51
|
Animal Models of Lassa Fever. Pathogens 2020; 9:pathogens9030197. [PMID: 32155851 PMCID: PMC7157617 DOI: 10.3390/pathogens9030197] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/31/2022] Open
Abstract
Lassa virus (LASV), the causative agent of Lassa fever, is estimated to be responsible for up to 300,000 new infections and 5000 deaths each year across Western Africa. The most recent 2018 and 2019 Nigerian outbreaks featured alarmingly high fatality rates of up to 25.4%. In addition to the severity and high fatality of the disease, a significant population of survivors suffer from long-term sequelae, such as sensorineural hearing loss, resulting in a huge socioeconomic burden in endemic regions. There are no Food and Drug Administration (FDA)-approved vaccines, and therapeutics remain extremely limited for Lassa fever. Development of countermeasures depends on relevant animal models that can develop a disease strongly mimicking the pathogenic features of Lassa fever in humans. The objective of this review is to evaluate the currently available animal models for LASV infection with an emphasis on their pathogenic and histologic characteristics as well as recent advances in the development of a suitable rodent model. This information may facilitate the development of an improved animal model for understanding disease pathogenesis of Lassa fever and for vaccine or antiviral testing.
Collapse
|
52
|
Ibrahim AEC, van Dolleweerd CJ, Drake PMW, Ma JKC. Development of a minigenome cassette for Lettuce necrotic yellows virus: A first step in rescuing a plant cytorhabdovirus. PLoS One 2020; 15:e0229877. [PMID: 32134974 PMCID: PMC7058326 DOI: 10.1371/journal.pone.0229877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/15/2020] [Indexed: 12/12/2022] Open
Abstract
Rhabdoviruses are enveloped negative-sense RNA viruses that have numerous biotechnological applications. However, recovering plant rhabdoviruses from cDNA remains difficult due to technical difficulties such as the need for concurrent in planta expression of the viral genome together with the viral nucleoprotein (N), phosphoprotein (P) and RNA-dependent RNA polymerase (L) and viral genome instability in E. coli. Here, we developed a negative-sense minigenome cassette for Lettuce necrotic yellows virus (LNYV). We introduced introns into the unstable viral ORF and employed Agrobacterium tumefaciens to co-infiltrate Nicotiana with the genes for the N, P, and L proteins together with the minigenome cassette. The minigenome cassette included the Discosoma sp. red fluorescent protein gene (DsRed) cloned in the negative-sense between the viral trailer and leader sequences which were placed between hammerhead and hepatitis delta ribozymes. In planta DsRed expression was demonstrated by western blotting while the appropriate splicing of introduced introns was confirmed by sequencing of RT-PCR product.
Collapse
Affiliation(s)
- Ahmad E. C. Ibrahim
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Craig J. van Dolleweerd
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Pascal M. W. Drake
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Julian K-C. Ma
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| |
Collapse
|
53
|
Monoclonal Antibodies with Neutralizing Activity and Fc-Effector Functions against the Machupo Virus Glycoprotein. J Virol 2020; 94:JVI.01741-19. [PMID: 31801871 DOI: 10.1128/jvi.01741-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/01/2019] [Indexed: 12/17/2022] Open
Abstract
Machupo virus (MACV), the causative agent of Bolivian hemorrhagic fever (BHF), is a New World arenavirus that was first isolated in Bolivia from a human spleen in 1963. Due to the lack of a specific vaccine or therapy, this virus is considered a major risk to public health and is classified as a category A priority pathogen by the U.S. National Institutes of Health. In this study, we used DNA vaccination against the MACV glycoprotein precursor complex (GPC) and murine hybridoma technology to generate 25 mouse monoclonal antibodies (MAbs) against the GPC of MACV. Out of 25 MAbs, five were found to have potent neutralization activity in vitro against a recombinant vesicular stomatitis virus expressing MACV GPC (VSV-MACV) as well as against authentic MACV. Furthermore, the five neutralizing MAbs exhibited strong antibody-dependent cellular cytotoxicity (ADCC) activity in a reporter assay. When tested in vivo using VSV-MACV in a Stat2-/- mouse model, three MAbs significantly lowered viral loads in the spleen. Our work provides valuable insights into epitopes targeted by neutralizing antibodies that could be potent targets for vaccines and therapeutics and shed light on the importance of effector functions in immunity against MACV.IMPORTANCE MACV infections are a significant public health concern and lead to high case fatality rates. No specific treatment or vaccine for MACV infections exist. However, cases of Junin virus infection, a related virus, can be treated with convalescent-phase serum. This indicates that a MAb-based therapy for MACV could be effective. Here, we describe several MAbs that neutralize MACV and could be used for this purpose.
Collapse
|
54
|
Cross RW, Xu R, Matassov D, Hamm S, Latham TE, Gerardi CS, Nowak RM, Geisbert JB, Ota-Setlik A, Agans KN, Luckay A, Witko SE, Soukieh L, Deer DJ, Mire CE, Feldmann H, Happi C, Fenton KA, Eldridge JH, Geisbert TW. Quadrivalent VesiculoVax vaccine protects nonhuman primates from viral-induced hemorrhagic fever and death. J Clin Invest 2020; 130:539-551. [PMID: 31820871 PMCID: PMC6934204 DOI: 10.1172/jci131958] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/10/2019] [Indexed: 02/04/2023] Open
Abstract
Recent occurrences of filoviruses and the arenavirus Lassa virus (LASV) in overlapping endemic areas of Africa highlight the need for a prophylactic vaccine that would confer protection against all of these viruses that cause lethal hemorrhagic fever (HF). We developed a quadrivalent formulation of VesiculoVax that contains recombinant vesicular stomatitis virus (rVSV) vectors expressing filovirus glycoproteins and that also contains a rVSV vector expressing the glycoprotein of a lineage IV strain of LASV. Cynomolgus macaques were vaccinated twice with the quadrivalent formulation, followed by challenge 28 days after the boost vaccination with each of the 3 corresponding filoviruses (Ebola, Sudan, Marburg) or a heterologous contemporary lineage II strain of LASV. Serum IgG and neutralizing antibody responses specific for all 4 glycoproteins were detected in all vaccinated animals. A modest and balanced cell-mediated immune response specific for the glycoproteins was also detected in most of the vaccinated macaques. Regardless of the level of total glycoprotein-specific immune response detected after vaccination, all immunized animals were protected from disease and death following lethal challenges. These findings indicate that vaccination with attenuated rVSV vectors each expressing a single HF virus glycoprotein may provide protection against those filoviruses and LASV most commonly responsible for outbreaks of severe HF in Africa.
Collapse
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | | | - Stefan Hamm
- Department of Viral Vaccine Discovery, Profectus BioSciences Inc., Pearl River, New York, USA
| | | | | | - Rebecca M. Nowak
- Department of Viral Vaccine Discovery, Profectus BioSciences Inc., Pearl River, New York, USA
| | - Joan B. Geisbert
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Krystle N. Agans
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | | | | | - Daniel J. Deer
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Chad E. Mire
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | - Christian Happi
- Department of Biological Sciences and African Center of Excellence for Genomics of Infectious Diseases, Redeemer’s University, Edo, Nigeria
| | - Karla A. Fenton
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - John H. Eldridge
- Department of Immunology
- Department of Viral Vaccine Development, and
- Department of Viral Vaccine Discovery, Profectus BioSciences Inc., Pearl River, New York, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
55
|
Mateo M, Reynard S, Carnec X, Journeaux A, Baillet N, Schaeffer J, Picard C, Legras-Lachuer C, Allan R, Perthame E, Hillion KH, Pietrosemoli N, Dillies MA, Barrot L, Vallve A, Barron S, Fellmann L, Gaillard JC, Armengaud J, Carbonnelle C, Raoul H, Tangy F, Baize S. Vaccines inducing immunity to Lassa virus glycoprotein and nucleoprotein protect macaques after a single shot. Sci Transl Med 2019; 11:11/512/eaaw3163. [DOI: 10.1126/scitranslmed.aaw3163] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/16/2019] [Accepted: 08/02/2019] [Indexed: 01/01/2023]
Abstract
Lassa fever is a major threat in Western Africa. The large number of people living at risk for this disease calls for the development of a vaccine against Lassa virus (LASV). We generated live-attenuated LASV vaccines based on measles virus and Mopeia virus platforms and expressing different LASV antigens, with the aim to develop a vaccine able to protect after a single shot. We compared the efficacy of these vaccines against LASV in cynomolgus monkeys. The vaccines were well tolerated and protected the animals from LASV infection and disease after a single immunization but with varying efficacy. Analysis of the immune responses showed that complete protection was associated with robust secondary T cell and antibody responses against LASV. Transcriptomic and proteomic analyses showed an early activation of innate immunity and T cell priming after immunization with the most effective vaccines, with changes detectable as early as 2 days after immunization. The most efficacious vaccine candidate, a measles vector simultaneously expressing LASV glycoprotein and nucleoprotein, has been selected for further clinical evaluation.
Collapse
|
56
|
Kainulainen MH, Spengler JR, Welch SR, Coleman-McCray JD, Harmon JR, Klena JD, Nichol ST, Albariño CG, Spiropoulou CF. Use of a Scalable Replicon-Particle Vaccine to Protect Against Lethal Lassa Virus Infection in the Guinea Pig Model. J Infect Dis 2019; 217:1957-1966. [PMID: 29800368 DOI: 10.1093/infdis/jiy123] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/01/2018] [Indexed: 11/14/2022] Open
Abstract
Lassa fever is a viral zoonosis that can be transmitted from person to person, especially in the hospital setting. The disease is endemic to several countries in West Africa and can be a major contributor to morbidity and mortality in affected areas. There are no approved vaccines to prevent Lassa virus infection. In this work, we present a vaccine candidate that combines the scalability and efficacy benefits of a live vaccine with the safety benefits of single-cycle replication. The system consists of Lassa virus replicon particles devoid of the virus essential glycoprotein gene, and a cell line that expresses the glycoprotein products, enabling efficient vaccine propagation. Guinea pigs vaccinated with these particles showed no clinical reaction to the inoculum and were protected against fever, weight loss, and lethality after infection with Lassa virus.
Collapse
Affiliation(s)
- Markus H Kainulainen
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - JoAnn D Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica R Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - John D Klena
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - César G Albariño
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
57
|
Fathi A, Dahlke C, Addo MM. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum Vaccin Immunother 2019; 15:2269-2285. [PMID: 31368826 PMCID: PMC6816421 DOI: 10.1080/21645515.2019.1649532] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The devastating Ebola virus (EBOV) outbreak in West Africa in 2013-2016 has flagged the need for the timely development of vaccines for high-threat pathogens. To be better prepared for new epidemics, the WHO has compiled a list of priority pathogens that are likely to cause future outbreaks and for which R&D efforts are, therefore, paramount (R&D Blueprint: https://www.who.int/blueprint/priority-diseases/en/ ). To this end, the detailed characterization of vaccine platforms is needed. The vesicular stomatitis virus (VSV) has been established as a robust vaccine vector backbone for infectious diseases for well over a decade. The recent clinical trials testing the vaccine candidate VSV-EBOV against EBOV disease now have added a substantial amount of clinical data and suggest VSV to be an ideal vaccine vector candidate for outbreak pathogens. In this review, we discuss insights gained from the clinical VSV-EBOV vaccine trials as well as from animal studies investigating vaccine candidates for Blueprint pathogens.
Collapse
Affiliation(s)
- Anahita Fathi
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Christine Dahlke
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Marylyn M Addo
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| |
Collapse
|
58
|
Salvato MS, Domi A, Guzmán-Cardozo C, Medina-Moreno S, Zapata JC, Hsu H, McCurley N, Basu R, Hauser M, Hellerstein M, Guirakhoo F. A Single Dose of Modified Vaccinia Ankara Expressing Lassa Virus-like Particles Protects Mice from Lethal Intra-cerebral Virus Challenge. Pathogens 2019; 8:E133. [PMID: 31466243 PMCID: PMC6789566 DOI: 10.3390/pathogens8030133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022] Open
Abstract
Lassa fever surpasses Ebola, Marburg, and all other hemorrhagic fevers except Dengue in its public health impact. Caused by Lassa virus (LASV), the disease is a scourge on populations in endemic areas of West Africa, where reported incidence is higher. Here, we report construction, characterization, and preclinical efficacy of a novel recombinant vaccine candidate GEO-LM01. Constructed in the Modified Vaccinia Ankara (MVA) vector, GEO-LM01 expresses the glycoprotein precursor (GPC) and zinc-binding matrix protein (Z) from the prototype Josiah strain lineage IV. When expressed together, GP and Z form Virus-Like Particles (VLPs) in cell culture. Immunogenicity and efficacy of GEO-LM01 was tested in a mouse challenge model. A single intramuscular dose of GEO-LM01 protected 100% of CBA/J mice challenged with a lethal dose of ML29, a Mopeia/Lassa reassortant virus, delivered directly into the brain. In contrast, all control animals died within one week. The vaccine induced low levels of antibodies but Lassa-specific CD4+ and CD8+ T cell responses. This is the first report showing that a single dose of a replication-deficient MVA vector can confer full protection against a lethal challenge with ML29 virus.
Collapse
Affiliation(s)
- Maria S Salvato
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | | - Juan Carlos Zapata
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Haoting Hsu
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Nathanael McCurley
- Office of Technology Licensing and Commercialization, Georgia State University, Atlanta, GA 30303, USA
| | - Rahul Basu
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | | | | | |
Collapse
|
59
|
Tang-Huau TL, Feldmann H, Rosenke K. Animal models for Lassa virus infection. Curr Opin Virol 2019; 37:112-117. [PMID: 31442921 DOI: 10.1016/j.coviro.2019.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/20/2022]
Abstract
In humans, Lassa virus infection can result in disease with hemorrhagic manifestations and high fatality rates. There are no approved treatments or vaccines available and the inherent danger of studying Lassa virus means it can only be studied in high containment labs (BSL4). Under these conditions, mouse models are becoming an important instrument in the study of Lassa virus infection, disease and host responses. While guinea pigs and non-human primates are the critical components in assessing treatments and vaccines and have recently been used with great affect in this capacity.
Collapse
Affiliation(s)
- Tsing-Lee Tang-Huau
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - H Feldmann
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA; Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - K Rosenke
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA.
| |
Collapse
|
60
|
Monath TP. A short history of Lassa fever: the first 10-15 years after discovery. Curr Opin Virol 2019; 37:77-83. [PMID: 31323506 DOI: 10.1016/j.coviro.2019.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/06/2019] [Indexed: 01/10/2023]
Abstract
This brief review is focused on the events surrounding the initial discovery of a new viral hemorrhagic fever in 1969 and the subsequent 10-15 years during which a substantial understanding of the disease was gained. In 1969, a series of sequential life-threating or fatal infections occurred among health care workers in Nigeria and the laboratory scientist who isolated and characterized the causative agent. The agent, Lassa virus was named after the geographical location of the first recognized human case. The new virus was shown to be related to lymphocytic choriomeningitis and to previously unclassified neotropical viruses, including Argentine and Bolivian hemorrhagic fevers, and a new taxonomic grouping, the Arenaviruses, was proposed. In 1970-72, three further epidemics occurred in Nigeria, Liberia and Sierra Leone, the first two involved nosocomial transmission, and the third was a community-based outbreak, during which the rodent reservoir host was identified. In 1976, a long-term research project commenced in Sierra Leone, which produced a rich body of data from prospectively designed studies on the clinical features, transmission, and treatment of the disease.
Collapse
Affiliation(s)
- Thomas P Monath
- Crozet BioPharma LLC, 94 Jackson Road Suite 108, Devens, MA 01434, United States.
| |
Collapse
|
61
|
Vesicular Stomatitis Virus-Based Vaccines Provide Cross-Protection against Andes and Sin Nombre Viruses. Viruses 2019; 11:v11070645. [PMID: 31337019 PMCID: PMC6669828 DOI: 10.3390/v11070645] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/24/2019] [Accepted: 07/12/2019] [Indexed: 01/13/2023] Open
Abstract
Andes virus (ANDV) and Sin Nombre virus (SNV) are the main causative agents responsible for hantavirus cardiopulmonary syndrome (HCPS) in the Americas. HCPS is a severe respiratory disease with a high fatality rate for which there are no approved therapeutics or vaccines available. Some vaccine approaches for HCPS have been tested in preclinical models, but none have been tested in infectious models in regard to their ability to protect against multiple species of HCPS-causing viruses. Here, we utilize recombinant vesicular stomatitis virus-based (VSV) vaccines for Andes virus (ANDV) and Sin Nombre virus (SNV) and assess their ability to provide cross-protection in infectious challenge models. We show that, while both rVSVΔG/ANDVGPC and rVSVΔG/SNVGPC display attenuated growth as compared to wild type VSV, each vaccine is able to induce a cross-reactive antibody response. Both vaccines protected against both homologous and heterologous challenge with ANDV and SNV and prevented HCPS in a lethal ANDV challenge model. This study provides evidence that the development of a single vaccine against HCPS-causing hantaviruses could provide protection against multiple agents.
Collapse
|
62
|
Jiang J, Banglore P, Cashman KA, Schmaljohn CS, Schultheis K, Pugh H, Nguyen J, Humeau LM, Broderick KE, Ramos SJ. Immunogenicity of a protective intradermal DNA vaccine against lassa virus in cynomolgus macaques. Hum Vaccin Immunother 2019; 15:2066-2074. [PMID: 31071008 PMCID: PMC6773375 DOI: 10.1080/21645515.2019.1616499] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Lassa virus (LASV) is a hemorrhagic fever virus of the Arenaviridae family with high rates of mortality and co-morbidities, including chronic seizures and permanent bilateral or unilateral deafness. LASV is endemic in West Africa and Lassa fever accounts for 10-16% of hospitalizations annually in parts of Sierra Leone and Liberia according to the CDC. An ongoing outbreak in Nigeria has resulted in 144 deaths in 568 cases confirmed as LASV as of November 2018, with many more suspected, highlighting the urgent need for a vaccine to prevent this severe disease. We previously reported on a DNA vaccine encoding a codon-optimized LASV glycoprotein precursor gene, pLASV-GPC, which completely protects Guinea pigs and nonhuman primates (NHPs) against viremia, clinical disease, and death following lethal LASV challenge. Herein we report on the immunogenicity profile of the LASV DNA vaccine in protected NHPs. Antigen-specific binding antibodies were generated in 100% (6/6) NHPs after two immunizations with pLASV-GPC. These antibodies bound predominantly to the assembled LASV glycoprotein complex and had robust neutralizing activity in a pseudovirus assay. pLASV-GPC DNA-immunized NHPs (5/6) also developed T cell responses as measured by IFNγ ELISpot assay. These results revealed that the pLASV-GPC DNA vaccine is capable of generating functional, LASV-specific T cell and antibody responses, and the assays developed in this study will provide a framework to identify correlates of protection and characterize immune responses in future clinical trials.
Collapse
Affiliation(s)
- Jingjing Jiang
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA
| | - Preeti Banglore
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA
| | - Kathleen A. Cashman
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Connie S. Schmaljohn
- Office of the Chief Scientists, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | | | - Holly Pugh
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA
| | - Jacklyn Nguyen
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA
| | - Laurent M. Humeau
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA
| | - Kate E. Broderick
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA
| | - Stephanie J. Ramos
- Research & Development, Inovio Pharmaceuticals Inc, Plymouth Meeting, PA, USA,CONTACT Stephanie J. Ramos 10480 Wateridge Circle, San Diego, CA 92121, USA
| |
Collapse
|
63
|
Stein DR, Sroga P, Warner BM, Deschambault Y, Poliquin G, Safronetz D. Evaluating Temperature Sensitivity of Vesicular Stomatitis Virus-Based Vaccines. Emerg Infect Dis 2019; 25:1563-1566. [PMID: 31141474 PMCID: PMC6649338 DOI: 10.3201/eid2508.190281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Use of the vesicular stomatitis virus (VSV)-based Ebola virus vaccine during outbreaks and the potential use of a similar VSV-based Lassa virus vaccine has raised questions about the vaccines' stability should the cold chain fail. We demonstrated that current cold chain conditions might tolerate significant variances without affecting efficacy.
Collapse
|
64
|
Vaccine platforms for the prevention of Lassa fever. Immunol Lett 2019; 215:1-11. [PMID: 31026485 PMCID: PMC7132387 DOI: 10.1016/j.imlet.2019.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 12/19/2022]
Abstract
The epidemiological significance of Lassa fever in West Africa is discussed. Viral ecology, pathology, and immunobiology of Lassa virus infection is described. Multiple vaccine candidates have been tested in pre-clinical models. Lassa fever vaccine candidates have yet to progress to clinical trials. Five platform technologies have been selected for expedited development.
Lassa fever is an acute viral haemorrhagic illness caused by Lassa virus (LASV), which is endemic throughout much of West Africa. The virus primarily circulates in the Mastomys natalensis reservoir and is transmitted to humans through contact with infectious rodents or their secretions; human-to-human transmission is documented as well. With the exception of Dengue fever, LASV has the highest human impact of any haemorrhagic fever virus. On-going outbreaks in Nigeria have resulted in unprecedented mortality. Consequently, the World Health Organization (WHO) has listed LASV as a high priority pathogen for the development of treatments and prophylactics. Currently, there are no licensed vaccines to protect against LASV infection. Although numerous candidates have demonstrated efficacy in animal models, to date, only a single candidate has advanced to clinical trials. Lassa fever vaccine development efforts have been hindered by the high cost of biocontainment requirements, the absence of established correlates of protection, and uncertainty regarding the extent to which animal models are predictive of vaccine efficacy in humans. This review briefly discusses the epidemiology and biology of LASV infection and highlights recent progress in vaccine development.
Collapse
|
65
|
Genzer SC, Huynh T, Coleman-Mccray JD, Harmon JR, Welch SR, Spengler JR. Hematology and Clinical Chemistry Reference Intervals for Inbred Strain 13/n Guinea Pigs ( Cavia Porcellus ). JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2019; 58:293-303. [PMID: 31010455 DOI: 10.30802/aalas-jaalas-18-000118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Inbred Strain 13/N Guinea Pigs are Frequently Used As Animal Models in Studies of Emerging and High-pathogenicity Viruses. To Date, Clinical Reference Intervals Have Not Been Established for Hematology and Clinical Chemistry Parameters in This Strain. We Obtained Whole-blood Samples from the Cranial Vena Cava of Healthy Strain 13/N Colony Animals for Inhouse Cbc and Clinical Chemistry Analyses. Analyte Values Were Investigated to Determine Subpopulation Differences According to Age and Sex. Glucose, Albumin, Alp, Lymphocyte Percentage, Hgb, and Mchc Decreased with Age, Whereas Neutrophil and Monocyte Percentages, Bun, Creatinine, Calcium, and Amylase Increased with Age. Total Protein and Wbc Counts Increased Over the First 300 D of Life Before Stabilizing. Across All Age Categories, Female Guinea Pigs Consistently Had Lower Rbc, Hct, Hgb, Alt, Alp, and Amylase Levels and Higher Mcv Values Than Males. These Trends Were Strongest in Adults (age, 151 Through 900 D). Most Parameters Stabilized by 300 D; Previous Studies Used 60 D or 120 D As Adult Age and 90 to 120 D As Sexual Maturity. We Recommend Age Group Definitions of 0 Through 150 D for Juveniles, 151 Through 900 D for Adults, and Older Than 900 D for Geriatric Adult Strain 13/N Guinea Pigs.
Collapse
Affiliation(s)
- Sarah C Genzer
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, Georgia;,
| | - Thanhthao Huynh
- Infectious Diseases Pathology Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Joann D Coleman-Mccray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica R Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
66
|
Stein DR, Warner BM, Soule G, Tierney K, Frost KL, Booth S, Safronetz D. A recombinant vesicular stomatitis-based Lassa fever vaccine elicits rapid and long-term protection from lethal Lassa virus infection in guinea pigs. NPJ Vaccines 2019; 4:8. [PMID: 30774999 PMCID: PMC6368541 DOI: 10.1038/s41541-019-0104-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022] Open
Abstract
The World Health Organization has identified Lassa virus (LASV) as one of the top five pathogens to cause a severe outbreak in the near future. This study assesses the ability of a leading vaccine candidate, recombinant Vesicular stomatitis virus expressing LASV glycoprotein (VSVΔG/LASVGPC), and its ability to induce rapid and long-term immunity to lethal guinea pig-adapted LASV (GPA-LASV). Outbred guinea pigs were vaccinated with a single dose of VSVΔG/LASVGPC followed by a lethal challenge of GPA-LASV at 7, 14, 25, 189, and 355 days post-vaccination. Statistically significant rapid and long-term protection was achieved at all time points with 100% protection at days 7 and 14 post-vaccination. While 83 and 87% protection were achieved at 25 days and 6 months post-vaccination, respectively. When guinea pigs were challenged one year after vaccination 71% protection was achieved. Notable infectious virus was isolated from the serum and tissues of some but not all animals. Total LASVGPC-specific IgG titers were also measured on a monthly basis leading up to LASV challenge however, it is unclear if antibody alone correlates with short and long term survival. These studies confirm that a single dose of VSVΔG/LASVGPC can induce rapid and long-term protection from LASV infection in an aggressive outbred model of infection, and supports further development in non-human primates. Lassa virus (LASV) is an emerging pathogen that can be associated with high case fatality but for which no clinically-approved vaccine currently exists. David Safronetz and colleagues at the Public Health Agency of Canada and the University of Manitoba investigate the efficacy of a single dose of a recombinant vaccine of LASV glycoproteins vectorized into vesicular stomatitis virus (VSVΔG/LASVGPC). Using guinea pigs lethally challenged with LASV, the protective efficacy of VSVΔG/LASVGPC and LASV-specific IgG is assessed at a number of time points out to approximately one year after vaccination. VSVΔG/LASVGPC elicits stable LASV glycoprotein-specific antibody production and durable protection from lethal LASV challenge, with 71% of animals surviving even at one year following vaccination and complete protection being afforded at earlier (weeks) time points. This pre-clinical model demonstrates the stable protection that can be established by a single dose of VSVΔG/LASVGPC.
Collapse
Affiliation(s)
- Derek R Stein
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada
| | - Bryce M Warner
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada.,2Department of Medical Microbiology, University of Manitoba, Winnipeg, MB Canada
| | - Geoff Soule
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada
| | - Kevin Tierney
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada
| | - Kathy L Frost
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada
| | - Stephanie Booth
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada
| | - David Safronetz
- 1Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB Canada.,2Department of Medical Microbiology, University of Manitoba, Winnipeg, MB Canada
| |
Collapse
|
67
|
Mazzola LT, Kelly-Cirino C. Diagnostics for Lassa fever virus: a genetically diverse pathogen found in low-resource settings. BMJ Glob Health 2019; 4:e001116. [PMID: 30899575 PMCID: PMC6407561 DOI: 10.1136/bmjgh-2018-001116] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 11/18/2022] Open
Abstract
Lassa fever virus (LASV) causes acute viral haemorrhagic fever with symptoms similar to those seen with Ebola virus infections. LASV is endemic to West Africa and is transmitted through contact with excretions of infected Mastomys natalensis rodents and other rodent species. Due to a high fatality rate, lack of treatment options and difficulties with prevention and control, LASV is one of the high-priority pathogens included in the WHO R&D Blueprint. The WHO LASV vaccine strategy relies on availability of effective diagnostic tests. Current diagnostics for LASV include in-house and commercial (primarily research-only) laboratory-based serological and nucleic acid amplification tests. There are two commercially available (for research use only) rapid diagnostic tests (RDTs), and a number of multiplex panels for differential detection of LASV infection from other endemic diseases with similar symptoms have been evaluated. However, a number of diagnostic gaps remain. Lineage detection is a challenge due to the genomic diversity of LASV, as pan-lineage sensitivity for both molecular and immunological detection is necessary for surveillance and outbreak response. While pan-lineage ELISA and RDTs are commercially available (for research use only), validation and external quality assessment (EQA) is needed to confirm detection sensitivity for all known or relevant strains. Variable sensitivity of LASV PCR tests also highlights the need for improved validation and EQA. Given that LASV outbreaks typically occur in low-resource settings, more options for point-of-care testing would be valuable. These requirements should be taken into account in target product profiles for improved LASV diagnostics.
Collapse
|
68
|
Lukashevich IS, Paessler S, de la Torre JC. Lassa virus diversity and feasibility for universal prophylactic vaccine. F1000Res 2019; 8. [PMID: 30774934 PMCID: PMC6357994 DOI: 10.12688/f1000research.16989.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Lassa virus (LASV) is a highly prevalent mammarenavirus in West Africa and is maintained in nature in a persistently infected rodent host, Mastomys natalensis, which is widely spread in sub-Saharan Africa. LASV infection of humans can cause Lassa fever (LF), a disease associated with high morbidity and significant mortality. Recent evidence indicates an LASV expansion outside its traditional endemic areas. In 2017, the World Health Organization (WHO) included LASV in top-priority pathogens and released a Target Product Profile (TPP) for vaccine development. Likewise, in 2018, the US Food and Drug Administration added LF to a priority review voucher program to encourage the development of preventive and therapeutics measures. In this article, we review recent progress in LASV vaccine research and development with a focus on the impact of LASV genetic and biological diversity on the design and development of vaccine candidates meeting the WHO's TPP for an LASV vaccine.
Collapse
Affiliation(s)
- Igor S Lukashevich
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40292, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology IMM-6, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
69
|
CSV2018: The 2nd Symposium of the Canadian Society for Virology. Viruses 2019; 11:v11010079. [PMID: 30669273 PMCID: PMC6356965 DOI: 10.3390/v11010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 11/16/2022] Open
Abstract
The 2nd Symposium of the Canadian Society for Virology (CSV2018) was held in June 2018 in Halifax, Nova Scotia, Canada, as a featured event marking the 200th anniversary of Dalhousie University. CSV2018 attracted 175 attendees from across Canada and around the world, more than double the number that attended the first CSV symposium two years earlier. CSV2018 provided a forum to discuss a wide range of topics in virology including human, veterinary, plant, and microbial pathogens. Invited keynote speakers included David Kelvin (Dalhousie University and Shantou University Medical College) who provided a historical perspective on influenza on the 100th anniversary of the 1918 pandemic; Sylvain Moineau (Université Laval) who described CRISPR-Cas systems and anti-CRISPR proteins in warfare between bacteriophages and their host microbes; and Kate O’Brien (then from Johns Hopkins University, now relocated to the World Health Organization where she is Director of Immunization, Vaccines and Biologicals), who discussed the underlying viral etiology for pneumonia in the developing world, and the evidence for respiratory syncytial virus (RSV) as a primary cause. Reflecting a strong commitment of Canadian virologists to science communication, CSV2018 featured the launch of Halifax’s first annual Soapbox Science event to enable public engagement with female scientists, and the live-taping of the 499th episode of the This Week in Virology (TWIV) podcast, hosted by Vincent Racaniello (Columbia University) and science writer Alan Dove. TWIV featured interviews of CSV co-founders Nathalie Grandvaux (Université de Montréal) and Craig McCormick (Dalhousie University), who discussed the origins and objectives of the new society; Ryan Noyce (University of Alberta), who discussed technical and ethical considerations of synthetic virology; and Kate O’Brien, who discussed vaccines and global health. Finally, because CSV seeks to provide a better future for the next generation of Canadian virologists, the symposium featured a large number of oral and poster presentations from trainees and closed with the awarding of presentation prizes to trainees, followed by a tour of the Halifax Citadel National Historic Site and an evening of entertainment at the historic Alexander Keith’s Brewery.
Collapse
|
70
|
Ibrahim A, Odon V, Kormelink R. Plant Viruses in Plant Molecular Pharming: Toward the Use of Enveloped Viruses. FRONTIERS IN PLANT SCIENCE 2019; 10:803. [PMID: 31275344 PMCID: PMC6594412 DOI: 10.3389/fpls.2019.00803] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/04/2019] [Indexed: 05/03/2023]
Abstract
Plant molecular pharming has emerged as a reliable platform for recombinant protein expression providing a safe and low-cost alternative to bacterial and mammalian cells-based systems. Simultaneously, plant viruses have evolved from pathogens to molecular tools for recombinant protein expression, chimaeric viral vaccine production, and lately, as nanoagents for drug delivery. This review summarizes the genesis of viral vectors and agroinfection, the development of non-enveloped viruses for various biotechnological applications, and the on-going research on enveloped plant viruses.
Collapse
|
71
|
Madu IG, Files M, Gharaibeh DN, Moore AL, Jung KH, Gowen BB, Dai D, Jones KF, Tyavanagimatt SR, Burgeson JR, Korth MJ, Bedard KM, Iadonato SP, Amberg SM. A potent Lassa virus antiviral targets an arenavirus virulence determinant. PLoS Pathog 2018; 14:e1007439. [PMID: 30576397 PMCID: PMC6322784 DOI: 10.1371/journal.ppat.1007439] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 01/07/2019] [Accepted: 10/25/2018] [Indexed: 11/25/2022] Open
Abstract
Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses. Lassa fever is a viral hemorrhagic fever disease that is transmitted to humans primarily through contact with the urine or feces of infected rodents. The disease is endemic in West Africa, and an unusually large outbreak occurred in Nigeria in early 2018. The case fatality rate was 25% among confirmed cases, underscoring the need for an effective antiviral therapy. Here, we evaluated the small-molecule drug LHF-535, which targets the arenavirus envelope glycoprotein, for broad-spectrum activity against Lassa viruses of different lineages and related arenaviruses that cause hemorrhagic fever diseases in South America. We also selected for LHF-535-resistant viruses and characterized their genotype and phenotype. Using a combination of surrogate systems and wild-type viruses, we determined that all tested Lassa virus strains and New World hemorrhagic fever arenaviruses were sensitive to LHF-535. Sensitivity to the drug was modulated by specific amino acid changes in the viral envelope glycoprotein, and the majority of emerging drug-resistant viruses were attenuated for virulence. Similarly, the live-attenuated vaccine strain for Argentine hemorrhagic fever was also resistant to LHF-535. These findings indicate that LHF-535 targets a viral virulence determinant, the mutation of which may result in the emergence of drug-resistant viruses, but with reduced capacity for virulence.
Collapse
Affiliation(s)
- Ikenna G. Madu
- Kineta, Inc., Seattle, Washington, United States of America
| | - Megan Files
- Kineta, Inc., Seattle, Washington, United States of America
| | - Dima N. Gharaibeh
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | - Amy L. Moore
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | - Kie-Hoon Jung
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Brian B. Gowen
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Dongcheng Dai
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | - Kevin F. Jones
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | | | - James R. Burgeson
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | | | | | | | - Sean M. Amberg
- Kineta, Inc., Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
72
|
Lundstrom K. Self-Replicating RNA Viruses for RNA Therapeutics. Molecules 2018; 23:molecules23123310. [PMID: 30551668 PMCID: PMC6321401 DOI: 10.3390/molecules23123310] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
Self-replicating single-stranded RNA viruses such as alphaviruses, flaviviruses, measles viruses, and rhabdoviruses provide efficient delivery and high-level expression of therapeutic genes due to their high capacity of RNA replication. This has contributed to novel approaches for therapeutic applications including vaccine development and gene therapy-based immunotherapy. Numerous studies in animal tumor models have demonstrated that self-replicating RNA viral vectors can generate antibody responses against infectious agents and tumor cells. Moreover, protection against challenges with pathogenic Ebola virus was obtained in primates immunized with alphaviruses and flaviviruses. Similarly, vaccinated animals have been demonstrated to withstand challenges with lethal doses of tumor cells. Furthermore, clinical trials have been conducted for several indications with self-amplifying RNA viruses. In this context, alphaviruses have been subjected to phase I clinical trials for a cytomegalovirus vaccine generating neutralizing antibodies in healthy volunteers, and for antigen delivery to dendritic cells providing clinically relevant antibody responses in cancer patients, respectively. Likewise, rhabdovirus particles have been subjected to phase I/II clinical trials showing good safety and immunogenicity against Ebola virus. Rhabdoviruses have generated promising results in phase III trials against Ebola virus. The purpose of this review is to summarize the achievements of using self-replicating RNA viruses for RNA therapy based on preclinical animal studies and clinical trials in humans.
Collapse
|
73
|
Caì Y, Iwasaki M, Beitzel BF, Yú S, Postnikova EN, Cubitt B, DeWald LE, Radoshitzky SR, Bollinger L, Jahrling PB, Palacios GF, de la Torre JC, Kuhn JH. Recombinant Lassa Virus Expressing Green Fluorescent Protein as a Tool for High-Throughput Drug Screens and Neutralizing Antibody Assays. Viruses 2018; 10:v10110655. [PMID: 30463334 PMCID: PMC6266387 DOI: 10.3390/v10110655] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 01/30/2023] Open
Abstract
Lassa virus (LASV), a mammarenavirus, infects an estimated 100,000–300,000 individuals yearly in western Africa and frequently causes lethal disease. Currently, no LASV-specific antivirals or vaccines are commercially available for prevention or treatment of Lassa fever, the disease caused by LASV. The development of medical countermeasure screening platforms is a crucial step to yield licensable products. Using reverse genetics, we generated a recombinant wild-type LASV (rLASV-WT) and a modified version thereof encoding a cleavable green fluorescent protein (GFP) as a reporter for rapid and quantitative detection of infection (rLASV-GFP). Both rLASV-WT and wild-type LASV exhibited similar growth kinetics in cultured cells, whereas growth of rLASV-GFP was slightly impaired. GFP reporter expression by rLASV-GFP remained stable over several serial passages in Vero cells. Using two well-characterized broad-spectrum antivirals known to inhibit LASV infection, favipiravir and ribavirin, we demonstrate that rLASV-GFP is a suitable screening tool for the identification of LASV infection inhibitors. Building on these findings, we established a rLASV-GFP-based high-throughput drug discovery screen and an rLASV-GFP-based antibody neutralization assay. Both platforms, now available as a standard tool at the IRF-Frederick (an international resource), will accelerate anti-LASV medical countermeasure discovery and reduce costs of antiviral screens in maximum containment laboratories.
Collapse
Affiliation(s)
- Yíngyún Caì
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| | - Masaharu Iwasaki
- Department of Immunology and Microbial Science, The Scripps Research Institute (TSRI), 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA.
| | - Brett F Beitzel
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA.
| | - Shuīqìng Yú
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| | - Elena N Postnikova
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| | - Beatrice Cubitt
- Department of Immunology and Microbial Science, The Scripps Research Institute (TSRI), 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA.
| | - Lisa Evans DeWald
- Integrated Research Facility at Fort Detrick (IRF-Frederick), 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.
| | - Laura Bollinger
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| | - Peter B Jahrling
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| | - Gustavo F Palacios
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA.
| | - Juan C de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute (TSRI), 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA.
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick (IRF-Frederick), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
74
|
Zapata JC, Medina-Moreno S, Guzmán-Cardozo C, Salvato MS. Improving the Breadth of the Host's Immune Response to Lassa Virus. Pathogens 2018; 7:E84. [PMID: 30373278 PMCID: PMC6313495 DOI: 10.3390/pathogens7040084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/20/2018] [Accepted: 10/24/2018] [Indexed: 12/21/2022] Open
Abstract
In 2017, the global Coalition for Epidemic Preparedness (CEPI) declared Lassa virus disease to be one of the world's foremost biothreats. In January 2018, World Health Organization experts met to address the Lassa biothreat. It was commonly recognized that the diversity of Lassa virus (LASV) isolated from West African patient samples was far greater than that of the Ebola isolates from the West African epidemic of 2013⁻2016. Thus, vaccines produced against Lassa virus disease face the added challenge that they must be broadly-protective against a wide variety of LASV. In this review, we discuss what is known about the immune response to Lassa infection. We also discuss the approaches used to make broadly-protective influenza vaccines and how they could be applied to developing broad vaccine coverage against LASV disease. Recent advances in AIDS research are also potentially applicable to the design of broadly-protective medical countermeasures against LASV disease.
Collapse
Affiliation(s)
- Juan Carlos Zapata
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Sandra Medina-Moreno
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Camila Guzmán-Cardozo
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Maria S Salvato
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
75
|
Abreu-Mota T, Hagen KR, Cooper K, Jahrling PB, Tan G, Wirblich C, Johnson RF, Schnell MJ. Non-neutralizing antibodies elicited by recombinant Lassa-Rabies vaccine are critical for protection against Lassa fever. Nat Commun 2018; 9:4223. [PMID: 30310067 PMCID: PMC6181965 DOI: 10.1038/s41467-018-06741-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/10/2018] [Indexed: 11/20/2022] Open
Abstract
Lassa fever (LF), caused by Lassa virus (LASV), is a viral hemorrhagic fever for which no approved vaccine or potent antiviral treatment is available. LF is a WHO priority disease and, together with rabies, a major health burden in West Africa. Here we present the development and characterization of an inactivated recombinant LASV and rabies vaccine candidate (LASSARAB) that expresses a codon-optimized LASV glycoprotein (coGPC) and is adjuvanted by a TLR-4 agonist (GLA-SE). LASSARAB elicits lasting humoral response against LASV and RABV in both mouse and guinea pig models, and it protects both guinea pigs and mice against LF. We also demonstrate a previously unexplored role for non-neutralizing LASV GPC-specific antibodies as a major mechanism of protection by LASSARAB against LF through antibody-dependent cellular functions. Overall, these findings demonstrate an effective inactivated LF vaccine and elucidate a novel humoral correlate of protection for LF.
Collapse
Affiliation(s)
- Tiago Abreu-Mota
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, 19107, USA
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, 4710-057, Portugal
| | - Katie R Hagen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick MD, 21702, USA
| | - Kurt Cooper
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick MD, 21702, USA
| | - Peter B Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick MD, 21702, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD, 20892, USA
| | - Gene Tan
- Infectious Disease, The J. Craig Venter Institute, La Jolla, CA, 92037, USA
- Department of Medicine, University of California, San Diego, La Jolla CA, 92037, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD, 20892, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, 19107, USA.
- Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
76
|
Warner BM, Safronetz D, Stein DR. Current research for a vaccine against Lassa hemorrhagic fever virus. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2519-2527. [PMID: 30147299 PMCID: PMC6097522 DOI: 10.2147/dddt.s147276] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lassa virus (LASV) is a rodent-borne arenavirus endemic to several West African countries that causes Lassa fever (LF). LF is typically mild but it can cause severe disease characterized by hemorrhagic fever and multi-organ failure. A current outbreak of LASV in Nigeria has seen greater than 300 cases with a case fatality rate of 22%. Currently, there are limited treatment options and no vaccine candidates are approved to prevent LASV infection. The Coalition for Epidemic Preparedness Innovations has identified LASV as an emerging pathogen of high consequence and this has resulted in a push for several preclinical vaccine candidates to be advanced toward clinical trials. Here, we discuss several important aspects of LASV infection including immunobiology, immune evasion, and correlates of protection against LF, which have been identified through animal models and human infections. In addition, we discuss several vaccine candidates that have shown efficacy in animal models that could be advanced toward clinical trials. The increased fatality rate seen in the recent LASV outbreak in Nigeria highlights the importance of developing effective treatment and prevention strategies against LF. The spike in LASV cases seen in West Africa has the potential for increased mortality and human-to-human transmission, making the development and testing of effective vaccines for LASV critical.
Collapse
Affiliation(s)
- Bryce M Warner
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - David Safronetz
- Zoonotic Diseases and Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada,
| | - Derek R Stein
- Zoonotic Diseases and Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada,
| |
Collapse
|
77
|
Li A, Yu J, Lu M, Ma Y, Attia Z, Shan C, Xue M, Liang X, Craig K, Makadiya N, He JJ, Jennings R, Shi PY, Peeples ME, Liu SL, Boyaka PN, Li J. A Zika virus vaccine expressing premembrane-envelope-NS1 polyprotein. Nat Commun 2018; 9:3067. [PMID: 30076287 PMCID: PMC6076265 DOI: 10.1038/s41467-018-05276-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 06/19/2018] [Indexed: 02/08/2023] Open
Abstract
Current efforts to develop Zika virus (ZIKV) subunit vaccines have been focused on pre-membrane (prM) and envelope (E) proteins, but the role of NS1 in ZIKV-specific immune response and protection is poorly understood. Here, we develop an attenuated recombinant vesicular stomatitis virus (rVSV)-based vaccine expressing ZIKV prM-E-NS1 as a polyprotein. This vectored vaccine candidate is attenuated in mice, where a single immunization induces ZIKV-specific antibody and T cell immune responses that provide protection against ZIKV challenge. Co-expression of prM, E, and NS1 induces significantly higher levels of Th2 and Th17 cytokine responses than prM-E. In addition, NS1 alone is capable of conferring partial protection against ZIKV infection in mice even though it does not induce neutralizing antibodies. These results demonstrate that attenuated rVSV co-expressing prM, E, and NS1 is a promising vaccine candidate for protection against ZIKV infection and highlights an important role for NS1 in ZIKV-specific cellular immune responses.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Genetic Vectors/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Polyproteins/genetics
- Polyproteins/immunology
- Th17 Cells/metabolism
- Th2 Cells/metabolism
- Vaccination
- Vaccines, Attenuated
- Vaccines, DNA/immunology
- Vaccines, Synthetic
- Vesiculovirus/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Nonstructural Proteins/genetics
- Viral Nonstructural Proteins/immunology
- Viral Nonstructural Proteins/metabolism
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Zika Virus/genetics
- Zika Virus/immunology
- Zika Virus/metabolism
- Zika Virus Infection/immunology
- Zika Virus Infection/prevention & control
Collapse
Affiliation(s)
- Anzhong Li
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Jingyou Yu
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
- Center for Retrovirus Research, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Yuanmei Ma
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Chao Shan
- Department of Biochemistry & Molecular Biology, Department of Pharmacology & Toxicology, and Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Miaoge Xue
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Kelsey Craig
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Nirajkumar Makadiya
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Jennifer J He
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Ryan Jennings
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, Department of Pharmacology & Toxicology, and Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, 370W. 9th Ave., Columbus, OH, 43210, USA
| | - Shan-Lu Liu
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
- Center for Retrovirus Research, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
- Infectious Diseases Institute, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
- Infectious Diseases Institute, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA.
- Infectious Diseases Institute, The Ohio State University, 1925 Coffey Road, Columbus, OH, 43210, USA.
| |
Collapse
|
78
|
Screening and Identification of Lassa Virus Entry Inhibitors from an FDA-Approved Drug Library. J Virol 2018; 92:JVI.00954-18. [PMID: 29899092 PMCID: PMC6069169 DOI: 10.1128/jvi.00954-18] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/25/2022] Open
Abstract
Lassa virus (LASV) belongs to the Mammarenavirus genus (family Arenaviridae) and causes severe hemorrhagic fever in humans. At present, there are no Food and Drug Administration (FDA)-approved drugs or vaccines specific for LASV. Here, high-throughput screening of an FDA-approved drug library was performed against LASV entry by using pseudotype virus bearing LASV envelope glycoprotein (GPC). Two hit compounds, lacidipine and phenothrin, were identified as LASV entry inhibitors in the micromolar range. A mechanistic study revealed that both compounds inhibited LASV entry by blocking low-pH-induced membrane fusion. Accordingly, lacidipine showed virucidal effects on the pseudotype virus of LASV. Adaptive mutant analyses demonstrated that replacement of T40, located in the ectodomain of the stable-signal peptide (SSP), with lysine (K) conferred LASV resistance to lacidipine. Furthermore, lacidipine showed antiviral activity against LASV, the closely related Mopeia virus (MOPV), and the New World arenavirus Guanarito virus (GTOV). Drug-resistant variants indicated that V36M in the ectodomain of the SSP mutant and V436A in the transmembrane domain of the GP2 mutant conferred GTOV resistance to lacidipine, suggesting the interface between SSP and GP2 is the target of lacidipine. This study shows that lacidipine is a candidate for LASV therapy, reinforcing the notion that the SSP-GP2 interface provides an entry-targeted platform for arenavirus inhibitor design.IMPORTANCE Currently, there is no approved therapy to treat Lassa fever; therefore, repurposing of approved drugs will accelerate the development of a therapeutic stratagem. In this study, we screened an FDA-approved library of drugs and identified two compounds, lacidipine and phenothrin, which inhibited Lassa virus entry by blocking low-pH-induced membrane fusion. Additionally, both compounds extended their inhibition against the entry of Guanarito virus, and the viral targets were identified as the SSP-GP2 interface.
Collapse
|
79
|
Welch SR, Scholte FEM, Albariño CG, Kainulainen MH, Coleman-McCray JD, Guerrero LW, Chakrabarti AK, Klena JD, Nichol ST, Spengler JR, Spiropoulou CF. The S Genome Segment Is Sufficient to Maintain Pathogenicity in Intra-Clade Lassa Virus Reassortants in a Guinea Pig Model. Front Cell Infect Microbiol 2018; 8:240. [PMID: 30050872 PMCID: PMC6050391 DOI: 10.3389/fcimb.2018.00240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/21/2018] [Indexed: 11/25/2022] Open
Abstract
Genome reassortment in Lassa virus (LASV) has been reported in nature, but phenotypic consequences of this phenomenon are not well described. Here we characterize, both in vitro and in vivo, reassortment between 2 LASV strains: the prototypic 1976 Josiah strain and a more recently isolated 2015 Liberian strain. In vitro analysis showed that although cis- and trans-acting elements of viral RNA synthesis were compatible between strains, reassortants demonstrated different levels of viral replication. These differences were also apparent in vivo, as reassortants varied in pathogenicity in the guinea pig model of LASV infection. The reassortant variant containing the Josiah S segment retained the virulence of the parental Josiah strain, but the reassortant variant containing the S segment of the Liberian isolate was highly attenuated compared to both parental strains. Contrary to observations in reassortants between LASV and other arenavirus species, which suggest that L segment-encoded factors are responsible for virulence, these studies highlight a role for S segment-encoded virulence factors in disease, and also suggest that inefficient interactions between proteins of heterologous strains may limit the prevalence of reassortant LASV variants in nature.
Collapse
Affiliation(s)
- Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Florine E M Scholte
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - César G Albariño
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Markus H Kainulainen
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - JoAnn D Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Lisa Wiggleton Guerrero
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Ayan K Chakrabarti
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - John D Klena
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| |
Collapse
|
80
|
Mendoza EJ, Warner B, Kobinger G, Ogden NH, Safronetz D. Baited vaccines: A strategy to mitigate rodent-borne viral zoonoses in humans. Zoonoses Public Health 2018; 65:711-727. [PMID: 29931738 DOI: 10.1111/zph.12487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/05/2018] [Accepted: 05/21/2018] [Indexed: 11/27/2022]
Abstract
Rodents serve as the natural reservoir and vector for a variety of pathogens, some of which are responsible for severe and life-threatening disease in humans. Despite the significant impact in humans many of these viruses, including Old and New World hantaviruses as well as Arenaviruses, most have no specific vaccine or therapeutic to treat or prevent human infection. The recent success of wildlife vaccines to mitigate rabies in animal populations offers interesting insight into the use of similar strategies for other zoonotic agents of human disease. In this review, we discuss the notion of using baited vaccines as a means to interrupt the transmission of viral pathogens between rodent reservoirs and to susceptible human hosts.
Collapse
Affiliation(s)
- Emelissa J Mendoza
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Bryce Warner
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gary Kobinger
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.,Centre Hospitalier de l'Université Laval, Quebec City, Quebec, Canada
| | - Nicholas H Ogden
- Public Health Risk Sciences Division, National Microbiology Laboratory, Public Health Agency of Canada, St-Hyacinthe, Quebec, Canada
| | - David Safronetz
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
81
|
Suder E, Furuyama W, Feldmann H, Marzi A, de Wit E. The vesicular stomatitis virus-based Ebola virus vaccine: From concept to clinical trials. Hum Vaccin Immunother 2018; 14:2107-2113. [PMID: 29757706 PMCID: PMC6183239 DOI: 10.1080/21645515.2018.1473698] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/30/2018] [Indexed: 10/25/2022] Open
Abstract
The devastating Ebola virus (EBOV) epidemic in West Africa in 2013-2016 accelerated the progress of several vaccines and antivirals through clinical trials, including the replication-competent vesicular stomatitis virus-based vaccine expressing the EBOV glycoprotein (VSV-EBOV). Extensive preclinical testing in animal models demonstrated the prophylactic and post-exposure efficacy of this vaccine, identified the mechanism of protection, and suggested it was safe for human use. Based on these data, VSV-EBOV was extensively tested in phase 1-3 clinical trials in North America, Europe and Africa. Although some side effects of vaccination were observed, these clinical trials showed that the VSV-EBOV was safe and immunogenic in humans. Moreover, the data supported the use of VSV-EBOV as an emergency vaccine in individuals at risk for Ebola virus disease. In this review, we summarize the results of the extensive preclinical and clinical testing of the VSV-EBOV vaccine.
Collapse
MESH Headings
- Animals
- Clinical Trials as Topic
- Drug Carriers
- Drug Evaluation, Preclinical
- Drug-Related Side Effects and Adverse Reactions/epidemiology
- Drug-Related Side Effects and Adverse Reactions/pathology
- Ebola Vaccines/administration & dosage
- Ebola Vaccines/genetics
- Ebola Vaccines/immunology
- Ebola Vaccines/isolation & purification
- Hemorrhagic Fever, Ebola/prevention & control
- Humans
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
- Vesiculovirus/genetics
Collapse
Affiliation(s)
- Ellen Suder
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Wakako Furuyama
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| |
Collapse
|
82
|
Carnec X, Mateo M, Page A, Reynard S, Hortion J, Picard C, Yekwa E, Barrot L, Barron S, Vallve A, Raoul H, Carbonnelle C, Ferron F, Baize S. A Vaccine Platform against Arenaviruses Based on a Recombinant Hyperattenuated Mopeia Virus Expressing Heterologous Glycoproteins. J Virol 2018; 92:e02230-17. [PMID: 29593043 PMCID: PMC5974477 DOI: 10.1128/jvi.02230-17] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/22/2018] [Indexed: 11/20/2022] Open
Abstract
Several Old World and New World arenaviruses are responsible for severe endemic and epidemic hemorrhagic fevers, whereas other members of the Arenaviridae family are nonpathogenic. To date, no approved vaccines, antivirals, or specific treatments are available, except for Junín virus. However, protection of nonhuman primates against Lassa fever virus (LASV) is possible through the inoculation of the closely related but nonpathogenic Mopeia virus (MOPV) before challenge with LASV. We reasoned that this virus, modified by using reverse genetics, would represent the basis for the generation of a vaccine platform against LASV and other pathogenic arenaviruses. After showing evidence of exoribonuclease (ExoN) activity in NP of MOPV, we found that this activity was essential for multiplication in antigen-presenting cells. The introduction of multiple mutations in the ExoN site of MOPV NP generated a hyperattenuated strain (MOPVExoN6b) that is (i) genetically stable over passages, (ii) has increased immunogenic properties compared to those of MOPV, and (iii) still promotes a strong type I interferon (IFN) response. MOPVExoN6b was further modified to harbor the envelope glycoproteins of heterologous pathogenic arenaviruses, such as LASV or Lujo, Machupo, Guanarito, Chapare, or Sabia virus in order to broaden specific antigenicity while preserving the hyperattenuated characteristics of the parental strain. Our MOPV-based vaccine candidate for LASV, MOPEVACLASV, was used in a one-shot immunization assay in nonhuman primates and fully protected them from a lethal challenge with LASV. Thus, our hyperattenuated strain of MOPV constitutes a promising new live-attenuated vaccine platform to immunize against several, if not all, pathogenic arenaviruses.IMPORTANCE Arenaviruses are emerging pathogens transmitted to humans by rodents and responsible for endemic and epidemic hemorrhagic fevers of global concern. Nonspecific symptoms associated with the onset of infection make these viruses difficult to distinguish from other endemic pathogens. Moreover, the unavailability of rapid diagnosis in the field delays the identification of the virus and early care for treatment and favors spreading. The vaccination of exposed populations would be of great help to decrease morbidity and human-to-human transmission. Using reverse genetics, we generated a vaccine platform for pathogenic arenaviruses based on a modified and hyperattenuated strain of the nonpathogenic Mopeia virus and showed that the Lassa virus candidate fully protected nonhuman primates from a lethal challenge. These results showed that a rationally designed recombinant MOPV-based vaccine is safe, immunogenic, and efficacious in nonhuman primates.
Collapse
Affiliation(s)
- Xavier Carnec
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| | - Mathieu Mateo
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| | - Audrey Page
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| | - Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| | - Jimmy Hortion
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| | - Caroline Picard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| | - Elsie Yekwa
- CNRS, Architecture et Fonction des Macromolécules Biologiques UMR 7257, Aix-Marseille Université, Marseille, France
| | - Laura Barrot
- Laboratoire P4 Jean Mérieux-INSERM, US003, INSERM, Lyon, France
| | - Stéphane Barron
- Laboratoire P4 Jean Mérieux-INSERM, US003, INSERM, Lyon, France
| | - Audrey Vallve
- Laboratoire P4 Jean Mérieux-INSERM, US003, INSERM, Lyon, France
| | - Hervé Raoul
- Laboratoire P4 Jean Mérieux-INSERM, US003, INSERM, Lyon, France
| | | | - François Ferron
- CNRS, Architecture et Fonction des Macromolécules Biologiques UMR 7257, Aix-Marseille Université, Marseille, France
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie (INSERM, CNRS, ENS Lyon, Université Lyon I), Lyon, France
| |
Collapse
|
83
|
Abstract
Lassa virus (LASV) is a persistent global health threat that causes about half a million cases of Lassa fever each year in Western Africa. Although most cases are mild, the disease can cause significant morbidity and results in as many as 5,000 deaths per year. Since 2015, Nigeria has been experiencing a severe and extended outbreak of Lassa fever, raising concerns that it could spill over into other countries and reach a magnitude similar to the West African Ebola outbreak of 2013-2016. Despite the burden that Lassa fever places on public health, both in Africa and around the world, there are still no clinically-approved therapeutics or vaccines to treat or prevent it. Nevertheless, a number of promising candidate vaccines have been developed over the last several years, and there is a growing political and social determination to drive at least one of these candidates towards licensure. This paper describes a LASV vaccine candidate that is being developed at Canada's National Microbiology Laboratory. Based on the same live attenuated vesicular stomatitis virus (VSV) vaccine platform that was used to produce the successful Ebola virus vaccine, the VSV-based LASV vaccine has been shown to elicit a potent and protective immune response against LASV. The vaccine shows 100% protection in the "gold-standard" nonhuman primate model of Lassa fever, inducing both humoral and cellular immune responses. Moreover, studies have shown that a single vaccination may offer universal protection against numerous different strains of the virus, and additional studies have shown that immunization with the VSV platform appears to be unaffected by pre-existing immunity to VSV. The next step in the development of the VSV-based LASV vaccine is phase I human clinical trials to assess vaccine safety and dosage.
Collapse
|
84
|
Zemp F, Rajwani J, Mahoney DJ. Rhabdoviruses as vaccine platforms for infectious disease and cancer. Biotechnol Genet Eng Rev 2018; 34:122-138. [PMID: 29781359 DOI: 10.1080/02648725.2018.1474320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The family Rhabdoviridae (RV) comprises a large, genetically diverse collection of single-stranded, negative sense RNA viruses from the order Mononegavirales. Several RV members are being developed as live-attenuated vaccine vectors for the prevention or treatment of infectious disease and cancer. These include the prototype recombinant Vesicular Stomatitis Virus (rVSV) and the more recently developed recombinant Maraba Virus, both species within the genus Vesiculoviridae. A relatively strong safety profile in humans, robust immunogenicity and genetic malleability are key features that make the RV family attractive vaccine platforms. Currently, the rVSV vector is in preclinical development for vaccination against numerous high-priority infectious diseases, with clinical evaluation underway for HIV/AIDS and Ebola virus disease. Indeed, the success of the rVSV-ZEBOV vaccine during the 2014-15 Ebola virus outbreak in West Africa highlights the therapeutic potential of rVSV as a vaccine vector for acute, life-threatening viral illnesses. The rVSV and rMaraba platforms are also being tested as 'oncolytic' cancer vaccines in a series of phase 1-2 clinical trials, after being proven effective at eliciting immune-mediated tumour regression in preclinical mouse models. In this review, we discuss the biological and genetic features that make RVs attractive vaccine platforms and the development and ongoing testing of rVSV and rMaraba strains as vaccine vectors for infectious disease and cancer.
Collapse
Affiliation(s)
- Franz Zemp
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada
| | - Jahanara Rajwani
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| | - Douglas J Mahoney
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,c Department of Microbiology, Immunology and Infectious Disease , Faculty of Medicine , University of Calgary , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| |
Collapse
|
85
|
Antibodies to the Glycoprotein GP2 Subunit Cross-React between Old and New World Arenaviruses. mSphere 2018; 3:3/3/e00189-18. [PMID: 29720525 PMCID: PMC5932378 DOI: 10.1128/msphere.00189-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022] Open
Abstract
Arenaviruses pose a major public health threat and cause numerous infections in humans each year. Although most viruses belonging to this family do not cause disease in humans, some arenaviruses, such as Lassa virus and Machupo virus, are the etiological agents of lethal hemorrhagic fevers. The absence of a currently licensed vaccine and the highly pathogenic nature of these viruses both make the necessity of developing viable vaccines and therapeutics all the more urgent. Arenaviruses have a single glycoprotein on the surface of virions, the glycoprotein complex (GPC), and this protein can be used as a target for vaccine development. Here, we describe immunization strategies to generate monoclonal antibodies (MAbs) that cross-react between the glycoprotein complexes of both Old World and New World arenaviruses. Several monoclonal antibodies isolated from immunized mice were highly cross-reactive, binding a range of Old World arenavirus glycoproteins, including that of Lassa virus. One such monoclonal antibody, KL-AV-2A1, bound to GPCs of both New World and Old World viruses, including Lassa and Machupo viruses. These cross-reactive antibodies bound to epitopes present on the glycoprotein 2 subunit of the glycoprotein complex, which is relatively conserved among arenaviruses. Monoclonal antibodies binding to these epitopes, however, did not inhibit viral entry as they failed to neutralize a replication-competent vesicular stomatitis virus pseudotyped with the Lassa virus glycoprotein complex in vitro In addition, no protection from virus challenge was observed in in vivo mouse models. Even so, these monoclonal antibodies might still prove to be useful in the development of clinical and diagnostic assays.IMPORTANCE Several viruses in the Arenaviridae family infect humans and cause severe hemorrhagic fevers which lead to high case fatality rates. Due to their pathogenicity and geographic tropisms, these viruses remain very understudied. As a result, an effective vaccine or therapy is urgently needed. Here, we describe efforts to produce cross-reactive monoclonal antibodies that bind to both New and Old World arenaviruses. All of our MAbs seem to be nonneutralizing and nonprotective and target subunit 2 of the glycoprotein. Due to the lack of reagents such as recombinant glycoproteins and antibodies for rapid detection assays, our MAbs could be beneficial as analytic and diagnostic tools.
Collapse
|
86
|
Takayama-Ito M, Lim CK, Yamaguchi Y, Posadas-Herrera G, Kato H, Iizuka I, Islam MT, Morimoto K, Saijo M. Replication-incompetent rabies virus vector harboring glycoprotein gene of lymphocytic choriomeningitis virus (LCMV) protects mice from LCMV challenge. PLoS Negl Trop Dis 2018; 12:e0006398. [PMID: 29659579 PMCID: PMC5901774 DOI: 10.1371/journal.pntd.0006398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/21/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Lymphocytic choriomeningitis virus (LCMV) causes a variety of diseases, including asymptomatic infections, meningitis, and congenital infections in the fetus of infected mother. The development of a safe and effective vaccine against LCMV is imperative. This study aims to develop a new candidate vaccine against LCMV using a recombinant replication-incompetent rabies virus (RV) vector. METHODOLOGY/PRINCIPAL FINDINGS In this study, we have generated a recombinant deficient RV expressing the LCMV glycoprotein precursor (GPC) (RVΔP-LCMV/GPC) which is lacking the RV-P gene. RVΔP-LCMV/GPC is able to propagate only in cells expressing the RV-P protein. In contrast, the LCMV-GPC can be expressed in general cells, which do not express RV-P protein. The ability of RVΔP-LCMV/GPC to protect mice from LCMV infection and induce cellular immunity was assessed. Mice inoculated intraperitoneally with RVΔP-LCMV/GPC showed higher survival rates (88.2%) than those inoculated with the parental recombinant RV-P gene-deficient RV (RVΔP) (7.7%) following a LCMV challenge. Neutralizing antibody (NAb) against LCMV was not induced, even in the sera of surviving mice. CD8+ T-cell depletion significantly reduced the survival rates of RVΔP-LCMV/GPC-inoculated mice after the LCMV challenge. These results suggest that CD8+ T cells play a major role in the observed protection against LCMV. In contrast, NAbs against RV were strongly induced in sera of mice inoculated with either RVΔP-LCMV/GPC or RVΔP. In safety tests, suckling mice inoculated intracerebrally with RVΔP-LCMV/GPC showed no symptoms. CONCLUSIONS/SIGNIFICANCE These results show RVΔP-LCMV/GPC might be a promising candidate vaccine with dual efficacy, protecting against both RV and LCMV.
Collapse
Affiliation(s)
- Mutsuyo Takayama-Ito
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Chang-Kweng Lim
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Yukie Yamaguchi
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Guillermo Posadas-Herrera
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Hirofumi Kato
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
- Division of Global Infectious Diseases, Department of Infection and Epidemiology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Itoe Iizuka
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Md. Taimur Islam
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
- Laboratory of Virology and Viral Infections, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Kyonancho, Musashino-shi, Tokyo, Japan
| | - Kinjiro Morimoto
- Faculty of Pharmacy, Yasuda Women's University, Yasuhigashi, Asaminami, Hiroshima, Japan
| | - Masayuki Saijo
- Department of virology I, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
87
|
Baseline mapping of Lassa fever virology, epidemiology and vaccine research and development. NPJ Vaccines 2018; 3:11. [PMID: 29581897 PMCID: PMC5861057 DOI: 10.1038/s41541-018-0049-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/12/2018] [Accepted: 02/16/2018] [Indexed: 01/14/2023] Open
Abstract
Lassa fever (LF) is a zoonotic disease associated with acute and potentially fatal hemorrhagic illness caused by the Lassa virus (LASV), a member of the family Arenaviridae. It is generally assumed that a single infection with LASV will produce life-long protective immunity. This suggests that protective immunity induced by vaccination is an achievable goal and that cell-mediated immunity may play a more important role in protection, at least following natural infection. Seropositive individuals in endemic regions have been shown to have LASV-specific T cells recognizing epitopes for nucleocapsid protein (NP) and glycoprotein precursor (GPC), suggesting that these will be important vaccine immunogens. The role of neutralizing antibodies in protective immunity is still equivocal as recent studies suggest a role for neutralizing antibodies. There is extensive genetic heterogeneity among LASV strains that is of concern in the development of assays to detect and identify all four LASV lineages. Furthermore, the gene disparity may complicate the synthesis of effective vaccines that will provide protection across multiple lineages. Non-human primate models of LASV infection are considered the gold standard for recapitulation of human LF. The most promising vaccine candidates to date are the ML29 (a live attenuated reassortant of Mopeia and LASV), vesicular stomatitis virus (VSV) and vaccinia-vectored platforms based on their ability to induce protection following single doses, high rates of survival following challenge, and the use of live virus platforms. To date no LASV vaccine candidates have undergone clinical evaluation.
Collapse
|
88
|
An LASV GPC pseudotyped virus based reporter system enables evaluation of vaccines in mice under non-BSL-4 conditions. Vaccine 2017; 35:5172-5178. [PMID: 28797730 DOI: 10.1016/j.vaccine.2017.07.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/16/2017] [Accepted: 07/27/2017] [Indexed: 12/23/2022]
Abstract
Lassa virus (LASV) causes a severe hemorrhagic fever endemic throughout western Africa. Because of the ability to cause lethal disease in humans, limited treatment options, and potential as a bioweapon, the need for vaccines to prevent LASV epidemic is urgent. However, LASV vaccine development has been hindered by the lack of appropriate small animal models for efficacy evaluation independent of biosafety level four (BSL-4) facilities. Here we generated an LASV-glycoprotein precursor (GPC)-pseudotyped Human immunodeficiency virus containing firefly luciferase (Fluc) reporter gene as surrogate to develop a bioluminescent-imaging-based BALB/c mouse model for one-round infection under non-BSL-4 conditions, in which the bioluminescent intensity of Fluc was utilized as endpoint when evaluating vaccine efficacy. Electron microscopy analysis demonstrated that LASV GPC pseudotyped virus appeared structurally similar to native virion. Meanwhile, we constructed DNA vaccine (pSV1.0-LASVGPC) and pseudoparticle-based vaccine (LASVpp) that displayed conformational GPC protein of LASV strain Josiah to vaccinate BALB/c mice using intramuscular electroporation and by intraperitoneal routes, respectively. Vaccinated mice in LASVpp alone and DNA prime+LASVpp boost schedules were protected against 100 AID50 of LASV pseudovirus challenge, and it was found that in vivo efficiencies correlated with their anti-LASV neutralizing activities and MCP-1 cytokine levels in serum sampled before infection. The bioluminescence pseudovirus infection model can be useful tool for the preliminary evaluation of immunogenicity and efficacy of vaccine candidates against LASV outside of BSL-4 containments, and the results with pseudoparticle-based vaccine provided very helpful information for LASV vaccine design.
Collapse
|
89
|
Abstract
The author reviews the foundation of the Coalition for Epidemic Preparedness and Innovations and the choices it has made for funding of vaccine development against epidemic diseases. He comments on those decisions as well as proposing how CEPI could remain relevant for the long term.
Collapse
Affiliation(s)
- Stanley A Plotkin
- a Emeritus Professor of Pediatrics , University of Pennsylvania ; Vaxconsult, Doylestown , PA , USA
| |
Collapse
|
90
|
Abstract
Two of the most important contemporary emerging viruses that affect human health in Africa are Ebola virus (EBOV) and Lassa virus (LASV). The 2013-2016 West African outbreak of EBOV was responsible for more than 11,000 deaths, primarily in Guinea, Sierra Leone and Liberia. LASV is constantly emerging in these and surrounding West African countries, with an estimate of more than 500,000 cases of Lassa fever, and approximately 5,000 deaths, annually. Both EBOV and LASV are zoonotic, and human infection often results in a severe haemorrhagic fever in both cases. However, the contribution of specific immune responses to disease differs between EBOV and LASV. This Review examines innate and adaptive immune responses to these viruses with the goal of delineating responses that are associated with protective versus pathogenic outcomes.
Collapse
|
91
|
Replicon RNA Viral Vectors as Vaccines. Vaccines (Basel) 2016; 4:vaccines4040039. [PMID: 27827980 PMCID: PMC5192359 DOI: 10.3390/vaccines4040039] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 12/22/2022] Open
Abstract
Single-stranded RNA viruses of both positive and negative polarity have been used as vectors for vaccine development. In this context, alphaviruses, flaviviruses, measles virus and rhabdoviruses have been engineered for expression of surface protein genes and antigens. Administration of replicon RNA vectors has resulted in strong immune responses and generation of neutralizing antibodies in various animal models. Immunization of mice, chicken, pigs and primates with virus-like particles, naked RNA or layered DNA/RNA plasmids has provided protection against challenges with lethal doses of infectious agents and administered tumor cells. Both prophylactic and therapeutic efficacy has been achieved in cancer immunotherapy. Moreover, recombinant particles and replicon RNAs have been encapsulated by liposomes to improve delivery and targeting. Replicon RNA vectors have also been subjected to clinical trials. Overall, immunization with self-replicating RNA viruses provides high transient expression levels of antigens resulting in generation of neutralizing antibody responses and protection against lethal challenges under safe conditions.
Collapse
|
92
|
Pushko P, Lukashevich IS, Weaver SC, Tretyakova I. DNA-launched live-attenuated vaccines for biodefense applications. Expert Rev Vaccines 2016; 15:1223-34. [PMID: 27055100 PMCID: PMC5033646 DOI: 10.1080/14760584.2016.1175943] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A novel vaccine platform uses DNA immunization to launch live-attenuated virus vaccines in vivo. This technology has been applied for vaccine development against positive-strand RNA viruses with global public health impact including alphaviruses and flaviviruses. The DNA-launched vaccine represents the recombinant plasmid that encodes the full-length genomic RNA of live-attenuated virus downstream from a eukaryotic promoter. When administered in vivo, the genomic RNA of live-attenuated virus is transcribed. The RNA initiates limited replication of a genetically defined, live-attenuated vaccine virus in the tissues of the vaccine recipient, thereby inducing a protective immune response. This platform combines the strengths of reverse genetics, DNA immunization and the advantages of live-attenuated vaccines, resulting in a reduced chance of genetic reversions, increased safety, and improved immunization. With this vaccine technology, the field of DNA vaccines is expanded from those that express subunit antigens to include a novel type of DNA vaccines that launch live-attenuated viruses.
Collapse
Affiliation(s)
- Peter Pushko
- Medigen, Inc. 8420 Gas House Pike Suite S, Frederick, MD 21701, USA
| | - Igor S. Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, Center for Predictive Medicine and Emerging Infectious Diseases, University of Louisville, 505 S Hancock St., Louisville, KY 40202, USA
| | - Scott C. Weaver
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development and Department of Microbiology and Immunology, University of Texas Medical Branch, GNL, 301 University Blvd., Galveston, TX 77555, USA
| | - Irina Tretyakova
- Medigen, Inc. 8420 Gas House Pike Suite S, Frederick, MD 21701, USA
| |
Collapse
|
93
|
Goba A, Khan SH, Fonnie M, Fullah M, Moigboi A, Kovoma A, Sinnah V, Yoko N, Rogers H, Safai S, Momoh M, Koroma V, Kamara FK, Konowu E, Yillah M, French I, Mustapha I, Kanneh F, Foday M, McCarthy H, Kallon T, Kallon M, Naiebu J, Sellu J, Jalloh AA, Gbakie M, Kanneh L, Massaly JLB, Kargbo D, Kargbo B, Vandi M, Gbetuwa M, Gevao SM, Sandi JD, Jalloh SC, Grant DS, Blyden SO, Crozier I, Schieffelin JS, McLellan SL, Jacob ST, Boisen ML, Hartnett JN, Cross RW, Branco LM, Andersen KG, Yozwiak NL, Gire SK, Tariyal R, Park DJ, Haislip AM, Bishop CM, Melnik LI, Gallaher WR, Wimley WC, He J, Shaffer JG, Sullivan BM, Grillo S, Oman S, Garry CE, Edwards DR, McCormick SJ, Elliott DH, Rouelle JA, Kannadka CB, Reyna AA, Bradley BT, Yu H, Yenni RE, Hastie KM, Geisbert JB, Kulakosky PC, Wilson RB, Oldstone MBA, Pitts KR, Henderson LA, Robinson JE, Geisbert TW, Saphire EO, Happi CT, Asogun DA, Sabeti PC, Garry RF. An Outbreak of Ebola Virus Disease in the Lassa Fever Zone. J Infect Dis 2016; 214:S110-S121. [PMID: 27402779 DOI: 10.1093/infdis/jiw239] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Kenema Government Hospital (KGH) has developed an advanced clinical and laboratory research capacity to manage the threat of Lassa fever, a viral hemorrhagic fever (VHF). The 2013-2016 Ebola virus (EBOV) disease (EVD) outbreak is the first to have occurred in an area close to a facility with established clinical and laboratory capacity for study of VHFs. METHODS Because of its proximity to the epicenter of the EVD outbreak, which began in Guinea in March 2014, the KGH Lassa fever Team mobilized to establish EBOV surveillance and diagnostic capabilities. RESULTS Augustine Goba, director of the KGH Lassa laboratory, diagnosed the first documented case of EVD in Sierra Leone, on 25 May 2014. Thereafter, KGH received and cared for numbers of patients with EVD that quickly overwhelmed the capacity for safe management. Numerous healthcare workers contracted and lost their lives to EVD. The vast majority of subsequent EVD cases in West Africa can be traced back to a single transmission chain that includes this first diagnosed case. CONCLUSIONS Responding to the challenges of confronting 2 hemorrhagic fever viruses will require continued investments in the development of countermeasures (vaccines, therapeutic agents, and diagnostic assays), infrastructure, and human resources.
Collapse
Affiliation(s)
- Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - S Humarr Khan
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Mbalu Fonnie
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Mohamed Fullah
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Alex Moigboi
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Alice Kovoma
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Vandi Sinnah
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Nancy Yoko
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Hawa Rogers
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Siddiki Safai
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Mambu Momoh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | | | - Edwin Konowu
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Mohamed Yillah
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Issa French
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | | | - Momoh Foday
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | - Tiangay Kallon
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | - Jenneh Naiebu
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | - Abdul A Jalloh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Michael Gbakie
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Lansana Kanneh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | | | | | | | | | | | - John D Sandi
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | | | - Donald S Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital Ministry of Health and Sanitation
| | | | - Ian Crozier
- World Health Organization Sierra Leone Ebola Response Team, Freetown, Sierra Leone Infectious Diseases Institute, Mulago Hospital Complex, Kampala, Uganda
| | - John S Schieffelin
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Susan L McLellan
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine Department of Tropical Medicine
| | - Shevin T Jacob
- Division of Allergy and Infectious Diseases, University of Washington, Seattle
| | - Matt L Boisen
- Corgenix, Broomfield, Colorado Zalgen Labs, Germantown, Maryland
| | | | - Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston
| | | | | | | | | | | | | | | | | | | | - William R Gallaher
- Department of Microbiology, Immunology, and Parasitology, LSU Health Mockingbird Nature Research Group, Pearl River, Louisiana
| | | | - Jing He
- Department of Biochemistry, Tulane University
| | - Jeffrey G Shaffer
- Department of Biostatistics and Bioinformatics, Tulane School of Public Health and Tropical Medicine
| | | | - Sonia Grillo
- Naval Engineering Facilities Command, Naples, Italy
| | | | - Courtney E Garry
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine Autoimmune Technologies, New Orleans
| | | | | | - Deborah H Elliott
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Julie A Rouelle
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Chandrika B Kannadka
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Ashley A Reyna
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Benjamin T Bradley
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Haini Yu
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | | | | | - Joan B Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston
| | | | | | | | | | | | - James E Robinson
- Section of Infectious Disease, Department of Pediatrics Section of Infectious Disease, Department of Internal Medicine, School of Medicine
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla
| | - Christian T Happi
- Redeemer's University, Ede Irrua Specialist Teaching Hospital, Nigeria
| | | | - Pardis C Sabeti
- Broad Institute of MIT and Harvard Department of Organismic and Evolutionary Biology, Center for Systems Biology, Harvard University, Cambridge, Massachusetts
| | - Robert F Garry
- Department of Microbiology and Immunology Zalgen Labs, Germantown, Maryland
| | | |
Collapse
|
94
|
Abstract
INTRODUCTION Lassa virus (LASV), the most prominent human pathogen of the Arenaviridae, is transmitted to humans from infected rodents and can cause Lassa Fever (LF). The sizeable disease burden in West Africa, numerous imported LF cases worldwide, and the possibility that LASV can be used as an agent of biological warfare make a strong case for vaccine development. There are no licensed LASV vaccines and the antiviral treatment is limited to an off-label use of ribavirin that is only partially effective. AREAS COVERED LASV vaccine development is hampered by high cost of biocontainment requirement, the absence of appropriate small animal models, genetic diversity of LASV species, and by high HIV-1 prevalence in LASV endemic areas. Over the past 15 years several vaccine platforms have been developed. Natural history of LASV and pathogenesis of the disease provide strong justification for replication-competent (RC) vaccine as one of the most feasible approaches to control LF. Development of LASV vaccine candidates based on reassortant, recombinant, and alphavirus replicon technologies is covered in this review. Expert commentary: Two lead RC vaccine candidates, reassortant ML29 and recombinant VSV/LASV, have been successfully tested in non-human primates and have been recommended by international vaccine experts for rapid clinical development. Both platforms have powerful molecular tools to further secure safety, improve immunogenicity, and cross-protection. These platforms are well positioned to design multivalent vaccines to protect against all LASV strains citculatrd in West Africa. The regulatory pathway of Candid #1, the first live-attenuated arenaviral vaccine against Argentine hemorrhagic, will be a reasonable guideline for LASV vaccine efficacy trials.
Collapse
Affiliation(s)
- Igor S Lukashevich
- a Department of Pharmacology and Toxicology, School of Medicine, and the Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases , University of Louisville , Louisville , KY , USA
| | | |
Collapse
|
95
|
Abstract
RNA viruses are characterized by their efficient capacity to replicate at high levels in mammalian cells leading to high expression of foreign genes and making them attractive candidates for vectors engineered for vaccine development and gene therapy. Particularly, alphaviruses, flaviviruses, rhabdoviruses and measles viruses have been applied for immunization against infectious agents and tumors. Application of replicon RNA, DNA/RNA-layered vectors and replication-deficient viral particles have provided strong immune responses and protection against challenges with lethal doses of viral pathogens or tumor cells. Moreover, tumor regression has been obtained when RNA replicons have been administered in the form of RNA, DNA and viral particles, including replication-proficient oncolytic particles.
Collapse
|
96
|
Single-Vector, Single-Injection Recombinant Vesicular Stomatitis Virus Vaccines Against High-Containment Viruses. Methods Mol Biol 2016; 1403:295-311. [PMID: 27076138 DOI: 10.1007/978-1-4939-3387-7_16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
There are many avenues for making an effective vaccine against viruses. Depending on the virus these can include one of the following: inactivation of whole virions; attenuation of viruses; recombinant viral proteins; non-replication-competent virus particles; or surrogate virus vector systems such as vesicular stomatitis virus (VSV). VSV is a prototypic enveloped animal virus that has been used for over four decades to study virus replication, entry, and assembly due to its ability to replicate to high titers in a wide variety of mammalian and insect cells. The use of reverse genetics to recover infectious and single-cycle replicating VSV from plasmid DNA transfected in cell culture began a revolution in the study of recombinant VSV (rVSV). This platform can be manipulated to study the viral genetic sequences and proteins important in the virus life cycle. Additionally, foreign genes can be inserted between naturally occurring or generated start/stop signals and polyadenylation sites within the VSV genome. VSV has a tolerance for foreign gene expression which has led to numerous rVSVs reported in the literature. Of particular interest are the very effective single-dose rVSV vaccine vectors against high-containment viruses such as filoviruses, henipaviruses, and arenaviruses. Herein we describe the methods for selecting foreign antigenic genes, selecting the location within the VSV genome for insertion, generation of rVSV using reverse genetics, and proper vaccine study designs.
Collapse
|
97
|
The broad-spectrum antiviral favipiravir protects guinea pigs from lethal Lassa virus infection post-disease onset. Sci Rep 2015; 5:14775. [PMID: 26456301 PMCID: PMC4600983 DOI: 10.1038/srep14775] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/07/2015] [Indexed: 01/25/2023] Open
Abstract
With up to 500,000 infections annually, Lassa virus (LASV), the cause of Lassa fever, is one of the most prevalent etiological agents of viral hemorrhagic fever (VHF) in humans. LASV is endemic in several West African countries with sporadic cases and prolonged outbreaks observed most commonly in Sierra Leone, Liberia, Guinea and Nigeria. Additionally several cases of Lassa fever have been imported into North America, Europe and Asia making LASV a global threat to public health. Despite this, currently no approved therapeutic or vaccine exists to treat or prevent LASV infections. Here, using a passaged strain of LASV that is uniformly lethal in Hartley guinea pigs, we demonstrate that favipiravir, a broad-spectrum antiviral agent and leading treatment option for influenza, has potent activity against LASV infection. In this model, once daily treatment with favipiravir significantly reduced viral titers in tissue samples and reduced mortality rates when compared with animals receiving vehicle-only or ribavirin, the current standard of care for Lassa fever. Favipiravir remained highly effective against lethal LASV infection when treatments were initiated nine days post-infection, a time when animals were demonstrating advanced signs of disease. These results support the further preclinical evaluation of favipiravir for Lassa fever and other VHFs.
Collapse
|