1
|
Li B, Zhang J, He T, Yuan H, Wu H, Wang P, Wu C. PRR adjuvants restrain high stability peptides presentation on APCs. eLife 2024; 13:RP99173. [PMID: 39475096 PMCID: PMC11524579 DOI: 10.7554/elife.99173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Adjuvants can affect APCs function and boost adaptive immune responses post-vaccination. However, whether they modulate the specificity of immune responses, particularly immunodominant epitope responses, and the mechanisms of regulating antigen processing and presentation remain poorly defined. Here, using overlapping synthetic peptides, we screened the dominant epitopes of Th1 responses in mice post-vaccination with different adjuvants and found that the adjuvants altered the antigen-specific CD4+ T-cell immunodominant epitope hierarchy. MHC-II immunopeptidomes demonstrated that the peptide repertoires presented by APCs were significantly altered by the adjuvants. Unexpectedly, no novel peptide presentation was detected after adjuvant treatment, whereas peptides with high binding stability for MHC-II presented in the control group were missing after adjuvant stimulation, particularly in the MPLA- and CpG-stimulated groups. The low-stability peptide present in the adjuvant groups effectively elicited robust T-cell responses and formed immune memory. Collectively, our results suggest that adjuvants (MPLA and CpG) inhibit high-stability peptide presentation instead of revealing cryptic epitopes, which may alter the specificity of CD4+ T-cell-dominant epitope responses. The capacity of adjuvants to modify peptide-MHC (pMHC) stability and antigen-specific T-cell immunodominant epitope responses has fundamental implications for the selection of suitable adjuvants in the vaccine design process and epitope vaccine development.
Collapse
Affiliation(s)
- Bin Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| | - Jin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| | - Taojun He
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| | - Hanmei Yuan
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| | - Hui Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen UniversityShenzhenChina
| |
Collapse
|
2
|
Piccaro G, Aquino G, Gigantino V, Tirelli V, Sanchez M, Iorio E, Matarese G, Cassone A, Palma C. Mycobacterium tuberculosis antigen 85B modifies BCG-induced antituberculosis immunity and favors pathogen survival. J Leukoc Biol 2024; 115:1053-1069. [PMID: 38242866 DOI: 10.1093/jleuko/qiae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Tuberculosis is one of the deadliest infectious diseases worldwide. Mycobacterium tuberculosis has developed strategies not only to evade host immunity but also to manipulate it for its survival. We investigated whether Mycobacterium tuberculosis exploited the immunogenicity of Ag85B, one of its major secretory proteins, to redirect host antituberculosis immunity to its advantage. We found that administration of Ag85B protein to mice vaccinated with Bacillus Calmette-Guérin impaired the protection elicited by vaccination, causing a more severe infection when mice were challenged with Mycobacterium tuberculosis. Ag85B administration reduced Bacillus Calmette-Guérin-induced CD4 T-cell activation and IFN-γ, CCL-4, and IL-22 production in response to Mycobacterium tuberculosis-infected cells. On the other hand, it promoted robust Ag85B-responsive IFN-γ-producing CD4 T cells, expansion of a subset of IFN-γ/IL-10-producing CD4+FOXP3+Treg cells, differential activation of IL-17/IL-22 responses, and activation of regulatory and exhaustion pathways, including programmed death ligand 1 expression on macrophages. All this resulted in impaired intracellular Mycobacterium tuberculosis growth control by systemic immunity, both before and after the Mycobacterium tuberculosis challenge. Interestingly, Mycobacterium tuberculosis infection itself generated Ag85B-reactive inflammatory immune cells incapable of clearing Mycobacterium tuberculosis in both unvaccinated and Bacillus Calmette-Guérin-vaccinated mice. Our data suggest that Mycobacterium tuberculosis can exploit the strong immunogenicity of Ag85B to promote its own survival and spread. Since Ag85B is normally secreted by replicating bacteria and is commonly found in the lungs of the Mycobacterium tuberculosis-infected host, our findings may advance the understanding on the mechanisms of Mycobacterium tuberculosis pathogenesis and immune evasion.
Collapse
Affiliation(s)
- Giovanni Piccaro
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Valentina Tirelli
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Massimo Sanchez
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Egidio Iorio
- Core Facilities-High Resolution NMR Unit, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie mediche, Università di Napoli "Federico II," Via Sergio Pansini 5, 80131 Naples, Italy
| | - Antonio Cassone
- Polo d'innovazione della Genomica, Genetica e Biologia, Via Fiorentina 1, 53100 Siena, Italy
| | - Carla Palma
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
3
|
Next-Generation Bioinformatics Approaches and Resources for Coronavirus Vaccine Discovery and Development-A Perspective Review. Vaccines (Basel) 2021; 9:vaccines9080812. [PMID: 34451937 PMCID: PMC8402397 DOI: 10.3390/vaccines9080812] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022] Open
Abstract
COVID-19 is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To fight this pandemic, which has caused a massive death toll around the globe, researchers are putting efforts into developing an effective vaccine against the pathogen. As genome sequencing projects for several coronavirus strains have been completed, a detailed investigation of the functions of the proteins and their 3D structures has gained increasing attention. These high throughput data are a valuable resource for accelerating the emerging field of immuno-informatics, which is primarily aimed toward the identification of potential antigenic epitopes in viral proteins that can be targeted for the development of a vaccine construct eliciting a high immune response. Bioinformatics platforms and various computational tools and databases are also essential for the identification of promising vaccine targets making the best use of genomic resources, for further experimental validation. The present review focuses on the various stages of the vaccine development process and the vaccines available for COVID-19. Additionally, recent advances in genomic platforms and publicly available bioinformatics resources in coronavirus vaccine discovery together with related immunoinformatics databases and advances in technology are discussed.
Collapse
|
4
|
Comprehensive Screening of Mouse T-Cell Epitopes in Human Herpesvirus 6B Glycoprotein H/L/Q1/Q2 Tetramer Complex. J Immunol Res 2020; 2020:4697529. [PMID: 32775465 PMCID: PMC7399772 DOI: 10.1155/2020/4697529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 11/21/2022] Open
Abstract
Human herpesvirus 6 (HHV-6) infects over 90% of people. The HHV-6 subtype, HHV-6B in particular, is often associated with exanthem subitum in early childhood. Exanthem subitum is usually self-limiting and good prognosis disease; however, some infants primarily infected with HHV-6B develop encephalitis/encephalopathy, and half of the patients developed encephalopathy reported to have neurological sequelae. Furthermore, after primary infection, HHV-6B remains in a latent state and sometimes reactivated in immunosuppressed patients, causing life-threatening severe encephalopathy. However, effective immunotherapies or vaccines for controlling HHV-6B infection and reactivation have not yet been established. Recently, we have found that the HHV-6B tetrameric glycoprotein (g) complex, gH/gL/gQ1/gQ2 is a promising vaccine candidate, and currently under preclinical development. To confirm our vaccine candidate protein complex induce detectable T-cell responses, in this study, we comprehensively screened CD4+ and CD8+ T-cell epitopes in the gH/gL/gQ1/gQ2 tetrameric complex protein in mice immunisation model. Both BALB/c and C57BL/6 mice were immunised with the tetrameric complex protein or plasmid DNA encoding gH, gL, gQ1, and gQ2, and then restimulated with 162 20-mer peptides covering the whole gH/gL/gQ1/gQ2 sequences; multiple CD4+ and CD8+ T-cell-stimulating peptides were identified in both BALB/c and C57BL/6 mice. Our study demonstrates that gH/gL/gQ1/gQ2 tetramer-targeted vaccination has potential to induce T-cell responses in two different strains of mice and supports the future development and application of T-cell-inducing vaccine and immunotherapies against HHV-6B.
Collapse
|
5
|
Pastore G, Carraro M, Pettini E, Nolfi E, Medaglini D, Ciabattini A. Optimized Protocol for the Detection of Multifunctional Epitope-Specific CD4 + T Cells Combining MHC-II Tetramer and Intracellular Cytokine Staining Technologies. Front Immunol 2019; 10:2304. [PMID: 31649661 PMCID: PMC6794358 DOI: 10.3389/fimmu.2019.02304] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
Analysis of multifunctional CD4+ T cells is fundamental for characterizing the immune responses to vaccination or infection. Major histocompatibility complex (MHC)/peptide tetramers represent a powerful technology for the detection of antigen-specific T cells by specific binding to their T-cell receptor, and their combination with functional assays is fundamental for characterizing the antigen-specific immune response. Here we optimized a protocol for the detection of multiple intracellular cytokines within epitope-specific CD4+ T cells identified by the MHC class II tetramer technology. The optimal procedure for assessing the functional activity of tetramer-binding CD4+ T cells was based on the simultaneous intracellular staining with both MHC tetramers and cytokine-specific antibodies upon in vitro restimulation of cells with the vaccine antigen. The protocol was selected among procedures that differently combine the steps of cellular restimulation and tetramer staining with intracellular cytokine labeling. This method can be applied to better understand the complex functional profile of CD4+ T-cell responses upon vaccination or infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Abebe F. Synergy between Th1 and Th2 responses during Mycobacterium tuberculosis infection: A review of current understanding. Int Rev Immunol 2019; 38:172-179. [PMID: 31244354 DOI: 10.1080/08830185.2019.1632842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Induction of Th1 (cell-mediated) immunity and associated production of IFN-γ by CD4+ T cells has been widely used as a marker of protective immunity against tuberculosis (TB). This is based on two assumptions. The first is the widely accepted view that Mycobacterium tuberculosis (Mtb), the causative agent of TB is an obligate intracellular pathogen, and the second is based on the Th1/Th2 paradigm, which posits that polarization of CD4+ T cells into type1 (cell-mediated) and type 2 (humoral) is central for proper induction of protective immunity against pathogens. However, almost all licensed vaccines currently in use are primarily anti-body based whether intracellular or extra-cellular. In addition, converging data from both animal models and humans indicate that the production of IFN-γ alone is not sufficient to confer protection against TB. In addition, a substantial body of the literature suggests that, in addition to Th1 cells, antibody classes and sub-classes are protective against TB. In a recent study, we have shown that there is a synergy between IFN-γ (cell-mediated) and IgA (humoral) in human population in an endemic setting. In this review, current data from both animal and human studies that support mixed Th1 and Th2 responses that are protective against Mtb and other pathogens are presented.
Collapse
Affiliation(s)
- Fekadu Abebe
- University of Oslo, Faculty of Medicine, Institute of Health and Society , Oslo , Norway
| |
Collapse
|
7
|
Garnica O, Das K, Devasundaram S, Dhandayuthapani S. Enhanced delivery of Mycobacterium tuberculosis antigens to antigen presenting cells using RVG peptide. Tuberculosis (Edinb) 2019; 116S:S34-S41. [PMID: 31064713 DOI: 10.1016/j.tube.2019.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 10/26/2022]
Abstract
Among the various strategies to improve vaccines against infectious diseases, targeting of antigens to dendritic cells (DCs), which are professional antigen presenting cells (APCs), has received increased attention in recent years. Here, we investigated whether a synthetic peptide region named RVG, originated from Rabies Virus Glycoprotein that binds to the α-7 subunit of the nicotinic acetylcholine receptors (AchR-α7) of APCs, could be used for the delivery of Mycobacterium tuberculosis (Mtb) peptide antigens to DCs and macrophages. Mouse bone marrow derived DCs (BMDCs) and human THP-1 macrophages stimulated with RVG fused peptide epitopes 85B241 and 85B96 (represent Ag85B241-256 and Ag85B96-111, respectively) from antigen 85B (Ag85B) of Mtb showed enhanced antigen presentation as compared to unfused peptide epitopes and BCG. Further, BMDCs stimulated with RVG fused 85B241 showed higher levels of IL-12 positive cells. Consistent with in vitro data, splenocytes of mice immunized with RVG-85B241 showed increased number of antigen specific IFN-γ, IL-2, and TNF-α producing cells in relation to splenocytes from mice immunized with 85B241 alone. These results suggest that RVG may be a promising tool to develop effective alternate vaccines against tuberculosis (TB).
Collapse
Affiliation(s)
- Omar Garnica
- Center of Emphasis in Infectious Diseases and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Kishore Das
- Center of Emphasis in Infectious Diseases and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Santhi Devasundaram
- Center of Emphasis in Infectious Diseases and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Subramanian Dhandayuthapani
- Center of Emphasis in Infectious Diseases and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA; Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA.
| |
Collapse
|
8
|
Lu Y, Kang J, Ning H, Wang L, Xu Y, Xue Y, Xu Z, Wu X, Bai Y. Immunological characteristics of Mycobacterium tuberculosis subunit vaccines immunized through different routes. Microb Pathog 2018; 125:84-92. [PMID: 30195646 DOI: 10.1016/j.micpath.2018.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 09/02/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022]
Abstract
Tuberculosis is chronic infectious disease caused by Mycobacterium tuberculosis (M.tb) that is prevalent worldwide. Several specific antigens, such as Antigen 85B (Ag85B) and 6 kDa early secretory antigenic target (ESAT-6) protein of M.tb, are listed as some of the candidate subunit vaccines against M.tb. ESAT-6, as a virulent factor and differential gene in M.tb, shows insufficient immunogenicity in animal model. In order to investigate the ways to improve the immunogenicity of ESAT-6, we immunized ESAT-6 by subcutaneous and intramuscular routes with different adjuvants. We found that ESAT-6 immunized alone did not induce significant humoral immunity in both immunization routes. However, subcutaneous immunization of ESAT-6 plus incomplete Freund's adjuvant can induce a significant humoral immune response, enhanced proliferation and elevated secretion of IFN-γ from splenocytes. Intramuscular immunization of ESAT-6 plus adjuvant aluminum salt or poly(I:C) did not enhance humoral and cellular immune responses. Therefore, it is concluded that immunization of ESAT-6 subcutaneously plus incomplete Freund's adjuvant induces stronger humoral and cellular immune responses, which can be considered of ESAT-6 as a subunit vaccine in further research against tuberculosis.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/immunology
- Cell Proliferation
- Guinea Pigs
- Immunity, Cellular
- Immunity, Humoral
- Injections, Intramuscular
- Injections, Subcutaneous
- Interferon-gamma/metabolism
- Leukocytes, Mononuclear/immunology
- Mice
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Yanzhi Lu
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Jian Kang
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Huanhuan Ning
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Lifei Wang
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Yanhui Xu
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Ying Xue
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Zhikai Xu
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China
| | - Xingan Wu
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China.
| | - Yinlan Bai
- Department of Microbiology, College of Basic Medical Sciences, The Fourth Military Medical University, Shaanxi Province, China.
| |
Collapse
|
9
|
|
10
|
Vujadinovic M, Wunderlich K, Callendret B, Koning M, Vermeulen M, Sanders B, van der Helm E, Gecgel A, Spek D, de Boer K, Stalknecht M, Serroyen J, Grazia Pau M, Schuitemaker H, Zahn R, Custers J, Vellinga J. Adenoviral Type 35 and 26 Vectors with a Bidirectional Expression Cassette in the E1 Region Show an Improved Genetic Stability Profile and Potent Transgene-Specific Immune Response. Hum Gene Ther 2017; 29:337-351. [PMID: 28816084 DOI: 10.1089/hum.2017.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetic vaccines based on replication-incompetent adenoviral (AdV) vectors are currently in clinical development. Monovalent AdV vectors express one antigen from an expression cassette placed in most cases in the E1 region. For many vaccines, inclusion of several antigens is necessary in order to raise protective immunity and/or target more than one pathogen or pathogen strain. On the basis of the current technology, a mix of several monovalent vectors can be employed. However, a mix of the standard monovalent AdV vectors may not be optimal with respect to manufacturing costs and the final dose per vector in humans. Alternatively, a variety of bivalent recombinant AdV vector approaches is described in the literature. It remains unclear whether all strategies are equally suitable for clinical development while preserving all the beneficial properties of the monovalent AdV (e.g., immunogenic potency). Therefore, a thorough assessment of different bivalent AdV strategies was performed in a head-to-head fashion compared with the monovalent benchmark. The vectors were tested for rescue efficiency, genetic stability, transgene expression, and potency to induce transgene-specific immune responses. We report that the vector expressing multiple antigens from a bidirectional expression cassette in E1 shows a better genetic stability profile and a potent transgene-specific immune response compared with the other tested bivalent vectors.
Collapse
Affiliation(s)
- Marija Vujadinovic
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Kerstin Wunderlich
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Benoit Callendret
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Marina Koning
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Mark Vermeulen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Barbara Sanders
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Esmeralda van der Helm
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Adile Gecgel
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Dirk Spek
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Karin de Boer
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Masha Stalknecht
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jan Serroyen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Maria Grazia Pau
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Hanneke Schuitemaker
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Roland Zahn
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jerome Custers
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jort Vellinga
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| |
Collapse
|
11
|
Cunningham AL, Garçon N, Leo O, Friedland LR, Strugnell R, Laupèze B, Doherty M, Stern P. Vaccine development: From concept to early clinical testing. Vaccine 2016; 34:6655-6664. [DOI: 10.1016/j.vaccine.2016.10.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/21/2016] [Accepted: 10/04/2016] [Indexed: 12/21/2022]
|
12
|
Adenovirally-Induced Polyfunctional T Cells Do Not Necessarily Recognize the Infected Target: Lessons from a Phase I Trial of the AERAS-402 Vaccine. Sci Rep 2016; 6:36355. [PMID: 27805026 PMCID: PMC5141283 DOI: 10.1038/srep36355] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 10/13/2016] [Indexed: 11/08/2022] Open
Abstract
The development of a vaccine for Mycobacterium tuberculosis (Mtb) has been impeded by the absence of correlates of protective immunity. One correlate would be the ability of cells induced by vaccination to recognize the Mtb-infected cell. AERAS-402 is a replication-deficient serotype 35 adenovirus containing DNA expressing a fusion protein of Mtb antigens 85A, 85B and TB10.4. We undertook a phase I double-blind, randomized placebo controlled trial of vaccination with AERAS-402 following BCG. Analysis of the vaccine-induced immune response revealed strong antigen-specific polyfunctional CD4+ and CD8+ T cell responses. However, analysis of the vaccine-induced CD8+ T cells revealed that in many instances these cells did not recognize the Mtb-infected cell. Our findings highlight the measurement of vaccine-induced, polyfunctional T cells may not reflect the extent or degree to which these cells are capable of identifying the Mtb-infected cell and correspondingly, the value of detailed experimental medicine studies early in vaccine development.
Collapse
|
13
|
Jeyanathan M, Thanthrige-Don N, Afkhami S, Lai R, Damjanovic D, Zganiacz A, Feng X, Yao XD, Rosenthal KL, Medina MF, Gauldie J, Ertl HC, Xing Z. Novel chimpanzee adenovirus-vectored respiratory mucosal tuberculosis vaccine: overcoming local anti-human adenovirus immunity for potent TB protection. Mucosal Immunol 2015; 8:1373-87. [PMID: 25872483 DOI: 10.1038/mi.2015.29] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 03/20/2015] [Indexed: 02/07/2023]
Abstract
Pulmonary tuberculosis (TB) remains to be a major global health problem despite many decades of parenteral use of Bacillus Calmette-Guérin (BCG) vaccine. Developing safe and effective respiratory mucosal TB vaccines represents a unique challenge. Over the past decade or so, the human serotype 5 adenovirus (AdHu5)-based TB vaccine has emerged as one of the most promising candidates based on a plethora of preclinical and early clinical studies. However, anti-AdHu5 immunity widely present in the lung of humans poses a serious gap and limitation to its real-world applications. In this study we have developed a novel chimpanzee adenovirus 68 (AdCh68)-vectored TB vaccine amenable to the respiratory route of vaccination. We have evaluated AdCh68-based TB vaccine for its safety, T-cell immunogenicity, and protective efficacy in relevant animal models of human pulmonary TB with or without parenteral BCG priming. We have also compared AdCh68-based TB vaccine with its AdHu5 counterpart in both naive animals and those with preexisting anti-AdHu5 immunity in the lung. We provide compelling evidence that AdCh68-based TB vaccine is not only safe when delivered to the respiratory tract but, importantly, is also superior to its AdHu5 counterpart in induction of T-cell responses and immune protection, and limiting lung immunopathology in the presence of preexisting anti-AdHu5 immunity in the lung. Our findings thus suggest AdCh68-based TB vaccine to be an ideal candidate for respiratory mucosal immunization, endorsing its further clinical development in humans.
Collapse
Affiliation(s)
- M Jeyanathan
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - N Thanthrige-Don
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - S Afkhami
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - R Lai
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - D Damjanovic
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - A Zganiacz
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - X Feng
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - X-D Yao
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - K L Rosenthal
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - M Fe Medina
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - J Gauldie
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - H C Ertl
- Department of Immunology, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Z Xing
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
14
|
Prota G, Christensen D, Andersen P, Medaglini D, Ciabattini A. Peptide-specific T helper cells identified by MHC class II tetramers differentiate into several subtypes upon immunization with CAF01 adjuvanted H56 tuberculosis vaccine formulation. Vaccine 2015; 33:6823-30. [PMID: 26494626 DOI: 10.1016/j.vaccine.2015.09.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 07/17/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
CD4(+) T-cell priming is an essential step in vaccination due to the key role of T helper cells in driving both effector and memory immune responses. Here we have characterized in C57BL/6 mice the T helper subtype differentiation among tetramer-specific CD4(+) T cells primed by subcutaneous immunization with the tuberculosis vaccine antigen H56 plus the adjuvant CAF01. Peptide-specific population identified by the MHC class II tetramers differentiated into several T helper subtypes upon antigen encounter, and the frequency of subpopulations differed according to their localization. Th1 (CXCR3(+)T-bet(+)), Tfh (CXCR5(+)PD-1(+)Bcl-6(+)) and RORγt(+) cells were induced in the lymph nodes draining the immunization site (dLN), while Th1 cells were the predominant subtype in the spleen. In addition, CD4(+) T cells co-expressing multiple T-cell lineage-specifying transcription factors were also detected. In the lungs, most of the tetramer-binding T cells were RORγt(+), while Tfh and Th1 cells were absent. After boosting, a higher frequency of tetramer-binding cells co-expressing the markers CD44 and CD127 was detected compared to primed cells, and cells showed a prevalent Th1 phenotype in both dLN and spleens, while Tfh cells were significantly reduced. In conclusion, these data demonstrate that parenteral immunization with H56 and CAF01 elicits a distribution of antigen-specific CD4(+) T cells in both lymphoid tissues and lungs, and gives rise to multiple T helper subtypes, that differ depending on localization and following reactivation.
Collapse
Affiliation(s)
- Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy.
| |
Collapse
|
15
|
Damjanovic D, Khera A, Afkhami S, Lai R, Zganiacz A, Jeyanathan M, Xing Z. Age at Mycobacterium bovis BCG Priming Has Limited Impact on Anti-Tuberculosis Immunity Boosted by Respiratory Mucosal AdHu5Ag85A Immunization in a Murine Model. PLoS One 2015; 10:e0131175. [PMID: 26098423 PMCID: PMC4476612 DOI: 10.1371/journal.pone.0131175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/31/2015] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB) remains a global pandemic despite the use of Bacillus Calmette-Guérin (BCG) vaccine, partly because BCG fails to effectively control adult pulmonary TB. The introduction of novel boost vaccines such as the human Adenovirus 5-vectored AdHu5Ag85A could improve and prolong the protective immunity of BCG immunization. Age at which BCG immunization is implemented varies greatly worldwide, and research is ongoing to discover the optimal stage during childhood to administer the vaccine, as well as when to boost the immune response with potential novel vaccines. Using a murine model of subcutaneous BCG immunization followed by intranasal AdHu5Ag85A boosting, we investigated the impact of age at BCG immunization on protective efficacy of BCG prime and AdHu5Ag85A boost immunization-mediated protection. Our results showed that age at parenteral BCG priming has limited impact on the efficacy of BCG prime-AdHu5Ag85A respiratory mucosal boost immunization-enhanced protection. However, when BCG immunization was delayed until the maturity of the immune system, longer sustained memory T cells were generated and resulted in enhanced boosting effect on T cells of AdHu5Ag85A respiratory mucosal immunization. Our findings hold implications for the design of new TB immunization protocols for humans.
Collapse
Affiliation(s)
- Daniela Damjanovic
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Amandeep Khera
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rocky Lai
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
16
|
Abstract
The tuberculosis (TB) pandemic continues to rampage despite widespread use of the BCG (Bacillus Calmette-Guérin) vaccine. Novel vaccination strategies are urgently needed to arrest global transmission and prevent the uncontrolled development of multidrug-resistant forms of Mycobacterium tuberculosis. Over the last two decades, considerable progress has been made in the field of vaccine development with numerous innovative preclinical candidates and more than a dozen vaccines in clinical trials. These vaccines are developed either as boosters of the current BCG vaccine or as novel prime vaccines to replace BCG. Given the enormous prevalence of latent TB infection, vaccines that are protective on top of an already established infection remain a high priority and a significant scientific challenge. Here we discuss the current state of TB vaccine research and development, our understanding of the underlying immunology, and the requirements for an efficient TB vaccine.
Collapse
|
17
|
Junqueira-Kipnis AP, Marques Neto LM, Kipnis A. Role of Fused Mycobacterium tuberculosis Immunogens and Adjuvants in Modern Tuberculosis Vaccines. Front Immunol 2014; 5:188. [PMID: 24795730 PMCID: PMC4005953 DOI: 10.3389/fimmu.2014.00188] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 04/09/2014] [Indexed: 11/13/2022] Open
Abstract
Several approaches have been developed to improve or replace the only available vaccine for tuberculosis (TB), BCG (Bacille Calmette Guerin). The development of subunit protein vaccines is a promising strategy because it combines specificity and safety. In addition, subunit protein vaccines can be designed to have selected immune epitopes associated with immunomodulating components to drive the appropriate immune response. However, the limited antigens present in subunit vaccines reduce their capacity to stimulate a complete immune response compared with vaccines composed of live attenuated or killed microorganisms. This deficiency can be compensated by the incorporation of adjuvants in the vaccine formulation. The fusion of adjuvants with Mycobacterium tuberculosis (Mtb) proteins or immune epitopes has the potential to become the new frontier in the TB vaccine development field. Researchers have addressed this approach by fusing the immune epitopes of their vaccines with molecules such as interleukins, lipids, lipoproteins, and immune stimulatory peptides, which have the potential to enhance the immune response. The fused molecules are being tested as subunit vaccines alone or within live attenuated vector contexts. Therefore, the objectives of this review are to discuss the association of Mtb fusion proteins with adjuvants; Mtb immunogens fused with adjuvants; and cytokine fusion with Mtb proteins and live recombinant vectors expressing cytokines. The incorporation of adjuvant molecules in a vaccine can be complex, and developing a stable fusion with proteins is a challenging task. Overall, the fusion of adjuvants with Mtb epitopes, despite the limited number of studies, is a promising field in vaccine development.
Collapse
Affiliation(s)
- Ana Paula Junqueira-Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia , Brazil
| | - Lázaro Moreira Marques Neto
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia , Brazil
| | - André Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia , Brazil
| |
Collapse
|
18
|
Lindenstrøm T, Aagaard C, Christensen D, Agger EM, Andersen P. High-frequency vaccine-induced CD8⁺ T cells specific for an epitope naturally processed during infection with Mycobacterium tuberculosis do not confer protection. Eur J Immunol 2014; 44:1699-709. [PMID: 24677089 DOI: 10.1002/eji.201344358] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/03/2014] [Accepted: 02/11/2014] [Indexed: 12/26/2022]
Abstract
Relatively few MHC class I epitopes have been identified from Mycobacterium tuberculosis, but during the late stage of infection, CD8(+) T-cell responses to these epitopes are often primed at an extraordinary high frequency. Although clearly available for recognition during infection, their role in resistance to mycobacterial infections still remain unclear. As an alternative to DNA and viral vaccination platforms, we have exploited a novel CD8(+) T-cell-inducing adjuvant, cationic adjuvant formulation 05 (dimethyldioctadecylammonium/trehalose dibehenate/poly (inositic:cytidylic) acid), to prime high-frequency CD8 responses to the immunodominant H2-K(b) -restricted IMYNYPAM epitope contained in the vaccine Ag tuberculosis (TB)10.4/Rv0288/ESX-H (where ESX is mycobacterial type VII secretion system). We report that the amino acid C-terminal to this minimal epitope plays a decisive role in proteasomal cleavage and epitope priming. The primary structure of TB10.4 is suboptimal for proteasomal processing of the epitope and amino acid substitutions in the flanking region markedly increased epitope-specific CD8(+) T-cell responses. One of the optimized sequences was contained in the closely related TB10.3/Rv3019c/ESX-R Ag and when recombinantly expressed and administered in the cationic adjuvant formulation 05 adjuvant, this Ag promoted very high CD8(+) T-cell responses. This abundant T-cell response was functionally active but provided no protection against challenge, suggesting that CD8(+) T cells play a limited role in protection against M. tuberculosis in the mouse model.
Collapse
Affiliation(s)
- Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| | | | | | | | | |
Collapse
|
19
|
Ivanyi J. Function and Potentials of M. tuberculosis Epitopes. Front Immunol 2014; 5:107. [PMID: 24715888 PMCID: PMC3970012 DOI: 10.3389/fimmu.2014.00107] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/03/2014] [Indexed: 11/30/2022] Open
Abstract
Study of the function of epitopes of Mycobacterium tuberculosis antigens contributed significantly toward better understanding of the immunopathogenesis and to efforts for improving infection and disease control. Characterization of genetically permissively presented immunodominant epitopes has implications for the evolution of the host–parasite relationship, development of immunodiagnostic tests, and subunit prophylactic vaccines. Knowledge of the determinants of cross-sensitization, relevant to other pathogenic or environmental mycobacteria and to host constituents has advanced. Epitope-defined IFNγ assay kits became established for the specific detection of infection with tubercle bacilli both in humans and cattle. The CD4 T-cell epitope repertoire was found to be more narrow in patients with active disease than in latently infected subjects. However, differential diagnosis of active TB could not be made reliably merely on the basis of epitope recognition. The mechanisms by which HLA polymorphism can influence the development of multibacillary tuberculosis (TB) need further analysis of epitopes, recognized by Th2 helper cells for B-cell responses. Future vaccine development would benefit from better definition of protective epitopes and from improved construction and formulation of subunits with enhanced immunogenicity. Epitope-defined serology, due to its operational advantages is suitable for active case finding in selected high disease incidence populations, aiming for an early detection of infectious cases and hence for reducing the transmission of infection. The existing knowledge of HLA class I binding epitopes could be the basis for the construction of T-cell receptor-like ligands for immunotherapeutic application. Continued analysis of the functions of mycobacterial epitopes, recognized by T cells and antibodies, remains a fertile avenue in TB research.
Collapse
Affiliation(s)
- Juraj Ivanyi
- Guy's Hospital, Kings College London , London , UK
| |
Collapse
|
20
|
Fraser CK, Diener KR, Brown MP, Hayball JD. Improving vaccines by incorporating immunological coadjuvants. Expert Rev Vaccines 2014; 6:559-78. [PMID: 17669010 DOI: 10.1586/14760584.6.4.559] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
While vaccination continues to be the most successful interventionist health policy to date, infectious disease remains a significant cause of death worldwide. A primary reason that vaccination is not able to generate effective immunity is a lack of appropriate adjuvants capable of initiating the desired immune response. Adjuvant combinations can potentially overcome this problem; however, the possible permutations to consider, which include the route and kinetics of vaccination, as well as combinations of adjuvants, are practically limitless. This review aims to summarize the current understanding of adjuvants and related immunological processes and how this knowledge can and has been applied to the strategic selection of adjuvant combinations as components of vaccines against human infectious disease.
Collapse
Affiliation(s)
- Cara K Fraser
- Experimental Therapeutics Laboratory, Hanson Institute, and School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Australia.
| | | | | | | |
Collapse
|
21
|
Beverley PCL, Sridhar S, Lalvani A, Tchilian EZ. Harnessing local and systemic immunity for vaccines against tuberculosis. Mucosal Immunol 2014; 7:20-6. [PMID: 24253104 DOI: 10.1038/mi.2013.99] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/09/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
Abstract
The lung is the portal of entry for Mycobacterium tuberculosis (Mtb) and animal experimental evidence indicates that local immune defense mechanisms are crucial for protective immunity. Immunization via the lower respiratory tract efficiently induces a dividing, activated, antigen-dependent, lung-resident, memory T-cell population, which is partly recoverable by bronchoalveolar lavage. These cells can inhibit the growth of Mtb in the lungs immediately after infection. Delivery of appropriate signals to the lung innate immune system is critical for induction of effective local immunity. In contrast after parenteral immunization, antigen-specific cells may be found in lung tissue but few are recoverable by lavage and inhibition of mycobacterial growth is delayed. Harnessing both local and systemic immunity can provide highly effective protection in animal models and the evidence suggests that taken in aggregate, multiple animal models may predict the success of novel vaccine strategies in humans.
Collapse
Affiliation(s)
- P C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S Sridhar
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - A Lalvani
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - E Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Marongiu L, Donini M, Toffali L, Zenaro E, Dusi S. ESAT-6 and HspX improve the effectiveness of BCG to induce human dendritic cells-dependent Th1 and NK cells activation. PLoS One 2013; 8:e75684. [PMID: 24130733 PMCID: PMC3794045 DOI: 10.1371/journal.pone.0075684] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 08/16/2013] [Indexed: 11/19/2022] Open
Abstract
The limited efficacy of the BCG vaccine against tuberculosis is partly due to the missing expression of immunogenic proteins. We analyzed whether the addition to BCG of ESAT-6 and HspX, two Mycobacterium tuberculosis (Mtb) antigens, could enhance its capacity to activate human dendritic cells (DCs). BCG showed a weak ability to induce DC maturation, cytokine release, and CD4(+) lymphocytes and NK cells activation. The addition of ESAT-6 or HspX alone to BCG-stimulated DC did not improve these processes, whereas their simultaneous addition enhanced BCG-dependent DC maturation and cytokine release, as well as the ability of BCG-treated DCs to stimulate IFN-γ release and CD69 expression by CD4(+) lymphocytes and NK cells. Addition of TLR2-blocking antibody decreased IL-12 release by BCG-stimulated DCs incubated with ESAT-6 and HspX, as well as IFN-γ secretion by CD4(+) lymphocytes co-cultured with these cells. Moreover, HspX and ESAT-6 improved the capacity of BCG-treated DCs to induce the expression of memory phenotype marker CD45RO in naïve CD4(+) T cells. Our results indicate that ESAT-6 and HspX cooperation enables BCG-treated human DCs to induce T lymphocyte and NK cell-mediated immune responses through TLR2-dependent IL-12 secretion. Therefore ESAT-6 and HspX represent good candidates for improving the effectiveness of BCG vaccination.
Collapse
Affiliation(s)
- Laura Marongiu
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
| | - Marta Donini
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
| | - Lara Toffali
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
| | - Elena Zenaro
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
| | - Stefano Dusi
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, Verona, Italy
- * E-mail:
| |
Collapse
|
23
|
Baldwin SL, Ching LK, Pine SO, Moutaftsi M, Lucas E, Vallur A, Orr MT, Bertholet S, Reed SG, Coler RN. Protection against tuberculosis with homologous or heterologous protein/vector vaccine approaches is not dependent on CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:2514-2525. [PMID: 23904160 DOI: 10.4049/jimmunol.1301161] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Considerable effort has been directed to develop Mycobacterium tuberculosis vaccines to boost bacille Calmette-Guérin or for those who cannot be immunized with bacille Calmette-Guérin. We hypothesized that CD4(+) and CD8(+) T cell responses with a heterologous prime/boost vaccine approach could induce long-lived vaccine efficacy against M. tuberculosis in C57BL/6 mice. We produced an adenovirus vector expressing ID93 (Ad5-ID93) for induction of CD8 T cells to use with our candidate tuberculosis vaccine, ID93/glucopyranosyl lipid adjuvant (GLA)-stable emulsion (SE), which induces potent Th1 CD4 T cells. Ad5-ID93 generates ID93-specific CD8(+) T cell responses and induces protection against M. tuberculosis. When Ad5-ID93 is administered in a prime-boost strategy with ID93/GLA-SE, both CD4(+) and CD8(+) T cells are generated and provide protection against M. tuberculosis. In a MHC class I-deficient mouse model, all groups including the Ad5-ID93 group elicited an Ag-specific CD4(+) T cell response and significantly fewer Ag-specific CD8(+) T cells, but were still protected against M. tuberculosis, suggesting that CD4(+) Th1 T cells could compensate for the loss of CD8(+) T cells. Lastly, the order of the heterologous immunizations was critical. Long-lived vaccine protection was observed only when Ad5-ID93 was given as the boost following an ID93/GLA-SE prime. The homologous ID93/GLA-SE prime/boost regimen also induced long-lived protection. One of the correlates of protection between these two approaches was an increase in the total number of ID93-specific IFN-γ-producing CD4(+) T cells 6 mo following the last immunization. Our findings provide insight into the development of vaccines not only for tuberculosis, but other diseases requiring T cell immunity.
Collapse
Affiliation(s)
- Susan L Baldwin
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Lance K Ching
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Samuel O Pine
- Allergan, Inc. 2525 Dupont Dr., Irvine, CA USA 92612
| | - Magdalini Moutaftsi
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Elyse Lucas
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Aarthy Vallur
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | - Mark T Orr
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102
| | | | - Steven G Reed
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102.,Department of Global Health, University of Washington, Seattle, WA, USA 98195.,Immune Design Corp., 1124 Columbia Street, Suite 700, Seattle, WA, USA 98104
| | - Rhea N Coler
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Suite 400, Seattle, WA, USA 98102.,Department of Global Health, University of Washington, Seattle, WA, USA 98195
| |
Collapse
|
24
|
Tchilian E, Ahuja D, Hey A, Jiang S, Beverley P. Immunization with different formulations of Mycobacterium tuberculosis antigen 85A induces immune responses with different specificity and protective efficacy. Vaccine 2013; 31:4624-31. [PMID: 23896422 PMCID: PMC3898716 DOI: 10.1016/j.vaccine.2013.07.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/25/2013] [Accepted: 07/16/2013] [Indexed: 11/21/2022]
Abstract
Immunization intranasally with Mtb antigen 85A is more protective than parenterally. Three 85A vaccines platforms induce responses with differing epitope specificity. Responses to the CD8 85A70–78 but not the CD8 85A145–152 epitope are protective.
To test the relative efficacy of CD4 and CD8T cells in mediating protective immunity to Mycobacterium tuberculosis (Mtb), we compared three immunization regimes designed to induce preferentially each subset. BALB/c mice were immunized intranasally (i.n.) or parenterally with antigen 85A either in a recombinant Adenoviral vector (Ad85A), as recombinant protein (r85A) or as a set of overlapping 15mer peptides (p85A). For the first time we show that i.n. immunization with overlapping 85A synthetic peptides as well as Ad85A or r85A can provide protection against Mtb challenge. For all forms of the antigen, i.n. induces greater protection against Mtb challenge than parenteral immunization. Ad85A induces a predominantly CD8T cell response against the 85A70–78 epitope, r85A a CD4 response to 85A99–118 and p85A a balanced CD4/CD8 response to the CD4 85A99–118 and CD8 85A145–152 epitopes. Immune responses to CD4 85A99–118 and CD8 85A70–78 but not CD8 85A145–152 are protective. Although Ad85A induces a strong response to the protective CD8 85A70–78 epitope, we could not induce any response to this epitope by peptide immunization. These results show that although peptide immunization can induce protective immunity to Mtb challenge, it can also induce a response to a non-protective epitope in antigen 85A, indicating that the specificity of an immune response may be more important for protection against Mtb than its magnitude. These findings have important implications for the application of such vaccines in humans.
Collapse
Affiliation(s)
- Elma Tchilian
- University of Oxford, The Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK.
| | | | | | | | | |
Collapse
|
25
|
Tsolaki AG, Nagy J, Leiva S, Kishore U, Rosenkrands I, Robertson BD. Mycobacterium tuberculosis antigen 85B and ESAT-6 expressed as a recombinant fusion protein in Mycobacterium smegmatis elicits cell-mediated immune response in a murine vaccination model. Mol Immunol 2013; 54:278-83. [DOI: 10.1016/j.molimm.2012.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 11/22/2012] [Accepted: 11/27/2012] [Indexed: 11/25/2022]
|
26
|
Feng G, Jiang Q, Xia M, Lu Y, Qiu W, Zhao D, Lu L, Peng G, Wang Y. Enhanced immune response and protective effects of nano-chitosan-based DNA vaccine encoding T cell epitopes of Esat-6 and FL against Mycobacterium tuberculosis infection. PLoS One 2013; 8:e61135. [PMID: 23637790 PMCID: PMC3634041 DOI: 10.1371/journal.pone.0061135] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 03/05/2013] [Indexed: 12/12/2022] Open
Abstract
Development of a novel and effective vaccine against Mycobacterium tuberculosis (M.tb) is a challenging for preventing TB infection. In this study, a novel nanoparticle-based recombinant DNA vaccine was developed, which contains Esat-6 three T cell epitopes (Esat-6/3e) and fms-like tyrosine kinase 3 ligand (FL) genes (termed Esat-6/3e-FL), and was enveloped with chitosan (CS) nanoparticles (nano-chitosan). The immunologic and protective efficacy of the nano-chitosan-based DNA vaccine (termed nano-Esat-6/3e-FL) was assessed in C57BL/6 mice after intramuscular prime vaccination with the plasmids DNA and nasal boost with the Esat-6/3e peptides. The results showed that the immunized mice remarkably elicited enhanced T cell responses and protection against M.tb H37Rv challenge. These findings indicate that the nano-chitosan can significantly elevate the immunologic and protective effects of the DNA vaccine, and the nano-Esat-6/3e-FL is a useful vaccine for preventing M.tb infection in mice.
Collapse
Affiliation(s)
- Ganzhu Feng
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Qingtao Jiang
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Mei Xia
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Yanlai Lu
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Wen Qiu
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Dan Zhao
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Liwei Lu
- Department of Pathology, Hong Kong University, Hong Kong, China
| | - Guangyong Peng
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Yingwei Wang
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Park H, Adamson L, Ha T, Mullen K, Hagen SI, Nogueron A, Sylwester AW, Axthelm MK, Legasse A, Piatak M, Lifson JD, McElrath JM, Picker LJ, Seder RA. Polyinosinic-polycytidylic acid is the most effective TLR adjuvant for SIV Gag protein-induced T cell responses in nonhuman primates. THE JOURNAL OF IMMUNOLOGY 2013; 190:4103-15. [PMID: 23509365 DOI: 10.4049/jimmunol.1202958] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prime-boost immunization with heterologous vaccines elicits potent cellular immunity. In this study, we assessed the influence of various TLR ligands on SIV Gag-specific T cell immunity and protection following prime-boost immunization. Rhesus macaques (RMs) were primed with SIV Gag protein emulsified in Montanide ISA51 with or without TLR3 (polyinosinic-polycytidylic acid [poly-IC]), TLR4 (monophosphoryl lipid A), TLR7/8 (3M-012), TLR9 (CpG), or TLR3 (poly-IC) combined with TLR7/8 ligands, then boosted with replication defective adenovirus 5 expressing SIV Gag (rAd5-Gag). After priming, RMs that received SIV Gag protein plus poly-IC developed significantly higher frequencies of SIV Gag-specific CD4(+) Th1 responses in blood and bronchoalveolar lavage (BAL) fluid lymphocytes compared with all other adjuvants, and low-level SIV Gag-specific CD8(+) T cell responses. After the rAd5-Gag boost, the magnitude and breadth of SIV Gag-specific CD8(+) T cell responses were significantly increased in RM primed with SIV Gag protein plus poly-IC, with or without the TLR7/8 ligand, or CpG. However, the anamnestic, SIV Gag-specific CD8(+) T cell response to SIVmac251 challenge was not significantly enhanced by SIV Gag protein priming with any of the adjuvants. In contrast, the anamnestic SIV Gag-specific CD4(+) T cell response in BAL was enhanced by SIV Gag protein priming with poly-IC or CpG, which correlated with partial control of early viral replication after SIVmac251 challenge. These results demonstrate that prime-boost vaccination with SIV Gag protein/poly-IC improves magnitude, breadth, and durability of CD4(+) T cell immune responses, which could have a role in the control of SIV viral replication.
Collapse
Affiliation(s)
- Haesun Park
- Department of Pathology, Vaccine and Gene Therapy Institute, and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abebe F. Is interferon-gamma the right marker for bacille Calmette-Guérin-induced immune protection? The missing link in our understanding of tuberculosis immunology. Clin Exp Immunol 2012; 169:213-9. [PMID: 22861360 DOI: 10.1111/j.1365-2249.2012.04614.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Bacille Calmette-Guérin (BCG), developed a century ago, is the only licensed tuberculosis (TB) vaccine in use to date. The protective efficacy of BCG against TB varies with no apparent protection in some population, and mechanisms of its immune protection is poorly known, and yet BCG is the most widely used vaccine, with more than 4 billion BCG-vaccinated children globally. BCG is probably the only licensed vaccine currently in use believed to mediate immune protection through the production of interferon (IFN)-γ by CD4 T cells, which in turn activates macrophages to kill Mycobacterium tuberculosis (Mtb). Currently, a number of new TB candidate vaccines are in different phases of clinical trial. The majority of these new vaccines are either recombinant forms of BCG or prime boosters of BCG (rBCG) and their immunogenicity is tested using BCG as a benchmark by measuring specific IFN-γ produced by CD4(+) T cells as a protective immune marker. However, some recent studies that examined mechanisms of immune protection of BCG in animals and humans have reported a lack of correlation between IFN-γ production by CD4 cells and BCG-induced immune protection. These studies point to the fact that there is a missing link in our understanding of TB immunology. Conversely, there is emerging evidence that other T cell subsets (gammadelta, γδ), CD8(+) T cells and natural killer (NK) cells may play a vital role in immune protection against Mtb infection and BCG-induced immune protection. γδ T cells and NK cells, which were considered to be part of the innate immunity in the past, have been shown to develop immunological memory upon re-encounter with the same pathogen. In this paper, the controversy over the role of IFN-γ as a marker for protective immunity against TB, and emerging data on the role of γδ T cells, CD8(+) and NK cells in TB immunology, will be presented.
Collapse
Affiliation(s)
- F Abebe
- Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
29
|
You Q, Wu Y, Wu Y, Wei W, Wang C, Jiang D, Yu X, Zhang X, Wang Y, Tang Z, Jiang C, Kong W. Immunogenicity and protective efficacy of heterologous prime-boost regimens with mycobacterial vaccines and recombinant adenovirus- and poxvirus-vectored vaccines against murine tuberculosis. Int J Infect Dis 2012; 16:e816-25. [PMID: 22921259 DOI: 10.1016/j.ijid.2012.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/03/2012] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES To evaluate regimens using bacillus Calmette-Guérin (BCG) or recombinant BCG (rBCG) overexpressing Ag85B for priming, followed by boosting with a modified vaccinia virus Ankara strain (MVA) and/or adenovirus vector (AD) expressing an Ag85B-ESAT6 fusion protein. METHODS Cellular and humoral immune responses were determined after subcutaneous vaccination, which was employed to trigger systemic immunity against intravenous infection in a mouse model of tuberculosis (TB). Bacterial loads and lung histology were evaluated. RESULTS The relative IgG2a and IgG1 antibody levels indicated that the viral-vectored vaccines generated a T-helper type 1 (Th1)-biased response after two doses of viral boost vaccinations. Boosting BCG-primed mice with viral vaccines induced a Th1 immune response that included both CD4 and CD8 T-cells generating antigen-specific interferon-gamma (IFN-γ) and CD8 T cytotoxic activity. Only mice vaccinated with two different viral boosters after BCG priming exhibited a significant reduction in bacterial burden in the lung after challenge. Histology examinations confirmed the attenuation of lung damage and more compact granulomas. After mycobacteria priming, boosting with AD85B-E6 followed by MVA85B-E6 afforded better protection than the reverse order of administration of the viral vectors. CONCLUSIONS This study demonstrates the potential of multiple heterologous viral booster vaccines, although the exact correlates of protection and optimal regimens should be further investigated for the rational design of future vaccine strategies.
Collapse
Affiliation(s)
- Qingrui You
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Gaoxin District Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Beamer GL, Cyktor J, Flaherty DK, Stromberg PC, Carruthers B, Turner J. CBA/J mice generate protective immunity to soluble Ag85 but fail to respond efficiently to Ag85 during natural Mycobacterium tuberculosis infection. Eur J Immunol 2012; 42:870-9. [PMID: 22531914 DOI: 10.1002/eji.201142054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In CBA/J mice, susceptibility to Mycobacterium tuberculosis (M.tb) is associated with low interferon-gamma (IFN-γ) responses to antigens (Antigen 85 (Ag85) and early secreted antigenic target-6 (ESAT-6)) that have been defined as immunodominant. Here, we asked whether the failure of CBA/J mice to recognize Ag85 is a consequence of M.tb infection or whether CBA/J mice have a general defect in generating specific T-cell responses to this protein antigen. We compared CBA/J mice during primary M.tb infection, Ag85 vaccination followed by M.tb challenge, or M.tb memory immune mice for their capacity to generate Ag85-specific IFN-γ responses and to control M.tb infection. CBA/J mice did not respond efficiently to Ag85 in the context of natural infection or re-infection. In contrast, CBA/J mice could generate Ag85-specific IFN-γ responses and protective immunity when this antigen was delivered as a soluble protein. Our data indicate that although M.tb infection of CBA/J mice does not drive an Ag85 response, these mice can fully and protectively respond to Ag85 if it is delivered as a vaccine. The data from this experimental model suggest that the Ag85-containing vaccines in clinical trials should protect M.tb susceptible humans.
Collapse
Affiliation(s)
- Gillian L Beamer
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | |
Collapse
|
31
|
Esparza-González SC, Troy A, Troudt J, Loera-Arias MJ, Villatoro-Hernández J, Torres-López E, Ancer-Rodríguez J, Gutiérrez-Puente Y, Muñoz-Maldonado G, Saucedo-Cárdenas O, Montes-de-Oca-Luna R, Izzo A. Recombinant adenovirus delivery of calreticulin-ESAT-6 produces an antigen-specific immune response but no protection against a Mycobacterium tuberculosis challenge. Scand J Immunol 2012; 75:259-65. [PMID: 22010821 DOI: 10.1111/j.1365-3083.2011.02655.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Bacillus Calmette-Guerin (BCG) has failed to efficaciously control the worldwide spread of the disease. New vaccine development targets virulence antigens of Mycobacterium tuberculosis that are deleted in Mycobacterium bovis BCG. Immunization with ESAT-6 and CFP10 provides protection against M. tuberculosis in a murine infection model. Further, previous studies have shown that calreticulin increases the cell-mediated immune responses to antigens. Therefore, to test whether calreticulin enhances the immune response against M. tuberculosis antigens, we fused ESAT-6 to calreticulin and constructed a recombinant replication-deficient adenovirus to express the resulting fusion protein (AdCRT-ESAT-6). The adjuvant effect of calreticulin was assayed by measuring cytokine responses specific to ESAT-6. Recombinant adenovirus expressing the fusion protein produced higher levels of interferon-γ and tumour necrosis factor-α in response to ESAT-6. This immune response was not improved by the addition of CFP-10 to the CRT-ESAT-6 fusion protein (AdCRT-ESAT-6-CFP10). Mice immunized with these recombinant adenoviruses did not decrease the mycobacterial burden after low-dose aerosol infection with M. tuberculosis. We conclude that calreticulin can be used as an adjuvant to enhance the immune response against mycobacterial antigens, but it is not enough to protect against tuberculosis.
Collapse
Affiliation(s)
- S C Esparza-González
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León (UANL), Monterrey N. L., México
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hansen J, Lindenstrøm T, Lindberg-Levin J, Aagaard C, Andersen P, Agger EM. CAF05: cationic liposomes that incorporate synthetic cord factor and poly(I:C) induce CTL immunity and reduce tumor burden in mice. Cancer Immunol Immunother 2012; 61:893-903. [PMID: 22095092 PMCID: PMC11028613 DOI: 10.1007/s00262-011-1156-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 11/03/2011] [Indexed: 01/13/2023]
Abstract
Considerable effort has been put into targeting tumors through therapeutic vaccination using dendritic cell-, DNA-, protein-, or peptide-based vaccines. Purified peptides and proteins are generally not immunogenic and need to be administered with an adjuvant that will trigger an appropriate immune response. Safe adjuvants that favor induction of tumor reactive CD8(+) T cells with the capacity to directly kill tumor cells are therefore a high priority. We have previously reported on the effect and mechanism of a cationic adjuvant formulation, CAF01, which incorporates synthetic mycobacterial cord factor and primes protective Th1, Th17, and antibody responses in animal models of bacterial, viral, and parasitic infections. The CAF01 adjuvant is currently in clinical trial. Using CAF01 as a backbone, we recently demonstrated that incorporating the TLR3 ligand polyinosinic/polycytidylic acid [poly(I:C)] primes CD8(+) T cells specific to the SIINFEKL epitope of the model antigen ovalbumin. In the present study, we demonstrate that CAF01/poly(I:C), termed cationic adjuvant formulation 05 or CAF05, can induce CD8(+) T cells that efficiently lyse target cells and significantly reduce tumor growth in two different mouse tumor models: lung B16-OVA melanoma expressing ovalbumin and the self-antigen TRP2, and subcutaneous TC-1 tumors expressing the human papillomavirus-16 protein E7.
Collapse
Affiliation(s)
- Jon Hansen
- Infectious Disease Immunology, Statens Serum Institut, Building 81/306, Copenhagen S, Denmark.
| | | | | | | | | | | |
Collapse
|
33
|
Scriba TJ, Tameris M, Smit E, van der Merwe L, Hughes EJ, Kadira B, Mauff K, Moyo S, Brittain N, Lawrie A, Mulenga H, de Kock M, Makhethe L, Janse van Rensburg E, Gelderbloem S, Veldsman A, Hatherill M, Geldenhuys H, Hill AVS, Hawkridge A, Hussey GD, Hanekom WA, McShane H, Mahomed H. A phase IIa trial of the new tuberculosis vaccine, MVA85A, in HIV- and/or Mycobacterium tuberculosis-infected adults. Am J Respir Crit Care Med 2012; 185:769-78. [PMID: 22281831 DOI: 10.1164/rccm.201108-1548oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Novel tuberculosis (TB) vaccines should be safe and effective in populations infected with Mycobacterium tuberculosis (M.tb) and/or HIV for effective TB control. OBJECTIVE To determine the safety and immunogenicity of MVA85A, a novel TB vaccine, among M.tb- and/or HIV-infected persons in a setting where TB and HIV are endemic. METHODS An open-label, phase IIa trial was conducted in 48 adults with M.tb and/or HIV infection. Safety and immunogenicity were analyzed up to 52 weeks after intradermal vaccination with 5 × 10(7) plaque-forming units of MVA85A. Specific T-cell responses were characterized by IFN-γ enzyme-linked immunospot and whole blood intracellular cytokine staining assays. MEASUREMENTS AND MAIN RESULTS MVA85A was well tolerated and no vaccine-related serious adverse events were recorded. MVA85A induced robust and durable response of mostly polyfunctional CD4(+) T cells, coexpressing IFN-γ, tumor necrosis factor-α, and IL-2. Magnitudes of pre- and postvaccination T-cell responses were lower in HIV-infected, compared with HIV-uninfected, vaccinees. No significant effect of antiretroviral therapy on immunogenicity of MVA85A was observed. CONCLUSIONS MVA85A was safe and immunogenic in persons with HIV and/or M.tb infection. These results support further evaluation of safety and efficacy of this vaccine for prevention of TB in these target populations.
Collapse
Affiliation(s)
- Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine, and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
You Q, Jiang C, Wu Y, Yu X, Chen Y, Zhang X, Wei W, Wang Y, Tang Z, Jiang D, Wu Y, Wang C, Meng X, Zhao X, Kong W. Subcutaneous Administration of Modified Vaccinia Virus Ankara Expressing an Ag85B-ESAT6 Fusion Protein, but Not an Adenovirus-Based Vaccine, Protects Mice Against Intravenous Challenge with Mycobacterium tuberculosis. Scand J Immunol 2011; 75:77-84. [DOI: 10.1111/j.1365-3083.2011.02629.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
35
|
Roche CM, Smith A, Lindsey DR, Meher A, Schluns K, Arora A, Armitige LY, Jagannath C. The ΔfbpA attenuated candidate vaccine from Mycobacterium tuberculosis, H37Rv primes for a stronger T-bet dependent Th1 immunity in mice. Tuberculosis (Edinb) 2011; 91 Suppl 1:S96-104. [PMID: 22082615 DOI: 10.1016/j.tube.2011.10.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ΔfbpA candidate vaccine derived from Mycobacterium tuberculosis (H37Rv) (Mtb) protects mice better than BCG against tuberculosis, and we investigated the hypothesis that ΔfbpA may induce a stronger Th1 immunity. Since T-bet transcription factor regulates Th1 immunity, mice infected with ΔfbpA, BCG vaccine and related mycobacteria were analyzed for T-bet positive T cells. Mouse dendritic cells (DCs) or macrophages were also pulsed with excretory-secreted antigens (ES; Antigen-85B, ESAT-6 and CFP10) and cocultured with T cells from immunized or naïve mice and tested for in vitro induction of T-bet and IFN-γ. In both models, ΔfbpA mutant induced a stronger response of T-bet(+)CD4 T cells, which correlated with an increased expansion of IFN-γ(+)CD4 T cells in vivo and in vitro. When DCs pulsed with ES antigens were allowed to stimulate T cells, ESAT-6 and CFP-10 failed to induce a recall expansion of T-bet(+)IFN-γ(+)CD4 T cells from BCG vaccinated mice. Thus, deletion of RD1 in BCG seems to reduce its ability to induce T-bet and induce stronger Th1 immunity. Finally, mice were vaccinated with ΔfbpA and BCG and challenged with virulent Mtb for evaluation of protection and T cell expansion. ΔfbpA vaccinated mice showed a rapid and stronger expansion of CD4(+)CXCR3(+) IFN-γ(+) T cells in the lungs of Mtb challenged mice, compared to those which had BCG vaccine. ΔfbpA immunized mice also showed a better decline of the Mtb bacterial counts of the lungs. Mtb derived ΔfbpA candidate vaccine therefore induces qualitatively better T-bet dependent Th1 immunity than BCG vaccine.
Collapse
Affiliation(s)
- Cherie M Roche
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, 6431 Fannin, MSB 2.200, Houston, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Currently there are no sufficiently validated biomarkers to aid the evaluation of new tuberculosis vaccine candidates, the improvement of tuberculosis diagnostics or the development of more effective and shorter treatment regimens. To date, the detection of Mycobacterium tuberculosis or its products has not been able to adequately address these needs. Understanding the interplay between the host immune system and M. tuberculosis may provide a platform for the identification of suitable biomarkers, through both unbiased and targeted hypothesis-driven approaches. Here, we review immunological markers, their relation to M. tuberculosis infection stages and their potential use in the fight against tuberculosis.
Collapse
|
37
|
Liu J, Zhang S, Tan S, Zheng B, Gao GF. Revival of the identification of cytotoxic T-lymphocyte epitopes for immunological diagnosis, therapy and vaccine development. Exp Biol Med (Maywood) 2011; 236:253-67. [PMID: 21330360 DOI: 10.1258/ebm.2010.010278] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Immunogenic T-cell epitopes have a central role in the cellular immunity against pathogens and tumors. However, in the early stage of cellular immunity studies, it was complicated and time-consuming to identify and characterize T-cell epitopes. Currently, the epitope screening is experiencing renewed enthusiasm due to advances in novel techniques and theories. Moreover, the application of T-cell epitope-based diagnoses for tuberculosis and new data on epitope-based vaccine development have also revived the field. There is a growing knowledge on the emphasis of epitope-stimulated T-cell immune responses in the elimination of pathogens and tumors. In this review, we outline the significance of the identification and characterization of T-cell epitopes. We also summarize the methods and strategies for epitope definition and, more importantly, address the relevance of cytotoxic T-lymphocyte epitopes to clinical diagnoses, therapy and vaccine development.
Collapse
Affiliation(s)
- Jun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | |
Collapse
|
38
|
Woodworth JS, Shin D, Volman M, Nunes-Alves C, Fortune SM, Behar SM. Mycobacterium tuberculosis directs immunofocusing of CD8+ T cell responses despite vaccination. THE JOURNAL OF IMMUNOLOGY 2010; 186:1627-37. [PMID: 21178003 DOI: 10.4049/jimmunol.1002911] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Vaccines that elicit T cell responses try to mimic protective memory T cell immunity after infection by increasing the frequency of Ag-specific T cells in the immune repertoire. However, the factors that determine immunodominance during infection and after vaccination and the relation between immunodominance and protection are incompletely understood. We previously identified TB10.4(20-28) as an immunodominant epitope recognized by H2-K(d)-restricted CD8(+) T cells after M. tuberculosis infection. Here we report a second epitope, EspA(150-158), that is recognized by a substantial number of pulmonary CD8(+) T cells. The relative abundance of these T cells in the naive repertoire only partially predicts their relative frequency after M. tuberculosis infection. Furthermore, although vaccination with recombinant vaccinia virus expressing these epitopes changes their relative immunodominance in the preinfection T cell repertoire, this change is transient after challenge with M. tuberculosis. We speculate that factors intrinsic to the chronic nature of M. tuberculosis infection establishes the hierarchy of immunodominance and may explain the failure of some vaccines to provide protection.
Collapse
Affiliation(s)
- Joshua S Woodworth
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
39
|
Billeskov R, Grandal MV, Poulsen C, Christensen JP, Winther N, Vingsbo-Lundberg C, Hoang TTKT, van Deurs B, Song YH, Aagaard C, Andersen P, Dietrich J. Difference in TB10.4 T-cell epitope recognition following immunization with recombinant TB10.4, BCG or infection with Mycobacterium tuberculosis. Eur J Immunol 2010; 40:1342-54. [PMID: 20186878 DOI: 10.1002/eji.200939830] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most novel vaccines against infectious diseases are based on recombinant Ag; however, only few studies have compared Ag-specific immune responses induced by natural infection with that induced by the same Ag in a recombinant form. Here, we studied the epitope recognition pattern of the tuberculosis vaccine Ag, TB10.4, in a recombinant form, or when expressed by the pathogen Mycobacterium tuberculosis (M.tb), or by the current anti-tuberculosis vaccine, Mycobacterium bovis BCG. We showed that BCG and M.tb induced a similar CD4+ T-cell specific TB10.4 epitope-pattern, which differed completely from that induced by recombinant TB10.4. This difference was not due to post-translational modifications of TB10.4 or because TB10.4 is secreted from BCG and M.tb as a complex with Rv0287. In addition, BCG and TB10.4/CAF01 were both taken up by DC and macrophages in vivo, and in vitro uptake experiments revealed that both TB10.4 and BCG were transported to Lamp+-compartments. BCG and TB10.4 however, were directed to different types of Lamp+-compartments in the same APC, which may lead to different epitope recognition patterns. In conclusion, we show that different vectors can induce completely different recognition of the same protein.
Collapse
Affiliation(s)
- Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gallant CJ, Cobat A, Simkin L, Black GF, Stanley K, Hughes J, Doherty TM, Hanekom WA, Eley B, Beyers N, Jaïs JP, van Helden P, Abel L, Alcaïs A, Hoal EG, Schurr E. Tuberculin Skin Test and In Vitro Assays Provide Complementary Measures of Antimycobacterial Immunity in Children and Adolescents. Chest 2010; 137:1071-7. [DOI: 10.1378/chest.09-1852] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
41
|
Baumgartner CK, Malherbe LP. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology 2010; 130:16-22. [PMID: 20331477 DOI: 10.1111/j.1365-2567.2010.03265.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
New vaccines based on soluble recombinant antigens (Ags) require adjuvants to elicit long-lasting protective humoral and cellular immunity. Despite the importance of CD4 T helper cells for the generation of long-lived memory B and CD8 T cells, the impact of adjuvants on CD4 T-cell responses is still poorly understood. Adjuvants are known to promote dendritic cell (DC) maturation and migration to secondary lymphoid organs where they present foreign peptides bound to class II major histocompatibility complex molecules (pMHCII) to naïve CD4 T cells. Random and imprecise rearrangements of genetic elements during thymic development ensure that a vast amount of T-cell receptors (TCRs) are present in the naïve CD4 T-cell repertoire. Ag-specific CD4 T cells are selected from this vast pre-immune repertoire based on the affinity of their TCR for pMHCII. Here, we review the evidence demonstrating a link between the adjuvant and the specificity and clonotypic diversity of the CD4 T-cell response, and consider the potential mechanisms at play.
Collapse
|
42
|
Tena-Coki NG, Scriba TJ, Peteni N, Eley B, Wilkinson RJ, Andersen P, Hanekom WA, Kampmann B. CD4 and CD8 T-cell responses to mycobacterial antigens in African children. Am J Respir Crit Care Med 2010; 182:120-9. [PMID: 20224065 DOI: 10.1164/rccm.200912-1862oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The current tuberculosis (TB) vaccine, bacille Calmette-Guérin (BCG), does not provide adequate protection against TB disease in children. Furthermore, more efficacious TB vaccines are needed for children with immunodeficiencies such as HIV infection, who are at highest risk of disease. OBJECTIVES To characterize mycobacteria-specific T cells in children who might benefit from vaccination against TB, focusing on responses to antigens contained in novel TB vaccines. METHODS Whole blood was collected from three groups of BCG-vaccinated children: HIV-seronegative children receiving TB treatment (n = 30), HIV-infected children (n = 30), and HIV-unexposed healthy children (n = 30). Blood was stimulated with Ag85B and TB10.4, or purified protein derivative, and T-cell cytokine production by CD4 and CD8 was determined by flow cytometry. The memory phenotype of antigen-specific CD4 and CD8 T cells was also determined. MEASUREMENTS AND MAIN RESULTS Mycobacteria-specific CD4 and CD8 T-cell responses were detectable in all three groups of children. Children receiving TB treatment had significantly higher frequencies of antigen-specific CD4 T cells compared with HIV-infected children (P = 0.0176). No significant differences in magnitude, function, or phenotype of specific T cells were observed in HIV-infected children compared with healthy control subjects. CD4 T cells expressing IFN-gamma, IL-2, or both expressed a CD45RA(-)CCR7(-)CD27(+/-) effector memory phenotype. Mycobacteria-specific CD8 T cells expressed mostly IFN-gamma in all groups of children; these cells expressed CD45RA(-)CCR7(-)CD27(+/-) or CD45RA(+)CCR7(-)CD27(+/-) effector memory phenotypes. CONCLUSIONS Mycobacteria-specific T-cell responses could be demonstrated in all groups of children, suggesting that the responses could be boosted by new TB vaccines currently in clinical trials.
Collapse
Affiliation(s)
- Nontobeko G Tena-Coki
- Institute of Infectious Diseases and Molecular Medicine, South African Tuberculosis Vaccine Initiative, and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Kagina BMN, Abel B, Bowmaker M, Scriba TJ, Gelderbloem S, Smit E, Erasmus M, Nene N, Walzl G, Black G, Hussey GD, Hesseling AC, Hanekom WA. Delaying BCG vaccination from birth to 10 weeks of age may result in an enhanced memory CD4 T cell response. Vaccine 2009; 27:5488-95. [PMID: 19616494 PMCID: PMC2745558 DOI: 10.1016/j.vaccine.2009.06.103] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 06/29/2009] [Accepted: 06/30/2009] [Indexed: 12/01/2022]
Abstract
BACKGROUND In most tuberculosis (TB) endemic countries, bacillus Calmette-Guérin (BCG) is usually given around birth to prevent severe TB in infants. The neonatal immune system is immature. Our hypothesis was that delaying BCG vaccination from birth to 10 weeks of age would enhance the vaccine-induced immune response. METHODS In a randomized clinical trial, BCG was administered intradermally either at birth (n=25) or at 10 weeks of age (n=21). Ten weeks after vaccination, and at 1 year of age, vaccine-specific CD4 and CD8 T cell responses were measured with a whole blood intracellular cytokine assay. RESULTS Infants who received delayed BCG vaccination demonstrated higher frequencies of BCG-specific CD4 T cells, particularly polyfunctional T cells co-expressing IFN-gamma, TNF-alpha and IL-2, and most strikingly at 1 year of age. CONCLUSIONS Delaying BCG vaccination from birth to 10 weeks of age enhances the quantitative and qualitative BCG-specific T cell response, when measured at 1 year of age.
Collapse
Affiliation(s)
- Benjamin M. N. Kagina
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Brian Abel
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Mark Bowmaker
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Sebastian Gelderbloem
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Erica Smit
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Mzwandile Erasmus
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla Nene
- Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, South Africa
| | - Gerhard Walzl
- Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, South Africa
| | - Gillian Black
- Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, South Africa
| | - Gregory D. Hussey
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Anneke C. Hesseling
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Willem A. Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
45
|
Silva BDDS, da Silva EB, do Nascimento IP, dos Reis MCG, Kipnis A, Junqueira-Kipnis AP. MPT-51/CpG DNA vaccine protects mice against Mycobacterium tuberculosis. Vaccine 2009; 27:4402-7. [DOI: 10.1016/j.vaccine.2009.05.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 05/06/2009] [Accepted: 05/14/2009] [Indexed: 11/16/2022]
|
46
|
Elvang T, Christensen JP, Billeskov R, Thi Kim Thanh Hoang T, Holst P, Thomsen AR, Andersen P, Dietrich J. CD4 and CD8 T cell responses to the M. tuberculosis Ag85B-TB10.4 promoted by adjuvanted subunit, adenovector or heterologous prime boost vaccination. PLoS One 2009; 4:e5139. [PMID: 19357780 PMCID: PMC2663846 DOI: 10.1371/journal.pone.0005139] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 03/16/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although CD4 T cells are crucial for defense against M.tb, it is still not clear whether the optimal response against M.tb in fact involves both CD4 and CD8 T cells. To test this, we used a new vaccine strategy that generated a strong balanced T cell response consisting of both CD4 and CD8 T cells. METHODS AND FINDINGS To compare CD4 and CD8 responses against Ag85B-TB10.4 (H4), H4 was delivered as a subunit vaccine in cationic liposomes (CAF01), expressed in Ad5 (Ad-H4) or as a heterologous prime boost vaccination. H4/CAF01 induced primarily CD4 T cells and Ad-H4 gave predominantly a CD8 T cell response. In contrast, the heterologous prime boost combination resulted in augmentation of both the CD4 and CD8 response. The majority (>40%) of the CD4 T cells induced by the heterologous prime boost protocol were polyfunctional, and expressed IFN-gamma(+), IL-2(+), and TNF-alpha(+), whereas most of the CD8 T cells expressed IFN-gamma(+) and TNF-alpha(+) and possessed strong cytotoxic potential. The heterologous prime boost protocol also gave an increase in protective efficacy against M.tb challenge compared to H4/CAF01 and Ad-H4. Both the H4 specific CD4 and CD8 T cells were recruited to the site of infection, at the onset of infection. However, compared to CD8 T cells, CD4 T cells showed more extensive recruitment and were the main T cell subset proliferating at the site of infection. CONCLUSIONS/SIGNIFICANCE Heterologous prime boost based on H4, produced an additive effect on the priming of CD4 and CD8 cells and in terms of the protective capacity of the vaccine, and therefore represent an interesting new vaccine strategy against M.tb. However, CD4 and CD8 T cells respond very differently to live M.tb challenge, in a manner which supports the consensus that CD4 T cells do play the major role during the early stages of an M.tb infection.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Cytokines/immunology
- Cytotoxicity, Immunologic
- Female
- Genetic Vectors
- Humans
- Immunization, Secondary/methods
- Lung/cytology
- Lung/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Phenotype
- Spleen/cytology
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/genetics
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Tara Elvang
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
- Institute of Medical Microbiology and Immunology, the Panum Institute, Copenhagen, Denmark
| | - Jan P. Christensen
- Institute of Medical Microbiology and Immunology, the Panum Institute, Copenhagen, Denmark
| | - Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | | | - Peter Holst
- Institute of Medical Microbiology and Immunology, the Panum Institute, Copenhagen, Denmark
| | - Allan Randrup Thomsen
- Institute of Medical Microbiology and Immunology, the Panum Institute, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
47
|
Agger EM, Rosenkrands I, Hansen J, Brahimi K, Vandahl BS, Aagaard C, Werninghaus K, Kirschning C, Lang R, Christensen D, Theisen M, Follmann F, Andersen P. Cationic liposomes formulated with synthetic mycobacterial cordfactor (CAF01): a versatile adjuvant for vaccines with different immunological requirements. PLoS One 2008; 3:e3116. [PMID: 18776936 PMCID: PMC2525815 DOI: 10.1371/journal.pone.0003116] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Accepted: 08/11/2008] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND It is now emerging that for vaccines against a range of diseases including influenza, malaria and HIV, the induction of a humoral response is insufficient and a substantial complementary cell-mediated immune response is necessary for adequate protection. Furthermore, for some diseases such as tuberculosis, a cellular response seems to be the sole effector mechanism required for protection. The development of new adjuvants capable of inducing highly complex immune responses with strong antigen-specific T-cell responses in addition to antibodies is therefore urgently needed. METHODS AND FINDINGS Herein, we describe a cationic adjuvant formulation (CAF01) consisting of DDA as a delivery vehicle and synthetic mycobacterial cordfactor as immunomodulator. CAF01 primes strong and complex immune responses and using ovalbumin as a model vaccine antigen in mice, antigen specific cell-mediated- and humoral responses were obtained at a level clearly above a range of currently used adjuvants (Aluminium, monophosphoryl lipid A, CFA/IFA, Montanide). This response occurs through Toll-like receptor 2, 3, 4 and 7-independent pathways whereas the response is partly reduced in MyD88-deficient mice. In three animal models of diseases with markedly different immunological requirement; Mycobacterium tuberculosis (cell-mediated), Chlamydia trachomatis (cell-mediated/humoral) and malaria (humoral) immunization with CAF01-based vaccines elicited significant protective immunity against challenge. CONCLUSION CAF01 is potentially a suitable adjuvant for a wide range of diseases including targets requiring both CMI and humoral immune responses for protection.
Collapse
Affiliation(s)
- Else Marie Agger
- Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hansen J, Jensen K, Follmann F, Agger E, Theisen M, Andersen P. Liposome Delivery ofChlamydia muridarumMajor Outer Membrane Protein Primes a Th1 Response That Protects against Genital Chlamydial Infection in a Mouse Model1. J Infect Dis 2008; 198:758-67. [DOI: 10.1086/590670] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
49
|
Heterologous boosting of recombinant adenoviral prime immunization with a novel vesicular stomatitis virus-vectored tuberculosis vaccine. Mol Ther 2008; 16:1161-9. [PMID: 18388911 PMCID: PMC7185538 DOI: 10.1038/mt.2008.59] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pulmonary tuberculosis (TB) remains a serious health problem worldwide. Effective vaccination strategies are needed. We report the development of a novel TB vaccine using vesicular stomatitis virus (VSV) as a viral vector system to express Ag85A. VSVAg85A was shown to be immunogenic when given to mice by either an intranasal or an intramuscular (i.m.) route. Although distinct T-cell profiles resulted from both routes of immunization, only intranasal delivery generated a mucosal T-cell response that was protective upon pulmonary Mycobacterium tuberculosis (M.tb) challenge. While this protection manifested at an early time-point after immunization, it was not sustained. The potential of VSVAg85A to be used as a mucosal booster for parenteral priming by an adenoviral TB vaccine expressing Ag85A (AdAg85A) was investigated. VSVAg85A immunization markedly boosted antigen-specific T-cell responses in the airway lumen while also augmenting immune activation in the systemic compartment, after AdAg85A priming. This translated into significantly better protective efficacy against pulmonary challenge with M.tb than either vaccine used alone. Our study therefore suggests that VSV as a vector system is a promising candidate to be used in a heterologous viral prime-boost immunization regimen against intracellular bacterial infection.
Collapse
|
50
|
Vaccine-elicited 10-kilodalton culture filtrate protein-specific CD8+ T cells are sufficient to mediate protection against Mycobacterium tuberculosis infection. Infect Immun 2008; 76:2249-55. [PMID: 18332205 DOI: 10.1128/iai.00024-08] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The 10-kDa culture filtrate protein (CFP-10) and 6-kDa early secretory antigen of T cells (ESAT-6) are secreted in abundance by Mycobacterium tuberculosis and are frequently recognized by T cells from infected people. The genes encoding these proteins have been deleted from the genome of the vaccine strain Mycobacterium bovis bacillus Calmette-Guérin (BCG), and it is hypothesized that these proteins are important targets of protective immunity. Indeed, vaccination with ESAT-6 elicits protective CD4+ T cells in C57BL/6 mice. We have previously shown that M. tuberculosis infection of C3H mice elicits CFP-10-specific CD8+ and CD4+ T cells. Here we demonstrate that immunization with a CFP-10 DNA vaccine stimulates a specific T-cell response only to the H-2K(k)-restricted epitope CFP-10(32-39). These CFP-10(32-39)-specific CD8+ cells undergo a rapid expansion and accumulate in the lung following challenge of immunized mice with aerosolized M. tuberculosis. Protective immunity is induced by CFP-10 DNA vaccination as measured by a CFU reduction in the lung and spleen 4 and 8 weeks after challenge with M. tuberculosis. These data demonstrate that CFP-10 is a protective antigen and that CFP-10(32-39)-specific CD8+ T cells elicited by vaccination are sufficient to mediate protection against tuberculosis.
Collapse
|