Several arenaviruses cause hemorrhagic fever (HF) disease in humans and pose a serious threat to public health within their endemic regions. Thus, the New World arenavirus Junin (JUNV) is the etiological agent of Argentine HF (AHF), a severe illness with hemorrhagic and neurological manifestations and a case fatality of 15-30%. In addition, JUNV is very stable, highly infectious by aerosol and causes high morbidity and mortality at low dose, features that make it highly suitable as potential biological weapon. Concerns about arenavirus infections are aggravated by the lack of licensed vaccines in the US and current anti-arenavirus therapies being limited to the use of the nucleoside analogue ribavirin, which is only partially effective and can cause significant toxicity. Accordingly, the development of antiviral drugs against JUNV has been considered as one of the top priorities within the Implementation Plan of the HHS Public Health Emergency Medical Countermeasures Enterprise (PHEMCE). Therefore, medical countermeasures against JUNV infections are eligible for acquisition by BARDA for the Strategic National Stockpile. We have developed a reverse genetic system for JUNV and rescued infectious viruses, both Candid1 and the pathogenic Romero strain, entirely from cloned cDNAs. This advance has provided us with the unique opportunity to generate a genetically well-defined seeds of Candid1 (rJUNV/Candid1cSD) and Romero (rJUNV/Romero) strains of JUNV to be utilized in immunogenicity and efficacy studies. This proposal is designed to provide a comprehensive characterization of the stability, immunogenic and protective properties of rJUNV/Candid1cSD as a first and necessary step for the development of a Candid 1-based vaccine able to meet the safety and efficacy criteria required for future FDA licensing. For this we propose the following specific aims: 1. Determine the genetic and phenotypic stability of rJUNV/Candid1cSD. We propose to generate and characterize a Master Virus Seed (MVS) of rCandid1cSD. We will use the MVS from rCandid1cSD to conduct independent serial passages in different cell substrates, including FDA approved cell lines for vaccine production. Infectious progenies from selected passages from each series will be characterized genetically (genome sequencing) and phenotypically (growth properties and virulence). Genetic and phenotypic information derived from these studies will be used to generate novel rCandid1 strains optimized for genetic and phenotypic stability. 2. To determine the safety, immunogenicity and efficacy of rJUNV/Candid1cSD in guinea pigs. We propose to conduct a comprehensive assessment of the virulence of our genetically well-defined MVS rCandid1cSD in guinea pigs. For this, we will determine clinical parameters associated with immunization of rCandid1cSD including: body temperature and weight, overall activity, assessment of hematological and clinical pathology data covering kidney and liver function, and protection against lethal challenge with JUNV. These studies will also include relevant viral variants identified during studies aimed at assessing the genetic and phenotypic stability of the MVS. 3. Determine the efficacy of rJUNV/Candid1cSD against JUNV in common marmoset (Callithrix jacchus). We propose to evaluate the protective efficacy of rJUNV/Candid1cSD against HF disease caused by the pathogenic rJUNV/Romero strain in the common marmoset (Callithrix jacchus) animal model of JUNV infection. Results from these studies will be correlated with immunogenicity data obtained from studies described in aim 2.

Public Health Relevance

This study will generate a system for production of recombinant vaccine against Argentine hemorrhagic fever, Candid1#. Additionally, we will generate data concerning its safety and stability in cells and animals and will test its immunogenicity and efficacy in two animal models. We propose a novel system for vaccine production against hemorrhagic fever viruses in order to increase our ability to prevent some of the most severe infectious diseases should it be introduced into the USA.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
1R01AI093445-01
Application #
8076684
Study Section
Special Emphasis Panel (ZAI1-NLE-M (J3))
Program Officer
Repik, Patricia M
Project Start
2011-04-04
Project End
2016-03-31
Budget Start
2011-04-04
Budget End
2012-03-31
Support Year
1
Fiscal Year
2011
Total Cost
$901,836
Indirect Cost
Name
University of Texas Medical Br Galveston
Department
Pathology
Type
Schools of Medicine
DUNS #
800771149
City
Galveston
State
TX
Country
United States
Zip Code
77555
Mateer, Elizabeth J; Paessler, Slobodan; Huang, Cheng (2018) Visualization of Double-Stranded RNA Colocalizing With Pattern Recognition Receptors in Arenavirus Infected Cells. Front Cell Infect Microbiol 8:251
Huang, Cheng; Kolokoltsova, Olga A; Mateer, Elizabeth J et al. (2017) Highly Pathogenic New World Arenavirus Infection Activates the Pattern Recognition Receptor Protein Kinase R without Attenuating Virus Replication in Human Cells. J Virol 91:
Manning, John T; Seregin, Alexey V; Yun, Nadezhda E et al. (2017) Absence of an N-Linked Glycosylation Motif in the Glycoprotein of the Live-Attenuated Argentine Hemorrhagic Fever Vaccine, Candid #1, Results in Its Improper Processing, and Reduced Surface Expression. Front Cell Infect Microbiol 7:20
Koma, Takaaki; Huang, Cheng; Aronson, Judith F et al. (2016) The Ectodomain of Glycoprotein from the Candid#1 Vaccine Strain of Junin Virus Rendered Machupo Virus Partially Attenuated in Mice Lacking IFN-??/? Receptor. PLoS Negl Trop Dis 10:e0004969
Koma, Takaaki; Patterson, Michael; Huang, Cheng et al. (2016) Machupo Virus Expressing GPC of the Candid#1 Vaccine Strain of Junin Virus Is Highly Attenuated and Immunogenic. J Virol 90:1290-7
Huang, Cheng; Kolokoltsova, Olga A; Yun, Nadezhda E et al. (2015) Highly Pathogenic New World and Old World Human Arenaviruses Induce Distinct Interferon Responses in Human Cells. J Virol 89:7079-88
Seregin, Alexey V; Yun, Nadezhda E; Miller, Milagros et al. (2015) The glycoprotein precursor gene of Junin virus determines the virulence of the Romero strain and the attenuation of the Candid #1 strain in a representative animal model of Argentine hemorrhagic fever. J Virol 89:5949-56
Patterson, Michael; Grant, Ashley; Paessler, Slobodan (2014) Epidemiology and pathogenesis of Bolivian hemorrhagic fever. Curr Opin Virol 5:82-90
Huang, Cheng; Walker, Aida G; Grant, Ashley M et al. (2014) Potent inhibition of Junín virus infection by interferon in murine cells. PLoS Negl Trop Dis 8:e2933
Patterson, Michael; Seregin, Alexey; Huang, Cheng et al. (2014) Rescue of a recombinant Machupo virus from cloned cDNAs and in vivo characterization in interferon (??/?) receptor double knockout mice. J Virol 88:1914-23

Showing the most recent 10 out of 15 publications