1
|
Zhang B, Mao H, Zhu H, Guo J, Zhou P, Ma Z. Response to HIV-1 gp160-carrying recombinant virus HSV-1 and HIV-1 VLP combined vaccine in BALB/c mice. Front Microbiol 2023; 14:1136664. [PMID: 37007461 PMCID: PMC10063819 DOI: 10.3389/fmicb.2023.1136664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
Human immunodeficiency virus (HIV) induced AIDS causes a large number of infections and deaths worldwide every year, still no vaccines are available to prevent infection. Recombinant herpes simplex virus type 1 (HSV-1) vector-based vaccines coding the target proteins of other pathogens have been widely used for disease control. Here, a recombinant virus with HIV-1 gp160 gene integration into the internal reverse (IR) region-deleted HSV-1 vector (HSV-BAC), was obtained by bacterial artificial chromosome (BAC) technology, and its immunogenicity investigated in BALB/c mice. The result showed similar replication ability of the HSV-BAC-based recombinant virus and wild type. Furthermore, humoral and cellular immune response showed superiority of intraperitoneal (IP) administration, compared to intranasally (IN), subcutaneous (SC) and intramuscularly (IM), that evidenced by production of significant antibody and T cell responses. More importantly, in a prime-boost combination study murine model, the recombinant viruses prime followed by HIV-1 VLP boost induced stronger and broader immune responses than single virus or protein vaccination in a similar vaccination regimen. Antibody production was sufficient with huge potential for viral clearance, along with efficient T-cell activation, which were evaluated by the enzyme-linked immunosorbent assay (ELISA) and flow cytometry (FC). Overall, these findings expose the value of combining different vaccine vectors and modalities to improve immunogenicity and breadth against different HIV-1 antigens.
Collapse
Affiliation(s)
- Beibei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Ürümqi, Xinjiang, China
| | - Hongyan Mao
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Ürümqi, Xinjiang, China
| | - Hongjuan Zhu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Ürümqi, Xinjiang, China
| | - Jingxia Guo
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Ürümqi, Xinjiang, China
| | - Paul Zhou
- Unit of Antiviral Immunity and Genetic Therapy, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhenghai Ma
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Ürümqi, Xinjiang, China
- *Correspondence: Zhenghai Ma,
| |
Collapse
|
2
|
Toledo NPL, Li H, Omange RW, Dacoba TG, Crecente-Campo J, Schalk D, Kashem MA, Rakasz E, Schultz-Darken N, Li Q, Whitney JB, Alonso MJ, Plummer FA, Luo M. Cervico-Vaginal Inflammatory Cytokine and Chemokine Responses to Two Different SIV Immunogens. Front Immunol 2020; 11:1935. [PMID: 32983121 PMCID: PMC7477078 DOI: 10.3389/fimmu.2020.01935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022] Open
Abstract
Studies have shown that vaccine vectors and route of immunization can differentially activate different arms of the immune system. However, the effects of different HIV vaccine immunogens on mucosal inflammation have not yet been studied. Because mucosal sites are the primary route of HIV infection, we evaluated the cervico-vaginal inflammatory cytokine and chemokine levels of Mauritian cynomolgus macaques following immunization and boost using two different SIV vaccine immunogens. The PCS vaccine delivers 12 20-amino acid peptides overlapping the 12 protease cleavage sites, and the Gag/Env vaccine delivers the full Gag and full Env proteins of simian immunodeficiency virus. We showed that the PCS vaccine prime and boosts induced short-lived, lower level increases of a few pro-inflammatory/chemotactic cytokines. In the PCS-vaccine group only the levels of MCP-1 were significantly increased above the baseline (P = 0.0078, Week 6; P = 0.0078, Week 17; P = 0.0234; Week 51) following multiple boosts. In contrast, immunizations with the Gag/Env vaccine persistently increased the levels of multiple cytokines/chemokines. In the Gag/Env group, higher than baseline levels were consistently observed for IL-8 (P = 0.0078, Week 16; P = 0.0078, Week 17; P = 0.0156, Week 52), IL-1β (P = 0.0234, Week 16; P = 0.0156, Week 17; P = 0.0156, Week 52), and MIP-1α (P = 0.0313, Week 16; P = 0.0156, Week 17; P = 0.0313, Week 52). Over time, repeated boosts altered the relative levels of these cytokines between the Gag/Env and PCS vaccine group. 18 weeks after final boost with a higher dosage, IP-10 levels (P = 0.0313) in the Gag/Env group remained higher than baseline. Thus, the influence of vaccine immunogens on mucosal inflammation needs to be considered when developing and evaluating candidate HIV vaccines.
Collapse
Affiliation(s)
- Nikki P L Toledo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Robert W Omange
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Dane Schalk
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, WI, United States
| | - Mohammad A Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Eva Rakasz
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, WI, United States
| | - Nancy Schultz-Darken
- Scientific Protocol Implementation Unit, Wisconsin National Primate Research Center, Madison, WI, United States
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Maria J Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| |
Collapse
|
3
|
Velashjerdi Farahani S, Reza Aghasadeghi M, Memarnejadian A, Faezi S, Shahosseini Z, Mahdavi M. Naloxone/alum mixture a potent adjuvant for HIV-1 vaccine: induction of cellular and poly-isotypic humoral immune responses. Pathog Glob Health 2016; 110:39-47. [PMID: 26403975 DOI: 10.1179/2047773215y.0000000035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In the present study we used a fusion peptide from HIV-1 p24 and Nef as vaccine model and adjuvant activity of Naloxone/alum mixture was evaluated in a peptide vaccine model. HIV-1 p24-Nef fusion peptide was synthesized. Female BALB/c mice were divided into five groups. The first group immunized subcutaneously with the p24-Nef fusion peptide adjuvanted with Naloxone/alum mixture and boosted with same protocol. The second was immunized with fusion peptide adjuvanted in alum. The control groups were injected with NLX (Group 3), Alum (Group 4), or PBS (Groups 5) under the same conditions. To determine the type of induced immune response, sera and splenocytes were analyzed by commercial ELISA method for total IgG and isotypes and cytokine secretion (IL-4 & IFN-γ), respectively. We have also used the ELISPOT assay to monitor changes in the frequency of IFN-γ-producing T cells. The proliferation of T cells was assessed using Brdu method and T-cell cytotoxicity was assessed with CFSE method. Immunization of mice with HIV-1 p24-Nef fusion peptide formulated in Naloxone/alum mixture significantly increased lymphocyte proliferation and shifted cytokine responses toward Th1 profile compared to all other groups. Analysis of humoral immune responses revealed that administration of HIV-1 p24-Nef fusion peptide with Naloxone/alum mixture significantly increased specific IgG responses and also increased IgG1,IgG2a, IgG2b, IgG3, and IgM vs. alum-adjuvanted vaccine groups. Naloxone/alum mixture as an adjuvant could improve cellular and humoral immune response for HIV vaccine model and this adjuvant maybe useful for HIV vaccine model in human clinical trial.
Collapse
Affiliation(s)
- Sima Velashjerdi Farahani
- a Department of Immunology , Pasteur Institute of Iran , Tehran , Iran.,b Faculty of Sciences, Department of Microbiology , Zanjan Islamic Azad University , Zanjan , Iran
| | | | - Arash Memarnejadian
- c Department of Hepatitis and AIDS , Pasteur Institute of Iran , Tehran , Iran
| | - Sobhan Faezi
- d Department of Mycobacteriology and Pulmonary Research , Pasteur Institute of Iran , Tehran , Iran
| | - Zahra Shahosseini
- e Department of Virology , Pasteur Institute of Iran , Tehran , Iran
| | - Mehdi Mahdavi
- a Department of Immunology , Pasteur Institute of Iran , Tehran , Iran
| |
Collapse
|
4
|
Lentiviral Protein Transfer Vectors Are an Efficient Vaccine Platform and Induce a Strong Antigen-Specific Cytotoxic T Cell Response. J Virol 2015; 89:9044-60. [PMID: 26085166 DOI: 10.1128/jvi.00844-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/14/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED To induce and trigger innate and adaptive immune responses, antigen-presenting cells (APCs) take up and process antigens. Retroviral particles are capable of transferring not only genetic information but also foreign cargo proteins when they are genetically fused to viral structural proteins. Here, we demonstrate the capacity of lentiviral protein transfer vectors (PTVs) for targeted antigen transfer directly into APCs and thereby induction of cytotoxic T cell responses. Targeting of lentiviral PTVs to APCs can be achieved analogously to gene transfer vectors by pseudotyping the particles with truncated wild-type measles virus (MV) glycoproteins (GPs), which use human SLAM (signaling lymphocyte activation molecule) as a main entry receptor. SLAM is expressed on stimulated lymphocytes and APCs, including dendritic cells. SLAM-targeted PTVs transferred the reporter protein green fluorescent protein (GFP) or Cre recombinase with strict receptor specificity into SLAM-expressing CHO and B cell lines, in contrast to broadly transducing vesicular stomatitis virus G protein (VSV-G) pseudotyped PTVs. Primary myeloid dendritic cells (mDCs) incubated with targeted or nontargeted ovalbumin (Ova)-transferring PTVs stimulated Ova-specific T lymphocytes, especially CD8(+) T cells. Administration of Ova-PTVs into SLAM-transgenic and control mice confirmed the observed predominant induction of antigen-specific CD8(+) T cells and demonstrated the capacity of protein transfer vectors as suitable vaccines for the induction of antigen-specific immune responses. IMPORTANCE This study demonstrates the specificity and efficacy of antigen transfer by SLAM-targeted and nontargeted lentiviral protein transfer vectors into antigen-presenting cells to trigger antigen-specific immune responses in vitro and in vivo. The observed predominant activation of antigen-specific CD8(+) T cells indicates the suitability of SLAM-targeted and also nontargeted PTVs as a vaccine for the induction of cytotoxic immune responses. Since cytotoxic CD8(+) T lymphocytes are a mainstay of antitumoral immune responses, PTVs could be engineered for the transfer of specific tumor antigens provoking tailored antitumoral immunity. Therefore, PTVs can be used as safe and efficient alternatives to gene transfer vectors or live attenuated replicating vector platforms, avoiding genotoxicity or general toxicity in highly immunocompromised patients, respectively. Thereby, the potential for easy envelope exchange allows the circumventing of neutralizing antibodies, e.g., during repeated boost immunizations.
Collapse
|
5
|
Pfaller CK, Cattaneo R, Schnell MJ. Reverse genetics of Mononegavirales: How they work, new vaccines, and new cancer therapeutics. Virology 2015; 479-480:331-44. [PMID: 25702088 DOI: 10.1016/j.virol.2015.01.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 01/26/2015] [Accepted: 01/30/2015] [Indexed: 12/24/2022]
Abstract
The order Mononegavirales includes five families: Bornaviridae, Filoviridae, Nyamaviridae, Paramyxoviridae, and Rhabdoviridae. The genome of these viruses is one molecule of negative-sense single strand RNA coding for five to ten genes in a conserved order. The RNA is not infectious until packaged by the nucleocapsid protein and transcribed by the polymerase and co-factors. Reverse genetics approaches have answered fundamental questions about the biology of Mononegavirales. The lack of icosahedral symmetry and modular organization in the genome of these viruses has facilitated engineering of viruses expressing fluorescent proteins, and these fluorescent proteins have provided important insights about the molecular and cellular basis of tissue tropism and pathogenesis. Studies have assessed the relevance for virulence of different receptors and the interactions with cellular proteins governing the innate immune responses. Research has also analyzed the mechanisms of attenuation. Based on these findings, ongoing clinical trials are exploring new live attenuated vaccines and the use of viruses re-engineered as cancer therapeutics.
Collapse
Affiliation(s)
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Philadelphia, PA 19107, USA; Jefferson Vaccine Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
6
|
Ondondo BO. The influence of delivery vectors on HIV vaccine efficacy. Front Microbiol 2014; 5:439. [PMID: 25202303 PMCID: PMC4141443 DOI: 10.3389/fmicb.2014.00439] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/03/2014] [Indexed: 12/31/2022] Open
Abstract
Development of an effective HIV/AIDS vaccine remains a big challenge, largely due to the enormous HIV diversity which propels immune escape. Thus novel vaccine strategies are targeting multiple variants of conserved antibody and T cell epitopic regions which would incur a huge fitness cost to the virus in the event of mutational escape. Besides immunogen design, the delivery modality is critical for vaccine potency and efficacy, and should be carefully selected in order to not only maximize transgene expression, but to also enhance the immuno-stimulatory potential to activate innate and adaptive immune systems. To date, five HIV vaccine candidates have been evaluated for efficacy and protection from acquisition was only achieved in a small proportion of vaccinees in the RV144 study which used a canarypox vector for delivery. Conversely, in the STEP study (HVTN 502) where human adenovirus serotype 5 (Ad5) was used, strong immune responses were induced but vaccination was more associated with increased risk of HIV acquisition than protection in vaccinees with pre-existing Ad5 immunity. The possibility that pre-existing immunity to a highly promising delivery vector may alter the natural course of HIV to increase acquisition risk is quite worrisome and a huge setback for HIV vaccine development. Thus, HIV vaccine development efforts are now geared toward delivery platforms which attain superior immunogenicity while concurrently limiting potential catastrophic effects likely to arise from pre-existing immunity or vector-related immuno-modulation. However, it still remains unclear whether it is poor immunogenicity of HIV antigens or substandard immunological potency of the safer delivery vectors that has limited the success of HIV vaccines. This article discusses some of the promising delivery vectors to be harnessed for improved HIV vaccine efficacy.
Collapse
Affiliation(s)
- Beatrice O Ondondo
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford Oxford, UK
| |
Collapse
|
7
|
Kwon KC, Verma D, Singh ND, Herzog R, Daniell H. Oral delivery of human biopharmaceuticals, autoantigens and vaccine antigens bioencapsulated in plant cells. Adv Drug Deliv Rev 2013; 65:782-99. [PMID: 23099275 PMCID: PMC3582797 DOI: 10.1016/j.addr.2012.10.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 09/26/2012] [Accepted: 10/17/2012] [Indexed: 12/19/2022]
Abstract
Among 12billion injections administered annually, unsafe delivery leads to >20million infections and >100million reactions. In an emerging new concept, freeze-dried plant cells (lettuce) expressing vaccine antigens/biopharmaceuticals are protected in the stomach from acids/enzymes but are released to the immune or blood circulatory system when plant cell walls are digested by microbes that colonize the gut. Vaccine antigens bioencapsulated in plant cells upon oral delivery after priming, conferred both mucosal and systemic immunity and protection against bacterial, viral or protozoan pathogens or toxin challenge. Oral delivery of autoantigens was effective against complications of type 1 diabetes and hemophilia, by developing tolerance. Oral delivery of proinsulin or exendin-4 expressed in plant cells regulated blood glucose levels similar to injections. Therefore, this new platform offers a low cost alternative to deliver different therapeutic proteins to combat infectious or inherited diseases by eliminating inactivated pathogens, expensive purification, cold storage/transportation and sterile injections.
Collapse
Affiliation(s)
- Kwang-Chul Kwon
- Department of Molecular Biology and Microbiology, College of Medicine, University of Central Florida, Biomolecular Science Building, Orlando, FL 32816-2364, USA
| | - Dheeraj Verma
- Department of Molecular Biology and Microbiology, College of Medicine, University of Central Florida, Biomolecular Science Building, Orlando, FL 32816-2364, USA
| | - Nameirakpam D. Singh
- Department of Molecular Biology and Microbiology, College of Medicine, University of Central Florida, Biomolecular Science Building, Orlando, FL 32816-2364, USA
| | - Roland Herzog
- Department of Pediatrics, College of Medicine, University of Florida, Cancer and Genetics Research Complex, 2033 Mowry Road, Gainesville, FL 32610, USA
| | - Henry Daniell
- Department of Molecular Biology and Microbiology, College of Medicine, University of Central Florida, Biomolecular Science Building, Orlando, FL 32816-2364, USA
| |
Collapse
|
8
|
Knuschke T, Sokolova V, Rotan O, Wadwa M, Tenbusch M, Hansen W, Staeheli P, Epple M, Buer J, Westendorf AM. Immunization with Biodegradable Nanoparticles Efficiently Induces Cellular Immunity and Protects against Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2013; 190:6221-9. [DOI: 10.4049/jimmunol.1202654] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
9
|
The synergistic effect of combined immunization with a DNA vaccine and chimeric yellow fever/dengue virus leads to strong protection against dengue. PLoS One 2013; 8:e58357. [PMID: 23472186 PMCID: PMC3589436 DOI: 10.1371/journal.pone.0058357] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 02/06/2013] [Indexed: 12/13/2022] Open
Abstract
The dengue envelope glycoprotein (E) is the major component of virion surface and its ectodomain is composed of domains I, II and III. This protein is the main target for the development of a dengue vaccine with induction of neutralizing antibodies. In the present work, we tested two different vaccination strategies, with combined immunizations in a prime/booster regimen or simultaneous inoculation with a DNA vaccine (pE1D2) and a chimeric yellow fever/dengue 2 virus (YF17D-D2). The pE1D2 DNA vaccine encodes the ectodomain of the envelope DENV2 protein fused to t-PA signal peptide, while the YF17D-D2 was constructed by replacing the prM and E genes from the 17D yellow fever vaccine virus by those from DENV2. Balb/c mice were inoculated with these two vaccines by different prime/booster or simultaneous immunization protocols and most of them induced a synergistic effect on the elicited immune response, mainly in neutralizing antibody production. Furthermore, combined immunization remarkably increased protection against a lethal dose of DENV2, when compared to each vaccine administered alone. Results also revealed that immunization with the DNA vaccine, regardless of the combination with the chimeric virus, induced a robust cell immune response, with production of IFN-γ by CD8+ T lymphocytes.
Collapse
|
10
|
Kar UK, Jiang J, Champion CI, Salehi S, Srivastava M, Sharma S, Rabizadeh S, Niazi K, Kickhoefer V, Rome LH, Kelly KA. Vault nanocapsules as adjuvants favor cell-mediated over antibody-mediated immune responses following immunization of mice. PLoS One 2012; 7:e38553. [PMID: 22808011 PMCID: PMC3394761 DOI: 10.1371/journal.pone.0038553] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 05/11/2012] [Indexed: 01/12/2023] Open
Abstract
Background Modifications of adjuvants that induce cell-mediated over antibody-mediated immunity is desired for development of vaccines. Nanocapsules have been found to be viable adjuvants and are amenable to engineering for desired immune responses. We previously showed that natural nanocapsules called vaults can be genetically engineered to elicit Th1 immunity and protection from a mucosal bacterial infection. The purpose of our study was to characterize immunity produced in response to OVA within vault nanoparticles and compare it to another nanocarrier. Methodology and Principal Findings We characterized immunity resulting from immunization with the model antigen, ovalbumin (OVA) encased in vault nanocapsules and liposomes. We measured OVA responsive CD8+ and CD4+ memory T cell responses, cytokine production and antibody titers in vitro and in vivo. We found that immunization with OVA contain in vaults induced a greater number of anti-OVA CD8+ memory T cells and production of IFNγ plus CD4+ memory T cells. Also, modification of the vault body could change the immune response compared to OVA encased in liposomes. Conclusions/Significance These experiments show that vault nanocapsules induced strong anti-OVA CD8+ and CD4+ T cell memory responses and modest antibody production, which markedly differed from the immune response induced by liposomes. We also found that the vault nanocapsule could be modified to change antibody isotypes in vivo. Thus it is possible to create a vault nanocapsule vaccine that can result in the unique combination of immunogen-responsive CD8+ and CD4+ T cell immunity coupled with an IgG1 response for future development of vault nanocapsule-based vaccines against antigens for human pathogens and cancer.
Collapse
Affiliation(s)
- Upendra K. Kar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Janina Jiang
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Cheryl I. Champion
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Sahar Salehi
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Minu Srivastava
- Molecular Medicine Laboratory, Veteran’s Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Sherven Sharma
- Molecular Medicine Laboratory, Veteran’s Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
| | - Shahrooz Rabizadeh
- Department of Bioengineering, Samueli School of Engineering, University of California Los Angeles, Los Angeles, California, United States of America
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kayvan Niazi
- Department of Bioengineering, Samueli School of Engineering, University of California Los Angeles, Los Angeles, California, United States of America
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Valerie Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Leonard H. Rome
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kathleen A. Kelly
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
11
|
Cox JH, Ferrari MG, Earl P, Lane JR, Jagodzinski LL, Polonis VR, Kuta EG, Boyer JD, Ratto-Kim S, Eller LA, Pham DT, Hart L, Montefiori D, Ferrari G, Parrish S, Weiner DB, Moss B, Kim JH, Birx D, VanCott TC. Inclusion of a CRF01_AE HIV envelope protein boost with a DNA/MVA prime-boost vaccine: Impact on humoral and cellular immunogenicity and viral load reduction after SHIV-E challenge. Vaccine 2012; 30:1830-40. [PMID: 22234262 PMCID: PMC3324265 DOI: 10.1016/j.vaccine.2011.12.131] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 12/21/2011] [Accepted: 12/28/2011] [Indexed: 01/13/2023]
Abstract
The current study assessed the immunogenicity and protective efficacy of various prime-boost vaccine regimens in rhesus macaques using combinations of recombinant DNA (rDNA), recombinant MVA (rMVA), and subunit gp140 protein. The rDNA and rMVA vectors were constructed to express Env from HIV-1 subtype CRF01_AE and Gag-Pol from CRF01_AE or SIVmac 239. One of the rMVAs, MVA/CMDR, has been recently tested in humans. Immunizations were administered at months 0 and 1 (prime) and months 3 and 6 (boost). After priming, HIV env-specific serum IgG was detected in monkeys receiving gp140 alone or rMVA but not in those receiving rDNA. Titers were enhanced in these groups after boosting either with gp140 alone or with rMVA plus gp140. The groups that received the rDNA prime developed env-specific IgG after boosting with rMVA with or without gp140. HIV Env-specific serum IgG binding antibodies were elicited more frequently and of higher titer, and breadth of neutralizing antibodies was increased with the inclusion of the subunit Env boost. T cell responses were measured by tetramer binding to Gag p11c in Mamu-A*01 macaques, and by IFN-γ ELISPOT assay to SIV-Gag. T cell responses were induced after vaccination with the highest responses seen in macaques immunized with rDNA and rMVA. Macaques were challenged intravenously with a novel SHIV-E virus (SIVmac239 Gag-Pol with an HIV-1 subtype E-Env CAR402). Post challenge with SHIV-E, antibody titers were boosted in all groups and peaked at 4 weeks. Robust T cell responses were seen in all groups post challenge and in macaques immunized with rDNA and rMVA a clear boosting of responses was seen. A greater than two-log drop in RNA copies/ml at peak viremia and earlier set point was achieved in macaques primed with rDNA, and boosted with rMVA/SHIV-AE plus gp140. Post challenge viremia in macaques immunized with other regimens was not significantly different to that of controls. These results demonstrate that a gp140 subunit and inclusion of SIV Gag-Pol may be critical for control of SHIV post challenge.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Antibodies, Neutralizing/blood
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Products, gag/immunology
- Gene Products, pol/immunology
- HIV Antibodies/blood
- HIV-1/immunology
- Immunity, Cellular
- Immunity, Humoral
- Immunization, Secondary
- Immunoglobulin G/blood
- Macaca mulatta
- Male
- Simian Immunodeficiency Virus/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- env Gene Products, Human Immunodeficiency Virus/immunology
Collapse
|
12
|
The immune response to a vesicular stomatitis virus vaccine vector is independent of particulate antigen secretion and protein turnover rate. J Virol 2012; 86:4253-61. [PMID: 22345454 DOI: 10.1128/jvi.05991-11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Vesicular stomatitis virus (VSV) is a highly cytopathic virus being developed as a vaccine vector due to its ability to induce strong protective T cell and antibody responses after a single dose. However, little is known regarding the mechanisms underlying the potent immune responses elicited by VSV. We previously generated a VSV vector expressing the hepatitis B virus middle envelope surface glycoprotein (MS) that induces strong MS-specific T cell and antibody responses in mice. After synthesis in the cytoplasm, the MS protein translocates to the endoplasmic reticulum, where it forms subviral particles that are secreted from the cell. To better understand the contributions of secreted and intracellular protein to the VSV-induced immune response, we produced a vector expressing a secretion-deficient MS mutant (MS(C69A)) and compared the immunogenicity of this vector to that of the wild-type VSV-MS vector in mice. As expected, the MS(C69A) protein was not secreted from VSV-infected cells and displayed enhanced proteasome-mediated degradation. Surprisingly, despite these differences in intracellular protein processing, the T cell and antibody responses generated to MS(C69A) were comparable to those elicited by virus expressing wild-type MS protein. Therefore, when it is expressed from VSV, the immune responses to MS are independent of particulate antigen secretion and the turnover rate of cytoplasmic protein. These results are consistent with a model in which the immune responses to VSV are strongly influenced by the replication cycle of the vector and demonstrate that characteristics of the vector have the capacity to affect vaccine efficacy more than do the properties of the antigen itself.
Collapse
|
13
|
Gonzalez-Rabade N, McGowan EG, Zhou F, McCabe MS, Bock R, Dix PJ, Gray JC, Ma JKC. Immunogenicity of chloroplast-derived HIV-1 p24 and a p24-Nef fusion protein following subcutaneous and oral administration in mice. PLANT BIOTECHNOLOGY JOURNAL 2011; 9:629-38. [PMID: 21443546 DOI: 10.1111/j.1467-7652.2011.00609.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
High-level expression of foreign proteins in chloroplasts of transplastomic plants provides excellent opportunities for the development of oral vaccines against a range of debilitating or fatal diseases. The HIV-1 capsid protein p24 and a fusion of p24 with the negative regulatory protein Nef (p24-Nef) accumulate to ∼4% and ∼40% of the total soluble protein of leaves of transplastomic tobacco (Nicotiana tabacum L.) plants. This study has investigated the immunogenicity in mice of these two HIV-1 proteins, using cholera toxin B subunit as an adjuvant. Subcutaneous immunization with purified chloroplast-derived p24 elicited a strong antigen-specific serum IgG response, comparable to that produced by Escherichia coli-derived p24. Oral administration of a partially purified preparation of chloroplast-derived p24-Nef fusion protein, used as a booster after subcutaneous injection with either p24 or Nef, also elicited strong antigen-specific serum IgG responses. Both IgG1 and IgG2a subtypes, associated with cell-mediated Th1 and humoral Th2 responses, respectively, were found in sera after subcutaneous and oral administration. These results indicate that chloroplast-derived HIV-1 p24-Nef is a promising candidate as a component of a subunit vaccine delivered by oral boosting, after subcutaneous priming by injection of p24 and/or Nef.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Adjuvants, Immunologic/administration & dosage
- Administration, Oral
- Animals
- Chloroplasts/genetics
- Chloroplasts/immunology
- Female
- HIV Core Protein p24/administration & dosage
- HIV Core Protein p24/genetics
- HIV Core Protein p24/immunology
- Immunity, Humoral/immunology
- Immunization, Secondary
- Injections, Subcutaneous
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nicotine/toxicity
- Plants, Genetically Modified/genetics
- Nicotiana/genetics
- nef Gene Products, Human Immunodeficiency Virus/administration & dosage
- nef Gene Products, Human Immunodeficiency Virus/genetics
- nef Gene Products, Human Immunodeficiency Virus/immunology
Collapse
|
14
|
Significant protection against high-dose simian immunodeficiency virus challenge conferred by a new prime-boost vaccine regimen. J Virol 2011; 85:5764-72. [PMID: 21490100 DOI: 10.1128/jvi.00342-11] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We constructed vaccine vectors based on live recombinant vesicular stomatitis virus (VSV) and a Semliki Forest virus (SFV) replicon (SFVG) that propagates through expression of the VSV glycoprotein (G). These vectors expressing simian immunodeficiency virus (SIV) Gag and Env proteins were used to vaccinate rhesus macaques with a new heterologous prime-boost regimen designed to optimize induction of antibody. Six vaccinated animals and six controls were then given a high-dose mucosal challenge with the diverse SIVsmE660 quasispecies. All control animals became infected and had peak viral RNA loads of 10(6) to 10(8) copies/ml. In contrast, four of the vaccinees showed significant (P = 0.03) apparent sterilizing immunity and no detectable viral loads. Subsequent CD8(+) T cell depletion confirmed the absence of SIV infection in these animals. The two other vaccinees had peak viral loads of 7 × 10(5) and 8 × 10(3) copies/ml, levels below those of all of the controls, and showed undetectable virus loads by day 42 postchallenge. The vaccine regimen induced high-titer prechallenge serum neutralizing antibodies (nAbs) to some cloned SIVsmE660 Env proteins, but antibodies able to neutralize the challenge virus swarm were not detected. The cellular immune responses induced by the vaccine were generally weak and did not correlate with protection. Although the immune correlates of protection are not yet clear, the heterologous VSV/SFVG prime-boost is clearly a potent vaccine regimen for inducing virus nAbs and protection against a heterogeneous viral swarm.
Collapse
|
15
|
Moon JJ, Suh H, Bershteyn A, Stephan MT, Liu H, Huang B, Sohail M, Luo S, Um SH, Khant H, Goodwin JT, Ramos J, Chiu W, Irvine DJ. Interbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responses. NATURE MATERIALS 2011; 10:243-51. [PMID: 21336265 PMCID: PMC3077947 DOI: 10.1038/nmat2960] [Citation(s) in RCA: 424] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 01/11/2011] [Indexed: 05/17/2023]
Abstract
Vaccines based on recombinant proteins avoid the toxicity and antivector immunity associated with live vaccine (for example, viral) vectors, but their immunogenicity is poor, particularly for CD8(+) T-cell responses. Synthetic particles carrying antigens and adjuvant molecules have been developed to enhance subunit vaccines, but in general these materials have failed to elicit CD8(+) T-cell responses comparable to those for live vectors in preclinical animal models. Here, we describe interbilayer-crosslinked multilamellar vesicles formed by crosslinking headgroups of adjacent lipid bilayers within multilamellar vesicles. Interbilayer-crosslinked vesicles stably entrapped protein antigens in the vesicle core and lipid-based immunostimulatory molecules in the vesicle walls under extracellular conditions, but exhibited rapid release in the presence of endolysosomal lipases. We found that these antigen/adjuvant-carrying vesicles form an extremely potent whole-protein vaccine, eliciting endogenous T-cell and antibody responses comparable to those for the strongest vaccine vectors. These materials should enable a range of subunit vaccines and provide new possibilities for therapeutic protein delivery.
Collapse
Affiliation(s)
- James J Moon
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
A major hurdle in the development of a global HIV-1 vaccine is viral diversity. For close to three decades, HIV vaccine development has focused on either the induction of T cell immune responses or antibody responses, and only rarely on both components. After the failure of the STEP trial, the scientific community concluded that a T cell-based vaccine would likely not be protective if the T cell immune responses were elicited against only a few dominant epitopes. Similarly, for vaccines focusing on antibody responses, one of the main criticisms after VaxGen's failed Phase III trials was on the limited antigen breadth included in the two formulations used. The successes of polyvalent vaccine approaches against other antigenically variable pathogens encourage implementation of the same approach for the design of HIV-1 vaccines. A review of the existing HIV-1 vaccination approaches based on the polyvalent principle is included here to provide a historical perspective for the current effort of developing a polyvalent HIV-1 vaccine. Results summarized in this review provide a clear indication that the polyvalent approach is a viable one for the future development of an effective HIV vaccine.
Collapse
Affiliation(s)
- Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | | | |
Collapse
|
17
|
Potent vesicular stomatitis virus-based avian influenza vaccines provide long-term sterilizing immunity against heterologous challenge. J Virol 2010; 84:4611-8. [PMID: 20181720 DOI: 10.1128/jvi.02637-09] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The emergence in 1997 and continuance today of a highly lethal H5N1 avian influenza virus (AIV) causing human disease has raised concern about an impending pandemic and the need for a vaccine to prepare for such an occurrence. We previously generated an efficacious vesicular stomatitis virus (VSV)-based AIV vaccine expressing H5 hemagglutinin (HA) from the fifth genomic position of VSV (J. A. Schwartz et al., Virology 366:166-173, 2007). Here we have generated and characterized VSV-based vaccines that express the A/Hong Kong/156/1997 (clade 0) H5 HA from the first position of the VSV genome. These vectors induce broadly cross-neutralizing antibodies against homologous and heterologous H5N1 viruses of different clades in mice. The vaccines provide complete protection against morbidity and mortality after heterologous challenge with clade 0 and clade 1 strains in animals even 1 year after vaccination. Postchallenge pulmonary virus loads show that these vectors provide sterilizing immunity. Therefore, VSV-based AIV vaccines are potent, broadly cross-protective pandemic vaccine candidates.
Collapse
|
18
|
Brown SA, Surman SL, Sealy R, Jones BG, Slobod KS, Branum K, Lockey TD, Howlett N, Freiden P, Flynn P, Hurwitz JL. Heterologous Prime-Boost HIV-1 Vaccination Regimens in Pre-Clinical and Clinical Trials. Viruses 2010; 2:435-467. [PMID: 20407589 PMCID: PMC2855973 DOI: 10.3390/v2020435] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 12/21/2022] Open
Abstract
Currently, there are more than 30 million people infected with HIV-1 and thousands more are infected each day. Vaccination is the single most effective mechanism for prevention of viral disease, and after more than 25 years of research, one vaccine has shown somewhat encouraging results in an advanced clinical efficacy trial. A modified intent-to-treat analysis of trial results showed that infection was approximately 30% lower in the vaccine group compared to the placebo group. The vaccine was administered using a heterologous prime-boost regimen in which both target antigens and delivery vehicles were changed during the course of inoculations. Here we examine the complexity of heterologous prime-boost immunizations. We show that the use of different delivery vehicles in prime and boost inoculations can help to avert the inhibitory effects caused by vector-specific immune responses. We also show that the introduction of new antigens into boost inoculations can be advantageous, demonstrating that the effect of `original antigenic sin' is not absolute. Pre-clinical and clinical studies are reviewed, including our own work with a three-vector vaccination regimen using recombinant DNA, virus (Sendai virus or vaccinia virus) and protein. Promising preliminary results suggest that the heterologous prime-boost strategy may possibly provide a foundation for the future prevention of HIV-1 infections in humans.
Collapse
Affiliation(s)
- Scott A. Brown
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (S.A.B.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Sherri L. Surman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Robert Sealy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Karen S. Slobod
- Early Development, Novartis Vaccines and Diagnostics, 350 Mass Ave. Cambridge, MA 02139, USA; E-Mail: (K.S.S.)
| | - Kristen Branum
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Timothy D. Lockey
- Department of Therapeutics, Production and Quality, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (T.D.L.)
| | - Nanna Howlett
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Pamela Freiden
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
| | - Patricia Flynn
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
- Department of Pediatrics, University of Tennessee, Memphis, TN 38163, USA
| | - Julia L. Hurwitz
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mail: (S.A.B.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; E-Mails: (S.L.S.); (R.S.); (B.G.J.); (K.B.); (N.H.); (P.F.); (P.F.)
- Department of Pathology, University of Tennessee, Memphis, TN 38163, USA
| |
Collapse
|
19
|
Brandsma JL, Shlyankevich M, Su Y, Zelterman D, Rose JK, Buonocore L. Reversal of papilloma growth in rabbits therapeutically vaccinated against E6 with naked DNA and/or vesicular stomatitis virus vectors. Vaccine 2009; 28:8345-51. [PMID: 19615481 DOI: 10.1016/j.vaccine.2009.04.082] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 03/27/2009] [Accepted: 04/03/2009] [Indexed: 02/02/2023]
Abstract
Persistent infection with high-risk human papillomaviruses (HPVs) is the greatest risk factor for the development of HPV-associated cancers. In this study rabbits bearing persistent and potentially malignant papillomas were used to test the efficacy of vaccination with a recombinant DNA and/or vesicular stomatitis virus (VSV) targeting the cottontail rabbit papillomavirus (CRPV) E6 protein. Immune responses were primed with either vector and boosted twice with the homologous or heterologous E6 vector. Over the course of 18 weeks, E6 vaccination reduced papilloma volumes to one third the volume in the controls, and the rabbits boosted with an heterologous vector tended to mount stronger responses. Small and medium-sized papillomas responded significantly but only slightly better than large papillomas. Finally the initial papilloma burden per rabbit, ranging from <100 mm(3) to >1000 mm(3), was not prognostic of antitumor efficacy. In summary both E6 vaccines elicited significant therapeutic immunity, and their sequential use tended to be advantageous.
Collapse
Affiliation(s)
- Janet L Brandsma
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Barefoot B, Thornburg NJ, Barouch DH, Yu JS, Sample C, Johnston RE, Liao HX, Kepler TB, Haynes BF, Ramsburg E. Comparison of multiple vaccine vectors in a single heterologous prime-boost trial. Vaccine 2008; 26:6108-18. [PMID: 18809447 DOI: 10.1016/j.vaccine.2008.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 08/29/2008] [Accepted: 09/01/2008] [Indexed: 12/22/2022]
Abstract
The prevention of infectious disease via prophylactic immunization is a mainstay of global public health efforts. Vaccine design would be facilitated by a better understanding of the type and durability of immune responses generated by different vaccine vectors. We report here the results of a comparative immunogenicity trial of six different vaccine vectors expressing the same insert antigen, cowpox virus B5 (CPXV-B5). Of those vectors tested, recombinant adenovirus (rAd5) was the most immunogenic, inducing the highest titer anti-B5 antibodies and conferring protection from sublethal vaccinia virus challenge in mice after a single immunization. We tested select heterologous prime-boost combinations and identified recombinant vesicular stomatitis virus (rVSV) and recombinant Venezuelan equine encephalitis virus replicons (VRP) as the most synergistic regimen. Comparative data such as those presented here are critical to efforts to generate protective vaccines for emerging infectious diseases as well as for biothreat agents.
Collapse
Affiliation(s)
- Brice Barefoot
- Duke Human Vaccine Institute, Duke University School of Medicine, DU Medical Center, 102 Research Drive, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Primary CD8+ T-cell response to soluble ovalbumin is improved by chloroquine treatment in vivo. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:1497-504. [PMID: 18753338 DOI: 10.1128/cvi.00166-08] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The efficiency of cross-presentation of exogenous antigens by dendritic cells (DCs) would seem to be related to the level of antigen escape from massive degradation mediated by lysosomal proteases in an acidic environment. Here, we demonstrate that a short course of treatment with chloroquine in mice during primary immunization with soluble antigens improved the cross-priming of naïve CD8(+) T lymphocytes in vivo. More specifically, priming of chloroquine-treated mice with soluble ovalbumin (OVA), OVA associated with alum, or OVA pulsed on DCs was more effective in inducing OVA-specific CD8(+) T lymphocytes than was priming of untreated mice. We conclude that chloroquine treatment improves the cross-presentation capacity of DCs and thus the size of effector and memory CD8(+) T cells during vaccination.
Collapse
|
22
|
Immune response in the absence of neurovirulence in mice infected with m protein mutant vesicular stomatitis virus. J Virol 2008; 82:9273-7. [PMID: 18614644 DOI: 10.1128/jvi.00915-08] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Matrix (M) protein mutants of vesicular stomatitis virus (VSV), such as rM51R-M virus, are less virulent than wild-type (wt) VSV strains due to their inability to suppress innate immunity. Studies presented here show that when inoculated intranasally into mice, rM51R-M virus was cleared from nasal mucosa by day 2 postinfection and was attenuated for spread to the central nervous system, in contrast to wt VSV, thus accounting for its reduced virulence. However, it stimulated an antibody response similar to that in mice infected with the wt virus, indicating that it has the ability to induce adaptive immunity in vivo without causing disease. These results support the use of M protein mutants of VSV as vaccine vectors.
Collapse
|
23
|
Wilson SR, Wilson JH, Buonocore L, Palin A, Rose JK, Reuter JD. Intranasal immunization with recombinant vesicular stomatitis virus expressing murine cytomegalovirus glycoprotein B induces humoral and cellular immunity. Comp Med 2008; 58:129-139. [PMID: 18524170 PMCID: PMC2703170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 11/10/2007] [Accepted: 11/29/2007] [Indexed: 05/26/2023]
Abstract
Cytomegalovirus is a leading cause of morbidity and mortality among neonatal and immunocompromised patients. The use of vaccine prophylaxis continues to be an effective approach to reducing viral infections and their associated diseases. Murine cytomegalovirus (mCMV) has proven to be a valuable animal model in determining the efficacy of newly developed vaccine strategies in vivo. Live recombinant vesicular stomatitis viruses (rVSV) have successfully been used as vaccine vectors for several viruses to induce strong humoral and cellular immunity. We tested the ability of intranasal immunization with an rVSV expressing the major envelope protein of mCMV, glycoprotein B (gB), to protect against challenge with mCMV in a mouse model. rVSV-gB-infected cells showed strong cytoplasmic and cell surface expression of gB, and neutralizing antibodies to gB were present in mice after a single intranasal vaccination of VSV-gB. After challenge with mCMV, recovery of live virus and viral DNA was significantly reduced in immunized mice. In addition, primed splenocytes produced a CD8+ IFNgamma response to gB. The ability to induce an immune response to a gene product through mucosal vaccination with rVSV-gB represents a potentially effective approach to limiting CMV-induced disease.
Collapse
Affiliation(s)
- Steven R Wilson
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Single-dose, therapeutic vaccination of mice with vesicular stomatitis virus expressing human papillomavirus type 16 E7 protein. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:817-24. [PMID: 18337377 DOI: 10.1128/cvi.00343-07] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We are developing recombinant attenuated vesicular stomatitis virus (VSV) as a vaccine vector to generate humoral and cell-mediated immune responses. Here, we explore the use of VSV vaccines for cancer immunotherapy. Immunotherapy targeting high-risk human papillomavirus (HPV) lesions has the potential to benefit HPV-infected individuals and cervical cancer patients by generating cytotoxic T cells that kill tumor cells that express viral antigens. A single dose of VSV expressing the HPV type 16 (HPV16) E7 oncogene was used for therapeutic vaccination of mice bearing TC-1 syngeneic tumors, which express HPV16 E7. HPV16 E7-specific T cells were generated and displayed cytotoxic activity against the tumor cells. By 14 days postvaccination, average tumor volumes were 10-fold less in the vaccinated group than in mice that received the empty-vector VSV, and regression of preexisting tumors occurred in some cases. This antitumor effect was CD8 T-cell dependent. Our results demonstrate antitumor responses to HPV16 E7 and suggest that recombinant-VSV-based vaccination should be explored as a therapeutic strategy for cervical carcinoma and other HPV-associated cancers.
Collapse
|
25
|
Matrix protein mediated shutdown of host cell metabolism limits vesicular stomatitis virus-induced interferon-alpha responses to plasmacytoid dendritic cells. Immunobiology 2007; 212:887-94. [PMID: 18086387 DOI: 10.1016/j.imbio.2007.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 09/28/2007] [Indexed: 11/22/2022]
Abstract
Upon infection with many different viruses, plasmacytoid dendritic cells (pDC) produce large amounts of type I interferon (IFN-alpha/beta). To address why upon vesicular stomatitis virus (VSV) infection pDC, but not conventional myeloid DC (mDC), are induced to produce IFN-alpha, pDC and mDC were differentiated from bone marrow cells (BM-DC). Upon VSV infection BM-pDC produced IFN-alpha, whereas BM-mDC did not. Notably, upon infection with VSV-M2, a VSV variant expressing a M51R mutant matrix (M) protein that showed a reduced sequestration of host cell metabolism, BM-pDC and BM-mDC mounted massive IFN-alpha responses. Both DC subsets showed comparable RNA levels of retinoic acid inducible gene-I (RIG-I) and Toll-like receptor (TLR) 7 and were able to respond upon triggering with double-stranded RNA (dsRNA) or single-stranded RNA (ssRNA) analogs. Moreover, upon VSV-M2 infection IFN-alpha production by both DC subsets was largely dependent on viral replication. Interestingly, upon virus infection BM-pDC, but not BM-mDC, up-regulated mRNA levels of nuclear export factors Nup96/98, probably reflecting cellular mechanisms to circumvent viral escape strategies. Collectively, these results indicated that cell types induced to produce IFN-alpha upon viral infection are not primarily defined by cellular receptor configurations but rather by complex virus/host cell interactions.
Collapse
|
26
|
Attenuation of recombinant vesicular stomatitis virus-human immunodeficiency virus type 1 vaccine vectors by gene translocations and g gene truncation reduces neurovirulence and enhances immunogenicity in mice. J Virol 2007; 82:207-19. [PMID: 17942549 DOI: 10.1128/jvi.01515-07] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recombinant vesicular stomatitis virus (rVSV) has shown great potential as a new viral vector for vaccination. However, the prototypic rVSV vector described previously was found to be insufficiently attenuated for clinical evaluation when assessed for neurovirulence in nonhuman primates. Here, we describe the attenuation, neurovirulence, and immunogenicity of rVSV vectors expressing human immunodeficiency virus type 1 Gag. These rVSV vectors were attenuated by combinations of the following manipulations: N gene translocations (N4), G gene truncations (CT1 or CT9), noncytopathic M gene mutations (Mncp), and positioning of the gag gene into the first position of the viral genome (gag1). The resulting N4CT1-gag1, N4CT9-gag1, and MncpCT1-gag1 vectors demonstrated dramatically reduced neurovirulence in mice following direct intracranial inoculation. Surprisingly, in spite of a very high level of attenuation, the N4CT1-gag1 and N4CT9-gag1 vectors generated robust Gag-specific immune responses following intramuscular immunization that were equivalent to or greater than immune responses generated by the more virulent prototypic vectors. MncpCT1-gag1 also induced Gag-specific immune responses following intramuscular immunization that were equivalent to immune responses generated by the prototypic rVSV vector. Placement of the gag gene in the first position of the VSV genome was associated with increased in vitro expression of Gag protein, in vivo expression of Gag mRNA, and enhanced immunogenicity of the vector. These findings demonstrate that through directed manipulation of the rVSV genome, vectors that have reduced neurovirulence and enhanced immunogenicity can be made.
Collapse
|
27
|
Vogel LN, Roberts A, Paddock CD, Genrich GL, Lamirande EW, Kapadia SU, Rose JK, Zaki SR, Subbarao K. Utility of the aged BALB/c mouse model to demonstrate prevention and control strategies for severe acute respiratory syndrome coronavirus (SARS-CoV). Vaccine 2006; 25:2173-9. [PMID: 17227689 PMCID: PMC1847333 DOI: 10.1016/j.vaccine.2006.11.055] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Revised: 11/13/2006] [Accepted: 11/21/2006] [Indexed: 01/12/2023]
Abstract
The causative agent of Severe Acute Respiratory Syndrome (SARS) was identified as a coronavirus (CoV) following the outbreak of 2002–2003. There are currently no licensed vaccines or treatments for SARS-CoV infections. Potential prevention and control strategies that show promise in vitro must be evaluated in animal models. The aged BALB/c mouse model for SARS supports a high level of viral replication in association with clinical illness and disease that mimics SARS in the elderly. We tested two preventive strategies, vaccination and passive transfer of serum antibody, to determine the extent of protection achieved against SARS-CoV challenge in this model. These approaches were able to achieve or induce antibody titers sufficient to reduce viral load, protect from weight loss and reduce or eliminate histopathologic changes in the lungs of aged mice. This study validates the utility of the aged BALB/c mouse model for evaluation of the efficacy of vaccines and immunoprophylaxis.
Collapse
Affiliation(s)
- Leatrice N Vogel
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bozac A, Berto E, Vasquez F, Grandi P, Caputo A, Manservigi R, Ensoli B, Marconi P. Expression of human immunodeficiency virus type 1 tat from a replication-deficient herpes simplex type 1 vector induces antigen-specific T cell responses. Vaccine 2006; 24:7148-58. [PMID: 16884834 DOI: 10.1016/j.vaccine.2006.06.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 06/23/2006] [Accepted: 06/26/2006] [Indexed: 10/24/2022]
Abstract
Herpes simplex type-1 virus (HSV-1) based vectors have been widely used in different gene therapy approaches and also as experimental vaccines against HSV-1 infection. Recent advances in the HSV-1 technology do support the use of replication defective HSV-1 as vaccine vectors for delivery of foreign antigens. We have examined the ability of a recombinant replication-defective HSV-1 vector expressing the HIV-1 Tat protein to induce long-term Tat-specific immune responses in the Balb/c murine model. The results showed that vector administration by the subcutaneous route elicits anti-Tat specific T-cell mediated immune responses in mice characterized by the presence of the Tat-specific cytotoxic activity and production of high levels of IFN-gamma.
Collapse
Affiliation(s)
- Aleksandra Bozac
- University of Ferrara, Department of Experimental and Diagnostic Medicine, Section of Microbiology, Via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Clarke DK, Cooper D, Egan MA, Hendry RM, Parks CL, Udem SA. Recombinant vesicular stomatitis virus as an HIV-1 vaccine vector. ACTA ACUST UNITED AC 2006; 28:239-53. [PMID: 16977404 PMCID: PMC7079905 DOI: 10.1007/s00281-006-0042-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2006] [Accepted: 06/16/2006] [Indexed: 11/27/2022]
Abstract
Recombinant vesicular stomatitis virus (rVSV) is currently under evaluation as a human immunodeficiency virus (HIV)-1 vaccine vector. The most compelling reasons to develop rVSV as a vaccine vector include a very low seroprevalence in humans, the ability to infect and robustly express foreign antigens in a broad range of cells, and vigorous growth in continuous cell lines used for vaccine manufacture. Numerous preclinical studies with rVSV vectors expressing antigens from a variety of human pathogens have demonstrated the versatility, flexibility, and potential efficacy of the rVSV vaccine platform. When administered to nonhuman primates (NHPs), rVSV vectors expressing HIV-1 Gag and Env elicited robust HIV-1-specific cellular and humoral immune responses, and animals immunized with rVSV vectors expressing simian immunodeficiency virus (SIV) Gag and HIV Env were protected from AIDS after challenge with a pathogenic SIV/HIV recombinant. However, results from an exploratory neurovirulence study in NHPs indicated that these prototypic rVSV vectors might not be adequately attenuated for widespread use in human populations. To address this safety concern, a variety of different attenuation strategies, designed to produce a range of further attenuated rVSV vectors, are currently under investigation. Additional modifications of further attenuated rVSV vectors to upregulate expression of HIV-1 antigens and coexpress molecular adjuvants are also being developed in an effort to balance immunogenicity and attenuation.
Collapse
Affiliation(s)
- David K. Clarke
- Department of Vaccines Discovery Research, Wyeth Research, Wyeth, 401 North Middletown Road, Pearl River, NY 10965 USA
| | - David Cooper
- Department of Vaccines Discovery Research, Wyeth Research, Wyeth, 401 North Middletown Road, Pearl River, NY 10965 USA
| | - Michael A. Egan
- Department of Vaccines Discovery Research, Wyeth Research, Wyeth, 401 North Middletown Road, Pearl River, NY 10965 USA
| | - R. Michael Hendry
- Department of Vaccines Discovery Research, Wyeth Research, Wyeth, 401 North Middletown Road, Pearl River, NY 10965 USA
| | - Christopher L. Parks
- Department of Vaccines Discovery Research, Wyeth Research, Wyeth, 401 North Middletown Road, Pearl River, NY 10965 USA
| | - Stephen A. Udem
- Department of Vaccines Discovery Research, Wyeth Research, Wyeth, 401 North Middletown Road, Pearl River, NY 10965 USA
- Present Address: International AIDS Vaccine Initiative, 110 William Street, 27th Floor, New York, NY 10038-3901 USA
| |
Collapse
|
30
|
Brandsma JL, Shlyankevich M, Buonocore L, Roberts A, Becker SM, Rose JK. Therapeutic efficacy of vesicular stomatitis virus-based E6 vaccination in rabbits. Vaccine 2006; 25:751-62. [PMID: 16962690 DOI: 10.1016/j.vaccine.2006.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Revised: 08/04/2006] [Accepted: 08/09/2006] [Indexed: 10/24/2022]
Abstract
Millions of people worldwide are currently infected with human papillomaviruses (HPVs). A therapeutic HPV vaccine would have widespread applicability because HPV-associated lesions are difficult to treat and may progress to carcinoma. We developed three attenuated VSV recombinants expressing the cottontail rabbit papillomavirus (CRPV) early protein E6 for use as vaccines. In cultured cells, two vectors expressed different levels of the E6 protein, and one expressed a ubiquitin-E6 fusion protein. All three were tested for therapeutic efficacy in the cottontail rabbit papillomavirus (CRPV)-rabbit model. Mock vaccination had no effect on papilloma growth. In contrast, inoculation with any of the VSV-E6 vaccines reduced the rate of papilloma growth to as little as 24% the rate in the controls. In five experiments, these effects were achieved after a single immunization. Furthermore, complete papilloma regression occurred in some rabbits observed for 4 months. A VSV-based papillomavirus E6 vaccine could have significant advantages over other therapeutic HPV vaccine candidates described to date.
Collapse
Affiliation(s)
- Janet L Brandsma
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520-8016, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Vingert B, Adotevi O, Patin D, Jung S, Shrikant P, Freyburger L, Eppolito C, Sapoznikov A, Amessou M, Quintin-Colonna F, Fridman WH, Johannes L, Tartour E. The Shiga toxin B-subunit targets antigen in vivo to dendritic cells and elicits anti-tumor immunity. Eur J Immunol 2006; 36:1124-35. [PMID: 16568496 DOI: 10.1002/eji.200535443] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The non-toxic B-subunit of Shiga toxin (STxB) interacts with the glycolipid Gb3, which is preferentially expressed on dendritic cells (DC) and B cells. After administration of STxB chemically coupled to OVA (STxB-OVA) in mice, we showed that the immunodominant OVA(257-264) peptide restricted by K(b) molecules is specifically presented by CD11c+ CD8alpha- DC, some of them displaying a mature phenotype. Using mice carrying a transgene encoding a diphtheria toxin receptor (DTR) under the control of the murine CD11c promoter, which allows inducible ablation of DC, we showed that DC are required for efficient priming of CTL after STxB-OVA vaccination. Immunization of mice with STxB-OVA induced OVA-specific CD8+ T cells detected ex vivo; these cells were long lasting, since they could be detected even 91 days after the last immunization and were composed of both central and memory T cells. Vaccination of mice with STxB-OVA and STxB coupled to E7, a protein derived from HPV16, inhibited tumor growth in prophylactic and therapeutic experiments. This effect was mainly mediated by CD8+ T cells. STxB therefore appears to be a powerful carrier directly targeting DC in vivo, resulting in a strong and durable CTL response associated with tumor protection.
Collapse
Affiliation(s)
- Benoit Vingert
- INSERM U255, Université René Descartes, Unité d'Immunologie Biologique, Hopital Européen Georges Pompidou AP-HP, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Nayak BP, Sailaja G, Jabbar AM. Augmenting the immunogenicity of DNA vaccines: role of plasmid-encoded Flt-3 ligand, as a molecular adjuvant in genetic vaccination. Virology 2006; 348:277-88. [PMID: 16563456 DOI: 10.1016/j.virol.2006.02.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Revised: 02/10/2006] [Accepted: 02/10/2006] [Indexed: 12/22/2022]
Abstract
In this study, we have taken advantage of the unique property of a potent dendritic cell (DC) growth factor, Flt-3 ligand (FL), which could act as a vaccine adjuvant. Accordingly, a single injection of plasmid DNA coding for soluble FL (FLex) was shown to induce large numbers of DCs in various tissue compartments and was critical for generating high frequencies of antigen-specific (HIV gp120 and LCMV NP) immune responses in mice. Interestingly, this enhanced level of immune response is strictly dependent on the co-delivery (i.m.) of the DNA vaccines and hFLex DNA to mice harboring large numbers of DCs. The high frequencies of antigen-specific CD8(+) T cells were largely associated with the expansion phase of DCs in vivo. However, DC expansion and immune enhancement have not reciprocally maintained a linear correlation, suggesting that other factors, cytokines/chemokines, which have the potential to modulate the microenvironment of DCs, could influence immunological outcome in this vaccination modality.
Collapse
Affiliation(s)
- Bishnu P Nayak
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | | | | |
Collapse
|
33
|
Ramsburg E, Publicover J, Buonocore L, Poholek A, Robek M, Palin A, Rose JK. A vesicular stomatitis virus recombinant expressing granulocyte-macrophage colony-stimulating factor induces enhanced T-cell responses and is highly attenuated for replication in animals. J Virol 2006; 79:15043-53. [PMID: 16306575 PMCID: PMC1316007 DOI: 10.1128/jvi.79.24.15043-15053.2005] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Live attenuated vectors based on recombinant vesicular stomatitis viruses (rVSVs) expressing foreign antigens are highly effective vaccines in animal models. In this study, we report that an rVSV (VSV-GMCSF1) expressing high levels of murine granulocyte-macrophage colony-stimulating factor (GM-CSF) from the first position in the viral genome is highly attenuated in terms of viral dissemination and pathogenesis after intranasal delivery to mice. However, this highly attenuated virus generated antibody and T-cell responses equivalent to those induced by a control virus expressing enhanced green fluorescent protein (EGFP) from the first position (VSV-EGFP1). The better containment and clearance of VSV-GMCSF1 may be due to enhanced recruitment of macrophages to the site of infection but is not explained by a greater induction of interferons. The primary CD8 T-cell and neutralizing antibody responses to VSV-GMCSF1 were equivalent to those generated by VSV-EGFP1, while the CD8 T-cell memory and recall responses to the vector were enhanced in mice infected with VSV-GMCSF1. It is likely that the GM-CSF produced by immunization with this virus results in an enhanced recruitment of antigen-presenting cells, leading to better acute and long-term T-cell responses. This recruitment appears to cancel out any negative effect of viral attenuation on immunogenicity.
Collapse
Affiliation(s)
- Elizabeth Ramsburg
- Department of Pathology, Yale University School of Medicine, 310 Cedar St. (LH302), New Haven, CT 06510, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Ahmed M, Brzoza KL, Hiltbold EM. Matrix protein mutant of vesicular stomatitis virus stimulates maturation of myeloid dendritic cells. J Virol 2006; 80:2194-205. [PMID: 16474127 PMCID: PMC1395366 DOI: 10.1128/jvi.80.5.2194-2205.2006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 12/02/2005] [Indexed: 12/29/2022] Open
Abstract
Matrix (M) protein mutants of vesicular stomatitis virus have recently been used as oncolytic viruses for tumor therapies and are being developed as vaccine vectors for heterologous antigens. Because dendritic cell (DC) maturation is an important correlate of tumor immunosurveillance and vaccine efficacy, we sought to determine the ability of a recombinant M protein mutant virus (rM51R-M virus) to mature DC in vitro. We have previously shown that rM51R-M virus is defective at inhibiting host gene expression in several cell lines compared to its recombinant wild-type counterpart, rwt virus. Therefore, rM51R-M virus allows the expression of genes involved in antiviral responses, such as the type I interferon (IFN) gene. Our results demonstrate that, in contrast to the rwt virus, rM51R-M virus induced the maturation of myeloid DC (mDC) populations, as indicated by an increase in the surface expression of CD40, CD80, and CD86 as well as the secretion of interleukin-12 (IL-12), IL-6, and type I IFN. In addition, mDC infected with rM51R-M virus effectively activated naïve T cells in vitro, whereas rwt virus-infected mDC were defective in antigen presentation. The inability of rwt virus to induce mDC maturation was correlated with the inhibition of host gene expression in rwt virus-infected cells. Our studies also indicated that the production of costimulatory molecules on mDC by rM51R-M virus was dependent on the type I IFN receptor, while maturation induced by this virus was largely independent of MyD88. These data indicate that rM51R-M virus effectively stimulates the maturation of mDC and has the potential to promote effective T-cell responses to vector-expressed antigens, activate DC at tumor sites during therapy, and aid in tumor immunosurveillance and destruction.
Collapse
Affiliation(s)
- Maryam Ahmed
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | |
Collapse
|
35
|
Cristillo AD, Wang S, Caskey MS, Unangst T, Hocker L, He L, Hudacik L, Whitney S, Keen T, Chou THW, Shen S, Joshi S, Kalyanaraman VS, Nair B, Markham P, Lu S, Pal R. Preclinical evaluation of cellular immune responses elicited by a polyvalent DNA prime/protein boost HIV-1 vaccine. Virology 2005; 346:151-68. [PMID: 16325880 DOI: 10.1016/j.virol.2005.10.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 09/19/2005] [Accepted: 10/21/2005] [Indexed: 10/25/2022]
Abstract
While DNA vaccines have been shown to prime cellular immune responses, levels are often low in nonhuman primates or humans. Hence, efforts have been directed toward boosting responses by combining DNA with different vaccination modalities. To this end, a polyvalent DNA prime/protein boost vaccine, consisting of codon optimized HIV-1 env (A, B, C, E) and gag (C) and homologous gp120 proteins in QS-21, was evaluated in rhesus macaques and BALB/c mice. Humoral and cellular responses, detected following DNA immunization, were increased following protein boost in macaques and mice. In dissecting cellular immune responses in mice, protein-enhanced responses were found to be mediated by CD4+ and CD8+ T cells with a Th1 cytokine bias. Our study reveals that, in addition to augmenting humoral responses, protein boosting of DNA-primed animals augments cellular immune responses mediated by CD8+ CTL, CD4+ T-helper cells and Th1 cytokines; thus, offering much promise in controlling HIV-1 in vaccinees.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories, Department of Cell Biology, 5510 Nicholson Lane, Kensington, MD 20895, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Jiang P, Liu Y, Yin X, Yuan F, Nie Y, Luo M, Aihua Z, Liyin D, Ding M, Deng H. Elicitation of neutralizing antibodies by intranasal administration of recombinant vesicular stomatitis virus expressing human immunodeficiency virus type 1 gp120. Biochem Biophys Res Commun 2005; 339:526-32. [PMID: 16313884 PMCID: PMC7092882 DOI: 10.1016/j.bbrc.2005.11.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2005] [Accepted: 11/14/2005] [Indexed: 12/01/2022]
Abstract
Recombinant viral vectors are useful tools for AIDS vaccine development. However, expression of HIV-1 envelope genes using viral vectors has not been successful in the induction of potent neutralizing antibodies in vivo. We took advantage of the strong immunogenicity of vesicular stomatitis virus (VSV)-based vector and expressed HIV-1 HXB2 gp120 gene in the recombinant VSV. Our results showed that HIV-1 gp120 protein expressed by the recombinant VSV retained the native conformation of the protein to some degree and was recognized by two well-characterized broad anti-HIV-1 neutralizing monoclonal antibodies b12, 2G12. We further showed that only one time intranasal immunization with the recombinant VSV led to production of anti-HIV-1 anti-sera in mice. In addition, we found that the anti-sera had the ability to neutralize not only HXB2 envelope-pseudotyped HIV-1 viruses but also HIV-1 pseudotyped viruses with JRFL envelopes. These results suggest that HIV-1 gp120 expressed by the recombinant VSV, in combination with the route of intranasal administration, is an effective strategy to evaluate the immunogenicity of HIV-1 envelope protein and its variants in mice.
Collapse
|
37
|
Publicover J, Ramsburg E, Rose JK. A single-cycle vaccine vector based on vesicular stomatitis virus can induce immune responses comparable to those generated by a replication-competent vector. J Virol 2005; 79:13231-8. [PMID: 16227246 PMCID: PMC1262593 DOI: 10.1128/jvi.79.21.13231-13238.2005] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Live attenuated vaccine vectors based on recombinant vesicular stomatitis virus (VSV) are effective in several viral disease models. In this study, we asked if a VSV vector capable of only a single cycle of replication might be an effective alternative to replication-competent VSV vectors. We compared the cellular immune responses to human immunodeficiency virus (HIV) envelope protein (Env) expressed by replication-competent and single-cycle VSV vectors and also examined the antibody response to Env. The single-cycle vector was grown by complementation with VSV G protein and then tested initially for immunogenicity when given by four different routes. When given by the intramuscular route in mice, we found that the single-cycle vector was equivalent to the replication-competent VSV vector in generating high-level primary and memory CD8 T-cell responses as well as antibody responses to Env. Cellular responses were analyzed using major histocompatibility complex class I tetramers and direct measurement of cytotoxic T-lymphocyte activity in vivo. We also found that the recall responses after boosting were equivalent in animals vaccinated with replication-competent or single-cycle vectors. Additionally, we observed recall and heightened memory responses after boosting animals with a single-cycle vector complemented with G protein from a different vesiculovirus. Because expression of HIV Env by G-deleted VSV might allow replication in human cells expressing CD4, we generated a single-cycle VSV recombinant expressing a secreted form of the HIV Env protein. This virus was just as effective as the recombinant expressing the membrane-anchored Env protein at producing CD8 T cells and antibody responses.
Collapse
Affiliation(s)
- Jean Publicover
- Section of Microbial Pathogenesis, Yale University School of Medicine, 310 Cedar St. (LH 315), New Haven, CT 06510, USA
| | | | | |
Collapse
|
38
|
McGettigan JP, Koser ML, McKenna PM, Smith ME, Marvin JM, Eisenlohr LC, Dietzschold B, Schnell MJ. Enhanced humoral HIV-1-specific immune responses generated from recombinant rhabdoviral-based vaccine vectors co-expressing HIV-1 proteins and IL-2. Virology 2005; 344:363-77. [PMID: 16226782 DOI: 10.1016/j.virol.2005.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 08/10/2005] [Accepted: 09/06/2005] [Indexed: 12/23/2022]
Abstract
Recombinant rabies virus (RV) vaccine strain-based vectors expressing HIV-1 antigens have been shown to induce strong and long-lasting cellular but modest humoral responses against the expressed antigens in mice. However, an effective vaccine against HIV-1 may require stronger responses, and the development of such an immune response may depend on the presence of certain cytokines at the time of the inoculation. Here, we describe several new RV-based vaccine vehicles expressing HIV-1 Gag or envelope (Env) and murine IL-2 or IL-4. Cells infected with recombinant RVs expressed high levels of functional IL-2 or IL-4 in culture supernatants in addition to HIV-1 proteins. The recombinant RV expressing IL-4 was highly attenuated in a cytokine-independent manner, indicating that the insertion of two foreign genes into the RV genome is mainly responsible for the attenuation observed. The expression of IL-4 resulted in a decrease in the cellular immune response against HIV-1 Gag and Env when compared with the parental virus not expressing IL-4 and only 2 of 20 mice seroconverted to HIV-1 Env after two inoculations. The IL-2-expressing RV was completely apathogenic after direct intracranial inoculation of mice. In addition, mice immunized with IL-2 maintained strong anti-HIV-1 Gag and Env cellular responses and consistently induced seroconversion against HIV-1 Env after two inoculations. This suggests the potential use of IL-2 in RV-based HIV-1 vaccine strategies, which may require the induction of both arms of the immune response.
Collapse
Affiliation(s)
- James P McGettigan
- Department of Microbiology and Immunology, Jefferson Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 330, Philadelphia, PA 19107-6799, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Finke S, Conzelmann KK. Recombinant rhabdoviruses: vectors for vaccine development and gene therapy. Curr Top Microbiol Immunol 2005; 292:165-200. [PMID: 15981472 DOI: 10.1007/3-540-27485-5_8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The establishment of methods to recover rhabdoviruses from cDNA, so-called reverse genetics systems, has made it possible to genetically engineer rhabdoviruses and to study all aspects of the virus life cycle by introducing defined mutations into the viral genomes. It has also opened the way to make use of the viruses in biomedical applications such as vaccination, gene therapy, or oncolytic virotherapy. The typical gene expression mode of rhabdoviruses, a high genetic stability, and the propensity to tolerate changes in the virus envelope have made rhabdoviruses attractive, targetable gene expression vectors. This chapter provides an overview on the possibilities to manipulate biological properties of the rhabdoviruses that may be important for further development of vaccine vectors and examples of recombinant rhabdoviruses expressing foreign genes and antigens.
Collapse
Affiliation(s)
- S Finke
- Max von Pettenkofer-Institut & Genzentrum, Ludwig-Maximilians-Universität, Feodor-Lynen-Str. 25, 81377 Munich, Germany.
| | | |
Collapse
|
40
|
Sundbäck M, Douagi I, Dayaraj C, Forsell MNE, Nordström EKL, McInerney GM, Spångberg K, Tjäder L, Bonin E, Sundström M, Liljeström P, Karlsson Hedestam GB. Efficient expansion of HIV-1-specific T cell responses by homologous immunization with recombinant Semliki Forest virus particles. Virology 2005; 341:190-202. [PMID: 16098555 DOI: 10.1016/j.virol.2005.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Revised: 06/01/2005] [Accepted: 07/14/2005] [Indexed: 11/16/2022]
Abstract
Vaccines based on recombinant viruses represent a promising strategy for the development of a prophylactic vaccine against HIV-1. However, despite a proven capacity to stimulate potent HIV-1-specific immune responses, viral systems have limited utility in homologous prime-boost regimens due to the generation of anti-vector immune responses. It is therefore important to develop a diverse set of vaccine candidates that can be combined in different heterologous prime-boost regimens and/or to identify a vaccine candidate that is less sensitive to anti-vector mediated immunity. In this report, we describe the design and pre-clinical immunogenicity of a Semliki Forest virus-based vaccine, VREP-C, encoding Indian origin HIV-1 clade C antigens. We show that a single immunization with VREP-C stimulates HIV-1-specific IFNgamma ELISPOT responses, which were efficiently boosted by a second and a third homologous VREP-C immunization resulting in highly potent cytotoxic T cell responses. These results suggest that VREP-C may be a valuable component of a future prophylactic vaccine against HIV-1.
Collapse
Affiliation(s)
- Maria Sundbäck
- Microbiology and Tumor Biology Center, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Webster DE, Thomas MC, Pickering R, Whyte A, Dry IB, Gorry PR, Wesselingh SL. Is there a role for plant‐made vaccines in the prevention of HIV/AIDS? Immunol Cell Biol 2005; 83:239-47. [PMID: 15877601 DOI: 10.1111/j.1440-1711.2005.01341.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although educational programs have had some impact, immunization against HIV will be necessary to control the AIDS pandemic. To be effective, vaccination will need to be accessible and affordable, directed against multiple antigens, and delivered in multiple doses. Plant-based vaccines that are heat-stable and easy to produce and administer are suited to this type of strategy. Pilot studies by a number of groups have demonstrated that plant viral expression systems can produce HIV antigens in quantities that are appropriate for use in vaccines. In addition, these plant-made HIV antigens have been shown to be immunogenic. However, given the need for potent cross-clade humoral and T-cell immunity for protection against HIV, and the uncertainty surrounding the efficacy of protein subunit vaccines, it is most likely that plant-made HIV vaccines will find their niche as booster immunizations in prime-boost vaccination schedules.
Collapse
MESH Headings
- Acquired Immunodeficiency Syndrome/immunology
- Acquired Immunodeficiency Syndrome/prevention & control
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- HIV Infections/immunology
- HIV Infections/prevention & control
- Humans
- Immune Tolerance/immunology
- Immunity, Cellular/immunology
- Mice
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/metabolism
- Vaccines, Edible/administration & dosage
- Vaccines, Edible/biosynthesis
- Vaccines, Edible/genetics
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/genetics
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- Diane E Webster
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
42
|
Tan GS, McKenna PM, Koser ML, McLinden R, Kim JH, McGettigan JP, Schnell MJ. Strong cellular and humoral anti-HIV Env immune responses induced by a heterologous rhabdoviral prime-boost approach. Virology 2005; 331:82-93. [PMID: 15582655 DOI: 10.1016/j.virol.2004.10.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Revised: 09/15/2004] [Accepted: 10/08/2004] [Indexed: 11/20/2022]
Abstract
Recombinant rhabdovirus vectors expressing human immunodeficiency virus (HIV) and/or simian immunodeficiency virus (SIV) proteins have been shown to induce strong immune responses in mice and rhesus macaques. However, the finding that such responses protect rhesus macaques from AIDS-like disease but not from infection indicates that further improvements for these vectors are needed. Here, we designed a prime-boost schedule consisting of a rabies virus (RV) vaccine strain and a recombinant vesicular stomatitis virus (VSV) both expressing HIV Envelope (Env). Mice were primed and boosted with the two vaccine vehicles by different routes and in different combinations. Mucosal and systemic humoral responses were assessed using enzyme linked immunosorbent assay (ELISA) while the cellular immune response was determined by an IFN-gamma ELISPOT assay. We found that an immunization combination of RV and VSV elicited the highest titers of anti-Env antibodies and the greatest amount of Env-specific IFN-gamma secreting cells pre- and post-challenge with a recombinant vaccinia virus expressing HIV(89.6) Env. Furthermore, intramuscular immunization did not induce antigen-specific mucosal antibodies while intranasal inoculation stimulated vector-specific IgA antibodies in vaginal washings and serum. Our results show that it is feasible to elicit robust cellular and humoral anti-HIV responses using two different live attenuated Rhabdovirus vectors to sequentially prime and boost.
Collapse
Affiliation(s)
- Gene S Tan
- Department of Microbiology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Mucosal Immunity and Vaccines Against Simian Immunodeficiency Virus and Human Immunodeficiency Virus. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50056-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
44
|
Rosenthal KL. Recombinant Live Viral Vectors as Vaccines for Mucosal Immunity. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50061-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
45
|
Abstract
A major challenge for immunologists has been the development of vaccines designed to emphasize cellular immune responses. One particularly promising approach is the prime-boost strategy, which has been shown to generate high levels of T-cell memory in animal models. Recently, several papers have highlighted the power of prime-boost strategies in eliciting protective cellular immunity to a variety of pathogens and have demonstrated efficacy in humans. Coupled with recent advances in our understanding of the mechanisms underlying the generation, maintenance and recall of T-cell memory, the field is poised to make tremendous progress. This Review discusses the impact of these recent developments on the future of prime-boost vaccine strategies.
Collapse
Affiliation(s)
- David L Woodland
- Trudeau Institute, 154 Algonquin Ave., Saranac Lake, NY 12983, USA.
| |
Collapse
|
46
|
Ramsburg E, Rose NF, Marx PA, Mefford M, Nixon DF, Moretto WJ, Montefiori D, Earl P, Moss B, Rose JK. Highly effective control of an AIDS virus challenge in macaques by using vesicular stomatitis virus and modified vaccinia virus Ankara vaccine vectors in a single-boost protocol. J Virol 2004; 78:3930-40. [PMID: 15047809 PMCID: PMC374300 DOI: 10.1128/jvi.78.8.3930-3940.2004] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have shown that vaccination and boosting of rhesus macaques with attenuated vesicular stomatitis virus (VSV) vectors encoding Env and Gag proteins of simian immunodeficiency virus-human immunodeficiency virus (SHIV) hybrid viruses protect rhesus macaques from AIDS after challenge with the highly pathogenic SHIV 89.6P (23). In the present study, we compared the effectiveness of a single prime-boost protocol consisting of VSV vectors expressing SHIV Env, Gag, and Pol proteins to that of a protocol consisting of a VSV vector prime followed with a single boost with modified vaccinia virus Ankara (MVA) expressing the same SHIV proteins. After challenge with SHIV 89.6P, MVA-boosted animals controlled peak challenge viral loads to less than 2 x 10(6) copies/ml (a level significantly lower than that seen with VSV-boosted animals and lower than those reported for other vaccine studies employing the same challenge). MVA-boosted animals have shown excellent preservation of CD4(+) T cells, while two of four VSV-boosted animals have shown significant loss of CD4(+) T cells. The improved protection in MVA-boosted animals correlates with trends toward stronger prechallenge CD8(+)-T-cell responses to SHIV antigens and stronger postchallenge SHIV-neutralizing antibody production.
Collapse
|
47
|
Shoji Y, Inoue S, Nakamichi K, Kurane I, Sakai T, Morimoto K. Generation and characterization of P gene-deficient rabies virus. Virology 2004; 318:295-305. [PMID: 14972555 DOI: 10.1016/j.virol.2003.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2003] [Revised: 10/01/2003] [Accepted: 10/03/2003] [Indexed: 10/26/2022]
Abstract
Rabies virus (RV) deficient in the P gene was generated by reverse genetics from cDNA of HEP-Flury strain lacking the entire P gene. The defective virus was propagated and amplified by rescue of virus, using a cell line that complemented the functions of the deficient gene. The P gene-deficient (def-P) virus replicated its genome and produced progeny viruses in the cell lines that constitutively expressed the P protein, although it grew at a slightly retarded rate compared to the parental strain. In contrast, no progeny virus was produced in the infected host when the def-P virus-infected cells that did not express the P protein. However, we found that the def-P virus had the ability to perform primary transcription (by the virion-associated polymerase) in the infected host without de novo P protein synthesis. The def-P virus was apathogenic in adult and suckling mice, even when inoculated intracranially. Inoculation of def-P virus in mice induced high levels of virus-neutralizing antibody (VNA) and conferred protective immunity against a lethal rabies infection. These results demonstrate the potential utility of gene-deficient virus as a novel live attenuated rabies vaccine.
Collapse
Affiliation(s)
- Youko Shoji
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Giri M, Ugen KE, Weiner DB. DNA vaccines against human immunodeficiency virus type 1 in the past decade. Clin Microbiol Rev 2004; 17:370-89. [PMID: 15084506 PMCID: PMC387404 DOI: 10.1128/cmr.17.2.370-389.2004] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This article reviews advances in the field of human immunodeficiency virus type 1 (HIV-1) and AIDS vaccine development over the last decade, with an emphasis on the DNA vaccination approach. Despite the discovery of HIV-1 and AIDS in humans nearly 20 years ago, there is no vaccine yet that can prevent HIV-1 infection. The focus has shifted toward developing vaccines that can control virus replication and disease progression by eliciting broadly cross-reactive T-cell responses. Among several approaches evaluated, the DNA-based modality has shown considerable promise in terms of its ability to elicit cellular immune responses in primate studies. Of great importance are efforts aimed at improvement of the potency of this modality in the clinic. The review discusses principles of DNA vaccine design and the various mechanisms of plasmid-encoded antigen presentation. The review also outlines current DNA-based vaccine strategies and vectors that have successfully been shown to control virus replication and slow disease progression in animal models. Finally, it lists recent strategies that have been developed as well as novel approaches under consideration to enhance the immunogenicity of plasmid-encoded HIV-1 antigen in various animal models.
Collapse
Affiliation(s)
- Malavika Giri
- Immunology Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
49
|
Jaffray A, Shephard E, van Harmelen J, Williamson C, Williamson AL, Rybicki EP. Human immunodeficiency virus type 1 subtype C Gag virus-like particle boost substantially improves the immune response to a subtype C gag DNA vaccine in mice. J Gen Virol 2004; 85:409-413. [PMID: 14769898 DOI: 10.1099/vir.0.19396-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) subtype C is the predominant HIV in southern Africa, and is the target of a number of recent vaccine candidates. It has been proposed that a heterologous prime/boost vaccination strategy may result in stronger, broader and more prolonged immune responses. Since HIV-1 Gag Pr55 polyprotein can assemble into virus-like particles (VLPs) which have been shown to induce a strong cellular immune response in animals, we showed that a typical southern African subtype C Pr55 protein expressed in insect cells via recombinant baculovirus could form VLPs. We then used the baculovirus-produced VLPs as a boost to a subtype C HIV-1 gag DNA prime vaccination in mice. This study shows that a low dose of HIV-1 subtype C Gag VLPs can significantly boost the immune response to a single subtype C gag DNA inoculation in mice. These results suggest a possible vaccination regimen for humans.
Collapse
Affiliation(s)
- Ann Jaffray
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Rondebosch 7701, South Africa
| | - Enid Shephard
- MRC Liver Research Centre, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Joanne van Harmelen
- Division of Virology, University of Cape Town, Observatory 7925, South Africa
| | - Carolyn Williamson
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Division of Virology, University of Cape Town, Observatory 7925, South Africa
| | - Anna-Lise Williamson
- National Health Laboratory Service, University of Cape Town, Observatory 7925, South Africa
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Division of Virology, University of Cape Town, Observatory 7925, South Africa
| | - Edward P Rybicki
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
50
|
Garulli B, Kawaoka Y, Castrucci MR. Mucosal and systemic immune responses to a human immunodeficiency virus type 1 epitope induced upon vaginal infection with a recombinant influenza A virus. J Virol 2004; 78:1020-5. [PMID: 14694134 PMCID: PMC368805 DOI: 10.1128/jvi.78.2.1020-1025.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The humoral and cellular immune responses in the genital mucosa likely play an important role in the prevention of sexually transmitted infections, including infection with human immunodeficiency virus type 1 (HIV-1). Here we show that vaginal infection of progesterone-treated BALB/c mice with a recombinant influenza virus bearing the immunodominant P18IIIB cytotoxic T-lymphocyte (CTL) epitope of the gp160 envelope protein from an HIV-1 IIIB isolate (P18IIIB; RIQRGPGRAFVTIGK) can induce a specific immune response in regional mucosal lymph nodes, as well as in a systemic site (the spleen). A single inoculation of mice with the recombinant influenza virus induced long-lasting (at least 5 months) antigen-specific CTL memory detectable as a rapid recall of effector CTLs upon vaginal infection with recombinant vaccinia virus expressing HIV-1 IIIB envelope gene products. Long-term antigen-specific CTL memory was also induced and maintained in distant mucosal tissues when mice were intranasally immunized with the recombinant influenza virus. These results indicate that mucosal immunization and, in particular, local vaginal immunization with recombinant influenza virus can provide strong, durable immune responses in the female genital tract of mice.
Collapse
Affiliation(s)
- Bruno Garulli
- Laboratory of Virology, Istituto Superiore di Sanità, 00161 Rome, Italy
| | | | | |
Collapse
|