Kaposi's sarcoma-associated herpesvirus (KSHV) has been consistently identified in Kaposi's sarcoma (KS) tumors, primary effusion lymphoma (PEL), and Multicentric Castleman's disease. Although classical KS has a low prevalence rate worldwide, the more aggressive endemic KS, seen primarily in Africa, accounts for nearly half of the reported cancers in some regions and is the leading cause of cancer death in those areas. Despite being a pressing human health problem, there has been little or no activity so far to develop protective and/or therapeutic vaccines against KSHV infection and its associated diseases. A current major barrier for the in vivo studies of KSHV replication, persistence, pathogenesis, and ultimately vaccine is the lack of a proper animal model. We recently performed the first successful zoonotic or cross-species transmission of KSHV into common marmosets, a New World primate (Callithrix jacchus, Cj). This preliminary study has shown that: (1) common marmosets intravenously inoculated with recombinant rKSHV.219 rapidly sero-converted and maintained a high anti-KSHV antibody response over a long period of time;(2) common marmosets infected with rKSHV.219 via the oral and/or intranasal routes also rapidly sero-converted and maintained an anti-KSHV antibody response;and (3) KSHV DNA and the latent nuclear antigen protein are readily detectable in infected animals. This is the first animal model that significantly recapitulates the important aspects of KSHV infection in humans, thus providing a unique opportunity in developing potential vaccine strategies against KSHV infection. The most impressive vaccine protections achieved to date against virus- associated diseases primarily utilize persisting, live, attenuated viruses. This may be due to the duration of the immune response elicited through the continuous expression of viral proteins from a persisting viral genome in the infected host. Thus, we will consider developing a persistent, attenuated KSHV to serve as the vaccine vector. The goal of this study is two-folds: Firstly, to develop a genetically modified replication-competent, non-pathogenic KSHV strain to be used as a vaccine candidate. Secondly, to further build up the primate model of KSHV infection and to develop immunological and virological assays for the vaccination and challenge experiments. This proposal is highly innovative and a successful outcome should prove to be a major discovery that significantly impacts the understanding of the controls of KSHV infection to ultimately reveal novel protective and/or therapeutic vaccine strategies.

Public Health Relevance

Kaposi's sarcoma-associated herpesvirus (KSHV) has been consistently identified in Kaposi's sarcoma tumors, primary effusion lymphoma, and Multicentric Castleman's disease. Although classical KS has a low prevalence rate worldwide, the more aggressive endemic KS, seen primarily in Africa, accounts for nearly half of the reported cancers in some regions and is the leading cause of cancer death in those areas. Despite being a pressing human health problem, there has been little or no activity so far to develop protective and/or therapeutic vaccines against KSHV infection and its associated diseases. The proposed research is directed toward investigating the potential development of live-attenuated KSHV as a vaccine candidate. This proposal is highly innovative and a successful outcome should prove to be a major discovery that significantly impacts the understanding of the controls of KSHV infection to ultimately reveal novel protective and/or therapeutic vaccine strategies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
High Impact Research and Research Infrastructure Programs (RC2)
Project #
1RC2CA148616-01
Application #
7852415
Study Section
Special Emphasis Panel (ZCA1-GRB-I (O9))
Program Officer
Read-Connole, Elizabeth Lee
Project Start
2009-09-30
Project End
2011-08-31
Budget Start
2009-09-30
Budget End
2010-08-31
Support Year
1
Fiscal Year
2009
Total Cost
$500,000
Indirect Cost
Name
University of Southern California
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
072933393
City
Los Angeles
State
CA
Country
United States
Zip Code
90089
Seo, Gil Ju; Yang, Aerin; Tan, Brandon et al. (2015) Akt Kinase-Mediated Checkpoint of cGAS DNA Sensing Pathway. Cell Rep 13:440-9
Jeong, Joseph H; Bhatia, Ayesha; Toth, Zsolt et al. (2011) TPL2/COT/MAP3K8 (TPL2) activation promotes androgen depletion-independent (ADI) prostate cancer growth. PLoS One 6:e16205
Lee, Hye-Ra; Choi, Won-Chan; Lee, Stacy et al. (2011) Bilateral inhibition of HAUSP deubiquitinase by a viral interferon regulatory factor protein. Nat Struct Mol Biol 18:1336-44
Inn, Kyung-Soo; Lee, Sun-Hwa; Rathbun, Jessica Y et al. (2011) Inhibition of RIG-I-mediated signaling by Kaposi's sarcoma-associated herpesvirus-encoded deubiquitinase ORF64. J Virol 85:10899-904
Toth, Zsolt; Maglinte, Dennis T; Lee, Sun Hwa et al. (2010) Epigenetic analysis of KSHV latent and lytic genomes. PLoS Pathog 6:e1001013
Lee, Hye-Ra; Lee, Stacy; Chaudhary, Preet M et al. (2010) Immune evasion by Kaposi's sarcoma-associated herpesvirus. Future Microbiol 5:1349-65