1
|
Golas G, Park BS, Wong SW. Glycoproteins gM and gN are indispensable factors for rhesus macaque rhadinovirus replication and spread but can be reconstituted by KSHV chimeras. J Virol 2025; 99:e0192224. [PMID: 39998253 PMCID: PMC11915806 DOI: 10.1128/jvi.01922-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Rhesus macaque rhadinovirus (RRV) is a primate gamma-2 herpesvirus (rhadinovirus) closely related to Kaposi sarcoma-associated herpesvirus (KSHV), the human oncovirus that causes Kaposi sarcoma. Like other herpesviruses, KSHV and RRV encode numerous envelope glycoproteins involved in cell attachment, entry, as well as assembly and release of progeny virions from infected cells. Two glycoproteins postulated to form a complex and reported to be virus-neutralizing targets are glycoproteins M (gM) and N (gN). To investigate gM and gN in rhadinovirus infection, we utilized infectious and pathogenic bacterial artificial chromosomes (BAC). RRV BACmids with nonsense mutations introduced into gM or gN did not yield an infectious virus. However, when gM or gN of RRV were exchanged for gM or gN from KSHV, each of the KSHV-chimeric RRV BACmids restored virus replication and infectious spread. Interestingly, we also discovered that the substitution of KSHVgM into the RRV BACmid was associated with attenuation in viral spread, an effect that was not countered by a double-chimeric virus. In contrast, the substitution of RRV gN into a KSHV BACmid negatively affected the assembly of KSHV, independent of gM/gN complex formation. Therefore, here, we revealed that in KSHV and RRV, gM and gN are interchangeable, contribute to crucial functions for viral assembly and spread, and have evolved in a virus-specific manner. Although more research is needed to define the roles of gM and gN, our work establishes the first glycoprotein-chimeric viruses for KSHV and RRV, which can now be used to corroborate gM/gN as targets for a cancer vaccine.IMPORTANCEKaposi sarcoma (KS) is a human cancer caused by KSHV and is one of the most frequently occurring cancers in HIV/AIDS patients, as well as in regions where KSHV is endemic. In this report, we have constructed and authenticated the first KSHV glycoprotein-encoding chimeric viruses for evaluations in the RRV/rhesus macaque model and have also uncovered fundamental roles for the glycoproteins gM and gN. Our work is significant by successfully bridging the human-specific, species barrier that has previously restricted preclinical evaluations of the KSHV glycoproteins as vaccine targets in vivo. Although there is no KSHV-specific animal model that is widely used, these KSHV-chimeric viruses may be useful as tools to guide future vaccine design and strategy as vaccine candidates progress toward clinical trials.
Collapse
Affiliation(s)
- Gavin Golas
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Byung S Park
- Biostatistics Shared Resource, Oregon Health & Science University, Knight Cancer Institute, Portland, Oregon, USA
- Biostatistics and Bioinformatics Core, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Scott W Wong
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Banerjee A, Dass D, Mukherjee S, Kaul M, Harshithkumar R, Bagchi P, Mukherjee A. The 'Oma's of the Gammas-Cancerogenesis by γ-Herpesviruses. Viruses 2024; 16:1928. [PMID: 39772235 PMCID: PMC11680331 DOI: 10.3390/v16121928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/03/2025] Open
Abstract
Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV), which are the only members of the gamma(γ) herpesviruses, are oncogenic viruses that significantly contribute to the development of various human cancers, such as Burkitt's lymphoma, nasopharyngeal carcinoma, Hodgkin's lymphoma, Kaposi's sarcoma, and primary effusion lymphoma. Oncogenesis triggered by γ-herpesviruses involves complex interactions between viral genetics, host cellular mechanisms, and immune evasion strategies. At the genetic level, crucial viral oncogenes participate in the disruption of cell signaling, leading to uncontrolled proliferation and inhibition of apoptosis. These viral proteins can modulate several cellular pathways, including the NF-κB and JAK/STAT pathways, which play essential roles in cell survival and inflammation. Epigenetic modifications further contribute to EBV- and KSHV-mediated cancerogenesis. Both EBV and KSHV manipulate host cell DNA methylation, histone modification, and chromatin remodeling, the interplay of which contribute to the elevation of oncogene expression and the silencing of the tumor suppressor genes. Immune factors also play a pivotal role in the development of cancer. The γ-herpesviruses have evolved intricate immune evasion strategies, including the manipulation of the major histocompatibility complex (MHC) and the release of cytokines, allowing infected cells to evade immune detection and destruction. In addition, a compromised immune system, such as in HIV/AIDS patients, significantly increases the risk of cancers associated with EBV and KSHV. This review aims to provide a comprehensive overview of the genetic, epigenetic, and immune mechanisms by which γ-herpesviruses drive cancerogenesis, highlighting key molecular pathways and potential therapeutic targets.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Debashree Dass
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Soumik Mukherjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Mollina Kaul
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - R. Harshithkumar
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Parikshit Bagchi
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anupam Mukherjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
- AcSIR—Academy of Scientific & Innovative Research, Ghaziabad 201002, UP, India
| |
Collapse
|
3
|
Yang WS, Kim D, Kang S, Lai CJ, Cha I, Chang PC, Jung JU. Development of KSHV vaccine platforms and chimeric MHV68-K-K8.1 glycoprotein for evaluating the in vivo immunogenicity and efficacy of KSHV vaccine candidates. mBio 2024; 15:e0291324. [PMID: 39475238 PMCID: PMC11633179 DOI: 10.1128/mbio.02913-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 12/12/2024] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV)/human herpesvirus 8 is an etiological agent of Kaposi's Sarcoma, multicentric Castleman's disease, and primary effusion lymphoma. Considering the high seroprevalence reaching up to 80% in sub-Saharan Africa, an effective vaccine is crucial for preventing KSHV infection. However, vaccine development has been limited due to the lack of an effective animal model that supports KSHV infection. Murine Herpesvirus 68 (MHV68), a natural mouse pathogen persisting lifelong post-infection, presents a promising model for KSHV infection. In this study, we developed KSHV vaccine and a chimeric MHV68 carrying the KSHV glycoprotein, serving as a surrogate challenge virus for testing KSHV vaccines in a mouse model. Among KSHV virion glycoproteins, K8.1 is the most abundant envelope glycoprotein with the highest immunogenicity. We developed two K8.1 vaccines: K8.1 mRNA-lipid nanoparticle (LNP) vaccine and K8.126-87-Ferritin (FT) nanoparticle vaccines. Both induced humoral responses in immunized mice, whereas K8.1 mRNA LNP also induced T cell responses. Using BACmid-mediated homologous recombination, the MHV68 M7 (gp150) gene was replaced with KSHV K8.1 gene to generate chimeric MHV68-K-K8.1. MHV68-K-K8.1 established acute and latent infection in the lungs and spleens of infected mice, respectively. Mice immunized with K8.1 mRNA LNP or K8.126-87-FT showed a reduction of MHV68-K-K8.1 titer but not MHV68 wild type (WT) titer in the lung. In addition, viral reactivation of MHV68-K-K8.1 was also significantly reduced in K8.1 mRNA LNP-immunized mice. This study demonstrates the effectiveness of two vaccine candidates in providing immunity against KSHV K8.1 and introduces a surrogate MHV68 system for evaluating vaccine efficacy in vivo.IMPORTANCEKaposi's sarcoma-associated herpesvirus (KSHV) is a prevalent virus that establishes lifelong persistent infection in humans and is linked to several malignancies. While antiretroviral therapy has reduced Kaposi's Sarcoma (KS) complications in people with HIV, KS still affects individuals with well-controlled HIV, older men without HIV, and transplant recipients. Despite its significant impact on human health, however, research on KSHV vaccine has been limited, mainly due to the lack of interest and the absence of a suitable animal model. This study addresses these challenges by developing KSHV K8.1 vaccine with two platforms, mRNA lipid nanoparticle (LNP) and FT nanoparticle. Additionally, chimeric virus, MHV68-K-K8.1, was created to evaluate KSHV vaccine efficacy in vivo. Vaccination of K8.1 mRNA LNP or K8.126-87-FT significantly reduced MHV68-K-K8.1 titers. Developing an effective KSHV vaccine requires an innovative approach to ensure safety and efficacy, especially for the immunocompromised population and people with limited healthcare resources. This study could be a potential blueprint for future KSHV vaccine development.
Collapse
MESH Headings
- Animals
- Mice
- Herpesvirus 8, Human/immunology
- Herpesvirus 8, Human/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Disease Models, Animal
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Female
- Vaccine Development
- Rhadinovirus/genetics
- Rhadinovirus/immunology
- Nanoparticles/chemistry
- Humans
- Herpesviridae Infections/prevention & control
- Herpesviridae Infections/immunology
- Herpesviridae Infections/virology
- Immunogenicity, Vaccine
- Herpesvirus Vaccines/immunology
- Herpesvirus Vaccines/administration & dosage
- Herpesvirus Vaccines/genetics
- Vaccine Efficacy
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Mice, Inbred C57BL
- Sarcoma, Kaposi/virology
- Sarcoma, Kaposi/immunology
- Sarcoma, Kaposi/prevention & control
- Liposomes
- Viral Proteins
Collapse
Affiliation(s)
- Wan-Shan Yang
- Department of Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute ,Cleveland Clinic, Cleveland, Ohio, USA
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Dokyun Kim
- Department of Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute ,Cleveland Clinic, Cleveland, Ohio, USA
| | - Soowon Kang
- Department of Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute ,Cleveland Clinic, Cleveland, Ohio, USA
| | - Chih-Jen Lai
- Department of Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute ,Cleveland Clinic, Cleveland, Ohio, USA
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Inho Cha
- Department of Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Pei-Ching Chang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jae U. Jung
- Department of Cancer Biology and Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Global Center for Pathogen Research and Human Health, Lerner Research Institute ,Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
4
|
Singh RK, Torne AS, Robertson ES. Hypoxic reactivation of Kaposi's sarcoma associated herpesvirus. CELL INSIGHT 2024; 3:100200. [PMID: 39391006 PMCID: PMC11466537 DOI: 10.1016/j.cellin.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024]
Abstract
Hypoxic reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) refers to the phenomenon under low oxygen where the virus goes from latent to lytic replication. Typically, healthy cells generally cease cell division and DNA replication under hypoxic conditions due to limited resources, and the presence of physiological inhibitors. This restricted replication under hypoxic conditions is considered an employed strategy of the cell to minimize energy consumption. However, cancerous cells continuously replicate and divide in hypoxic conditions by reprogramming several aspects of their cell physiology, including but not limited to metabolism, cell cycle, DNA replication, transcription, translation, and the epigenome. KSHV infection, similar to cancerous cells, is known to bypass hypoxia-induced restrictions and undergo reactivation to produce progeny viruses. In previous studies we have mapped several aspects of cell physiology that are manipulated by KSHV through its latent antigens during hypoxic conditions, which allows for a permissive environment for its replication. We discuss the major strategies utilized by KSHV to bypass hypoxia-induced repression. We also describe the KSHV-encoded antigens responsible for modulating these cellular processes important for successful viral replication and persistence in hypoxia.
Collapse
Affiliation(s)
- Rajnish Kumar Singh
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Atharva S Torne
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| |
Collapse
|
5
|
Byren HC, Holzer A, Müller A, Korn K, Backovic M, Harrer EG, Bergmann S, Harrer T, Neipel F. Increased human herpesvirus-8 neutralizing response during remission from Kaposi's sarcoma. J Gen Virol 2024; 105. [PMID: 39565219 DOI: 10.1099/jgv.0.002044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
Human herpesvirus-8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus, is a human oncogenic herpesvirus that is responsible for several diseases including Kaposi's sarcoma (KS). KS prevalence varies dramatically, although emergence increases considerably with human immunodeficiency virus -1 (HIV-1) co-infection, making it one of the most common cancers in HIV-1 patients and sub-Saharan African men, even prior to the HIV-1 epidemic in Africa. Studies have shown that neutralizing antibodies exist in HHV-8-infected sera, which are most likely targeted to viral lytic surface glycoproteins, such as glycoprotein K8.1 (gpK8.1) and gHgL. Fifty-eight HHV-8-positive serum samples were tested for the levels of gpK8.1- and gHgL-binding antibodies and in vitro HHV-8-neutralizing capacity. Each sample was then categorized according to the disease status, which included asymptomatic infection, active KS and remission from KS, and the three measured parameters were compared between the disease groups. We show that neutralizing capacity in infected patient sera increases with remission of KS. Interestingly, antibodies targeting gpK8.1, but not gHgL, were also found to be increased during active disease and remission. Comparison of neutralizing capacity and antibody levels on an individual patient basis revealed that antibody levels, primarily targeting gHgL, are correlated with serum neutralizing response in sub-lingual Kaposi sarcoma (SLK) cells. Adsorption of gHgL or gpK8.1 antibodies from human sera removed the neutralizing response in SLK cells, although some non-specific removal of antibodies from the sera means that this result should be interpreted with caution. Taken collectively, these results suggest that glycoproteins, such as gHgL, are targets for neutralizing antibodies. Furthermore, our data imply that recovery from KS is associated with increased neutralizing capacity, suggesting that neutralizing antibodies may contribute to KS resolution. However, it is vital for further work to be completed in order to elucidate this relationship.
Collapse
Affiliation(s)
- Hannah C Byren
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Angela Holzer
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Alexander Müller
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Klaus Korn
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Marija Backovic
- Pasteur Institute, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Ellen G Harrer
- Infectious Diseases and Immunodeficiency Section, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Silke Bergmann
- Infectious Diseases and Immunodeficiency Section, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Thomas Harrer
- Infectious Diseases and Immunodeficiency Section, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Frank Neipel
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| |
Collapse
|
6
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Durocher Y. CHO cells for virus-like particle and subunit vaccine manufacturing. Vaccine 2024; 42:2530-2542. [PMID: 38503664 DOI: 10.1016/j.vaccine.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Chinese Hamster Ovary (CHO) cells, employed primarily for manufacturing monoclonal antibodies and other recombinant protein (r-protein) therapeutics, are emerging as a promising host for vaccine antigen production. This is exemplified by the recently approved CHO cell-derived subunit vaccines (SUV) against respiratory syncytial virus (RSV) and varicella-zoster virus (VZV), as well as the enveloped virus-like particle (eVLP) vaccine against hepatitis B virus (HBV). Here, we summarize the design, production, and immunogenicity features of these vaccine and review the most recent progress of other CHO-derived vaccines in pre-clinical and clinical development. We also discuss the challenges associated with vaccine production in CHO cells, with a focus on ensuring viral clearance for eVLP products.
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC H2X 3Y7, Canada.
| |
Collapse
|
7
|
Zhong L, Zhang W, Krummenacher C, Chen Y, Zheng Q, Zhao Q, Zeng MS, Xia N, Zeng YX, Xu M, Zhang X. Targeting herpesvirus entry complex and fusogen glycoproteins with prophylactic and therapeutic agents. Trends Microbiol 2023:S0966-842X(23)00077-X. [DOI: 10.1016/j.tim.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 04/03/2023]
|
8
|
Lam AK, Roshan R, Miley W, Labo N, Zhen J, Kurland AP, Cheng C, Huang H, Teng PL, Harelson C, Gong D, Tam YK, Radu CG, Epeldegui M, Johnson JR, Zhou ZH, Whitby D, Wu TT. Immunization of Mice with Virus-Like Vesicles of Kaposi Sarcoma-Associated Herpesvirus Reveals a Role for Antibodies Targeting ORF4 in Activating Complement-Mediated Neutralization. J Virol 2023; 97:e0160022. [PMID: 36757205 PMCID: PMC9972917 DOI: 10.1128/jvi.01600-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Infection by Kaposi sarcoma-associated herpesvirus (KSHV) can cause severe consequences, such as cancers and lymphoproliferative diseases. Whole inactivated viruses (WIV) with chemically destroyed genetic materials have been used as antigens in several licensed vaccines. During KSHV productive replication, virus-like vesicles (VLVs) that lack capsids and viral genomes are generated along with virions. Here, we investigated the immunogenicity of KSHV VLVs produced from a viral mutant that was defective in capsid formation and DNA packaging. Mice immunized with adjuvanted VLVs generated KSHV-specific T cell and antibody responses. Neutralization of KSHV infection by the VLV immune serum was low but was markedly enhanced in the presence of the complement system. Complement-enhanced neutralization and complement deposition on KSHV-infected cells was dependent on antibodies targeting viral open reading frame 4 (ORF4). However, limited complement-mediated enhancement was detected in the sera of a small cohort of KSHV-infected humans which contained few neutralizing antibodies. Therefore, vaccination that induces antibody effector functions can potentially improve infection-induced humoral immunity. Overall, our study highlights a potential benefit of engaging complement-mediated antibody functions in future KSHV vaccine development. IMPORTANCE KSHV is a virus that can lead to cancer after infection. A vaccine that prevents KSHV infection or transmission would be helpful in preventing the development of these cancers. We investigated KSHV VLV as an immunogen for vaccination. We determined that antibodies targeting the viral protein ORF4 induced by VLV immunization could engage the complement system and neutralize viral infection. However, ORF4-specific antibodies were seldom detected in the sera of KSHV-infected humans. Moreover, these human sera did not potently trigger complement-mediated neutralization, indicating an improvement that immunization can confer. Our study suggests a new antibody-mediated mechanism to control KSHV infection and underscores the benefit of activating the complement system in a future KSHV vaccine.
Collapse
Affiliation(s)
- Alex K. Lam
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Romin Roshan
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Wendell Miley
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Nazzarena Labo
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James Zhen
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Andrew P. Kurland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Celine Cheng
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Haigen Huang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Pu-Lin Teng
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Claire Harelson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Danyang Gong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, British Columbia, Canada
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Marta Epeldegui
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jeffrey R. Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Z. Hong Zhou
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Denise Whitby
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
9
|
Co-Infection of the Epstein-Barr Virus and the Kaposi Sarcoma-Associated Herpesvirus. Viruses 2022; 14:v14122709. [PMID: 36560713 PMCID: PMC9782805 DOI: 10.3390/v14122709] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
The two human tumor viruses, Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV), have been mostly studied in isolation. Recent studies suggest that co-infection with both viruses as observed in one of their associated malignancies, namely primary effusion lymphoma (PEL), might also be required for KSHV persistence. In this review, we discuss how EBV and KSHV might support each other for persistence and lymphomagenesis. Moreover, we summarize what is known about their innate and adaptive immune control which both seem to be required to ensure asymptomatic persistent co-infection with these two human tumor viruses. A better understanding of this immune control might allow us to prepare for vaccination against EBV and KSHV in the future.
Collapse
|
10
|
Casper C, Corey L, Cohen JI, Damania B, Gershon AA, Kaslow DC, Krug LT, Martin J, Mbulaiteye SM, Mocarski ES, Moore PS, Ogembo JG, Phipps W, Whitby D, Wood C. KSHV (HHV8) vaccine: promises and potential pitfalls for a new anti-cancer vaccine. NPJ Vaccines 2022; 7:108. [PMID: 36127367 PMCID: PMC9488886 DOI: 10.1038/s41541-022-00535-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/02/2022] [Indexed: 01/25/2023] Open
Abstract
Seven viruses cause at least 15% of the total cancer burden. Viral cancers have been described as the "low-hanging fruit" that can be potentially prevented or treated by new vaccines that would alter the course of global human cancer. Kaposi sarcoma herpesvirus (KSHV or HHV8) is the sole cause of Kaposi sarcoma, which primarily afflicts resource-poor and socially marginalized populations. This review summarizes a recent NIH-sponsored workshop's findings on the epidemiology and biology of KSHV as an overlooked but potentially vaccine-preventable infection. The unique epidemiology of this virus provides opportunities to prevent its cancers if an effective, inexpensive, and well-tolerated vaccine can be developed and delivered.
Collapse
Affiliation(s)
- Corey Casper
- Infectious Disease Research Institute, 1616 Eastlake Ave. East, Suite 400, Seattle, WA, 98102, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institutes of Health, Bldg. 50, Room 6134, 50 South Drive, MSC8007, Bethesda, MD, 20892-8007, USA
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center & Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, US
| | - Anne A Gershon
- Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY10032, US
| | - David C Kaslow
- PATH Essential Medicines, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA, USA
| | - Laurie T Krug
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Jeffrey Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Sam M Mbulaiteye
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, NIH, HHS, 9609 Medical Center Dr, Rm. 6E118 MSC 3330, Bethesda, MD, 20892, USA
| | | | - Patrick S Moore
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Warren Phipps
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Denise Whitby
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Charles Wood
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| |
Collapse
|
11
|
Tornesello AL, Tagliamonte M, Buonaguro FM, Tornesello ML, Buonaguro L. Virus-like Particles as Preventive and Therapeutic Cancer Vaccines. Vaccines (Basel) 2022; 10:227. [PMID: 35214685 PMCID: PMC8879290 DOI: 10.3390/vaccines10020227] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/04/2022] Open
Abstract
Virus-like particles (VLPs) are self-assembled viral protein complexes that mimic the native virus structure without being infectious. VLPs, similarly to wild type viruses, are able to efficiently target and activate dendritic cells (DCs) triggering the B and T cell immunities. Therefore, VLPs hold great promise for the development of effective and affordable vaccines in infectious diseases and cancers. Vaccine formulations based on VLPs, compared to other nanoparticles, have the advantage of incorporating multiple antigens derived from different proteins. Moreover, such antigens can be functionalized by chemical modifications without affecting the structural conformation or the antigenicity. This review summarizes the current status of preventive and therapeutic VLP-based vaccines developed against human oncoviruses as well as cancers.
Collapse
Affiliation(s)
- Anna Lucia Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (F.M.B.); (M.L.T.)
| | - Maria Tagliamonte
- Innovative Immunological Models, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.T.); (L.B.)
| | - Franco M. Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (F.M.B.); (M.L.T.)
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (F.M.B.); (M.L.T.)
| | - Luigi Buonaguro
- Innovative Immunological Models, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.T.); (L.B.)
| |
Collapse
|
12
|
A Pentavalent Epstein-Barr Virus-Like Particle Vaccine Elicits High Titers of Neutralizing Antibodies against Epstein-Barr Virus Infection in Immunized Rabbits. Vaccines (Basel) 2020; 8:vaccines8020169. [PMID: 32268575 PMCID: PMC7349562 DOI: 10.3390/vaccines8020169] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/16/2022] Open
Abstract
Primary infection with Epstein-Barr virus (EBV) is associated with acute infectious mononucleosis, whereas persistent infection is associated with chronic diseases such as autoimmune diseases and various types of cancer. Indeed, approximately 2% of all new cancer cases occurring annually worldwide are EBV-associated. Currently, there is no licensed EBV prophylactic vaccine. Selection of appropriate viral protein subunits is critical for development of an effective vaccine. Although the major EBV surface glycoprotein gp350/220 (gp350) has been proposed as an important prophylactic vaccine target, attempts to develop a potent vaccine based on gp350 alone have shown limited success in the clinic. We provide data showing that five EBV glycoproteins (gp350, gB, gp42, gH, and gL) involved in viral entry and infection can successfully be incorporated on the surface of EBV-like particles (EBV-LPs). These EBV-LPs, when administered together with aluminum hydroxide and monophosphoryl lipid A as adjuvants to New Zealand white rabbits, elicited EBV glycoprotein-specific antibodies capable of neutralizing viral infection in vitro in both B cells and epithelial cells, better than soluble gp350 ectodomain. Our findings suggest that a pentavalent EBV-LP formulation might be an ideal candidate for development as a safe and immunogenic EBV vaccine.
Collapse
|
13
|
The Kaposi's Sarcoma-Associated Herpesvirus (KSHV) gH/gL Complex Is the Predominant Neutralizing Antigenic Determinant in KSHV-Infected Individuals. Viruses 2020; 12:v12030256. [PMID: 32111001 PMCID: PMC7150787 DOI: 10.3390/v12030256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/14/2020] [Accepted: 02/23/2020] [Indexed: 12/24/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi’s sarcoma (KS), one of the most prevalent cancers of people living with HIV/AIDS in sub-Saharan Africa. The seroprevalence for KSHV is high in the region, and no prophylactic vaccine against the virus is available. In this study, we characterized the antigenic targets of KSHV-specific neutralizing antibodies (nAbs) in asymptomatic KSHV-infected individuals and KS patients with high nAbs titers. We quantified the extent to which various KSHV envelope glycoproteins (gB, ORF28, ORF68, gH, gL, gM, gN and gpK8.1) adsorbed/removed KSHV-specific nAbs from the plasma of infected individuals. Our study revealed that plasma from a majority of KSHV neutralizers recognizes multiple viral glycoproteins. Moreover, the breadth of nAbs responses against these viral glycoproteins varies among endemic KS, epidemic KS and asymptomatic KSHV-infected individuals. Importantly, among the KSHV glycoproteins, the gH/gL complex, but neither gH nor gL alone, showed the highest adsorption of KSHV-specific nAbs. This activity was detected in 80% of the KSHV-infected individuals regardless of their KS status. The findings suggest that the gH/gL complex is the predominant antigenic determinant of KSHV-specific nAbs. Therefore, gH/gL is a potential target for development of KSHV prophylactic vaccines.
Collapse
|
14
|
Kaposi Sarcoma-Associated Herpesvirus Glycoprotein H Is Indispensable for Infection of Epithelial, Endothelial, and Fibroblast Cell Types. J Virol 2019; 93:JVI.00630-19. [PMID: 31142670 DOI: 10.1128/jvi.00630-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is an emerging pathogen and is the causative infectious agent of Kaposi sarcoma and two malignancies of B cell origin. To date, there is no licensed KSHV vaccine. Development of an effective vaccine against KSHV continues to be limited by a poor understanding of how the virus initiates acute primary infection in vivo in diverse human cell types. The role of glycoprotein H (gH) in herpesvirus entry mechanisms remains largely unresolved. To characterize the requirement for KSHV gH in the viral life cycle and in determination of cell tropism, we generated and characterized a mutant KSHV in which expression of gH was abrogated. Using a bacterial artificial chromosome containing a complete recombinant KSHV genome and recombinant DNA technology, we inserted stop codons into the gH coding region. We used electron microscopy to reveal that the gH-null mutant virus assembled and exited from cells normally, compared to wild-type virus. Using purified virions, we assessed infectivity of the gH-null mutant in diverse mammalian cell types in vitro Unlike wild-type virus or a gH-containing revertant, the gH-null mutant was unable to infect any of the epithelial, endothelial, or fibroblast cell types tested. However, its ability to infect B cells was equivocal and remains to be investigated in vivo due to generally poor infectivity in vitro Together, these results suggest that gH is critical for KSHV infection of highly permissive cell types, including epithelial, endothelial, and fibroblast cells.IMPORTANCE All homologues of herpesvirus gH studied to date have been implicated in playing an essential role in viral infection of diverse permissive cell types. However, the role of gH in the mechanism of KSHV infection remains largely unresolved. In this study, we generated a gH-null mutant KSHV and provided evidence that deficiency of gH expression did not affect viral particle assembly or egress. Using the gH-null mutant, we showed that gH was indispensable for KSHV infection of epithelial, endothelial, and fibroblast cells in vitro This suggests that gH is an important target for the development of a KSHV prophylactic vaccine to prevent initial viral infection.
Collapse
|
15
|
Mulama DH, Mutsvunguma LZ, Totonchy J, Ye P, Foley J, Escalante GM, Rodriguez E, Nabiee R, Muniraju M, Wussow F, Barasa AK, Ogembo JG. A multivalent Kaposi sarcoma-associated herpesvirus-like particle vaccine capable of eliciting high titers of neutralizing antibodies in immunized rabbits. Vaccine 2019; 37:4184-4194. [PMID: 31201053 DOI: 10.1016/j.vaccine.2019.04.071] [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] [Received: 03/19/2019] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022]
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is an emerging pathogen and the causative agent of multiple cancers in immunocompromised patients. To date, there is no licensed prophylactic KSHV vaccine. In this study, we generated a novel subunit vaccine that incorporates four key KSHV envelope glycoproteins required for viral entry in diverse cell types (gpK8.1, gB, and gH/gL) into a single multivalent KSHV-like particle (KSHV-LP). Purified KSHV-LPs were similar in size, shape, and morphology to KSHV virions. Vaccination of rabbits with adjuvanted KSHV-LPs generated strong glycoprotein-specific antibody responses, and purified immunoglobulins from KSHV-LP-immunized rabbits neutralized KSHV infection in epithelial, endothelial, fibroblast, and B cell lines (60-90% at the highest concentration tested). These findings suggest that KSHV-LPs may be an ideal platform for developing a safe and effective prophylactic KSHV vaccine. We envision performing future studies in animal models that are susceptible to KSHV infection, to determine correlates of immune protection in vivo.
Collapse
Affiliation(s)
- David H Mulama
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States; Biological Sciences Department, Masinde Muliro University of Science and Technology, Kakamega, Kenya
| | - Lorraine Z Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | | | - Peng Ye
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Joslyn Foley
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Gabriela M Escalante
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Ramina Nabiee
- Chapman University, School of Pharmacy, Irvine, CA, United States
| | - Murali Muniraju
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Felix Wussow
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Anne K Barasa
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States; Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States.
| |
Collapse
|
16
|
Abstract
Kaposi sarcoma (KS) gained public attention as an AIDS-defining malignancy; its appearance on the skin was a highly stigmatizing sign of HIV infection during the height of the AIDS epidemic. The widespread introduction of effective antiretrovirals to control HIV by restoring immunocompetence reduced the prevalence of AIDS-related KS, although KS does occur in individuals with well-controlled HIV infection. KS also presents in individuals without HIV infection in older men (classic KS), in sub-Saharan Africa (endemic KS) and in transplant recipients (iatrogenic KS). The aetiologic agent of KS is KS herpesvirus (KSHV; also known as human herpesvirus-8), and viral proteins can induce KS-associated cellular changes that enable the virus to evade the host immune system and allow the infected cell to survive and proliferate despite viral infection. Currently, most cases of KS occur in sub-Saharan Africa, where KSHV infection is prevalent owing to transmission by saliva in childhood compounded by the ongoing AIDS epidemic. Treatment for early AIDS-related KS in previously untreated patients should start with the control of HIV with antiretrovirals, which frequently results in KS regression. In advanced-stage KS, chemotherapy with pegylated liposomal doxorubicin or paclitaxel is the most common treatment, although it is seldom curative. In sub-Saharan Africa, KS continues to have a poor prognosis. Newer treatments for KS based on the mechanisms of its pathogenesis are being explored.
Collapse
Affiliation(s)
- Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Blossom Damania
- Department of Microbiology and Immunology, Lineberger Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - Jeffrey Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Mark Bower
- National Centre for HIV Malignancy, Chelsea & Westminster Hospital, London, UK
| | - Denise Whitby
- Leidos Biomedical Research, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| |
Collapse
|