1
|
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
|
2
|
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
|
3
|
Rossi SL, Russell-Lodrigue KE, Plante KS, Bergren NA, Gorchakov R, Roy CJ, Weaver SC. Rationally Attenuated Vaccines for Venezuelan Equine Encephalitis Protect Against Epidemic Strains with a Single Dose. Vaccines (Basel) 2020; 8:E497. [PMID: 32887313 PMCID: PMC7563393 DOI: 10.3390/vaccines8030497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is a re-emerging virus of human, agriculture, and bioweapon threat importance. No FDA-approved treatment is available to combat Venezuelan equine encephalitis in humans, prompting the need to create a vaccine that is safe, efficacious, and cannot be replicated in the mosquito vector. Here we describe the use of a serotype ID VEEV (ZPC-738) vaccine with an internal ribosome entry site (IRES) to alter gene expression patterns. This ZPC/IRES vaccine was genetically engineered in two ways based on the position of the IRES insertion to create a vaccine that is safe and efficacious. After a single dose, both versions of the ZPC/IRES vaccine elicited neutralizing antibody responses in mice and non-human primates after a single dose, with more robust responses produced by version 2. Further, all mice and primates were protected from viremia following VEEV challenge. These vaccines were also safer in neonatal mice than the current investigational new drug vaccine, TC-83. These results show that IRES-based attenuation of alphavirus genomes consistently produce promising vaccine candidates, with VEEV/IRES version 2 showing promise for further development.
Collapse
Affiliation(s)
- Shannan L. Rossi
- Department of Pathology and Microbiology and Immunology, Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Kenneth S. Plante
- Department of Microbiology and Immunology and World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Nicholas A. Bergren
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Rodion Gorchakov
- Department of Health, Safety and Environment, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia;
| | - Chad J. Roy
- Tulane National Primate Research Center, Covington, LA 70433, USA; (K.E.R.-L.); (C.J.R.)
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Scott C. Weaver
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
4
|
Johnson DM, Sokoloski KJ, Jokinen JD, Pfeffer TL, Chu YK, Adcock RS, Chung D, Tretyakova I, Pushko P, Lukashevich IS. Advanced Safety and Genetic Stability in Mice of a Novel DNA-Launched Venezuelan Equine Encephalitis Virus Vaccine with Rearranged Structural Genes. Vaccines (Basel) 2020; 8:vaccines8010114. [PMID: 32121666 PMCID: PMC7157698 DOI: 10.3390/vaccines8010114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022] Open
Abstract
The safety and genetic stability of V4020, a novel Venezuelan Equine Encephalitis Virus (VEEV) vaccine based on the investigational VEEV TC-83 strain, was evaluated in mice. V4020 was generated from infectious DNA, contains a stabilizing mutation in the E2-120 glycoprotein, and includes rearrangement of structural genes. After intracranial inoculation (IC), replication of V4020 was more attenuated than TC-83, as documented by low clinical scores, inflammation, viral load in brain, and earlier viral clearance. During the first 9 days post-inoculation (DPI), genes involved in inflammation, cytokine signaling, adaptive immune responses, and apoptosis were upregulated in both groups. However, the magnitude of upregulation was greater in TC-83 than V4020 mice, and this pattern persisted till 13 DPI, while V4020 gene expression profiles declined to mock-infected levels. In addition, genetic markers of macrophages, DCs, and microglia were strongly upregulated in TC-83 mice. During five serial passages in the brain, less severe clinical manifestations and a lower viral load were observed in V4020 mice and all animals survived. In contrast, 13.3% of mice met euthanasia criteria during the passages in TC-83 group. At 2 DPI, RNA-Seq analysis of brain tissues revealed that V4020 mice had lower rates of mutations throughout five passages. A higher synonymous mutation ratio was observed in the nsP4 (RdRP) gene of TC-83 compared to V4020 mice. At 2 DPI, both viruses induced different expression profiles of host genes involved in neuro-regeneration. Taken together, these results provide evidence for the improved safety and genetic stability of the experimental V4020 VEEV vaccine in a murine model.
Collapse
Affiliation(s)
- Dylan M. Johnson
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (K.J.S.); (D.C.)
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
- Correspondence: (D.M.J.); (I.S.L.)
| | - Kevin J. Sokoloski
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (K.J.S.); (D.C.)
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
| | - Jenny D. Jokinen
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Tia L. Pfeffer
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Yong-Kyu Chu
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
| | - Robert S. Adcock
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
| | - Donghoon Chung
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (K.J.S.); (D.C.)
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
| | | | - Peter Pushko
- Medigen, Inc., Frederick, MD 21701, USA; (I.T.); (P.P.)
| | - Igor S. Lukashevich
- Center for Predictive Medicine, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (T.L.P.); (Y.-K.C.); (R.S.A.)
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
- Correspondence: (D.M.J.); (I.S.L.)
| |
Collapse
|
5
|
Tretyakova I, Plante KS, Rossi SL, Lawrence WS, Peel JE, Gudjohnsen S, Wang E, Mirchandani D, Tibbens A, Lamichhane TN, Lukashevich IS, Comer JE, Weaver SC, Pushko P. Venezuelan equine encephalitis vaccine with rearranged genome resists reversion and protects non-human primates from viremia after aerosol challenge. Vaccine 2020; 38:3378-3386. [PMID: 32085953 DOI: 10.1016/j.vaccine.2020.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023]
Abstract
Live-attenuated V4020 vaccine for Venezuelan equine encephalitis virus (VEEV) containing attenuating rearrangement of the virus structural genes was evaluated in a non-human primate model for immunogenicity and protective efficacy against aerosol challenge with wild-type VEEV. The genomic RNA of V4020 vaccine virus was encoded in the pMG4020 plasmid under control of the CMV promoter and contained the capsid gene downstream from the glycoprotein genes. It also included attenuating mutations from the VEE TC83 vaccine, with E2-120Arg substitution genetically engineered to prevent reversion mutations. The population of V4020 vaccine virus derived from pMG4020-transfected Vero cells was characterized by next generation sequencing (NGS) and indicated no detectable genetic reversions. Cynomolgus macaques were vaccinated with V4020 vaccine virus. After one or two vaccinations including by intramuscular route, high levels of virus-neutralizing antibodies were confirmed with no viremia or apparent adverse reactions to vaccinations. The protective effect of vaccination was evaluated using an aerosol challenge with VEEV. After challenge, macaques had no detectable viremia, demonstrating a protective effect of vaccination with live V4020 VEEV vaccine.
Collapse
Affiliation(s)
- Irina Tretyakova
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD 21701, USA.
| | - Kenneth S Plante
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA; World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - William S Lawrence
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennifer E Peel
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sif Gudjohnsen
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA; World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Eryu Wang
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Divya Mirchandani
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA; World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Alexander Tibbens
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD 21701, USA
| | - Tek N Lamichhane
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD 21701, USA
| | - Igor S Lukashevich
- Department of Pharmacology and Toxicology, University of Louisville, 505 S Hancock St., Louisville, KY 40202, USA
| | - Jason E Comer
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA; World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Scott C Weaver
- Institute for Human Infections and Immunity and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA; World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Peter Pushko
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD 21701, USA.
| |
Collapse
|
6
|
Burke CW, Froude JW, Rossi F, White CE, Moyer CL, Ennis J, Pitt ML, Streatfield S, Jones RM, Musiychuk K, Kervinen J, Zeitlin L, Yusibov V, Glass PJ. Therapeutic monoclonal antibody treatment protects nonhuman primates from severe Venezuelan equine encephalitis virus disease after aerosol exposure. PLoS Pathog 2019; 15:e1008157. [PMID: 31790515 PMCID: PMC6907853 DOI: 10.1371/journal.ppat.1008157] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/12/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
There are no FDA licensed vaccines or therapeutics for Venezuelan equine encephalitis virus (VEEV) which causes a debilitating acute febrile illness in humans that can progress to encephalitis. Previous studies demonstrated that murine and macaque monoclonal antibodies (mAbs) provide prophylactic and therapeutic efficacy against VEEV peripheral and aerosol challenge in mice. Additionally, humanized versions of two neutralizing mAbs specific for the E2 glycoprotein, 1A3B-7 and 1A4A-1, administered singly protected mice against aerosolized VEEV. However, no studies have demonstrated protection in nonhuman primate (NHP) models of VEEV infection. Here, we evaluated a chimeric antibody 1A3B-7 (c1A3B-7) containing mouse variable regions on a human IgG framework and a humanized antibody 1A4A-1 containing a serum half-life extension modification (Hu-1A4A-1-YTE) for their post-exposure efficacy in NHPs exposed to aerosolized VEEV. Approximately 24 hours after exposure, NHPs were administered a single bolus intravenous mAb. Control NHPs had typical biomarkers of VEEV infection including measurable viremia, fever, and lymphopenia. In contrast, c1A3B-7 treated NHPs had significant reductions in viremia and lymphopenia and on average approximately 50% reduction in fever. Although not statistically significant, Hu-1A4A-1-YTE administration did result in reductions in viremia and fever duration. Delay of treatment with c1A3B-7 to 48 hours post-exposure still provided NHPs protection from severe VEE disease through reductions in viremia and fever. These results demonstrate that post-exposure administration of c1A3B-7 protected macaques from development of severe VEE disease even when administered 48 hours following aerosol exposure and describe the first evaluations of VEEV-specific mAbs for post-exposure prophylactic use in NHPs. Viral mutations were identified in one NHP after c1A3B-7 treatment administered 24 hrs after virus exposure. This suggests that a cocktail-based therapy, or an alternative mAb against an epitope that cannot mutate without resulting in loss of viral fitness may be necessary for a highly effective therapeutic.
Collapse
Affiliation(s)
- Crystal W. Burke
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Jeffery W. Froude
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Franco Rossi
- Center of Aerobiological Sciences, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Charles E. White
- Biostatisics Branch, US Army Medical Research Institute of Infectious Disease, Fort Detrick Maryland, United States of America
| | - Crystal L. Moyer
- Mapp Biopharmaceutical, Inc., San Diego, California, United States of America
| | - Jane Ennis
- Mapp Biopharmaceutical, Inc., San Diego, California, United States of America
| | - M. Louise Pitt
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Stephen Streatfield
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - R. Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Konstantin Musiychuk
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Jukka Kervinen
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, California, United States of America
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Pamela J. Glass
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| |
Collapse
|
7
|
Rusnak JM, Glass PJ, Weaver SC, Sabourin CL, Glenn AM, Klimstra W, Badorrek CS, Nasar F, Ward LA. Approach to Strain Selection and the Propagation of Viral Stocks for Venezuelan Equine Encephalitis Virus Vaccine Efficacy Testing under the Animal Rule. Viruses 2019; 11:v11090807. [PMID: 31480472 PMCID: PMC6784384 DOI: 10.3390/v11090807] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/23/2019] [Accepted: 08/30/2019] [Indexed: 12/21/2022] Open
Abstract
Licensure of a vaccine to protect against aerosolized Venezuelan equine encephalitis virus (VEEV) requires use of the U.S. Food and Drug Administration (FDA) Animal Rule to assess vaccine efficacy as human studies are not feasible or ethical. An approach to selecting VEEV challenge strains for use under the Animal Rule was developed, taking into account Department of Defense (DOD) vaccine requirements, FDA Animal Rule guidelines, strain availability, and lessons learned from the generation of filovirus challenge agents within the Filovirus Animal Nonclinical Group (FANG). Initial down-selection to VEEV IAB and IC epizootic varieties was based on the DOD objective for vaccine protection in a bioterrorism event. The subsequent down-selection of VEEV IAB and IC isolates was based on isolate availability, origin, virulence, culture and animal passage history, known disease progression in animal models, relevancy to human disease, and ability to generate sufficient challenge material. Methods for the propagation of viral stocks (use of uncloned (wild-type), plaque-cloned, versus cDNA-cloned virus) to minimize variability in the potency of the resulting challenge materials were also reviewed. The presented processes for VEEV strain selection and the propagation of viral stocks may serve as a template for animal model development product testing under the Animal Rule to other viral vaccine programs. This manuscript is based on the culmination of work presented at the “Alphavirus Workshop” organized and hosted by the Joint Vaccine Acquisition Program (JVAP) on 15 December 2014 at Fort Detrick, Maryland, USA.
Collapse
Affiliation(s)
- Janice M Rusnak
- Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager-Medical Countermeasure Systems (JMP-MCS), Joint Vaccine Acquisition Program (JVAP), 1564 Freedman Drive, Fort Detrick, MD 21702, USA.
| | - Pamela J Glass
- Department of Virology, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, MD 21702, USA
| | - Scott C Weaver
- Institute for Human Infections and Immunity, World Reference Center for Emerging Viruses and Arboviruses and Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Carol L Sabourin
- Battelle Biomedical Research Center, 1425 Plain City-Georgesville Road, West Jefferson, OH 43162, USA
| | - Andrew M Glenn
- Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager-Medical Countermeasure Systems (JMP-MCS), Joint Vaccine Acquisition Program (JVAP), 1564 Freedman Drive, Fort Detrick, MD 21702, USA
| | - William Klimstra
- Center for Vaccine Research, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA
| | - Christopher S Badorrek
- Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager-Medical Countermeasure Systems (JMP-MCS), Joint Vaccine Acquisition Program (JVAP), 1564 Freedman Drive, Fort Detrick, MD 21702, USA
| | - Farooq Nasar
- Department of Virology, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, MD 21702, USA
| | - Lucy A Ward
- Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager-Medical Countermeasure Systems (JMP-MCS), Joint Vaccine Acquisition Program (JVAP), 1564 Freedman Drive, Fort Detrick, MD 21702, USA
| |
Collapse
|
8
|
Novel DNA-launched Venezuelan equine encephalitis virus vaccine with rearranged genome. Vaccine 2019; 37:3317-3325. [PMID: 31072736 DOI: 10.1016/j.vaccine.2019.04.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/20/2019] [Accepted: 04/24/2019] [Indexed: 11/23/2022]
Abstract
Novel live-attenuated V4020 vaccine was prepared for Venezuelan equine encephalitis virus (VEEV), an alphavirus from the Togaviridae family. The genome of V4020 virus was rearranged, with the capsid gene expressed using a duplicate subgenomic promoter downstream from the glycoprotein genes. V4020 also included both attenuating mutations from the TC83 VEEV vaccine secured by mutagenesis to prevent reversion mutations. The full-length infectious RNA of V4020 vaccine virus was expressed from pMG4020 plasmid downstream from the CMV promoter and launched replication of live-attenuated V4020 in vitro or in vivo. BALB/c mice vaccinated with a single dose of V4020 virus or with pMG4020 plasmid had no adverse reactions to vaccinations and developed high titers of neutralizing antibodies. After challenge with the wild type VEEV, vaccinated mice survived with no morbidity, while all unvaccinated controls succumbed to lethal infection. Intracranial injections in mice showed attenuated replication of V4020 vaccine virus as compared to the TC83. We conclude that V4020 vaccine has safety advantage over TC83, while provides equivalent protection in a mouse VEEV challenge model.
Collapse
|
9
|
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
|
10
|
Abstract
Zoonotic viruses circulate as swarms in animal reservoirs and can emerge into human populations, causing epidemics that adversely affect public health. Portable, safe, and effective vaccine platforms are needed in the context of these outbreak and emergence situations. In this work, we report the generation and characterization of an alphavirus replicon vaccine platform based on a non-select agent, attenuated Venezuelan equine encephalitis (VEE) virus vaccine, strain 3526 (VRP 3526). Using both noroviruses and coronaviruses as model systems, we demonstrate the utility of the VRP 3526 platform in the generation of recombinant proteins, production of virus-like particles, and in vivo efficacy as a vaccine against emergent viruses. Importantly, packaging under biosafety level 2 (BSL2) conditions distinguishes VRP 3526 from previously reported alphavirus platforms and makes this approach accessible to the majority of laboratories around the world. In addition, improved outcomes in the vulnerable aged models as well as against heterologous challenge suggest improved efficacy compared to that of previously attenuated VRP approaches. Taking these results together, the VRP 3526 platform represents a safe and highly portable system that can be rapidly deployed under BSL2 conditions for generation of candidate vaccines against emerging microbial pathogens. IMPORTANCE While VEE virus replicon particles provide a robust, established platform for antigen expression and vaccination, its utility has been limited by the requirement for high-containment-level facilities for production and packaging. In this work, we utilize an attenuated vaccine strain capable of use at lower biocontainment level but retaining the capacity of the wild-type replicon particle. Importantly, the new replicon platform provides equal protection for aged mice and following heterologous challenge, which distinguishes it from other attenuated replicon platforms. Together, the new system represents a highly portable, safe system for use in the context of disease emergence.
Collapse
|
11
|
|
12
|
Gupta P, Sharma A, Han J, Yang A, Bhomia M, Knollmann-Ritschel B, Puri RK, Maheshwari RK. Differential host gene responses from infection with neurovirulent and partially-neurovirulent strains of Venezuelan equine encephalitis virus. BMC Infect Dis 2017; 17:309. [PMID: 28446152 PMCID: PMC5405508 DOI: 10.1186/s12879-017-2355-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 03/28/2017] [Indexed: 12/21/2022] Open
Abstract
Background Venezuelan equine encephalitis virus (VEEV) is an alphavirus in the family Togaviridae. VEEV causes a bi-phasic illness in mice where primary replication in lymphoid organs is followed by entry into the central nervous system (CNS). The CNS phase of infection is marked by encephalitis and large scale neuronal death ultimately resulting in death. Molecular determinants of VEEV neurovirulence are not well understood. In this study, host gene expression response to highly neurovirulent VEEV (V3000 strain) infection was compared with that of a partially neurovirulent VEEV (V3034 strain) to identify host factors associated with VEEV neurovirulence. Methods Whole genome microarrays were performed to identify the significantly modulated genes. Microarray observations were classified into three categories i.e., genes that were similarly modulated against both V3000 and V3034 infections, and genes that were uniquely modulated in infection with V3034 or V3000. Histologic sections of spleen and brain were evaluated by hematoxylin and eosin stains from all the mice. Results V3000 infection induced a greater degree of pathology in both the spleen and brain tissue of infected mice compared to V3034 infection. Genes commonly modulated in the spleens after V3000 or V3034 infection were associated with innate immune responses, inflammation and antigen presentation, however, V3000 induced a gene response profile that suggests a stronger inflammatory and apoptotic response compared to V3034. In the brain, both the strains of VEEV induced an innate immune response reflected by an upregulation of the genes involved in antigen presentation, interferon response, and inflammation. Similar to the spleen, V3000 was found to induce a stronger inflammatory response than V3034 in terms of induction of pro-inflammatory genes and associated pathways. Ccl2, Ccl5, Ccl6, and Ly6 were uniquely upregulated in V3000 infected mouse brains and correlated with the extensive inflammation observed in the brain. Conclusion The common gene profile identified from V3000 and V3034 exposure can help in understanding a generalized host response to VEEV infection. Inflammatory genes that were uniquely identified in mouse brains with V3000 infection will help in better understanding the lethal neurovirulence of VEEV. Future studies are needed to explore the roles played by the genes identified in VEEV induced encephalitis. Electronic supplementary material The online version of this article (doi:10.1186/s12879-017-2355-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paridhi Gupta
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Anuj Sharma
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Jing Han
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA
| | - Amy Yang
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA
| | - Manish Bhomia
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Barbara Knollmann-Ritschel
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Raj K Puri
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA
| | - Radha K Maheshwari
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| |
Collapse
|
13
|
Salimi H, Cain MD, Klein RS. Encephalitic Arboviruses: Emergence, Clinical Presentation, and Neuropathogenesis. Neurotherapeutics 2016; 13:514-34. [PMID: 27220616 PMCID: PMC4965410 DOI: 10.1007/s13311-016-0443-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Arboviruses are arthropod-borne viruses that exhibit worldwide distribution, contributing to systemic and neurologic infections in a variety of geographical locations. Arboviruses are transmitted to vertebral hosts during blood feedings by mosquitoes, ticks, biting flies, mites, and nits. While the majority of arboviral infections do not lead to neuroinvasive forms of disease, they are among the most severe infectious risks to the health of the human central nervous system. The neurologic diseases caused by arboviruses include meningitis, encephalitis, myelitis, encephalomyelitis, neuritis, and myositis in which virus- and immune-mediated injury may lead to severe, persisting neurologic deficits or death. Here we will review the major families of emerging arboviruses that cause neurologic infections, their neuropathogenesis and host neuroimmunologic responses, and current strategies for treatment and prevention of neurologic infections they cause.
Collapse
Affiliation(s)
- Hamid Salimi
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew D Cain
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Robyn S Klein
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
14
|
Abstract
Inactivated vaccines have been used for over a century to induce protection against viral pathogens. This established approach of vaccine production is relatively straightforward to achieve and there is an augmented safety profile as compared to their live counterparts. Today, there are six viral pathogens for which licensed inactivated vaccines are available with many more in development. Here, we describe the principles of viral inactivation and the application of these principles to vaccine development. Specifically emphasized are the manufacturing procedure and the accompanying assays, of which assays used for monitoring the inactivation process and preservation of neutralizing epitopes, are pivotal. Novel inactivated vaccines in development and the hurdles they face for licensure are also discussed as well as the (dis)advantages of inactivation over the other vaccine production methodologies.
Collapse
|
15
|
Voss K, Amaya M, Mueller C, Roberts B, Kehn-Hall K, Bailey C, Petricoin E, Narayanan A. Inhibition of host extracellular signal-regulated kinase (ERK) activation decreases new world alphavirus multiplication in infected cells. Virology 2014; 468-470:490-503. [PMID: 25261871 PMCID: PMC7127730 DOI: 10.1016/j.virol.2014.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/31/2014] [Accepted: 09/06/2014] [Indexed: 01/13/2023]
Abstract
New World alphaviruses belonging to the family Togaviridae are classified as emerging infectious agents and Category B select agents. Our study is focused on the role of the host extracellular signal-regulated kinase (ERK) in the infectious process of New World alphaviruses. Infection of human cells by Venezuelan equine encephalitis virus (VEEV) results in the activation of the ERK-signaling cascade. Inhibition of ERK1/2 by the small molecule inhibitor Ag-126 results in inhibition of viral multiplication. Ag-126-mediated inhibition of VEEV was due to potential effects on early and late stages of the infectious process. While expression of viral proteins was down-regulated in Ag-126 treated cells, we did not observe any influence of Ag-126 on the nuclear distribution of capsid. Finally, Ag-126 exerted a broad-spectrum inhibitory effect on New World alphavirus multiplication, thus indicating that the host kinase, ERK, is a broad-spectrum candidate for development of novel therapeutics against New World alphaviruses. VEEV infection activated multiple components of the ERK signaling cascade. Inhibition of ERK activation using Ag-126 inhibited VEEV multiplication. Activation of ERK by Ceramide C6 increased infectious titers of TC-83. Ag-126 inhibited virulent strains of all New World alphaviruses. Ag-126 treatment increased percent survival of infected cells.
Collapse
Affiliation(s)
- Kelsey Voss
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA, USA
| | - Moushimi Amaya
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA, USA
| | - Claudius Mueller
- Center for Applied Proteomics and Personalized Medicine, George Mason University, 10900 University Boulevard, Manassas, VA, USA
| | - Brian Roberts
- Leidos Health Life Sciences, 5202 Presidents Court, Suite 110, Frederick, MD, USA
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA, USA
| | - Charles Bailey
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA, USA
| | - Emanuel Petricoin
- Center for Applied Proteomics and Personalized Medicine, George Mason University, 10900 University Boulevard, Manassas, VA, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, 10650 Pyramid Place, Manassas, VA, USA.
| |
Collapse
|
16
|
Second generation inactivated eastern equine encephalitis virus vaccine candidates protect mice against a lethal aerosol challenge. PLoS One 2014; 9:e104708. [PMID: 25116127 PMCID: PMC4130539 DOI: 10.1371/journal.pone.0104708] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/16/2014] [Indexed: 11/23/2022] Open
Abstract
Currently, there are no FDA-licensed vaccines or therapeutics for eastern equine encephalitis virus (EEEV) for human use. We recently developed several methods to inactivate CVEV1219, a chimeric live-attenuated eastern equine encephalitis virus (EEEV). Dosage and schedule studies were conducted to evaluate the immunogenicity and protective efficacy of three potential second-generation inactivated EEEV (iEEEV) vaccine candidates in mice: formalin-inactivated CVEV1219 (fCVEV1219), INA-inactivated CVEV1219 (iCVEV1219) and gamma-irradiated CVEV1219 (gCVEV1219). Both fCVEV1219 and gCVEV1219 provided partial to complete protection against an aerosol challenge when administered by different routes and schedules at various doses, while iCVEV1219 was unable to provide substantial protection against an aerosol challenge by any route, dose, or schedule tested. When evaluating antibody responses, neutralizing antibody, not virus specific IgG or IgA, was the best correlate of protection. The results of these studies suggest that both fCVEV1219 and gCVEV1219 should be evaluated further and considered for advancement as potential second-generation inactivated vaccine candidates for EEEV.
Collapse
|
17
|
Monath TP. Vaccines against diseases transmitted from animals to humans: a one health paradigm. Vaccine 2013; 31:5321-38. [PMID: 24060567 PMCID: PMC7130581 DOI: 10.1016/j.vaccine.2013.09.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/08/2013] [Accepted: 09/16/2013] [Indexed: 10/28/2022]
Abstract
This review focuses on the immunization of animals as a means of preventing human diseases (zoonoses). Three frameworks for the use of vaccines in this context are described, and examples are provided of successes and failures. Framework I vaccines are used for protection of humans and economically valuable animals, where neither plays a role in the transmission cycle. The benefit of collaborations between animal health and human health industries and regulators in developing such products is discussed, and one example (West Nile vaccine) of a single product developed for use in animals and humans is described. Framework II vaccines are indicated for domesticated animals as a means of preventing disease in both animals and humans. The agents of concern are transmitted directly or indirectly (e.g. via arthropod vectors) from animals to humans. A number of examples of the use of Framework II vaccines are provided, e.g. against brucellosis, Escherichia coli O157, rabies, Rift Valley fever, Venezuelan equine encephalitis, and Hendra virus. Framework III vaccines are used to immunize wild animals as a means of preventing transmission of disease agents to humans and domesticated animals. Examples are reservoir-targeted, oral bait rabies, Mycobacterium bovis and Lyme disease vaccines. Given the speed and lost cost of veterinary vaccine development, some interventions based on the immunization of animals could lead to rapid and relatively inexpensive advances in public health. Opportunities for vaccine-based approaches to preventing zoonotic and emerging diseases that integrate veterinary and human medicine (the One Health paradigm) are emphasized.
Collapse
Affiliation(s)
- Thomas P Monath
- One Health Initiative Pro Bono Team, United States(1); Austria; PaxVax Inc., United States.
| |
Collapse
|
18
|
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
|
19
|
|
20
|
Tretyakova I, Lukashevich IS, Glass P, Wang E, Weaver S, Pushko P. Novel vaccine against Venezuelan equine encephalitis combines advantages of DNA immunization and a live attenuated vaccine. Vaccine 2012; 31:1019-25. [PMID: 23287629 DOI: 10.1016/j.vaccine.2012.12.050] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 12/12/2012] [Accepted: 12/17/2012] [Indexed: 11/18/2022]
Abstract
DNA vaccines combine remarkable genetic and chemical stability with proven safety and efficacy in animal models, while remaining less immunogenic in humans. In contrast, live-attenuated vaccines have the advantage of inducing rapid, robust, long-term immunity after a single-dose vaccination. Here we describe novel iDNA vaccine technology that is based on an infectious DNA platform and combines advantages of DNA and live attenuated vaccines. We applied this technology for vaccination against infection with Venezuelan equine encephalitis virus (VEEV), an alphavirus from the Togaviridae family. The iDNA vaccine is based on transcription of the full-length genomic RNA of the TC-83 live-attenuated virus from plasmid DNA in vivo. The in vivo-generated viral RNA initiates limited replication of the vaccine virus, which in turn leads to efficient immunization. This technology allows the plasmid DNA to launch a live-attenuated vaccine in vitro or in vivo. Less than 10 ng of pTC83 iDNA encoding the full-length genomic RNA of the TC-83 vaccine strain initiated replication of the vaccine virus in vitro. In order to evaluate this approach in vivo, BALB/c mice were vaccinated with a single dose of pTC83 iDNA. After vaccination, all mice seroconverted with no adverse reactions. Four weeks after immunization, animals were challenged with the lethal epidemic strain of VEEV. All iDNA-vaccinated mice were protected from fatal disease, while all unvaccinated controls succumbed to infection and died. To our knowledge, this is the first example of launching a clinical live-attenuated vaccine from recombinant plasmid DNA in vivo.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Encephalitis Virus, Venezuelan Equine/immunology
- Encephalomyelitis, Venezuelan Equine/immunology
- Encephalomyelitis, Venezuelan Equine/prevention & control
- Female
- Mice
- Mice, Inbred BALB C
- Survival Analysis
- Vaccination/methods
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Irina Tretyakova
- Medigen, Inc., 4539 Metropolitan Court, Frederick, MD 21704, USA
| | | | | | | | | | | |
Collapse
|
21
|
Rülker T, Voß L, Thullier P, O' Brien LM, Pelat T, Perkins SD, Langermann C, Schirrmann T, Dübel S, Marschall HJ, Hust M, Hülseweh B. Isolation and characterisation of a human-like antibody fragment (scFv) that inactivates VEEV in vitro and in vivo. PLoS One 2012; 7:e37242. [PMID: 22666347 PMCID: PMC3364240 DOI: 10.1371/journal.pone.0037242] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/18/2012] [Indexed: 11/18/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) belongs to the Alphavirus genus and several species of this family are pathogenic to humans. The viruses are classified as potential agents of biological warfare and terrorism and sensitive detection as well as effective prophylaxis and antiviral therapies are required.In this work, we describe the isolation of the anti-VEEV single chain Fragment variable (scFv), ToR67-3B4, from a non-human primate (NHP) antibody gene library. We report its recloning into the bivalent scFv-Fc format and further immunological and biochemical characterisation.The scFv-Fc ToR67-3B4 recognised viable as well as formalin and ß-propionolactone (ß-Pl) inactivated virus particles and could be applied for immunoblot analysis of VEEV proteins and immuno-histochemistry of VEEV infected cells. It detected specifically the viral E1 envelope protein of VEEV but did not react with reduced viral glycoprotein preparations suggesting that recognition depends upon conformational epitopes. The recombinant antibody was able to detect multiple VEEV subtypes and displayed only marginal cross-reactivity to other Alphavirus species except for EEEV. In addition, the scFv-Fc fusion described here might be of therapeutic use since it successfully inactivated VEEV in a murine disease model. When the recombinant antibody was administered 6 hours post challenge, 80% to 100% of mice survived lethal VEEV IA/B or IE infection. Forty to sixty percent of mice survived when scFv-Fc ToR67-3B4 was applied 6 hours post challenge with VEEV subtypes II and former IIIA. In combination with E2-neutralising antibodies the NHP antibody isolated here could significantly improve passive protection as well as generic therapy of VEE.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/isolation & purification
- Cloning, Molecular
- Encephalitis Virus, Venezuelan Equine/immunology
- Gene Library
- Genetic Vectors/genetics
- Humans
- Immunization, Passive
- Macaca fascicularis
- Male
- Mice
- Mice, Inbred BALB C
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/isolation & purification
- Sequence Analysis
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/isolation & purification
Collapse
Affiliation(s)
- Torsten Rülker
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Luzie Voß
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| | - Philippe Thullier
- Centre de Recherche du Service de Santé des Armées (CRSSA-IRBA), La Tronche, France
| | - Lyn M. O' Brien
- Defence Science and Technology Laboratory, Biomedical Sciences Department, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - Thibaut Pelat
- Centre de Recherche du Service de Santé des Armées (CRSSA-IRBA), La Tronche, France
| | - Stuart D. Perkins
- Defence Science and Technology Laboratory, Biomedical Sciences Department, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - Claudia Langermann
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Hans-Jürgen Marschall
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| |
Collapse
|
22
|
O'Brien LM, Goodchild SA, Phillpotts RJ, Perkins SD. A humanised murine monoclonal antibody protects mice from Venezuelan equine encephalitis virus, Everglades virus and Mucambo virus when administered up to 48 h after airborne challenge. Virology 2012; 426:100-5. [PMID: 22341308 DOI: 10.1016/j.virol.2012.01.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 01/03/2012] [Accepted: 01/30/2012] [Indexed: 11/18/2022]
Abstract
Currently there are no licensed antiviral treatments for the Alphaviruses Venezuelan equine encephalitis virus (VEEV), Everglades virus and Mucambo virus. We previously developed a humanised version of the mouse monoclonal antibody 1A3B-7 (Hu1A3B-7) which exhibited a wide range of reactivity in vitro and was able to protect mice from infection with VEEV. Continued work with the humanised antibody has now demonstrated that it has the potential to be a new human therapeutic. Hu1A3B-7 successfully protected mice from infection with multiple Alphaviruses. The effectiveness of the humanisation process was determined by assessing proliferation responses in human T-cells to peptides derived from the murine and humanised versions of the V(H) and V(L) domains. This analysis showed that the number of human T-cell epitopes within the humanised antibody had been substantially reduced, indicating that Hu1A3B-7 may have reduced immunogenicity in vivo.
Collapse
Affiliation(s)
- Lyn M O'Brien
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 0JQ, UK.
| | | | | | | |
Collapse
|
23
|
Aguilar PV, Estrada-Franco JG, Navarro-Lopez R, Ferro C, Haddow AD, Weaver SC. Endemic Venezuelan equine encephalitis in the Americas: hidden under the dengue umbrella. Future Virol 2011. [DOI: 10.2217/fvl.11.50] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Venezuelan equine encephalitis (VEE) is an emerging infectious disease in Latin America. Outbreaks have been recorded for decades in countries with enzootic circulation, and the recent implementation of surveillance systems has allowed the detection of additional human cases in countries and areas with previously unknown VEE activity. Clinically, VEE is indistinguishable from dengue and other arboviral diseases and confirmatory diagnosis requires the use of specialized laboratory tests that are difficult to afford in resource-limited regions. Thus, the disease burden of endemic VEE in developing countries remains largely unknown, but recent surveillance suggests that it may represent up to 10% of the dengue burden in neotropical cities, or tens-of-thousands of cases per year throughout Latin America. The potential emergence of epizootic viruses from enzootic progenitors further highlights the need to strengthen surveillance activities, identify mosquito vectors and reservoirs and develop effective strategies to control the disease. In this article, we provide an overview of the current status of endemic VEE that results from spillover of the enzootic cycles, and we discuss public health measures for disease control as well as future avenues for VEE research.
Collapse
Affiliation(s)
- Patricia V Aguilar
- Center for Tropical Diseases, Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jose G Estrada-Franco
- Center for Tropical Diseases, Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Roberto Navarro-Lopez
- Comision Mexico-Estados Unidos para la Prevencion de la Fiebre Aftosa & Otras Enfermedades Exoticas de los Animales, Mexico City, Mexico
| | | | - Andrew D Haddow
- Center for Tropical Diseases, Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | | |
Collapse
|
24
|
Aguilar PV, Estrada-Franco JG, Navarro-Lopez R, Ferro C, Haddow AD, Weaver SC. Endemic Venezuelan equine encephalitis in the Americas: hidden under the dengue umbrella. Future Virol 2011; 6:721-740. [PMID: 21765860 DOI: 10.2217/fvl.11.5] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Venezuelan equine encephalitis (VEE) is an emerging infectious disease in Latin America. Outbreaks have been recorded for decades in countries with enzootic circulation, and the recent implementation of surveillance systems has allowed the detection of additional human cases in countries and areas with previously unknown VEE activity. Clinically, VEE is indistinguishable from dengue and other arboviral diseases and confirmatory diagnosis requires the use of specialized laboratory tests that are difficult to afford in resource-limited regions. Thus, the disease burden of endemic VEE in developing countries remains largely unknown, but recent surveillance suggests that it may represent up to 10% of the dengue burden in neotropical cities, or tens-of-thousands of cases per year throughout Latin America. The potential emergence of epizootic viruses from enzootic progenitors further highlights the need to strengthen surveillance activities, identify mosquito vectors and reservoirs and develop effective strategies to control the disease. In this article, we provide an overview of the current status of endemic VEE that results from spillover of the enzootic cycles, and we discuss public health measures for disease control as well as future avenues for VEE research.
Collapse
Affiliation(s)
- Patricia V Aguilar
- Center for Tropical Diseases, Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | |
Collapse
|