applications. In order to address this objective, the generated Next Generation Sequencing (NGS) data forms the basis for targeted genome editing strategies, employing known enzymes of various cellular machinery, in generating organisms with specifically selected phenotypes. This review focuses primarily on CRISPR/Cas9 technology in the context of its advantages over Zinc finger proteins (ZNF) and Transcription activator-like effector nucleases (TALEN) and meganucleases mutagenesis strategies, for use in agricultural and veterinary applications. This review will describe the application of CRISPR/Cas9 in creating modified organisms with custom-made properties, without the undesired non-targeted effects associated with virus vector vaccines and bioactive molecules produced in bacterial systems. Examples of the successful and unsuccessful applications of this technology to plants, animals and microorganisms are provided, as well as an in-depth look into possible future trends and applications in vaccine development, disease resistance and enhanced phenotypic traits will be discussed.
Collapse
Affiliation(s)
- Ali Mazloum
- Federal Center for Animal Health, Vladimir, Russia
| | - Maxim Karagyaur
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | | | - Antoinette van Schalkwyk
- Agricultural Research Council-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
- Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Ma Jun
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Fu Qiang
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | | |
Collapse
|
2
|
Orlova OV, Glazkova DV, Bogoslovskaya EV, Shipulin GA, Yudin SM. Development of Modified Vaccinia Virus Ankara-Based Vaccines: Advantages and Applications. Vaccines (Basel) 2022; 10:vaccines10091516. [PMID: 36146594 PMCID: PMC9503770 DOI: 10.3390/vaccines10091516] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is a promising viral vector for vaccine development. MVA is well studied and has been widely used for vaccination against smallpox in Germany. This review describes the history of the origin of the virus and its properties as a vaccine, including a high safety profile. In recent years, MVA has found its place as a vector for the creation of vaccines against various diseases. To date, a large number of vaccine candidates based on the MVA vector have already been developed, many of which have been tested in preclinical and clinical studies. We discuss data on the immunogenicity and efficacy of some of these vaccines.
Collapse
|
3
|
Chaudhary O, Wang L, Bose D, Narayan V, Yeh MT, Carville A, Clements JD, Andino R, Kozlowski PA, Aldovini A. Comparative Evaluation of Prophylactic SIV Vaccination Modalities Administered to the Oral Cavity. AIDS Res Hum Retroviruses 2020; 36:984-997. [PMID: 32962398 PMCID: PMC7703093 DOI: 10.1089/aid.2020.0157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Attempts to develop a protective human immunodeficiency virus (HIV) vaccine have had limited success, especially in terms of inducing protective antibodies capable of neutralizing different viral strains. As HIV transmission occurs mainly via mucosal surfaces, HIV replicates significantly in the gastrointestinal tract, and the oral route of vaccination is a very convenient one to implement worldwide, we explored three SIV vaccine modalities administered orally and composed of simian immunodeficiency virus (SIV) DNA priming with different boosting immunogens, with the goal of evaluating whether they could provide lasting humoral and cellular responses, including at mucosal surfaces that are sites of HIV entry. Twenty-four Cynomolgus macaques (CyM) were primed with replication-incompetent SIV DNA provirus and divided into three groups for the following booster vaccinations, all administered in the oral cavity: Group 1 with recombinant SIV gp140 and Escherichia coli heat-labile toxin adjuvant dmLT, Group 2 with recombinant SIV-Oral Poliovirus (SIV-OPV), and Group 3 with recombinant SIV-modified vaccinia ankara (SIV-MVA). Cell-mediated responses were measured using blood, lymph node, rectal and vaginal mononuclear cells. Significant levels of systemic and mucosal T-cell responses against Gag and Env were observed in all groups. Some SIV-specific plasma IgG, rectal and salivary IgA antibodies were generated, mainly in animals that received SIV DNA + SIV-MVA, but no vaginal IgA was detected. Susceptibility to infection after SIVmac251 challenge was similar in vaccinated and nonvaccinated animals, but acute infection viremia levels were lower in the group that received SIV DNA + SIV-MVA. Nonvaccinated CyM maintained central memory and total CD4+ T-cell levels in the normal range during the 5 months of postinfection follow-up as did the vaccinated animals, precluding evaluation of vaccine impact on disease progression. We conclude that the oral cavity vaccination tested in these regimens can stimulate cell-mediated immunity systemically and mucosally, but humoral response stimulation was limited with the doses and the vaccine platforms used.
Collapse
Affiliation(s)
- Omkar Chaudhary
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Lingyun Wang
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepanwita Bose
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivek Narayan
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ming Te Yeh
- Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | | | - John D. Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Raul Andino
- Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Anna Aldovini
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Leite Pereira A, Jouhault Q, Marcos Lopez E, Cosma A, Lambotte O, Le Grand R, Lehmann MH, Tchitchek N. Modulation of Cell Surface Receptor Expression by Modified Vaccinia Virus Ankara in Leukocytes of Healthy and HIV-Infected Individuals. Front Immunol 2020; 11:2096. [PMID: 33013882 PMCID: PMC7506042 DOI: 10.3389/fimmu.2020.02096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/03/2020] [Indexed: 11/19/2022] Open
Abstract
Viral vectors are increasingly used as delivery means to induce a specific immunity in humans and animals. However, they also impact the immune system, and it depends on the given context whether this is beneficial or not. The attenuated vaccinia virus strain modified vaccinia virus Ankara (MVA) has been used as a viral vector in clinical studies intended to treat and prevent cancer and infectious diseases. The adjuvant property of MVA is thought to be due to its capability to stimulate innate immunity. Here, we confirmed that MVA induces interleukin-8 (IL-8), and this chemokine was upregulated significantly more in monocytes and HLA-DRbright dendritic cells (DCs) of HIV-infected patients on combined antiretroviral therapy (ART) than in cells of healthy persons. The effect of MVA on cell surface receptors is mostly unknown. Using mass cytometry profiling, we investigated the expression of 17 cell surface receptors in leukocytes after ex vivo infection of human whole-blood samples with MVA. We found that MVA downregulates most of the characteristic cell surface markers in particular types of leukocytes. In contrast, C-X-C motif chemokine receptor 4 (CXCR4) was significantly upregulated in each leukocyte type of healthy persons. Additionally, we detected a relative higher cell surface expression of the HIV-1 co-receptors C-C motif chemokine receptor 5 (CCR5) and CXCR4 in leukocytes of HIV-ART patients than in healthy persons. Importantly, we showed that MVA infection significantly downregulated CCR5 in CD4+ T cells, CD8+ T cells, B cells, and three different DC populations. CD86, a costimulatory molecule for T cells, was significantly upregulated in HLA-DRbright DCs after MVA infection of whole blood from HIV-ART patients. However, MVA was unable to downregulate cell surface expression of CD11b and CD32 in monocytes and neutrophils of HIV-ART patients to the same extent as in monocytes and neutrophils of healthy persons. In summary, MVA modulates the expression of many different kinds of cell surface receptors in leukocytes, which can vary in cells originating from persons previously infected with other pathogens.
Collapse
Affiliation(s)
- Adrien Leite Pereira
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Quentin Jouhault
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Ernesto Marcos Lopez
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Antonio Cosma
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Olivier Lambotte
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France.,INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France.,APHP, Service de Médecine Interne et Immunologie Clinique, Hôpitaux Universitaires Paris Saclay, Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Michael H Lehmann
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nicolas Tchitchek
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure, CEA-Université Paris Sud 11, Fontenay-aux-Roses, France
| |
Collapse
|
5
|
Salvato MS, Domi A, Guzmán-Cardozo C, Medina-Moreno S, Zapata JC, Hsu H, McCurley N, Basu R, Hauser M, Hellerstein M, Guirakhoo F. A Single Dose of Modified Vaccinia Ankara Expressing Lassa Virus-like Particles Protects Mice from Lethal Intra-cerebral Virus Challenge. Pathogens 2019; 8:E133. [PMID: 31466243 PMCID: PMC6789566 DOI: 10.3390/pathogens8030133] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022] Open
Abstract
Lassa fever surpasses Ebola, Marburg, and all other hemorrhagic fevers except Dengue in its public health impact. Caused by Lassa virus (LASV), the disease is a scourge on populations in endemic areas of West Africa, where reported incidence is higher. Here, we report construction, characterization, and preclinical efficacy of a novel recombinant vaccine candidate GEO-LM01. Constructed in the Modified Vaccinia Ankara (MVA) vector, GEO-LM01 expresses the glycoprotein precursor (GPC) and zinc-binding matrix protein (Z) from the prototype Josiah strain lineage IV. When expressed together, GP and Z form Virus-Like Particles (VLPs) in cell culture. Immunogenicity and efficacy of GEO-LM01 was tested in a mouse challenge model. A single intramuscular dose of GEO-LM01 protected 100% of CBA/J mice challenged with a lethal dose of ML29, a Mopeia/Lassa reassortant virus, delivered directly into the brain. In contrast, all control animals died within one week. The vaccine induced low levels of antibodies but Lassa-specific CD4+ and CD8+ T cell responses. This is the first report showing that a single dose of a replication-deficient MVA vector can confer full protection against a lethal challenge with ML29 virus.
Collapse
Affiliation(s)
- Maria S Salvato
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | | - Juan Carlos Zapata
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Haoting Hsu
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Nathanael McCurley
- Office of Technology Licensing and Commercialization, Georgia State University, Atlanta, GA 30303, USA
| | - Rahul Basu
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | | | | | |
Collapse
|
6
|
Lymph node conduits transport virions for rapid T cell activation. Nat Immunol 2019; 20:602-612. [PMID: 30886418 PMCID: PMC6474694 DOI: 10.1038/s41590-019-0342-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 02/02/2019] [Indexed: 11/29/2022]
Abstract
Despite intense interest in antiviral T cell priming, the routes of virion movement in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined virion and infected-cell locations after vaccinia and Zika virus administration. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Using confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus-infected cells deeper in the LN, leading to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T cells. Virions can access lymph node conduits.
Collapse
|
7
|
Abstract
Chikungunya is a clinically and economically important arbovirus that has spread globally in the twenty-first century. While uncommonly fatal, infection with the virus can lead to incapacitating arthralgia that can persist for months to years. The adverse impacts of viral spread are most severe in developing low- and middle-income countries in which medical infrastructure is insufficient and manual labor is an economic driver. Unfortunately, no prophylactic or therapeutic treatments are approved for human use to combat the virus. Historically, vaccination has proven to be the most efficient and successful strategy for protecting populations and eradicating infectious disease. A large and diverse range of promising vaccination approaches for use against Chikungunya has emerged in recent years and been shown to safely elicit protective immune responses in animal models and humans. Importantly, many of these are based on technologies that have been clinically approved for use against other pathogens. Furthermore, clinical trials are currently ongoing for a subset of these. The purpose of this review is to provide a description of the relevant immunobiology of Chikungunya infection, to present immune-stimulating technologies that have been successfully employed to protect against infection, and discuss priorities and challenges regarding the future development of a vaccine for clinical use.
Collapse
|
8
|
O'Leary MP, Choi AH, Kim SI, Chaurasiya S, Lu J, Park AK, Woo Y, Warner SG, Fong Y, Chen NG. Novel oncolytic chimeric orthopoxvirus causes regression of pancreatic cancer xenografts and exhibits abscopal effect at a single low dose. J Transl Med 2018; 16:110. [PMID: 29699566 PMCID: PMC5918769 DOI: 10.1186/s12967-018-1483-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) has been increasing by 0.5% per year in the United States. PDAC portends a dismal prognosis and novel therapies are needed. This study describes the generation and characterization of a novel oncolytic chimeric orthopoxvirus for the treatment of pancreatic cancer. Methods After chimerization and high-throughput screening, CF33 was chosen from 100 new chimeric orthopoxvirus isolates for its ability to kill pancreatic cancer cells. In vitro cytotoxicity was assayed in six pancreatic cancer cell lines. In vivo efficacy and toxicity were evaluated in PANC-1 and MIA PaCa-2 xenograft models. Results CF33 caused rapid killing of six pancreatic cancer cells lines in vitro, releasing damage-associated molecular patterns, and regression of PANC-1 injected and non-injected distant xenografts in vivo after a single low intratumoral dose of 103 plaque-forming units. Using luciferase imaging, CF33 was noted to preferentially replicate in tumors which corresponds to the low viral titers found in solid organs. Conclusion The low dose of CF33 required to treat pancreatic cancer in this preclinical study may ease the manufacturing and dosing challenges currently facing oncolytic viral therapy.
Collapse
Affiliation(s)
- Michael P O'Leary
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Audrey H Choi
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Sang-In Kim
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Shyambabu Chaurasiya
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Jianming Lu
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Anthony K Park
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Yanghee Woo
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Susanne G Warner
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Yuman Fong
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA.,Center for Gene Therapy, Department of Hematologic and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Nanhai G Chen
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA. .,Center for Gene Therapy, Department of Hematologic and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA. .,Gene Editing and Viral Vector Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
9
|
Okoli A, Okeke MI, Tryland M, Moens U. CRISPR/Cas9-Advancing Orthopoxvirus Genome Editing for Vaccine and Vector Development. Viruses 2018; 10:E50. [PMID: 29361752 PMCID: PMC5795463 DOI: 10.3390/v10010050] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/17/2022] Open
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR)/associated protein 9 (Cas9) technology is revolutionizing genome editing approaches. Its high efficiency, specificity, versatility, flexibility, simplicity and low cost have made the CRISPR/Cas9 system preferable to other guided site-specific nuclease-based systems such as TALENs (Transcription Activator-like Effector Nucleases) and ZFNs (Zinc Finger Nucleases) in genome editing of viruses. CRISPR/Cas9 is presently being applied in constructing viral mutants, preventing virus infections, eradicating proviral DNA, and inhibiting viral replication in infected cells. The successful adaptation of CRISPR/Cas9 to editing the genome of Vaccinia virus paves the way for its application in editing other vaccine/vector-relevant orthopoxvirus (OPXV) strains. Thus, CRISPR/Cas9 can be used to resolve some of the major hindrances to the development of OPXV-based recombinant vaccines and vectors, including sub-optimal immunogenicity; transgene and genome instability; reversion of attenuation; potential of spread of transgenes to wildtype strains and close contacts, which are important biosafety and risk assessment considerations. In this article, we review the published literature on the application of CRISPR/Cas9 in virus genome editing and discuss the potentials of CRISPR/Cas9 in advancing OPXV-based recombinant vaccines and vectors. We also discuss the application of CRISPR/Cas9 in combating viruses of clinical relevance, the limitations of CRISPR/Cas9 and the current strategies to overcome them.
Collapse
Affiliation(s)
- Arinze Okoli
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
| | - Malachy I Okeke
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
| | - Morten Tryland
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
- Artic Infection Biology, Department of Artic and Marine Biology, The Artic University of Norway, N-9037 Tromsø, Norway.
| | - Ugo Moens
- Molecular Inflammation Research Group, Institute of Medical Biology, The Arctic University of Norway, N-9037 Tromsø, Norway.
| |
Collapse
|
10
|
Dowall SD, Carroll MW, Hewson R. Development of vaccines against Crimean-Congo haemorrhagic fever virus. Vaccine 2017; 35:6015-6023. [PMID: 28687403 PMCID: PMC5637709 DOI: 10.1016/j.vaccine.2017.05.031] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/24/2017] [Accepted: 05/10/2017] [Indexed: 11/19/2022]
Abstract
Crimean-Congo haemorrhagic fever virus (CCHFV) is a deadly human pathogen of the utmost seriousness being highly lethal causing devastating disease symptoms that result in intense and prolonged suffering to those infected. During the past 40years, this virus has repeatedly caused sporadic outbreaks responsible for relatively low numbers of human casualties, but with an alarming fatality rate of up to 80% in clinically infected patients. CCHFV is transmitted to humans by Hyalomma ticks and contact with the blood of viremic livestock, additionally cases of human-to-human transmission are not uncommon in nosocomial settings. The incidence of CCHF closely matches the geographical range of permissive ticks, which are widespread throughout Africa, Asia, the Middle East and Europe. As such, CCHFV is the most widespread tick-borne virus on earth. It is a concern that recent data shows the geographic distribution of Hyalomma ticks is expanding. Migratory birds are also disseminating Hyalomma ticks into more northerly parts of Europe thus potentially exposing naïve human populations to CCHFV. The virus has been imported into the UK on two occasions in the last five years with the first fatal case being confirmed in 2012. A licensed vaccine to CCHF is not available. In this review, we discuss the background and complications surrounding this limitation and examine the current status and recent advances in the development of vaccines against CCHFV.
Collapse
Affiliation(s)
- Stuart D Dowall
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Miles W Carroll
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Roger Hewson
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| |
Collapse
|
11
|
|
12
|
van den Doel P, Volz A, Roose JM, Sewbalaksing VD, Pijlman GP, van Middelkoop I, Duiverman V, van de Wetering E, Sutter G, Osterhaus ADME, Martina BEE. Recombinant modified vaccinia virus Ankara expressing glycoprotein E2 of Chikungunya virus protects AG129 mice against lethal challenge. PLoS Negl Trop Dis 2014; 8:e3101. [PMID: 25188230 PMCID: PMC4154657 DOI: 10.1371/journal.pntd.0003101] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 07/07/2014] [Indexed: 01/04/2023] Open
Abstract
Chikungunya virus (CHIKV) infection is characterized by rash, acute high fever, chills, headache, nausea, photophobia, vomiting, and severe polyarthralgia. There is evidence that arthralgia can persist for years and result in long-term discomfort. Neurologic disease with fatal outcome has been documented, although at low incidences. The CHIKV RNA genome encodes five structural proteins (C, E1, E2, E3 and 6K). The E1 spike protein drives the fusion process within the cytoplasm, while the E2 protein is believed to interact with cellular receptors and therefore most probably constitutes the target of neutralizing antibodies. We have constructed recombinant Modified Vaccinia Ankara (MVA) expressing E3E2, 6KE1, or the entire CHIKV envelope polyprotein cassette E3E26KE1. MVA is an appropriate platform because of its demonstrated clinical safety and its suitability for expression of various heterologous proteins. After completing the immunization scheme, animals were challenged with CHIV-S27. Immunization of AG129 mice with MVAs expressing E2 or E3E26KE1 elicited neutralizing antibodies in all animals and provided 100% protection against lethal disease. In contrast, 75% of the animals immunized with 6KE1 were protected against lethal infection. In conclusion, MVA expressing the glycoprotein E2 of CHIKV represents as an immunogenic and effective candidate vaccine against CHIKV infections.
Collapse
Affiliation(s)
- Petra van den Doel
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Asisa Volz
- Institute for Infectious Diseases and Zoonoses, University of Munich LMU, Munich, Germany
| | - Jouke M. Roose
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | | | - Vincent Duiverman
- Erasmus Medical Center Laboratory Animal Science Center (EDC), Rotterdam, The Netherlands
| | - Eva van de Wetering
- Erasmus Medical Center Laboratory Animal Science Center (EDC), Rotterdam, The Netherlands
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, University of Munich LMU, Munich, Germany
| | - Albert D. M. E. Osterhaus
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Artemis One Health, Utrecht, The Netherlands
| | - Byron E. E. Martina
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Artemis One Health, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
13
|
Clinical development of Modified Vaccinia virus Ankara vaccines. Vaccine 2013; 31:4241-6. [PMID: 23523410 DOI: 10.1016/j.vaccine.2013.03.020] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/21/2022]
Abstract
The smallpox vaccine Vaccinia was successfully used to eradicate smallpox, but although very effective, it was a very reactogenic vaccine and responsible for the deaths of one or two people per million vaccinated. Modified Vaccinia virus Ankara (MVA) is a replication-deficient and attenuated derivative, also used in the smallpox eradication campaign and now being developed as a recombinant viral vector to produce vaccines against infectious diseases and cancer. Many clinical trials of these new vaccines have been conducted, and the findings of these trials are reviewed here. The safety of MVA is now well documented, immunogenicity is influenced by the dose and vaccination regimen, and information on the efficacy of MVA-vectored vaccines is now beginning to accumulate.
Collapse
|
14
|
Cottingham MG, Carroll MW. Recombinant MVA vaccines: dispelling the myths. Vaccine 2013; 31:4247-51. [PMID: 23523407 DOI: 10.1016/j.vaccine.2013.03.021] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 03/11/2013] [Indexed: 12/22/2022]
Abstract
Diseases such as HIV/AIDS, tuberculosis, malaria and cancer are prime targets for prophylactic or therapeutic vaccination, but have proven partially or wholly resistant to traditional approaches to vaccine design. New vaccines based on recombinant viral vectors expressing a foreign antigen are under intense development for these and other indications. One of the most advanced and most promising vectors is the attenuated, non-replicating poxvirus MVA (modified vaccinia virus Ankara), a safer derivative of the uniquely successful smallpox vaccine. Despite the ability of recombinant MVA to induce potent humoral and cellular immune responses against transgenic antigen in humans, especially when used as the latter element of a heterologous prime-boost regimen, doubts are occasionally expressed about the ultimate feasibility of this approach. In this review, five common misconceptions over recombinant MVA are discussed, and evidence is cited to show that recombinant MVA is at least sufficiently genetically stable, manufacturable, safe, and immunogenic (even in the face of prior anti-vector immunity) to warrant reasonable hope over the feasibility of large-scale deployment, should useful levels of protection against target pathogens, or therapeutic benefit for cancer, be demonstrated in efficacy trials.
Collapse
Affiliation(s)
- Matthew G Cottingham
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ, UK.
| | | |
Collapse
|
15
|
Immunogenicity of a vaccine regimen composed of simian immunodeficiency virus DNA, rMVA, and viral particles administered to female rhesus macaques via four different mucosal routes. J Virol 2013; 87:4738-50. [PMID: 23408627 DOI: 10.1128/jvi.03531-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A comparative evaluation of the immunity stimulated with a vaccine regimen that includes simian immunodeficiency virus (SIV), interleukin 2 (IL-2), and IL-15 DNAs, recombinant modified vaccinia virus Ankara (rMVA), and inactivated SIVmac239 particles administered into the oral and nasal cavities, small intestine, and vagina was carried out in female rhesus macaques to determine the best route to induce diverse anti-SIV immunity that may be critical to protection from SIV infection and disease. All four immunizations generated mucosal SIV-specific IgA. Oral immunization was as effective as vaginal immunization in inducing SIV-specific IgA in vaginal secretions and generated greater IgA responses in rectal secretions and saliva samples compared to the other immunization routes. All four immunizations stimulated systemic T-cell responses against Gag and Env, albeit to a different extent, with oral immunization providing greater magnitude and nasal immunization providing wider functional heterogeneity. SIV-specific T cells producing gamma interferon (IFN-γ) dominated these responses. Limited levels of SIV-specific IgG antibodies were detected in plasma samples, and no SIV-specific IgG antibodies were detected in secretions. Vaccination also induced CD4(+) and CD8(+) T-cell responses in the rectal and vaginal mucosa with greater functional heterogeneity than in blood samples. Rectal T-cell responses were significantly greater in the orally vaccinated animals than in the other animals. The most balanced, diverse, and higher-magnitude vaginal T-cell responses were observed after intestinal vaccination. Significantly higher CD8(+) granzyme B-positive T-cell responses were observed systemically after intestinal vaccination and in rectal cells after oral immunization. The majority of SIV-specific T cells that produced granzyme B did not produce cytokines. Of the immunization routes tested, oral vaccination provided the most diverse and significant response to the vaccine.
Collapse
|
16
|
O'Connell RJ, Kim JH, Corey L, Michael NL. Human immunodeficiency virus vaccine trials. Cold Spring Harb Perspect Med 2012; 2:a007351. [PMID: 23209178 PMCID: PMC3543076 DOI: 10.1101/cshperspect.a007351] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
More than 2 million AIDS-related deaths occurred globally in 2008, and more than 33 million people are living with HIV/AIDS. Despite promising advances in prevention, an estimated 2.7 million new HIV infections occurred in that year, so that for every two patients placed on combination antiretroviral treatment, five people became infected. The pandemic poses a formidable challenge to the development, progress, and stability of global society 30 years after it was recognized. Experimental preventive HIV-1 vaccines have been administered to more than 44,000 human volunteers in more than 187 separate trials since 1987. Only five candidate vaccine strategies have been advanced to efficacy testing. The recombinant glycoprotein (rgp)120 subunit vaccines, AIDSVAX B/B and AIDSVAX B/E, and the Merck Adenovirus serotype (Ad)5 viral-vector expressing HIV-1 Gag, Pol, and Nef failed to show a reduction in infection rate or lowering of postinfection viral set point. Most recently, a phase III trial that tested a heterologous prime-boost vaccine combination of ALVAC-HIV vCP1521 and bivalent rgp120 (AIDSVAX B/E) showed 31% efficacy in protection from infection among community-risk Thai participants. A fifth efficacy trial testing a DNA/recombinant(r) Ad5 prime-boost combination is currently under way. We review the clinical trials of HIV vaccines that have provided insight into human immunogenicity or efficacy in preventing HIV-1 infection.
Collapse
Affiliation(s)
- Robert J O'Connell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | | | |
Collapse
|
17
|
Zanetti FA, Del Médico Zajac MP, Taboga OA, Calamante G. Evaluation of modified vaccinia virus Ankara expressing VP2 protein of infectious bursal disease virus as an immunogen in chickens. J Vet Sci 2012; 13:199-201. [PMID: 22705743 PMCID: PMC3386346 DOI: 10.4142/jvs.2012.13.2.199] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A recombinant modified vaccinia Ankara (MVA) virus expressing mature viral protein 2 (VP2) of the infectious bursal disease virus (IBDV) was constructed to develop MVA-based vaccines for poultry. We demonstrated that this recombinant virus was able to induce a specific immune response by observing the production of anti-IBDV-seroneutralizing antibodies in specific pathogen-free chickens. Besides, as the epitopes of VP2 responsible to induce IBDV-neutralizing antibodies are discontinuous, our results suggest that VP2 protein expressed from MVA-VP2 maintained the correct conformational structure. To our knowledge, this is the first report on the usefulness of MVA-based vectors for developing recombinant vaccines for poultry.
Collapse
Affiliation(s)
- Flavia Adriana Zanetti
- Consejo Nacional de Investigaciones Científicas y Técnicas, Rivadavia 1917 (C1033AAJ) Ciudad Autónoma de Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
18
|
Pollara JJ, Spesock AH, Pickup DJ, Laster SM, Petty ITD. Production of prostaglandin E₂ in response to infection with modified vaccinia Ankara virus. Virology 2012; 428:146-55. [PMID: 22534090 DOI: 10.1016/j.virol.2012.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 03/26/2012] [Indexed: 11/26/2022]
Abstract
Prostaglandin E₂ (PGE₂) is an arachidonic acid (AA)-derived signaling molecule that can influence host immune responses to infection or vaccination. In this study, we investigated PGE₂ production in vitro by cells infected with the poxvirus vaccine strain, modified vaccinia Ankara virus (MVA). Human THP-1 cells, murine bone marrow-derived dendritic cells, and murine C3HA fibroblasts all accumulated PGE₂ to high levels in culture supernatants upon infection with MVA. We also demonstrated that MVA induced the release of AA from infected cells, and this was, most unusually, independent of host cytosolic phospholipase A₂ activity. The accumulation of AA and PGE₂ was dependent on viral gene expression, but independent of canonical NF-κB signaling via p65/RelA. The production of PGE₂ required host cyclooxygenase-2 (COX-2) activity, and COX-2 protein accumulated during MVA infection. The results of this study provide insight into a novel aspect of MVA biology that may affect the efficacy of MVA-based vaccines.
Collapse
Affiliation(s)
- Justin J Pollara
- Department of Microbiology, North Carolina State University, Raleigh, NC 27695, USA
| | | | | | | | | |
Collapse
|
19
|
Top S, Foucras G, Deplanche M, Rives G, Calvalido J, Comtet L, Bertagnoli S, Meyer G. Myxomavirus as a vector for the immunisation of sheep: Protection study against challenge with bluetongue virus. Vaccine 2012; 30:1609-16. [DOI: 10.1016/j.vaccine.2011.12.108] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 12/15/2011] [Accepted: 12/22/2011] [Indexed: 10/14/2022]
|
20
|
Rahbar R, Rogers E, Murooka T, Kislinger T, Fish EN. Glomulin: a permissivity factor for vaccinia virus infection. J Interferon Cytokine Res 2012; 32:127-37. [PMID: 22280104 DOI: 10.1089/jir.2011.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In earlier studies we provided evidence that vaccinia virus (VACV) phosphorylation-activation of host cell signaling effectors is critical for subsequent viral replication. In this report, using mass spectrometry-based proteomics, we have identified 387 host cell proteins that co-immunoprecipitate with VACV in infected, permissive PM1.CCR5 human T cells. Among these, glomulin was distinguishable based on its known interaction with a tyrosine kinase receptor, c-Met, its ability to become tyrosine-phosphorylated, and its association with signaling effectors. siRNA knockdown of glomulin expression in PM1.CCR5 T cells reduces VACV infection. Glomulin interacts with the inactive, nonphosphorylated form of c-MET. We demonstrate that treatment of PM1.CCR5 T cells with a c-Met phosphorylation inhibitor leads to a significant reduction in VACV infectivity. Additionally, inhibition of phosphorylation of c-Met abrogates VACV-inducible phosphorylation of Erk 1/2 and IRS-2, signaling effectors identified as critical for VACV infection. These data identify glomulin as a permissivity factor for VACV infection and as a potential therapeutic target for inhibition of VACV infection.
Collapse
Affiliation(s)
- Ramtin Rahbar
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
21
|
Kulkarni V, Jalah R, Ganneru B, Bergamaschi C, Alicea C, von Gegerfelt A, Patel V, Zhang GM, Chowdhury B, Broderick KE, Sardesai NY, Valentin A, Rosati M, Felber BK, Pavlakis GN. Comparison of immune responses generated by optimized DNA vaccination against SIV antigens in mice and macaques. Vaccine 2011; 29:6742-54. [PMID: 21195080 PMCID: PMC3115438 DOI: 10.1016/j.vaccine.2010.12.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Optimized DNA vectors were constructed comprising the proteome of SIV including the structural, enzymatic, regulatory, and accessory proteins. In addition to native antigens as produced by the virus, fusion proteins and modified antigens with altered secretion, cellular localization and stability characteristics were generated. The DNA vectors were tested for expression upon transfection in human cells. In addition, the vectors were tested either alone or in combinations in mice and macaques, which provided an opportunity to compare immune responses in two animal models. DNA only immunization using intramuscular injection in the absence or presence of in vivo electroporation did not alter the phenotype of the induced T cell responses in mice. Although several fusion proteins induced immune responses to all the components of a polyprotein, we noted fusion proteins that abrogated immune response to some of the components. Since the expression levels of such fusion proteins were not affected, these data suggest that the immune recognition of certain components was altered by the fusion. Testing different DNA vectors in mice and macaques revealed that a combination of DNAs producing different forms of the same antigen generated more balanced immune responses, a desirable feature for an optimal AIDS vaccine.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/immunology
- Animals
- Antigens, Viral/immunology
- Cloning, Molecular
- Electroporation
- Enzyme-Linked Immunospot Assay
- Female
- Flow Cytometry
- Gene Products, env/genetics
- Gene Products, env/immunology
- Gene Products, env/metabolism
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Gene Products, gag/metabolism
- Genetic Vectors
- HEK293 Cells
- HIV-1/genetics
- HIV-1/immunology
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Interferon-gamma/immunology
- Macaca mulatta
- Mice
- Mice, Inbred BALB C
- Plasmids/genetics
- Plasmids/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- SAIDS Vaccines/administration & dosage
- SAIDS Vaccines/immunology
- Simian Immunodeficiency Virus/genetics
- Simian Immunodeficiency Virus/immunology
- Transfection
- Vaccination
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Rashmi Jalah
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Brunda Ganneru
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Cristina Bergamaschi
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Agneta von Gegerfelt
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Vainav Patel
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Gen-Mu Zhang
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Bhabadeb Chowdhury
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | | | | | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| |
Collapse
|
22
|
Abstract
Since the eradication of smallpox, immunological studies of smallpox vaccine (vaccinia virus) have provided great gains in understanding immunology as well as depecting smallpox vaccine-derived immune responses. Despite this wealth of knowledge, two confounding problems remain. First, that surviving smallpox provides an individual with lifelong protective immunity, whereas vaccination may not, and second, that specific molecular correlates of protection against smallpox remain vague. Historical literature on smallpox and contemporary studies of other human infections with orthopoxviruses, such as monkeypox, indicate that vaccination may lower disease risks, but it does not provide complete protection against infection in all individuals. Factors impacting protective immunity include longevity of immunologic memory post-vaccination, challenge dose and differences in the challenge viruses, as well as route of exposure. This article discusses historical information regarding smallpox attack rates during outbreaks and contemporary views on aspects of vaccines and naturally occurring orthopoxvirus outbreaks as related to vaccine-derived immunity.
Collapse
Affiliation(s)
| | - Mary G Reynolds
- Centers for Disease Control & Prevention, Division of High Consequence Pathogens & Pathology, Poxvirus & Rabies Branch, Poxivirus Program, 1600 Clifton Road, Atlanta, GA 30339, USA
| |
Collapse
|
23
|
Abern M, Kaufman HL, Latchamsetty K. An update on TroVax for the treatment of progressive castration-resistant prostate cancer. Onco Targets Ther 2011; 4:33-41. [PMID: 21691576 PMCID: PMC3116792 DOI: 10.2147/ott.s14271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Indexed: 11/23/2022] Open
Abstract
Prostate cancer is a common human malignancy with few effective therapeutic options for treating advanced castration-resistant disease. The potential therapeutic effectiveness of immunotherapy and vaccines, in particular, has gained popularity based on the identification of prostate-associated antigens, potent expression vectors for vaccination, and data from recent clinical trials. A modified vaccinia Ankara (MVA) virus expressing 5T4, a tumor-associated glycoprotein, has shown promise in preclinical studies and clinical trials in patients with colorectal and renal cell carcinoma. This review will discuss the rationale for immunotherapy in prostate cancer and describe preclinical and limited clinical data in prostate cancer for the MVA-5T4 (TroVax®) vaccine.
Collapse
Affiliation(s)
- Michael Abern
- Department of Urology, Rush University Medical Center, Chicago, IL, USA
| | | | | |
Collapse
|
24
|
Manrique M, Kozlowski PA, Cobo-Molinos A, Wang SW, Wilson RL, Montefiori DC, Mansfield KG, Carville A, Aldovini A. Long-term control of simian immunodeficiency virus mac251 viremia to undetectable levels in half of infected female rhesus macaques nasally vaccinated with simian immunodeficiency virus DNA/recombinant modified vaccinia virus Ankara. THE JOURNAL OF IMMUNOLOGY 2011; 186:3581-93. [PMID: 21317390 DOI: 10.4049/jimmunol.1002594] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The efficacy of two SIV DNA plus recombinant modified vaccinia virus Ankara nasal vaccine regimens, one combined with plasmids expressing IL-2 and IL-15, the other with plasmids expressing GM-CSF, IL-12, and TNF-α, which may better stimulate humoral responses, was evaluated in two female rhesus macaque groups. Vaccination stimulated significant SIV-specific mucosal and systemic cell-mediated immunity in both groups, whereas SIV-specific IgA titers were sporadic and IgG titers negative. All vaccinated animals, except one, became infected after intravaginal SIV(mac251) low-dose challenge. Half of the vaccinated, infected animals (7/13) promptly controlled virus replication to undetectable viremia for the duration of the trial (130 wk) and displayed virological and immunological phenotypes similar to those of exposed, uninfected individuals. When all vaccinated animals were considered, a 3-log viremia reduction was observed, compared with controls. The excellent viral replication containment achieved in vaccinated animals translated into significant preservation of circulating α4β7(high+)/CD4(+) T cells and of circulating and mucosal CD4(+)/C(M) T cells and in reduced immune activation. A more significant long-term survival was also observed in these animals. Median survival was 72 wk for the control group, whereas >50% of the vaccinated animals were still disease free 130 wk postchallenge, when the trial was closed. There was a statistically significant correlation between levels of CD4(+)/IFN-γ(+) and CD8(+)/IFN-γ(+) T cell percentages on the day of challenge and the control of viremia at week 60 postchallenge or survival. Postchallenge immunological correlates of protection were systemic anti-SIV Gag + Env CD4(+)/IL-2(+), CD4(+)/IFN-γ(+), and CD8(+)/TNF-α(+) T cells and vaginal anti-SIV Gag + Env CD8(+) T cell total monofunctional responses.
Collapse
Affiliation(s)
- Mariana Manrique
- Department of Medicine, Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Paris RM, Kim JH, Robb ML, Michael NL. Prime-boost immunization with poxvirus or adenovirus vectors as a strategy to develop a protective vaccine for HIV-1. Expert Rev Vaccines 2010; 9:1055-69. [PMID: 20822348 DOI: 10.1586/erv.10.106] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Challenges in the development of an effective HIV-1 vaccine are myriad with significant hurdles posed by viral diversity, the lack of a human correlate of protection and difficulty in creating immunogens capable of eliciting broadly neutralizing antibodies. The implicit requirement for novel approaches to these problems has resulted in vaccine candidates designed to elicit cellular and/or humoral immune responses, to include recombinant DNA, viral and bacterial vectors, and subunit proteins. Here, we review data from clinical studies primarily of poxvirus and adenovirus vector vaccines, used in a heterologous prime-boost combination strategy. Currently, this strategy appears to hold the most promise for an effective vaccine based on results from immunogenicity testing and nonhuman primate challenge models, as well as the modest efficacy recently observed in the Thai prime-boost trial.
Collapse
Affiliation(s)
- Robert M Paris
- US Military HIV Research Program (MHRP), Department of Retrovirology, Armed Forces Research Institute of Medical Sciences, 315/6 Rajvithi Road, Bangkok, 10400, Thailand.
| | | | | | | |
Collapse
|
26
|
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
|
27
|
Zheng Y, Ourmanov I, Goeken RM, Whitted S, Brown CR, Buckler-White A, Iyengar R, Plishka RJ, Hirsch VM. Correction of a carboxyl terminal simian immunodeficiency virus Nef frameshift mutation restores virus replication in macaques. Virology 2010; 401:207-14. [PMID: 20303562 PMCID: PMC3418331 DOI: 10.1016/j.virol.2010.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 11/16/2009] [Accepted: 02/22/2010] [Indexed: 10/19/2022]
Abstract
Previous studies demonstrated that the nef gene is a critical determinant of the pathogenicity of simian immunodeficiency virus (SIV) in macaques. In the present study, we evaluated the effect of a spontaneous frameshift mutation in the C-terminus of the nef gene of the minimally pathogenic SIVsmH4i clone. This clone exhibited a single nucleotide deletion in the nef gene relative to pathogenic SIV clones that resulted in a frameshift and addition of 46 amino acids to the C-terminus of Nef. We generated a corrected version of this clone, SIVsmH4i Nef+ that restored Nef protein expression. Inoculation of macaques with SIVsmH4i resulted in delayed and low levels of peak viremia. This contrasted with improved kinetics and robust peak viremia in macaques inoculated with the corrected version. Despite the restoration of in vivo replication ability, neither clone resulted in memory CD4+ T cell loss or disease in a period of two years.
Collapse
Affiliation(s)
- Yanfang Zheng
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ilnour Ourmanov
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert M. Goeken
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sonya Whitted
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Charles R. Brown
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alicia Buckler-White
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ranjini Iyengar
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald J. Plishka
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vanessa M. Hirsch
- Laboratory of Molecular Microbiology, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Abstract
AbstractSwine influenza is an important contagious disease in pigs caused by influenza A viruses. Although only three subtypes of influenza A viruses, H1N1, H1N2 and H3N2, predominantly infect pigs worldwide, it is still a big challenge for vaccine manufacturers to produce efficacious vaccines for the prevention and control of swine influenza. Swine influenza viruses not only cause significant economic losses for the swine industry, but are also important zoonotic pathogens. Vaccination is still one of the most important and effective strategies to prevent and control influenza for both the animal and human population. In this review, we will discuss the current status of swine influenza worldwide as well as current and future options to control this economically important swine disease.
Collapse
|
29
|
MVA recombinants expressing the fusion and hemagglutinin genes of PPRV protects goats against virulent challenge. Indian J Microbiol 2010; 50:266-74. [PMID: 23100840 DOI: 10.1007/s12088-010-0026-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 04/03/2009] [Indexed: 10/19/2022] Open
Abstract
Peste des Petits Ruminants (PPR) is a highly contagious animal disease caused by the Peste des Petits Ruminants virus (PPRV) belonging to the genus morbillivirus and family Paramyxoviridae. The disease results in high morbidity and mortality in goats, sheep and in some small wild ruminants. The presence of large number of small ruminants reared in endemic areas makes PPR a notorious disease threatening the livelihood of poor farmers. Conventional vaccination using a live, attenuated vaccine gives adequate protection but cannot be used in case of eradication of the disease due to difficulty in differentiation of infected animals from the vaccinated ones.In the present study, we constructed two recombinant viruses using attenuated Modified Vaccinia virus Ankara virus (MVA) namely MVA-F and MVA-H expressing the full length PPRV fusion (F) and hemagglutinin (H) glycoproteins, respectively. Goats were vaccinated intramuscularly with 105 plaque forming units (PFU) each of the recombinant viruses and a live attenuated vaccine (RAKSHA PPR) and challenged 4 months later with PPRV challenge virus (10(3) goat LD(50)). All goats were completely protected from the clinical disease. This study gave an indication that mass vaccination of small ruminants with either of the above or both recombinant inexpensive virus vaccines could help in possible eradication of PPRV from endemic countries like India and subsequent seromonitoring of the disease for differentiation of infected animals from vaccinated ones.
Collapse
|
30
|
Stech J. Several hemagglutinins of same serotype for induction of broad immunity against influenza A virus antigenic drift variants: WO2008048984. Expert Opin Ther Pat 2010; 20:447-50. [PMID: 20180625 DOI: 10.1517/13543770903547855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The respiratory disease influenza gives rise to severe public health concerns. During inter-pandemic periods, the constant problem of the annually recurring seasonal influenza is perpetuated by the ability of influenza viruses to alter their surface antigens continuously (antigenic drift). Therefore, vaccines eliciting broad immunity against drift variants still remain a major objective in vaccine development. The patent WO2008048984 evaluated in this article claims an approach which aims to elicit homosubtypic protection against drift variants by simultaneous vaccination with several hemagglutinins (HAs) of the same serotype. The proposed multivalent vaccine based on simultaneous administration of several HAs, the results obtained from mice immunization studies and the implications of this concept are discussed in light of their relevance to application in humans. This proof-of-principle study suggests that a multivalent HA vaccine could elicit broad protection against drifted virus variants of one HA subtype. In the future, the dependence of broad efficacy on large antigenic distances among the HAs used for immunization as well as the antigenic distance between the HAs administered to that of the challenge virus, the immunological correlates of broad efficacy, and the suitability of this concept for domestic animals and humans remain to be investigated.
Collapse
Affiliation(s)
- Jürgen Stech
- Friedrich-Loeffler-Institute, Institute of Molecular Biology, Federal Research Institute for Animal Health, Suedufer 10, Greifswald - Insel Riems, 17493, Germany.
| |
Collapse
|
31
|
Pandey A, Singh N, Sambhara S, Mittal SK. Egg-independent vaccine strategies for highly pathogenic H5N1 influenza viruses. HUMAN VACCINES 2010; 6:178-88. [PMID: 19875936 PMCID: PMC2888842 DOI: 10.4161/hv.6.2.9899] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of a highly pathogenic H5N1 influenza virus in Hong Kong in 1997 and the subsequent appearance of other H5N1 strains and their spread to several countries in southeast Asia, Africa, the Middle East and Europe has evoked fear of a global influenza pandemic. Vaccines offer the best hope to combat the threat of an influenza pandemic. However, the global demand for a pandemic vaccine cannot be fulfilled by the current egg-based vaccine manufacturing strategies, thus creating a need to explore alternative technologies for vaccine production and delivery. Several egg-independent vaccine approaches such as cell culture-derived whole virus or subvirion vaccines, recombinant protein-based vaccines, virus-like particle (VLP) vaccines, DNA vaccines and viral vector-based vaccines are currently being investigated and appear promising both in preclinical and clinical studies. The present review will highlight the various egg-independent alternative vaccine approaches for pandemic influenza.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Correspondence: Suresh K. Mittal, Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA, Tel: 765-496-2894, Fax: 765-494-9830, , Suryaprakash Sambhara, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA, Tel: 404-639-3800, Fax: 404-639-5180,
| |
Collapse
|
32
|
Modified H5 promoter improves stability of insert genes while maintaining immunogenicity during extended passage of genetically engineered MVA vaccines. Vaccine 2009; 28:1547-57. [PMID: 19969118 PMCID: PMC2821965 DOI: 10.1016/j.vaccine.2009.11.056] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 11/17/2009] [Accepted: 11/20/2009] [Indexed: 11/22/2022]
Abstract
We have engineered recombinant (r) Modified Vaccinia Ankara (MVA) to express multiple antigens under the control of either of two related vaccinia synthetic promoters (pSyn) with early and late transcriptional activity or the modified H5 (mH5) promoter which has predominant early activity. We sequentially passaged these constructs and analyzed their genetic stability by qPCR, and concluded that rMVA expressing multiple antigens using the mH5 promoter exhibit remarkable genetic stability and maintain potent immunogenicity after serial passage. In contrast, rMVA expressing antigens using engineered vaccinia synthetic E/L (pSyn I or II) promoters are genetically unstable. Progressive accumulation of antigen loss variants resulted in a viral preparation with lower immunogenicity after serial passage. Metabolic labeling, followed by cold chase revealed little difference in stability of proteins expressed from mH5 or pSyn promoter constructs. We conclude that maintenance of genetic stability which is achieved using mH5, though not with pSyn promoters, is linked to timing, not the magnitude of expression levels of foreign antigen, which is more closely associated with immunogenicity of the vaccine.
Collapse
|
33
|
Modified vaccinia Ankara strains with identical coding sequences actually represent complex mixtures of viruses that determine the biological properties of each strain. Vaccine 2009; 27:7442-50. [DOI: 10.1016/j.vaccine.2009.05.095] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 05/26/2009] [Accepted: 05/31/2009] [Indexed: 11/21/2022]
|
34
|
Nasal DNA-MVA SIV vaccination provides more significant protection from progression to AIDS than a similar intramuscular vaccination. Mucosal Immunol 2009; 2:536-50. [PMID: 19741603 DOI: 10.1038/mi.2009.103] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Preventive human immunodeficiency virus (HIV) vaccination may require induction of virus-specific immune responses at mucosal sites to contain viral infection locally after exposure, as most HIV infections occur through mucosal surfaces. We compared the efficacy of an intranasal or intramuscular Simian immunodeficiency virus (SIV)+ interleukin (IL)-2+IL-15 DNA/SIV-MVA (modified vaccinia virus Ankara) vaccination in preventing disease progression in SIVmac251 intrarectally challenged rhesus macaques. SIV-specific rectal IgA responses were more significantly persistent in nasally vaccinated than in intramuscularly vaccinated animals. No significant differences were observed in the magnitude of systemic T-cell responses between the two groups, although the nasal immunization induced more significant anti-SIV T-cell responses in the colorectal mucosa. After challenge, CD4(+) central memory (C(M)) T-cell preservation and significant disease-delay were observed in both vaccination groups. However, nasally vaccinated animals had more significant early preservation of circulating and colorectal CD4(+) C(M) T cells, of circulating CD4(+)/alpha4beta7(+) effector memory (E(M)) T cells, and a longer disease-free interval when compared with the intramuscularly vaccinated or control groups. Regardless of vaccination status, long-term viremia control and preservation of CD4(+) C(M) T cells was detected in animals with significantly higher systemic CD8(+)/tumor necrosis factor (TNF)-alpha(+) and CD8(+)/interferon (IFN)-gamma(+) T-cell responses and higher SIV-specific CD4(+)/IL-2(+) responses in colorectal T cells.
Collapse
|
35
|
Kreijtz J, Suezer Y, de Mutsert G, van den Brand J, van Amerongen G, Schnierle B, Kuiken T, Fouchier R, Löwer J, Osterhaus A, Sutter G, Rimmelzwaan G. Preclinical evaluation of a modified vaccinia virus Ankara (MVA)-based vaccine against influenza A/H5N1 viruses. Vaccine 2009; 27:6296-9. [DOI: 10.1016/j.vaccine.2009.03.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 03/03/2009] [Accepted: 03/09/2009] [Indexed: 11/15/2022]
|
36
|
Gudmundsdotter L, Nilsson C, Brave A, Hejdeman B, Earl P, Moss B, Robb M, Cox J, Michael N, Marovich M, Biberfeld G, Sandström E, Wahren B. Recombinant Modified Vaccinia Ankara (MVA) effectively boosts DNA-primed HIV-specific immune responses in humans despite pre-existing vaccinia immunity. Vaccine 2009; 27:4468-74. [PMID: 19450644 PMCID: PMC4788966 DOI: 10.1016/j.vaccine.2009.05.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2009] [Revised: 04/27/2009] [Accepted: 05/10/2009] [Indexed: 10/20/2022]
Abstract
The presence of vector-specific immune responses may hamper the induction of responses to a foreign antigen encoded by the vector. We evaluated the impact of pre-existing immunity to vaccinia virus on the induction of HIV-specific responses after immunization of healthy volunteers with a HIV-1 DNA prime-MVA boost vaccine. Following three priming immunizations with HIV-1 DNA plasmids, the volunteers were boosted with a single injection of recombinant MVA encoding HIV-1 proteins. Pre-existing immunity to vaccinia virus did not reduce the proportion of individuals who responded to HIV-1, but did lower the magnitude of responses. Our results suggest that vaccinia-based vectors can be used to efficiently induce immune responses to vectored HIV-1 antigens, even in individuals with pre-existing immunity to vaccinia virus.
Collapse
Affiliation(s)
- Lindvi Gudmundsdotter
- Karolinska Institutet and Swedish Institute for Infectious Disease Control, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hodge JW, Higgins J, Schlom J. Harnessing the unique local immunostimulatory properties of modified vaccinia Ankara (MVA) virus to generate superior tumor-specific immune responses and antitumor activity in a diversified prime and boost vaccine regimen. Vaccine 2009; 27:4475-82. [PMID: 19450631 PMCID: PMC3518379 DOI: 10.1016/j.vaccine.2009.05.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 04/28/2009] [Accepted: 05/10/2009] [Indexed: 10/20/2022]
Abstract
Recombinant poxviruses expressing tumor-associated antigens (TAAs) are currently being evaluated in clinical trials as an approach to treat various cancers. We have previously generated poxviral vectors expressing a TAA and a TRIad of COstimulatory Molecules (B7-1, ICAM-1, and LFA-3; TRICOM) as transgenes, including replication competent recombinant vaccinia (rV) or replication-defective modified vaccinia Ankara (MVA), to prime tumor-specific immune responses, and a replication-defective recombinant fowlpox (rF) to boost these responses. MVA is a potentially safer, replication-defective form of vaccinia virus with unique immunostimulatory properties that could make it a superior priming vaccine. Here, an MVA vector encoding a tumor antigen (CEA) and TRICOM was utilized (rMVA). A single rMVA-CEA/TRICOM vaccination induced greater expression of several serum cytokines associated with enhanced T-cell immunity than that seen with vaccinia. We hypothesized that this effect might "precondition" the vaccination site for a more effective boost. An rMVA-CEA/TRICOM prime followed 7 days later (but not 30 days later) by an rF-CEA/TRICOM boost at the same injection site (but not at a distal site) induced more potent CEA-specific T-cell responses, and superior CEA-specific immunity and antitumor activity, than rV-CEA/TRICOM followed by rF-CEA/TRICOM. This preconditioning effect was also observed using a heterologous antigen model, where priming with rMVA-CEA/TRICOM followed 7 days later by rF-LacZ/TRICOM enhanced beta-gal-specific immunity compared to rF-LacZ/TRICOM only. The studies reported here show for the first time that priming with rMVA followed 7 days later by an rF boost at the same injection site, versus a distal site, generates superior tumor-specific immunity and antitumor activity.
Collapse
Affiliation(s)
| | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
38
|
Aravindaram K, Kuo TY, Lan CW, Yu HH, Wang PH, Chen YS, Chen GHC, Yang NS. Protective immunity against porcine circovirus 2 in mice induced by a gene-based combination vaccination. J Gene Med 2009; 11:288-301. [PMID: 19194994 DOI: 10.1002/jgm.1300] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Porcine circovirus type 2 (PCV2) is the primary cause of an emerging swine disease, postweaning multisystemic wasting syndrome, that is responsible for economic losses. To develop an effective vaccine for PCV2, we evaluated a heterologous prime-boost vaccine approach, using a gene gun-mediated naked DNA vector as a priming and modified vaccinia virus ankara (MVA) as a booster, in Balb/c mice. METHODS Three open reading frames (ORF) of PCV2 viral samples from infected pigs were amplified, and gene gun-mediated DNA priming vaccination was performed followed by boosts with MVA vectors expressing the same ORFs of PCV2. After vaccination, mice were challenged with PCV2 virus, and virus titers in the lungs and lymph nodes were measured. RESULTS The combination of ORF-2 and -3 in this gene-based vaccine strategy resulted in high antibody titers and virus neutralization activity in serum, reduced PCV2 virus load, and reduced levels of apoptosis in the lungs. No cross-reaction was observed between ORF-1 and -2, but weak cross-reaction was observed between ORF-1 and -3, and between ORF-2 and -3. Following vaccination, expression of chemokines, macrophage inflammatory protein-1beta and regulated upon activation, normal T cell expressed and secreted, increased significantly. The expression of T helper 1-type cytokine (interferon-gamma) and specific lysis of PCV2-infected cells increased; concomitantly, the level of T helper 2-type cytokine (interleukin-10) decreased in test mice. The expression of tumor necrosis factor-alpha and granulocyte-macrophage colony-stimulating factor increased significantly in mice vaccinated with ORF-2/-3, and with ORF-1/-2/-3. CONCLUSIONS This prime-boost vaccination strategy, using a gene gun for DNA priming and recombinant MVA for boosts, may be an attractive vaccine strategy against PCV2 infection in swine.
Collapse
Affiliation(s)
- Kandan Aravindaram
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kreijtz JHCM, Suezer Y, de Mutsert G, van den Brand JMA, van Amerongen G, Schnierle BS, Kuiken T, Fouchier RAM, Löwer J, Osterhaus ADME, Sutter G, Rimmelzwaan GF. Recombinant modified vaccinia virus Ankara expressing the hemagglutinin gene confers protection against homologous and heterologous H5N1 influenza virus infections in macaques. J Infect Dis 2009; 199:405-13. [PMID: 19061423 DOI: 10.1086/595984] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Highly pathogenic avian influenza viruses of the H5N1 subtype have been responsible for an increasing number of infections in humans since 2003. More than 60% of infected individuals die, and new infections are reported frequently. In light of the pandemic threat caused by these events, the rapid availability of safe and effective vaccines is desirable. Modified vaccinia virus Ankara (MVA) expressing the hemagglutinin (HA) gene of H5N1 viruses is a promising candidate vaccine that induced protective immunity against infection with homologous and heterologous H5N1 influenza virus in mice. METHODS In the present study, we evaluated a recombinant MVA vector expressing the HA gene of H5N1 influenza virus A/Vietnam/1194/04 (MVA-HA-VN/04) in nonhuman primates. Cynomolgus macaques were immunized twice and then were challenged with influenza virus A/Vietnam/1194/04 (clade 1) or A/Indonesia/5/05 (clade 2.1) to assess the level of protective immunity. RESULTS Immunization with MVA-HA-VN/04 induced (cross-reactive) antibodies and prevented virus replication in the upper and lower respiratory tract and the development of severe necrotizing bronchointerstitial pneumonia. CONCLUSION Therefore, MVA-HA-VN/04 is a promising vaccine candidate for the induction of protective immunity against highly pathogenic H5N1 avian influenza viruses in humans.
Collapse
Affiliation(s)
- J H C M Kreijtz
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pignolet B, Boullier S, Gelfi J, Bozzetti M, Russo P, Foulon E, Meyer G, Delverdier M, Foucras G, Bertagnoli S. Safety and immunogenicity of myxoma virus as a new viral vector for small ruminants. J Gen Virol 2008; 89:1371-1379. [PMID: 18474552 DOI: 10.1099/vir.0.83595-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Myxoma virus (MYXV), a leporide-specific poxvirus, represents an attractive candidate for the generation of safe and non-replicative vaccine vectors for other species. With the aim of developing new recombinant vaccines for ruminants, we evaluated the safety and the immunogenicity of recombinant MYXV in sheep. In vitro studies indicated that ovine primary fibroblasts were not permissive for MYXV and that infection of ovine peripheral blood mononuclear cells occurred at a low rate. Although non-specific activation significantly improved the susceptibility of lymphocytes, MYXV infection remained abortive. Histological and immunohistochemical examination at the inoculation sites revealed the development of an inflammatory process and allowed the detection of sparse infected cells in the dermis. In addition, inoculated sheep developed an antibody response directed against MYXV and the product of the transgene. Overall, these results provide the first line of evidence on the potential of MYXV as a viral vector for ruminants.
Collapse
Affiliation(s)
- Béatrice Pignolet
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Séverine Boullier
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Jacqueline Gelfi
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Marjorie Bozzetti
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Pierre Russo
- AFSSA LERPRA les Templiers, 105 route des Chappes, F-06902 Sophia Antipolis, France
| | - Eliane Foulon
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Gilles Meyer
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Maxence Delverdier
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Gilles Foucras
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| | - Stéphane Bertagnoli
- Université de Toulouse, ENVT, UMR 1225, F-31076 Toulouse, France.,INRA, UMR 1225, F-31076 Toulouse, France
| |
Collapse
|
41
|
Design, construction, and characterization of a multigenic modified vaccinia Ankara candidate vaccine against human immunodeficiency virus type 1 subtype C/B'. J Acquir Immune Defic Syndr 2008; 47:412-21. [PMID: 18209682 DOI: 10.1097/qai.0b013e3181651bb2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The rapid spread of HIV-1 underscores the urgent need to develop an effective vaccine. Using modified vaccinia Ankara (MVA) as a vector, we designed and constructed a multigenic candidate vaccine against a recombinant C/B' subtype of HIV-1 that is dominant in southwest China. Five HIV-1 genes (gag, pol, DeltaV2env, tat, and nef) were introduced into 2 separate regions of the MVA genome using modified single- and dual-promoter insertion vectors. Recombinant MVA was selected by immunofluorescence double-staining and foci purification. The end product is a single recombinant MVA, termed ADMVA, that expresses HIV-1 DeltaV2Env and fusion proteins Gag-Pol and Nef-Tat. By in vitro analyses, all expected HIV-1 proteins were expressed in infected chicken embryo fibroblasts and various human cell lines. Additionally, 2 sequential intramuscular injections of 10(6) 50% tissue infectious culture dose (TCID50) of ADMVA into BALB/c and B6 x B10 mice elicited broad cell-mediated immune responses against all 5 viral proteins as determined by interferon-gamma enzyme immunospot assays. The number of spot-forming cells was in the range of 200 to 800 per million splenocytes, and both CD4 and CD8 T-cell responses were detected. Moreover, high serum titers (>1:20,000) of antibodies against HIV-1 gp120 were also elicited. The magnitude of immune responses correlated with the dose of ADMVA, and the vaccine caused no overt adverse consequences, up to 10(7) TCID50 per injection. ADMVA has since been advanced into clinical trials. A phase 1 study has been completed, and a prime-boost with ADVAX (see accompanying article) is now underway.
Collapse
|
42
|
Paul S, Geist M, Dott K, Snary D, Taylor-Papadimitriou J, Acres B, Silvestre N, Kieny MP, Balloul JM. Specific tumor cell targeting by a recombinant MVA expressing a functional single chain antibody on the surface of intracellular mature virus (IMV) particles. Viral Immunol 2008; 20:664-71. [PMID: 18158739 DOI: 10.1089/vim.2007.0058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinant vaccinia virus with tumor cell specificity may provide a versatile tool either for direct lysis of cancer cells or for the targeted transfer of genes encoding immunomodulatory or toxic molecules. We report the expression of a tumor-specific single-chain antibody on the surface of intracellular mature vaccinia virus particles (IMV). The wild-type p14 externally membrane-associated protein p14 (A27L gene), which is not required for viral binding and replication, was replaced by p14 fusion molecules carrying a single-chain antibody directed against the tumor-associated antigen MUC-1. MUC-1 mucin is an epithelial cell antigen whose aberrant expression plays a role in autoimmunity and tumor immunity in the majority of human carcinomas and multiple myeloma. Fusion protein carrying the single-chain antibody at the NH2-terminal position was expressed and exposed at the envelope of the corresponding recombinant virus. The construct containing the antibody was able to bind a MUC-1 specific 60mer peptide. Moreover, targeted virus infects MUC-1-expressing cells in vitro more efficiently.
Collapse
|
43
|
Manrique M, Micewicz E, Kozlowski PA, Wang SW, Aurora D, Wilson RL, Ghebremichael M, Mazzara G, Montefiori D, Carville A, Mansfield KG, Aldovini A. DNA-MVA vaccine protection after X4 SHIV challenge in macaques correlates with day-of-challenge antiviral CD4+ cell-mediated immunity levels and postchallenge preservation of CD4+ T cell memory. AIDS Res Hum Retroviruses 2008; 24:505-19. [PMID: 18373436 PMCID: PMC2677999 DOI: 10.1089/aid.2007.0191] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The ability of vaccines to induce immunity both in mucosal and systemic compartments may be required for prevention of HIV infection and AIDS. We compared DNA-MVA vaccination regimens adjuvanted by IL-12 DNA, administered intramuscularly and nasally or only nasally. Most of the vaccinated Rhesus macaques developed mucosal and systemic humoral and cell-mediated SHIV-specific immune responses. Stimulation of mucosal anti-Env IgA responses was limited. After rectal challenge with SHIV 89.6P, all vaccinated and naive animals became infected. However, most of the vaccinated animals showed significant control of viremia and protection from CD4(+) T cell loss and AIDS progression compared to the control animals. The levels of CD4(+) and CD8(+) T cell virus-specific responses measured on the day of challenge correlated with the level of viremia control observed later during the chronic infection. Postchallenge viremia levels inversely correlated with the preservation of SHIV-specific CD4(+)/IL-2(+) and CD8(+)/TNF-alpha(+) T cells but not with CD4(+)/IFN-gamma(+) T cells measured over time after challenge. We also found that during the early chronic infection SHIV vaccination permitted a more significant preservation of both naive and memory CD4(+) T cells compared to controls. In addition, we observed a more significant and prolonged preservation of memory CD4(+) T cells after SHIV vaccination and challenge than that observed after SIV vaccination and challenge. As the antiviral immunity stimulated by vaccination is present in the memory CD4(+) T cell subpopulations, its more limited targeting by SHIV compared to SIV may explain the better control of X4 tropic SHIV than R5 tropic SIVs by vaccination.
Collapse
Affiliation(s)
- Mariana Manrique
- Department of Medicine, Children's Hospital Boston, Department of Pediatrics, Harvard Medical School, Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Stanford MM, McFadden G. Myxoma virus and oncolytic virotherapy: a new biologic weapon in the war against cancer. Expert Opin Biol Ther 2007; 7:1415-25. [PMID: 17727330 DOI: 10.1517/14712598.7.9.1415] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Oncolytic virotherapy is an innovative alternative to more conventional cancer therapies. The ability of some viruses to specifically target and kill malignant cancerous cells while leaving normal tissue unscathed has opened a large repertoire of new and selective cancer killing therapeutic candidates. Poxviruses, such as vaccinia virus, have a long history of use in humans as live vaccines and have more recently been studied as potential platforms for delivery of immunotherapeutics and attenuated variants of vaccinia have been explored as oncolytic candidates. In contrast, the poxvirus myxoma virus is a novel oncolytic candidate that has no history of use in humans directly, as it has a distinct and absolute host species tropism to lagomorphs (rabbits). Myxoma virus has been recently shown to be able to also selectively infect and kill human tumor cells, a unique tropism that is linked to dysregulated intracellular signalling pathways found in the majority of human cancers. This review outlines the existing knowledge on the tropism of myxoma virus for human cancer cells, as well as preclinical data exhibiting its ability to infect and clear tumors in animal models of cancer. This is an exciting new therapeutic option for treating cancer, and myxoma virus joins a growing group of oncolytic virus candidates that are being developed as a new class of cancer therapies in man.
Collapse
Affiliation(s)
- Marianne M Stanford
- University of Western Ontario, Biotherapeutics Research Group, Robarts Research Institute, Department of Microbiology and Immunology, London, Ontario, N6G 2V4, Canada
| | | |
Collapse
|
45
|
Pignolet B, Duteyrat JL, Allemandou A, Gelfi J, Foucras G, Bertagnoli S. In vitro permissivity of bovine cells for wild-type and vaccinal myxoma virus strains. Virol J 2007; 4:94. [PMID: 17900332 PMCID: PMC2045666 DOI: 10.1186/1743-422x-4-94] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 09/27/2007] [Indexed: 11/10/2022] Open
Abstract
Myxoma virus (MYXV), a leporide-specific poxvirus, represents an attractive candidate for the generation of safe, non-replicative vaccine vector for non-host species. However, there is very little information concerning infection of non-laboratory animals species cells with MYXV. In this study, we investigated interactions between bovine cells and respectively a wild type strain (T1) and a vaccinal strain (SG33) of MYXV. We showed that bovine KOP-R, BT and MDBK cell lines do not support MYXV production. Electron microscopy observations of BT-infected cells revealed the low efficiency of viral entry and the production of defective virions. In addition, infection of bovine peripheral blood mononuclear cells (PBMC) occurred at a very low level, even following non-specific activation, and was always abortive. We did not observe significant differences between the wild type strain and the vaccinal strain of MYXV, indicating that SG33 could be used for new bovine vaccination strategies.
Collapse
Affiliation(s)
- Béatrice Pignolet
- Laboratory « Interactions Hôtes-Virus et Vaccinologie », UMR 1225 INRA-ENVT, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des capelles, Toulouse F-31076, France
| | - Jean-Luc Duteyrat
- Laboratory « Interactions Hôtes-Virus et Vaccinologie », UMR 1225 INRA-ENVT, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des capelles, Toulouse F-31076, France
- Centre de Microscopie Electronique Appliquée à la Biologie, Faculté de Médecine de Rangueil, 133 route de Narbonne, Toulouse, F-31062, France
| | - Aude Allemandou
- Laboratory « Interactions Hôtes-Virus et Vaccinologie », UMR 1225 INRA-ENVT, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des capelles, Toulouse F-31076, France
- Centre de Microscopie Electronique Appliquée à la Biologie, Faculté de Médecine de Rangueil, 133 route de Narbonne, Toulouse, F-31062, France
| | - Jacqueline Gelfi
- Laboratory « Interactions Hôtes-Virus et Vaccinologie », UMR 1225 INRA-ENVT, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des capelles, Toulouse F-31076, France
| | - Gilles Foucras
- laboratory « Résistome des ruminants », UMR 1225 INRA-ENVT, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des capelles, Toulouse F-31076, France
| | - Stéphane Bertagnoli
- Laboratory « Interactions Hôtes-Virus et Vaccinologie », UMR 1225 INRA-ENVT, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des capelles, Toulouse F-31076, France
| |
Collapse
|
46
|
Santra S, Sun Y, Parvani JG, Philippon V, Wyand MS, Manson K, Gomez-Yafal A, Mazzara G, Panicali D, Markham PD, Montefiori DC, Letvin NL. Heterologous prime/boost immunization of rhesus monkeys by using diverse poxvirus vectors. J Virol 2007; 81:8563-70. [PMID: 17553898 PMCID: PMC1951337 DOI: 10.1128/jvi.00744-07] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Accepted: 05/23/2007] [Indexed: 11/20/2022] Open
Abstract
As the diversity of potential immunogens increases within certain classes of vectors, the possibility has arisen of employing heterologous prime/boost immunizations using diverse members of the same family of vectors. The present study was initiated to explore the use of divergent pox vectors in a prime/boost regimen to elicit high-frequency cellular immune responses to human immunodeficiency virus type 1 envelope and simian immunodeficiency virus gag in rhesus monkeys. We demonstrated that monkeys vaccinated with a recombinant modified vaccinia virus Ankara (rMVA) prime/recombinant fowlpox virus (rFPV) boost regimen and monkeys vaccinated with a recombinant vaccinia virus prime/rFPV boost regimen developed comparable cellular immune responses that were greater in magnitude than those elicited by a homologous prime/boost with rMVA. Nevertheless, comparable magnitude recall cellular immune responses were observed in monkeys vaccinated with heterologous and homologous recombinant poxvirus following challenge with the CXCR4-tropic SHIV-89.6P. Consistent with this finding, comparable levels of containment of viral replication and CD4(+) T-lymphocyte preservation were seen in these groups of recombinant poxvirus-vaccinated monkeys. This study supports further exploration of combining recombinant vectors of the same family in prime/boost immunization strategies to optimize vaccine-elicited cellular immune responses.
Collapse
Affiliation(s)
- Sampa Santra
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Guerra S, Nájera JL, González JM, López-Fernández LA, Climent N, Gatell JM, Gallart T, Esteban M. Distinct gene expression profiling after infection of immature human monocyte-derived dendritic cells by the attenuated poxvirus vectors MVA and NYVAC. J Virol 2007; 81:8707-21. [PMID: 17537851 PMCID: PMC1951336 DOI: 10.1128/jvi.00444-07] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although recombinants based on the attenuated poxvirus vectors MVA and NYVAC are currently in clinical trials, the nature of the genes triggered by these vectors in antigen-presenting cells is poorly characterized. Using microarray technology and various analysis conditions, we compared specific changes in gene expression profiling following MVA and NYVAC infection of immature human monocyte-derived dendritic cells (MDDC). Microarray analysis was performed at 6 h postinfection, since these viruses induced extensive cytopathic effects, rRNA breakdown, and apoptosis at late times postinfection. MVA- and NYVAC-infected MDDC shared upregulation of 195 genes compared to uninfected cells: MVA specifically upregulated 359 genes, and NYVAC upregulated 165 genes. Microarray comparison of NYVAC and MVA infection revealed 544 genes with distinct expression patterns after poxvirus infection and 283 genes specifically upregulated after MVA infection. Both vectors upregulated genes for cytokines, cytokine receptors, chemokines, chemokine receptors, and molecules involved in antigen uptake and processing, including major histocompatibility complex genes. mRNA levels for interleukin 12beta (IL-12beta), beta interferon, and tumor necrosis factor alpha were higher after MVA infection than after NYVAC infection. The expression profiles of transcription factors such as NF-kappaB/Rel and STAT were regulated similarly by both viruses; in contrast, OASL, MDA5, and IRIG-I expression increased only during MVA infection. Type I interferon, IL-6, and Toll-like receptor pathways were specifically induced after MVA infection. Following MVA or NYVAC infection in MDDC, we found similarities as well as differences between these virus strains in the expression of cellular genes with immunological function, which should have an impact when these vectors are used as recombinant vaccines.
Collapse
Affiliation(s)
- Susana Guerra
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Campus Universidad Autónoma, E-28049 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhu W, Fang Q, Zhuang K, Wang H, Yu W, Zhou J, Liu L, Tien P, Zhang L, Chen Z. The attenuation of vaccinia Tian Tan strain by the removal of the viral M1L-K2L genes. J Virol Methods 2007; 144:17-26. [PMID: 17459491 PMCID: PMC7112875 DOI: 10.1016/j.jviromet.2007.03.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 03/12/2007] [Accepted: 03/14/2007] [Indexed: 11/25/2022]
Abstract
To generate a safe vaccinia Tian Tan (VTT)-based vaccine vector, it is necessary to develop a method to attenuate the virus. A modified VTT (MVTT(2-GFP)) was constructed by replacing the viral M1L-K2L genes with a GFP gene. In comparison to the parental VTT, MVTT(2-GFP) lost its replication capacity in rabbit RK13 and human HeLa cell lines. The life cycle of viral replication was blocked at different stages in these two cell lines as determined by electron microscope examination. MVTT(2-GFP) was less virulent than VTT for 100-fold by measuring mouse body weight loss after intranasal viral inoculation and for 340-fold by determining the intracranial LD(50) value in mice. The foreign GFP gene was stable genetically after 10 rounds of passage in Vero cells. Importantly, MVTT(2-GFP) elicited both humoral and cell-mediated immune responses to the GFP gene in mice. With two intramuscular inoculations of 10(5)PFU virus, the anti-GFP antibody reciprocal endpoint titer reached over 700 as determined by an ELISA. The number of IFN-gamma secreting T cells reached over 350SFU per million splenocytes against a CD8+ T cell-specific epitope of GFP. Collectively, the removal of the M1L-K2L genes is a useful method to generate an attenuated vaccinia Tian Tan vaccine vector.
Collapse
Affiliation(s)
- Weijun Zhu
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Qing Fang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Ke Zhuang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Haibo Wang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Wenbo Yu
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Jingying Zhou
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Li Liu
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Po Tien
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
| | - Linqi Zhang
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
- AIDS Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Zhiwei Chen
- Modern Virology Research Center and AIDS Center, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei 430072, PR China
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
- Corresponding author at: Aaron Diamond AIDS Research Center, 455 First Avenue, New York, NY 10016, USA. Tel.: +1 212 448 5031; fax: +1 212 725 1126.
| |
Collapse
|
49
|
Berry N, Stebbings R, Brown S, Christian P, Thorstensson R, Ahmed RK, Davis L, Ferguson D, D'Arcy N, Elsley W, Hull R, Lines J, Wade-Evans A, Stott J, Almond N. Immunological responses and viral modulatory effects of vaccination with recombinant modified vaccinia virus Ankara (rMVA) expressing structural and regulatory transgenes of simian immunodeficiency virus (SIVmac32H/J5M). J Med Primatol 2007; 36:80-94. [PMID: 17493138 DOI: 10.1111/j.1600-0684.2007.00216.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND The immunogenicity and protective efficacy of recombinant modified vaccinia virus Ankara (rMVA) vectors expressing structural (gag/pol, env) and regulatory (tat, rev, nef) genes of SIVmac251/32H-J5 (rMVA-J5) were assessed. METHODS Immunization with rMVA constructs (2.5 x 10(7) IU) 32, 20 and 8 weeks pre-challenge was compared with 32 and 20 weeks but with a final boost 8 weeks pre-challenge with 2 x 10(6) fixed-inactivated HSC-F4 cells infected with SIVmac32H. Controls received rMVA vectors expressing an irrelevant transgene or were naïve challenge controls. All received 10 MID(50) SIVmac32H/J5 intravenously. RESULTS Vaccinates immunized with rMVA-J5 exhibited significant, albeit transient, control of peak primary viraemia despite inconsistent and variable immune responses elicted by vaccination. Humoral and cellular responses to Env were most consistent, with lower responses to Nef, Rev and Tat. Increasing titres of anti-vaccinia neutralizing antibodies reflected the number and dose of rMVA inoculations. CONCLUSIONS Improved combinations of viral vectors are required to elicit appropriate immune responses to control viral replication.
Collapse
Affiliation(s)
- N Berry
- Division of Retrovirology, National Institute for Biological Standards and Control, South Mimms, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Naik AM, Chalikonda S, McCart JA, Xu H, Guo ZS, Langham G, Gardner D, Mocellin S, Lokshin AE, Moss B, Alexander HR, Bartlett DL. Intravenous and isolated limb perfusion delivery of wild type and a tumor-selective replicating mutant vaccinia virus in nonhuman primates. Hum Gene Ther 2006; 17:31-45. [PMID: 16409123 DOI: 10.1089/hum.2006.17.31] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In this study we examine the safety, feasibility, and biodistribution of a tumor-selective mutant vaccinia (vvDD) and wild-type WR (vF13) vaccinia after delivery via intradermal or intravenous infection or isolated limb perfusion (ILP) in rhesus macaques. By intradermal inoculation, 10(6) PFU of vvDD caused a minimal skin reaction whereas vF13 caused marked erythema and necrosis with a peak indurated area of 108 cm2. By intravenous delivery, vvDD caused no clinical symptoms of viremia and no viral recovery from tissues, serum, saliva, urine, or feces. In contrast, vF13 caused symptoms of lethargy, anorexia, fever, and signs of viremia. Delivery of vF13 via ILP resulted in numerous cutaneous pox lesions localized solely to the perfused limb with high viral recovery in the perfused skin and muscle. ILP with vvDD resulted in no visible pox lesions and no clinical signs or symptoms of viremia. No long-term toxicity was identified after ILP with 10(9) PFU of vvDD, and no virus was recovered from any tissue, serum, saliva, urine, or fecal sample. These results suggest that vvDD appears to be safe in primates, and thus vvDD should be further investigated for clinical trial in human cancer patients.
Collapse
Affiliation(s)
- Arpana M Naik
- Center for Cancer Research, Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|