1
|
Cain M, Huang Q, Sanchez S, Ly H, Liang Y. Evaluating Antigen- and Vector-Specific Immune Responses of a Recombinant Pichinde Virus-Based Vaccine Expressing the Lymphocytic Choriomeningitis Virus Nucleoprotein. Vaccines (Basel) 2024; 12:1450. [PMID: 39772112 PMCID: PMC11680116 DOI: 10.3390/vaccines12121450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Live viral vector-based vaccines are known to elicit strong immune responses, but their use can be limited by anti-vector immunity. Here, we analyzed the immunological responses of a live-attenuated recombinant Pichinde virus (PICV) vector platform (rP18tri). METHODS To evaluate anti-PICV immunity in the development of vaccine antigen-specific immune responses, we generated a rP18tri-based vaccine expressing the lymphocytic choriomeningitis virus (LCMV) nucleoprotein (NP) and administered four doses of this rP18tri-NPLCMV vaccine to mice. Using MHC-I tetramers to detect PICV NP38-45 and LCMV NP396-404 epitope-specific CD8+ T cells, we monitored vector- and vaccine-antigen-specific immune responses after each vaccination dose. RESULTS LCMV NP396-404-specific effector and memory CD8+ T cells were detected after the first dose and peaked after the second dose, whereas PICV NP38-45-specific memory CD8+ T cells increased with each dose. PICV-binding IgG antibodies peaked after the second dose, while anti-PICV neutralizing antibodies (NAbs) remained low even after the fourth dose. Immunization with the rP18tri-NPLCMV vaccine significantly reduced LCMV viral titers in a chronic LCMV Clone 13 infection model, demonstrating the protective role of LCMV NP-specific T cells. CONCLUSION These findings provide important insights into the antigen- and vector-specific immunity of the rP18tri-NPLCMV vaccine and support the development of NP-based vaccines against arenavirus pathogens.
Collapse
Affiliation(s)
| | | | | | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA; (M.C.); (Q.H.)
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA; (M.C.); (Q.H.)
| |
Collapse
|
2
|
Santos LC, Fernandes AMS, Alves IA, Serafini MR, Silva LDSE, de Freitas HF, Leite LCC, Santos CC. Trends in Viral Vector-Based Vaccines for Tuberculosis: A Patent Review (2010-2023). Vaccines (Basel) 2024; 12:876. [PMID: 39204002 PMCID: PMC11359462 DOI: 10.3390/vaccines12080876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Tuberculosis (TB) is an ancient global public health problem. Several strategies have been applied to develop new and more effective vaccines against TB, from attenuated or inactivated mycobacteria to recombinant subunit or genetic vaccines, including viral vectors. This review aimed to evaluate patents filed between 2010 and 2023 for TB vaccine candidates. It focuses on viral vector-based strategies. A search was carried out in Espacenet, using the descriptors "mycobacterium and tuberculosis" and the classification A61K39. Of the 411 patents preliminarily identified, the majority were related to subunit vaccines, with 10 patents based on viral vector platforms selected in this study. Most of the identified patents belong to the United States or China, with a concentration of patent filings between 2013 and 2023. Adenoviruses were the most explored viral vectors, and the most common immunodominant Mycobacterium tuberculosis (Mtb) antigens were present in all the selected patents. The majority of patents were tested in mouse models by intranasal or subcutaneous route of immunization. In the coming years, an increased use of this platform for prophylactic and/or therapeutic approaches for TB and other diseases is expected. Along with this, expanding knowledge about the safety of this technology is essential to advance its use.
Collapse
Affiliation(s)
- Lana C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Antônio Márcio Santana Fernandes
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | - Izabel Almeida Alves
- Departamento do Medicamento, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil;
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Estado da Bahia, Salvador 41150-000, BA, Brazil
| | - Mairim Russo Serafini
- Departamento de Farmácia, Universidade Federal do Sergipe, São Cristóvão 49100-000, SE, Brazil;
| | - Leandra da Silva e Silva
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
| | | | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, SP, Brazil;
| | - Carina C. Santos
- Serviço de Imunologia das Doenças Infecciosas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil; (L.C.S.); (A.M.S.F.); (L.d.S.e.S.)
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador 40170-115, BA, Brazil
| |
Collapse
|
3
|
Sharma B, Bekerman E, Truong H, Lee J, Gamez-Guerrero M, Boopathy A, Mital R, Huang KB, Ahmadi-Erber S, Wimmer R, Schulha S, Lauterbach H, Orlinger K, Suthram S, Lewis MG, Blair W, Makadzange T, Geleziunas R, Murry JP, Schmidt S. Arenavirus-Based Vectors Generate Robust SIV Immunity in Non-Human Primates. Vaccines (Basel) 2024; 12:735. [PMID: 39066373 PMCID: PMC11281402 DOI: 10.3390/vaccines12070735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Arenavirus-based vectors are being investigated as therapeutic vaccine candidates with the potential to elicit robust CD8 T-cell responses. We compared the immunogenicity of replicating (artPICV and artLCMV) and non-replicating (rPICV and rLCMV) arenavirus-based vectors expressing simian immunodeficiency virus (SIV) Gag and Envelope (Env) immunogens in treatment-naïve non-human primates. Heterologous regimens with non-replicating and replicating vectors elicited more robust SIV IFN-γ responses than a homologous regimen, and replicating vectors elicited significantly higher cellular immunogenicity than non-replicating vectors. The heterologous regimen elicited high anti-Env antibody titers when administered intravenously, with replicating vectors inducing significantly higher titers than non-replicating vectors. Intramuscular immunization resulted in more durable antibody responses than intravenous immunization for both vector platforms, with no difference between the replicating and non-replicating vectors. Overall, both replicating and non-replicating arenavirus vectors generated robust T- and B-cell-mediated immunity to SIV antigens in treatment-naïve non-human primates, supporting further evaluation of these vectors in a clinical setting for HIV therapy.
Collapse
Affiliation(s)
| | | | - Hoa Truong
- Gilead Sciences, Inc., Foster City, CA 94404, USA
| | - Johnny Lee
- Gilead Sciences, Inc., Foster City, CA 94404, USA
| | | | | | - Rohit Mital
- Gilead Sciences, Inc., Foster City, CA 94404, USA
| | | | | | | | | | | | | | | | | | - Wade Blair
- Gilead Sciences, Inc., Foster City, CA 94404, USA
| | | | | | | | | |
Collapse
|
4
|
Berger A, Pedersen J, Kowatsch MM, Scholte F, Lafrance MA, Azizi H, Li Y, Gomez A, Wade M, Fausther-Bovendo H, de La Vega MA, Jelinski J, Babuadze G, Nepveu-Traversy ME, Lamarre C, Racine T, Kang CY, Gaillet B, Garnier A, Gilbert R, Kamen A, Yao XJ, Fowke KR, Arts E, Kobinger G. Impact of Recombinant VSV-HIV Prime, DNA-Boost Vaccine Candidates on Immunogenicity and Viremia on SHIV-Infected Rhesus Macaques. Vaccines (Basel) 2024; 12:369. [PMID: 38675751 PMCID: PMC11053682 DOI: 10.3390/vaccines12040369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Currently, no effective vaccine to prevent human immunodeficiency virus (HIV) infection is available, and various platforms are being examined. The vesicular stomatitis virus (VSV) vaccine vehicle can induce robust humoral and cell-mediated immune responses, making it a suitable candidate for the development of an HIV vaccine. Here, we analyze the protective immunological impacts of recombinant VSV vaccine vectors that express chimeric HIV Envelope proteins (Env) in rhesus macaques. To improve the immunogenicity of these VSV-HIV Env vaccine candidates, we generated chimeric Envs containing the transmembrane and cytoplasmic tail of the simian immunodeficiency virus (SIV), which increases surface Env on the particle. Additionally, the Ebola virus glycoprotein was added to the VSV-HIV vaccine particles to divert tropism from CD4 T cells and enhance their replications both in vitro and in vivo. Animals were boosted with DNA constructs that encoded matching antigens. Vaccinated animals developed non-neutralizing antibody responses against both the HIV Env and the Ebola virus glycoprotein (EBOV GP) as well as systemic memory T-cell activation. However, these responses were not associated with observable protection against simian-HIV (SHIV) infection following repeated high-dose intra-rectal SHIV SF162p3 challenges.
Collapse
Affiliation(s)
- Alice Berger
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Jannie Pedersen
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Monika M. Kowatsch
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (M.M.K.); (K.R.F.)
| | - Florine Scholte
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Marc-Alexandre Lafrance
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Hiva Azizi
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Yue Li
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada; (Y.L.); (C.-Y.K.); (E.A.)
| | - Alejandro Gomez
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Matthew Wade
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Hugues Fausther-Bovendo
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Marc-Antoine de La Vega
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Joseph Jelinski
- Galveston National Laboratory, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - George Babuadze
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | | | - Claude Lamarre
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Unversité Laval, Quebec, QC G1V 0A6, Canada; (A.B.); (J.P.); (F.S.); (M.-A.L.); (H.A.); (A.G.); (M.W.); (H.F.-B.); (M.-A.d.L.V.); (G.B.); (C.L.)
| | - Trina Racine
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec, QC G1E 6W2, Canada; (T.R.); (X.-J.Y.)
| | - Chil-Yong Kang
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada; (Y.L.); (C.-Y.K.); (E.A.)
| | - Bruno Gaillet
- Department of Chemical Engineering, Faculty of Science and Engineering, Laval University, Quebec, QC G1V 0A6, Canada; (B.G.); (A.G.)
| | - Alain Garnier
- Department of Chemical Engineering, Faculty of Science and Engineering, Laval University, Quebec, QC G1V 0A6, Canada; (B.G.); (A.G.)
| | - Rénald Gilbert
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council, Montreal, QC H4P 2R2, Canada;
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Xiao-Jian Yao
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec, QC G1E 6W2, Canada; (T.R.); (X.-J.Y.)
| | - Keith R. Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (M.M.K.); (K.R.F.)
| | - Eric Arts
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada; (Y.L.); (C.-Y.K.); (E.A.)
| | - Gary Kobinger
- Galveston National Laboratory, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| |
Collapse
|
5
|
Belnoue E, Vogelzang A, Nieuwenhuizen NE, Krzyzaniak MA, Darbre S, Kreutzfeldt M, Wagner I, Merkler D, Lambert PH, Kaufmann SHE, Siegrist CA, Pinschewer DD. Replication-Deficient Lymphocytic Choriomeningitis Virus-Vectored Vaccine Candidate for the Induction of T Cell Immunity against Mycobacterium tuberculosis. Int J Mol Sci 2022; 23:2700. [PMID: 35269842 PMCID: PMC8911050 DOI: 10.3390/ijms23052700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 11/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) represents a major burden to global health, and refined vaccines are needed. Replication-deficient lymphocytic choriomeningitis virus (rLCMV)-based vaccine vectors against cytomegalovirus have proven safe for human use and elicited robust T cell responses in a large proportion of vaccine recipients. Here, we developed an rLCMV vaccine expressing the Mtb antigens TB10.4 and Ag85B. In mice, rLCMV elicited high frequencies of polyfunctional Mtb-specific CD8 and CD4 T cell responses. CD8 but not CD4 T cells were efficiently boosted upon vector re-vaccination. High-frequency responses were also observed in neonatally vaccinated mice, and co-administration of rLCMV with Expanded Program of Immunization (EPI) vaccines did not result in substantial reciprocal interference. Importantly, rLCMV immunization significantly reduced the lung Mtb burden upon aerosol challenge, resulting in improved lung ventilation. Protection was associated with increased CD8 T cell recruitment but reduced CD4 T cell infiltration upon Mtb challenge. When combining rLCMV with BCG vaccination in a heterologous prime-boost regimen, responses to the rLCMV-encoded Mtb antigens were further augmented, but protection was not significantly different from rLCMV or BCG vaccination alone. This work suggests that rLCMV may show utility for neonatal and/or adult vaccination efforts against pulmonary tuberculosis.
Collapse
Affiliation(s)
- Elodie Belnoue
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Alexis Vogelzang
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; (A.V.); (N.E.N.); (S.H.E.K.)
| | - Natalie E. Nieuwenhuizen
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; (A.V.); (N.E.N.); (S.H.E.K.)
| | - Magdalena A. Krzyzaniak
- Division of Experimental Virology, Department of Biomedicine, University of Basel, 4003 Basel, Switzerland;
| | - Stephanie Darbre
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva 4, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva 4, Switzerland
| | - Paul-Henri Lambert
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; (A.V.); (N.E.N.); (S.H.E.K.)
| | - Claire-Anne Siegrist
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Daniel D. Pinschewer
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
- Division of Experimental Virology, Department of Biomedicine, University of Basel, 4003 Basel, Switzerland;
| |
Collapse
|
6
|
Law JLM, Logan M, Joyce MA, Landi A, Hockman D, Crawford K, Johnson J, LaChance G, Saffran HA, Shields J, Hobart E, Brassard R, Arutyunova E, Pabbaraju K, Croxen M, Tipples G, Lemieux MJ, Tyrrell DL, Houghton M. SARS-COV-2 recombinant Receptor-Binding-Domain (RBD) induces neutralizing antibodies against variant strains of SARS-CoV-2 and SARS-CoV-1. Vaccine 2021; 39:5769-5779. [PMID: 34481699 PMCID: PMC8387217 DOI: 10.1016/j.vaccine.2021.08.081] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022]
Abstract
SARS-CoV-2 is the etiological agent of COVID19. There are currently several licensed vaccines approved for human use and most of them target the spike protein in the virion envelope to induce protective immunity. Recently, variants that spread more quickly have emerged. There is evidence that some of these variants are less sensitive to neutralization in vitro, but it is not clear whether they can evade vaccine induced protection. In this study, we tested SARS-CoV-2 spike RBD as a vaccine antigen and explored the effect of formulation with Alum/MPLA or AddaS03 adjuvants. Our results show that RBD induces high titers of neutralizing antibodies and activates strong cellular immune responses. There is also significant cross-neutralization of variants B.1.1.7 and B.1.351 and to a lesser extent, SARS-CoV-1. These results indicate that recombinant RBD can be a viable candidate as a stand-alone vaccine or as a booster shot to diversify our strategy for COVID19 protection.
Collapse
Affiliation(s)
- John Lok Man Law
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| | - Michael Logan
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Michael A Joyce
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Abdolamir Landi
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Darren Hockman
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Kevin Crawford
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Janelle Johnson
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Gerald LaChance
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Holly A Saffran
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Justin Shields
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Eve Hobart
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Raelynn Brassard
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Elena Arutyunova
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | | | | | - Graham Tipples
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada; Alberta Precision Laboratories, Edmonton, Canada
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - D Lorne Tyrrell
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Michael Houghton
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
7
|
Hokello J, Sharma AL, Tyagi M. An Update on the HIV DNA Vaccine Strategy. Vaccines (Basel) 2021; 9:vaccines9060605. [PMID: 34198789 PMCID: PMC8226902 DOI: 10.3390/vaccines9060605] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 01/24/2023] Open
Abstract
In 2020, the global prevalence of human immunodeficiency virus (HIV) infection was estimated to be 38 million, and a total of 690,000 people died from acquired immunodeficiency syndrome (AIDS)–related complications. Notably, around 12.6 million people living with HIIV/AIDS did not have access to life-saving treatment. The advent of the highly active antiretroviral therapy (HAART) in the mid-1990s remarkably enhanced the life expectancy of people living with HIV/AIDS as a result of improved immune functions. However, HAART has several drawbacks, especially when it is not used properly, including a high risk for the development of drug resistance, as well as undesirable side effects such as lipodystrophy and endocrine dysfunctions, which result in HAART intolerability. HAART is also not curative. Furthermore, new HIV infections continue to occur globally at a high rate, with an estimated 1.7 million new infections occurring in 2018 alone. Therefore, there is still an urgent need for an affordable, effective, and readily available preventive vaccine against HIV/AIDS. Despite this urgent need, however, progress toward an effective HIV vaccine has been modest over the last four decades. Reasons for this slow progress are mainly associated with the unique aspects of HIV itself and its ability to rapidly mutate, targeting immune cells and escape host immune responses. Several approaches to an HIV vaccine have been undertaken. However, this review will mainly discuss progress made, including the pre-clinical and clinical trials involving vector-based HIV DNA vaccines and the use of integrating lentiviral vectors in HIV vaccine development. We concluded by recommending particularly the use of integrase-defective lentiviral vectors, owing to their safety profiles, as one of the promising vectors in HIV DNA vaccine strategies both for prophylactic and therapeutic HIV vaccines.
Collapse
Affiliation(s)
- Joseph Hokello
- Department of Microbiology and Immunology, Faculty of Biomedical Sciences, Kampala International University-Western Campus, P.O. Box 71, Bushenyi 0256, Uganda;
| | | | - Mudit Tyagi
- Center for Translational Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA;
- Correspondence:
| |
Collapse
|
8
|
Bonilla WV, Kirchhammer N, Marx AF, Kallert SM, Krzyzaniak MA, Lu M, Darbre S, Schmidt S, Raguz J, Berka U, Vincenti I, Pauzuolis M, Kerber R, Hoepner S, Günther S, Magnus C, Merkler D, Orlinger KK, Zippelius A, Pinschewer DD. Heterologous arenavirus vector prime-boost overrules self-tolerance for efficient tumor-specific CD8 T cell attack. CELL REPORTS MEDICINE 2021; 2:100209. [PMID: 33763654 PMCID: PMC7974551 DOI: 10.1016/j.xcrm.2021.100209] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/16/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Therapeutic vaccination regimens inducing clinically effective tumor-specific CD8+ T lymphocyte (CTL) responses are an unmet medical need. We engineer two distantly related arenaviruses, Pichinde virus and lymphocytic choriomeningitis virus, for therapeutic cancer vaccination. In mice, life-replicating vector formats of these two viruses delivering a self-antigen in a heterologous prime-boost regimen induce tumor-specific CTL responses up to 50% of the circulating CD8 T cell pool. This CTL attack eliminates established solid tumors in a significant proportion of animals, accompanied by protection against tumor rechallenge. The magnitude of CTL responses is alarmin driven and requires combining two genealogically distantly related arenaviruses. Vector-neutralizing antibodies do not inhibit booster immunizations by the same vector or by closely related vectors. Rather, CTL immunodominance hierarchies favor vector backbone-targeted responses at the expense of self-reactive CTLs. These findings establish an arenavirus-based immunotherapy regimen that allows reshuffling of immunodominance hierarchies and breaking self-directed tolerance for efficient tumor control. Engineered arenaviruses induce potent tumor self-specific CD8 T cell (CTL) response Combinations of distantly but not closely related arenavirus vectors eliminate tumors Vector backbone-targeted CTL responses compete against tumor self-reactive CTLs Optimized vector combinations reshuffle immunodominance to break self-tolerance
Collapse
Affiliation(s)
- Weldy V Bonilla
- University of Basel, Department of Biomedicine, Basel, Switzerland
| | | | | | - Sandra M Kallert
- University of Basel, Department of Biomedicine, Basel, Switzerland
| | | | - Min Lu
- University of Basel, Department of Biomedicine, Basel, Switzerland
| | - Stéphanie Darbre
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | | | | | - Ilena Vincenti
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Mindaugas Pauzuolis
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Romy Kerber
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sabine Hoepner
- Tumor Immunology, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Carsten Magnus
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Division of Clinical Pathology, University Hospitals of Geneva, Geneva, Switzerland
| | | | - Alfred Zippelius
- University of Basel, Department of Biomedicine, Basel, Switzerland.,Medical Oncology, University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
9
|
Ring SS, Królik M, Hartmann F, Schmidt E, Ali OH, Ludewig B, Kochanek S, Flatz L. Heterologous Prime Boost Vaccination Induces Protective Melanoma-Specific CD8 + T Cell Responses. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:179-187. [PMID: 33209978 PMCID: PMC7658660 DOI: 10.1016/j.omto.2020.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022]
Abstract
Cancer vaccination aims at inducing an adaptive immune response against tumor-derived antigens. In this study, we utilize recombinant human adenovirus serotype 5 (rAd5) and recombinant lymphocytic choriomeningitis virus (rLCMV)-based vectors expressing the melanocyte differentiation antigen gp100. In contrast to single or homologous vaccination, a heterologous prime boost vaccination starting with a rAd5-gp100 prime immunization followed by a rLCMV-gp100 boost injection induces a high magnitude of polyfunctional gp100-specific CD8+ T cells. Our data indicate that an optimal T cell induction is dependent on the order and interval of the vaccinations. A prophylactic prime boost vaccination with rAd5- and rLCMV-gp100 protects mice from a B16.F10 melanoma challenge. In the therapeutic setting, combination of the vaccination with low-dose cyclophosphamide showed a synergistic effect and significantly delayed tumor growth. Our findings suggest that heterologous viral vector prime boost immunizations can mediate tumor control in a mouse melanoma model.
Collapse
Affiliation(s)
- Sandra S Ring
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Michał Królik
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Fabienne Hartmann
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Erika Schmidt
- Department of Gene Therapy, Ulm University, Helmholtzstrasse 8, 89081 Ulm, Germany
| | - Omar Hasan Ali
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Stefan Kochanek
- Department of Gene Therapy, Ulm University, Helmholtzstrasse 8, 89081 Ulm, Germany
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland.,Department of Oncology and Hematology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Department of Dermatology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| |
Collapse
|
10
|
Schwendinger M, Thiry G, De Vos B, Leroux-Roels G, Bruhwyler J, Huygens A, Ganeff C, Buchinger H, Orlinger KK, Pinschewer DD, Monath TP, Lilja AE. A Randomized Dose-Escalating Phase I Trial of a Replication-Deficient Lymphocytic Choriomeningitis Virus Vector-Based Vaccine Against Human Cytomegalovirus. J Infect Dis 2020; 225:1399-1410. [PMID: 32313928 PMCID: PMC9016443 DOI: 10.1093/infdis/jiaa121] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/20/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND A vaccine (HB-101) consisting of 2 nonreplicating lymphocytic choriomeningitis virus (LCMV) vectors expressing the human cytomegalovirus antigens glycoprotein B (gB) and the 65-kD phosphoprotein (pp65), respectively, is in development to prevent cytomegalovirus infection. METHODS HB-101 was tested in cytomegalovirus-naive, healthy adults in a randomized, double-blind, placebo-controlled, dose-escalation Phase I trial. Fifty-four subjects received low, medium, or high dose of HB-101 or placebo by intramuscular administration at Month 0, 1, and 3. Safety and immunogenicity were the respective primary and secondary endpoints. Subjects were followed for 12 months after the initial immunization. RESULTS Vaccination was associated with transient mild to moderate adverse events. HB-101 administration induced dose-dependent gB- and pp65-specific cellular responses, dominated by pp65-specific CD8 T cells, a high fraction of which were polyfunctional. Two administrations were sufficient to elicit dose-dependent gB-binding and cytomegalovirus-neutralizing antibodies (Abs). Cytomegalovirus-specific immune responses were boosted after each administration. Only 1 of 42 vaccine recipients mounted a transient LCMV vector-neutralizing Ab response. CONCLUSIONS HB-101 was well tolerated and induced cytomegalovirus-specific polyfunctional CD8 T-cell and neutralizing Ab responses in the majority of subjects. Lack of vector-neutralizing Ab responses should facilitate booster vaccinations. These results justify further clinical evaluation of this vaccine candidate.
Collapse
Affiliation(s)
| | - Georges Thiry
- Hookipa Pharma Inc., New York, New York, USA.,Senergues Consult, Saint Etienne de Maurs, France
| | - Beatrice De Vos
- Bejamad bvba, Consultancy Office Pharmaceutical Sciences, Dworp, Belgium
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | - Daniel D Pinschewer
- Hookipa Pharma Inc., New York, New York, USA.,University of Basel, Department of Biomedicine - Haus Petersplatz, Basel, Switzerland
| | - Thomas P Monath
- Hookipa Pharma Inc., New York, New York, USA.,Crozet BioPharma LLC, Devens, Massachusetts, USA
| | | |
Collapse
|
11
|
Abstract
The unprecedented challenges of developing effective vaccines against intracellular pathogens such as HIV, malaria, and tuberculosis have resulted in more rational approaches to vaccine development. Apart from the recent advances in the design and selection of improved epitopes and adjuvants, there are also ongoing efforts to optimize delivery platforms. The unprecedented challenges of developing effective vaccines against intracellular pathogens such as HIV, malaria, and tuberculosis have resulted in more rational approaches to vaccine development. Apart from the recent advances in the design and selection of improved epitopes and adjuvants, there are also ongoing efforts to optimize delivery platforms. Viral vectors are the best-characterized delivery tools because of their intrinsic adjuvant capability, unique cellular tropism, and ability to trigger robust adaptive immune responses. However, a known limitation of viral vectors is preexisting immunity, and ongoing efforts are aimed at developing novel vector platforms with lower seroprevalence. It is also becoming increasingly clear that different vectors, even those derived from phylogenetically similar viruses, can elicit substantially distinct immune responses, in terms of quantity, quality, and location, which can ultimately affect immune protection. This review provides a summary of the status of viral vector development for HIV vaccines, with a particular focus on novel viral vectors and the types of adaptive immune responses that they induce.
Collapse
|
12
|
Kallert SM, Darbre S, Bonilla WV, Kreutzfeldt M, Page N, Müller P, Kreuzaler M, Lu M, Favre S, Kreppel F, Löhning M, Luther SA, Zippelius A, Merkler D, Pinschewer DD. Replicating viral vector platform exploits alarmin signals for potent CD8 + T cell-mediated tumour immunotherapy. Nat Commun 2017; 8:15327. [PMID: 28548102 PMCID: PMC5458557 DOI: 10.1038/ncomms15327] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/22/2017] [Indexed: 12/27/2022] Open
Abstract
Viral infections lead to alarmin release and elicit potent cytotoxic effector T lymphocyte (CTLeff) responses. Conversely, the induction of protective tumour-specific CTLeff and their recruitment into the tumour remain challenging tasks. Here we show that lymphocytic choriomeningitis virus (LCMV) can be engineered to serve as a replication competent, stably-attenuated immunotherapy vector (artLCMV). artLCMV delivers tumour-associated antigens to dendritic cells for efficient CTL priming. Unlike replication-deficient vectors, artLCMV targets also lymphoid tissue stroma cells expressing the alarmin interleukin-33. By triggering interleukin-33 signals, artLCMV elicits CTLeff responses of higher magnitude and functionality than those induced by replication-deficient vectors. Superior anti-tumour efficacy of artLCMV immunotherapy depends on interleukin-33 signalling, and a massive CTLeff influx triggers an
inflammatory conversion of the tumour microenvironment. Our observations suggest that replicating viral delivery systems can release alarmins for improved anti-tumour efficacy. These mechanistic insights may outweigh safety concerns around replicating viral vectors in cancer immunotherapy. Viruses trigger potent cytotoxic T cell responses, whereas anti-tumour immunity has been difficult to establish. Here the authors engineer a replicating viral delivery system for tumour-associated antigens, which induces alarmin release, innate activation and protective anti-tumour immunity in mice.
Collapse
Affiliation(s)
- Sandra M Kallert
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Petersplatz 10, 4009 Basel, Switzerland
| | - Stephanie Darbre
- Departement de Pathologie et Immunologie, Centre Médical Universitaire, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Weldy V Bonilla
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Petersplatz 10, 4009 Basel, Switzerland
| | - Mario Kreutzfeldt
- Departement de Pathologie et Immunologie, Centre Médical Universitaire, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Nicolas Page
- Departement de Pathologie et Immunologie, Centre Médical Universitaire, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Philipp Müller
- Department of Biomedicine, University Hospital and University of Basel, Hebelstr. 20, 4031 Basel, Switzerland
| | - Matthias Kreuzaler
- Department of Biomedicine, University Hospital and University of Basel, Hebelstr. 20, 4031 Basel, Switzerland
| | - Min Lu
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Petersplatz 10, 4009 Basel, Switzerland
| | - Stéphanie Favre
- Department of Biochemistry, Center for Immunity and Infection Lausanne, University of Lausanne, Chemin des Boveresses 144, 1066 Epalinges, Switzerland
| | - Florian Kreppel
- Witten/Herdecke University (UW/H), Faculty of Health/School of Medicine, Stockumer Str. 10, 58453 Witten, Germany
| | - Max Löhning
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Sanjiv A Luther
- Department of Biochemistry, Center for Immunity and Infection Lausanne, University of Lausanne, Chemin des Boveresses 144, 1066 Epalinges, Switzerland
| | - Alfred Zippelius
- Department of Biomedicine, University Hospital and University of Basel, Hebelstr. 20, 4031 Basel, Switzerland.,Department of Medical Oncology, University Hospital Basel, Hebelstr. 20, 4031 Basel, Switzerland
| | - Doron Merkler
- Departement de Pathologie et Immunologie, Centre Médical Universitaire, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Petersplatz 10, 4009 Basel, Switzerland
| |
Collapse
|
13
|
Penaloza MacMaster P, Shields JL, Alayo QA, Cabral C, Jimenez J, Mondesir J, Chandrashekar A, Cabral JM, Lim M, Iampietro MJ, Provine NM, Bricault CA, Seaman M, Orlinger K, Aspoeck A, Fuhrmann G, Lilja AE, Monath T, Mangeat B, Pinschewer DD, Barouch DH. Development of novel replication-defective lymphocytic choriomeningitis virus vectors expressing SIV antigens. Vaccine 2016; 35:1-9. [PMID: 27899229 DOI: 10.1016/j.vaccine.2016.11.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/06/2016] [Accepted: 11/18/2016] [Indexed: 01/23/2023]
Abstract
An important focus in vaccine research is the design of vaccine vectors with low seroprevalence and high immunogenicity. Replication-incompetent lymphocytic choriomeningitis virus (rLCMV) vectors do not elicit vector-neutralizing antibody responses, and homologous prime-boost regimens with rLCMV vectors induce boostable and protective T cell responses to model antigens in mice. However, cellular and humoral immune responses following homologous rLCMV vaccine regimens have not been rigorously evaluated in non-human primates (NHPs). To test whether rLCMV vectors constitute an effective vaccine platform in NHPs, we developed rLCMV vectors expressing SIVmac239 Env and Gag antigens and assessed their immunogenicity in mice and cynomolgus macaques. Immunization with rLCMV vaccine vectors expressing SIV Env and Gag was effective at generating SIV-specific T cell and antibody responses in both mice and NHPs. Epitope mapping using SIV Env in C57BL/6 mice demonstrated that rLCMV vectors induced sustained poly-functional responses to both dominant and subdominant epitopes. Our results suggest the potential of rLCMV vectors as vaccine candidates. Future SIV challenge experiments in rhesus macaques will be needed to assess immune protection by these vaccine vectors.
Collapse
Affiliation(s)
- Pablo Penaloza MacMaster
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer L Shields
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Quazim A Alayo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Crystal Cabral
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Jessica Jimenez
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Jade Mondesir
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Joseph M Cabral
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Matthew Lim
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - M Justin Iampietro
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Nicholas M Provine
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Christine A Bricault
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Michael Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Klaus Orlinger
- Hookipa Biotech AG Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Andreas Aspoeck
- Hookipa Biotech AG Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Gerhard Fuhrmann
- Hookipa Biotech AG Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Anders E Lilja
- Hookipa Biotech AG Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Thomas Monath
- Hookipa Biotech AG Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Bastien Mangeat
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland
| | - Daniel D Pinschewer
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Boston, MA 02114, USA.
| |
Collapse
|
14
|
Cheng C, Wang L, Ko SY, Kong WP, Schmidt SD, Gall JGD, Colloca S, Seder RA, Mascola JR, Nabel GJ. Combination recombinant simian or chimpanzee adenoviral vectors for vaccine development. Vaccine 2015; 33:7344-7351. [PMID: 26514419 PMCID: PMC11059210 DOI: 10.1016/j.vaccine.2015.10.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/01/2015] [Accepted: 10/07/2015] [Indexed: 11/25/2022]
Abstract
Recombinant adenoviral vector (rAd)-based vaccines are currently being developed for several infectious diseases and cancer therapy, but pre-existing seroprevalence to such vectors may prevent their use in broad human populations. In this study, we investigated the potential of low seroprevalence non-human primate rAd vectors to stimulate cellular and humoral responses using HIV/SIV Env glycoprotein (gp) as the representative antigen. Mice were immunized with novel simian or chimpanzee rAd (rSAV or rChAd) vectors encoding HIV gp or SIV gp by single immunization or in heterologous prime/boost combinations (DNA/rAd; rAd/rAd; rAd/NYVAC or rAd/rLCM), and adaptive immunity was assessed. Among the rSAV and rChAd tested, rSAV16 or rChAd3 vector alone generated the most potent immune responses. The DNA/rSAV regimen also generated immune responses similar to the DNA/rAd5 regimen. rChAd63/rChAd3 and rChAd3 /NYVAC induced similar or even higher levels of CD4+ and CD8+ T-cell and IgG responses as compared to rAd28/rAd5, one of the most potent combinations of human rAds. The optimized vaccine regimen stimulated improved cellular immune responses and neutralizing antibodies against HIV compared to the DNA/rAd5 regimen. Based on these results, this type of novel rAd vector and its prime/boost combination regimens represent promising candidates for vaccine development.
Collapse
MESH Headings
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- AIDS Vaccines/isolation & purification
- Adenoviruses, Simian/genetics
- Adenoviruses, Simian/immunology
- Animals
- Antibodies, Viral/blood
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Drug Carriers
- Drug Discovery/methods
- Female
- Genetic Vectors
- Immunoglobulin G/blood
- Mice, Inbred BALB C
- SAIDS Vaccines/genetics
- SAIDS Vaccines/immunology
- SAIDS Vaccines/isolation & purification
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/isolation & purification
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Viral Vaccines/isolation & purification
Collapse
Affiliation(s)
- Cheng Cheng
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - Lingshu Wang
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - Sung-Youl Ko
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - Wing-Pui Kong
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - Stephen D Schmidt
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - Jason G D Gall
- GenVec, Inc., 65 West Watkins Mill Rd., Gaithersburg, MD 20878, United States.
| | | | - Robert A Seder
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - John R Mascola
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| | - Gary J Nabel
- Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, United States.
| |
Collapse
|
15
|
The Nucleoprotein Is Required for Lymphocytic Choriomeningitis Virus-Based Vaccine Vector Immunogenicity. J Virol 2015; 89:11734-8. [PMID: 26355095 DOI: 10.1128/jvi.01613-15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/06/2015] [Indexed: 12/14/2022] Open
Abstract
Recombinant glycoprotein-deficient lymphocytic choriomeningitis virus-based vaccine vectors (rLCMV/ΔGP) are potent CD8(+) T cell inducers. To investigate the underlying molecular requirements, we generated a nucleoprotein-deficient vector counterpart (rLCMV/ΔNP). NP but not GP is a minimal trans-acting factor for viral transcription and genome replication. We found that, unlike rLCMV/ΔGP, rLCMV/ΔNP failed to elicit detectable CD8(+) T cell responses unless NP was trans complemented in a transgenic host. Hence, NP-dependent intracellular gene expression is essential for LCMV vector immunogenicity.
Collapse
|
16
|
Breakthrough of SIV strain smE660 challenge in SIV strain mac239-vaccinated rhesus macaques despite potent autologous neutralizing antibody responses. Proc Natl Acad Sci U S A 2015; 112:10780-5. [PMID: 26261312 DOI: 10.1073/pnas.1509731112] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Although the correlates of immunological protection from human immunodeficiency virus or simian immunodeficiency virus infection remain incompletely understood, it is generally believed that medium to high titers of serum neutralizing antibodies (nAbs) against the challenge virus will prevent infection. This paradigm is based on a series of studies in which passive transfer of HIV-specific nAbs protected rhesus macaques (RMs) from subsequent mucosal challenge with a chimeric human/simian immunodeficiency virus. However, it is unknown whether nAb titers define protection in the setting of active immunization. Here we determined serum nAb titers against breakthrough transmitted/founder (T/F) SIVsmE660-derived envelope glycoprotein (Env) variants from 14 RMs immunized with SIVmac239-based DNA-prime/modified vaccinia virus Ankara-boost vaccine regimens that included GM-CSF or CD40L adjuvants and conferred significant but incomplete protection against repeated low-dose intrarectal challenge. A single Env variant established infection in all RMs except one, with no identifiable genetic signature associated with vaccination breakthrough compared with T/F Envs from four unvaccinated monkeys. Breakthrough T/F Env pseudoviruses were potently neutralized in vitro by heterologous pooled serum from chronically SIVsmE660-infected monkeys at IC50 titers exceeding 1:1,000,000. Remarkably, the T/F Env pseudoviruses from 13 of 14 monkeys were also susceptible to neutralization by autologous prechallenge serum at in vitro IC50 titers ranging from 1:742-1:10,832. These titers were similar to those observed in vaccinated RMs that remained uninfected. These data suggest that the relationship between serum nAb titers and protection from mucosal SIV challenge in the setting of active immunization is more complex than previously recognized, warranting further studies into the balance between immune activation, target cell availability, and protective antibody responses.
Collapse
|
17
|
Characterization and Implementation of a Diverse Simian Immunodeficiency Virus SIVsm Envelope Panel in the Assessment of Neutralizing Antibody Breadth Elicited in Rhesus Macaques by Multimodal Vaccines Expressing the SIVmac239 Envelope. J Virol 2015; 89:8130-51. [PMID: 26018167 DOI: 10.1128/jvi.01221-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 09/03/2014] [Indexed: 02/02/2023] Open
Abstract
UNLABELLED Antibodies that can neutralize diverse viral strains are likely to be an important component of a protective human immunodeficiency virus type 1 (HIV-1) vaccine. To this end, preclinical simian immunodeficiency virus (SIV)-based nonhuman primate immunization regimens have been designed to evaluate and enhance antibody-mediated protection. However, these trials often rely on a limited selection of SIV strains with extreme neutralization phenotypes to assess vaccine-elicited antibody activity. To mirror the viral panels used to assess HIV-1 antibody breadth, we created and characterized a novel panel of 14 genetically and phenotypically diverse SIVsm envelope (Env) glycoproteins. To assess the utility of this panel, we characterized the neutralizing activity elicited by four SIVmac239 envelope-expressing DNA/modified vaccinia virus Ankara vector- and protein-based vaccination regimens that included the immunomodulatory adjuvants granulocyte-macrophage colony-stimulating factor, Toll-like receptor (TLR) ligands, and CD40 ligand. The SIVsm Env panel exhibited a spectrum of neutralization sensitivity to SIV-infected plasma pools and monoclonal antibodies, allowing categorization into three tiers. Pooled sera from 91 rhesus macaques immunized in the four trials consistently neutralized only the highly sensitive tier 1a SIVsm Envs, regardless of the immunization regimen. The inability of vaccine-mediated antibodies to neutralize the moderately resistant tier 1b and tier 2 SIVsm Envs defined here suggests that those antibodies were directed toward epitopes that are not accessible on most SIVsm Envs. To achieve a broader and more effective neutralization profile in preclinical vaccine studies that is relevant to known features of HIV-1 neutralization, more emphasis should be placed on optimizing the Env immunogen, as the neutralization profile achieved by the addition of adjuvants does not appear to supersede the neutralizing antibody profile determined by the immunogen. IMPORTANCE Many in the HIV/AIDS vaccine field believe that the ability to elicit broadly neutralizing antibodies capable of blocking genetically diverse HIV-1 variants is a critical component of a protective vaccine. Various SIV-based nonhuman primate vaccine studies have investigated ways to improve antibody-mediated protection against a heterologous SIV challenge, including administering adjuvants that might stimulate a greater neutralization breadth. Using a novel SIV neutralization panel and samples from four rhesus macaque vaccine trials designed for cross comparison, we show that different regimens expressing the same SIV envelope immunogen consistently elicit antibodies that neutralize only the very sensitive tier 1a SIV variants. The results argue that the neutralizing antibody profile elicited by a vaccine is primarily determined by the envelope immunogen and is not substantially broadened by including adjuvants, resulting in the conclusion that the envelope immunogen itself should be the primary consideration in efforts to elicit antibodies with greater neutralization breadth.
Collapse
|
18
|
Vaccine-Induced Linear Epitope-Specific Antibodies to Simian Immunodeficiency Virus SIVmac239 Envelope Are Distinct from Those Induced to the Human Immunodeficiency Virus Type 1 Envelope in Nonhuman Primates. J Virol 2015; 89:8643-50. [PMID: 26018159 DOI: 10.1128/jvi.03635-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/17/2015] [Indexed: 01/04/2023] Open
Abstract
To evaluate antibody specificities induced by simian immunodeficiency virus (SIV) versus human immunodeficiency virus type 1 (HIV-1) envelope antigens in nonhuman primate (NHP), we profiled binding antibody responses to linear epitopes in NHP studies with HIV-1 or SIV immunogens. We found that, overall, HIV-1 Env IgG responses were dominated by V3, with the notable exception of the responses to the vaccine strain A244 Env that were dominated by V2, whereas the anti-SIVmac239 Env responses were dominated by V2 regardless of the vaccine regimen.
Collapse
|
19
|
Schell JB, Bahl K, Folta-Stogniew E, Rose N, Buonocore L, Marx PA, Gambhira R, Rose JK. Antigenic requirement for Gag in a vaccine that protects against high-dose mucosal challenge with simian immunodeficiency virus. Virology 2015; 476:405-412. [PMID: 25591175 DOI: 10.1016/j.virol.2014.12.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/16/2014] [Accepted: 12/17/2014] [Indexed: 11/28/2022]
Abstract
We reported previously on a vaccine approach that conferred apparent sterilizing immunity to SIVsmE660. The vaccine regimen employed a prime-boost using vectors based on recombinant vesicular stomatitis virus (VSV) and an alphavirus replicon expressing either SIV Gag or SIV Env. In the current study, we tested the ability of vectors expressing only the SIVsmE660 Env protein to protect macaques against the same high-dose mucosal challenge. Animals developed neutralizing antibody levels comparable to or greater than seen in the previous vaccine study. When the vaccinated animals were challenged with the same high-dose of SIVsmE660, all became infected. While average peak viral loads in animals were slightly lower than those of previous controls, the viral set points were not significantly different. These data indicate that Gag, or the combination of Gag and Env are required for the generation of apparent sterilizing immunity to the SIVsmE660 challenge.
Collapse
Affiliation(s)
- John B Schell
- Yale University School of Medicine, New Haven, CT, United States
| | - Kapil Bahl
- Yale University School of Medicine, New Haven, CT, United States
| | - Ewa Folta-Stogniew
- Yale University School of Medicine, New Haven, CT, United States; Keck Biophysical Resource Facility, New Haven, CT, United States
| | - Nina Rose
- Yale University School of Medicine, New Haven, CT, United States
| | - Linda Buonocore
- Yale University School of Medicine, New Haven, CT, United States
| | - Preston A Marx
- Tulane National Primate Research Center, Covington, LA, United States
| | - Ratish Gambhira
- Tulane National Primate Research Center, Covington, LA, United States
| | - John K Rose
- Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
20
|
Xu H, Wang X, Veazey RS. Simian Immunodeficiency Virus Infection and Mucosal Immunity. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
21
|
Regoes RR, Magnus C. The role of chance in primate lentiviral infectivity: from protomer to host organism. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 129:327-51. [PMID: 25595809 DOI: 10.1016/bs.pmbts.2014.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Infection is best described as a stochastic process. Whether a host becomes infected upon exposure has a strong random element. The same applies to cells exposed to virions. In this review, we show how the mathematical formalism for stochastic processes has been used to describe and understand the infection by the Human and Simian Immunodeficiency Virus on different levels. We survey quantitative studies on the establishment of infection in the host (the organismal level) and on the infection of target cells (the cellular and molecular level). We then discuss how a synthesis of the approaches across these levels could give rise to a predictive framework for assessing the efficacy of microbicides and vaccines.
Collapse
Affiliation(s)
- Roland R Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland.
| | - Carsten Magnus
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Roychoudhuri R, Lefebvre F, Honda M, Pan L, Ji Y, Klebanoff CA, Nichols CN, Fourati S, Hegazy AN, Goulet JP, Gattinoni L, Nabel GJ, Gilliet M, Cameron M, Restifo NP, Sékaly RP, Flatz L. Transcriptional profiles reveal a stepwise developmental program of memory CD8(+) T cell differentiation. Vaccine 2014; 33:914-23. [PMID: 25446821 DOI: 10.1016/j.vaccine.2014.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/11/2014] [Accepted: 10/06/2014] [Indexed: 11/30/2022]
Abstract
The generation of CD8(+) T-cell memory is a major aim of vaccination. While distinct subsets of CD8(+) T-cells are generated following immunization that differ in their ability to confer long-term immunity against infection, the transcriptional profiles of these subsets within endogenous vaccine-induced CD8(+) T cell responses have not been resolved. Here, we measure global transcriptional profiles of endogenous effector (TEFF), effector memory (TEM) and central memory (TCM) CD8(+) T-cells arising from immunization with three distinct prime-boost vaccine regimens. While a proportion of transcripts were uniquely regulated within distinct CD8(+) T cell populations, we observed progressive up- or down-regulation in the expression of a majority of differentially expressed transcripts when subsets were compared in the order TN>TCM>TEM>TEFF. Strikingly, when we compared global differences in gene expression between TN, TCM, TEM and TEFF cells with known transcriptional changes that result when CD8(+) T cells repetitively encounter antigen, our analysis overwhelmingly favored a model whereby cumulative antigen stimulation drives differentiation specifically from TN>TCM>TEM>TEFF and this was common to all vaccines tested. These findings provide insight into the molecular basis of immunological memory and identify potential biomarkers for characterization of vaccine-induced responses and prediction of vaccine efficacy.
Collapse
Affiliation(s)
- Rahul Roychoudhuri
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | - Mitsuo Honda
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Pan
- Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987, USA
| | - Yun Ji
- National Cancer Institute (NCI), National Institutes of Bethesda, Bethesda, MD 20892, USA
| | | | - Carmen N Nichols
- Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987, USA
| | - Slim Fourati
- Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987, USA
| | - Ahmed N Hegazy
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Luca Gattinoni
- National Cancer Institute (NCI), National Institutes of Bethesda, Bethesda, MD 20892, USA
| | - Gary J Nabel
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michel Gilliet
- Department of Dermatology, University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Mark Cameron
- Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987, USA
| | - Nicholas P Restifo
- National Cancer Institute (NCI), National Institutes of Bethesda, Bethesda, MD 20892, USA
| | - Rafick P Sékaly
- Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987, USA.
| | - Lukas Flatz
- Department of Dermatology, University Hospital CHUV, 1011 Lausanne, Switzerland; Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| |
Collapse
|
23
|
Abstract
Recent advances in the immunology, pathogenesis, and prevention of human immunodeficiency virus (HIV) infection continue to reveal clues to the mechanisms involved in the progressive immunodeficiency attributed to infection, but more importantly have shed light on the correlates of immunity to infection and disease progression. HIV selectively infects, eliminates, and/or dysregulates several key cells of the human immune system, thwarting multiple arms of the host immune response, and inflicting severe damage to mucosal barriers, resulting in tissue infiltration of 'symbiotic' intestinal bacteria and viruses that essentially become opportunistic infections promoting systemic immune activation. This leads to activation and recruitment or more target cells for perpetuating HIV infection, resulting in persistent, high-level viral replication in lymphoid tissues, rapid evolution of resistant strains, and continued evasion of immune responses. However, vaccine studies and studies of spontaneous controllers are finally providing correlates of immunity from protection and disease progression, including virus-specific CD4(+) T-cell responses, binding anti-bodies, innate immune responses, and generation of antibodies with potent antibody-dependent cell-mediated cytotoxicity activity. Emerging correlates of immunity indicate that prevention of HIV infection may be possible through effective vaccine strategies that protect and stimulate key regulatory cells and immune responses in susceptible hosts. Furthermore, immune therapies specifically directed toward boosting specific aspects of the immune system may eventually lead to a cure for HIV-infected patients.
Collapse
Affiliation(s)
- Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA
| | | | | |
Collapse
|
24
|
Roederer M, Keele BF, Schmidt SD, Mason RD, Welles HC, Fischer W, Labranche C, Foulds KE, Louder MK, Yang ZY, Todd JPM, Buzby AP, Mach LV, Shen L, Seaton KE, Ward BM, Bailer RT, Gottardo R, Gu W, Ferrari G, Alam SM, Denny TN, Montefiori DC, Tomaras GD, Korber BT, Nason MC, Seder RA, Koup RA, Letvin NL, Rao SS, Nabel GJ, Mascola JR. Immunological and virological mechanisms of vaccine-mediated protection against SIV and HIV. Nature 2014; 505:502-8. [PMID: 24352234 PMCID: PMC3946913 DOI: 10.1038/nature12893] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 11/21/2013] [Indexed: 02/07/2023]
Abstract
A major challenge for the development of a highly effective AIDS vaccine is the identification of mechanisms of protective immunity. To address this question, we used a nonhuman primate challenge model with simian immunodeficiency virus (SIV). We show that antibodies to the SIV envelope are necessary and sufficient to prevent infection. Moreover, sequencing of viruses from breakthrough infections revealed selective pressure against neutralization-sensitive viruses; we identified a two-amino-acid signature that alters antigenicity and confers neutralization resistance. A similar signature confers resistance of human immunodeficiency virus (HIV)-1 to neutralization by monoclonal antibodies against variable regions 1 and 2 (V1V2), suggesting that SIV and HIV share a fundamental mechanism of immune escape from vaccine-elicited or naturally elicited antibodies. These analyses provide insight into the limited efficacy seen in HIV vaccine trials.
Collapse
Affiliation(s)
- Mario Roederer
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Brandon F Keele
- SAIC-Frederick, Frederick National Laboratory, NIH, Frederick, Maryland 21702, USA
| | | | | | - Hugh C Welles
- 1] Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA [2] George Washington University, Washington DC 20052, USA
| | - Will Fischer
- Los Alamos National Laboratories, Los Alamos, New Mexico 87545, USA
| | - Celia Labranche
- Department of Surgery, Duke University, Durham, North Carolina 27710, USA
| | - Kathryn E Foulds
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Mark K Louder
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Zhi-Yong Yang
- 1] Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA [2] Sanofi-Pasteur, Cambridge, Massachusetts 02139, USA
| | - John-Paul M Todd
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Adam P Buzby
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA
| | - Linh V Mach
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA
| | - Ling Shen
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA
| | - Kelly E Seaton
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
| | - Brandy M Ward
- Department of Surgery, Duke University, Durham, North Carolina 27710, USA
| | - Robert T Bailer
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Raphael Gottardo
- Fred Hutchison Cancer Research Center, Seattle, Washington 98109, USA
| | - Wenjuan Gu
- Biostatistics Research Branch, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Guido Ferrari
- Department of Surgery, Duke University, Durham, North Carolina 27710, USA
| | - S Munir Alam
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
| | - Thomas N Denny
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
| | - David C Montefiori
- Department of Surgery, Duke University, Durham, North Carolina 27710, USA
| | - Georgia D Tomaras
- Human Vaccine Institute, Duke University, Durham, North Carolina 27710, USA
| | - Bette T Korber
- Los Alamos National Laboratories, Los Alamos, New Mexico 87545, USA
| | - Martha C Nason
- Biostatistics Research Branch, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Robert A Seder
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Richard A Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Norman L Letvin
- 1] Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115, USA [2]
| | - Srinivas S Rao
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Gary J Nabel
- 1] Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA [2] Sanofi-Pasteur, Cambridge, Massachusetts 02139, USA
| | - John R Mascola
- Vaccine Research Center, NIAID, NIH, Bethesda, Maryland 20892, USA
| |
Collapse
|
25
|
HIV vaccine research and discovery in the nonhuman primates model: a unified theory in acquisition prevention and control of SIV infection. Curr Opin HIV AIDS 2013; 8:288-94. [PMID: 23666390 DOI: 10.1097/coh.0b013e328361cfe8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Here we highlight the latest advances in HIV vaccine concepts that will expand our knowledge on how to elicit effective acquisition-prevention and/or control of simian immunodeficiency virus (SIV) replication in the nonhuman primate (NHP) model. RECENT FINDINGS In the context of the promising analyses from the RV144 Thai Trial and the effective control of SIV replication exerted by rhCMV-(SIV) elicited EM CD8 T cells, the HIV field has recently shifted toward vaccine concepts that combine protection from acquisition with effective control of SIV replication. Current studies in the NHP model have demonstrated the efficacy of HIV-neutralizing antibodies via passive transfer, the potential importance of the CD4 Tfh subset, the ability to effectively model the RV144 vaccine trial and the capacity of an Ad26 prime and modified vaccinia Ankara virus boost to elicit Env-specific antibody and cellular responses that both limit acquisition and control heterologous SIVmac251 challenge. SUMMARY The latest work in the NHP model suggests that the next generation HIV-1 vaccines should aim to provoke a comprehensive adaptive immune response for both prevention of SIV acquisition as well as control of replication in breakthrough infection.
Collapse
|
26
|
Kulkarni V, Rosati M, Valentin A, Jalah R, Alicea C, Yu L, Guan Y, Shen X, Tomaras GD, LaBranche C, Montefiori DC, Irene C, Prattipati R, Pinter A, Sullivan SM, Pavlakis GN, Felber BK. Vaccination with Vaxfectin(®) adjuvanted SIV DNA induces long-lasting humoral immune responses able to reduce SIVmac251 Viremia. Hum Vaccin Immunother 2013; 9:2069-80. [PMID: 23820294 DOI: 10.4161/hv.25442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We evaluated the immunogenicity and efficacy of Vaxfectin(®) adjuvanted SIV DNA vaccines in mice and macaques. Vaccination of mice with Vaxfectin(®) adjuvanted SIV gag DNA induced higher humoral immune responses than administration of unadjuvanted DNA, whereas similar levels of cellular immunity were elicited. Vaxfectin(®) adjuvanted SIVmac251 gag and env DNA immunization of rhesus macaques was used to examine magnitude, durability, and efficacy of humoral immunity. Vaccinated macaques elicited potent neutralizing antibodies able to cross-neutralize the heterologous SIVsmE660 Env. We found remarkable durability of Gag and Env humoral responses, sustained during ~2 y of follow-up. The Env-specific antibody responses induced by Vaxfectin(®) adjuvanted env DNA vaccination disseminated into mucosal tissues, as demonstrated by their presence in saliva, including responses to the V1-V2 region, and rectal fluids. The efficacy of the immune responses was evaluated upon intrarectal challenge with low repeated dose SIVmac251. Although 2 of the 3 vaccinees became infected, these animals showed significantly lower peak virus loads and lower chronic viremia than non-immunized infected controls. Thus, Vaxfectin(®) adjuvanted DNA is a promising vaccine approach for inducing potent immune responses able to control the highly pathogenic SIVmac251.
Collapse
Affiliation(s)
- Viraj Kulkarni
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Margherita Rosati
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Antonio Valentin
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Rashmi Jalah
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Lei Yu
- Institute of Human Virology and Department of Microbiology & Immunology; University of Maryland School of Medicine; Baltimore, MD USA
| | - Yongjun Guan
- Institute of Human Virology and Department of Microbiology & Immunology; University of Maryland School of Medicine; Baltimore, MD USA
| | | | | | | | | | - Carmela Irene
- Public Health Research Institute; University of Medicine and Dentistry of New Jersey; Newark, NJ USA
| | - Rajasekhar Prattipati
- Public Health Research Institute; University of Medicine and Dentistry of New Jersey; Newark, NJ USA
| | - Abraham Pinter
- Public Health Research Institute; University of Medicine and Dentistry of New Jersey; Newark, NJ USA
| | | | - George N Pavlakis
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| |
Collapse
|
27
|
Li J, Valentin A, Kulkarni V, Rosati M, Beach RK, Alicea C, Hannaman D, Reed SG, Felber BK, Pavlakis GN. HIV/SIV DNA vaccine combined with protein in a co-immunization protocol elicits highest humoral responses to envelope in mice and macaques. Vaccine 2013; 31:3747-55. [PMID: 23624057 DOI: 10.1016/j.vaccine.2013.04.037] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/05/2013] [Accepted: 04/15/2013] [Indexed: 12/27/2022]
Abstract
Vaccination with HIV/SIV DNAs elicits potent T-cell responses. To improve humoral immune responses, we combined DNA and protein in a co-immunization protocol using in vivo electroporation in mice and macaques. DNA&protein co-immunization induced higher antibody responses than DNA or protein alone, or DNA prime/protein boost in mice. DNA&protein co-immunization induced similar levels of cellular responses as those obtained by DNA only vaccination. The inclusion of SIV or HIV Env gp120 protein did not impair the development of cellular immune responses elicited by DNA present in the vaccine regimen. In macaques, the DNA&protein co-immunization regimen also elicited higher levels of humoral responses with broader cross-neutralizing activity. Despite the improved immunogenicity of DNA&protein co-immunization, the protein formulation with the EM-005 (GLA-SE) adjuvant further increased the anti-Env humoral responses. Dissecting the contribution of EM-005, we found that its administration upregulated the expression of co-stimulatory molecules and stimulated cytokine production, especially IL-6, by dendritic cells in vivo. These terminally differentiated, mature, dendritic cells possibly promote higher levels of humoral responses, supporting the inclusion of the EM-005 adjuvant with the vaccine. Thus, DNA&protein co-immunization is a promising strategy to improve the rapidity of development, magnitude and potency of the humoral immune responses.
Collapse
Affiliation(s)
- Jinyao Li
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mucosal priming with a replicating-vaccinia virus-based vaccine elicits protective immunity to simian immunodeficiency virus challenge in rhesus monkeys. J Virol 2013; 87:5669-77. [PMID: 23487457 DOI: 10.1128/jvi.03247-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mucosal surfaces are not targeted by most human immunodeficiency virus type 1 (HIV-1) vaccines, despite being major routes for HIV-1 transmission. Here we report a novel vaccination regimen consisting of a mucosal prime with a modified replicating vaccinia virus Tiantan strain (MVTT(SIVgpe)) and an intramuscular boost with a nonreplicating adenovirus strain (Ad5(SIVgpe)). This regimen elicited robust cellular immune responses with enhanced magnitudes, sustainability, and polyfunctionality, as well as higher titers of neutralizing antibodies against the simian immunodeficiency virus SIV(mac1A11) in rhesus monkeys. The reductions in peak and set-point viral loads were significant in most animals, with one other animal being protected fully from high-dose intrarectal inoculation of SIV(mac239). Furthermore, the animals vaccinated with this regimen were healthy, while ~75% of control animals developed simian AIDS. The protective effects correlated with the vaccine-elicited SIV-specific CD8(+) T cell responses against Gag and Pol. Our study provides a novel strategy for developing an HIV-1 vaccine by using the combination of a replicating vector and mucosal priming.
Collapse
|
29
|
Heterogeneity in neutralization sensitivities of viruses comprising the simian immunodeficiency virus SIVsmE660 isolate and vaccine challenge stock. J Virol 2013; 87:5477-92. [PMID: 23468494 DOI: 10.1128/jvi.03419-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The sooty mangabey-derived simian immunodeficiency virus (SIV) strain E660 (SIVsmE660) is a genetically heterogeneous, pathogenic isolate that is commonly used as a vaccine challenge strain in the nonhuman primate (NHP) model of human immunodeficiency virus type 1 (HIV-1) infection. Though it is often employed to assess antibody-based vaccine strategies, its sensitivity to antibody-mediated neutralization has not been well characterized. Here, we utilize single-genome sequencing and infectivity assays to analyze the neutralization sensitivity of the uncloned SIVsmE660 isolate, individual viruses comprising the isolate, and transmitted/founder (T/F) viruses arising from low-dose mucosal inoculation of macaques with the isolate. We found that the SIVsmE660 isolate overall was highly sensitive to neutralization by SIV-infected macaque plasma samples (50% inhibitory concentration [IC50] < 10(-5)) and monoclonal antibodies targeting V3 (IC50 < 0.01 μg/ml), CD4-induced (IC50 < 0.1 μg/ml), CD4 binding site (IC50 ~ 1 μg/ml), and V4 (IC50, ~5 μg/ml) epitopes. In comparison, SIVmac251 and SIVmac239 were highly resistant to neutralization by these same antibodies. Differences in neutralization sensitivity between SIVsmE660 and SIVmac251/239 were not dependent on the cell type in which virus was produced or tested. These findings indicate that in comparison to SIVmac251/239 and primary HIV-1 viruses, SIVsmE660 generally exhibits substantially less masking of antigenically conserved Env epitopes. Interestingly, we identified a minor population of viruses (~10%) in both the SIVsmE660 isolate and T/F viruses arising from it that were substantially more resistant (>1,000-fold) to antibody neutralization and another fraction (~20%) that was intermediate in neutralization resistance. These findings may explain the variable natural history and variable protection afforded by heterologous Env-based vaccines in rhesus macaques challenged by high-dose versus low-dose SIVsmE660 inoculation regimens.
Collapse
|
30
|
Conformational epitope consisting of the V3 and V4 loops as a target for potent and broad neutralization of simian immunodeficiency viruses. J Virol 2013; 87:5424-36. [PMID: 23468483 DOI: 10.1128/jvi.00201-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inducing neutralizing antibodies (NAb) is the key to developing a protective vaccine against human immunodeficiency virus type 1 (HIV-1). To clarify the neutralization mechanism of simian immunodeficiency virus (SIV), we analyzed NAb B404, which showed potent and broad neutralizing activity against various SIV strains. In 4 SIVsmH635FC-infected macaques, B404-like antibodies using the specific VH3 gene with a long complementarity-determining region 3 loop and λ light chain were the major NAbs in terms of the number and neutralizing potency. This biased NAb induction was observed in all 4 SIVsmH635FC-infected macaques but not in 2 macaques infected with a SIV mix, suggesting that induction of B404-like NAbs depended on the inoculated virus. Analysis using Env mutants revealed that the V3 and V4 loops were critical for B404 binding. The reactivity to the B404 epitope on trimeric, but not monomeric, Env was enhanced by CD4 ligation. The B404-resistant variant, which was induced by passages with increasing concentrations of B404, accumulated amino acid substitutions in the C2 region of gp120. Molecular dynamics simulations of the gp120 outer domains indicated that the C2 mutations could effectively alter the structural dynamics of the V3/V4 loops and their neighboring regions. These results suggest that a conformational epitope consisting of the V3 and V4 loops is the target for potent and broad neutralization of SIV. Identifying the new neutralizing epitope, as well as specifying the VH3 gene used for epitope recognition, will help to develop HIV-1 vaccines.
Collapse
|
31
|
Patel V, Jalah R, Kulkarni V, Valentin A, Rosati M, Alicea C, von Gegerfelt A, Huang W, Guan Y, Keele BF, Bess JW, Piatak M, Lifson JD, Williams WT, Shen X, Tomaras GD, Amara RR, Robinson HL, Johnson W, Broderick KE, Sardesai NY, Venzon DJ, Hirsch VM, Felber BK, Pavlakis GN. DNA and virus particle vaccination protects against acquisition and confers control of viremia upon heterologous simian immunodeficiency virus challenge. Proc Natl Acad Sci U S A 2013; 110:2975-80. [PMID: 23359688 PMCID: PMC3581900 DOI: 10.1073/pnas.1215393110] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We have previously shown that macaques vaccinated with DNA vectors expressing SIVmac239 antigens developed potent immune responses able to reduce viremia upon high-dose SIVmac251 challenge. To further improve vaccine-induced immunity and protection, we combined the SIVmac239 DNA vaccine with protein immunization using inactivated SIVmac239 viral particles as protein source. Twenty-six weeks after the last vaccination, the animals were challenged intrarectally at weekly intervals with a titrated dose of the heterologous SIVsmE660. Two of DNA-protein coimmunized macaques did not become infected after 14 challenges, but all controls were infected by 11 challenges. Vaccinated macaques showed modest protection from SIVsmE660 acquisition compared with naïve controls (P = 0.050; stratified for TRIM5α genotype). Vaccinees had significantly lower peak (1.6 log, P = 0.0048) and chronic phase viremia (P = 0.044), with 73% of the vaccinees suppressing viral replication to levels below assay detection during the 40-wk follow-up. Vaccine-induced immune responses associated significantly with virus control: binding antibody titers and the presence of rectal IgG to SIVsmE660 Env correlated with delayed SIVsmE660 acquisition; SIV-specific cytotoxic T cells, prechallenge CD4(+) effector memory, and postchallenge CD8(+) transitional memory cells correlated with control of viremia. Thus, SIVmac239 DNA and protein-based vaccine protocols were able to achieve high, persistent, broad, and effective cellular and humoral immune responses able to delay heterologous SIVsmE660 infection and to provide long-term control of viremia. These studies support a role of DNA and protein-based vaccines for development of an efficacious HIV/AIDS vaccine.
Collapse
Affiliation(s)
| | - Rashmi Jalah
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | | | | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | | - Wensheng Huang
- Department of Microbiology, and Immunology, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Yongjun Guan
- Department of Microbiology, and Immunology, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Julian W. Bess
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Michael Piatak
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Science Applications International Corporation-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | | | | | | - Rama R. Amara
- Yerkes National Primate Center, Emory University, Atlanta, GA 30329
| | | | | | | | | | - David J. Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20852; and
| | - Vanessa M. Hirsch
- Nonhuman Primate Virology Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20814
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | |
Collapse
|
32
|
|
33
|
Zhou X, Ramachandran S, Mann M, Popkin DL. Role of lymphocytic choriomeningitis virus (LCMV) in understanding viral immunology: past, present and future. Viruses 2012; 4:2650-69. [PMID: 23202498 PMCID: PMC3509666 DOI: 10.3390/v4112650] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 10/18/2012] [Accepted: 10/24/2012] [Indexed: 11/16/2022] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a common infection of rodents first identified over eighty years ago in St. Louis, MO, U.S.A. It is best known for its application in immunological studies. The history of LCMV closely correlates with the development of modern immunology. With the use of LCMV as a model pathogen several key concepts have emerged: Major Histocompatibility Complex (MHC) restriction, T cell memory, persistent infections, T cell exhaustion and the key role of immune pathology in disease. Given the phenomenal infrastructure within this field (e.g., defined immunodominant and subdominant epitopes to all T cell receptor specificities as well as the cognate tetramers for enumeration in vivo) the study of LCMV remains an active and productive platform for biological research across the globe to this day. Here we present a historical primer that highlights several breakthroughs since the discovery of LCMV. Next, we highlight current research in the field and conclude with our predictions for future directions in the remarkable field of LCMV research.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Dermatology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; (X.Z.); (S.R.); (M.M.)
| | - Srividya Ramachandran
- Department of Dermatology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; (X.Z.); (S.R.); (M.M.)
| | - Margaret Mann
- Department of Dermatology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; (X.Z.); (S.R.); (M.M.)
| | - Daniel L. Popkin
- Department of Dermatology, Pathology, Microbiology & Molecular Biology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
34
|
Klasse PJ, Moore JP. Good CoP, bad CoP? Interrogating the immune responses to primate lentiviral vaccines. Retrovirology 2012; 9:80. [PMID: 23025660 PMCID: PMC3484039 DOI: 10.1186/1742-4690-9-80] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 09/14/2012] [Indexed: 11/17/2022] Open
Abstract
Correlates of protection (CoPs) against infection by primate lentiviruses remain undefined. Modest protection against HIV-1 was observed in one human vaccine trial, whereas previous trials and vaccine-challenge experiments in non-human primates have yielded inconsistent but intriguing results. Although high levels of neutralizing antibodies are known to protect macaques from mucosal and intravenous viral challenges, antibody or other adaptive immune responses associated with protection might also be mere markers of innate immunity or susceptibility. Specific strategies for augmenting the design of both human trials and animal experiments could help to identify mechanistic correlates of protection and clarify the influences of confounding factors. Robust protection may, however, require the combined actions of immune responses and other host factors, thereby limiting what inferences can be drawn from statistical associations. Here, we discuss how to analyze immune protection against primate lentiviruses, and how host factors could influence both the elicitation and effectiveness of vaccine-induced responses.
Collapse
Affiliation(s)
- Per Johan Klasse
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornel University, 1300 York Avenue, Box 62, New York, NY 10065-4896, USA.
| | | |
Collapse
|