1
|
Zhang X, Sun Y, Zhang J, Zhang J, Wang J, Hu C, Wang Y, Hu F, Cai S, He Y, Liu Y, Sun Y, Yang S, Jiang D, Yang K. Construction and evaluation of glycoprotein-based nucleic acid vaccines for Marburg virus. Med Microbiol Immunol 2024; 214:1. [PMID: 39607590 DOI: 10.1007/s00430-024-00811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Marburg virus (MARV) is a zoonotic virus that can infect humans and non-human primates (NHPs) and lead to a fatal Marburg hemorrhagic fever (MHF), while there is no approved vaccine or antiviral treatment for MHF. The nucleic acid vaccine has unique advantages, including fast and simple preparation, easy to follow the virus mutation situation, and less adverse reactions. Therefore, we constructed the DNA and mRNA candidate vaccines based on codon-optimized MARV glycoprotein sequence, and evaluated the immune effect in mice through ELISA, ELISpot, and Flow cytometry. After the second booster immunization, both of the candidate vaccines induced strong humoral immune response, enhanced T cell response, and elicited neutralizing antibodies. Notably, DNA candidate vaccine induced stronger humoral immune response, while mRNA candidate vaccine elicited higher levels of IFN-γ and IL-4. In addition, transcriptome analysis revealed that the candidate vaccines activated immune response related pathways. Our study shed new light on the nucleic acid vaccines for MARV and further confirmed the potential of nucleic acid vaccine for future MHF prevention and control.
Collapse
MESH Headings
- Animals
- Marburgvirus/immunology
- Marburgvirus/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Marburg Virus Disease/prevention & control
- Marburg Virus Disease/immunology
- Mice
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Glycoproteins/immunology
- Glycoproteins/genetics
- Female
- Mice, Inbred BALB C
- T-Lymphocytes/immunology
- Immunity, Humoral
- mRNA Vaccines
- Immunization, Secondary
- Interleukin-4/metabolism
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/administration & dosage
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- Interferon-gamma/metabolism
Collapse
Affiliation(s)
- Xiyang Zhang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
- Military Medical Innovation Center, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China
| | - Yubo Sun
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Jiaxing Zhang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Junqi Zhang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Jing Wang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Chenchen Hu
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Yueyue Wang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Feiming Hu
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Sirui Cai
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Yuanli He
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Yang Liu
- Institute of AIDS Prevention and Control, Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, 710054, China
| | - Yuanjie Sun
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Shuya Yang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China
| | - Dongbo Jiang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China.
| | - Kun Yang
- Department of Immunology, The Key Laboratory of Bio-hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (the Fourth Military Medical University), Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
2
|
de La Vega MA, XIII A, Massey CS, Spengler JR, Kobinger GP, Woolsey C. An update on nonhuman primate usage for drug and vaccine evaluation against filoviruses. Expert Opin Drug Discov 2024; 19:1185-1211. [PMID: 39090822 PMCID: PMC11466704 DOI: 10.1080/17460441.2024.2386100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Due to their faithful recapitulation of human disease, nonhuman primates (NHPs) are considered the gold standard for evaluating drugs against Ebolavirus and other filoviruses. The long-term goal is to reduce the reliance on NHPs with more ethical alternatives. In silico simulations and organoid models have the potential to revolutionize drug testing by providing accurate, human-based systems that mimic disease processes and drug responses without the ethical concerns associated with animal testing. However, as these emerging technologies are still in their developmental infancy, NHP models are presently needed for late-stage evaluation of filovirus vaccines and drugs, as they provide critical insights into the efficacy and safety of new medical countermeasures. AREAS COVERED In this review, the authors introduce available NHP models and examine the existing literature on drug discovery for all medically significant filoviruses in corresponding models. EXPERT OPINION A deliberate shift toward animal-free models is desired to align with the 3Rs of animal research. In the short term, the use of NHP models can be refined and reduced by enhancing replicability and publishing negative data. Replacement involves a gradual transition, beginning with the selection and optimization of better small animal models; advancing organoid systems, and using in silico models to accurately predict immunological outcomes.
Collapse
Affiliation(s)
- Marc-Antoine de La Vega
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Ara XIII
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Christopher S. Massey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Jessica R. Spengler
- Viral Special Pathogens Branch and Infectious Diseases
Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for
Disease Control and Prevention, Atlanta, GA
| | - Gary P. Kobinger
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| |
Collapse
|
3
|
Zhang X, Sun Y, Zhang J, Wei H, Wang J, Hu C, Liu Y, Cai S, Yuan Q, Wang Y, Sun Y, Yang S, Jiang D, Yang K. Lysosome-Associated Membrane Protein Targeting Strategy Improved Immunogenicity of Glycoprotein-Based DNA Vaccine for Marburg Virus. Vaccines (Basel) 2024; 12:1013. [PMID: 39340043 PMCID: PMC11436145 DOI: 10.3390/vaccines12091013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Marburg hemorrhagic fever (MHF) is a fatal infectious disease caused by Marburg virus (MARV) infection, and MARV has been identified as a priority pathogen for vaccine development by the WHO. The glycoprotein (GP) of MARV mediates viral adhesion and invasion of host cells and therefore can be used as an effective target for vaccine development. Moreover, DNA vaccines have unique advantages, such as simple construction processes, low production costs, and few adverse reactions, but their immunogenicity may decrease due to the poor absorption rate of plasmids. Lysosome-associated membrane protein 1 (LAMP1) can direct antigens to lysosomes and endosomes and has great potential for improving the immunogenicity of nucleic acid vaccines. Therefore, we constructed a DNA vaccine based on a codon-optimized MARV GP (ID MF939097.1) fused with LAMP1 and explored the effect of a LAMP targeting strategy on improving the immunogenicity of the MARV DNA vaccine. ELISA, ELISpot, and flow cytometry revealed that the introduction of LAMP1 into the MARV DNA candidate vaccine improved the humoral and cellular immune response, enhanced the secretion of cytokines, and established long-term immune protection. Transcriptome analysis revealed that the LAMP targeting strategy significantly enriched antigen processing and presentation-related pathways, especially the MHC class II-related pathway, in the candidate vaccine. Our study broadens the strategic vision for enhanced DNA vaccine design and provides a promising candidate vaccine for MHF prevention.
Collapse
Affiliation(s)
- Xiyang Zhang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
- Military Medical Innovation Center, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Yubo Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Junqi Zhang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Hengzheng Wei
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Jing Wang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Chenchen Hu
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Yang Liu
- Institute of AIDS Prevention and Control, Shaanxi Provincial Center for Disease Control and Prevention, Xi'an 710054, China
| | - Sirui Cai
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Qinghong Yuan
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Yueyue Wang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Yuanjie Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Shuya Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Dongbo Jiang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| | - Kun Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (The Fourth Military Medical University), Xi'an 710032, China
| |
Collapse
|
4
|
Han L, Song S, Feng H, Ma J, Wei W, Si F. A roadmap for developing Venezuelan equine encephalitis virus (VEEV) vaccines: Lessons from the past, strategies for the future. Int J Biol Macromol 2023:125514. [PMID: 37353130 DOI: 10.1016/j.ijbiomac.2023.125514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Venezuelan equine encephalitis (VEE) is a zoonotic infectious disease caused by the Venezuelan equine encephalitis virus (VEEV), which can lead to severe central nervous system infections in both humans and animals. At present, the medical community does not possess a viable means of addressing VEE, rendering the prevention of the virus a matter of paramount importance. Regarding the prevention and control of VEEV, the implementation of a vaccination program has been recognized as the most efficient strategy. Nevertheless, there are currently no licensed vaccines or drugs available for human use against VEEV. This imperative has led to a surge of interest in vaccine research, with VEEV being a prime focus for researchers in the field. In this paper, we initially present a comprehensive overview of the current taxonomic classification of VEEV and the cellular infection mechanism of the virus. Subsequently, we provide a detailed introduction of the prominent VEEV vaccine types presently available, including inactivated vaccines, live attenuated vaccines, genetic, and virus-like particle vaccines. Moreover, we emphasize the challenges that current VEEV vaccine development faces and suggest urgent measures that must be taken to overcome these obstacles. Notably, based on our latest research, we propose the feasibility of incorporation codon usage bias strategies to create the novel VEEV vaccine. Finally, we prose several areas that future VEEV vaccine development should focus on. Our objective is to encourage collaboration between the medical and veterinary communities, expedite the translation of existing vaccines from laboratory to clinical applications, while also preparing for future outbreaks of new VEEV variants.
Collapse
Affiliation(s)
- Lulu Han
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China; Huaihe Hospital of Henan University, Clinical Medical College of Henan University, Kai Feng 475000, China
| | - Shuai Song
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, PR China
| | - Huilin Feng
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences of Henan University, Kai Feng 475000, China
| | - Jing Ma
- Huaihe Hospital of Henan University, Clinical Medical College of Henan University, Kai Feng 475000, China
| | - Wenqiang Wei
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences of Henan University, Kai Feng 475000, China.
| | - Fusheng Si
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China.
| |
Collapse
|
5
|
Zhang HQ, Zhang QY, Yuan ZM, Zhang B. The potential epidemic threat of Ebola virus and the development of a preventive vaccine. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:67-78. [DOI: 10.1016/j.jobb.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
6
|
History and impact of the mouse-adapted Ebola virus model. Antiviral Res 2023; 210:105493. [PMID: 36567023 DOI: 10.1016/j.antiviral.2022.105493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Ebola virus (EBOV) is a member of the filoviridae family, which are comprised of negative sense, enveloped RNA hemorrhagic fever viruses that can cause severe disease and high lethality rates. These viruses require BSL-4 containment laboratories for study. Early studies of EBOV pathogenesis relied heavily on the use of nonhuman primates, which are expensive and cumbersome to handle in large numbers. Guinea pig models were also developed, but even to this day limited reagents are available in this model. In 1998, Mike Bray and colleagues developed a mouse-adapted EBOV (maEBOV) that caused lethality in adult immunocompetent mice. This model had significant advantages, including being inexpensive, allowing for higher animal numbers for statistical analysis, availability of reagents for studying pathogenesis, and availability of a vast array of genetically modified strains. The model has been used to test vaccines, therapeutic drugs, EBOV mutants, and pathogenesis, and its importance is demonstrated by the hundreds of citations referencing the original publication. This review will cover the history of the maEBOV model and its use in filovirus research.
Collapse
|
7
|
Hunegnaw R, Honko AN, Wang L, Carr D, Murray T, Shi W, Nguyen L, Storm N, Dulan CNM, Foulds KE, Agans KN, Cross RW, Geisbert JB, Cheng C, Ploquin A, Stanley DA, Geisbert TW, Nabel GJ, Sullivan NJ. A single-shot ChAd3-MARV vaccine confers rapid and durable protection against Marburg virus in nonhuman primates. Sci Transl Med 2022; 14:eabq6364. [PMID: 36516269 DOI: 10.1126/scitranslmed.abq6364] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Marburg virus (MARV) causes a severe hemorrhagic fever disease in primates with mortality rates in humans of up to 90%. MARV has been identified as a category A bioterrorism agent by the Centers for Disease Control and Prevention (CDC) and priority pathogen A by the National Institute of Allergy and Infectious Diseases (NIAID), needing urgent research and development of countermeasures because of the high public health risk it poses. The recent cases of MARV in West Africa underscore the substantial outbreak potential of this virus. The potential for cross-border spread, as had occurred during the 2014-2016 Ebola virus outbreak, illustrates the critical need for MARV vaccines. To support regulatory approval of the chimpanzee adenovirus 3 (ChAd3)-MARV vaccine that has completed phase 1 trials, we showed that the nonreplicating ChAd3 vector, which has a demonstrated safety profile in humans, protected against a uniformly lethal challenge with MARV/Ang. Protective immunity was achieved within 7 days of vaccination and was maintained through 1 year after vaccination. Antigen-specific antibodies were an immune correlate of protection in the acute challenge model, and their concentration was predictive of protection. These results demonstrate that a single-shot ChAd3-MARV vaccine generated a protective immune response that was both rapid and durable with an immune correlate of protection that will support advanced clinical development.
Collapse
Affiliation(s)
- Ruth Hunegnaw
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anna N Honko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Derick Carr
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Tamar Murray
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Wei Shi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Lam Nguyen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nadia Storm
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Caitlyn N M Dulan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Krystle N Agans
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Joan B Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cheng Cheng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Aurélie Ploquin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Daphne A Stanley
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gary J Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Abir MH, Rahman T, Das A, Etu SN, Nafiz IH, Rakib A, Mitra S, Emran TB, Dhama K, Islam A, Siyadatpanah A, Mahmud S, Kim B, Hassan MM. Pathogenicity and virulence of Marburg virus. Virulence 2022; 13:609-633. [PMID: 35363588 PMCID: PMC8986239 DOI: 10.1080/21505594.2022.2054760] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/13/2022] [Indexed: 12/25/2022] Open
Abstract
Marburg virus (MARV) has been a major concern since 1967, with two major outbreaks occurring in 1998 and 2004. Infection from MARV results in severe hemorrhagic fever, causing organ dysfunction and death. Exposure to fruit bats in caves and mines, and human-to-human transmission had major roles in the amplification of MARV outbreaks in African countries. The high fatality rate of up to 90% demands the broad study of MARV diseases (MVD) that correspond with MARV infection. Since large outbreaks are rare for MARV, clinical investigations are often inadequate for providing the substantial data necessary to determine the treatment of MARV disease. Therefore, an overall review may contribute to minimizing the limitations associated with future medical research and improve the clinical management of MVD. In this review, we sought to analyze and amalgamate significant information regarding MARV disease epidemics, pathophysiology, and management approaches to provide a better understanding of this deadly virus and the associated infection.
Collapse
Affiliation(s)
- Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Silvia Naznin Etu
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ahmed Rakib
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Ariful Islam
- EcoHealth Alliance, New York, NY, USA
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Victoria, Australia
| | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand, Iran
| | - Shafi Mahmud
- Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Bonlgee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Mohammad Mahmudul Hassan
- Queensland Alliance for One Health Sciences, School of Veterinary Sciences, The University of Queensland, Gatton, Australia
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| |
Collapse
|
9
|
A DNA vaccine targeting VEE virus delivered by needle-free jet-injection protects macaques against aerosol challenge. NPJ Vaccines 2022; 7:46. [PMID: 35459271 PMCID: PMC9033795 DOI: 10.1038/s41541-022-00469-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/18/2022] [Indexed: 11/09/2022] Open
Abstract
We have previously shown that DNA vaccines expressing codon optimized alphavirus envelope glycoprotein genes protect both mice and nonhuman primates from viral challenge when delivered by particle-mediated epidermal delivery (PMED) or intramuscular (IM) electroporation (EP). Another technology with fewer logistical drawbacks is disposable syringe jet injection (DSJI) devices developed by PharmaJet, Inc. These needle-free jet injection systems are spring-powered and capable of delivering vaccines either IM or into the dermis (ID). Here, we evaluated the immunogenicity of our Venezuelan equine encephalitis virus (VEEV) DNA vaccine delivered by either the IM- or ID-DSJI devices in nonhuman primates. The protective efficacy was assessed following aerosol challenge. We found that a prime and single boost by either the IM or ID route resulted in humoral and cellular immune responses that provided significant protection against disease and viremia. Although the ID route utilized one-fifth the DNA dose used in the IM route of vaccination, and the measured humoral and cellular immune responses trended lower, the level of protection was high and performed as well as the IM route for several clinical endpoints.
Collapse
|
10
|
Affiliation(s)
- Fang Zhao
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yun He
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hongzhou Lu
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Torres-Ruesta A, Chee RSL, Ng LF. Insights into Antibody-Mediated Alphavirus Immunity and Vaccine Development Landscape. Microorganisms 2021; 9:microorganisms9050899. [PMID: 33922370 PMCID: PMC8145166 DOI: 10.3390/microorganisms9050899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022] Open
Abstract
Alphaviruses are mosquito-borne pathogens distributed worldwide in tropical and temperate areas causing a wide range of symptoms ranging from inflammatory arthritis-like manifestations to the induction of encephalitis in humans. Historically, large outbreaks in susceptible populations have been recorded followed by the development of protective long-lasting antibody responses suggesting a potential advantageous role for a vaccine. Although the current understanding of alphavirus antibody-mediated immunity has been mainly gathered in natural and experimental settings of chikungunya virus (CHIKV) infection, little is known about the humoral responses triggered by other emerging alphaviruses. This knowledge is needed to improve serology-based diagnostic tests and the development of highly effective cross-protective vaccines. Here, we review the role of antibody-mediated immunity upon arthritogenic and neurotropic alphavirus infections, and the current research efforts for the development of vaccines as a tool to control future alphavirus outbreaks.
Collapse
Affiliation(s)
- Anthony Torres-Ruesta
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.T.-R.); (R.S.-L.C.)
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Rhonda Sin-Ling Chee
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.T.-R.); (R.S.-L.C.)
| | - Lisa F.P. Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.T.-R.); (R.S.-L.C.)
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Correspondence: ; Tel.: +65-6407-0028
| |
Collapse
|
12
|
Kortepeter MG, Dierberg K, Shenoy ES, Cieslak TJ. Marburg virus disease: A summary for clinicians. Int J Infect Dis 2020; 99:233-242. [PMID: 32758690 PMCID: PMC7397931 DOI: 10.1016/j.ijid.2020.07.042] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/09/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES This article summarizes the countermeasures for Marburg virus disease, focusing on pathogenesis, clinical features and diagnostics. There is an emphasis on therapies and vaccines that have demonstrated, through their evaluation in nonhuman primates (NHPs) and/or in humans, potential for use in an emergency situation. METHODS A standardized literature review was conducted on vaccines and treatments for Marburg virus disease, with a focus on human and nonhuman primate data published in the last five years. More detail on the methods that were used is summarized in a companion methods paper. RESULTS The study identified six treatments and four vaccine platforms that have demonstrated, through their efficacy in NHPs, potential benefit for treating or preventing infection in humans. CONCLUSION Succinct summaries of Marburg countermeasures are provided to give the busy clinician a head start in reviewing the literature if faced with a patient with Marburg virus disease. Links to other authoritative sources of information are also provided.
Collapse
|
13
|
Stromberg ZR, Fischer W, Bradfute SB, Kubicek-Sutherland JZ, Hraber P. Vaccine Advances against Venezuelan, Eastern, and Western Equine Encephalitis Viruses. Vaccines (Basel) 2020; 8:vaccines8020273. [PMID: 32503232 PMCID: PMC7350001 DOI: 10.3390/vaccines8020273] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 01/21/2023] Open
Abstract
Vaccinations are a crucial intervention in combating infectious diseases. The three neurotropic Alphaviruses, Eastern (EEEV), Venezuelan (VEEV), and Western (WEEV) equine encephalitis viruses, are pathogens of interest for animal health, public health, and biological defense. In both equines and humans, these viruses can cause febrile illness that may progress to encephalitis. Currently, there are no licensed treatments or vaccines available for these viruses in humans. Experimental vaccines have shown variable efficacy and may cause severe adverse effects. Here, we outline recent strategies used to generate vaccines against EEEV, VEEV, and WEEV with an emphasis on virus-vectored and plasmid DNA delivery. Despite candidate vaccines protecting against one of the three viruses, few studies have demonstrated an effective trivalent vaccine. We evaluated the potential of published vaccines to generate cross-reactive protective responses by comparing DNA vaccine sequences to a set of EEEV, VEEV, and WEEV genomes and determining the vaccine coverages of potential epitopes. Finally, we discuss future directions in the development of vaccines to combat EEEV, VEEV, and WEEV.
Collapse
Affiliation(s)
- Zachary R. Stromberg
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM 505, USA; (Z.R.S.); (J.Z.K.-S.)
| | - Will Fischer
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 505, USA;
| | - Steven B. Bradfute
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 505, USA;
| | - Jessica Z. Kubicek-Sutherland
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM 505, USA; (Z.R.S.); (J.Z.K.-S.)
| | - Peter Hraber
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 505, USA;
- Correspondence:
| |
Collapse
|
14
|
Suschak JJ, Schmaljohn CS. Vaccines against Ebola virus and Marburg virus: recent advances and promising candidates. Hum Vaccin Immunother 2019; 15:2359-2377. [PMID: 31589088 DOI: 10.1080/21645515.2019.1651140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The filoviruses Ebola virus and Marburg virus are among the most dangerous pathogens in the world. Both viruses cause viral hemorrhagic fever, with case fatality rates of up to 90%. Historically, filovirus outbreaks had been relatively small, with only a few hundred cases reported. However, the recent West African Ebola virus outbreak underscored the threat that filoviruses pose. The three year-long outbreak resulted in 28,646 Ebola virus infections and 11,323 deaths. The lack of Food and Drug Administration (FDA) licensed vaccines and antiviral drugs hindered early efforts to contain the outbreak. In response, the global scientific community has spurred the advanced development of many filovirus vaccine candidates. Novel vaccine platforms, such as viral vectors and DNA vaccines, have emerged, leading to the investigation of candidate vaccines that have demonstrated protective efficacy in small animal and nonhuman primate studies. Here, we will discuss several of these vaccine platforms with a particular focus on approaches that have advanced into clinical development.
Collapse
Affiliation(s)
- John J Suschak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Connie S Schmaljohn
- Headquarters Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
15
|
Sharma A, Knollmann-Ritschel B. Current Understanding of the Molecular Basis of Venezuelan Equine Encephalitis Virus Pathogenesis and Vaccine Development. Viruses 2019; 11:v11020164. [PMID: 30781656 PMCID: PMC6410161 DOI: 10.3390/v11020164] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/30/2019] [Accepted: 02/07/2019] [Indexed: 12/30/2022] Open
Abstract
Dedication This review is dedicated in the memory of Dr Radha K. Maheshwari, a great mentor and colleague, whose passion for research and student training has left a lasting effect on this manuscript and many other works. Abstract Venezuelan equine encephalitis virus (VEEV) is an alphavirus in the family Togaviridae. VEEV is highly infectious in aerosol form and a known bio-warfare agent that can cause severe encephalitis in humans. Periodic outbreaks of VEEV occur predominantly in Central and South America. Increased interest in VEEV has resulted in a more thorough understanding of the pathogenesis of this disease. Inflammation plays a paradoxical role of antiviral response as well as development of lethal encephalitis through an interplay between the host and viral factors that dictate virus replication. VEEV has efficient replication machinery that adapts to overcome deleterious mutations in the viral genome or improve interactions with host factors. In the last few decades there has been ongoing development of various VEEV vaccine candidates addressing the shortcomings of the current investigational new drugs or approved vaccines. We review the current understanding of the molecular basis of VEEV pathogenesis and discuss various types of vaccine candidates.
Collapse
Affiliation(s)
- Anuj Sharma
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | |
Collapse
|
16
|
Self-Amplifying RNA Vaccines for Venezuelan Equine Encephalitis Virus Induce Robust Protective Immunogenicity in Mice. Mol Ther 2019; 27:850-865. [PMID: 30770173 DOI: 10.1016/j.ymthe.2018.12.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/18/2018] [Accepted: 12/27/2018] [Indexed: 01/21/2023] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is a known biological defense threat. A live-attenuated investigational vaccine, TC-83, is available, but it has a high non-response rate and can also cause severe reactogenicity. We generated two novel VEE vaccine candidates using self-amplifying mRNA (SAM). LAV-CNE is a live-attenuated VEE SAM vaccine formulated with synthetic cationic nanoemulsion (CNE) and carrying the RNA genome of TC-83. IAV-CNE is an irreversibly-attenuated VEE SAM vaccine formulated with CNE, delivering a TC-83 genome lacking the capsid gene. LAV-CNE launches a TC-83 infection cycle in vaccinated subjects but eliminates the need for live-attenuated vaccine production and potentially reduces manufacturing time and complexity. IAV-CNE produces a single cycle of RNA amplification and antigen expression without generating infectious viruses in subjects, thereby creating a potentially safer alternative to live-attenuated vaccine. Here, we demonstrated that mice vaccinated with LAV-CNE elicited immune responses similar to those of TC-83, providing 100% protection against aerosol VEEV challenge. IAV-CNE was also immunogenic, resulting in significant protection against VEEV challenge. These studies demonstrate the proof of concept for using the SAM platform to streamline the development of effective attenuated vaccines against VEEV and closely related alphavirus pathogens such as western and eastern equine encephalitis and Chikungunya viruses.
Collapse
|
17
|
Suschak JJ, Bagley K, Six C, Shoemaker CJ, Kwilas S, Spik KW, Dupuy LC, Schmaljohn CS. The genetic adjuvant IL-12 enhances the protective efficacy of a DNA vaccine for Venezuelan equine encephalitis virus delivered by intramuscular injection in mice. Antiviral Res 2018; 159:113-121. [PMID: 30268913 DOI: 10.1016/j.antiviral.2018.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 01/01/2023]
Abstract
We have previously shown that DNA vaccines expressing codon-optimized alphavirus envelope glycoprotein genes protect both mice and non-human primates from viral challenge when delivered by intramuscular electroporation (IM-EP). To determine if we could achieve equivalent immunogenicity and protective efficacy in the absence of electroporation, we co-delivered our Venezuelan equine encephalitis virus (VEEV) DNA vaccine with DNA plasmids expressing genetic adjuvants designed to augment immune responses. We tested the Th1-inducing cytokine IL-12 as well as the granulocyte growth factor GM-CSF, both of which have demonstrated significant adjuvant effect when included in clinical DNA vaccine formulations. Additionally, as multiple reports have described the necessity of IFN-αβ in DNA vaccine immunogenicity, we tested vaccine plasmids encoding a potent stimulator of the IFN-αβ pathway. Our data suggest that IM vaccination of mice with plasmid DNA encoding genetic adjuvants enhances VEEV vaccine immunogenicity, resulting in improved T cell responses, as well as skewing of the anti-VEEV IgG antibody isotype. Additionally, IM vaccination of VEEV DNA vaccine and IL-12 provided complete protection against aerosol VEEV challenge. Overall, our data suggest that co-delivery of genetic adjuvants with alphavirus DNA vaccines using IM delivery can influence the type of immune response obtained and provide comparable protective immunity to that achieved by IM-EP delivery of the vaccine without adjuvants.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Encephalitis Virus, Venezuelan Equine
- Encephalomyelitis, Venezuelan Equine/immunology
- Encephalomyelitis, Venezuelan Equine/prevention & control
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Immunogenicity, Vaccine
- Injections, Intramuscular
- Interleukin-12/genetics
- Interleukin-12/immunology
- Mice
- Mice, Inbred BALB C
- Vaccines, DNA/immunology
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- John J Suschak
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | - Carolyn Six
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Charles J Shoemaker
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Steven Kwilas
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Kristin W Spik
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Lesley C Dupuy
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Connie S Schmaljohn
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| |
Collapse
|
18
|
Abstract
The West African Ebola virus (EBOV) epidemic has fast-tracked countermeasures for this rare, emerging zoonotic pathogen. Until 2013-2014, most EBOV vaccine candidates were stalled between the preclinical and clinical milestones on the path to licensure, because of funding problems, lack of interest from pharmaceutical companies, and competing priorities in public health. The unprecedented and devastating epidemic propelled vaccine candidates toward clinical trials that were initiated near the end of the active response to the outbreak. Those trials did not have a major impact on the epidemic but provided invaluable data on vaccine safety, immunogenicity, and, to a limited degree, even efficacy in humans. There are plenty of lessons to learn from these trials, some of which are addressed in this review. Better preparation is essential to executing an effective response to EBOV in the future; yet, the first indications of waning interest are already noticeable.
Collapse
Affiliation(s)
- Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba 93E 0J9, Canada
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA
| | - Andrea Marzi
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| |
Collapse
|
19
|
A Multiagent Alphavirus DNA Vaccine Delivered by Intramuscular Electroporation Elicits Robust and Durable Virus-Specific Immune Responses in Mice and Rabbits and Completely Protects Mice against Lethal Venezuelan, Western, and Eastern Equine Encephalitis Virus Aerosol Challenges. J Immunol Res 2018; 2018:8521060. [PMID: 29967804 PMCID: PMC6008678 DOI: 10.1155/2018/8521060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/26/2018] [Indexed: 11/17/2022] Open
Abstract
There remains a need for vaccines that can safely and effectively protect against the biological threat agents Venezuelan (VEEV), western (WEEV), and eastern (EEEV) equine encephalitis virus. Previously, we demonstrated that a VEEV DNA vaccine that was optimized for increased antigen expression and delivered by intramuscular (IM) electroporation (EP) elicited robust and durable virus-specific antibody responses in multiple animal species and provided complete protection against VEEV aerosol challenge in mice and nonhuman primates. Here, we performed a comparative evaluation of the immunogenicity and protective efficacy of individual optimized VEEV, WEEV, and EEEV DNA vaccines with that of a 1 : 1 : 1 mixture of these vaccines, which we have termed the 3-EEV DNA vaccine, when delivered by IM EP. The individual DNA vaccines and the 3-EEV DNA vaccine elicited robust and durable virus-specific antibody responses in mice and rabbits and completely protected mice from homologous VEEV, WEEV, and EEEV aerosol challenges. Taken together, the results from these studies demonstrate that the individual VEEV, WEEV, and EEEV DNA vaccines and the 3-EEV DNA vaccine delivered by IM EP provide an effective means of eliciting protection against lethal encephalitic alphavirus infections in a murine model and represent viable next-generation vaccine candidates that warrant further development.
Collapse
|
20
|
|
21
|
Abstract
The filoviruses, Ebola virus (EBOV), and Marburg virus (MARV), are among the most pathogenic viruses known to man and the causative agents of viral hemorrhagic fever outbreaks in Africa with case fatality rates of up to 90%. Nearly 30,000 infections were observed in the latest EBOV epidemic in West Africa; previous outbreaks were much smaller, typically only affecting less than a few hundred people. Compared to other diseases such as AIDS or Malaria with millions of cases annually, filovirus hemorrhagic fever (FHF) is one of the neglected infectious diseases. There are no licensed vaccines or therapeutics available to treat EBOV and MARV infections; therefore, these pathogens can only be handled in maximum containment laboratories and are classified as select agents. Under these limitations, a very few laboratories worldwide conducted basic research and countermeasure development for EBOV and MARV since their respective discoveries in 1967 (MARV) and 1976 (EBOV). In this review, we discuss several vaccine platforms against EBOV and MARV, which have been assessed for their protective efficacy in animal models of FHF. The focus is on the most promising approaches, which were accelerated in clinical development (phase I-III trials) during the EBOV epidemic in West Africa.
Collapse
Affiliation(s)
- Pierce Reynolds
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
22
|
Bounds CE, Terry FE, Moise L, Hannaman D, Martin WD, De Groot AS, Suschak JJ, Dupuy LC, Schmaljohn CS. An immunoinformatics-derived DNA vaccine encoding human class II T cell epitopes of Ebola virus, Sudan virus, and Venezuelan equine encephalitis virus is immunogenic in HLA transgenic mice. Hum Vaccin Immunother 2017; 13:2824-2836. [PMID: 28575582 PMCID: PMC5718811 DOI: 10.1080/21645515.2017.1329788] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Immunoinformatics tools were used to predict human leukocyte antigen (HLA) class II-restricted T cell epitopes within the envelope glycoproteins and nucleocapsid proteins of Ebola virus (EBOV) and Sudan virus (SUDV) and the structural proteins of Venezuelan equine encephalitis virus (VEEV). Selected epitopes were tested for binding to soluble HLA molecules representing 5 class II alleles (DRB1*0101, DRB1*0301, DRB1*0401, DRB1*0701, and DRB1*1501). All but one of the 25 tested peptides bound to at least one of the DRB1 alleles, and 4 of the peptides bound at least moderately or weakly to all 5 DRB1 alleles. Additional algorithms were used to design a single "string-of-beads" expression construct with 44 selected epitopes arranged to avoid creation of spurious junctional epitopes. Seventeen of these 44 predicted epitopes were conserved between the major histocompatibility complex (MHC) of humans and mice, allowing initial testing in mice. BALB/c mice vaccinated with the multi-epitope construct developed statistically significant cellular immune responses to EBOV, SUDV, and VEEV peptides as measured by interferon (IFN)-γ ELISpot assays. Significant levels of antibodies to VEEV, but not EBOV, were also detected in vaccinated BALB/c mice. To assess immunogenicity in the context of a human MHC, HLA-DR3 transgenic mice were vaccinated with the multi-epitope construct and boosted with a mixture of the 25 peptides used in the binding assays. The vaccinated HLA-DR3 mice developed significant cellular immune responses to 4 of the 25 (16%) tested individual class II peptides as measured by IFN-γ ELISpot assays. In addition, these mice developed antibodies against EBOV and VEEV as measured by ELISA. While a low but significant level of protection was observed in vaccinated transgenic mice after aerosol exposure to VEEV, no protection was observed after intraperitoneal challenge with mouse-adapted EBOV. These studies provide proof of concept for the use of an informatics approach to design a multi-agent, multi-epitope immunogen and provide a basis for further testing aimed at focusing immune responses toward desired protective T cell epitopes.
Collapse
Affiliation(s)
- Callie E Bounds
- a United States Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | | | - Leonard Moise
- b EpiVax, Inc. , Providence , RI , USA.,c Institute for Immunology and Informatics , University of Rhode Island , Providence , RI , USA
| | - Drew Hannaman
- d Ichor Medical Systems, Inc. , San Diego , CA , USA
| | | | - Anne S De Groot
- b EpiVax, Inc. , Providence , RI , USA.,c Institute for Immunology and Informatics , University of Rhode Island , Providence , RI , USA
| | - John J Suschak
- a United States Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Lesley C Dupuy
- a United States Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Connie S Schmaljohn
- a United States Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
23
|
Sharma R, Jangid K, Anuradha. Ebola Vaccine: How Far are we? J Clin Diagn Res 2017; 11:DE01-DE04. [PMID: 28658761 PMCID: PMC5483663 DOI: 10.7860/jcdr/2017/22184.9863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 02/05/2017] [Indexed: 11/24/2022]
Abstract
Ebola viruses have been identified as an emerging threat as it causes severe haemorrhagic fever in human with mortality rates ranging from 50 to 90%. In addition to being a global health concern, the virus also is considered a potential biological threat agent. As for now, no licensed vaccine is available for pre or post exposure treatment. Recent epidemic of this disease in South Africa has led to concern towards development of an effective vaccine on a priority basis. This review is an attempt to look upon current progress in the development of Ebola virus vaccines and highlights strategies that have the greatest potential for commercial development.
Collapse
Affiliation(s)
- Rajani Sharma
- Senior Resident, Department of Microbiology, PGIMER and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Ketki Jangid
- Senior Resident, Department of Microbiology, PGIMER and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Anuradha
- Assistant Professor, Department of Microbiology, PGIMER and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| |
Collapse
|
24
|
Abstract
For 40 years ebolaviruses have been responsible for sporadic outbreaks of severe and often fatal hemorrhagic fever in humans and nonhuman primates. In December 2013 an unprecedented Zaire ebolavirus epidemic began in West Africa. Although "patient zero" has finally been reached after 2 years, the virus is again causing disease in the region. Currently there are no licensed vaccines or therapeutic countermeasures against ebolaviruses; however, the epidemic in West Africa has focused attention on the potential vaccine platforms developed over the past 15 years. There has been remarkable progress using a variety of platforms including DNA, subunit, and several viral vector approaches, replicating and non-replicating, which have shown varying degrees of protective efficacy in the "gold-standard" nonhuman primate models for Ebolavirus infections. A number of these vaccine platforms have moved into clinical trials over the past year with the hope of finding an efficacious vaccine to prevent future outbreaks/epidemics of Ebola hemorrhagic fever on the scale of the West African epidemic.
Collapse
Affiliation(s)
- Chad E Mire
- a Galveston National Laboratory, and Department of Microbiology and Immunology , University of Texas Medical Branch , Galveston , TX , USA
| | - Thomas W Geisbert
- a Galveston National Laboratory, and Department of Microbiology and Immunology , University of Texas Medical Branch , Galveston , TX , USA
| | - Heinz Feldmann
- b Laboratory of Virology, Division of Intramural Research , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton , MT , USA
| | - Andrea Marzi
- b Laboratory of Virology, Division of Intramural Research , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton , MT , USA
| |
Collapse
|
25
|
Abstract
Long-term control of viral outbreaks requires the use of vaccines to impart acquired resistance and ensuing protection. In the wake of an epidemic, established immunity against a particular disease can limit spread and significantly decrease mortality. Creation of a safe and efficacious vaccine against Ebola virus (EBOV) has proven elusive so far, but various inventive strategies are now being employed to counteract the threat of outbreaks caused by EBOV and related filoviruses. Here, we present a current overview of progress in the field of Ebola virus vaccine development.
Collapse
Affiliation(s)
- Rohan Keshwara
- Department of Microbiology and Immunology, Sidney Kimmel Medical College,Thomas Jefferson University, Philadelphia, Pennsylvania 19107;
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College,Thomas Jefferson University, Philadelphia, Pennsylvania 19107;
| |
Collapse
|
26
|
Burk R, Bollinger L, Johnson JC, Wada J, Radoshitzky SR, Palacios G, Bavari S, Jahrling PB, Kuhn JH. Neglected filoviruses. FEMS Microbiol Rev 2016; 40:494-519. [PMID: 27268907 PMCID: PMC4931228 DOI: 10.1093/femsre/fuw010] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/06/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022] Open
Abstract
Eight viruses are currently assigned to the family Filoviridae Marburg virus, Sudan virus and, in particular, Ebola virus have received the most attention both by researchers and the public from 1967 to 2013. During this period, natural human filovirus disease outbreaks occurred sporadically in Equatorial Africa and, despite high case-fatality rates, never included more than several dozen to a few hundred infections per outbreak. Research emphasis shifted almost exclusively to Ebola virus in 2014, when this virus was identified as the cause of an outbreak that has thus far involved more than 28 646 people and caused more than 11 323 deaths in Western Africa. Consequently, major efforts are currently underway to develop licensed medical countermeasures against Ebola virus infection. However, the ecology of and mechanisms behind Ebola virus emergence are as little understood as they are for all other filoviruses. Consequently, the possibility of the future occurrence of a large disease outbreak caused by other less characterized filoviruses (i.e. Bundibugyo virus, Lloviu virus, Ravn virus, Reston virus and Taï Forest virus) is impossible to rule out. Yet, for many of these viruses, not even rudimentary research tools are available, let alone medical countermeasures. This review summarizes the current knowledge on these less well-characterized filoviruses.
Collapse
Affiliation(s)
- Robin Burk
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
- Department of Infectious Diseases, Virology, University of Heidelberg, 69120 Heidelberg, Baden-Württemberg, Germany
| | - Laura Bollinger
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Joshua C. Johnson
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Jiro Wada
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Sheli R. Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Gustavo Palacios
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Peter B. Jahrling
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| |
Collapse
|
27
|
Grant-Klein RJ, Altamura LA, Badger CV, Bounds CE, Van Deusen NM, Kwilas SA, Vu HA, Warfield KL, Hooper JW, Hannaman D, Dupuy LC, Schmaljohn CS. Codon-optimized filovirus DNA vaccines delivered by intramuscular electroporation protect cynomolgus macaques from lethal Ebola and Marburg virus challenges. Hum Vaccin Immunother 2016; 11:1991-2004. [PMID: 25996997 DOI: 10.1080/21645515.2015.1039757] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cynomolgus macaques were vaccinated by intramuscular electroporation with DNA plasmids expressing codon-optimized glycoprotein (GP) genes of Ebola virus (EBOV) or Marburg virus (MARV) or a combination of codon-optimized GP DNA vaccines for EBOV, MARV, Sudan virus and Ravn virus. When measured by ELISA, the individual vaccines elicited slightly higher IgG responses to EBOV or MARV than did the combination vaccines. No significant differences in immune responses of macaques given the individual or combination vaccines were measured by pseudovirion neutralization or IFN-γ ELISpot assays. Both the MARV and mixed vaccines were able to protect macaques from lethal MARV challenge (5/6 vs. 6/6). In contrast, a greater proportion of macaques vaccinated with the EBOV vaccine survived lethal EBOV challenge in comparison to those that received the mixed vaccine (5/6 vs. 1/6). EBOV challenge survivors had significantly higher pre-challenge neutralizing antibody titers than those that succumbed.
Collapse
Affiliation(s)
- Rebecca J Grant-Klein
- a United States Army Medical Research Institute of Infectious Diseases ; Fort Detrick , MD USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Fernando L, Qiu X, Melito PL, Williams KJN, Feldmann F, Feldmann H, Jones SM, Alimonti JB. Immune Response to Marburg Virus Angola Infection in Nonhuman Primates. J Infect Dis 2015; 212 Suppl 2:S234-41. [DOI: 10.1093/infdis/jiv095] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
29
|
Hartnett JN, Boisen ML, Oottamasathien D, Jones AB, Millett MM, Nelson DS, Muncy IJ, Goba A, Momoh M, Fullah M, Mire CE, Geisbert JB, Geisbert TW, Holton DL, Rouelle JA, Kannadka CB, Reyna AA, Moses LM, Khan SH, Gevao SM, Grant DS, Robinson JE, Happi C, Pitts KR, Garry RF, Branco LM. Current and emerging strategies for the diagnosis, prevention and treatment of Lassa fever. Future Virol 2015. [DOI: 10.2217/fvl.15.41] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ABSTRACT Lassa fever (LF) is a potentially fatal disease that affects an estimated 300,000–500,000 people in endemic areas of west Africa each year. Though past studies have identified fatality rates of 5–20% in patients suspected to have contracted Lassa virus (LASV), new studies using more precise clinical diagnoses and modern diagnostic assays show fatalities rates above 60% in acutely ill patients from endemic regions. Currently, there are no approved vaccines or therapeutics, and only one Comformité Européenne (CE) marked rapid immunodiagnostic for acute LASV infection. Therefore, preventing LASV transmission is the primary goal in endemic regions. Development of rapid immunodiagnostics and research into the efficacy of current treatment options continues toward saving lives in west Africa as well as creating a line of defense against the nefarious use of LASV in bioterrorism settings.
Collapse
Affiliation(s)
- Jessica N Hartnett
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Matthew L Boisen
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
- Corgenix Medical Corporation, Broomfield, CO 80020, USA
| | | | | | | | | | - Ivana J Muncy
- Corgenix Medical Corporation, Broomfield, CO 80020, USA
| | | | - Mambu Momoh
- Kenema Government Hospital, Kenema, Sierra Leone
- Eastern Polytechnic College, Kenema, Sierra Leone
| | | | - Chad E Mire
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Debra L Holton
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Julie A Rouelle
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Chandrika B Kannadka
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Ashley A Reyna
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Lina M Moses
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | | | - Sahr M Gevao
- Ministry of Health and Sanitation, Freetown, Sierra Leone
- University of Sierra Leone, Freetown, Sierra Leone
| | - Donald S Grant
- Kenema Government Hospital, Kenema, Sierra Leone
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - James E Robinson
- Department of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | - Kelly R Pitts
- Corgenix Medical Corporation, Broomfield, CO 80020, USA
| | - Robert F Garry
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
- Zalgen Labs, LLC, Germantown, MD 20876, USA
| | | | | |
Collapse
|
30
|
Ye L, Yang C. Development of vaccines for prevention of Ebola virus infection. Microbes Infect 2015; 17:98-108. [DOI: 10.1016/j.micinf.2014.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 01/25/2023]
|
31
|
Eliciting specific humoral immunity from a plasmid DNA encoding infectious bursal disease virus polyprotein gene fused with avian influenza virus hemagglutinin gene. J Virol Methods 2014; 211:36-42. [PMID: 25445883 DOI: 10.1016/j.jviromet.2014.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 10/16/2014] [Accepted: 10/21/2014] [Indexed: 11/22/2022]
Abstract
DNA vaccine coding for infectious bursal disease virus (IBDV) polyprotein gene and that for avian influenza virus (AIV) hemagglutinin (HA) gene have been shown to induce immunity and provide protection against the respective disease. The present study was carried out to determine whether an IBDV polyprotein gene-based DNA fused with AIV HA gene could trigger immune response to both IBDV and AIV. After transfection, VP2 and HA were detected in the cytoplasm and at cell membrane, respectively, by immunofluorescent antibody double staining method, suggesting the fusion strategy did not affect the location of protein expression. VP4 cleavage between VP2 and HA was confirmed by Western blot, indicating the fusion strategy did not affect VP4 function in transfected cells. After vaccination in chickens, the DNA construct VP24-HA/pcDNA induced ELISA and virus neutralizing antibodies against VP2 and hemagglutination inhibition antibody against the HA subtype. The results indicated that a single plasmid construct carrying IBDV VP243 gene-based DNA fused with AIV HA gene can elicit specific antibody responses to both IBDV and AIV by DNA vaccination.
Collapse
|
32
|
Abstract
Ebola hemorrhagic fever is one of the most fatal viral diseases worldwide affecting humans and nonhuman primates. Although infections only occur frequently in Central Africa, the virus has the potential to spread globally and is classified as a category A pathogen that could be misused as a bioterrorism agent. As of today there is no vaccine or treatment licensed to counteract Ebola virus infections. DNA, subunit and several viral vector approaches, replicating and non-replicating, have been tested as potential vaccine platforms and their protective efficacy has been evaluated in nonhuman primate models for Ebola virus infections, which closely resemble disease progression in humans. Though these vaccine platforms seem to confer protection through different mechanisms, several of them are efficacious against lethal disease in nonhuman primates attesting that vaccination against Ebola virus infections is feasible.
Collapse
Affiliation(s)
- Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana 59840, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana 59840, MT, USA
| |
Collapse
|
33
|
Warfield KL, Swenson DL, Demmin G, Bavari S. Filovirus-like particles as vaccines and discovery tools. Expert Rev Vaccines 2014; 4:429-40. [PMID: 16026254 DOI: 10.1586/14760584.4.3.429] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Ebola and Marburg viruses are members of the family Filoviridae, which cause severe hemorrhagic fevers in humans. Filovirus outbreaks have been sporadic, with mortality rates currently ranging from 30 to 90%. Unfortunately, there is no efficacious human therapy or vaccine available to treat disease caused by either Ebola or Marburg virus infection. Expression of the filovirus matrix protein, VP40, is sufficient to drive spontaneous production and release of virus-like particles (VLPs) that resemble the distinctively filamentous infectious virions. The addition of other filovirus proteins, including virion proteins (VP)24, 30 and 35 and glycoprotein, increases the efficiency of VLP production and results in particles containing multiple filovirus antigens. Vaccination with Ebola or Marburg VLPs containing glycoprotein and VP40 completely protects rodents from lethal challenge with the homologous virus. These candidate vaccines are currently being tested for immunogenicity and efficacy in nonhuman primates. Furthermore, the Ebola and Marburg VLPs are being used as a surrogate model to further understand the filovirus life cycle, with the goal of developing rationally designed vaccines and therapeutics. Thus, in addition to their use as a vaccine, VLPs are currently being used as tools to learn lessons about filovirus pathogenesis, immunology, replication and assembly requirements.
Collapse
Affiliation(s)
- Kelly L Warfield
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702-5011, USA.
| | | | | | | |
Collapse
|
34
|
|
35
|
Novel vaccination approaches against equine alphavirus encephalitides. Vaccine 2014; 32:311-9. [DOI: 10.1016/j.vaccine.2013.11.071] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/17/2013] [Accepted: 11/18/2013] [Indexed: 11/23/2022]
|
36
|
Nakayama E, Saijo M. Animal models for Ebola and Marburg virus infections. Front Microbiol 2013; 4:267. [PMID: 24046765 PMCID: PMC3763195 DOI: 10.3389/fmicb.2013.00267] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 08/19/2013] [Indexed: 11/16/2022] Open
Abstract
Ebola and Marburg hemorrhagic fevers (EHF and MHF) are caused by the Filoviridae family, Ebolavirus and Marburgvirus (ebolavirus and marburgvirus), respectively. These severe diseases have high mortality rates in humans. Although EHF and MHF are endemic to sub-Saharan Africa. A novel filovirus, Lloviu virus, which is genetically distinct from ebolavirus and marburgvirus, was recently discovered in Spain where filoviral hemorrhagic fever had never been reported. The virulence of this virus has not been determined. Ebolavirus and marburgvirus are classified as biosafety level-4 (BSL-4) pathogens and Category A agents, for which the US government requires preparedness in case of bioterrorism. Therefore, preventive measures against these viral hemorrhagic fevers should be prepared, not only in disease-endemic regions, but also in disease-free countries. Diagnostics, vaccines, and therapeutics need to be developed, and therefore the establishment of animal models for EHF and MHF is invaluable. Several animal models have been developed for EHF and MHF using non-human primates (NHPs) and rodents, which are crucial to understand pathophysiology and to develop diagnostics, vaccines, and therapeutics. Rhesus and cynomolgus macaques are representative models of filovirus infection as they exhibit remarkably similar symptoms to those observed in humans. However, the NHP models have practical and ethical problems that limit their experimental use. Furthermore, there are no inbred and genetically manipulated strains of NHP. Rodent models such as mouse, guinea pig, and hamster, have also been developed. However, these rodent models require adaptation of the virus to produce lethal disease and do not mirror all symptoms of human filovirus infection. This review article provides an outline of the clinical features of EHF and MHF in animals, including humans, and discusses how the animal models have been developed to study pathophysiology, vaccines, and therapeutics.
Collapse
Affiliation(s)
- Eri Nakayama
- Department of Virology 1, National Institute of Infectious Diseases Tokyo, Japan
| | | |
Collapse
|
37
|
Nagata LP, Wong JP, Hu WG, Wu JQ. Vaccines and therapeutics for the encephalitic alphaviruses. Future Virol 2013. [DOI: 10.2217/fvl.13.42] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This article is a review of vaccines and therapeutics in development for the encephalitic alphaviruses, which includes eastern equine encephalitis virus, western equine encephalitis virus and Venezuelan equine encephalitis virus. The encephalitic alphaviruses are endemic within regions in North and South America. Hosts are normally exposed after being bitten by infectious mosquitoes, and infection can develop into encephalitis in equines and humans with severe rates of morbidity and mortality. These viruses are also potential biological threat agents, being highly infectious via an aerosol route of exposure. In humans, equine encephalitis virus and western equine encephalitis virus are neurotropic viruses targeting the CNS and causing encephalitis. Mortality rates are 50 and 10%, respectively, for these viruses. On the other hand, Venezuelan equine encephalitis virus produces a systemic influenza-like illness with pathogenesis in the lungs and lymphoid tissue in adults and older children. The incidence of encephalitis is less than 5% in younger children with a case–mortality rate of 1%. The host response to virus infectivity is briefly discussed, along with a number of promising therapeutic and prophylactic approaches. These approaches can be broadly classified as: virus-specific, including vaccines, antibody therapy and gene-silencing oligonucleotides; or broad-spectrum, including interferon and activation of the host‘s innate immunity.
Collapse
Affiliation(s)
- Les P Nagata
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| | - Jonathan P Wong
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| | - Wei-gang Hu
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| | - Josh Q Wu
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| |
Collapse
|
38
|
Induction of broad cytotoxic T cells by protective DNA vaccination against Marburg and Ebola. Mol Ther 2013; 21:1432-44. [PMID: 23670573 DOI: 10.1038/mt.2013.61] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/12/2013] [Indexed: 12/24/2022] Open
Abstract
Marburg and Ebola hemorrhagic fevers have been described as the most virulent viral diseases known to man due to associative lethality rates of up to 90%. Death can occur within days to weeks of exposure and there is currently no licensed vaccine or therapeutic. Recent evidence suggests an important role for antiviral T cells in conferring protection, but little detailed analysis of this response as driven by a protective vaccine has been reported. We developed a synthetic polyvalent-filovirus DNA vaccine against Marburg marburgvirus (MARV), Zaire ebolavirus (ZEBOV), and Sudan ebolavirus (SUDV). Preclinical efficacy studies were performed in guinea pigs and mice using rodent-adapted viruses, whereas murine T-cell responses were extensively analyzed using a novel modified assay described herein. Vaccination was highly potent, elicited robust neutralizing antibodies, and completely protected against MARV and ZEBOV challenge. Comprehensive T-cell analysis revealed cytotoxic T lymphocytes (CTLs) of great magnitude, epitopic breadth, and Th1-type marker expression. This model provides an important preclinical tool for studying protective immune correlates that could be applied to existing platforms. Data herein support further evaluation of this enhanced gene-based approach in nonhuman primate studies for in depth analyses of T-cell epitopes in understanding protective efficacy.
Collapse
|
39
|
Tamilvanan T, Hopper W. High-throughput virtual screening and docking studies of matrix protein vp40 of ebola virus. Bioinformation 2013; 9:286-92. [PMID: 23559747 PMCID: PMC3607187 DOI: 10.6026/97320630009286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/23/2013] [Indexed: 11/23/2022] Open
Abstract
Ebolavirus, a member of the Filoviridae family of negative-sense RNA viruses, causes severe haemorrhagic fever leading up to 90% lethality. Ebolavirus matrix protein VP40 is involved in the virus assembly and budding process. The RNA binding pocket of VP40 is considered as the drug target site for structure based drug design. High Throughput Virtual Screening and molecular docking studies were employed to find the suitable inhibitors against VP40. Ten compounds showing good glide score and glide energy as well as interaction with specific amino acid residues were short listed as drug leads. These small molecule inhibitors could be potent inhibitors for VP40 matrix protein by blocking virus assembly and budding process.
Collapse
Affiliation(s)
- Thangaraju Tamilvanan
- Department of Bioinformatics, School of Bioengineering, Faculty of Engineering & Technology, SRM University, Kattankulathur,
603203, Tamil Nadu, India
| | - Waheeta Hopper
- Department of Bioinformatics, School of Bioengineering, Faculty of Engineering & Technology, SRM University, Kattankulathur,
603203, Tamil Nadu, India
| |
Collapse
|
40
|
|
41
|
Friedlander AM, Grabenstein JD, Brachman PS. Anthrax vaccines. Vaccines (Basel) 2013. [DOI: 10.1016/b978-1-4557-0090-5.00022-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
42
|
Abstract
In 1967, the first reported filovirus hemorrhagic fever outbreak took place in Germany and the former Yugoslavia. The causative agent that was identified during this outbreak, Marburg virus, is one of the most deadly human pathogens. This article provides a comprehensive overview of our current knowledge about Marburg virus disease ranging from ecology to pathogenesis and molecular biology.
Collapse
Affiliation(s)
- Kristina Brauburger
- Department of Microbiology, School of Medicine and National Emerging Infectious Diseases Laboratories Institute, Boston University, Boston, MA 02118, USA.
| | | | | | | |
Collapse
|
43
|
Bradfute SB, Warfield KL, Bray M. Mouse models for filovirus infections. Viruses 2012; 4:1477-508. [PMID: 23170168 PMCID: PMC3499815 DOI: 10.3390/v4091477] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/27/2012] [Accepted: 08/27/2012] [Indexed: 12/21/2022] Open
Abstract
The filoviruses marburg- and ebolaviruses can cause severe hemorrhagic fever (HF) in humans and nonhuman primates. Because many cases have occurred in geographical areas lacking a medical research infrastructure, most studies of the pathogenesis of filoviral HF, and all efforts to develop drugs and vaccines, have been carried out in biocontainment laboratories in non-endemic countries, using nonhuman primates (NHPs), guinea pigs and mice as animal models. NHPs appear to closely mirror filoviral HF in humans (based on limited clinical data), but only small numbers may be used in carefully regulated experiments; much research is therefore done in rodents. Because of their availability in large numbers and the existence of a wealth of reagents for biochemical and immunological testing, mice have become the preferred small animal model for filovirus research. Since the first experiments following the initial 1967 marburgvirus outbreak, wild-type or mouse-adapted viruses have been tested in immunocompetent or immunodeficient mice. In this paper, we review how these types of studies have been used to investigate the pathogenesis of filoviral disease, identify immune responses to infection and evaluate antiviral drugs and vaccines. We also discuss the strengths and weaknesses of murine models for filovirus research, and identify important questions for further study.
Collapse
Affiliation(s)
- Steven B. Bradfute
- Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-505-272-1433; Fax: +1-505-272-6995
| | - Kelly L. Warfield
- Vaccine Development, Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA;
| | - Mike Bray
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
44
|
Grant-Klein RJ, Van Deusen NM, Badger CV, Hannaman D, Dupuy LC, Schmaljohn CS. A multiagent filovirus DNA vaccine delivered by intramuscular electroporation completely protects mice from ebola and Marburg virus challenge. Hum Vaccin Immunother 2012; 8:1703-6. [PMID: 22922764 DOI: 10.4161/hv.21873] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We evaluated the immunogenicity and protective efficacy of DNA vaccines expressing the codon-optimized envelope glycoprotein genes of Zaire ebolavirus, Sudan ebolavirus, and Marburg marburgvirus (Musoke and Ravn). Intramuscular or intradermal delivery of the vaccines in BALB/c mice was performed using the TriGrid™ electroporation device. Mice that received DNA vaccines against the individual viruses developed robust glycoprotein-specific antibody titers as determined by ELISA and survived lethal viral challenge with no display of clinical signs of infection. Survival curve analysis revealed there was a statistically significant increase in survival compared to the control groups for both the Ebola and Ravn virus challenges. These data suggest that further analysis of the immune responses generated in the mice and additional protection studies in nonhuman primates are warranted.
Collapse
|
45
|
Abstract
INTRODUCTION Ebolaviruses cause severe viral hemorrhagic fever in humans and non-human primates (NHPs), with case fatality rates of up to 90%. Currently, neither a specific treatment nor a vaccine licensed for use in humans is available. However, a number of vaccine candidates have been developed in the last decade that are highly protective in NHPs, the gold standard animal model for ebola hemorrhagic fever. AREAS COVERED This review analyzes a number of scenarios for the use of ebolavirus vaccines, discusses the requirements for ebolavirus vaccines in these scenarios and describes current ebolavirus vaccines. Among these vaccines are recombinant adenoviruses, recombinant vesicular stomatitis viruses (VSVs), recombinant human parainfluenza viruses and virus-like particles. Interestingly, one of these vaccine platforms, based on recombinant VSVs, has also demonstrated post-exposure protection in NHPs. EXPERT OPINION The most pressing remaining challenge is now to move these vaccine candidates forward into human trials and toward licensure. In order to achieve this, it will be necessary to establish the mechanisms and correlates of protection for these vaccines, and to continue to demonstrate their safety, particularly in potentially immunocompromised populations. However, already now there is sufficient evidence that, from a scientific perspective, a vaccine protective against ebolaviruses is possible.
Collapse
Affiliation(s)
- Thomas Hoenen
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Division of Intramural Research, Rocky Mountain Laboratories, Disease Modelling and Transmission Unit - Laboratory of Virology , 2A120A, 903 S 4th St, Hamilton, MT, USA.
| | | | | |
Collapse
|
46
|
Albrecht MT, Eyles JE, Baillie LW, Keane-Myers AM. Immunogenicity and efficacy of an anthrax/plague DNA fusion vaccine in a mouse model. ACTA ACUST UNITED AC 2012; 65:505-9. [PMID: 22515653 DOI: 10.1111/j.1574-695x.2012.00974.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 04/02/2012] [Accepted: 04/03/2012] [Indexed: 11/27/2022]
Abstract
The efficacy of multi-agent DNA vaccines consisting of a truncated gene encoding Bacillus anthracis lethal factor (LFn) fused to either Yersinia pestis V antigen (V) or Y . pestis F1 was evaluated. A/J mice were immunized by gene gun and developed predominantly IgG1 responses that were fully protective against a lethal aerosolized B. anthracis spore challenge but required the presence of an additional DNA vaccine expressing anthrax protective antigen to boost survival against aerosolized Y. pestis.
Collapse
Affiliation(s)
- Mark T Albrecht
- Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, USA
| | | | | | | |
Collapse
|
47
|
Protective role of cytotoxic T lymphocytes in filovirus hemorrhagic fever. J Biomed Biotechnol 2011; 2011:984241. [PMID: 22253531 PMCID: PMC3255346 DOI: 10.1155/2011/984241] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 10/21/2011] [Indexed: 11/18/2022] Open
Abstract
Infection with many emerging viruses, such as the hemorrhagic fever disease caused by the filoviruses, Marburg (MARV), and Ebola virus (EBOV), leaves the host with a short timeframe in which to mouse a protective immune response. In lethal cases, uncontrolled viral replication and virus-induced immune dysregulation are too severe to overcome, and mortality is generally associated with a lack of notable immune responses. Vaccination studies in animals have demonstrated an association of IgG and neutralizing antibody responses against the protective glycoprotein antigen with survival from lethal challenge. More recently, studies in animal models of filovirus hemorrhagic fever have established that induction of a strong filovirus-specific cytotoxic T lymphocyte (CTL) response can facilitate complete viral clearance. In this review, we describe assays used to discover CTL responses after vaccination or live filovirus infection in both animal models and human clinical trials. Unfortunately, little data regarding CTL responses have been collected from infected human survivors, primarily due to the low frequency of disease and the inability to perform these studies in the field. Advancements in assays and technologies may allow these studies to occur during future outbreaks.
Collapse
|
48
|
Falzarano D, Feldmann F, Grolla A, Leung A, Ebihara H, Strong JE, Marzi A, Takada A, Jones S, Gren J, Geisbert J, Jones SM, Geisbert TW, Feldmann H. Single immunization with a monovalent vesicular stomatitis virus-based vaccine protects nonhuman primates against heterologous challenge with Bundibugyo ebolavirus. J Infect Dis 2011; 204 Suppl 3:S1082-9. [PMID: 21987745 DOI: 10.1093/infdis/jir350] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The recombinant vesicular stomatitis virus (rVSV) vector-based monovalent vaccine platform expressing a filovirus glycoprotein has been demonstrated to provide protection from lethal challenge with Ebola (EBOV) and Marburg (MARV) viruses both prophylactically and after exposure. This platform provides protection between heterologous strains within a species; however, protection from lethal challenge between species has been largely unsuccessful. To determine whether the rVSV-EBOV vaccines have the potential to provide protection against a newly emerging, phylogenetically related species, cynomolgus macaques were vaccinated with an rVSV vaccine expressing either the glycoprotein of Zaire ebolavirus (ZEBOV) or Côte d'Ivoire ebolavirus (CIEBOV) and then challenged with Bundibugyo ebolavirus (BEBOV), which was recently proposed as a new EBOV species following an outbreak in Uganda in 2007. A single vaccination with the ZEBOV-specific vaccine provided cross-protection (75% survival) against subsequent BEBOV challenge, whereas vaccination with the CIEBOV-specific vaccine resulted in an outcome similar to mock-immunized animals (33% and 25% survival, respectively). This demonstrates that monovalent rVSV-based vaccines may be useful against a newly emerging species; however, heterologous protection across species remains challenging and may depend on enhancing the immune responses either through booster immunizations or through the inclusion of multiple immunogens.
Collapse
Affiliation(s)
- Darryl Falzarano
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Warfield KL, Aman MJ. Advances in virus-like particle vaccines for filoviruses. J Infect Dis 2011; 204 Suppl 3:S1053-9. [PMID: 21987741 DOI: 10.1093/infdis/jir346] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ebola virus (EBOV) and Marburg virus (MARV) are among the deadliest human pathogens, with no vaccines or therapeutics available. Multiple vaccine platforms have been tested for efficacy as prophylactic pretreatments or therapeutics for prevention of filovirus hemorrhagic fever. Most successful vaccines are based on a virus-vectored approach expressing the protective glycoprotein (GP); protein-based subunit and DNA vaccines have been tested with moderate success. Virus-like particle (VLP) vaccines have realized promising results when tested in both rodents and nonhuman primates. VLPs rely on the natural properties of the viral matrix protein (VP) 40 to drive budding of filamentous particles that can also incorporate ≥ 1 other filovirus protein, including GP, VP24, and nucleoprotein (NP). Filovirus VLP vaccines have used particles containing 2 or 3 (GP and VP40, with or without NP) viral proteins generated in either mammalian or insect cells. Early studies successfully demonstrated efficacy of bivalent VLP vaccines in rodents; more recent studies have shown the ability of the VLP vaccines containing GP, NP, and VP40 to confer complete homologous protection against Ebola virus and Marburg virus in a prophylactic setting against in macaques. This review will discuss published work to date regarding development of the VLP vaccines for prevention of lethal filovirus hemorrhagic fever.
Collapse
Affiliation(s)
- Kelly L Warfield
- Vaccine Development, Integrated Biotherapeutics, 21 Firstfield Rd, Ste 100, Gaithersburg, MD 20878, USA.
| | | |
Collapse
|
50
|
Grant-Klein RJ, Altamura LA, Schmaljohn CS. Progress in recombinant DNA-derived vaccines for Lassa virus and filoviruses. Virus Res 2011; 162:148-61. [PMID: 21925552 DOI: 10.1016/j.virusres.2011.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/01/2011] [Accepted: 09/04/2011] [Indexed: 11/19/2022]
Abstract
Developing vaccines for highly pathogenic viruses such as those causing Lassa, Ebola, and Marburg hemorrhagic fevers is a daunting task due to both scientific and logistical constraints. Scientific hurdles to overcome include poorly defined relationships between pathogenicity and protective immune responses, genetic diversity of viruses, and safety in a target population that includes a large number of individuals with compromised immune systems. Logistical obstacles include the requirement for biosafety level-4 containment to study the authentic viruses, the poor public health infrastructure of the endemic disease areas, and the cost of developing these vaccines for use in non-lucrative markets. Recombinant DNA-based vaccine approaches offer promise of overcoming some of these issues. In this review, we consider the status of various recombinant DNA candidate vaccines against Lassa virus and filoviruses which have been tested in animals.
Collapse
Affiliation(s)
- Rebecca J Grant-Klein
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | | |
Collapse
|