1
|
Rodrigues KA, Zhang YJ, Aung A, Morgan DM, Maiorino L, Yousefpour P, Gibson G, Ozorowski G, Gregory JR, Amlashi P, Buckley M, Ward AB, Schief WR, Love JC, Irvine DJ. Vaccines combining slow delivery and follicle targeting of antigens increase germinal center B cell clonal diversity and clonal expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608655. [PMID: 39229011 PMCID: PMC11370361 DOI: 10.1101/2024.08.19.608655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Vaccines incorporating slow delivery, multivalent antigen display, or immunomodulation through adjuvants have an important role to play in shaping the humoral immune response. Here we analyzed mechanisms of action of a clinically relevant combination adjuvant strategy, where phosphoserine (pSer)-tagged immunogens bound to aluminum hydroxide (alum) adjuvant (promoting prolonged antigen delivery to draining lymph nodes) are combined with a potent saponin nanoparticle adjuvant termed SMNP (which alters lymph flow and antigen entry into lymph nodes). When employed with a stabilized HIV Env trimer antigen in mice, this combined adjuvant approach promoted substantial enhancements in germinal center (GC) and antibody responses relative to either adjuvant alone. Using scRNA-seq and scBCR-seq, we found that the alum-pSer/SMNP combination both increased the diversity of GC B cell clones and increased GC B cell clonal expansion, coincident with increases in the expression of Myc and the proportion of S-phase GC B cells. To gain insight into the source of these changes in the GC response, we analyzed antigen biodistribution and structural integrity in draining lymph nodes and found that the combination adjuvant approach, but not alum-pSer delivery or SMNP alone, promoted accumulation of highly intact antigen on follicular dendritic cells, reflecting an integration of the slow antigen delivery and altered lymph node uptake effects of these two adjuvants. These results demonstrate how adjuvants with complementary mechanisms of action impacting vaccine biodistribution and kinetics can synergize to enhance humoral immunity.
Collapse
Affiliation(s)
- Kristen A. Rodrigues
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Harvard-MIT Health Sciences and Technology Program, Institute for Medical Engineering and Science; Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Yiming J. Zhang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Duncan M. Morgan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Department of Chemical Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Parisa Yousefpour
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Grace Gibson
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Gabriel Ozorowski
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Justin R. Gregory
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Parastoo Amlashi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Maureen Buckley
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Andrew B. Ward
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - William R. Schief
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - J. Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Department of Chemical Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Howard Hughes Medical Institute; Chevy Chase, MD 20815 USA
| |
Collapse
|
2
|
Letafati A, Bahari M, Salahi Ardekani O, Nayerain Jazi N, Nikzad A, norouzi F, Mahdavi B, Aboofazeli A, Mozhgani SH. HTLV-1 vaccination Landscape: Current developments and challenges. Vaccine X 2024; 19:100525. [PMID: 39105133 PMCID: PMC11298643 DOI: 10.1016/j.jvacx.2024.100525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/23/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is a retrovirus that is distinguished for its correlation to myelopathy/tropical spastic paraparesis (HAM/TSP) and adult T-cell leukemia/lymphoma (ATLL). As well, HTLV-1 has been documented to have links with other inflammatory diseases, such as uveitis and dermatitis. According to the World Health Organization (WHO), the global distribution of HTLV-1 infection is estimated to extend between 5 and 10 million individuals. Recent efforts in HTLV-1 vaccine development primarily involve selecting viral components, such as antigens, from structural and non-structural proteins. These components are chosen to trigger a vigorous immune response from cytotoxic T lymphocytes (CTLs), helper T lymphocytes (HTLs), and B cells. Investigation into developing a vaccine against HTLV-1 is ongoing, and current surveys have explored several approaches, including viral vector vaccines, DNA vaccines, protein and peptide vaccines, dendritic cell-based vaccines, mRNA vaccines, and other platforms. Despite these investigations have shown promising results, challenges like the necessity for long-term protective immunity, addressing viral diversity, and managing potential side effects remain. It is critical to keep track of the progress made in HTLV-1 vaccination research to comprehend the development status and its possible impacts. The evolving nature of vaccine development underscores the importance of staying informed about advancements as we strive to combat HTLV-1-associated diseases through effective vaccination strategies. In this review, our goal is to provide an overview of the current status of HTLV-1 vaccination efforts, emphasizing the progress, challenges, and potential future directions in this vital area of research.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mahshid Bahari
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Negar Nayerain Jazi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Abuzar Nikzad
- Dipartimento di Chimica Organica e Industriale Universita’ di Milano, Milan, Italy
| | - Farnaz norouzi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Bahar Mahdavi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Amir Aboofazeli
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
3
|
Webale SK, Kilongosi M, Munyekenye G, Onyango D, Marwa I, Bowen N. HIV-1 Transmission Cluster in Injection Drug Users in Nairobi City, Kenya. Ethiop J Health Sci 2023; 33:203-210. [PMID: 37484179 PMCID: PMC10358376 DOI: 10.4314/ejhs.v33i2.4] [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: 10/27/2022] [Accepted: 12/20/2022] [Indexed: 07/25/2023] Open
Abstract
Background While there is a striking increase in the prevalence of HIV in injection drug users, information on envelope-gene subtypes and transmission clusters in injection drug users is scarce. Method In a cross-sectional study, 247 injection drug users were recruited via out-rich method. Deoxyribonucleic acid was extracted from dry blood spot samples, amplified by Polymerase Chain Reaction and sequenced. Subtyping was performed using COntext-based Modeling for Expeditious Typing (COMET) and Recombinant Identification Program (RIP) tools. Phylogenetic diversity and Transmission clusters were identified using MEGA version 6.0 and TreeLink, respectively. Results Overall, 42 (17.0%) injection drug users were sero-positive for HIV-1. Of the 37 samples successfully sequenced, 29 (78.4%) sequences were identified as A1, 6 (16.2%) as AG while 1 (2.7%) as A1/G/AE and A1/C recombinants. The HIV subtypes formed clusters with little genetic diversity. Conclusion The high HIV prevalence was associated with transmission clusters and diversity in subtypes indicating ongoing local transmission. Therefore, there is need for comprehensive HIV care tailored to this population.
Collapse
Affiliation(s)
- Sella K Webale
- School of Biological sciences, Maseno University, Maseno, Kenya
| | - Mark Kilongosi
- School of Health Sciences, Kirinyaga University, Kutus, Kenya
| | | | - David Onyango
- School of Biological sciences, Maseno University, Maseno, Kenya
| | | | - Nancy Bowen
- National HIV Reference Laboratories, Ministry of Health, Nairobi city, Kenya
| |
Collapse
|
4
|
Santana CS, Andrade FDO, da Silva GCS, Nascimento JODS, Campos RF, Giovanetti M, Santos LA, Gois LL, Alcantara LCJ, Barreto FK. Advances in preventive vaccine development against HTLV-1 infection: A systematic review of the last 35 years. Front Immunol 2023; 14:1073779. [PMID: 36860854 PMCID: PMC9968880 DOI: 10.3389/fimmu.2023.1073779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Introduction The Human T-lymphotropic virus type 1 (HTLV-1) was the first described human retrovirus. It is currently estimated that around 5 to 10 million people worldwide are infected with this virus. Despite its high prevalence, there is still no preventive vaccine against the HTLV-1 infection. It is known that vaccine development and large-scale immunization play an important role in global public health. To understand the advances in this field we performed a systematic review regarding the current progress in the development of a preventive vaccine against the HTLV-1 infection. Methods This review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA®) guidelines and was registered at the International Prospective Register of Systematic Reviews (PROSPERO). The search for articles was performed in PubMed, Lilacs, Embase and SciELO databases. From the 2,485 articles identified, 25 were selected according to the inclusion and exclusion criteria. Results The analysis of these articles indicated that potential vaccine designs in development are available, although there is still a paucity of studies in the human clinical trial phase. Discussion Although HTLV-1 was discovered almost 40 years ago, it remains a great challenge and a worldwide neglected threat. The scarcity of funding contributes decisively to the inconclusiveness of the vaccine development. The data summarized here intends to highlight the necessity to improve the current knowledge of this neglected retrovirus, encouraging for more studies on vaccine development aiming the to eliminate this human threat. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier (CRD42021270412).
Collapse
Affiliation(s)
- Carolina Souza Santana
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | | | | | | | - Raissa Frazão Campos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | - Marta Giovanetti
- Laboratório de Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou - Fiocruz, Belo Horizonte, Minas Gerais, Brazil.,Department of Science and Technology for Humans and the Environment, University of Campus Bio-Medico di Roma, Rome, Italy
| | - Luciane Amorim Santos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Luana Leandro Gois
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Luiz Carlos Júnior Alcantara
- Laboratório de Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou - Fiocruz, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Khouri Barreto
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| |
Collapse
|
5
|
USP10 regulates B cell response to SARS-CoV-2 or HIV-1 nanoparticle vaccines through deubiquitinating AID. Signal Transduct Target Ther 2022; 7:7. [PMID: 34983926 PMCID: PMC8724756 DOI: 10.1038/s41392-021-00858-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/28/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Activation-induced cytidine deaminase (AID) initiates class-switch recombination and somatic hypermutation (SHM) in antibody genes. Protein expression and activity are tightly controlled by various mechanisms. However, it remains unknown whether a signal from the extracellular environment directly affects the AID activity in the nucleus where it works. Here, we demonstrated that a deubiquitinase USP10, which specifically stabilizes nuclear AID protein, can translocate into the nucleus after AKT-mediated phosphorylation at its T674 within the NLS domain. Interestingly, the signals from BCR and TLR1/2 synergistically promoted this phosphorylation. The deficiency of USP10 in B cells significantly decreased AID protein levels, subsequently reducing neutralizing antibody production after immunization with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or human immunodeficiency virus type 1 (HIV-1) nanoparticle vaccines. Collectively, we demonstrated that USP10 functions as an integrator for both BCR and TLR signals and directly regulates nuclear AID activity. Its manipulation could be used for the development of vaccines and adjuvants.
Collapse
|
6
|
Ciccozzi M, Lai A, Zehender G, Borsetti A, Cella E, Ciotti M, Sagnelli E, Sagnelli C, Angeletti S. The phylogenetic approach for viral infectious disease evolution and epidemiology: An updating review. J Med Virol 2019; 91:1707-1724. [PMID: 31243773 DOI: 10.1002/jmv.25526] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022]
Abstract
In the last decade, the phylogenetic approach is recurrent in molecular evolutionary analysis. On 12 May, 2019, about 2 296 213 papers are found, but typing "phylogeny" or "epidemiology AND phylogeny" only 199 804 and 20 133 are retrieved, respectively. Molecular epidemiology in infectious diseases is widely used to define the source of infection as so as the ancestral relationships of individuals sampled from a population. Coalescent theory and phylogeographic analysis have had scientific application in several, recent pandemic events, and nosocomial outbreaks. Hepatitis viruses and immunodeficiency virus (human immunodeficiency virus) have been largely studied. Phylogenetic analysis has been recently applied on Polyomaviruses so as in the more recent outbreaks due to different arboviruses type as Zika and chikungunya viruses discovering the source of infection and the geographic spread. Data on sequences isolated by the microorganism are essential to apply the phylogenetic tools and research in the field of infectious disease phylodinamics is growing up. There is the need to apply molecular phylogenetic and evolutionary methods in areas out of infectious diseases, as translational genomics and personalized medicine. Lastly, the application of these tools in vaccine strategy so as in antibiotic and antiviral researchers are encouraged.
Collapse
Affiliation(s)
- Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Alessia Lai
- Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan, Italy
| | - Gianguglielmo Zehender
- Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan, Italy
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Roma, Italy
| | - Eleonora Cella
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Marco Ciotti
- Laboratory of Molecular Virology, Polyclinic Tor Vergata Foundation, Rome, Italy
| | - Evangelista Sagnelli
- Department of Mental Health and Public Medicine, Section of Infectious Diseases, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Caterina Sagnelli
- Department of Mental Health and Public Medicine, Section of Infectious Diseases, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| |
Collapse
|
7
|
Bancroft T, DeBuysscher BL, Weidle C, Schwartz A, Wall A, Gray MD, Feng J, Steach HR, Fitzpatrick KS, Gewe MM, Skog PD, Doyle-Cooper C, Ota T, Strong RK, Nemazee D, Pancera M, Stamatatos L, McGuire AT, Taylor JJ. Detection and activation of HIV broadly neutralizing antibody precursor B cells using anti-idiotypes. J Exp Med 2019; 216:2331-2347. [PMID: 31345930 PMCID: PMC6780997 DOI: 10.1084/jem.20190164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/29/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023] Open
Abstract
Many tested vaccines fail to provide protection against disease despite the induction of antibodies that bind the pathogen of interest. In light of this, there is much interest in rationally designed subunit vaccines that direct the antibody response to protective epitopes. Here, we produced a panel of anti-idiotype antibodies able to specifically recognize the inferred germline version of the human immunodeficiency virus 1 (HIV-1) broadly neutralizing antibody b12 (iglb12). We determined the crystal structure of two anti-idiotypes in complex with iglb12 and used these anti-idiotypes to identify rare naive human B cells expressing B cell receptors with similarity to iglb12. Immunization with a multimerized version of this anti-idiotype induced the proliferation of transgenic murine B cells expressing the iglb12 heavy chain in vivo, despite the presence of deletion and anergy within this population. Together, our data indicate that anti-idiotypes are a valuable tool for the study and induction of potentially protective antibodies.
Collapse
Affiliation(s)
- Tara Bancroft
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Blair L DeBuysscher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Connor Weidle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Allison Schwartz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Abigail Wall
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Matthew D Gray
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Junli Feng
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Holly R Steach
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Kristin S Fitzpatrick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mesfin M Gewe
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Patrick D Skog
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA
| | - Colleen Doyle-Cooper
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA
| | - Takayuki Ota
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA
| | - Roland K Strong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - David Nemazee
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Leonidas Stamatatos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA .,Department of Global Health, University of Washington, Seattle, WA
| | - Andrew T McGuire
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA .,Department of Global Health, University of Washington, Seattle, WA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA .,Department of Global Health, University of Washington, Seattle, WA.,Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
8
|
Krivorutchenko YL, Nosik DN, Maligina VY, Lobach OA, Andronovskaja IB, Kirsanova MA, Grishkovets VI. CYTOTOXIC PROPERTIES OF TRITERPENE SAPONIN TAUROSID SX1 AND ITS EFFECT ON HUMAN IMMUNODEFICIENCY VIRUS AND INFLUENZA VIRUS INFECTION IN MICE. Vopr Virusol 2018; 63:123-129. [PMID: 36494938 DOI: 10.18821/0507-4088-2018-63-3-123-129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Indexed: 12/13/2022]
Abstract
Triterpene saponin Taurosid Sx1 purified from leaves of the plant Crimean Ivy Hedera taurica Carr. (Araliaceae) was evaluated for its cytotoxic activity against lymphoblastoid cell lines MT-4, Jurkat-tat, U937, and human peripheral blood monocytes. The ability of saponin to influence HIV-1 replication was studied as well. In addition, the ability of Taurosid Sx1 to increase survival of mice infected with influenza virus А/WSN/1/33(H1N1) and its capacity for strengthening the immune responses in mice immunized with the influenza vaccine Grippol® have been studied. Taurosid Sx1 has been shown to inhibit MT-4 cell line at 25 μg ml-1 concentration, IC50 33,3 μmol l-1 (MTT assay). The saponin concentration of 5 μg ml-1 was non-toxic for all the cell lines studied and demonstrated a moderate inhibitory effect on HIV p24 production in Jurkat-tat cells. In the lower concentrations Taurosid Sx1 did not stimulate HIV p24 production. It was shown that oral administration of 200 μg Taurosid Sx1 to the influenza virus infected mice caused 1.5-fold increase in their survival. Taurosid Sx1 given orally amplified immunopotentiating ability of an intramusculary administered subunit influenza vaccine. Antibody production was significantly higher in animals fed Taurosid Sx1 after primary or secondary immunizatuion. In mice given 2 doses of vaccine, from 1 to 3 weeks apart, feeding 200 μg saponin resulted in 2 to 10-fold enhancement in production of anti-H1, anti-H3, and anti-inluenza type B hemagglutinin antibodies. Thus, Taurosid Sx1 can be considered as low-toxic promising immunopotentiating agent uncapable of enhancing HIV-1 replication.
Collapse
Affiliation(s)
- Y L Krivorutchenko
- Medical Academy named after S.I. Georgievsky, Federal State Autonomous Educational Institution of Higher Education «V. I. Vernadsky Crimean Federal University»
| | - D N Nosik
- D.I. Ivanovsky Institute of Virology, «National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya»
| | - V Y Maligina
- Medical Academy named after S.I. Georgievsky, Federal State Autonomous Educational Institution of Higher Education «V. I. Vernadsky Crimean Federal University»
| | - O A Lobach
- D.I. Ivanovsky Institute of Virology, «National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya»
| | - I B Andronovskaja
- Medical Academy named after S.I. Georgievsky, Federal State Autonomous Educational Institution of Higher Education «V. I. Vernadsky Crimean Federal University»
| | - M A Kirsanova
- Medical Academy named after S.I. Georgievsky, Federal State Autonomous Educational Institution of Higher Education «V. I. Vernadsky Crimean Federal University»
| | - V I Grishkovets
- Medical Academy named after S.I. Georgievsky, Federal State Autonomous Educational Institution of Higher Education «V. I. Vernadsky Crimean Federal University»
| |
Collapse
|
9
|
Subcutaneous administration CpG-ODNs acts as a potent adjuvant for an HIV-1-tat-based vaccine candidate to elicit cellular immunity in BALB/c mice. Biotechnol Lett 2018; 40:527-533. [PMID: 29313255 DOI: 10.1007/s10529-017-2497-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 12/20/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To evaluate the combined effects of CpG oligodeoxynucleotides (CpG-ODNs) adjuvant and subcutaneous injection route on efficacy of a HIV-1-tat DNA vaccine candidate using BALB/c mice as an animal model. RESULTS Evaluation of cellular and humoral immunity of mice injected subcutaneously with HIV-1-tat gene cloned into a pcDNA3.1 vector indicated that significant levels of IFN-γ cytokine secretion (900 pg/ml), lymphocyte proliferation (2.5 stimulation index) and IgG2a (1.45 absorbance 450 nm) production could be achieved. These indicators of stimulated cellular immunity were elicited 2 weeks after the last injection (P < 0.05). CONCLUSIONS Formulation of HIV-1-tat DNA vaccine candidate with CpG-ODNs as an adjuvant while administrated subcutaneously are a promising approach to induce effective cellular immunity responses against HIV-1 infection.
Collapse
|
10
|
Ngu LN, Nji NN, Ambada G, Ngoh AA, Njambe Priso GD, Tchadji JC, Lissom A, Magagoum SH, Sake CN, Tchouangueu TF, Chukwuma GO, Okoli AS, Sagnia B, Chukwuanukwu R, Tebit DM, Esimone CO, Waffo AB, Park CG, Überla K, Nchinda GW. Dendritic cell targeted HIV-1 gag protein vaccine provides help to a recombinant Newcastle disease virus vectored vaccine including mobilization of protective CD8 + T cells. IMMUNITY INFLAMMATION AND DISEASE 2017; 6:163-175. [PMID: 29205929 PMCID: PMC5818444 DOI: 10.1002/iid3.209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
Abstract
Introduction Recombinant Newcastle Disease virus (rNDV) vectored vaccines are safe mucosal applicable vaccines with intrinsic immune‐modulatory properties for the induction of efficient immunity. Like all viral vectored vaccines repeated inoculation via mucosal routes invariably results to immunity against viral vaccine vectors. To obviate immunity against viral vaccine vectors and improve the ability of rNDV vectored vaccines in inducing T cell immunity in murine air way we have directed dendritic cell targeted HIV‐1 gag protein (DEC‐Gag) vaccine; for the induction of helper CD4+ T cells to a Recombinant Newcastle disease virus expressing codon optimized HIV‐1 Gag P55 (rNDV‐L‐Gag) vaccine. Methods We do so through successive administration of anti‐DEC205‐gagP24 protein plus polyICLC (DEC‐Gag) vaccine and rNDV‐L‐Gag. First strong gag specific helper CD4+ T cells are induced in mice by selected targeting of anti‐DEC205‐gagP24 protein vaccine to dendritic cells (DC) in situ together with polyICLC as adjuvant. This targeting helped T cell immunity develop to a subsequent rNDV‐L‐Gag vaccine and improved both systemic and mucosal gag specific immunity. Results This sequential DEC‐Gag vaccine prime followed by an rNDV‐L‐gag boost results to improved viral vectored immunization in murine airway, including mobilization of protective CD8+ T cells to a pathogenic virus infection site. Conclusion Thus, complementary prime boost vaccination, in which prime and boost favor distinct types of T cell immunity, improves viral vectored immunization, including mobilization of protective CD8+T cells to a pathogenic virus infection site such as the murine airway.
Collapse
Affiliation(s)
- Loveline N Ngu
- Department of Biochemistry, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon.,Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon
| | - Nadesh N Nji
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon
| | - Georgia Ambada
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Apeh A Ngoh
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of biomedical sciences, University of Dschang, Dschang, Cameroon
| | - Ghislain D Njambe Priso
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Jules C Tchadji
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Abel Lissom
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Suzanne H Magagoum
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Animal Biology and Physiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Carol N Sake
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Microbiology, University of Yaounde One, P.O. Box 812, Yaounde, Cameroon
| | - Thibau F Tchouangueu
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of biochemistry, University of Dschang, Dschang, Cameroon
| | - George O Chukwuma
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Medical Laboratory Science College of Medicine, Nnewi Campus, Nnamdi Azikiwe University, Awka, Anambra
| | | | - Bertrand Sagnia
- Microbiology and Immunology Laboratory, CIRCB, Yaounde, Cameroon
| | - Rebecca Chukwuanukwu
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Department of Medical Laboratory Science College of Medicine, Nnewi Campus, Nnamdi Azikiwe University, Awka, Anambra
| | - Denis M Tebit
- Myles Thaler Center for AIDS and Human Retrovirus Research, Department of Microbiology, Immunology and Cancer Biology, Jordan Hall 7088, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22903, USA
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| | - Alain B Waffo
- Department of Biological Sciences # 223, Alabama State University, 1627, Hall Street, Montgomery, Alabama 36104, USA
| | - Chae G Park
- Laboratory of Immunology, Brain Korea 21 PLUS Project for Medical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York 10065, USA
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Erlangen, Germany
| | - Godwin W Nchinda
- Laboratory of Vaccinology/Biobanking of The Chantal Biya International Reference Center for research on the prevention and management of HIV/AIDS (CIRCB), BP 3077, Messa Yaounde, Cameroon.,Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
11
|
Identification of HIV-1-Based Virus-like Particles by Multifrequency Atomic Force Microscopy. Biophys J 2017; 111:1173-1179. [PMID: 27653476 DOI: 10.1016/j.bpj.2016.07.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 11/21/2022] Open
Abstract
Virus-like particles (VLPs) have become a promising platform for vaccine production. VLPs are formed by structural viral proteins that inherently self-assemble when expressed in a host cell. They represent a highly immunogenic and safe vaccine platform, due to the absence of the viral genome and its high protein density. One of the most important parameters in vaccine production is the quality of the product. A related bottleneck in VLP-based products is the presence of cellular vesicles as a major contaminant in the preparations, which will require the set up of techniques allowing for specific discrimination of VLPs from host vesicular bodies. In this work novel, to our knowledge, multifrequency (MF) atomic force microscopy (AFM) has permitted full structural nanophysical characterization by its access to the virus capsid of the HIV-based VLPs. The assessment of these particles by advanced amplitude modulation-frequency modulation (AM-FM) viscoelastic mapping mode has enhanced the imaging resolution of their nanomechanical properties, opening a new window for the study of the biophysical attributes of VLPs. Finally, the identification and differentiation of HIV-based VLPs from cellular vesicles has been performed under ambient conditions, providing, to our knowledge, novel methodology for the monitoring and quality control of VLPs.
Collapse
|
12
|
Behrens AJ, Seabright GE, Crispin M. Targeting Glycans of HIV Envelope Glycoproteins for Vaccine Design. CHEMICAL BIOLOGY OF GLYCOPROTEINS 2017. [DOI: 10.1039/9781782623823-00300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The surface of the envelope spike of the human immunodeficiency virus (HIV) is covered with a dense array of glycans, which is sufficient to impede the host antibody response while maintaining a window for receptor recognition. The glycan density significantly exceeds that typically observed on self glycoproteins and is sufficiently high to disrupt the maturation process of glycans, from oligomannose- to complex-type glycosylation, that normally occurs during glycoprotein transit through the secretory system. It is notable that this generates a degree of homogeneity not seen in the highly mutated protein moiety. The conserved, close glycan packing and divergences from default glycan processing give a window for immune recognition. Encouragingly, in a subset of individuals, broadly neutralizing antibodies (bNAbs) have been isolated that recognize these features and are protective in passive-transfer models. Here, we review the recent advances in our understanding of the glycan shield of HIV and outline the strategies that are being pursued to elicit glycan-binding bNAbs by vaccination.
Collapse
Affiliation(s)
- Anna-Janina Behrens
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| | - Gemma E. Seabright
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| | - Max Crispin
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford South Parks Road Oxford OX1 3QU UK
| |
Collapse
|
13
|
Wang N, Yuan Z, Niu W, Li Q, Guo J. Synthetic biology approach for the development of conditionally replicating HIV-1 vaccine. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2017; 92:455-462. [PMID: 28983143 PMCID: PMC5624719 DOI: 10.1002/jctb.5174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
While the combined antiretroviral therapy has resulted in a significant decrease in HIV-1 related morbidity and mortality, the HIV-1 pandemic has not been substantially averted. To curtail the 2.4 million new infections each year, a prophylactic HIV-1 vaccine is urgently needed. This review first summarizes four major completed clinical efficacy trials of prophylactic HIV-1 vaccine and their outcomes. Next, it discusses several other approaches that have not yet advanced to clinical efficacy trials, but provided valuable insights into vaccine design. Among them, live-attenuated vaccines (LAVs) provided excellent protection in a non-human primate model. However, safety concerns have precluded the current version of LAVs from clinical application. As the major component of this review, two synthetic biology approaches for improving the safety of HIV-1 LAVs through controlling HIV-1 replication are discussed. Particular focus is on a novel approach that uses unnatural amino acid-mediated suppression of amber nonsense codon to generate conditionally replicating HIV-1 variants. The objective is to attract more attention towards this promising research field and to provoke creative designs and innovative utilization of the two control strategies.
Collapse
Affiliation(s)
- Nanxi Wang
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Zhe Yuan
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Wei Niu
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Qingsheng Li
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| |
Collapse
|
14
|
Musumeci G, Bon I, Lembo D, Cagno V, Re MC, Signoretto C, Diani E, Lopalco L, Pastori C, Martin L, Ponchel G, Gibellini D, Bouchemal K. M48U1 and Tenofovir combination synergistically inhibits HIV infection in activated PBMCs and human cervicovaginal histocultures. Sci Rep 2017; 7:41018. [PMID: 28145455 PMCID: PMC5286506 DOI: 10.1038/srep41018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/15/2016] [Indexed: 11/25/2022] Open
Abstract
Microbicides are considered a promising strategy for preventing human immunodeficiency virus (HIV-1) transmission and disease. In this report, we first analyzed the antiviral activity of the miniCD4 M48U1 peptide formulated in hydroxyethylcellulose (HEC) hydrogel in activated peripheral blood mononuclear cells (PBMCs) infected with R5- and X4–tropic HIV-1 strains. The results demonstrate that M48U1 prevented infection by several HIV-1 strains including laboratory strains, and HIV-1 subtype B and C strains isolated from the activated PBMCs of patients. M48U1 also inhibited infection by two HIV-1 transmitted/founder infectious molecular clones (pREJO.c/2864 and pTHRO.c/2626). In addition, M48U1 was administered in association with tenofovir, and these two antiretroviral drugs synergistically inhibited HIV-1 infection. In the next series of experiments, we tested M48U1 alone or in combination with tenofovir in HEC hydrogel with an organ-like structure mimicking human cervicovaginal tissue. We demonstrated a strong antiviral effect in absence of significant tissue toxicity. Together, these results indicate that co-treatment with M48U1 plus tenofovir is an effective antiviral strategy that may be used as a new topical microbicide to prevent HIV-1 transmission.
Collapse
Affiliation(s)
- Giuseppina Musumeci
- Department of Experimental, Diagnostics and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, 40138 Bologna, Italy
| | - Isabella Bon
- Department of Experimental, Diagnostics and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, 40138 Bologna, Italy
| | - David Lembo
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy
| | - Valeria Cagno
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy
| | - Maria Carla Re
- Department of Experimental, Diagnostics and Specialty Medicine (DIMES), Microbiology Section, University of Bologna, 40138 Bologna, Italy
| | - Caterina Signoretto
- Department of Diagnostics and Public Health, Microbiology and Virology Unit, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Erica Diani
- Department of Diagnostics and Public Health, Microbiology and Virology Unit, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Lucia Lopalco
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudia Pastori
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Loïc Martin
- CEA, iBiTecS, Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), Gif sur Yvette, F-91191, France
| | - Gilles Ponchel
- Institut Galien Paris Sud, CNRS UMR 8612, Univ. Paris-Sud, Université Paris Saclay, 5 rue J-B. Clément, 92296, Châtenay-Malabry cedex, France
| | - Davide Gibellini
- Department of Diagnostics and Public Health, Microbiology and Virology Unit, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Kawthar Bouchemal
- Institut Galien Paris Sud, CNRS UMR 8612, Univ. Paris-Sud, Université Paris Saclay, 5 rue J-B. Clément, 92296, Châtenay-Malabry cedex, France
| |
Collapse
|
15
|
Escolano A, Steichen JM, Dosenovic P, Kulp DW, Golijanin J, Sok D, Freund NT, Gitlin AD, Oliveira T, Araki T, Lowe S, Chen ST, Heinemann J, Yao KH, Georgeson E, Saye-Francisco KL, Gazumyan A, Adachi Y, Kubitz M, Burton DR, Schief WR, Nussenzweig MC. Sequential Immunization Elicits Broadly Neutralizing Anti-HIV-1 Antibodies in Ig Knockin Mice. Cell 2016; 166:1445-1458.e12. [PMID: 27610569 DOI: 10.1016/j.cell.2016.07.030] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/20/2016] [Indexed: 12/14/2022]
Abstract
A vaccine that elicits broadly neutralizing antibodies (bNAbs) against HIV-1 is likely to be protective, but this has not been achieved. To explore immunization regimens that might elicit bNAbs, we produced and immunized mice expressing the predicted germline PGT121, a bNAb specific for the V3-loop and surrounding glycans on the HIV-1 spike. Priming with an epitope-modified immunogen designed to activate germline antibody-expressing B cells, followed by ELISA-guided boosting with a sequence of directional immunogens, native-like trimers with decreasing epitope modification, elicited heterologous tier-2-neutralizing responses. In contrast, repeated immunization with the priming immunogen did not. Antibody cloning confirmed elicitation of high levels of somatic mutation and tier-2-neutralizing antibodies resembling the authentic human bNAb. Our data establish that sequential immunization with specifically designed immunogens can induce high levels of somatic mutation and shepherd antibody maturation to produce bNAbs from their inferred germline precursors.
Collapse
Affiliation(s)
- Amelia Escolano
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Jon M Steichen
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Pia Dosenovic
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Daniel W Kulp
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jovana Golijanin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Devin Sok
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; International AIDS Vaccine Initiative, New York, NY 10004, USA
| | - Natalia T Freund
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Alexander D Gitlin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Thiago Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Tatsuya Araki
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Sarina Lowe
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Spencer T Chen
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Jennifer Heinemann
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Kai-Hui Yao
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Erik Georgeson
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Karen L Saye-Francisco
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Yumiko Adachi
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael Kubitz
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - William R Schief
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology and Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
16
|
Pfaender S, von Hahn T, Steinmann J, Ciesek S, Steinmann E. Prevention strategies for blood-borne viruses-in the Era of vaccines, direct acting antivirals and antiretroviral therapy. Rev Med Virol 2016; 26:330-9. [PMID: 27185010 PMCID: PMC5084801 DOI: 10.1002/rmv.1890] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 12/21/2022]
Abstract
Blood-borne viruses, such as hepatitis B virus, hepatitis C virus, human immunodeficiency virus, and the facultative blood-borne hepatitis E virus, are considered a major public health problem given that they are accountable for millions of deaths each year. Treatment options, including effective vaccine design, development of antiviral strategies and the implementation of antiretroviral therapy have improved substantially over the last couple of years and contribute to successful treatment and prevention of these infectious diseases. In this review, we summarise the current knowledge and concepts in prevention of transmission of these blood-borne viruses.
Collapse
Affiliation(s)
- Stephanie Pfaender
- Institute for Experimental Virology, Twincore, and Hannover Medical School Hannover, Germany, and Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Infectious Diseases and Pathobiology, University of Bern, 3012 Bern, Switzerland, Federal Department of Home Affairs, Institute of Virology and Immunology, 3012 Bern and 3147, Mittelhäusern, Switzerland
| | - Thomas von Hahn
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany
- German Center for Infection Research (DZIF) - Hannover-Braunschweig Site
- Institute for Molecular Biology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Joerg Steinmann
- Institute of Medical Microbiology, University Hospital Essen, Essen, Germany
| | - Sandra Ciesek
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eike Steinmann
- Institute for Experimental Virology, Twincore, and Hannover Medical School Hannover, Germany, and Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
17
|
Ji Z, Xie Z, Wang Q, Zhang Z, Gong T, Sun X. A Prime-Boost Strategy Combining Intravaginal and Intramuscular Administration of Homologous Adenovirus to Enhance Immune Response Against HIV-1 in Mice. Hum Gene Ther 2016; 27:219-29. [PMID: 26715124 DOI: 10.1089/hum.2015.153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Immune responses to HIV in the vaginal tract effectively trigger both systemic and mucosal protection, providing a double layer of defense. However, recombinant adenoviral (rAd) vectors delivered intravaginally do not effectively penetrate the mucus layer and vaginal epithelium, and instead are rapidly cleared. To overcome these barriers, we previously synthesized a novel cationic polyethylene glycol derivative that can self-assemble into nanocomplexes with rAd. These nanocomplexes can help rAd bypass the mucus layer and enhance mucosal immune response to the encoded antigen. However, the resulting cellular and humoral responses were still lower than those elicited by single intramuscular injection of rAd. Therefore, in the present study we investigated a new vaccination strategy involving intravaginal priming with our nanocomplexes, followed by an intramuscular boost with rAd-gag. Mice immunized in this way showed more potent humoral and cellular responses, as well as higher IgA levels, than animals primed and boosted intravaginally with nanocomplexes. These results show the promise of a prime-boost strategy for developing vaccine candidates against HIV.
Collapse
Affiliation(s)
- Zhonghua Ji
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, West China School of Pharmacy, Sichuan University , Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Zhaolu Xie
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, West China School of Pharmacy, Sichuan University , Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Qin Wang
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, West China School of Pharmacy, Sichuan University , Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, West China School of Pharmacy, Sichuan University , Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, West China School of Pharmacy, Sichuan University , Ministry of Education, Chengdu, Sichuan, People's Republic of China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Novel Drug Delivery Systems, West China School of Pharmacy, Sichuan University , Ministry of Education, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
18
|
Abstract
Vaccination is one of the cheapest health-care interventions that have saved more lives than any other drugs or therapies. Due to successful immunization programs we rarely hear about some of the common diseases of the early twentieth century including small pox and polio. Vaccination programs have also helped to increase food production notably poultry, cattle, and milk production due to lower incidence of infectious diseases in farm animals. Though vaccination programs have eradicated several diseases and increased the quality of life there are several diseases that have no effective vaccines. Currently there are no vaccines for cancer, neurodegenerative diseases, autoimmune diseases, as well as infectious diseases like tuberculosis, AIDS, and parasitic diseases including malaria. Abuse of antibiotics has resulted in the generation of several antibiotic-resistant bacterial strains; hence there is a need to develop novel vaccines for antibiotic-resistant microorganisms. Changes in climate is another concern for vaccinologists. Climate change could lead to generation of new strains of infectious microorganisms that would require development of novel vaccines. Use of conventional vaccination strategies to develop vaccines has severe limitations; hence innovative strategies are essential in the development of novel and effective vaccines.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA.
| | - Rima Dilbarova
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA.,College of Arts and Sciences, Drexel University, Philadelphia, PA, 19104, USA
| | | |
Collapse
|
19
|
Walsh SR, Moodie Z, Fiore-Gartland AJ, Morgan C, Wilck MB, Hammer SM, Buchbinder SP, Kalams SA, Goepfert PA, Mulligan MJ, Keefer MC, Baden LR, Swann EM, Grant S, Ahmed H, Li F, Hertz T, Self SG, Friedrich D, Frahm N, Liao HX, Montefiori DC, Tomaras GD, McElrath MJ, Hural J, Graham BS, Jin X. Vaccination With Heterologous HIV-1 Envelope Sequences and Heterologous Adenovirus Vectors Increases T-Cell Responses to Conserved Regions: HVTN 083. J Infect Dis 2015; 213:541-50. [PMID: 26475930 DOI: 10.1093/infdis/jiv496] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/09/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increasing the breadth of human immunodeficiency virus type 1 (HIV-1) vaccine-elicited immune responses or targeting conserved regions may improve coverage of circulating strains. HIV Vaccine Trials Network 083 tested whether cellular immune responses with these features are induced by prime-boost strategies, using heterologous vectors, heterologous inserts, or a combination of both. METHODS A total of 180 participants were randomly assigned to receive combinations of adenovirus vectors (Ad5 or Ad35) and HIV-1 envelope (Env) gene inserts (clade A or B) in a prime-boost regimen. RESULTS T-cell responses to heterologous and homologous insert regimens targeted a similar number of epitopes (ratio of means, 1.0; 95% confidence interval [CI], .6-1.6; P = .91), but heterologous insert regimens induced significantly more epitopes that were shared between EnvA and EnvB than homologous insert regimens (ratio of means, 2.7; 95% CI, 1.2-5.7; P = .01). Participants in the heterologous versus homologous insert groups had T-cell responses that targeted epitopes with greater evolutionary conservation (mean entropy [±SD], 0.32 ± 0.1 bits; P = .003), and epitopes recognized by responders provided higher coverage (49%; P = .035). Heterologous vector regimens had higher numbers of total, EnvA, and EnvB epitopes than homologous vector regimens (P = .02, .044, and .045, respectively). CONCLUSIONS These data demonstrate that vaccination with heterologous insert prime boosting increased T-cell responses to shared epitopes, while heterologous vector prime boosting increased the number of T-cell epitopes recognized. CLINICAL TRIALS REGISTRATION NCT01095224.
Collapse
Affiliation(s)
- Stephen R Walsh
- Division of Infectious Diseases, Brigham and Women's Hospital Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, Massachusetts
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | | | - Cecilia Morgan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Marissa B Wilck
- Division of Infectious Diseases, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts
| | | | | | - Spyros A Kalams
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | | | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts
| | | | - Shannon Grant
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Hasan Ahmed
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Fusheng Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Tomer Hertz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Steven G Self
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - David Friedrich
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center Department of Global Health, University of Washington, Seattle
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina
| | | | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center Department of Global Health, University of Washington, Seattle Departments of Medicine and Laboratory Medicine, University of Washington, Seattle
| | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Barney S Graham
- Dale and Betty Bumpers Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Xia Jin
- University of Rochester, New York
| | | |
Collapse
|
20
|
Voellmy R, Bloom DC, Vilaboa N. A novel approach for addressing diseases not yielding to effective vaccination? Immunization by replication-competent controlled virus. Expert Rev Vaccines 2015; 14:637-51. [PMID: 25676927 DOI: 10.1586/14760584.2015.1013941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Vaccination involves inoculation of a subject with a disabled disease-causing microbe or parts thereof. While vaccination has been highly successful, we still lack sufficiently effective vaccines for important infectious diseases. We propose that a more complete immune response than that elicited from a vaccine may be obtained from immunization with a disease-causing virus modified to subject replication-essential genes to the control of a gene switch activated by non-lethal heat in the presence of a drug-like compound. Upon inoculation, strictly localized replication of the virus would be triggered by a heat dose administered to the inoculation site. Activated virus would transiently replicate with an efficiency approaching that of the disease-causing virus and express all viral antigens. It may also vector heterologous antigens or control co-infecting microbes.
Collapse
Affiliation(s)
- Richard Voellmy
- Department of Physiological Sciences, University of Florida College of Veterinary Sciences, Gainesville, FL, USA
| | | | | |
Collapse
|
21
|
Hassapis KA, Stylianou DC, Kostrikis LG. Architectural insight into inovirus-associated vectors (IAVs) and development of IAV-based vaccines inducing humoral and cellular responses: implications in HIV-1 vaccines. Viruses 2014; 6:5047-76. [PMID: 25525909 PMCID: PMC4276942 DOI: 10.3390/v6125047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 12/21/2022] Open
Abstract
Inovirus-associated vectors (IAVs) are engineered, non-lytic, filamentous bacteriophages that are assembled primarily from thousands of copies of the major coat protein gp8 and just five copies of each of the four minor coat proteins gp3, gp6, gp7 and gp9. Inovirus display studies have shown that the architecture of inoviruses makes all coat proteins of the inoviral particle accessible to the outside. This particular feature of IAVs allows foreign antigenic peptides to be displayed on the outer surface of the virion fused to its coat proteins and for more than two decades has been exploited in many applications including antibody or peptide display libraries, drug design, and vaccine development against infectious and non-infectious diseases. As vaccine carriers, IAVs have been shown to elicit both a cellular and humoral response against various pathogens through the display of antibody epitopes on their coat proteins. Despite their high immunogenicity, the goal of developing an effective vaccine against HIV-1 has not yet materialized. One possible limitation of previous efforts was the use of broadly neutralizing antibodies, which exhibited autoreactivity properties. In the past five years, however, new, more potent broadly neutralizing antibodies that do not exhibit autoreactivity properties have been isolated from HIV-1 infected individuals, suggesting that vaccination strategies aimed at producing such broadly neutralizing antibodies may confer protection against infection. The utilization of these new, broadly neutralizing antibodies in combination with the architectural traits of IAVs have driven the current developments in the design of an inovirus-based vaccine against HIV-1. This article reviews the applications of IAVs in vaccine development, with particular emphasis on the design of inoviral-based vaccines against HIV-1.
Collapse
Affiliation(s)
- Kyriakos A Hassapis
- Department of Biological Sciences, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus.
| | - Dora C Stylianou
- Department of Biological Sciences, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus.
| | - Leondios G Kostrikis
- Department of Biological Sciences, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus.
| |
Collapse
|
22
|
Abstract
Although some success was achieved in recent years in HIV prevention, an effective vaccine remains the means with the most potential of curtailing HIV-1 infections worldwide. Despite multiple failed attempts, a recent HIV vaccine regimen demonstrated modest protection from infection. Although the protective efficacy in this trial was not sufficient to warrant licensure, it spurred renewed optimism in the field and has provided valuable insights for improving future vaccine designs. This review summarizes the pertinent details of vaccine development and discusses ways the field is moving forward to develop a vaccine to prevent HIV infection and disease progression.
Collapse
Affiliation(s)
- Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 908, 20th Street South, CCB 328, Birmingham, AL 35294, USA.
| | - Anju Bansal
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 845, 19th Street South, BBRB 557, Birmingham, AL 35294, USA
| |
Collapse
|
23
|
Schwartz JL. Fcgbp - A Potential Viral Trap in RV144. Open AIDS J 2014; 8:21-4. [PMID: 25246998 PMCID: PMC4166788 DOI: 10.2174/1874613601408010021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/08/2014] [Accepted: 07/10/2014] [Indexed: 12/22/2022] Open
Abstract
Years of extensive research have yielded much knowledge in many aspects of HIV-1 infection, treatments, and education. However, without a vaccine, the number of people infected worldwide continues to grow. The partial success of the Thai RV144 vaccine trial provides hope that a method of protection is indeed possible. Understanding the mechanism behind the protection is critical if we hope to achieve our goal of inhibiting new infections of HIV-1. We hypothesize that the Fc of IgG binding protein (Fcgbp) is associated with the protection observed in the RV144 vaccine trial. It has the ability to trap viral-antibody complexes in the mucosa by binding the Fc of IgG to Fcgbp. This property could be used in the form of a microbicide containing antibodies to a variety of HIV-1 epitopes to prevent sexual transmission of HIV-1. The aim of this paper is to stimulate further research into Fcgbp and its role in innate immunity.
Collapse
Affiliation(s)
- Jacquelyn L Schwartz
- Department of Physiology, University of Manitoba, 745 Bannatyne Ave., Winnipeg, Manitoba, R3E 0J9, Canada
| |
Collapse
|
24
|
Direct interrogation of viral peptides presented by the class I HLA of HIV-infected T cells. J Virol 2014; 88:12992-3004. [PMID: 25165114 DOI: 10.1128/jvi.01914-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Identification of CD8(+) cytotoxic T lymphocyte (CTL) epitopes has traditionally relied upon testing of overlapping peptide libraries for their reactivity with T cells in vitro. Here, we pursued deep ligand sequencing (DLS) as an alternative method of directly identifying those ligands that are epitopes presented to CTLs by the class I human leukocyte antigens (HLA) of infected cells. Soluble class I HLA-A*11:01 (sHLA) was gathered from HIV-1 NL4-3-infected human CD4(+) SUP-T1 cells. HLA-A*11:01 harvested from infected cells was immunoaffinity purified and acid boiled to release heavy and light chains from peptide ligands that were then recovered by size-exclusion filtration. The ligands were first fractionated by high-pH high-pressure liquid chromatography and then subjected to separation by nano-liquid chromatography (nano-LC)-mass spectrometry (MS) at low pH. Approximately 10 million ions were selected for sequencing by tandem mass spectrometry (MS/MS). HLA-A*11:01 ligand sequences were determined with PEAKS software and confirmed by comparison to spectra generated from synthetic peptides. DLS identified 42 viral ligands presented by HLA-A*11:01, and 37 of these were previously undetected. These data demonstrate that (i) HIV-1 Gag and Nef are extensively sampled, (ii) ligand length variants are prevalent, particularly within Gag and Nef hot spots where ligand sequences overlap, (iii) noncanonical ligands are T cell reactive, and (iv) HIV-1 ligands are derived from de novo synthesis rather than endocytic sampling. Next-generation immunotherapies must factor these nascent HIV-1 ligand length variants and the finding that CTL-reactive epitopes may be absent during infection of CD4(+) T cells into strategies designed to enhance T cell immunity. IMPORTANCE HIV-1 epitopes catalogued by the Los Alamos National Laboratory (LANL) have yielded limited success in vaccine trials. Because the HLA of infected cells have not previously been assessed for HIV-1 ligands, the objective here was to directly characterize the viral ligands that mark infected cells. Recovery of HLA-presented peptides from HIV-1-infected CD4(+) T cells and interrogation of the peptide cargo by mass spectrometric DLS show that typical and atypical viral ligands are efficiently presented by HLA and targeted by human CTLs. Nef and Gag ligands dominate the infected cell's antigenic profile, largely due to extensive ligand sampling from select hot spots within these viral proteins. Also, HIV-1 ligands are often longer than expected, and these length variants are quite antigenic. These findings emphasize that an HLA-based view of HIV-1 ligand presentation to CTLs provides previously unrealized information that may enhance the development of immune therapies and vaccines.
Collapse
|
25
|
Xie Z, Ji Z, Zhang Z, Gong T, Sun X. Adenoviral vectors coated with cationic PEG derivatives for intravaginal vaccination against HIV-1. Biomaterials 2014; 35:7896-908. [PMID: 24929620 DOI: 10.1016/j.biomaterials.2014.05.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/20/2014] [Indexed: 01/24/2023]
Abstract
Mucus layer coating the vaginal epithelium represents a barrier for intravaginally delivered recombined adenoviral (rAd) vectors, but it could be overcome by proper polyethylene glycol (PEG) modification. Here we synthesized two cationic PEG derivatives, amino-(EO)n/(AGE)m-Cyss (APCs). The polymers contained neutral linear PEG (2-5 kDa) to provide a hydrophilic surface and amine pendants to provide positive charge for coating negatively charged rAd by physical adsorption. Given proper molecular composition, the polymer (5k-APC) could coat rAd without causing aggregation, facilitating its mucus penetrating ability and enhancing gene expression both in vitro and in vivo. With HIVgag as the model antigen, the polymer-rAd complexes were administered intravaginally to elicit both systemic and mucosal immune responses. 5k-APC-rAd immunization elicited robust HIVgag-specific cellular responses and also induced higher antigen-specific serum IgG. More importantly, mice immunized with 5k-APC-rAd showed higher level of IgA in vaginal lavage fluid. These findings suggest that 5k-APC-rAd is a promising system for intravaginal immunization against infectious diseases such as HIV within the vaginal tract.
Collapse
Affiliation(s)
- Zhaolu Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhonghua Ji
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, PR China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, PR China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|