57 results on '"Jim Tartaglia"'
Search Results
2. Pentavalent HIV-1 vaccine protects against simian-human immunodeficiency virus challenge
- Author
-
Todd Bradley, Justin Pollara, Sampa Santra, Nathan Vandergrift, Srivamshi Pittala, Chris Bailey-Kellogg, Xiaoying Shen, Robert Parks, Derrick Goodman, Amanda Eaton, Harikrishnan Balachandran, Linh V. Mach, Kevin O. Saunders, Joshua A. Weiner, Richard Scearce, Laura L. Sutherland, Sanjay Phogat, Jim Tartaglia, Steven G. Reed, Shiu-Lok Hu, James F. Theis, Abraham Pinter, David C. Montefiori, Thomas B. Kepler, Kristina K. Peachman, Mangala Rao, Nelson L. Michael, Todd J. Suscovich, Galit Alter, Margaret E. Ackerman, M. Anthony Moody, Hua-Xin Liao, Georgia Tomaras, Guido Ferrari, Bette T. Korber, and Barton F. Haynes
- Subjects
Science - Abstract
A previous human HIV-1 vaccine clinical trial, boosting with HIV envelope protein from two strains, demonstrated moderate vaccine efficacy. Here, Bradleyet al. show that a pentavalent HIV envelope protein boost improves protection from viral challenge in non-human primates and they identify immune correlates of protection.
- Published
- 2017
- Full Text
- View/download PDF
3. Improved innate and adaptive immunostimulation by genetically modified HIV-1 protein expressing NYVAC vectors.
- Author
-
Esther D Quakkelaar, Anke Redeker, Elias K Haddad, Alexandre Harari, Stella Mayo McCaughey, Thomas Duhen, Abdelali Filali-Mouhim, Jean-Philippe Goulet, Nikki M Loof, Ferry Ossendorp, Beatriz Perdiguero, Paul Heinen, Carmen E Gomez, Karen V Kibler, David M Koelle, Rafick P Sékaly, Federica Sallusto, Antonio Lanzavecchia, Giuseppe Pantaleo, Mariano Esteban, Jim Tartaglia, Bertram L Jacobs, and Cornelis J M Melief
- Subjects
Medicine ,Science - Abstract
Attenuated poxviruses are safe and capable of expressing foreign antigens. Poxviruses are applied in veterinary vaccination and explored as candidate vaccines for humans. However, poxviruses express multiple genes encoding proteins that interfere with components of the innate and adaptive immune response. This manuscript describes two strategies aimed to improve the immunogenicity of the highly attenuated, host-range restricted poxvirus NYVAC: deletion of the viral gene encoding type-I interferon-binding protein and development of attenuated replication-competent NYVAC. We evaluated these newly generated NYVAC mutants, encoding HIV-1 env, gag, pol and nef, for their ability to stimulate HIV-specific CD8 T-cell responses in vitro from blood mononuclear cells of HIV-infected subjects. The new vectors were evaluated and compared to the parental NYVAC vector in dendritic cells (DCs), RNA expression arrays, HIV gag expression and cross-presentation assays in vitro. Deletion of type-I interferon-binding protein enhanced expression of interferon and interferon-induced genes in DCs, and increased maturation of infected DCs. Restoration of replication competence induced activation of pathways involving antigen processing and presentation. Also, replication-competent NYVAC showed increased Gag expression in infected cells, permitting enhanced cross-presentation to HIV-specific CD8 T cells and proliferation of HIV-specific memory CD8 T-cells in vitro. The recombinant NYVAC combining both modifications induced interferon-induced genes and genes involved in antigen processing and presentation, as well as increased Gag expression. This combined replication-competent NYVAC is a promising candidate for the next generation of HIV vaccines.
- Published
- 2011
- Full Text
- View/download PDF
4. The transcription factor CREB1 is a mechanistic driver of immunogenicity and reduced HIV-1 acquisition following ALVAC vaccination
- Author
-
Robert Parks, Kelly E. Seaton, Barton F. Haynes, Jim Tartaglia, Jeffrey Tomalka, Muhammad Bilal Latif, Georgia D. Tomaras, Slim Fourati, Ana María González, Adam Pelletier, Nelson L. Michael, Richard A. Koup, Rafick Pierre Sekaly, Peter Wilkinson, Genoveffa Franchini, Merlin L. Robb, Norman L. Letvin, Kathryn Furr, Sampa Santra, Michelle A. Lifton, Nicole L. Yates, Ashish Sharma, and Kevin R. Carlson
- Subjects
Innate immune system ,Immunogenicity ,Immunology ,Antigen presentation ,Simian immunodeficiency virus ,Biology ,medicine.disease_cause ,Acquired immune system ,Vaccine efficacy ,Vaccination ,Immune system ,medicine ,Immunology and Allergy - Abstract
Development of effective human immunodeficiency virus 1 (HIV-1) vaccines requires synergy between innate and adaptive immune cells. Here we show that induction of the transcription factor CREB1 and its target genes by the recombinant canarypox vector ALVAC + Alum augments immunogenicity in non-human primates (NHPs) and predicts reduced HIV-1 acquisition in the RV144 trial. These target genes include those encoding cytokines/chemokines associated with heightened protection from simian immunodeficiency virus challenge in NHPs. Expression of CREB1 target genes probably results from direct cGAMP (STING agonist)-modulated p-CREB1 activity that drives the recruitment of CD4+ T cells and B cells to the site of antigen presentation. Importantly, unlike NHPs immunized with ALVAC + Alum, those immunized with ALVAC + MF59, the regimen in the HVTN702 trial that showed no protection from HIV infection, exhibited significantly reduced CREB1 target gene expression. Our integrated systems biology approach has validated CREB1 as a critical driver of vaccine efficacy and highlights that adjuvants that trigger CREB1 signaling may be critical for efficacious HIV-1 vaccines. Understanding the mechanistic basis of vaccine efficacy is crucial to the development of next-generation vaccines. Sekaly and colleagues find that activation of the transcription factor CREB1 by the RV144 HIV-1 vaccine underpins the induction of robust adaptive immunity.
- Published
- 2021
- Full Text
- View/download PDF
5. Expansion of the HSV-2-specific T cell repertoire in skin after immunotherapeutic HSV-2 vaccine
- Author
-
Emily S. Ford, Alvason Li, Christine Johnston, Lichun Dong, Kurt Diem, Lichen Jing, Kerry J. Laing, Alexis Klock, Krithi Basu, Mariliis Ott, Jim Tartaglia, Sanjay Gurunathan, Jack L. Reid, Matyas Ecsedi, Aude G. Chapuis, Meei-Li Huang, Amalia S. Magaret, Jia Zhu, David M. Koelle, and Lawrence Corey
- Abstract
The skin at the site of HSV-2 reactivation is enriched for HSV-2 specific T cells. To evaluate whether an immunotherapeutic vaccine could elicit skin-based memory T cells we studied skin biopsies and HSV-2-reactive CD4+ T cells from peripheral blood mononuclear cells (PBMCs) by T-cell receptor (TCR) sequencing before and after vaccination with a replication-incompetent whole virus HSV-2 vaccine candidate (HSV529). The representation of HSV-2-reactive CD4+ T cell sequences from PBMCs increased from a median of 0.03% (range 0-0.09%) to 0.6% (range 0-1.3%) of the total skin TCR repertoire after the first vaccine dose. We found sustained expansion after vaccination in unique, skin-based T-cell clonotypes that were not detected in HSV-2-reactive CD4+ T cells isolated from PBMCs. While detection of skin clonotypes in the blood was related to abundance in the skin it was not related to expansion after vaccination. In one participant a switch in immunodominance was observed after vaccination with the emergence of a newly dominant TCRa/b pair in skin that was not detected in blood. We confirmed that the newly dominant clonotype was derived from an HSV-specific CD4+ T cell by creation of a synthetic TCR in a Jurkat-based cell line with a NR4A1-mNeonGreen reporter system. Our data indicate that the skin in areas of HSV-2 reactivation possesses an oligoclonal TCR repertoire that is distinct from the circulation with prominent clonotypes infrequently detected in the circulation by standard methods. Defining the influence of therapeutic vaccination on the HSV-2-specific TCR repertoire requires tissue-based evaluation.
- Published
- 2022
- Full Text
- View/download PDF
6. The transcription factor CREB1 is a mechanistic driver of immunogenicity and reduced HIV-1 acquisition following ALVAC vaccination
- Author
-
Jeffrey Alan, Tomalka, Adam Nicolas, Pelletier, Slim, Fourati, Muhammad Bilal, Latif, Ashish, Sharma, Kathryn, Furr, Kevin, Carlson, Michelle, Lifton, Ana, Gonzalez, Peter, Wilkinson, Genoveffa, Franchini, Robert, Parks, Norman, Letvin, Nicole, Yates, Kelly, Seaton, Georgia, Tomaras, Jim, Tartaglia, Merlin L, Robb, Nelson L, Michael, Richard, Koup, Barton, Haynes, Sampa, Santra, and Rafick Pierre, Sekaly
- Subjects
AIDS Vaccines ,CD4-Positive T-Lymphocytes ,Primates ,B-Lymphocytes ,Genetic Vectors ,Vaccination ,Gene Expression ,HIV Infections ,Viral Vaccines ,HIV Antibodies ,Immunogenicity, Vaccine ,Adjuvants, Immunologic ,HIV-1 ,Animals ,Humans ,Immunization ,Cyclic AMP Response Element-Binding Protein - Abstract
Development of effective human immunodeficiency virus 1 (HIV-1) vaccines requires synergy between innate and adaptive immune cells. Here we show that induction of the transcription factor CREB1 and its target genes by the recombinant canarypox vector ALVAC + Alum augments immunogenicity in non-human primates (NHPs) and predicts reduced HIV-1 acquisition in the RV144 trial. These target genes include those encoding cytokines/chemokines associated with heightened protection from simian immunodeficiency virus challenge in NHPs. Expression of CREB1 target genes probably results from direct cGAMP (STING agonist)-modulated p-CREB1 activity that drives the recruitment of CD4
- Published
- 2020
7. Pentavalent HIV-1 vaccine protects against simian-human immunodeficiency virus challenge
- Author
-
Linh Mach, Bette T. Korber, Robert Parks, Hua-Xin Liao, Steven G. Reed, Barton F. Haynes, Amanda Eaton, Georgia D. Tomaras, Richard M. Scearce, Nelson L. Michael, Xiaoying Shen, James F. Theis, Galit Alter, M. Anthony Moody, Kristina K. Peachman, Srivamshi Pittala, Shiu Lok Hu, Guido Ferrari, David C. Montefiori, Harikrishnan Balachandran, Mangala Rao, Sanjay Phogat, Derrick Goodman, Thomas B. Kepler, Laura L. Sutherland, Margaret E. Ackerman, Todd Bradley, Joshua A. Weiner, Kevin O. Saunders, Justin Pollara, Sampa Santra, Jim Tartaglia, Nathan Vandergrift, Abraham Pinter, Todd J. Suscovich, and Chris Bailey-Kellogg
- Subjects
0301 basic medicine ,Male ,Immunogen ,viruses ,General Physics and Astronomy ,HIV Antibodies ,HIV Envelope Protein gp120 ,Serology ,law.invention ,Epitopes ,0302 clinical medicine ,law ,Medicine ,Phylogeny ,Antibody-dependent cell-mediated cytotoxicity ,AIDS Vaccines ,Multidisciplinary ,biology ,virus diseases ,Recombinant Proteins ,3. Good health ,Killer Cells, Natural ,030220 oncology & carcinogenesis ,Recombinant DNA ,Regression Analysis ,Female ,Simian Immunodeficiency Virus ,Antibody ,Protein Binding ,Science ,General Biochemistry, Genetics and Molecular Biology ,Article ,Pentavalent vaccine ,03 medical and health sciences ,Phagocytosis ,Neutralization Tests ,Predictive Value of Tests ,Animals ,Humans ,business.industry ,General Chemistry ,Complement System Proteins ,Vaccine efficacy ,Virology ,Macaca mulatta ,030104 developmental biology ,Immunization ,Immunology ,Mutation ,biology.protein ,HIV-1 ,Leukocytes, Mononuclear ,business - Abstract
The RV144 Thai trial HIV-1 vaccine of recombinant poxvirus (ALVAC) and recombinant HIV-1 gp120 subtype B/subtype E (B/E) proteins demonstrated 31% vaccine efficacy. Here we design an ALVAC/Pentavalent B/E/E/E/E vaccine to increase the diversity of gp120 motifs in the immunogen to elicit a broader antibody response and enhance protection. We find that immunization of rhesus macaques with the pentavalent vaccine results in protection of 55% of pentavalent-vaccine-immunized macaques from simian–human immunodeficiency virus (SHIV) challenge. Systems serology of the antibody responses identifies plasma antibody binding to HIV-infected cells, peak ADCC antibody titres, NK cell-mediated ADCC and antibody-mediated activation of MIP-1β in NK cells as the four immunological parameters that best predict decreased infection risk that are improved by the pentavalent vaccine. Thus inclusion of additional gp120 immunogens to a pox-prime/protein boost regimen can augment antibody responses and enhance protection from a SHIV challenge in rhesus macaques., A previous human HIV-1 vaccine clinical trial, boosting with HIV envelope protein from two strains, demonstrated moderate vaccine efficacy. Here, Bradley et al. show that a pentavalent HIV envelope protein boost improves protection from viral challenge in non-human primates and they identify immune correlates of protection.
- Published
- 2017
8. Recombinant HIV-1 vaccine candidates based on replication-defective flavivirus vector
- Author
-
B. H. Oakes, Thierry Calandra, Chelsey Bennett, Kathryn E. Foulds, Ralf Wagner, Matthieu Perreau, Mark Parrington, Annette Ives, J. McElrath, Konstantin V. Pugachev, Bertram L. Jacobs, Thierry Roger, Georgia D. Tomaras, Mariano Esteban, Guido Ferrari, Nicole L. Yates, David C. Montefiori, Harold Kleanthous, Mario Roederer, Bryan T. Mayer, Karen V. Kibler, Giuseppe Pantaleo, S. Ding, M. Vaine, Sanjay Phogat, Benedikt Asbach, Beatriz Perdiguero, Shing Fen Kao, Deborah T. Weiss, A. G. Spies, Gregory J. Mize, Jim Tartaglia, Raphael Gottardo, Maryann Giel-Moloney, and Bill & Melinda Gates Foundation
- Subjects
0301 basic medicine ,viruses ,Genetic Vectors ,Heterologous ,lcsh:Medicine ,HIV Infections ,Cross Reactions ,Biology ,Article ,Virus ,law.invention ,Mice ,03 medical and health sciences ,0302 clinical medicine ,law ,Chlorocebus aethiops ,Animals ,030212 general & internal medicine ,Vector (molecular biology) ,Neutralizing antibody ,lcsh:Science ,Vero Cells ,AIDS Vaccines ,Mice, Inbred BALB C ,Vaccines, Synthetic ,Multidisciplinary ,Virulence ,Molecular medicine ,Molecular engineering ,Flavivirus ,Immunogenicity ,lcsh:R ,Defective Viruses ,virus diseases ,biology.organism_classification ,Antibodies, Neutralizing ,Macaca mulatta ,Virology ,Vaccination ,030104 developmental biology ,Preclinical research ,HIV-1 ,biology.protein ,Recombinant DNA ,Female ,lcsh:Q - Abstract
Multiple approaches utilizing viral and DNA vectors have shown promise in the development of an effective vaccine against HIV. In this study, an alternative replication-defective flavivirus vector, RepliVax (RV), was evaluated for the delivery of HIV-1 immunogens. Recombinant RV-HIV viruses were engineered to stably express clade C virus Gag and Env (gp120TM) proteins and propagated in Vero helper cells. RV-based vectors enabled efficient expression and correct maturation of Gag and gp120TM proteins, were apathogenic in a sensitive suckling mouse neurovirulence test, and were similar in immunogenicity to recombinant poxvirus NYVAC-HIV vectors in homologous or heterologous prime-boost combinations in mice. In a pilot NHP study, immunogenicity of RV-HIV viruses used as a prime or boost for DNA or NYVAC candidates was compared to a DNA prime/NYVAC boost benchmark scheme when administered together with adjuvanted gp120 protein. Similar neutralizing antibody titers, binding IgG titers measured against a broad panel of Env and Gag antigens, and ADCC responses were observed in the groups throughout the course of the study, and T cell responses were elicited. The entire data demonstrate that RV vectors have the potential as novel HIV-1 vaccine components for use in combination with other promising candidates to develop new effective vaccination strategies., The study was funded by the Bill & Melinda Gates Foundation (project nr OPP1040705) and we thank Dr. Pervin Amklesaria and Dr. Nina Russell (Gates Foundation) for their guidance of this study. We thank the Poxvirus T-cell Vaccine Discovery Consortium funded by the Bill & Melinda Gates Foundation (OPP38599) for the development and provision of NYVAC and DNA vectors; support for the ICS and antibody immune monitoring assays was provided by the Vaccine Immune Monitoring Centers (OPP1032325 & OPP1032144), and support for the statistical analysis from the Vaccine Immunology Statistical Center (OPP1032317), all part of the Collaboration for AIDS Vaccine Discovery.
- Published
- 2019
9. Comparison of Immunogenicity in Rhesus Macaques of Transmitted-Founder, HIV-1 Group M Consensus, and Trivalent Mosaic Envelope Vaccines Formulated as a DNA Prime, NYVAC, and Envelope Protein Boost
- Author
-
Barton F. Haynes, Giuseppe Pantaleo, Harikrishnan Balachandran, Hua-Xin Liao, Barbara K. Felber, David C. Montefiori, Mariano Esteban, Michael S. Seaman, Sampa Santra, Carmen E. Gómez, Margherita Rosati, Nathan Vandergrift, Kevin O. Saunders, Jim Tartaglia, Michelle A. Lifton, Sanjay Phogat, Beatriz Perdiguero, Robert Parks, George N. Pavlakis, Richard M. Scearce, Elena E. Giorgi, Karen V. Kibler, Krissey E. Lloyd, S. Munir Alam, Bertram L. Jacobs, Linh Mach, Bette T. Korber, Sandrine L. Hulot, Norman L. Letvin, and Laura L. Sutherland
- Subjects
CD4-Positive T-Lymphocytes ,Enzyme-Linked Immunospot Assay ,Immunogen ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,HIV Antibodies ,Microbiology ,Epitope ,Interferon-gamma ,Immune system ,Antigen ,Virology ,Vaccines and Antiviral Agents ,Consensus Sequence ,Vaccines, DNA ,medicine ,Animals ,Humans ,Aspartate Aminotransferases ,Antigens, Viral ,biology ,Immunogenicity ,ELISPOT ,Vaccination ,SAIDS Vaccines ,env Gene Products, Human Immunodeficiency Virus ,Antibodies, Neutralizing ,Macaca mulatta ,medicine.anatomical_structure ,Insect Science ,Vaccines, Subunit ,HIV-1 ,biology.protein ,Antibody - Abstract
An effective human immunodeficiency virus type 1 (HIV-1) vaccine must induce protective antibody responses, as well as CD4 + and CD8 + T cell responses, that can be effective despite extraordinary diversity of HIV-1. The consensus and mosaic immunogens are complete but artificial proteins, computationally designed to elicit immune responses with improved cross-reactive breadth, to attempt to overcome the challenge of global HIV diversity. In this study, we have compared the immunogenicity of a transmitted-founder (T/F) B clade Env (B.1059), a global group M consensus Env (Con-S), and a global trivalent mosaic Env protein in rhesus macaques. These antigens were delivered using a DNA prime-recombinant NYVAC (rNYVAC) vector and Env protein boost vaccination strategy. While Con-S Env was a single sequence, mosaic immunogens were a set of three Envs optimized to include the most common forms of potential T cell epitopes. Both Con-S and mosaic sequences retained common amino acids encompassed by both antibody and T cell epitopes and were central to globally circulating strains. Mosaics and Con-S Envs expressed as full-length proteins bound well to a number of neutralizing antibodies with discontinuous epitopes. Also, both consensus and mosaic immunogens induced significantly higher gamma interferon (IFN-γ) enzyme-linked immunosorbent spot assay (ELISpot) responses than B.1059 immunogen. Immunization with these proteins, particularly Con-S, also induced significantly higher neutralizing antibodies to viruses than B.1059 Env, primarily to tier 1 viruses. Both Con-S and mosaics stimulated more potent CD8-T cell responses against heterologous Envs than did B.1059. Both antibody and cellular data from this study strengthen the concept of using in silico -designed centralized immunogens for global HIV-1 vaccine development strategies. IMPORTANCE There is an increasing appreciation for the importance of vaccine-induced anti-Env antibody responses for preventing HIV-1 acquisition. This nonhuman primate study demonstrates that in silico -designed global HIV-1 immunogens, designed for a human clinical trial, are capable of eliciting not only T lymphocyte responses but also potent anti-Env antibody responses.
- Published
- 2015
- Full Text
- View/download PDF
10. HIV/AIDS Vaccine Candidates Based on Replication-Competent Recombinant Poxvirus NYVAC-C-KC Expressing Trimeric gp140 and Gag-Derived Virus-Like Particles or Lacking the Viral Molecule B19 That Inhibits Type I Interferon Activate Relevant HIV-1-Specific B and T Cell Immune Functions in Nonhuman Primates
- Author
-
Steven G. Self, Brian Burke, Susan W. Barnett, Song Ding, Beatriz Perdiguero, Karen V. Kibler, Guido Ferrari, Juan García-Arriaza, Raphael Gottardo, Anthony D. Cristillo, Ralf Wagner, Kathryn E. Foulds, Giuseppe Pantaleo, Jonathan L. Heeney, Mario Roederer, Michael S. Seaman, Bertram L. Jacobs, Georgia D. Tomaras, Nicole L. Yates, Mariano Esteban, Deborah E. Weiss, Carter Lee, Sanjay Phogat, Bhavesh Borate, Jim Tartaglia, and David C. Montefiori
- Subjects
CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,HIV Antigens ,viruses ,T cell ,Immunology ,HIV Infections ,Vaccinia virus ,CD8-Positive T-Lymphocytes ,HIV Antibodies ,HIV Envelope Protein gp120 ,Biology ,Microbiology ,Virus ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Antigen ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Vaccines, Virus-Like Particle ,030212 general & internal medicine ,Vector (molecular biology) ,Receptors, Interferon ,AIDS Vaccines ,Immunogenicity ,Vaccination ,Antibody-Dependent Cell Cytotoxicity ,env Gene Products, Human Immunodeficiency Virus ,virus diseases ,Antibodies, Neutralizing ,Macaca mulatta ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Insect Science ,Interferon Type I ,Vaccinia - Abstract
The nonreplicating attenuated poxvirus vector NYVAC expressing clade C(CN54) HIV-1 Env(gp120) and Gag-Pol-Nef antigens (NYVAC-C) showed limited immunogenicity in phase I clinical trials. To enhance the capacity of the NYVAC vector to trigger broad humoral responses and a more balanced activation of CD4 + and CD8 + T cells, here we compared the HIV-1-specific immunogenicity elicited in nonhuman primates immunized with two replicating NYVAC vectors that have been modified by the insertion of the K1L and C7L vaccinia virus host range genes and express the clade C(ZM96) trimeric HIV-1 gp140 protein or a Gag(ZM96)-Pol-Nef(CN54) polyprotein as Gag-derived virus-like particles (termed NYVAC-C-KC). Additionally, one NYVAC-C-KC vector was generated by deleting the viral gene B19R , an inhibitor of the type I interferon response (NYVAC-C-KC-ΔB19R). An immunization protocol mimicking that of the RV144 phase III clinical trial was used. Two groups of macaques received two doses of the corresponding NYVAC-C-KC vectors (weeks 0 and 4) and booster doses with NYVAC-C-KC vectors plus the clade C HIV-1 gp120 protein (weeks 12 and 24). The two replicating NYVAC-C-KC vectors induced enhanced and similar HIV-1-specific CD4 + and CD8 + T cell responses, similar levels of binding IgG antibodies, low levels of IgA antibodies, and high levels of antibody-dependent cellular cytotoxicity responses and HIV-1-neutralizing antibodies. Small differences within the NYVAC-C-KC-ΔB19R group were seen in the magnitude of CD4 + and CD8 + T cells, the induction of some cytokines, and the neutralization of some HIV-1 isolates. Thus, replication-competent NYVAC-C-KC vectors acquired relevant immunological properties as vaccine candidates against HIV/AIDS, and the viral B19 molecule exerts some control of immune functions. IMPORTANCE It is of special importance to find a safe and effective HIV/AIDS vaccine that can induce strong and broad T cell and humoral immune responses correlating with HIV-1 protection. Here we developed novel replicating poxvirus NYVAC-based HIV/AIDS vaccine candidates expressing clade C HIV-1 antigens, with one of them lacking the vaccinia virus B19 protein, an inhibitor of the type I interferon response. Immunization of nonhuman primates with these novel NYVAC-C-KC vectors and the protein component gp120 elicited high levels of T cell and humoral immune responses, with the vector containing a deletion in B19R inducing a trend toward a higher magnitude of CD4 + and CD8 + T cell responses and neutralization of some HIV-1 strains. These poxvirus vectors could be considered HIV/AIDS vaccine candidates based on their activation of potential immune correlates of protection.
- Published
- 2017
- Full Text
- View/download PDF
11. Antibodies with High Avidity to the gp120 Envelope Protein in Protection from Simian Immunodeficiency Virus SIV mac251 Acquisition in an Immunization Regimen That Mimics the RV-144 Thai Trial
- Author
-
Ranajit Pal, Poonam Pegu, Georgia D. Tomaras, Guido Ferrari, Nelson L. Michael, Yongjun Guan, Mangala Rao, Monica Vaccari, Jeffrey D. Lifson, Jim Tartaglia, David Venzon, Jerome H. Kim, Maria Grazia Ferrari, S. Munir Alam, Melvin N. Doster, Brandon F. Keele, Lauren Hudacik, Shari N. Gordon, David C. Montefiori, Claudio Fenizia, Erik Billings, Genoveffa Franchini, Stephen Whitney, and Donald Stablein
- Subjects
CD4-Positive T-Lymphocytes ,viruses ,Immunology ,Antibody Affinity ,Simian Acquired Immunodeficiency Syndrome ,CD8-Positive T-Lymphocytes ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Virus ,Viral Envelope Proteins ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Avidity ,RV 144 ,Membrane Glycoproteins ,SAIDS Vaccines ,virus diseases ,Simian immunodeficiency virus ,Vaccination ,Immunization ,Insect Science ,biology.protein ,Macaca ,Antibody - Abstract
The recombinant canarypox vector, ALVAC-HIV, together with human immunodeficiency virus (HIV) gp120 envelope glycoprotein, has protected 31.2% of Thai individuals from HIV acquisition in the RV144 HIV vaccine trial. This outcome was unexpected, given the limited ability of the vaccine components to induce CD8 + T-cell responses or broadly neutralizing antibodies. We vaccinated macaques with an immunization regimen intended to mimic the RV144 trial and exposed them intrarectally to a dose of the simian immunodeficiency virus SIV mac251 that transmits few virus variants, similar to HIV transmission to humans. Vaccination induced anti-envelope antibodies in all vaccinees and CD4 + and CD8 + T-cell responses. Three of the 11 macaques vaccinated with ALVAC-SIV/gp120 were protected from SIV mac251 acquisition, but the result was not significant. The remaining vaccinees were infected and progressed to disease. The magnitudes of vaccine-induced SIV mac251 -specific T-cell responses and binding antibodies were not significantly different between protected and infected animals. However, sera from protected animals had higher avidity antibodies to gp120, recognized the variable envelope regions V1/V2, and reduced SIV mac251 infectivity in cells that express high levels of α 4 β 7 integrins, suggesting a functional role of antibodies to V2. The current results emphasize the utility of determining the titer of repeated mucosal challenge in the preclinical evaluation of HIV vaccines.
- Published
- 2013
- Full Text
- View/download PDF
12. Magnitude and Breadth of the Neutralizing Antibody Response in the RV144 and Vax003 HIV-1 Vaccine Efficacy Trials
- Author
-
Merlin L. Robb, David C. Montefiori, Hasan Ahmed, Mark de Souza, Peter B. Gilbert, Jaranit Kaewkungwal, M. Anthony Moody, Mattia Bonsignori, Ying Huang, Faruk Sinangil, John C. Kappes, Jerome H. Kim, Victoria R. Polonis, Charla Andrews, Eric Sanders-Buell, Christina Ochsenbauer, Phillip W. Berman, Kelli Greene, Sorachai Nitayaphan, Carter Lee, Robert McLinden, Donald P. Francis, Punnee Pitisuttithum, Jim Tartaglia, Celia C. LaBranche, Haili Tang, Steve Self, Agnes Laurence-Chenine, Hongmei Gao, Barton F. Haynes, Chitraporn Karnasuta, Nelson L. Michael, Sodsai Tovanabutra, and Supachai Rerks-Ngarm
- Subjects
Human Immunodeficiency Virus Proteins ,Population ,HIV Infections ,HIV Antibodies ,Biology ,Gp41 ,Canarypox virus ,Virus ,Major Articles and Brief Reports ,Antigen ,Humans ,Immunology and Allergy ,Substance Abuse, Intravenous ,Neutralizing antibody ,education ,Immunization Schedule ,AIDS Vaccines ,education.field_of_study ,Antibodies, Monoclonal ,Thailand ,Entry into host ,Vaccine efficacy ,Antibodies, Neutralizing ,Virology ,Infectious Diseases ,AIDSVAX ,Immunology ,HIV-1 ,biology.protein ,HIV/AIDS ,Epitope Mapping - Abstract
It is widely believed that a neutralizing antibody (NAb) response of sufficient magnitude, breadth, and duration would be highly beneficial for human immunodeficiency virus type 1 (HIV-1) vaccines [1–3]. Indeed, Nabs protect against experimental challenge with simian human immunodeficiency virus (SHIV) in nonhuman primates [4–6], and they exert strong selective pressure on HIV-1 after infection in humans [7, 8]. Neutralization occurs when antibodies bind to functional envelope glycoprotein (Env) spikes on the virus surface to prevent entry into host cells [9–11]. Each Env spike consists of 3 surface gp120 molecules bound noncovalently to 3 transmembrane gp41 molecules [12, 13]. These glycoproteins exhibit an extraordinary degree of genetic and antigenic variability that poses major challenges for vaccine development [14, 15]. Moreover, the virus uses a number of mechanisms to evade NAbs [7, 12, 16]. As a result, a minor subset of circulating variants exhibit a highly neutralization-sensitive tier 1 phenotype and are often susceptible to vaccine-elicited NAbs, whereas most circulating strains exhibit a less sensitive tier 2 phenotype and have proven difficult to target with vaccines[1, 2, 15]. A recently completed HIV-1 vaccine efficacy trial in Thailand (RV144) showed that priming with a recombinant canarypox vector (vCP1521) and boosting with this vector plus bivalent gp120 protein (AIDSVAX B/E) can provide partial protection against the acquisition of HIV-1 infection in a community-based heterosexual population [17]. The same bivalent gp120 immunogen, when used alone and with an increased number of inoculations (Vax003 trial), showed no protection in a cohort of Thai injection drug users [18]. In addition, no overall protection was seen when a similar regimen of bivalent gp120 (AIDSVAX B/B) was used alone in a cohort of mostly men who have sex with men (MSM) in North America and the Netherlands (Vax004 trial) [19]. The gp120 protein component in all 3 clinical trials was designed to elicit NAbs [20, 21]. In Vax004, strong NAb responses were seen against a subset of tier 1 viruses, and sporadic weak responses were seen against tier 2 viruses [22]. Here we assessed the magnitude and breadth of NAb responses in RV144 and Vax003.
- Published
- 2012
- Full Text
- View/download PDF
13. Comparison of T cell immune responses induced by vectored HIV vaccines in non-human primates and humans
- Author
-
Christine Gaunt, John W. Shiver, Sheri Dubey, Jim Tartaglia, Liming Guan, Michael N. Robertson, Danilo R. Casimiro, Devan V. Mehrotra, Sanjay Gurunathan, Romnie Long, Kiersten Anderson, Kelly B. Collins, Aimin Tang, Andrew J. Bett, Suzanne Cole, Xiao Sun, Rose Fernandez, and Steve Meschino
- Subjects
Cellular immunity ,T-Lymphocytes ,viruses ,Immunization, Secondary ,HIV Infections ,Biology ,Adenoviridae ,Interferon-gamma ,Immune system ,Vaccines, DNA ,Animals ,Humans ,Vector (molecular biology) ,HIV vaccine ,AIDS Vaccines ,Immunity, Cellular ,Clinical Trials, Phase I as Topic ,General Veterinary ,General Immunology and Microbiology ,Immunogenicity ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,Genes, gag ,Macaca mulatta ,Virology ,Vaccination ,Infectious Diseases ,Immunization ,Models, Animal ,Lentivirus ,Immunology ,Molecular Medicine - Abstract
Following the disappointing outcome of the phase IIb test-of-concept step study in which Merck's adenovirus type 5 (Ad5) HIV-1 clade B gag/pol/nef vaccine failed to demonstrate efficacy in HIV high-risk individuals, an extensive review of the trial and preclinical studies which supported the trial is ongoing. One point of interest is how well preclinical nonhuman primate immunogenicity studies predicted what was observed in humans. Here we compare the HIV-1-specific cellular immune responses elicited in nonhuman primates and human clinical trial subjects to several HIV-1 vaccine candidates. We find that although rhesus macaques are immunologically more responsive to vaccination than humans, the hierarchy in potency of single-modality prime–boost regimens using several vector approaches (adenovirus, DNA, and pox vectors) was well predicted. Vaccine approaches using complex formulations such as novel adjuvants (DNA + CRL1005) or mixed-modality prime–boost (DNA/Ad5; Ad5/ALVAC) did not correlate as well between rhesus macaques and humans. Although the immunogenicity of the vaccines and vaccine regimens evaluated were not all accurately predicted, testing in rhesus macaques generally offers an indispensable tool for ranking the immunological potential of HIV-1 vaccine candidates.
- Published
- 2010
- Full Text
- View/download PDF
14. Preclinical Qualification of a New Multi-antigen Candidate Vaccine for Metastatic Melanoma
- Author
-
Thorsten U. Vogel, Ray Oomen, Danielle Salha, Neil L. Berinstein, Corey Lovitt, Tao Wen, Mark Parrington, Linong Zhang, Mei Tang, Lucian Visan, Bryan McNeil, Bill Bradley, Judy Caterini, Devender Sandhu, Belma Ljutic, Nancy Scollard, Shi-Xian Cao, Liwei He, Pamela Dunn, Jim Tartaglia, and Beata Gajewska
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,medicine.medical_treatment ,Immunology ,Antigen presentation ,Drug Evaluation, Preclinical ,Mice, Transgenic ,Poxviridae Infections ,Lymphocyte Activation ,Cancer Vaccines ,Mice ,Immune system ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,HLA-A2 Antigen ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Cloning, Molecular ,Neoplasm Metastasis ,Melanoma ,Cells, Cultured ,Pharmacology ,Melanoma-associated antigen ,business.industry ,Poxviridae ,Immunogenicity ,Viral Vaccine ,Viral Vaccines ,Immunotherapy ,business ,T-Lymphocytes, Cytotoxic - Abstract
New therapies are urgently required for the treatment of patients with melanoma. Here we describe the generation and preclinical evaluation of 3 new recombinant ALVAC(2) poxviruses vCP2264, vCP2291, and vCP2292 for their ability to induce the desired cellular immune responses against the encoded melanoma-associated antigens. This was done either in HLA-A2/K transgenic mice or using in vitro antigen-presentation studies. These studies demonstrated that the vaccine was able to induce HLA-A*0201-restricted T-cell responses against gp100 and NY-ESO-1, detectable directly ex vivo, in HLA-A2/K-transgenic mice. The in vitro antigen presentation studies, in the absence of appropriate animal models, demonstrated that target cells infected with the vaccine construct were lysed by MAGE-1, MAGE-3 or MART-1 peptide-specific T cells. These data indicate that ALVAC(2)-encoded melanoma-associated antigens can be properly processed and presented to induce antigen-specific cytotoxic T-cell responses. To enhance the immunogenicity of the melanoma antigens, a TRIad of COstimulatory Molecules (TRICOM) were also cloned into all 3 vectors. Increased in vitro proliferation and IFN-γ production was observed with all ALVAC(2) poxviruses encoding TRICOM, confirming the immune-enhancing effect of the ALVAC-encoded TRICOM. These studies demonstrated that all components of the vaccine were functionally active and provide a rationale for moving this candidate vaccine to the clinic.
- Published
- 2010
- Full Text
- View/download PDF
15. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses
- Author
-
Janice M. Moser, Rama Akondy, Elias A. Said, Bader Yassine-Diab, Rafick Pierre Sekaly, Aline Rinfret, Peter Wilkinson, Mark J. Cameron, Robert S. Balderas, Denis Gaucher, Nadia Kettaf, David J. Kelvin, Jim Tartaglia, Geneviève Boucher, Robert Clum, Elias K. Haddad, Bastian R. Angermann, Abdelali Filali-Mouhim, Giuseppe Pantaleo, René Therrien, Riyaz Mehta, Donald Drake, Roland Somogyi, Erika Castro, Larry D. Greller, and Younes Chouikh
- Subjects
Transcription, Genetic ,T-Lymphocytes ,animal diseases ,Interleukin-1beta ,Immunology ,Yellow fever vaccine ,chemical and pharmacologic phenomena ,Biology ,Article ,Immune System Phenomena ,Immune system ,Immunity ,medicine ,Humans ,Immunology and Allergy ,Gene Regulatory Networks ,B-Lymphocytes ,Cell Proliferation ,Flow Cytometry ,Gene Expression Profiling ,Gene Expression Regulation ,Immune System Processes ,Immunity, Innate ,Lymphocyte Subsets ,Vaccination ,Yellow Fever Vaccine ,Innate immune system ,CCL18 ,Articles ,biochemical phenomena, metabolism, and nutrition ,Acquired immune system ,Virology ,Immunization ,bacteria ,medicine.drug - Abstract
Correlates of immune-mediated protection to most viral and cancer vaccines are still unknown. This impedes the development of novel vaccines to incurable diseases such as HIV and cancer. In this study, we have used functional genomics and polychromatic flow cytometry to define the signature of the immune response to the yellow fever (YF) vaccine 17D (YF17D) in a cohort of 40 volunteers followed for up to 1 yr after vaccination. We show that immunization with YF17D leads to an integrated immune response that includes several effector arms of innate immunity, including complement, the inflammasome, and interferons, as well as adaptive immunity as shown by an early T cell response followed by a brisk and variable B cell response. Development of these responses is preceded, as demonstrated in three independent vaccination trials and in a novel in vitro system of primary immune responses (modular immune in vitro construct [MIMIC] system), by the coordinated up-regulation of transcripts for specific transcription factors, including STAT1, IRF7, and ETS2, which are upstream of the different effector arms of the immune response. These results clearly show that the immune response to a strong vaccine is preceded by coordinated induction of master transcription factors that lead to the development of a broad, polyfunctional, and persistent immune response that integrates all effector cells of the immune system.
- Published
- 2008
- Full Text
- View/download PDF
16. Adjuvant-dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition
- Author
-
Zhong-Min Ma, Nelson L. Michael, Donald N. Forthal, Genoveffa Franchini, Jim Tartaglia, Margaret E. Ackerman, Stephen Whitney, Mitzi M. Donaldson, Xiaoying Shen, Amy W. Chung, Chris Bailey-Kellogg, Mark J. Cameron, Shari N. Gordon, Marjorie Robert-Guroff, Guido Ferrari, Erik Billings, Rafick-Pierre Sekaly, Richard A. Koup, Nicolo Binello, Mario Roederer, Galit Alter, Eric P. Brown, Christopher J. Miller, Karen G Dowell, David S Quinn, Donald Stablein, Susan W. Barnett, Monica Vaccari, Brandon F. Keele, Francesca Caccuri, Kathryn E. Foulds, Luca Schifanella, Jerome H. Kim, DeVon Thompson, Melvin N. Doster, David C. Montefiori, Slim Fourati, Adrian B. McDermott, Vaniambadi S. Kalyanaraman, Mangala Rao, Frank Liang, Silvia Ratto-Kim, Diego A. Vargas-Inchaustegui, Georgia D. Tomaras, Poonam Pegu, Namal P.M. Liyanage, Maria Grazia Ferrari, Massimiliano Bissa, Karin Loré, Matthew Blackburn, Tran B. Phan, Sanjay Phogat, and David Venzon
- Subjects
Genetics and Molecular Biology (all) ,0301 basic medicine ,viruses ,medicine.medical_treatment ,Simian Acquired Immunodeficiency Syndrome ,Adaptive Immunity ,Medical and Health Sciences ,Medicine (all) ,Biochemistry, Genetics and Molecular Biology (all) ,Biochemistry ,Random Allocation ,Immunogenicity, Vaccine ,Viral Envelope Proteins ,Immunologic ,Innate ,Lymphocytes ,Mucosal ,Membrane Glycoproteins ,Immunogenicity ,Viral Vaccine ,Innate lymphoid cell ,Interleukin-17 ,Simian immunodeficiency virus ,SAIDS Vaccines ,virus diseases ,General Medicine ,Acquired immune system ,Alum Compounds ,Simian Immunodeficiency Virus ,Adjuvant ,Signal Transduction ,Immunology ,complex mixtures ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Adjuvants, Immunologic ,Immunity ,medicine ,Animals ,Adjuvants ,Immunity, Mucosal ,business.industry ,Vaccine trial ,Viral Vaccines ,Vaccine efficacy ,Macaca mulatta ,Immunity, Innate ,Good Health and Well Being ,030104 developmental biology ,Immunoglobulin G ,ras Proteins ,business ,Transcriptome ,Vaccine - Abstract
A recombinant vaccine containing Aventis Pasteur's canarypox vector (ALVAC)-HIV and gp120 alum decreased the risk of HIV acquisition in the RV144 vaccine trial. The substitution of alum with the more immunogenic MF59 adjuvant is under consideration for the next efficacy human trial. We found here that an ALVAC-simian immunodeficiency virus (SIV) and gp120 alum (ALVAC-SIV + gp120) equivalent vaccine, but not an ALVAC-SIV + gp120 MF59 vaccine, was efficacious in delaying the onset of SIVmac251 in rhesus macaques, despite the higher immunogenicity of the latter adjuvant. Vaccine efficacy was associated with alum-induced, but not with MF59-induced, envelope (Env)-dependent mucosal innate lymphoid cells (ILCs) that produce interleukin (IL)-17, as well as with mucosal IgG to the gp120 variable region 2 (V2) and the expression of 12 genes, ten of which are part of the RAS pathway. The association between RAS activation and vaccine efficacy was also observed in an independent efficacious SIV-vaccine approach. Whether RAS activation, mucosal ILCs and antibodies to V2 are also important hallmarks of HIV-vaccine efficacy in humans will require further studies.
- Published
- 2015
17. Head-to-Head Comparison of Poxvirus NYVAC and ALVAC Vectors Expressing Identical HIV-1 Clade C Immunogens in Prime-Boost Combination with Env Protein in Nonhuman Primates
- Author
-
Jim Tartaglia, Carter Lee, Brian Burke, Jonathan L. Heeney, Mario Roederer, Mariano Esteban, Michael S. Seaman, Nicole L. Yates, Giuseppe Pantaleo, Steve Self, Song Ding, Jiansheng Yao, Benedikt Asbach, Bertram L. Jacobs, Guido Ferrari, Karen V. Kibler, Georgia D. Tomaras, Deborah T. Weiss, Patrick Farrell, Juan García-Arriaza, Shing Fen Kao, Anthony D. Cristillo, Ralf Wagner, Natalie Hawkins, Sanjay Phogat, Kathryn E. Foulds, Celia C. LaBranche, David C. Montefiori, Susan W. Barnett, Xiaoying Shen, Beatriz Perdiguero, Adrian B. McDermott, Heeney, Jonathan [0000-0003-2702-1621], and Apollo - University of Cambridge Repository
- Subjects
Immunogen ,HIV Antigens ,viruses ,Immunology ,Genetic Vectors ,HIV Infections ,Chick Embryo ,Viral Plaque Assay ,V3 loop ,HIV Antibodies ,Microbiology ,Antigen ,Virology ,Vaccines and Antiviral Agents ,Animals ,Vector (molecular biology) ,HIV vaccine ,Promoter Regions, Genetic ,Antibody-dependent cell-mediated cytotoxicity ,AIDS Vaccines ,Vaccines, Synthetic ,biology ,Immunogenicity ,Poxviridae ,virus diseases ,Gene Products, env ,Viral Vaccines ,Antibodies, Neutralizing ,Macaca mulatta ,Insect Science ,biology.protein ,Antibody - Abstract
We compared the HIV-1-specific cellular and humoral immune responses elicited in rhesus macaques immunized with two poxvirus vectors (NYVAC and ALVAC) expressing the same HIV-1 antigens from clade C, Env gp140 as a trimeric cell-released protein and a Gag-Pol-Nef polyprotein as Gag-induced virus-like particles (VLPs) (referred to as NYVAC-C and ALVAC-C). The immunization protocol consisted of two doses of the corresponding poxvirus vector plus two doses of a combination of the poxvirus vector and a purified HIV-1 gp120 protein from clade C. This immunogenicity profile was also compared to that elicited by vaccine regimens consisting of two doses of the ALVAC vector expressing HIV-1 antigens from clades B/E (ALVAC-vCP1521) plus two doses of a combination of ALVAC-vCP1521 and HIV-1 gp120 protein from clades B/E (similar to the RV144 trial regimen) or clade C. The results showed that immunization of macaques with NYVAC-C stimulated at different times more potent HIV-1-specific CD4 + T-cell responses and induced a trend toward higher-magnitude HIV-1-specific CD8 + T-cell immune responses than did ALVAC-C. Furthermore, NYVAC-C induced a trend toward higher levels of binding IgG antibodies against clade C HIV-1 gp140, gp120, or murine leukemia virus (MuLV) gp70-scaffolded V1/V2 and toward best cross-clade-binding IgG responses against HIV-1 gp140 from clades A, B, and group M consensus, than did ALVAC-C. Of the linear binding IgG responses, most were directed against the V3 loop in all immunization groups. Additionally, NYVAC-C and ALVAC-C also induced similar levels of HIV-1-neutralizing antibodies and antibody-dependent cellular cytotoxicity (ADCC) responses. Interestingly, binding IgA antibody levels against HIV-1 gp120 or MuLV gp70-scaffolded V1/V2 were absent or very low in all immunization groups. Overall, these results provide a comprehensive survey of the immunogenicity of NYVAC versus ALVAC expressing HIV-1 antigens in nonhuman primates and indicate that NYVAC may represent an alternative candidate to ALVAC in the development of a future HIV-1 vaccine. IMPORTANCE The finding of a safe and effective HIV/AIDS vaccine immunogen is one of the main research priorities. Here, we generated two poxvirus-based HIV vaccine candidates (NYVAC and ALVAC vectors) expressing the same clade C HIV-1 antigens in separate vectors, and we analyzed in nonhuman primates their immunogenicity profiles. The results showed that immunization with NYVAC-C induced a trend toward higher HIV-1-specific cellular and humoral immune responses than did ALVAC-C, indicating that this new NYVAC vector could be a novel optimized HIV/AIDS vaccine candidate for human clinical trials.
- Published
- 2015
18. Systemic Immunization with an ALVAC-HIV-1/Protein Boost Vaccine Strategy Protects Rhesus Macaques from CD4 + T-Cell Loss and Reduces both Systemic and Mucosal Simian-Human Immunodeficiency Virus SHIV KU2 RNA Levels
- Author
-
Jim Tartaglia, Genoveffa Franchini, Nicolas Rose, Marcin Moniuszko, Lindsey Hocker, Janos Nacsa, Vaniambadi S. Kalyanaraman, Igor M. Belyakov, Sampa Santra, Yvette Edghill-Smith, Norman L. Letvin, Lauren Hudacik, Ranajit Pal, David C. Montefiori, Zdeněk Hel, David Venzon, Jay A. Berzofsky, Phillip D. Markham, and Robyn Washington Parks
- Subjects
Canarypox ,biology ,viruses ,Immunogenicity ,Immunology ,virus diseases ,Viremia ,Simian immunodeficiency virus ,medicine.disease_cause ,medicine.disease ,biology.organism_classification ,Microbiology ,Virology ,Virus ,Viral replication ,Insect Science ,Lentivirus ,medicine ,Viral load - Abstract
Due to the alarming spread of human immunodeficiency virus type 1 (HIV-1) infection worldwide, the development of a prophylactic vaccine is critical. Vaccine strategies for AIDS have included the use of structural and nonstructural HIV subunit proteins or peptides, naked DNA, bacterial and viral live vectors, or combinations of the above. Most of the vaccines developed thus far induce T-cell responses but are unable to induce neutralizing antibodies to the Env of primary HIV-1 isolates. The fact that a decrease in plasma viremia (9, 41) is concomitant with the appearance of virus-specific cytotoxic T lymphocytes (CTLs) and depletion of CD8+ T cells in simian immunodeficiency virus (SIV)-infected macaques or HIV-infected chimpanzees suggests that CD8+ T cells are able to partially control HIV-1/SIV replication (15, 36, 43, 52). Indeed, several “T-cell vaccines” based on DNA and live vectors confer protection from high-level replication of challenge viruses in rhesus macaques (2, 4, 8, 30, 31, 48). The extent of the decrease in viral replication induced by these vaccines is variable in nonhuman primates, and protection from disease appears to be dependent on the virulence of the virus used in the challenge experiments (28). The relative efficacy of poxvirus-based vaccine candidates with various degrees of attenuation has been extensively studied in rhesus macaques after challenge with SIV, SHIV, and HIV-2 isolates (1, 3, 8, 24, 25, 29-31, 48). These vaccine modalities elicit variable levels of cell-mediated immune response and prevent infection after challenge exposure to viruses with low virulence, such as HIV-2 (3, 25), and to other somewhat-attenuated SIV isolates (34, 35). Importantly, these vaccine modalities were also able to significantly reduce virus load after challenge with highly pathogenic SIV isolates (8, 29, 32, 48). An ALVAC-SIV vaccine encoding the gag, pol, and env genes of the SIVmac251 isolate was able to reduce plasma virus load during primary infection and conferred protection from CD4+ loss during both acute and chronic phases of infection after rectal exposure to a highly pathogenic SIVmac251 isolate (50). Among the pox vector-based vaccines, several ALVAC-based HIV-1 vaccines have been tested in phase I and II clinical trials and have been shown to be safe and immunogenic in humans (10, 14, 18, 21, 27). Whether the immunogenicity of these vaccine candidates will be sufficient to protect humans from HIV-1 remains unknown (5, 11, 44). The ongoing human phase III trial in Thailand will provide key information in this regard. Here, we designed a study to assess whether systemic immunization with recombinant canarypox expressing either HIV-1 gp120 or gp160 followed by a boost with either purified gp120 or gp140 proteins would confer protection after mucosal challenge with the pathogenic SHIVKU2. We also investigated whether the addition of the Tat protein of HIV-1 as an immunogen could provide better protection, since a few studies have demonstrated that immunization of macaques with Tat protein either alone or as a Nef-Tat fusion protein in a multicomponent subunit vaccine confers an advantage in a SHIV89.6P model (12, 13, 56) but not in other studies (53). Here, we found that immunization of macaques with this type of ALVAC-based vaccine formulation elicited both antibody and cellular responses and significantly decreased plasma viremia and CD4+ T-cell loss after rectal exposure to the SHIVKU2 isolate. Tat protein immunization had no additive effect on the reduction of virus load in vaccinated rhesus macaques, as also observed by others (42, 46, 53).
- Published
- 2006
- Full Text
- View/download PDF
19. Improved Vaccine Protection from Simian AIDS by the Addition of Nonstructural Simian Immunodeficiency Virus Genes
- Author
-
Elzbieta Tryniszewska, Janos Nacsa, Genoveffa Franchini, Zdeněk Hel, Jim Tartaglia, George N. Pavlakis, Phillip D. Markham, Barbara K. Felber, Mark G. Lewis, and Wen-Po Tsai
- Subjects
Genes, Viral ,T-Lymphocytes ,T cell ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Viremia ,Biology ,medicine.disease_cause ,Immune system ,medicine ,Animals ,Immunology and Allergy ,Antigens, Viral ,SAIDS Vaccines ,virus diseases ,Simian immunodeficiency virus ,medicine.disease ,Macaca mulatta ,Virology ,Vaccination ,medicine.anatomical_structure ,Viral replication ,Simian AIDS ,Simian Immunodeficiency Virus ,CD8 - Abstract
An HIV-1 vaccine able to induce broad CD4+ and CD8+ T cell responses may provide long-term control of viral replication. In this study we directly assess the relative benefit of immunization with vaccines expressing three structural Ags (Gag, Pol, and Env), three early regulatory proteins (Rev, Tat, and Nef), or a complex vaccine expressing all six Ags. The simultaneous administration of all six Ags during vaccination resulted in Ag competition manifested by a relative reduction of CD8+ T cell and lymphoproliferative responses to individual Ags. Despite the Ag competition, vaccination with all six Ags resulted in a delay in the onset and a decrease in the extent of acute viremia after mucosal challenge exposure to highly pathogenic SIVmac251. Reduced levels of acute viremia correlated with lower post-set point viremia and long-term control of infection. In immunized animals, virus-specific CD4+ T cell and lymphoproliferative responses were preserved during acute viremia, and the maintenance of these responses predicted the long-term virological outcome. Taken together, these results suggest that the breadth of the immune response is probably more important than high frequency responses to a limited number of epitopes. These data provide the first clear evidence of the importance of nonstructural HIV Ags as components of an HIV-1 vaccine.
- Published
- 2006
- Full Text
- View/download PDF
20. Heterologous Human Immunodeficiency Virus Type 1 Priming-Boosting Immunization Strategies Involving Replication-Defective Adenovirus and Poxvirus Vaccine Vectors
- Author
-
Andrew J. Bett, Romnie Long, Minchun Chen, Aimin Tang, Michael Chastain, Emilio A. Emini, Danilo R. Casimiro, John W. Shiver, Tong-Ming Fu, Mary-Ellen Davies, Sanjay Gurunathan, Troy McKelvey, Jim Tartaglia, and Keith A. Wilson
- Subjects
Cellular immunity ,T-Lymphocytes ,viruses ,Genetic Vectors ,Immunology ,Immunization, Secondary ,Gene Products, gag ,Heterologous ,HIV Infections ,Biology ,Virus Replication ,medicine.disease_cause ,complex mixtures ,Microbiology ,Virus ,Adenoviridae ,Interferon-gamma ,Immune system ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,Poxviridae ,AIDS Vaccines ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Macaca mulatta ,Vaccination ,Viral replication ,Insect Science ,HIV-1 ,Immunization - Abstract
We compared the human immunodeficiency virus type 1 (HIV-1)-specific cellular immune responses elicited in nonhuman primates by HIV-1 gag -expressing replication-defective adenovirus serotype 5 (Ad5) or poxvirus vectors, used either alone or in combination with each other. The responses arising from a heterologous Ad5 priming-poxvirus boosting regimen were significantly greater than those elicited by homologous regimens with the individual vectors or by a heterologous poxvirus priming-Ad5 boosting regimen. The heterologous Ad5 priming-poxvirus boosting approach may have potential utility in humans as a means of inducing high levels of cellular immunity.
- Published
- 2004
- Full Text
- View/download PDF
21. Poxvirus-based vaccine candidates for HIV: two decades of experience with special emphasis on canarypox vectors
- Author
-
Stanley A. Plotkin, Genoveffa Franchini, Lynn Baglyos, Sanjay Gurunathan, and Jim Tartaglia
- Subjects
Modified vaccinia Ankara ,Canarypox ,Immunology ,HIV Infections ,Biology ,Avipoxvirus ,chemistry.chemical_compound ,Drug Discovery ,Animals ,Humans ,Vector (molecular biology) ,HIV vaccine ,AIDS Vaccines ,Pharmacology ,Immunogenicity ,HIV ,biology.organism_classification ,Virology ,Disease Models, Animal ,chemistry ,HIV-2 ,HIV-1 ,Molecular Medicine ,Simian Immunodeficiency Virus ,Vaccinia ,Reassortant Viruses - Abstract
Poxvirus vectors have emerged as important vectors for licensed veterinary vaccines and candidate vaccines for humans. Vaccinia, highly-attenuated vaccinia strains and avipoxviruses have been assessed extensively in preclinical models, as well as in humans, to determine their immunogenicity and protective efficacy against HIV. The attenuated vaccinia strains and avipoxviruses have been shown to be safe and able to carry HIV genes and express their proteins to induce both antibodies and cellular immune responses. Preclinical studies show protection against HIV challenge. When using a live attenuated vector system, one must be cognizant of the potential for immune dampening because of vector-specific immunity. In this regard, avipoxviruses, such as canarypox, appear free of the inhibitory effects of vector immunity and repeated use. Unlike vaccinia-based vectors derived from classical vaccine strains, NYVAC and modified vaccinia Ankara may be less susceptible to this effect. In the coming 5 to 10 years, we will certainly know whether this class of vaccine candidates, either alone or in a prime-boost format with other vectors or proteins, will contribute to HIV disease management either from a preventive or therapeutic perspective. Additional Phase I and II studies, as well as human efficacy trials will provide new information. Furthermore, it is hoped that this body of data will contribute to a better understanding of the relevance of specific immunogenicity end points to protection and the predictive value of available animal models in HIV vaccine development.
- Published
- 2004
- Full Text
- View/download PDF
22. Local immunotherapy of spontaneous feline fibrosarcomas using recombinant poxviruses expressing interleukin 2 (IL2)
- Author
-
Tafani Jp, Jim Tartaglia, Therese-Marie Jourdier, Delisle F, Catherine Moste, Moingeon P, Devauchelle P, and Marie-Claude Bonnet
- Subjects
Male ,Skin Neoplasms ,Fibrosarcoma ,medicine.medical_treatment ,Genetic Vectors ,Context (language use) ,Canarypox virus ,Cat Diseases ,Recombinant virus ,Virus ,Viral vector ,chemistry.chemical_compound ,Dogs ,Genetics ,medicine ,Animals ,Poxviridae ,Luciferases ,Molecular Biology ,biology ,Viral Vaccines ,Immunotherapy ,biology.organism_classification ,Virology ,chemistry ,Animals, Domestic ,Cats ,Interleukin-2 ,Molecular Medicine ,Female ,Neoplasm Recurrence, Local ,Vaccinia - Abstract
We tested the canarypox virus vector ALVAC and the genetically attenuated vaccinia virus vector NYVAC as vehicles for achieving local immunomodulation in domestic animals bearing spontaneous tumours. Following intratumoral administration of ALVAC-, or NYVAC-luciferase in dogs with melanoma, it was demonstrated that viral recombinants remained localized along the needle track, with no virus detectable in the periphery of the tumour. Given these distribution characteristics and their well-documented safety profile, ALVAC- or NYVAC-based recombinants expressing feline or human IL2, respectively, were administered to domestic cats, in order to prevent the recurrence of spontaneous fibrosarcomas. In the absence of immunotherapy, tumour recurrence was observed in 61% of animals within a 12-month follow-up period after treatment with surgery and iridium-based radiotherapy. In contrast, only 39 and 28% of cats receiving either NYVAC-human IL2 or ALVAC-feline IL2, respectively, exhibited tumour recurrences. Based on such results, and in the context of ongoing clinical studies conducted in humans, we discuss the utilization of ALVAC- or NYVAC-based recombinants as viable therapeutic modalities for local immunotherapy or therapeutic vaccination against cancer, both in humans and companion animals.
- Published
- 2003
- Full Text
- View/download PDF
23. Modeling a Safer Smallpox Vaccination Regimen, for Human Immunodeficiency Virus Type 1–Infected Patients, in Immunocompromised Macaques
- Author
-
Genoveffa Franchini, Vanessa M. Hirsch, Derrick Martin, James G. McNally, Jim Tartaglia, Jay A. Berzofsky, Phil Markham, Mike Bray, Marcin Moniuszko, Yvette Edghill-Smith, Hana Golding, Tatiana S. Karpova, Wen-Po Tsai, Marsha J. Sowers, Elzbieta Tryniszewska, Jody Manischewitz, Mark G. Lewis, Igor M. Belyakov, David Venzon, Lisa R. King, Steven J. Snodgrass, Bernard Moss, Janos Nacsa, and John Parrish
- Subjects
viruses ,Simian Acquired Immunodeficiency Syndrome ,HIV Infections ,Vaccinia virus ,Antibodies, Viral ,Vaccines, Attenuated ,medicine.disease_cause ,complex mixtures ,Macaque ,Virus ,Dryvax ,Immunocompromised Host ,chemistry.chemical_compound ,Progressive vaccinia ,biology.animal ,medicine ,Animals ,Humans ,Immunology and Allergy ,Smallpox vaccine ,Immunization Schedule ,Skin ,biology ,Vaccination ,Viral Vaccines ,Simian immunodeficiency virus ,medicine.disease ,Macaca mulatta ,Virology ,Disease Models, Animal ,Infectious Diseases ,chemistry ,Immunology ,HIV-1 ,Simian Immunodeficiency Virus ,Vaccinia ,Smallpox Vaccine ,Smallpox - Abstract
We have modeled smallpox vaccination with Dryvax (Wyeth) in rhesus macaques that had depletion of CD4(+) T cells induced by infection with simian immunodeficiency virus or simian/human immunodeficiency virus. Smallpox vaccination induced significantly larger skin lesions in immunocompromised macaques than in healthy macaques. Unexpectedly, "progressive vaccinia" was infrequent. Vaccination of immunocompromised macaques with the genetically-engineered, replication-deficient poxvirus NYVAC, before or after retrovirus infection, was safe and lessened the severity of Dryvax-induced skin lesions. Neutralizing antibodies to vaccinia were induced by NYVAC, even in macaques with severe CD4(+) T cell depletion, and their titers inversely correlated with the time to complete resolution of the skin lesions. Together, these results provide the proof of concept, in macaque models that mirror human immunodeficiency virus type 1 infection, that a prime-boost approach with a highly attenuated poxvirus followed by Dryvax increases the safety of smallpox vaccination, and they highlight the importance of neutralizing antibodies in protection against virulent poxvirus.
- Published
- 2003
- Full Text
- View/download PDF
24. Both Mucosal and Systemic Routes of Immunization with the Live, Attenuated NYVAC/Simian Immunodeficiency Virus SIVgpeRecombinant Vaccine Result in Gag-Specific CD8+T-Cell Responses in Mucosal Tissues of Macaques
- Author
-
Genoveffa Franchini, Brian L. Kelsall, Andrew A. Lackner, Liljana Stevceva, Jim Tartaglia, Elzbieta Tryniszewska, Xavier Alvarez, Warren Strober, and Janos Nacsa
- Subjects
viruses ,animal diseases ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Gene Products, gag ,Vaccinia virus ,CD8-Positive T-Lymphocytes ,Vaccines, Attenuated ,medicine.disease_cause ,Injections, Intramuscular ,Microbiology ,Macaque ,Immune system ,Administration, Rectal ,Virology ,biology.animal ,medicine ,Animals ,Cytotoxic T cell ,Immunity, Mucosal ,Administration, Intranasal ,Vaccines, Synthetic ,biology ,Immunodominant Epitopes ,SAIDS Vaccines ,virus diseases ,Viral Vaccines ,Simian immunodeficiency virus ,Vaccination ,Insect Science ,Pathogenesis and Immunity ,Macaca ,Live vector vaccine ,Immunization ,Simian Immunodeficiency Virus ,Nasal administration ,CD8 - Abstract
As most human immunodeficiency virus (HIV) infection occurs via mucosal surfaces, an important goal of vaccination may be the induction of virus-specific immune responses at mucosal sites to contain viral infection early on. Here we designed a study in macaques carrying the major histocompatibility complex class I Mamu-A∗01 molecule to assess the capacity of the highly attenuated poxvirus NYVAC/simian immunodeficiency virus (SIV) SIVgpevaccine candidate administered by the intranasal, intramuscular, or intrarectal route to induce mucosal immunity. All macaques, including one naive macaque, were exposed to SIVmac251by the intrarectal route and sacrificed 48 h after infection. The kinetics of immune response at various time points following immunization with NYVAC/SIVgpeand the anamnestic response to SIVmac251at 48 h after challenge were assessed in blood, in serial rectal and vaginal biopsy samples, and in tissues at euthanasia with an SIVmacGag-specific tetramer. In addition, at euthanasia, antigen-specific cells producing gamma interferon or tumor necrosis factor alpha from the jejunum lamina propria were quantified in all macaques. Surprisingly, antigen-specific CD8+T cells were found in the mucosal tissues of all immunized macaques regardless of whether the vaccine was administered by a mucosal route (intranasal or intrarectal) or systemically. In addition, following mucosal SIVmac251challenge, antigen-specific responses were mainly confined to mucosal tissues, again regardless of the route of immunization. We conclude that immunization with a live vector vaccine results in the appearance of CD8+T-cell responses at mucosal sites even when the vaccine is delivered by nonmucosal routes.
- Published
- 2002
- Full Text
- View/download PDF
25. Vaccination of Macaques with Long-Standing SIVmac251 Infection Lowers the Viral Set Point After Cessation of Antiretroviral Therapy
- Author
-
Zdeněk Hel, David Venzon, Genoveffa Franchini, Elzbieta Tryniszewska, Robyn Washington Parks, Mark G. Lewis, Jim Tartaglia, David C. Montefiori, Marcin Moniuszko, Kendall A. Smith, Peter Silvera, and Janos Nacsa
- Subjects
CD4-Positive T-Lymphocytes ,viruses ,T cell ,Immunology ,CD4-CD8 Ratio ,Simian Acquired Immunodeficiency Syndrome ,Epitopes, T-Lymphocyte ,CD8-Positive T-Lymphocytes ,Biology ,Virus Replication ,Antiviral Agents ,Epitope ,Pharmacotherapy ,medicine ,Animals ,Immunology and Allergy ,Viremia ,Vector (molecular biology) ,Immunization Schedule ,SAIDS Vaccines ,Macaca mulatta ,Fusion protein ,Virology ,Vaccination ,medicine.anatomical_structure ,Interleukin-2 ,Drug Therapy, Combination ,Simian Immunodeficiency Virus ,CD8 - Abstract
A cohort of rhesus macaques with long-standing SIVmac251 infection (≥5 mo) was treated with continuous antiretroviral therapy (ART). A group of eight macaques was vaccinated with or without simultaneous administration of low dose IL-2 with the highly attenuated poxvirus vector (NYVAC) vaccine candidate expressing the SIVmac structural gag-pol-env (gpe) genes and a novel chimeric fusion protein derived from the rev-tat-nef (rtn) regulatory genes. Control groups consisted of mock-vaccinated macaques or animals treated only with IL-2. Vaccination significantly expanded both virus-specific CD4+ and CD8+ T cell responses, and IL-2 further increased the vaccine-induced response to an immunodominant Gag epitope. Following antiretroviral treatment interruption, the viral set point was significantly lower in vaccinated than in control macaques for at least 4 consecutive mo, and viral containment was inversely correlated with vaccine-induced, virus-specific CD4+ and CD8+ T cell responses. These data provide the proof of concept that therapeutic vaccination before cessation of ART may be a feasible approach in the clinical management of HIV-1 infection.
- Published
- 2002
- Full Text
- View/download PDF
26. Design and In Vivo Immunogenicity of a Polyvalent Vaccine Based on SIVmac Regulatory Genes
- Author
-
Tatiana S. Karpova, Genoveffa Franchini, Julie M. Johnson, John D. Altman, James G. McNally, Jim Tartaglia, V.S. Kalyanaraman, Zdeněk Hel, Robert Harrod, Wen-Po Tsai, Elzbieta Tryniszewska, Jake Fullen, and Barbara K. Felber
- Subjects
Recombinant Fusion Proteins ,animal diseases ,viruses ,Gene Products, gag ,Regulatory Sequences, Nucleic Acid ,Biology ,medicine.disease_cause ,Viral life cycle ,Genetics ,medicine ,Animals ,Humans ,Viral Regulatory and Accessory Proteins ,Molecular Biology ,Regulator gene ,Vaccines ,Vaccines, Synthetic ,Polyvalent Vaccine ,Immunogenicity ,virus diseases ,Cell Biology ,General Medicine ,Simian immunodeficiency virus ,Macaca mulatta ,Fusion protein ,Virology ,Genes, nef ,Genes, rev ,Viral Regulatory Proteins ,Genes, tat ,Regulatory sequence ,Simian Immunodeficiency Virus ,HeLa Cells - Abstract
Most vaccine modalities for human immunodeficiency virus type 1 (HIV-1) tested for immunogenicity and efficacy in the SIVmac (simian immunodeficiency virus) macaque model do not include the viral regulatory proteins. Because viral regulatory proteins are expressed early during the virus life cycle and represent an additional source of antigens, their inclusion as a vaccine component may increase the overall virus-specific immune response in vaccinees. However, at least two of the early proteins, Tat and Nef, may be immunosuppressive, limiting their usefulness as components of an SIV vaccine. We have constructed a polyvalent chimeric protein in which the open reading frames for Tat and Nef have been reassorted and the nuclear localization sequence for Tat and Rev and the myristoylation site for Nef have been removed. The resulting DNA plasmid (pDNA-SIV-Retanef) (pDNA-SIV-RTN) encodes a protein of 55 kDa (Retanef) that localizes at the steady state in the cytoplasma of transfected cells. Both the DNA-SIV-RTN and the highly attenuated recombinant poxvirus vector NYVAC-SIV-RTN were demonstrated to be immunogenic in SIVmac251-infected macaques treated with ART as well as in naive macaques. An equivalent strategy may be used for the generation of polyvalent antigens encoding the regulatory proteins in a HIV-1 vaccine candidate.
- Published
- 2002
- Full Text
- View/download PDF
27. Therapeutic vaccines against melanoma and colorectal cancer
- Author
-
Neil L. Berinstein, Brian H. Barber, Marie-Claude Bonnet, Jim Tartaglia, Michel Klein, and Philippe Moingeon
- Subjects
Cellular immunity ,Colorectal cancer ,T-Lymphocytes ,medicine.medical_treatment ,Genetic Vectors ,Canarypox virus ,Cancer Vaccines ,Avipoxvirus ,Antigen ,Antigens, Neoplasm ,Humans ,Medicine ,Melanoma ,Clinical Trials as Topic ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Public Health, Environmental and Occupational Health ,Cancer ,Immunotherapy ,medicine.disease ,Infectious Diseases ,Immunization ,Immunology ,Cancer research ,Cytokines ,Molecular Medicine ,Safety ,Colorectal Neoplasms ,business - Abstract
Our overall strategy is to develop multivalent recombinant vaccines capable of eliciting broad immune responses in patients with malignant melanoma or colorectal cancer. We report herein results from initial studies conducted in cancer patients to evaluate the effect of intratumoral administration of recombinant canarypox viruses carrying cytokine genes. Our current focus is on the induction of tumor-specific T-cell responses using a prime/boost immunization schedule with a unique vector system derived from the canary pox virus called ALVAC, in which we incorporate genes encoding Tumor Associated Antigens (TAAs) of interest. Clinical studies in colorectal cancer evaluating an ALVAC CEA candidate vaccine have shown that this approach is safe and can induce tumor-specific T cell responses. Additional clinical studies evaluating candidate vaccines against melanoma and colorectal cancer, targeting either the gp100, Mage 1, Mage 3 or p53 molecules are ongoing.
- Published
- 2001
- Full Text
- View/download PDF
28. Replication-Defective Canarypox (ALVAC) Vectors Effectively Activate Anti–Human Immunodeficiency Virus-1 Cytotoxic T Lymphocytes Present in Infected Patients: Implications for Antigen-Specific Immunotherapy
- Author
-
Jim Tartaglia, Guido Ferrari, Kent J. Weinhold, C. Berend, D. Moody, W.I. Cox, John Bartlett, J. F. Toso, Janet Ottinger, R. Dodge, and Enzo Paoletti
- Subjects
Canarypox ,Lymphocyte ,Immunology ,Cell Biology ,Hematology ,Biology ,biology.organism_classification ,Biochemistry ,Virology ,Defective virus ,CTL ,medicine.anatomical_structure ,Antigen ,Aldesleukin ,medicine ,Cytotoxic T cell ,CD8 - Abstract
In the attempt to develop immunotherapeutic strategies for acquired immunodeficiency syndrome capable of activating effector cells in an antigen-specific manner while maintaining the broadest possible T-cell repertoire, we evaluated two canarypox (ALVAC)-based vectors for their capacity to induce ex vivo activation/expansion of human immunodeficiency virus (HIV)-specific CD8+ cytotoxic lymphocyte precursors (CTLp) obtained from HIV-1–infected donors. These two vectors, vCP205 encoding HIV-1 gp120 + TM (28 amino acid transmembrane anchor sequence) in addition to Gag/protease and vCP300 encoding gp120 + Gag/protease as well as Nef and Pol CTL determinants, are pancytotropic but replication incompetent in mammalian cells. Bulk peripheral blood mononuclear cells (PBMCs) or enriched CD8+ T cells were stimulated for 10 days with autologous ALVAC-infected PBMCs in the presence of different cytokine combinations (interleukin-2 [IL-2], IL-4, IL-7, and IL-12). Activation by ALVAC constructs was highly antigen-specific, because vCP205 elicited only Env and Gag CTL, whereas vCP300 elicited broader reactivities against Env, Gag, Pol, and Nef determinants. The ALVAC activation of CTLp was IL-2 dependent and enhanced by the addition of IL-7, whereas IL-4 and IL-12 failed to augment cytotoxic reactivities elicited by these constructs. The expansion of enriched CD8+ T cells after activation with vCP300 was higher in patients with CD4 counts greater than 400 cells/μL. Two rounds of in vitro stimulation (IVS) with vCP300 resulted in nearly an eightfold expansion of CD8+ lymphocytes over a 25-day period. After the second IVS, an average 3.2-fold increase among the different antigen-specific CTL frequencies was achieved. These studies clearly show that HIV-recombinant ALVAC vectors represent powerful polyvalent antigenic stimuli for activation and expansion of the CD8 lymphocyte response that occurs as a result of HIV infection.
- Published
- 1997
- Full Text
- View/download PDF
29. Erratum: Corrigendum: Adjuvant-dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition
- Author
-
Maria Grazia Ferrari, Vaniambadi S. Kalyanaraman, Zhong-Min Ma, Brandon F. Keele, Mangala Rao, Genoveffa Franchini, Francesca Caccuri, Amy W. Chung, Mark J. Cameron, Chris Bailey-Kellogg, Margaret E. Ackerman, Adrian B. McDermott, Jim Tartaglia, Guido Ferrari, Richard A. Koup, Silvia Ratto-Kim, Stephen Whitney, Rafick-Pierre Sekaly, Mitzi M. Donaldson, Massimiliano Bissa, Diego A. Vargas-Inchaustegui, Georgia D. Tomaras, Nelson L. Michael, Christopher J. Miller, DeVon Thompson, David S Quinn, Luca Schifanella, Mario Roederer, Jerome H. Kim, Matthew Blackburn, Tran B. Phan, Melvin N. Doster, Donald Stablein, David Venzon, Slim Fourati, Kathryn E. Foulds, Susan W. Barnett, Monica Vaccari, Karin Loré, Donald N. Forthal, Frank Liang, David C. Montefiori, Poonam Pegu, Shari N. Gordon, Sanjay Phogat, Xiaoying Shen, Marjorie Robert-Guroff, Eric P. Brown, Karen G Dowell, Namal P.M. Liyanage, Nicolo Binello, Galit Alter, and Erik Billings
- Subjects
0301 basic medicine ,viruses ,medicine.medical_treatment ,virus diseases ,General Medicine ,Computational biology ,Biology ,complex mixtures ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Nat ,medicine ,Adjuvant - Abstract
A recombinant vaccine containing Aventis Pasteur’s canarypox vector (ALVAC)–HIV and gp120 alum decreased the risk of HIV acquisition in the RV144 vaccine trial. The substitution of alum with the more immunogenic MF59 adjuvant is under consideration for the next efficacy human trial. We found here that an ALVAC–simian immunodeficiency virus (SIV) and gp120 alum (ALVAC–SIV + gp120) equivalent vaccine, but not an ALVAC–SIV + gp120 MF59 vaccine, was efficacious in delaying the onset of SIVmac251 in rhesus macaques, despite the higher immunogenicity of the latter adjuvant. Vaccine efficacy was associated with alum-induced, but not with MF59-induced, envelope (Env)-dependent mucosal innate lymphoid cells (ILCs) that produce interleukin (IL)-17, as well as with mucosal IgG to the gp120 variable region 2 (V2) and the expression of 12 genes, ten of which are part of the RAS pathway. The association between RAS activation and vaccine efficacy was also observed in an independent efficacious SIV-vaccine approach. Whether RAS activation, mucosal ILCs and antibodies to V2 are also important hallmarks of HIV-vaccine efficacy in humans will require further studies.
- Published
- 2016
- Full Text
- View/download PDF
30. The Thai Phase III HIV Type 1 Vaccine trial (RV144) regimen induces antibodies that target conserved regions within the V2 loop of gp120
- Author
-
Nicos, Karasavvas, Erik, Billings, Mangala, Rao, Constance, Williams, Susan, Zolla-Pazner, Robert T, Bailer, Richard A, Koup, Sirinan, Madnote, Duangnapa, Arworn, Xiaoying, Shen, Georgia D, Tomaras, Jeffrey R, Currier, Mike, Jiang, Craig, Magaret, Charla, Andrews, Raphael, Gottardo, Peter, Gilbert, Timothy J, Cardozo, Supachai, Rerks-Ngarm, Sorachai, Nitayaphan, Punnee, Pitisuttithum, Jaranit, Kaewkungwal, Robert, Paris, Kelli, Greene, Hongmei, Gao, Sanjay, Gurunathan, Jim, Tartaglia, Faruk, Sinangil, Bette T, Korber, David C, Montefiori, John R, Mascola, Merlin L, Robb, Barton F, Haynes, Viseth, Ngauy, Nelson L, Michael, Jerome H, Kim, Mark S, de Souza, and Patricia, Morgan
- Subjects
Male ,Immunology ,Molecular Sequence Data ,Protein Array Analysis ,Epitopes, T-Lymphocyte ,Enzyme-Linked Immunosorbent Assay ,HIV Infections ,Biology ,HIV Antibodies ,HIV Envelope Protein gp120 ,Cell Line ,Virology ,Humans ,Amino Acid Sequence ,Peptide sequence ,AIDS Vaccines ,Vaccines ,Vaccine trial ,Vaccination ,Loop (topology) ,Regimen ,Infectious Diseases ,AIDSVAX ,biology.protein ,HIV-1 ,Female ,Peptide microarray ,Antibody - Abstract
The Thai Phase III clinical trial (RV144) showed modest efficacy in preventing HIV-1 acquisition. Plasma collected from HIV-1-uninfected trial participants completing all injections with ALVAC-HIV (vCP1521) prime and AIDSVAX B/E boost were tested for antibody responses against HIV-1 gp120 envelope (Env). Peptide microarray analysis from six HIV-1 subtypes and group M consensus showed that vaccination induced antibody responses to the second variable (V2) loop of gp120 of multiple subtypes. We further evaluated V2 responses by ELISA and surface plasmon resonance using cyclic (Cyc) and linear V2 loop peptides. Thirty-one of 32 vaccine recipients tested (97%) had antibody responses against Cyc V2 at 2 weeks postimmunization with a reciprocal geometric mean titer (GMT) of 1100 (range: 200–3200). The frequency of detecting plasma V2 antibodies declined to 19% at 28 weeks post-last injection (GMT: 110, range: 100–200). Antibody responses targeted the mid-region of the V2 loop that contains conserved epitopes and has the amino acid sequence KQKVHALFYKLDIVPI (HXB2 Numbering sequence 169–184). Valine at position 172 was critical for antibody binding. The frequency of V3 responses at 2 weeks postimmunization was modest (18/32, 56%) with a GMT of 185 (range: 100–800). In contrast, naturally infected HIV-1 individuals had a lower frequency of antibody responses to V2 (10/20, 50%; p=0.003) and a higher frequency of responses to V3 (19/20, 95%), with GMTs of 400 (range: 100–3200) and 3570 (range: 200–12,800), respectively. RV144 vaccination induced antibodies that targeted a region of the V2 loop that contains conserved epitopes. Early HIV-1 transmission events involve V2 loop interactions, raising the possibility that anti-V2 antibodies in RV144 may have contributed to viral inhibition.
- Published
- 2012
31. Design of an HIV Env antigen that binds with high affinity to antibodies against linear, conformational and broadly neutralizing epitopes within V1/V2
- Author
-
S Crawford, John R. Mascola, Punnee Pitisuttithum, R Park, Jaranit Kaewkungwal, Mattia Bonsignori, Melissa Cooper, Anthony M. Moody, Nicos Karasavvas, Kwan-Ki Hwang, Peter D. Kwong, Xusheng Lü, Jim Tartaglia, Gary J. Nabel, Nelson L. Michael, Kaifan Dai, Haiyan Chen, Susan Zolla-Pazner, R De, Sodsai Tovanabutra, Abraham Pinter, Jerome H. Kim, Zhi-Yong Yang, Georgia D. Tomaras, Thomas Lee Jeffries, Faruk Sinangil, Larry Liao, Marie Pancera, Sorachai Nitayaphan, Alam, Supachai Rerks-Ngarm, and Barton F. Haynes
- Subjects
lcsh:Immunologic diseases. Allergy ,biology ,Chemistry ,Human immunodeficiency virus (HIV) ,virus diseases ,medicine.disease_cause ,Virology ,Neutralizing epitope ,Protein structure ,Infectious Diseases ,Antigen ,biology.protein ,medicine ,Oral Presentation ,Antibody ,lcsh:RC581-607 - Abstract
Design of an HIV Env antigen that binds with high affinity to antibodies against linear, conformational and broadly neutralizing epitopes within V1/V2 L Liao, M Bonsignori, K Hwang, AM Moody, R Park, S Crawford, H Chen, TL Jeffries, M Cooper, X Lu, R De, N Karasavvas, S Rerks-Ngarm, S Nitayaphan, J Kaewkungwal, S Tovanabutra, P Pitisuttithum, J Tartaglia, F Sinangil, J Kim, NL Michael, GD Tomaras, Z Yang, K Dai, M Pancera, GJ Nabel, JR Mascola, PD Kwong, A Pinter, S Zolla-Pazner, MS Alam, BF Haynes
- Published
- 2012
32. Comparison of the depth of vaccine-elicited HIV-1 Env epitope-specific CD8+ T lymphocyte responses
- Author
-
Sandrine L. Hulot, Sampa Santra, Giuseppe Pantaleo, Jim Tartaglia, Carmen E. Gómez, Bertram L. Jacobs, Beatriz Perdiguero, Norman L. Letvin, Barton F. Haynes, Seaman, Bette T. Korber, and Mariano Esteban
- Subjects
lcsh:Immunologic diseases. Allergy ,Immunogen ,biology ,T-cell receptor ,chemical and pharmacologic phenomena ,Carboxyfluorescein succinimidyl ester ,T lymphocyte ,Virology ,Epitope ,chemistry.chemical_compound ,Infectious Diseases ,chemistry ,Poster Presentation ,MHC class I ,biology.protein ,Avidity ,lcsh:RC581-607 ,CD8 - Abstract
Background One of the major challenges in the development of an effective HIV-1 vaccine is the extraordinary genetic diversity of the virus. Immunizations of nonhuman primates using consensus and mosaic immunogens have been shown to elicit cross-reactive CD8+T lymphocyte responses that increase the depth of epitope recognition. However, one of the limitations of vaccine-induced epitope-specific CD8+T lymphocytes includes lack of protection against diverse strains and emergent forms of HIV-1 due to altered T cell receptor (TCR) affinity for variant peptide:MHC class I complexes. Methods In this study, we immunized a cohort of fifteen MamuA*01+ rhesus monkeys with either a 3-valent mosaic Env, group M consensus Env, or single clade B Env vaccine and compared the ability of the CD8+T lymphocyte populations elicited by each immunogen to recognize variants of an HIV-1 envelope epitope sequence p41A (YI9). We identified vaccine-induced CD8+T lymphocytes populations using tetramers constructed with 9 variants of p41A epitope. We assessed the ability of those variant peptides to activate CTL by measuring cytokine production and CD107a expression. We evaluated proliferation using carboxyfluorescein succinimidyl ester. Finally, we investigated the functional avidity of these CD8+T lymphocytes for the variant peptide:Mamu-A*01 complexes using surface plasmon resonance technology. Results Our data show that Env immunizations can generate cross-reactive CD8+T lymphocytes that recognize 2 of 9 (22%) of the variants of p41A epitope, with higher responses induced by the consensus and the 3-valent mosaic immunogens (variant from clade C and variant from clade A/E) compared to the single clade Env immunogen. Tetramer-binding data also show that CD8+ T lymphocytes from monkeys immunized with mosaic immunogen have a trend of higher binding to majority of the variant peptides tested.
- Published
- 2012
33. Antibodies to the envelope protein protect macaques from SIVmac251 acquisition in an immunization regimen that mimics the RV-144 Thai trial
- Author
-
Guido Ferrari, Ranajit Pal, Stephen Whitney, Y Guan, Melvin N. Doster, Brandon F. Keele, Lauren Hudacik, David C. Montefiori, Shari N. Gordon, Mangala Rao, Donald Stablein, E Billings, Nelson L. Michael, Jerome H. Kim, David Venzon, Jeffrey D. Lifson, Maria Grazia Ferrari, Claudio Fenizia, Jim Tartaglia, Genoveffa Franchini, and Poonam Pegu
- Subjects
biology ,business.industry ,virus diseases ,Canarypox Vector ,Virology ,Virus ,Regimen ,Infectious Diseases ,Immunization ,Immunology ,biology.protein ,Oral Presentation ,Medicine ,Antibody ,Hiv transmission ,business ,RV 144 ,CD8 - Abstract
Background The canarypox vector ALVAC-HIV, together with the HIV gp120 envelope, has protected 31.2% of Thai heterosexual individuals from HIV acquisition in the RV144 HIV vaccine trial. This outcome was unexpected, given the limited ability of the ALVAC-HIV vaccine component to induce CD8+T-cell responses, and of the HIVgp120 envelope to elicit broad neutralizing antibodies. Methods We vaccinated macaques with an immunization regimen that mimics the RV144 trial and exposed them to a mucosal dose of SIVmac251 that transmits few virus variants, similar to HIV transmission to humans. Results
- Published
- 2012
- Full Text
- View/download PDF
34. Extended evaluation of the virologic, immunologic, and clinical course of volunteers who acquired HIV-1 infection in a phase III vaccine trial of ALVAC-HIV and AIDSVAX B/E
- Author
-
Chirapa Eamsila, Jorge Flores, Donald P. Francis, Rapee Trichavaroj, Nampueng Churikanont, Chawetsan Namwat, Robert Paris, Sorachai Nitayaphan, Jaranit Kaewkungkal, Elizabeth Adams, Jim Tartaglia, Peter B. Gilbert, Merlin L. Robb, Mark de Souza, Chureeratana Bowonwatanuwong, Prasert Thongcharoen, Viseth Ngauy, Sanjay Gurunathan, Charla Andrews, Jerome H. Kim, Supamit Chunsutthiwat, Prayura Kunasol, Nakorn Premsri, Shuying S. Li, Nelson L. Michael, Robert J. O'Connell, and Supachai Rerks-Ngarm
- Subjects
Adult ,Male ,medicine.medical_specialty ,Time Factors ,HIV Infections ,Young Adult ,Risk-Taking ,Semen ,Internal medicine ,Antiretroviral Therapy, Highly Active ,Immunology and Allergy ,Medicine ,Humans ,Prospective Studies ,Prospective cohort study ,AIDS Vaccines ,business.industry ,Surrogate endpoint ,Vaccination ,Vaccine trial ,Viral Vaccines ,Viral Load ,Vaccine efficacy ,Thailand ,CD4 Lymphocyte Count ,Clinical trial ,Infectious Diseases ,AIDSVAX ,Immunology ,Vagina ,Disease Progression ,HIV-1 ,Linear Models ,Female ,business ,Viral load ,Follow-Up Studies - Abstract
Background The Thai Phase III Trial of ALVAC-HIV and AIDSVAX B/E showed an estimated vaccine efficacy (VE) of 31% to prevent acquisition of human immunodeficiency virus (HIV). Here we evaluated the effect of vaccination on disease progression after infection. Methods CD4(+) T-cell counts and HIV viral load (VL) were measured serially. The primary analysis evaluated vaccine efficacy (VEP) as the percent reduction (vaccine vs placebo) in cumulative probability of a primary composite endpoint of clinical and CD4(+) count components at prespecified time points after infection. Secondary analyses of biomarker-based endpoints were assessed using marginal mean and linear mixed models. Results There were 61 endpoints in the modified intent-to-treat cohort (mITT; n = 114). There was no evidence for efficacy at 30, 42, 54, and 60 months in the mITT and per protocol (n = 90) cohorts. Estimated VEP (mITT) was15.8% (-21.9, 41.8) at 60 months postinfection. There was weak evidence of lower VL and higher CD4(+) count at 60 and 66 months in the vaccine group. Lower mucosal VL was observed among vaccine recipients, primarily in semen (P = .04). Conclusions Vaccination did not affect the clinical course of HIV disease after infection. A potential vaccine effect on the genital mucosa warrants further study.
- Published
- 2012
35. Risk behaviour and time as covariates for efficacy of the HIV vaccine regimen ALVAC-HIV (vCP1521) and AIDSVAX B/E: a post-hoc analysis of the Thai phase 3 efficacy trial RV 144
- Author
-
Michael Benenson, Sanjay Gurunathan, Don Stablein, Supachai Rerks-Ngarm, Punnee Pitisuttithum, Prasert Thongcharoen, Jerome H. Kim, Sorachai Nitayaphan, Elizabeth Adams, Merlin L. Robb, Chirasak Khamboonruang, Jim Tartaglia, Peter B. Gilbert, Jaranit Kaewkungwal, Patricia Morgan, Donald P. Francis, Joseph Chiu, Prayura Kunasol, Nelson L. Michael, and Robert Paris
- Subjects
Adult ,Male ,medicine.medical_specialty ,Time Factors ,Adolescent ,HIV Infections ,Kaplan-Meier Estimate ,Statistics, Nonparametric ,law.invention ,Young Adult ,Risk-Taking ,Acquired immunodeficiency syndrome (AIDS) ,Randomized controlled trial ,law ,Internal medicine ,Surveys and Questionnaires ,Medicine ,Humans ,HIV vaccine ,Homosexuality, Male ,RV 144 ,Substance Abuse, Intravenous ,AIDS Vaccines ,business.industry ,Viral Load ,Vaccine efficacy ,medicine.disease ,Thailand ,Vaccination ,Infectious Diseases ,AIDSVAX ,Immunology ,Female ,business ,Viral load - Abstract
Summary Background The Thai phase 3 HIV vaccine trial RV 144 showed modest efficacy of a vaccine against HIV acquisition. Baseline variables of age, sex, marital status, and risk did not modify vaccine efficacy. We did a post-hoc analysis of the trial's data to investigate behavioural risk and efficacy every 6 months after vaccination. Methods RV 144 was a randomised, multicentre, double-blind, placebo-controlled efficacy trial testing the combination of the HIV vaccines ALVAC-HIV (vCP1521) and AIDSVAX B/E to prevent HIV infection or reduce setpoint viral load. Male and female volunteers aged 18–30 years were recruited from the community. In this post-hoc analysis of the modified intention-to-treat population (16 395 participants), HIV risk behaviour was assessed with a self-administered questionnaire at the time of initial vaccination in the trial and every 6 months thereafter for 3 years. We classified participants' behaviour as low, medium, or high risk. Both the acquisition endpoint and the early viral-load endpoint were examined for interactions with risk status over time and temporal effects after vaccination. Multiple proportional hazards regression models with treatment and time-varying risk covariates were analysed. Findings Risk of acquisition of HIV was low in each risk group, but 9187 (58·2%) participants reported higher-risk behaviour at least once during the study. Participants classified as high or increasing risk at least once during follow-up were compared with those who maintained low-risk or medium-risk behaviour as a time-varying covariate, and the interaction of risk status and acquisition efficacy was significant (p=0·01), with greater benefit in low-risk individuals. Vaccine efficacy seemed to peak early—cumulative vaccine efficacy was estimated to be 60·5% (95% CI 22–80) through the 12 months after initial vaccination—and declined quickly. Vaccination did not seem to affect viral load in either early or late infections. Interpretation Future HIV vaccine trials should recognise potential interactions between challenge intensity and risk heterogeneity in both population and treatment effects. The regimen tested in the RV 144 phase 3 trial might benefit from extended immunisation schedules. Funding US Army Medical Research and Materiel Command and Division of AIDS, National Institute of Allergy and Infectious Disease, National Institutes of Health.
- Published
- 2012
36. Poxvirus vector-based HIV vaccines
- Author
-
Jim Tartaglia, Bertram L. Jacobs, Giuseppe Pantaleo, and Mariano Esteban
- Subjects
viruses ,Immunology ,Genetic Vectors ,Human immunodeficiency virus (HIV) ,HIV Infections ,Vaccinia virus ,HIV Envelope Protein gp120 ,medicine.disease_cause ,Immune system ,Virology ,Medicine ,Humans ,Vector (molecular biology) ,HIV vaccine ,AIDS Vaccines ,Clinical Trials as Topic ,Vaccines, Synthetic ,Oncology (nursing) ,business.industry ,virus diseases ,Hematology ,biochemical phenomena, metabolism, and nutrition ,Thailand ,Vaccination ,Infectious Diseases ,Oncology ,Immunization ,HIV-1 ,business ,Synthetic immunology - Abstract
In this review, we will provide the scientific rationale for the use of poxvirus vectors in the field of HIV vaccines, the immunological profile of the vaccine-induced immune responses, an update on the current use of poxvirus vector-based vaccines in HIV vaccine clinical trials, and the development of new modified poxvirus vectors with improved immunological profile.An Ad5-HIV vaccine was tested in a phase IIb clinical trial (known as the Step trial). Vaccinations in the Step trial were discontinued because the vaccine did not show any effect on acquisition of infection and on viral load. After the disappointing failure of the Step trial, the field of HIV vaccine has regained enthusiasm and vigour due to the promising protective effect observed in the phase III efficacy trial (known as RV-144) performed in Thailand which has tested a poxvirus-gp120 combination.The RV-144 phase III has provided for the first time evidence that an HIV vaccine can prevent HIV infection. The results from the RV-144 trial are providing the scientific rationale for the future development of the HIV vaccine field and for designing future efficacy trials.
- Published
- 2010
37. Improved innate and adaptive immunostimulation by genetically modified HIV-1 protein expressing NYVAC vectors
- Author
-
Giuseppe Pantaleo, Esther D. Quakkelaar, Bertram L. Jacobs, Carmen E. Gómez, Paul P. Heinen, Nikki M. Loof, Elias K. Haddad, Thomas Duhen, Mariano Esteban, Abdelali Filali-Mouhim, Karen V. Kibler, Antonio Lanzavecchia, Alexandre Harari, David M. Koelle, Anke Redeker, Beatriz Perdiguero, Stella Mayo McCaughey, Federica Sallusto, Jim Tartaglia, Ferry Ossendorp, Jean Philippe Goulet, Rafick Pierre Sekaly, and Cornelis J. M. Melief
- Subjects
Viral Diseases ,Anatomy and Physiology ,viruses ,T-Lymphocytes ,Gene Expression ,Antigen Processing and Recognition ,Adaptive Immunity ,gag Gene Products, Human Immunodeficiency Virus ,Interferon ,Cricetinae ,Immune Physiology ,Cytotoxic T cell ,Immune Response ,Antigen Presentation ,Multidisciplinary ,Antigen processing ,T Cells ,Immunogenicity ,Vaccination ,virus diseases ,Acquired immune system ,Infectious Diseases ,Medicine ,Genetic Engineering ,medicine.drug ,Signal Transduction ,Research Article ,Science ,Immune Cells ,Antigen presentation ,Genetic Vectors ,Immunology ,Antigen-Presenting Cells ,Biology ,Cross Reactions ,Microbiology ,Viral Proteins ,Antigen ,Virology ,Vaccine Development ,medicine ,Animals ,Humans ,Antigens ,Cell Proliferation ,Poxviridae ,Immunity ,Interferon-alpha ,HIV ,Viral Vaccines ,Dendritic Cells ,Immunity, Innate ,Viral replication ,Gene Expression Regulation ,HIV-1 ,Immunization ,Clinical Immunology ,B7-2 Antigen ,t-cell responses immunodeficiency virus-infection broad species-specificity phase-i trial vaccinia virus immune-responses dendritic cells clade-c immunogenicity candidates ,Gene Deletion ,HeLa Cells - Abstract
Attenuated poxviruses are safe and capable of expressing foreign antigens. Poxviruses are applied in veterinary vaccination and explored as candidate vaccines for humans. However, poxviruses express multiple genes encoding proteins that interfere with components of the innate and adaptive immune response. This manuscript describes two strategies aimed to improve the immunogenicity of the highly attenuated, host-range restricted poxvirus NYVAC: deletion of the viral gene encoding type-I interferon-binding protein and development of attenuated replication-competent NYVAC. We evaluated these newly generated NYVAC mutants, encoding HIV-1 env, gag, pol and nef, for their ability to stimulate HIV-specific CD8 T-cell responses in vitro from blood mononuclear cells of HIV-infected subjects. The new vectors were evaluated and compared to the parental NYVAC vector in dendritic cells (DCs), RNA expression arrays, HIV gag expression and crosspresentation assays in vitro. Deletion of type-I interferon-binding protein enhanced expression of interferon and interferoninduced genes in DCs, and increased maturation of infected DCs. Restoration of replication competence induced activationof pathways involving antigen processing and presentation. Also, replication-competent NYVAC showed increased Gag expression in infected cells, permitting enhanced cross-presentation to HIV-specific CD8 T cells and proliferation of HIVspecific memory CD8 T-cells in vitro. The recombinant NYVAC combining both modifications induced interferon-induced genes and genes involved in antigen processing and presentation, as well as increased Gag expression. This combined replication-competent NYVAC is a promising candidate for the next generation of HIV vaccines.
- Published
- 2010
38. OA07-04 LB. Immunogenicity of ALVAC-HIV® (vCP1521) and AIDSVAX® B/E prime boose vaccination in RV144, the Thai Phase III HIV vaccine trial
- Author
-
Alexandra Schuetz, S Jongrakthaitae, de Souza, Jim Tartaglia, Nelson L. Michael, D Francis, Robert Paris, Len Dally, S Rerks-Ngam, Silvia Ratto-Kim, Y Phuang-ngem, S Nitayaphan, Jung-Hyun Kim, W Chuenarom, and Rapee Trichavaroj
- Subjects
lcsh:Immunologic diseases. Allergy ,biology ,business.industry ,viruses ,ELISPOT ,Immunogenicity ,virus diseases ,Peripheral blood mononuclear cell ,Virology ,Vaccination ,Infectious Diseases ,Immunization ,biology.protein ,Oral Presentation ,Cytotoxic T cell ,Medicine ,Antibody ,lcsh:RC581-607 ,business ,CD8 - Abstract
Methods A list of blinded samples from persons completing all 4 injections with either placebo or vaccine and remained HIV negative at the end of the trial was provided. Peripheral blood mononuclear cells (PBMC) or plasma were tested to CRF 01_AE and subtype B vaccine antigens in the following validated assays: (1) Interferon-gamma (IFN-γ) ELISpot; (2) IFN-γ/interleukin-2 intracellular cytokine staining (ICS); (3) Binding antibody (BAb). ELISpot and ICS assays measured responses to Env (92TH023) and Gag (LAI) peptide pools prior to and 6 months following the completion of immunization. BAb was measured using reciprocal dilution EIA to A244 and MN gp120 and BH10 p24 prior to and at 2 weeks following the completion of immunization. Results Data will be un-blinded to treatment assignment by October 2009. Analyses of post-injection responses to Env and Gag by ELISpot revealed an overall frequency of 14%, with Env responses (11%) predominating over Gag (5%). The overall frequency of ICS responses to HIV peptides in samples studied to date was 35% and was greater for CD4 (26%) than CD8 (9%) T cells, with responses to Env again predominating: 26% versus 1% Gag for CD4 and 6% Env versus 2% Gag for CD8 T cells. The frequency of BAb responses to p24 was 37% and was identical for CRF01_AE and MN gp120 (70%).
- Published
- 2009
- Full Text
- View/download PDF
39. Use of predictive markers of HIV disease progression in vaccine trials
- Author
-
Jim Tartaglia, Sanjay Gurunathan, Claude Meric, R El Habib, Stanley A. Plotkin, B. Dodet, Lynn Baglyos, and Lawrence Corey
- Subjects
Oncology ,medicine.medical_specialty ,HIV Infections ,Acquired immunodeficiency syndrome (AIDS) ,Internal medicine ,Clinical endpoint ,Medicine ,Animals ,Humans ,HIV vaccine ,AIDS Vaccines ,Clinical Trials as Topic ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Surrogate endpoint ,Public Health, Environmental and Occupational Health ,Vaccine trial ,Viral Load ,medicine.disease ,Vaccination ,Infectious Diseases ,Immunology ,Disease Progression ,HIV-1 ,Molecular Medicine ,RNA, Viral ,Viral disease ,business ,Viral load ,Biomarkers ,T-Lymphocytes, Cytotoxic - Abstract
Generating broadly neutralizing antibodies with candidate vaccines has remained an elusive goal. Consequently, vaccine candidates developed have aimed at eliciting cell-mediated immune effector activities (CMI) that could delay disease progression, and maybe also limit secondary transmission, by controlling virus replication. There is considerable discussion about what types of endpoints would constitute definable standardized clinical benefit to the individual that would result in licensure of these candidate vaccines. Identifying biomarkers that can be used as surrogates for clinical endpoints in randomized clinical trials would be useful, because it would shorten studies and reduce costs. Biological markers associated with disease progression and secondary transmission and that may be used as prognosis markers and surrogate endpoints in HIV vaccine trials have emerged from analyses of data from studies on natural history of HIV infection. Extensive literature is cited to support the use of plasma viral load as a primary endpoint for supporting licensure decisions. Overall, a significant result on viral load in a vaccine trial should be considered as a significant breakthrough for vaccines and be aggressively pursued with the caveat that such a result should rapidly be followed by well-defined studies to verify durable virological and immunological vaccine benefit, as well as ultimate clinical benefit. The review also provides perspectives on magnitude of viral load reduction, durability of viral load reduction for reduced progression of HIV disease.
- Published
- 2008
40. EV02: a Phase I trial to compare the safety and immunogenicity of HIV DNA-C prime-NYVAC-C boost to NYVAC-C alone
- Author
-
Donatella Ciuffreda, Hans Wolf, Pierre-Alexandre Bart, Jim Tartaglia, Wolfgang Stöhr, Elizabeth Brodnicki, Miranda Cowen, Tristan Barber, Romilda Gamboni, Marie-Joelle Frachette, Cristina Cellerai, Jonathan L. Heeney, Ralf Wagner, Christiane Moog, Ken Legg, Jonathan Weber, Giuseppe Pantaleo, Abdel Babiker, Séverine Burnet, Sheena McCormack, and Alexandre Harari
- Subjects
Adult ,Male ,Immunization, Secondary ,HIV Infections ,Asymptomatic ,Injections, Intramuscular ,law.invention ,Randomized controlled trial ,law ,medicine ,Humans ,Adverse effect ,Antigens, Viral ,AIDS Vaccines ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,Immunogenicity ,ELISPOT ,Public Health, Environmental and Occupational Health ,env Gene Products, Human Immunodeficiency Virus ,Viral Vaccines ,biology.organism_classification ,Regimen ,Infectious Diseases ,Alanine transaminase ,Drug Design ,Lentivirus ,Immunology ,biology.protein ,HIV-1 ,Molecular Medicine ,Female ,medicine.symptom ,Safety ,business - Abstract
Summary The aim of this randomised controlled trial was to see if the addition of 4 mg/ml DNA-C priming given by the intramuscular route at weeks 0 and 4 to NYVAC-C at weeks 20 and 24, safely increased the proportion of participants with HIV-specific T-cell responses measured by the interferon (IFN)-γ ELISpot assay at weeks 26 and/or 28 compared to NYVAC-C alone. Although 2 individuals discontinued after the first DNA-C due to adverse events (1 vaso-vagal; 1 transient, asymptomatic elevation in alanine transaminase), the vaccines were well tolerated. Three others failed to complete the regimen (1 changed her mind; 2 lost to follow-up). Of the 35 that completed the regimen 90% (18/20) in the DNA-C group had ELISpot responses compared to 33% (5/15) that received NYVAC-C alone ( p = 0.001). Responses were to envelope in the majority (21/23). Of the 9 individuals with responses to envelope and other peptides, 8 were in the DNA-C group. These promising results suggest that DNA-C was an effective priming agent, that merits further investigation.
- Published
- 2007
41. European Union and EDCTP strategy in the global context: recommendations for preventive HIV/AIDS vaccines research
- Author
-
Michael Hoelscher, Jim Tartaglia, Janneke van de Wijgert, Saladin Osmanov, Patrice Debré, Quentin J. Sattentau, Reinold E. Schmidt, Mario Clerici, Simonetta Di Fabio, Frances Gotch, Manuel Romaris, Coumba Toure, Clive M. Gray, Louise Pedneault, Arnd Hoeveler, Thomas Lehner, Jiri Mestecky, Global Health, and Infectious diseases
- Subjects
Economic growth ,Health Planning Guidelines ,Developing country ,Context (language use) ,Acquired immunodeficiency syndrome (AIDS) ,medicine ,Humans ,media_common.cataloged_instance ,European Union ,HIV vaccine ,European union ,Developing Countries ,media_common ,AIDS Vaccines ,Strategic planning ,Acquired Immunodeficiency Syndrome ,Clinical Trials as Topic ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Public Health, Environmental and Occupational Health ,HIV ,medicine.disease ,Infectious Diseases ,General partnership ,Immunology ,Molecular Medicine ,Safety ,business - Abstract
The European Commission (EC) has strong commitments and recognises the need to continue to ensure that HIV/AIDS research efforts receive global attention. The EC is facing this challenge in a global context and has made substantial investments together with European Developing Countries Clinical Trial Partnership (EDCTP) to formulate a program for the accomplishment of a scientific strategic plan promoting the European/African HIV vaccine development approach. The EC and EDCTP has convened a number of meetings by experts in basic and clinical virology, immunology, epidemiology, as well as industrial and regulatory representatives. The remit of the committee of experts was to define (1) objective criteria for selection of HIV candidates; (2) to determine criteria for selection of sites for clinical trials in Europe and Africa. The resulting consensus paper will guide the EC and EDCTP in developing HIV vaccine strategy and recommendations.
- Published
- 2005
42. Tumoral and immunologic response after vaccination of melanoma patients with an ALVAC virus encoding MAGE antigens recognized by T cells
- Author
-
Pierre van der Bruggen, Danielle Liénard, Philippe Moingeon, Daniel E. Speiser, Nicolas van Baren, Sylvie Negrier, Bernard Escudier, Pierre-Yves Dietrich, Vincent Brichard, Ralf G. Meyer, Gerd Ritter, Marie-Claude Bonnet, Dominique Maraninchi, Amir Khammari, Sophie Piperno-Neumann, Marie Marchand, Thierry Dorval, Brigitte Dréno, Susanne Osanto, Jim Tartaglia, Thierry Boon, and Pierre Coulie
- Subjects
Adult ,Male ,Cancer Research ,Vaccination schedule ,Enzyme-Linked Immunosorbent Assay ,Canarypox virus ,Recombinant virus ,Cancer Vaccines ,Virus ,Antigen ,Antigens, Neoplasm ,Medicine ,Humans ,Melanoma ,Aged ,Aged, 80 and over ,business.industry ,Immunogenicity ,Viral Vaccines ,Middle Aged ,Virology ,Neoplasm Proteins ,Vaccination ,Treatment Outcome ,Oncology ,Immunology ,Disease Progression ,Female ,Melanoma-Specific Antigens ,business ,T-Lymphocytes, Cytotoxic - Abstract
PurposeTo evaluate the toxicity, antitumoral effectiveness, and immunogenicity of repeated vaccinations with ALVAC miniMAGE-1/3, a recombinant canarypox virus containing a minigene encoding antigenic peptides MAGE-3168-176and MAGE-1161-169, which are presented by HLA-A1 and B35 on tumor cells and can be recognized by cytolytic T lymphocytes (CTLs).Materials and MethodsThe vaccination schedule comprised four sequential injections of the recombinant virus, followed by three booster vaccinations with the MAGE-3168-176and MAGE-1161-169peptides. The vaccines were administered, both intradermally and subcutaneously, at 3-week intervals.ResultsForty patients with advanced cancer were treated, including 37 melanoma patients. The vaccines were generally well tolerated with moderate adverse events, consisting mainly of transient inflammatory reactions at the virus injection sites. Among the 30 melanoma patients assessable for tumor response, a partial response was observed in one patient, and disease stabilization in two others. The remaining patients had progressive disease. Among the patients with stable or progressive disease, five showed evidence of tumor regression. A CTL response against the MAGE-3 vaccine antigen was detected in three of four patients with tumor regression, and in only one of 11 patients without regression.ConclusionRepeated vaccination with ALVAC miniMAGE-1/3 is associated with tumor regression and with a detectable CTL response in a minority of melanoma patients. There is a significant correlation between tumor regression and CTL response. The contribution of vaccine-induced CTL in the tumor regression process is discussed in view of the immunologic events that could be analyzed in detail in one patient.
- Published
- 2005
43. Characterization of functional and phenotypic changes in anti-Gag vaccine-induced T cell responses and their role in protection after HIV-1 infection
- Author
-
Sadie M. West, David Price, Robert B. Belshe, Mario Roederer, Kent J. Weinhold, Barbara Exley, David R. Ambrozak, Anju Bansal, Daniel C. Douek, Georgia D. Tomaras, Paul A. Goepfert, Zenaido T. Camacho, Vanessa Teaberry, Michael R. Betts, Guido Ferrari, Feng Gao, Richard A. Koup, Jim Tartaglia, and J. Michael Kilby
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,T cell ,T-Lymphocytes ,Molecular Sequence Data ,Gene Products, gag ,HIV Infections ,Disease ,Biology ,CD8-Positive T-Lymphocytes ,Epitope ,Virus ,HIV Seronegativity ,medicine ,Humans ,Amino Acid Sequence ,Allele ,HLA-B27 Antigen ,AIDS Vaccines ,Multidisciplinary ,Base Sequence ,virus diseases ,Biological Sciences ,Virology ,Phenotype ,R1 ,medicine.anatomical_structure ,Immunology ,DNA, Viral ,HIV-1 ,CD8 - Abstract
Worldwide HIV-1 vaccine efforts are guided by the principle that HIV-specific T cell responses may provide protection from infection or delay overt disease. However, no clear correlates of T cell-mediated immune protection have been identified. Here, we examine in a HLA-B27 + HIV seronegative vaccinee persistent HIV-specific vaccine-induced anti-Gag CD4 + and CD8 + T cell responses. Although these responses exhibited those characteristics (multifunctionality, appropriate memory phenotype, and targeting of epitopes associated with long-term nonprogression) predicted to correlate with protection from infection, the subject became HIV infected. After HIV infection, the vaccine-induced CD8 + T cells expanded, but both CD4 + and CD8 + T cell responses acquired the functional and phenotypic patterns characteristic of chronic HIV infection. The virus quickly escaped the vaccine-induced T cell response, and the subject progressed more rapidly than expected for someone expressing the HLA-B27 allele. These data suggest that control of HIV by vaccine-elicited HIV-specific T cell responses may be difficult, even when the T cell response has those characteristics predicted to provide optimal protection.
- Published
- 2005
44. Absence of immunodominant anti-Gag p17 (SL9) responses among Gag CTL-positive, HIV-uninfected vaccine recipients expressing the HLA-A*0201 allele
- Author
-
Kent J. Weinhold, Jim Tartaglia, Guido Ferrari, Michael R. Betts, Wesley Neal, Janet Ottinger, Richard A. Koup, Paul A. Goepfert, Bradley H. Edwards, Anizsa M. Jones, Susan Buchbinder, and M. Juliana McElrath
- Subjects
Canarypox ,HIV Antigens ,Immunology ,Immunization, Secondary ,Epitopes, T-Lymphocyte ,Gene Products, gag ,HIV Infections ,T-Cell Antigen Receptor Specificity ,Immunodominance ,Epitope ,Canarypox virus ,HIV Seronegativity ,HLA-A2 Antigen ,Vaccines, DNA ,Immunology and Allergy ,Humans ,HIV vaccine ,Alleles ,AIDS Vaccines ,Vaccines, Synthetic ,biology ,HLA-A Antigens ,Immunodominant Epitopes ,ELISPOT ,Immunogenicity ,Vaccination ,env Gene Products, Human Immunodeficiency Virus ,biology.organism_classification ,Virology ,Peptide Fragments ,Histocompatibility ,CTL ,HIV-1 ,T-Lymphocytes, Cytotoxic - Abstract
According to a number of previous reports, control of HIV replication in humans appears to be linked to the presence of anti-HIV-1 Gag-specific CD8 responses. During the chronic phase of HIV-1 infection, up to 75% of the HIV-infected individuals who express the histocompatibility leukocyte Ag (HLA)-A*0201 recognize the Gag p17 SLYNTVATL (aa residues 77–85) epitope (SL9). However, the role of the anti-SL9 CD8 CTL in controlling HIV-1 infection remains controversial. In this study we determined whether the pattern of SL9 immunodominance in uninfected, HLA-A*0201 HIV vaccine recipients is similar to that seen in chronically HIV-infected subjects. The presence of anti-SL9 responses was determined using a panel of highly sensitive cellular immunoassays, including peptide:MHC tetramer binding, IFN-γ ELISPOT, and cytokine flow cytometry. Thirteen HLA-A*0201 vaccinees with documented anti-Gag CD8 CTL reactivities were tested, and none had a detectable anti-SL9 response. These findings strongly suggest that the pattern of SL9 epitope immunodominance previously reported among chronically infected, HLA-A*0201-positive patients is not recapitulated in noninfected recipients of Gag-containing canarypox-based candidate vaccines and may be influenced by the relative immunogenicity of these constructs.
- Published
- 2004
45. Avipox-based simian immunodeficiency virus (SIV) vaccines elicit a high frequency of SIV-specific CD4+ and CD8+ T-cell responses in vaccinia-experienced SIVmac251-infected macaques
- Author
-
Genoveffa Franchini, Janos Nacsa, Antonia Radaelli, Jim Tartaglia, Dennis Panicali, Yvette Edghill-Smith, David Venzon, Carlo De Giuli Morghen, and Wen-Po Tsai
- Subjects
Fowlpox ,CD4-Positive T-Lymphocytes ,Cellular immunity ,Genotype ,viruses ,Simian Acquired Immunodeficiency Syndrome ,Gene Products, gag ,Gene Products, pol ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Antibodies, Viral ,Virus ,Avipoxvirus ,chemistry.chemical_compound ,medicine ,Vaccinia ,Animals ,Poxviridae ,Lymphocyte Count ,RNA, Messenger ,Immunization Schedule ,Immunity, Cellular ,General Veterinary ,General Immunology and Microbiology ,biology ,Public Health, Environmental and Occupational Health ,SAIDS Vaccines ,Simian immunodeficiency virus ,Group-specific antigen ,Vector vaccine ,medicine.disease ,biology.organism_classification ,Virology ,Macaca mulatta ,Infectious Diseases ,chemistry ,Immunology ,Molecular Medicine ,Cytokines ,RNA, Viral ,Simian Immunodeficiency Virus ,T-Lymphocytes, Cytotoxic - Abstract
The ability of ALVAC- or fowlpox-based simian immunodeficiency virus (SIV) vaccines to boost SIV-specific CD4+ and CD8+ T-cell responses was tested in 10 vaccinia-experienced macaques infected with SIVmac251. The CD8+ T-cell response to the dominant Gag 181–189 CM9 was quantitated in seven Mamu-A ∗ 01-positive macaques by tetramer staining, by ex vivo cytotoxic T-lymphocyte (CTL) activity, and by intracellular cytokine staining (ICS) with the specific Gag 181-189 CM9 peptide. The overall CD8+ T-cell response to Gag was assessed using a peptide pool encompassing the entire Gag protein followed by measurement of TNF-α production in ICS assay. Similarly, virus-specific CD4+ T-cell responses were measured by ICS for TNF-α following stimulation with the Gag-overlapping peptide and by proliferative response following stimulation with purified p27 Gag. The two vaccine modalities effectively boosted both CD4+ and CD8+ SIV-specific T-cell response despite prior exposure to the vaccinia-derivative NYVAC vector, suggesting that sequential boosting with either avipox-based vector vaccine candidate is a realistic approach in immune therapy of human immunodeficiency virus type 1 (HIV-1)-infected individuals.
- Published
- 2004
46. A novel chimeric Rev, Tat, and Nef (Retanef) antigen as a component of an SIV/HIV vaccine
- Author
-
Robert Harrod, Jake Fullen, Genoveffa Franchini, Jim Tartaglia, Julie M. Johnson, Elzbieta Tryniszewska, Zdeněk Hel, and Wen-Po Tsai
- Subjects
HIV Antigens ,animal diseases ,viruses ,Recombinant Fusion Proteins ,Genetic Vectors ,Molecular Sequence Data ,medicine.disease_cause ,Transfection ,Genes, env ,Epitope ,Virus ,Open Reading Frames ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Amino Acid Sequence ,HIV vaccine ,Antigens, Viral ,Vero Cells ,AIDS Vaccines ,Vaccines, Synthetic ,Expression vector ,General Veterinary ,General Immunology and Microbiology ,biology ,Base Sequence ,ELISPOT ,Immunogenicity ,Immunotoxins ,Vaccination ,Public Health, Environmental and Occupational Health ,SAIDS Vaccines ,virus diseases ,Proteins ,Simian immunodeficiency virus ,biology.organism_classification ,Virology ,Macaca mulatta ,Genes, nef ,Genes, rev ,Infectious Diseases ,Genes, tat ,Lentivirus ,HIV-1 ,Molecular Medicine ,Simian Immunodeficiency Virus ,HeLa Cells - Abstract
The human immunodeficiency virus type 1 (HIV-1) regulatory proteins Rev, Tat, and Nef are expressed at early time post-infection and represent attractive targets to be included in a vaccine candidate for AIDS. However, the putative immunosuppressive activities of some of these proteins may limit their immunogenicity. To circumvent these issues, a novel chimeric polyprotein vaccine candidate (Retanef), comprising genetically modified and re-assorted rev, tat, and nef open reading frames of simian immunodeficiency virus (SIV), was constructed and optimized for its expression in mammalian cells. Retanef encodes a protein of approximately 55 kDa localized primarily in the cytoplasm of transfected cells. The Retanef gene expressed in context of an eucaryotic expression vector (DNA-SIV-Retanef) or cloned into a highly attenuated poxvirus-based NYVAC vector (NYVAC-SIV-Retanef) was used to immunize either naive rhesus macaques or macaques chronically infected with SIVmac251 undergoing anti-retroviral therapy (ART). Three immunizations of naive macaques with DNA-SIV-Retanef followed by a single NYVAC-SIV-Retanef boost induced a response to the Mamu-A∗01-restricted Tat epitope (Tat_SL8, TTPESANL) demonstrated by staining with a specific tetramer and by direct cytolytic activity assays, as well as responses to Rev, Tat and Nef proteins demonstrated by ELISPOT assays using overlapping peptide pools encompassing the entire proteins. Immunization of infected macaques with either DNA-SIV-Retanef or NYVAC-SIV-Retanef expanded the frequency of Tat-specific tetramer-staining cells by two- to seven-fold. No adverse effects were observed in either naive or SIV-infected rhesus macaques. Thus, an analogous HIV-1-based chimeric vaccine may represent useful component of an HIV-1 vaccine.
- Published
- 2002
47. Enhanced multiepitope-based vaccines elicit CD8+ cytotoxic T cells against both immunodominant and cryptic epitopes
- Author
-
Stephen W. Davis, Guy Russo, John A. Tine, Jim Tartaglia, Anne Payne, Philippe Moingeon, Pierre Langlade Demoyen, François A. Lemonnier, and Hüseyin Firat
- Subjects
T cell ,Genetic Vectors ,Epitopes, T-Lymphocyte ,Mice, Transgenic ,Immunodominance ,Biology ,Cancer Vaccines ,Epitope ,Mice ,Immune system ,MART-1 Antigen ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,MHC class I ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Antigen Presentation ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,Immunodominant Epitopes ,Public Health, Environmental and Occupational Health ,Virology ,Neoplasm Proteins ,Infectious Diseases ,medicine.anatomical_structure ,Immunology ,biology.protein ,Molecular Medicine ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
A frequent issue in vaccinology is to elicit balanced T cell responses against both immunodominant and cryptic T cell epitopes, from one or several antigens presented at the same time to the immune system. Using HLA-A2.1.1 restricted epitopes from the Melan A/MART-1 or gp 100 melanoma-associated antigens as a model, we engineered a series of constructs in the ALVAC canarypox vector system: T cell epitopes were expressed either as linear polyepitopes (with or without spacers), or as minigenes encoding a single epitope. The latter were found to allow the best processing and presentation of most T cell epitopes, following infection by ALVAC recombinants of the HLA A2+ bladder carcinoma cell line and stimulation of epitope-specific human TIL lines. These various constructs were also used to immunize HLA-A2.1.1 HHD transgenic mice to compare their capacity to elicit T cells responses. Polyepitopes but also minigenes encoding wild-type epitopes could not elicit in a reliable manner balanced CTL responses against all target epitopes from gp100. We could rescue T cells responses against poorly immunogenic epitopes after introducing appropriate point mutations to enhance their interaction with MHC Class I molecules. Epitope enhancement within either polyepitope, multiepitopes (i.e. minigenes expressed under the control of separate promoters) or full length immunogens should be systematically considered when designing vaccines containing both cryptic and immunodominant target epitopes.
- Published
- 2002
48. Severe impairment of primary but not memory responses to influenza viral antigens in aged mice: costimulation in vivo partially reverses impaired primary immune responses
- Author
-
Renata Miranda, Jim Tartaglia, Suryaprakash Sambhara, Brian J. Underdown, David S. Burt, Anjna Kurichh, Olive James, and Michel Klein
- Subjects
Aging ,Immunology ,Genetic Vectors ,Vaccinia virus ,Biology ,Immune Dysfunction ,Antibodies, Viral ,Lymphocyte Activation ,Preventive vaccination ,Mice ,Immune system ,Adjuvants, Immunologic ,Orthomyxoviridae Infections ,In vivo ,Antigens, CD ,Animals ,Antigens, Viral ,Cells, Cultured ,Viral antigens ,Membrane Glycoproteins ,Virology ,Survival Analysis ,Up-Regulation ,Vaccination ,Influenza A virus ,Influenza Vaccines ,Mice, Inbred DBA ,B7-1 Antigen ,Female ,B7-2 Antigen ,Influenza virus vaccine ,Immunologic Memory ,T-Lymphocytes, Cytotoxic - Abstract
Profound alterations in humoral and cellular immune responses are a hallmark of aging, and understanding the immunobiology of aging is key to the success of preventive vaccination strategies. With aging, while recall or memory responses to influenza viral antigens for the most part remained unaltered, primary immune responses are severely impaired. The impaired primary responses are partly due to a lack of costimulation, as providing costimulation at the time of induction of primary immune responses against influenza virus vaccine partially reversed aged-related immune dysfunction and conferred enhanced protection. Inclusion of immunomodulators that up-regulate the expression of costimulatory molecules must be considered to improve the efficacy of vaccination in the elderly, particularly to novel immunogens.
- Published
- 2001
49. Modulation of the antibody response to the HIV envelope subunit by co-administration of infectious or heat-inactivated canarypoxvirus (ALVAC) preparations
- Author
-
Florence Boudet, Jim Tartaglia, Catherine Moste, Michel Chevalier, and Therese-Marie Jourdier
- Subjects
Male ,Canarypox ,Immunogen ,Neutrophils ,medicine.medical_treatment ,Guinea Pigs ,HIV Antibodies ,Immunoglobulin G ,Avipoxvirus ,HIV Envelope Protein gp160 ,Mice ,Antigen ,medicine ,Animals ,General Veterinary ,General Immunology and Microbiology ,biology ,Immunogenicity ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,Virology ,Recombinant Proteins ,Infectious Diseases ,Immunology ,biology.protein ,Molecular Medicine ,Female ,Antibody ,Adjuvant ,Chickens - Abstract
Poxviruses are large DNA viruses capable of infecting a broad range of animal species. Infection is generally accompanied by an inflammatory response in the host, the extent of which varies considerably with the specific poxvirus and host species. Regarding ALVAC, a poxvirus derived from the canarypox vaccine strain, Kanapox, and which represents a promising immunization vehicle in humans, nothing is known about its inflammatory capacity. The present study was aimed at documenting this issue in rodents, including mice and guinea pigs. It was then attempted to evaluate how such properties could influence the immunogenicity of an antigen concomitantly administered with ALVAC preparations using the HIV envelope subunit, rgp160, as the model immunogen. The results revealed that ALVAC, either infectious or heat-inactivated, induced in both animal species an early inflammatory response, as evidenced by a rapid migration of neutrophils to the site of inoculation. In parallel, the canarypoxvirus was shown to strongly adjuvant the co-administered immunogen, resulting in a marked increase in Env-specific IgG, IgG1 and particularly IgG2(a) serum titers. Of further interest, the heat-inactivated preparation of ALVAC retained this immunostimulatory activity. Whether or not a link between the inflammatory and immunomodulatory properties of ALVAC exists remains to be established, but such features are clearly interesting with respect to the potential use of ALVAC as an immunization vehicle.
- Published
- 2001
50. Recombinant viruses as a tool for therapeutic vaccination against human cancers
- Author
-
Moingeon P, Michel Klein, Jim Tartaglia, F Verdier, Marie-Claude Bonnet, A Lindberg, and Philippe Kourilsky
- Subjects
viruses ,medicine.medical_treatment ,Immunology ,Genetic Vectors ,Biology ,Cancer Vaccines ,Immune tolerance ,Viral vector ,Immune system ,Cancer immunotherapy ,Antigen ,Antigens, Neoplasm ,Neoplasms ,medicine ,Immune Tolerance ,Immunology and Allergy ,Humans ,Clinical Trials as Topic ,Vaccines, Synthetic ,Immunogenicity ,Immunotherapy, Active ,Immunotherapy ,Virology ,Immunization ,DNA, Viral - Abstract
Viral vectors can be used to express a variety of genes in vivo, that encode tumor associated antigens, cytokines, or accessory molecules. For vaccination purposes, the ideal viral vector should be safe and enable efficient presentation of expressed antigens to the immune system. It should also exhibit low intrinsic immunogenicity to allow for its re-administration in order to boost relevant specific immune responses. Furthermore, the vector system must meet criteria that enable its industrialization. The characteristics of the most promising viral vectors, including retroviruses, poxviruses, adenoviruses, adeno-associated viruses, herpes simplex viruses, and alphaviruses, will be reviewed in this communication. Such recombinant viruses have been successfully used in animal models as therapeutic cancer vaccines. Based on these encouraging results, a series of clinical studies, reviewed herein, have been undertaken. Human clinical trials, have as of today, allowed investigators to establish that recombinant viruses can be safely used in cancer patients, and that such recombinants can break immune tolerance against tumor-associated antigens. These promising results are now leading to improved immunization protocols associating recombinant viruses with alternate antigen-presentation platforms (prime-boost regimens), in order to elicit broad tumor-specific immune responses (humoral and cellular) against multiple target antigens.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.