1
|
Ivanyi J. Tuberculosis vaccination needs to avoid 'decoy' immune reactions. Tuberculosis (Edinb) 2020; 126:102021. [PMID: 33254012 DOI: 10.1016/j.tube.2020.102021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022]
Abstract
Current search for a new effective vaccine against tuberculosis involves selected antigens, vectors and adjuvants. These are being evaluated usually by their booster inoculation following priming with Bacillus Calmette-Guerin. The purpose of this article is to point out, that despite being attenuated of virulence, priming with BCG may still involve immune mechanisms, which are not favourable for protection against active disease. It is postulated, that the responsible 'decoy' constituents selected during the evolution of pathogenic tubercle bacilli may be involved in the evasion from bactericidal host resistance and stimulate immune responses of a cytokine phenotype, which lead to the transition from latent closed granulomas to reactivation with infectious lung cavities. The decoy mechanisms appear as favourable for most infected subjects but leading in a minority of cases to pathology which can effectively transmit the infection. It is proposed that construction and development of new vaccine candidates could benefit from avoiding decoy-type immune mechanisms.
Collapse
Affiliation(s)
- Juraj Ivanyi
- Centre for Host-Microbiome Interactions, Guy's Campus of Kings College London, SE1, 1UL, United kingdom.
| |
Collapse
|
2
|
Moradi B, Sankian M, Amini Y, Gholoobi A, Meshkat Z. A new DNA vaccine expressing HspX-PPE44-EsxV fusion antigens of Mycobacterium tuberculosis induced strong immune responses. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:909-914. [PMID: 32774813 PMCID: PMC7395183 DOI: 10.22038/ijbms.2020.38521.9171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Infection with tuberculosis (TB) is regarded as a major health issue. Due to the emergence of antibiotic resistance during TB treatment, prevention via vaccination is one of the most effective ways of controlling the infection. DNA vaccines are developed at a greater pace due to their ability in generating a long-lasting immune response, higher safety compared to the live vaccines, and relatively lower cost of production. In the present study, we evaluated a new DNA vaccine encoding the fusion HspX-PPE44-EsxV antigens, separately, and in combination with Bacillus Calmette-Guérin (BCG) administration, in a prime-boost method in mice. MATERIALS AND METHODS A novel DNA vaccine encoding HspX-PPE44-EsxV fusion antigen of Mycobacterium tuberculosis was constructed, and RT-PCR and Western blot analysis were performed to verify the expression of the antigen. Female BALB/c mice were divided into five groups (PBS, BCG, pcDNA3.1 (+) vector, pDNA/HspX-PPE44-EsxV vaccine, and the BCG-prime boost groups). In order to evaluate the immunogenicity of the recombinant vector, BALB/c mice were injected with 100 μg of pDNA at 2-week intervals. Then, cytokine assay was conducted using eBioscience ELISA kits (Ebioscience, AUT) according to manufacturers' instructions to evaluate the concentrations of IL-4, IL-12, TGF-β, and IFN-γ. RESULTS The concentrations of INF-γ, IL-12, and TGF-beta were significantly increased compared to the control groups (P<0.001). INF-γ and IL-12 production were increased significantly in pDNA/HspX-PPE44-EsxV+BCG group compared to pDNA/HspX-PPE44-EsxV group (P<0.001). CONCLUSION This study showed that the present DNA vaccine could induce a high level of specific cytokines in mice. It was also shown that using this DNA vaccine in a BCG prime-boost protocol can produce significant amounts of IFN-γ, IL-12, and TGF-β.
Collapse
Affiliation(s)
- Bagher Moradi
- Esfarayen Faculty of Medical Sciences, Esfarayen, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yousef Amini
- Infectious Diseases and Tropical Medicine Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Aida Gholoobi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Bickett TE, McLean J, Creissen E, Izzo L, Hagan C, Izzo AJ, Silva Angulo F, Izzo AA. Characterizing the BCG Induced Macrophage and Neutrophil Mechanisms for Defense Against Mycobacterium tuberculosis. Front Immunol 2020; 11:1202. [PMID: 32625209 PMCID: PMC7314953 DOI: 10.3389/fimmu.2020.01202] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022] Open
Abstract
The live attenuated Mycobacterium bovis strain, Bacille Calmette Guérin (BCG) is a potent innate immune stimulator. In the C57BL/6 mouse model of tuberculosis, BCG vaccination leads to a significant reduction of Mycobacterium tuberculosis burden after aerogenic infection. Our studies indicated that BCG induced protection against pulmonary tuberculosis was independent of T cells and present as early as 7 days after vaccination. This protection showed longevity, as it did not wane when conventional T cell and TNF-α deficient mice were infected 30 days post-vaccination. As BCG induced mycobacterial killing after 7 days, this study investigated the contributions of the innate immune system after BCG vaccination to better understand mechanisms required for mycobacterial killing. Subcutaneous BCG inoculation resulted in significant CD11b+F4/80+ monocyte subset recruitment into the lungs within 7 days. Further studies revealed that killing of mycobacteria was dependent on the viability of BCG, because irradiated BCG did not have the same effect. Although others have identified BCG as a facilitator of trained innate immunity, we found that BCG reduced the mycobacterial burden in the absence of mechanisms required for trained innate immunity, highlighting a role for macrophages and neutrophils for vaccine induced killing of M. tuberculosis.
Collapse
Affiliation(s)
- Thomas E Bickett
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Jennifer McLean
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Elizabeth Creissen
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Linda Izzo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Cassidy Hagan
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Antonio J Izzo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Fabiola Silva Angulo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Angelo A Izzo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
4
|
Moliva JI, Turner J, Torrelles JB. Immune Responses to Bacillus Calmette-Guérin Vaccination: Why Do They Fail to Protect against Mycobacterium tuberculosis? Front Immunol 2017; 8:407. [PMID: 28424703 PMCID: PMC5380737 DOI: 10.3389/fimmu.2017.00407] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis (TB), is the current leading cause of death due to a single infectious organism. Although curable, the broad emergence of multi-, extensive-, extreme-, and total-drug resistant strains of M.tb has hindered eradication efforts of this pathogen. Furthermore, computational models predict a quarter of the world’s population is infected with M.tb in a latent state, effectively serving as the largest reservoir for any human pathogen with the ability to cause significant morbidity and mortality. The World Health Organization has prioritized new strategies for improved vaccination programs; however, the lack of understanding of mycobacterial immunity has made it difficult to develop new successful vaccines. Currently, Mycobacterium bovis bacillus Calmette–Guérin (BCG) is the only vaccine approved for use to prevent TB. BCG is highly efficacious at preventing meningeal and miliary TB, but is at best 60% effective against the development of pulmonary TB in adults and wanes as we age. In this review, we provide a detailed summary on the innate immune response of macrophages, dendritic cells, and neutrophils in response to BCG vaccination. Additionally, we discuss adaptive immune responses generated by BCG vaccination, emphasizing their specific contributions to mycobacterial immunity. The success of future vaccines against TB will directly depend on our understanding of mycobacterial immunity.
Collapse
Affiliation(s)
- Juan I Moliva
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
5
|
Abstract
ABSTRACT
Immunological memory is a central feature of the adaptive immune system and a prerequisite for generating effective vaccines. Understanding long-term memory responses to
Mycobacterium tuberculosis
will thus provide us with valuable insights that can guide us in the search for a novel vaccine against tuberculosis (TB). For many years, triggering CD4 T cells and, in particular, those secreting interferon-γ has been the goal of most TB vaccine research, and numerous data from animals and humans support the key role of this subset in protective immunity. More recently, we have learned that the memory response required for effective control of
M. tuberculosis
is much more complex, probably involving several phenotypically different CD4 T cell subsets as well as other cell types that are yet to be defined. Herein, we describe recent insights into memory immunity to TB in the context of both animal models and the human infection. With the increasing amount of data generated from clinical testing of novel TB vaccines, we also summarize recent knowledge of vaccine-induced memory immunity.
Collapse
|
6
|
Chaubey KK, Gupta RD, Gupta S, Singh SV, Bhatia AK, Jayaraman S, Kumar N, Goel A, Rathore AS, Sahzad, Sohal JS, Stephen BJ, Singh M, Goyal M, Dhama K, Derakhshandeh A. Trends and advances in the diagnosis and control of paratuberculosis in domestic livestock. Vet Q 2016; 36:203-227. [PMID: 27356470 DOI: 10.1080/01652176.2016.1196508] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Paratuberculosis (pTB) is a chronic granulomatous enteritis caused by Mycobacterium avium subsp. paratuberculosis (MAP) in a wide variety of domestic and wild animals. Control of pTB is difficult due to the lack of sensitive, efficacious and cost-effective diagnostics and marker vaccines. Microscopy, culture, and PCR have been used for the screening of MAP infection in animals for quite a long time. Besides, giving variable sensitivity and specificity, these tests have not been considered ideal for large-scale screening of domestic livestock. Serological tests like ELISA easily detects anti-MAP antibodies. However, it cannot differentiate between the vaccinated and infected animals. Nanotechnology-based diagnostic tests are underway to improve the sensitivity and specificity. Newer generation diagnostic tests based on recombinant MAP secretory proteins would open new paradigm for the differentiation between infected and vaccinated animals and for early detection of the infection. Due to higher seroreactivity of secretory proteins vis-à-vis cellular proteins, the secretory proteins may be used as marker vaccine, which may aid in the control of pTB infection in animals. Secretory proteins can be potentially used to develop future diagnostics, surveillance and monitoring of the disease progression in animals and the marker vaccine for the control and eradication of pTB.
Collapse
Affiliation(s)
- Kundan Kumar Chaubey
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India.,b Department of Microbiology and Immunology , GLA University , Mathura , India
| | - Rinkoo Devi Gupta
- c Department of Life sciences and Biotechnology , South Asian University , New Delhi , India
| | - Saurabh Gupta
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India.,b Department of Microbiology and Immunology , GLA University , Mathura , India
| | - Shoor Vir Singh
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India
| | - Ashok Kumar Bhatia
- b Department of Microbiology and Immunology , GLA University , Mathura , India
| | - Sujata Jayaraman
- d Amity Institutes of Microbial Technology , Amity University , Jaipur , India
| | - Naveen Kumar
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India
| | - Anjana Goel
- b Department of Microbiology and Immunology , GLA University , Mathura , India
| | - Abhishek Singh Rathore
- c Department of Life sciences and Biotechnology , South Asian University , New Delhi , India
| | - Sahzad
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India
| | - Jagdip Singh Sohal
- d Amity Institutes of Microbial Technology , Amity University , Jaipur , India
| | - Bjorn John Stephen
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India
| | - Manju Singh
- a Microbiology Laboratory, Animal Health Division , Central Institute for Research on Goats , Mathura , India
| | - Manish Goyal
- e Division of Parasitology , Central Drug Research Institute , Lucknow , India
| | - Kuldeep Dhama
- f Pathology Division , Indian Veterinary Research Institute (IVRI) , Bareilly , India
| | - Abdollah Derakhshandeh
- g Department of Pathobiology, School of Veterinary Medicine , Shiraz University , Shiraz , Iran
| |
Collapse
|
7
|
Villarreal DO, Siefert RJ, Weiner DB. Alarmin IL-33 elicits potent TB-specific cell-mediated responses. Hum Vaccin Immunother 2016; 11:1954-60. [PMID: 26091147 DOI: 10.1080/21645515.2015.1026499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tuberculosis (TB) still remains a major public health issue despite the current available vaccine for TB, Bacille Calmette Guerin (BCG). An effective vaccine against TB remains a top priority in the fight against this pandemic bacterial infection. Adequate protection against TB is associated with the development of TH1-type and CD8(+) T cell responses. One alarmin cytokine, interleukin 33 (IL-33), has now been implicated in the development of both CD4(+) TH1 and CD8(+) T cell immunity. In this study, we determined whether the administration of IL-33 as an adjuvant, encoded in a DNA plasmid, could enhance the immunogenicity of a TB DNA vaccine. We report that the co-immunization of IL-33 with a DNA vaccine expressing the Mycobacterium Tuberculosis (Mtb) antigen 85B (Ag85B) induced robust Ag85B-specific IFNγ responses by ELISpot compared to Ag85B alone. Furthermore, these enhanced responses were characterized by higher frequencies of Ag85B-specific, multifunctional CD4(+) and CD8(+) T cells. Vaccination with IL-33 also increased the ability of the Ag85B-specific CD8(+) T cells to undergo degranulation and to secrete IFNγ and TNFα cytokines. These finding highlights IL-33 as a promising adjuvant to significantly improve the immunogenicity of TB DNA vaccines and support further study of this effective vaccine strategy against TB.
Collapse
Affiliation(s)
- Daniel O Villarreal
- a Department of Pathology and Laboratory Medicine ; Perelman School of Medicine; University of Pennsylvania ; Philadelphia , PA USA
| | | | | |
Collapse
|
8
|
Carpenter SM, Nunes-Alves C, Booty MG, Way SS, Behar SM. A Higher Activation Threshold of Memory CD8+ T Cells Has a Fitness Cost That Is Modified by TCR Affinity during Tuberculosis. PLoS Pathog 2016; 12:e1005380. [PMID: 26745507 PMCID: PMC4706326 DOI: 10.1371/journal.ppat.1005380] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/11/2015] [Indexed: 12/17/2022] Open
Abstract
T cell vaccines against Mycobacterium tuberculosis (Mtb) and other pathogens are based on the principle that memory T cells rapidly generate effector responses upon challenge, leading to pathogen clearance. Despite eliciting a robust memory CD8+ T cell response to the immunodominant Mtb antigen TB10.4 (EsxH), we find the increased frequency of TB10.4-specific CD8+ T cells conferred by vaccination to be short-lived after Mtb challenge. To compare memory and naïve CD8+ T cell function during their response to Mtb, we track their expansions using TB10.4-specific retrogenic CD8+ T cells. We find that the primary (naïve) response outnumbers the secondary (memory) response during Mtb challenge, an effect moderated by increased TCR affinity. To determine whether the expansion of polyclonal memory T cells is restrained following Mtb challenge, we used TCRβ deep sequencing to track TB10.4-specific CD8+ T cells after vaccination and subsequent challenge in intact mice. Successful memory T cells, defined by their clonal expansion after Mtb challenge, express similar CDR3β sequences suggesting TCR selection by antigen. Thus, both TCR-dependent and -independent factors affect the fitness of memory CD8+ responses. The impaired expansion of the majority of memory T cell clonotypes may explain why some TB vaccines have not provided better protection. CD8+ T cells are important for enforcing latency of tuberculosis, and for Mtb control in patients with HIV and low CD4 counts. While vaccines that primarily elicit CD4+ T cell responses have had difficulty preventing active pulmonary TB, a TB vaccine that elicits a potent memory CD8+ T cells is a logical alternative strategy. Memory T cells are thought to respond more rapidly than the primary (naïve) response. However, by directly comparing naïve and memory TCR retrogenic CD8+ T cells specific for the TB10.4 antigen during infection, we observe memory-derived T cells to be less fit than naïve-derived T cells. We relate the reduced fitness of memory CD8+ T cells to their lower sensitivity to antigen and show that fitness can be improved by increasing TCR affinity. Using a novel method for tracking CD8+ T cells elicited by vaccination during the response to Mtb aerosol challenge in intact mice, we observe the robust expansion of a new primary response as well as clonal selection of the secondary response, likely driven by TCR affinity. We propose that generating memory T cells with high affinities should be a goal of vaccination against TB.
Collapse
Affiliation(s)
- Stephen M. Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (SMC); (SMB)
| | - Cláudio Nunes-Alves
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Matthew G. Booty
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sing Sing Way
- Division of Infectious Diseases, Cincinnati Children’s Hospital, Cincinnati, Ohio, United States of America
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (SMC); (SMB)
| |
Collapse
|
9
|
Villarreal DO, Walters J, Laddy DJ, Yan J, Weiner DB. Multivalent TB vaccines targeting the esx gene family generate potent and broad cell-mediated immune responses superior to BCG. Hum Vaccin Immunother 2015; 10:2188-98. [PMID: 25424922 DOI: 10.4161/hv.29574] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Development of a broad-spectrum synthetic vaccine against TB would represent an important advance to the limited vaccine armamentarium against TB. It is believed that the esx family of TB antigens may represent important vaccine candidates. However, only 4 esx antigens have been studied as potential vaccine antigens. The challenge remains to develop a vaccine that simultaneously targets all 23 members of the esx family to induce enhanced broad-spectrum cell-mediated immunity. We sought to investigate if broader cellular immune responses could be induced using a multivalent DNA vaccine representing the esx family protein members delivered via electroporation. In this study, 15 designed esx antigens were created to cross target all members of the esx family. They were distributed into groups of 3 self-processing antigens each, resulting in 5 trivalent highly optimized DNA plasmids. Vaccination with all 5 constructs elicited robust antigen-specific IFN-γ responses to all encoded esx antigens and induced multifunctional CD4 Th1 and CD8 T cell responses. Importantly, we show that when all constructs are combined into a cocktail, the RSQ-15 vaccine, elicited substantial broad Ag-specific T cell responses to all esx antigens as compared with vaccination with BCG. Moreover, these vaccine-induced responses were highly cross-reactive with BCG encoded esx family members and were highly immune effective in a BCG DNA prime-boost format. Furthermore, we demonstrate the vaccine potential and immunopotent profile of several novel esx antigens never previously studied. These data highlight the likely importance of these novel immunogens for study as preventative or therapeutic synthetic TB vaccines in combination or as stand alone antigens.
Collapse
Affiliation(s)
- Daniel O Villarreal
- a Department of Pathology and Laboratory Medicine; University of Pennsylvania School of Medicine; Philadelphia, PA USA
| | | | | | | | | |
Collapse
|
10
|
Quest for correlates of protection against tuberculosis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:258-66. [PMID: 25589549 DOI: 10.1128/cvi.00721-14] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A major impediment to tuberculosis (TB) vaccine development is the lack of reliable correlates of immune protection or biomarkers that would predict vaccine efficacy. Gamma interferon (IFN-γ) produced by CD4(+) T cells and, recently, multifunctional CD4(+) T cells secreting IFN-γ, tumor necrosis factor (TNF), and interleukin-2 (IL-2) have been used in vaccine studies as a measurable immune parameter, reflecting activity of a vaccine and potentially predicting protection. However, accumulating experimental evidence suggests that host resistance against Mycobacterium tuberculosis infection is independent of IFN-γ and TNF secretion from CD4(+) T cells. Furthermore, the booster vaccine MVA85A, despite generating a high level of multifunctional CD4(+) T cell response in the host, failed to confer enhanced protection in vaccinated subjects. These findings suggest the need for identifying reliable correlates of protection to determine the efficacy of TB vaccine candidates. This article focuses on alternative pathways that mediate M. tuberculosis control and their potential for serving as markers of protection. The review also discusses the significance of investigating the natural human immune response to M. tuberculosis to identify the correlates of protection in vaccination.
Collapse
|
11
|
Geluk A, van den Eeden SJF, van Meijgaarden KE, Dijkman K, Franken KLMC, Ottenhoff THM. A multistage-polyepitope vaccine protects against Mycobacterium tuberculosis infection in HLA-DR3 transgenic mice. Vaccine 2012; 30:7513-21. [PMID: 23103299 DOI: 10.1016/j.vaccine.2012.10.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/03/2012] [Accepted: 10/13/2012] [Indexed: 10/27/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is responsible for almost 2 million deaths annually. BCG, currently the only TB vaccine, induces variable protection and does not protect against reactivation of latent TB. Thus, efficient vaccines to supplement BCG are required urgently. Since Mtb's proteome differs qualitatively and quantitatively during bacterial replication stages from that expressed during dormancy, improved TB vaccines should drive immune responses to Mtb antigens expressed during multiple stages of infection. Consequently, such "multistage" vaccines should be composed of (immunodominant) antigens expressed during different phases of Mtb infection. As a concept multistage vaccine, we constructed a polyepitope by fusing five HLA-DR3-restricted T-cell epitopes derived from different Mtb proteins either expressed highly by replicating bacteria (Ag85B, hsp65, 19 kDa lipoprotein), or abundantly expressed by dormant bacilli and recognized preferentially by TST(+) individuals (hsp16, Rv1733c). PBMC of HLA-DR3(+) but not HLA-DR3(-) cured TB patients and TST(+) individuals responded well to the multistage-polyepitope in vitro. The in vivo immunogenicity and protective efficacy of the multistage-polyepitope were analyzed using HLA-DR3 transgenic mice lacking endogenous murine class II as a model. Immunization with the multistage-polyepitope adjuvanted with CpG generated high IgG levels as well as polyfunctional CD4(+) T-cells producing IFN-γ, TNF and IL-2, specific for these HLA-DR3-restricted epitopes. Importantly, multistage-polyepitope immunization reduced the number of bacilli in the lungs after Mtb challenge when administered as prophylactic vaccine. Given the extensive repertoire of potential Mtb antigens available for immune recognition, the data of our model demonstrate the potential of multistage-polyepitope vaccines to protect against TB.
Collapse
Affiliation(s)
- Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands.
| | | | | | | | | | | |
Collapse
|
12
|
Zhang Y, Chen S, Li J, Liu Y, Hu Y, Cai H. Oral immunogenicity of potato-derived antigens to Mycobacterium tuberculosis in mice. Acta Biochim Biophys Sin (Shanghai) 2012; 44:823-30. [PMID: 22917938 DOI: 10.1093/abbs/gms068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The novel use of transgenic plants as vectors for the expression of viral and bacterial antigens has been increasingly tested as an alternative methodology for the production and delivery of experimental oral vaccines. Here, we examined the immunogenicity of combined plant-made vaccines that include four genes encoding immune-dominant antigens from Mycobacterium tuberculosis. Compared with the wild type and other control groups, mice treated with the combined plant-made vaccines showed significantly higher levels of interferon-γ and interleukin-2 production in response to all four proteins, and higher levels of antigen-specific CD4(+) and CD8(+) T-cell responses and immunoglobulin (Ig) G and IgA titers. These results suggest that combined plant-made vaccines can induce immunogenicity against M. tuberculosis through the induction of stronger Th1-associated immune responses. This is the first report of an orally delivered combined plant-made vaccine against tuberculosis priming an antigen-specific Th1 response, a comprehensive effect including both mucosal and systemic immune responses.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory of Protein and Plant Gene Research, Peking University, Beijing, China
| | | | | | | | | | | |
Collapse
|
13
|
Yuan W, Dong N, Zhang L, Liu J, Lin S, Xiang Z, Qiao H, Tong W, Qin C. Immunogenicity and protective efficacy of a tuberculosis DNA vaccine expressing a fusion protein of Ag85B-Esat6-HspX in mice. Vaccine 2012; 30:2490-7. [DOI: 10.1016/j.vaccine.2011.06.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 05/19/2011] [Accepted: 06/09/2011] [Indexed: 12/23/2022]
|
14
|
Parra M, Derrick SC, Yang A, Tian J, Kolibab K, Oakley M, Perera LP, Jacobs WR, Kumar S, Morris SL. Malaria infections do not compromise vaccine-induced immunity against tuberculosis in mice. PLoS One 2011; 6:e28164. [PMID: 22205939 PMCID: PMC3242757 DOI: 10.1371/journal.pone.0028164] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/02/2011] [Indexed: 11/18/2022] Open
Abstract
Background Given the considerable geographic overlap in the endemic regions for malaria and tuberculosis, it is probable that co-infections with Mycobacterium tuberculosis and Plasmodium species are prevalent. Thus, it is quite likely that both malaria and TB vaccines may be used in the same populations in endemic areas. While novel vaccines are currently being developed and tested individually against each of these pathogens, the efficacy of these vaccines has not been evaluated in co-infection models. To further assess the effectiveness of these new immunization strategies, we investigated whether co-infection with malaria would impact the anti-tuberculosis protection induced by four different types of TB vaccines in a mouse model of pulmonary tuberculosis. Principal Findings Here we show that the anti-tuberculosis protective immunity induced by four different tuberculosis vaccines was not impacted by a concurrent infection with Plasmodium yoelii NL, a nonlethal form of murine malaria. After an aerogenic challenge with virulent M. tuberculosis, the lung bacterial burdens of vaccinated animals were not statistically different in malaria infected and malaria naïve mice. Multi-parameter flow cytometric analysis showed that the frequency and the median fluorescence intensities (MFI) for specific multifunctional T (MFT) cells expressing IFN-γ, TNF-α, and/or IL-2 were suppressed by the presence of malaria parasites at 2 weeks following the malaria infection but was not affected after parasite clearance at 7 and 10 weeks post-challenge with P. yoelii NL. Conclusions Our data indicate that the effectiveness of novel TB vaccines in protecting against tuberculosis was unaffected by a primary malaria co-infection in a mouse model of pulmonary tuberculosis. While the activities of specific MFT cell subsets were reduced at elevated levels of malaria parasitemia, the T cell suppression was short-lived. Our findings have important relevance in developing strategies for the deployment of new TB vaccines in malaria endemic areas.
Collapse
Affiliation(s)
- Marcela Parra
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
| | - Steven C. Derrick
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
| | - Amy Yang
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
| | - JinHua Tian
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
| | - Kristopher Kolibab
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
| | - Miranda Oakley
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
| | - Liyanage P. Perera
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - William R. Jacobs
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Sanjai Kumar
- Office of Blood Research and Review, Center for Biologics Research and Review, USFDA, Bethesda, Maryland, United States of America
| | - Sheldon L. Morris
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
15
|
Woodworth JS, Shin D, Volman M, Nunes-Alves C, Fortune SM, Behar SM. Mycobacterium tuberculosis directs immunofocusing of CD8+ T cell responses despite vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:1627-37. [PMID: 21178003 PMCID: PMC3133636 DOI: 10.4049/jimmunol.1002911] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
- Program in Immunology, Harvard School of Public Health, Boston, MA 02115
| | - Daniel Shin
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Mattijs Volman
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Cláudio Nunes-Alves
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115
| | - Samuel M. Behar
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115
| |
Collapse
|
16
|
Mu J, Jeyanathan M, Shaler CR, Horvath C, Damjanovic D, Zganiacz A, Kugathasan K, McCormick S, Xing Z. Respiratory mucosal immunization with adenovirus gene transfer vector induces helper CD4 T cell-independent protective immunity. J Gene Med 2010; 12:693-704. [PMID: 20694950 DOI: 10.1002/jgm.1487] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Virus-vectored vaccine is a powerful activator of CD8 T cell-mediated immunity and is especially amenable to respiratory mucosal immunization, offering hopes for use in humans with diminished helper CD4 T cell function. However, whether virus-mediated mucosal immunization can produce immune protective CD8 T cells without the CD4 T cell help remains to be investigated. METHODS We used a replication-deficient adenovirus vector expressing an Mycobacterium tuberculosis antigen Ag85A for intranasal vaccination and evaluated its effect on CD8 T cell activation and protection in mice depleted of CD4 T cells. RESULTS Intranasal vaccination of CD4 T cell-depleted mice led to suboptimal generation of Ag-specific tetramer(+) or interferon (IFN)-gamma-producing CD8 T cells in the lung and spleen but this was observed mainly at the early time after vaccination. Reduced CD8 T cell priming was also accompanied by decreased CD8 T cell responses (CTL). Nevertheless, the ratio of Ag-specific CD8 T cells to IFN-gamma-producing CD8 T cells in CD4 T cell-depleted hosts remained comparable to that in CD4 T cell-competent hosts. Furthermore, the 'unhelped' CD8 T cells also displayed a similar immune phenotype as the 'helped' counterparts. The animals with 'unhelped' CD8 T cells were as well-protected from pulmonary M. tuberculosis challenge as those with 'helped' CD8 T cells in the absence of CD4 T cells. CONCLUSIONS The data obtained in the present study suggest that the fully immune protective CD8 T cells can still be generated by respiratory mucosal viral-mediated immunization without CD4 T cells and that CD8 T cells, 'helped' or 'unhelped', can confer significant protection against pulmonary tuberculosis independent of CD4 T cells.
Collapse
Affiliation(s)
- Jingyu Mu
- Department of Pathology & Molecular Medicine, Centre for Gene Therapeutics & M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
There has never been a greater need for a new protective tuberculosis vaccine. Bacille Calmette-Guerin remains the cornerstone of any vaccine strategy, but improving its immunogenicity and efficacy has now become an urgent global health priority. This review discusses the main vaccines currently in clinical development and other novel vaccine strategies in the pipeline. It addresses the key questions in vaccine design, including antigen selection, route of vaccine delivery and immune correlates of vaccine-induced protection. There is an opportunity to identify such correlates from ongoing and future Phase II/III trials and, as these emerge, they can be used to validate the most relevant and predictive animal models with which to develop the next generation of new vaccines.
Collapse
Affiliation(s)
- Angela M Minassian
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Level 2, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| | | |
Collapse
|
18
|
Ingolotti M, Kawalekar O, Shedlock DJ, Muthumani K, Weiner DB. DNA vaccines for targeting bacterial infections. Expert Rev Vaccines 2010; 9:747-63. [PMID: 20624048 PMCID: PMC2962930 DOI: 10.1586/erv.10.57] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DNA vaccination has been of great interest since its discovery in the 1990s due to its ability to elicit both humoral and cellular immune responses. DNA vaccines consist of a DNA plasmid containing a transgene that encodes the sequence of a target protein from a pathogen under the control of a eukaryotic promoter. This revolutionary technology has proven to be effective in animal models and four DNA vaccine products have recently been approved for veterinary use. Although few DNA vaccines against bacterial infections have been tested, the results are encouraging. Because of their versatility, safety and simplicity a wider range of organisms can be targeted by these vaccines, which shows their potential advantages to public health. This article describes the mechanism of action of DNA vaccines and their potential use for targeting bacterial infections. In addition, it provides an updated summary of the methods used to enhance immunogenicity from codon optimization and adjuvants to delivery techniques including electroporation and use of nanoparticles.
Collapse
Affiliation(s)
- Mariana Ingolotti
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Department of Anatomy, School of Biomedical Sciences, Austral University, Pilar, Buenos Aires, Argentina
| | - Omkar Kawalekar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Devon J Shedlock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - David B Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
19
|
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 PMCID: PMC2902756 DOI: 10.1164/rccm.200912-1862oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 03/09/2010] [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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Thomas J. Scriba
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Nomathemba Peteni
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Brian Eley
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Robert J. Wilkinson
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Peter Andersen
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Willem A. Hanekom
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Beate Kampmann
- 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; Wellcome Trust Centre for Clinical Tropical Medicine, Division of Medicine, and Academic Department of Paediatrics, Imperial College, London, United Kingdom; Staten Serum Institute, Copenhagen, Denmark; and MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| |
Collapse
|
20
|
Airway luminal T cells: a newcomer on the stage of TB vaccination strategies. Trends Immunol 2010; 31:247-52. [PMID: 20542470 DOI: 10.1016/j.it.2010.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 04/26/2010] [Accepted: 05/03/2010] [Indexed: 01/25/2023]
Abstract
Protection against pulmonary tuberculosis (TB) by vaccination is often ascribed to the presence of TB-reactive T cells in the lung before infection. Challenging this view, new studies analyzing vaccine-induced T cells in various tissue compartments after parenteral immunization suggest a poor correlation between the presence of anti-TB T cells in the lung interstitium and spleen before Mycobacterium tuberculosis exposure and protection. In contrast, respiratory mucosal immunization leads to distribution of T cells not only in the lung interstitium and spleen, but also in the airway lumen, and the presence of these cells correlates well with protection. Furthermore, airway luminal recruitment of parenteral vaccine-induced T cells in peripheral tissues prior to M. tuberculosis challenge restores protection. We propose that understanding the biology of airway luminal T cells holds important implications for developing effective TB vaccination strategies.
Collapse
|
21
|
Jeyanathan M, Mu J, McCormick S, Damjanovic D, Small CL, Shaler CR, Kugathasan K, Xing Z. Murine airway luminal antituberculosis memory CD8 T cells by mucosal immunization are maintained via antigen-driven in situ proliferation, independent of peripheral T cell recruitment. Am J Respir Crit Care Med 2009; 181:862-72. [PMID: 20019338 DOI: 10.1164/rccm.200910-1583oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The airway luminal memory CD8 T cells induced by respiratory mucosal immunization in a murine model have been found to be critical to antituberculosis immunity. However, the mechanisms of their maintenance on airway mucosal surface still remain poorly understood. OBJECTIVES Using a model of adenovirus-based intranasal immunization we investigated the immune property and the mechanisms of maintenance of airway luminal CD8 T cells. METHODS Immune properties of airway luminal Mycobacterium tuberculosis antigen-specific CD8 T cells were examined. Proliferation of airway luminal CD8 T cells was determined by in vivo T cell-labeling techniques. The role of peripheral T cell recruitment in maintaining airway luminal CD8 T cells was investigated by blocking lymphocyte trafficking from lymphoid and peripheral tissues. The requirement of M. tuberculosis antigens for in situ T cell proliferation was evaluated using a T cell transfer approach. An airway M. tuberculosis challenge model was used to study the relationship between CD8 T cell-mediated protection and peripheral T cell recruitment. MEASUREMENTS AND MAIN RESULTS Intranasal immunization leads to elicitation of persisting M. tuberculosis antigen-specific CD8 T cells in the airway lumen, which display an activated effector memory phenotype different from those in peripheral tissues. Airway luminal T cells continuously proliferate in an antigen-dependent manner, and can be maintained even in the absence of peripheral T cell recruitment. The lungs equipped with such CD8 T cells are protected from airway M. tuberculosis challenge independent of both peripheral T cell supply and CD4 T cells. CONCLUSIONS Vaccine-inducible airway luminal antituberculosis memory CD8 T cells are self-renewable in an antigen-dependent manner, and can be maintained independent of peripheral T cell supply.
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- Centre for Gene Therapeutics, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario L8N 3Z5, Canada
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Pagliari C, Pereira NV, Kanashiro L, Stegun FW, Croda J, Duarte MIS, Sotto MN. Characterization of cytotoxic immune response in skin and mucosal lesions of paracoccidioidomycosis. J Cutan Pathol 2009; 37:565-70. [PMID: 19702683 DOI: 10.1111/j.1600-0560.2009.01413.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND CD8+ T cells and natural killer (NK) cells are involved in the immune response against some pathogens. For this purpose, we investigated the in situ paracoccidioidomycosis (PCM) immune response addressing the participation of NK cells, CD8+ T cells, perforin and granzyme B expression. METHODS Sixty biopsies of PCM skin and mucosa were classified according to the presence of compact granulomas (G1), poorly organized granulomas (G2) and both kinds in the same lesion (G3). CD8+ T cells, NK cells, perforin and granzyme B were showed by immunohistochemistry. RESULTS CD8+ T cells were increased over NK cells in cutaneous G1 and G2 lesions. There was no difference regarding such cells in G3 lesions, although they were abundant in such lesions. In mucosa, CD8+ T cells were increased in number over NK cells in all groups. Granzyme B in skin increased in G2 and G3. The number of granzyme did not differ in mucosal lesions in the three groups. CONCLUSIONS CD8+ T cells and NK cells play a role in PCM cutaneous and mucosal lesions. The predominance of CD8+ T cells over NK cells may represent an effective response against the fungi. Moreover, the high number of granzyme B expressing cells corroborates this possibility.
Collapse
Affiliation(s)
- Carla Pagliari
- Laboratory of the Discipline of Pathology of Transmissible Diseases, Department of Pathology, Faculty of Medical Sciences, University of São Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
23
|
Sun R, Skeiky YAW, Izzo A, Dheenadhayalan V, Imam Z, Penn E, Stagliano K, Haddock S, Mueller S, Fulkerson J, Scanga C, Grover A, Derrick SC, Morris S, Hone DM, Horwitz MA, Kaufmann SHE, Sadoff JC. Novel recombinant BCG expressing perfringolysin O and the over-expression of key immunodominant antigens; pre-clinical characterization, safety and protection against challenge with Mycobacterium tuberculosis. Vaccine 2009; 27:4412-23. [PMID: 19500523 DOI: 10.1016/j.vaccine.2009.05.048] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 05/07/2009] [Accepted: 05/14/2009] [Indexed: 11/28/2022]
Affiliation(s)
- Ronggai Sun
- Aeras Global TB Vaccine Foundation, 1405 Research Blvd., Rockville, MD 20850, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
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: 10] [Impact Index Per Article: 0.6] [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]
|
25
|
Ryan AA, Nambiar JK, Wozniak TM, Roediger B, Shklovskaya E, Britton WJ, Fazekas de St. Groth B, Triccas JA. Antigen Load Governs the Differential Priming of CD8 T Cells in Response to the Bacille Calmette Guérin Vaccine orMycobacterium tuberculosisInfection. THE JOURNAL OF IMMUNOLOGY 2009; 182:7172-7. [DOI: 10.4049/jimmunol.0801694] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
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: 60] [Impact Index Per Article: 3.8] [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
|
27
|
Immunotherapy with combined DNA vaccines is an effective treatment for M. bovis infection in cattle. Vaccine 2009; 27:1317-22. [DOI: 10.1016/j.vaccine.2008.12.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 12/17/2008] [Accepted: 12/28/2008] [Indexed: 11/17/2022]
|
28
|
Therapeutic efficacy of a tuberculosis DNA vaccine encoding heat shock protein 65 of Mycobacterium tuberculosis and the human interleukin 2 fusion gene. Tuberculosis (Edinb) 2009; 89:54-61. [DOI: 10.1016/j.tube.2008.09.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2008] [Revised: 09/01/2008] [Accepted: 09/17/2008] [Indexed: 01/08/2023]
|
29
|
Woodworth JS, Wu Y, Behar SM. Mycobacterium tuberculosis-specific CD8+ T cells require perforin to kill target cells and provide protection in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:8595-603. [PMID: 19050279 PMCID: PMC3133658 DOI: 10.4049/jimmunol.181.12.8595] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Optimal immunity to Mycobacterium tuberculosis (Mtb) infection requires CD8(+) T cells, and several current Mtb vaccine candidates are being engineered to elicit enhanced CD8(+) T cell responses. However, the function of these T cells and the mechanism by which they provide protection is still unknown. We have previously shown that CD8(+) T cells specific for the mycobacterial Ags CFP10 and TB10.4 accumulate in the lungs of mice following Mtb infection and have cytolytic activity in vivo. In this study, we determine which cytolytic pathways are used by these CD8(+) T cells during Mtb infection. We find that Mtb-specific CD8(+) T cells lacking perforin have reduced cytolytic capacity in vivo. In the absence of perforin, the residual cytolytic activity is CD95 and TNFR dependent. This is particularly true in Mtb-infected lung tissue where disruption of both perforin and CD95 eliminates target cell lysis. Moreover, adoptive transfer of immune CD8(+) T cells isolated from wild-type, but not perforin-deficient mice, protect recipient mice from Mtb infection. We conclude that CD8(+) T cells elicited following Mtb infection use several cytolytic pathways in a hierarchical and compensatory manner dominated by perforin-mediated cytolysis. Finally, although several cytolytic pathways are available, adoptively transferred Mtb-specific CD8(+) T cells require perforin-mediated cytolysis to protect animals from infection. These data show that CD8(+) T cell-mediated protection during Mtb infection requires more than the secretion of IFN-gamma and specifically defines the CD8(+) cytolytic mechanisms utilized and required in vivo.
Collapse
MESH Headings
- Adoptive Transfer
- Amino Acid Sequence
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/microbiology
- Cytotoxicity Tests, Immunologic
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Female
- Histocompatibility Antigens Class I/administration & dosage
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/toxicity
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Mycobacterium tuberculosis/immunology
- Perforin/administration & dosage
- Perforin/deficiency
- Perforin/physiology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/microbiology
- T-Lymphocytes, Cytotoxic/transplantation
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/metabolism
- Tuberculosis, Pulmonary/prevention & control
Collapse
Affiliation(s)
- Joshua S. Woodworth
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Ying Wu
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Samuel M. Behar
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
30
|
Kathaperumal K, Kumanan V, McDonough S, Chen LH, Park SU, Moreira MAS, Akey B, Huntley J, Chang CF, Chang YF. Evaluation of immune responses and protective efficacy in a goat model following immunization with a coctail of recombinant antigens and a polyprotein of Mycobacterium avium subsp. paratuberculosis. Vaccine 2008; 27:123-35. [PMID: 18955101 DOI: 10.1016/j.vaccine.2008.10.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 09/30/2008] [Accepted: 10/02/2008] [Indexed: 01/27/2023]
Abstract
The protective efficacy of four recombinant antigens (85A, 85B, superoxide dismutase [SOD], and a fusion polypeptide [Map74F]) of Mycobacterium avium subsp. paratuberculosis (MAP) along with the adjuvant dimethydioctadecyl ammonium bromide (DDA) was assessed in a goat challenge model. Animals were immunized with the four antigens with adjuvant DDA (Group I, eight goat kids) or without the adjuvant (Group II, eight goat kids) or adjuvant only (Group III, nine goat kids). Animals were boostered 3 weeks after the primary vaccination and challenged 3 weeks after the booster. Significant antigen-specific lymphoproliferation was observed in the immunized animals 3 weeks after the booster immunization. This response increased further at 4 weeks after the booster. Similarly, antigen-specific IFN-gamma responses increased in the immunized animals 3 weeks after the booster. The response was significantly higher for 85A and Map74F at 10 weeks after primary vaccination (APV) in Group I animals compared to the other two groups. CD4+ T-cell populations were higher in the vaccinated animals from 6 to 10 weeks APV than those of the control animals. A significant increase in recombinant antigen-specific IFN-gamma gene expression was detected in the vaccinated animals. At necropsy (38 weeks APV), our multicomponent subunit vaccine imparted a significant protection in terms of reduction of MAP burden in target organs as compared to sham-immunized goats. This study indicates that our multicomponent subunit vaccine induced a good Th1 response and conferred protection against MAP infection in a goat challenge model.
Collapse
Affiliation(s)
- Kumanan Kathaperumal
- Animal Health Diagnostic Center, Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, UpTwoer Road, Ithaca, NY 14853, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mycobacterium bovis BCG immunization induces protective immunity against nine different Mycobacterium tuberculosis strains in mice. Infect Immun 2008; 76:5173-80. [PMID: 18710860 DOI: 10.1128/iai.00019-08] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Recent preclinical and epidemiologic studies have suggested that certain Mycobacterium tuberculosis genotypes (in particular, Beijing lineage strains) may be resistant to Mycobacterium bovis BCG vaccine-induced antituberculosis protective immunity. To investigate the strain specificity of BCG-induced protective responses in a murine model of pulmonary tuberculosis, C57BL/6 mice were vaccinated with BCG vaccine and then challenged 2 months later with one of nine M. tuberculosis isolates. Four of these strains were from the W-Beijing lineage (HN878, N4, NHN5, and ChS) while four were non-Beijing-type isolates (C913, CDC1551, NY669, and NY920). As a control, the WHO standard M. tuberculosis Erdman strain was evaluated in these vaccination/challenge experiments. To assess the protective responses evoked by BCG immunization, organ bacterial burdens and lung pathology were assessed in vaccinated and naïve mice at 4, 12, and 20 weeks postchallenge as well as during the day of infection. At 4 weeks after the aerosol challenge with each of these strains, significantly reduced bacterial growth in the lungs and spleens and significantly improved lung pathology were seen in all vaccinated animals compared to naïve controls. After 12 weeks, reduced organ bacterial burdens were detected in vaccinated animals infected with six of nine challenge strains. Although lung CFU values were lower in vaccinated mice for only three of nine groups at 20 weeks postchallenge, significantly decreased lung inflammation was seen in all immunized animals relative to controls at 20 weeks postchallenge. Taken together, these data demonstrate that BCG vaccination protects against infection with diverse M. tuberculosis strains in the mouse model of pulmonary tuberculosis and suggest that strain-specific resistance to BCG-induced protective immunity may be uncommon.
Collapse
|
32
|
Park SU, Kathaperumal K, McDonough S, Akey B, Huntley J, Bannantine JP, Chang YF. Immunization with a DNA vaccine cocktail induces a Th1 response and protects mice against Mycobacterium avium subsp. paratuberculosis challenge. Vaccine 2008; 26:4329-37. [PMID: 18582521 DOI: 10.1016/j.vaccine.2008.06.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 06/02/2008] [Accepted: 06/04/2008] [Indexed: 10/21/2022]
Abstract
Several antigens of Mycobacterium avium subsp. paratuberculosis have been studied as vaccine components and their immunogenicity has been evaluated. Previously, we reported that 85 antigen complex (85A, 85B, and 85C), superoxide dismutase (SOD), and 35kDa protein could induce significant lymphocyte proliferation as well as the elaboration of Th1-associated cytokines including interferon gamma (IFN-gamma), interleukin-2 (IL-2), IL-12 and tumor necrosis factor alpha (TNF-alpha). Based on these results, we cloned and expressed 85A, 85B, 85C, SOD, and 35kDa-protein genes into the eukaryotic expression plasmid pVR1020. C57BL/6 mice were immunized three times intramuscularly with the recombinant DNA cocktail and pVR1020 DNA alone as control. A significant reduction in the bacterial burden in the spleen and liver of mice immunized with the DNA cocktail as compared to the vector control group was found. Also, the relative severity of the liver and spleen histopathology paralleled the MAP culture results, more granulomas and acid-fast bacilli in the vector control animals. Moreover, mice immunized with the DNA cocktail developed both CD4(+) and CD8(+) T cell responses to the recombinant antigens and showed significant lymphocyte proliferation. The Th1 response related cytokine (IFN-gamma) levels increased in splenocytes obtained from immunized animals. These results indicate that the use of a recombinant DNA vaccine can provide protective immunity against mycobacterial infection by inducing a Th1 response.
Collapse
Affiliation(s)
- Sung-Un Park
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Chang-hong S, Xiao-wu W, Hai Z, Ting-fen Z, Li-Mei W, Zhi-kai X. Immune responses and protective efficacy of the gene vaccine expressing Ag85B and ESAT6 fusion protein from Mycobacterium tuberculosis. DNA Cell Biol 2008; 27:199-207. [PMID: 18163878 DOI: 10.1089/dna.2007.0648] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Genetic immunity is a new promising approach for the development of novel tuberculosis vaccines. In this study, it is shown that DNA vaccines expressing the fusion protein of antigen 85B (Ag85B) and early secreted antigenic target 6-kDa antigen (ESAT6) can induce high levels of specific IgG2a antibody subtype in the mice. With the prolongation of postimmunization time, the levels of IgG2a antibody decrease gradually. Although a high-level specific IgG2a antibody subtype is also elicited by classical BCG, the ratio of antibody subtypes IgG2a to IgG1 changes 4 weeks after immunization, and IgG1 is gradually shifted to the main antibody subtype. DNA vaccines also elicit cellular immunity as shown by specific spleen lymphocytes proliferation to Ag85B or ESAT6 protein and the production of high levels of IFN-gamma and IL-2, which is similar to that elicited by BCG. Vaccination of mice with DNA vaccines expressing the fusion protein Ag85B-ESAT6 results in a significant level of protection against the subsequent high-dose challenge with virulent Mycobacterium tuberculosis (MTB) H37Rv. Dramatic reduction in the number of MTB colony-forming units in the spleens and lungs is observed. Pathological examination showed that recombinant plasmid and BCG groups have only minor damage and organizational structures that are kept relatively complete, while in the control group, spleens and lungs are damaged seriously. Therefore, although the reducing degree of mycobacterial loads in the organ of mice immunized with recombinant plasmid is not more than that of BCG, through the analysis of pathological changes, we may conclude that the protective effect provided by DNA vaccine expressing the Ag85B-ESAT6 fusion protein is equivalent to that afforded by the classical BCG.
Collapse
Affiliation(s)
- Shi Chang-hong
- Lab Animal Center, The Fourth Military Medical University, Xi'an, China.
| | | | | | | | | | | |
Collapse
|
34
|
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
|
35
|
Abstract
Tuberculosis continues to cause considerable human morbidity and mortality worldwide, particularly in people coinfected with HIV. The emergence of multidrug resistance makes the medical treatment of tuberculosis even more difficult. Thus, the development of a tuberculosis vaccine is a global health priority. Here we review the data concerning the role of CD8+ T cells in immunity to tuberculosis and consider how CD8+ T cells can be elicited by vaccination. Many immunization strategies have the potential to elicit CD8+ T cells and we critically review the data supporting a role for vaccine-induced CD8+ T cells in protective immunity. The synergy between CD4+ and CD8+ T cells suggests that a vaccine that elicits both T-cell subsets has the best chance at preventing tuberculosis.
Collapse
Affiliation(s)
- Samuel M. Behar
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Smith Building Room 516C, One Jimmy Fund Way, Boston, MA 02115. Phone: (617)-525-1033, Fax: (617)-525-1010
| | - Joshua S.M. Woodworth
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Smith Building Room 516C, One Jimmy Fund Way, Boston, MA 02115. Phone: (617)-525-1065, Fax: (617)-525-1010
| | - Ying Wu
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Smith Building Room 516C, One Jimmy Fund Way, Boston, MA 02115. Phone: (617)-525-1042, Fax: (617)-525-1010
| |
Collapse
|
36
|
Radosevic K, Wieland CW, Rodriguez A, Weverling GJ, Mintardjo R, Gillissen G, Vogels R, Skeiky YAW, Hone DM, Sadoff JC, van der Poll T, Havenga M, Goudsmit J. Protective immune responses to a recombinant adenovirus type 35 tuberculosis vaccine in two mouse strains: CD4 and CD8 T-cell epitope mapping and role of gamma interferon. Infect Immun 2007; 75:4105-15. [PMID: 17526747 PMCID: PMC1951991 DOI: 10.1128/iai.00004-07] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There is an urgent need for an efficacious vaccine against tuberculosis (TB). Cellular immune responses are key to an effective protective response against TB. Recombinant adenovirus (rAd) vectors are especially suited to the induction of strong T-cell immunity and thus represent promising vaccine vehicles for the prevention of TB. We have previously reported on rAd vector serotype 35, the serotype of choice due to low preexisting immunity worldwide, which expresses a unique fusion protein of Mycobacterium tuberculosis antigens Ag85A, Ag85B, and TB10.4 (Ad35-TBS). Here, we demonstrate that Ad35-TBS confers protection against M. tuberculosis when administered to mice through either an intranasal or an intramuscular route. Histological evaluation of lung tissue corroborated the protection and, in addition, demonstrated differences between two mouse strains, with diffuse inflammation in BALB/c mice and distinct granuloma formation in C57BL/6 mice. Epitope mapping analysis in these mouse strains showed that the major T-cell epitopes are conserved in the artificial fusion protein, while three novel CD8 peptides were discovered. Using a defined set of T-cell epitopes, we reveal differences between the two mouse strains in the type of protective immune response, demonstrating that different antigen-specific gamma interferon (IFN-gamma)-producing T cells can provide protection against M. tuberculosis challenge. While in BALB/c (H-2(d)) mice, a dominant CD8 T-cell response was detected, in C57BL/6 (H-2(b)) mice, more balanced CD4/CD8 T-cell responses were observed, with a more pronounced CD4 response in the lungs. These results unify conflicting reports on the relative importance of CD4 versus CD8 T-cell responses in protection and emphasize the key role of IFN-gamma.
Collapse
MESH Headings
- Acyltransferases/genetics
- Acyltransferases/immunology
- Adenoviridae/genetics
- Administration, Intranasal
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Colony Count, Microbial
- Disease Models, Animal
- Epitope Mapping
- Epitopes, T-Lymphocyte/immunology
- Female
- Genetic Vectors
- Injections, Intramuscular
- Interferon-gamma/immunology
- Liver/microbiology
- Lung/microbiology
- Lung/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Spleen/microbiology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/genetics
- Tuberculosis Vaccines/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
|
37
|
Sierra VG. Is a new tuberculosis vaccine necessary and feasible? A Cuban opinion. Tuberculosis (Edinb) 2007; 86:169-78. [PMID: 16677860 DOI: 10.1016/j.tube.2006.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 03/08/2006] [Indexed: 10/24/2022]
Abstract
Mycobacterium tuberculosis kills more human beings worldwide than any other pathogen. An estimated two billion people are already infected with the bacterium. In 2006, tuberculosis (TB) will kill nearly one million more people than in 1992. In Cuba, TB is not a serious health problem any more and we are striving to eliminate it in the near future. The most widely applied human vaccine in the world is BCG. It is also a safe vaccine except when it is applied to immunocompromised persons. Its protective efficacy is a controversial topic. In spite of this, more than 80 years of experience with this vaccine has demonstrated that BCG is effective, at least in significantly preventing childhood TB, including the meningeal and disseminated forms of the disease, but does not protect against the predominant pulmonary form of the disease in adults, which means that our best TB vaccine now is inadequate; we therefore need a new vaccine. The following facts, apart from the experience with BCG, support discussion about the feasibility of a new and better TB vaccine: Less than 10% of the 2 billion TB infected persons develop active disease. It has been demonstrated that HIV + TB co-infection increases 30 times the risk of contracting active TB and it increases the risk of being killed by the bacterium. Some new vaccine candidates, now under development and evaluation, are showing promising results in preclinical studies, and a few of them have entered clinical trials. There seems to be a consensus that a new TB vaccine will be feasible, but some challenging issues must be positively solved, such as, the lack of universally accepted correlates for protection, improved diagnostics, and final vaccine efficacy evaluation conducted on large phase III clinical trials in underdeveloped countries. The ethical, economical, organizational and scientific questions involved in this global task are enormous, but feasible.
Collapse
|
38
|
Ngai P, McCormick S, Small C, Zhang X, Zganiacz A, Aoki N, Xing Z. Gamma interferon responses of CD4 and CD8 T-cell subsets are quantitatively different and independent of each other during pulmonary Mycobacterium bovis BCG infection. Infect Immun 2007; 75:2244-52. [PMID: 17307945 PMCID: PMC1865770 DOI: 10.1128/iai.00024-07] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Gamma interferon (IFN-gamma) is a key cytokine in host defense against intracellular mycobacterial infection. It has been believed that both CD4 and CD8 T cells are the primary sources of IFN-gamma. However, the relative contributions of CD4 and CD8 T-cell subsets to IFN-gamma production and the relationship between CD4 and CD8 T-cell activation have not been examined. By using a model of pulmonary mycobacterial infection and various immunodetection assays, we found that CD4 T cells mounted a much stronger IFN-gamma response than CD8 T cells at various times after mycobacterial infection, and this pronounced IFN-gamma production by CD4 T cells was attributed to both greater numbers of antigen-specific CD4 T cells and a greater IFN-gamma secretion capacity of these cells. By using major histocompatibility complex class II-deficient or CD4-deficient mice, we found that the lack of CD4 T cells did not negatively affect primary or secondary CD8 T-cell IFN-gamma responses. The CD8 T cells activated in the absence of CD4 T cells were capable of immune protection against secondary mycobacterial challenge. Our results suggest that, whereas both CD4 and CD8 T cells are capable of IFN-gamma production, the former represent a much greater cellular source of IFN-gamma. Moreover, during mycobacterial infection, CD8 T-cell IFN-gamma responses and activation are independent of CD4 T-cell activation.
Collapse
Affiliation(s)
- Patricia Ngai
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. West, Hamilton, Ontario L8N 3Z5, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Bennekov T, Dietrich J, Rosenkrands I, Stryhn A, Doherty TM, Andersen P. Alteration of epitope recognition pattern in Ag85B and ESAT-6 has a profound influence on vaccine-induced protection against Mycobacterium tuberculosis. Eur J Immunol 2007; 36:3346-55. [PMID: 17109467 DOI: 10.1002/eji.200636128] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To analyze the effect of vaccine delivery systems on antigen recognition and vaccine efficacy, we compared immune responses in mice immunized either with an adenovirus vector expressing a fusion of Ag85B and ESAT-6 or with the recombinant fusion protein in a liposomal adjuvant. Both vaccines induced high levels of antigen-specific IFN-gamma production. The adjuvanted protein vaccine induced primarily a CD4 T cell response directed to the epitope Ag85B(241-255) and gave efficient protection against subsequent Mycobacterium tuberculosis infection. In contrast, the adenoviral construct induced a strong CD8 response predominantly targeted to the epitope ESAT-6(15-29) and no significant protection against infection. Vaccination with the protein vaccine resulted in highly accelerated recall of Ag85B(241-255)-specific T cells immediately post M. tuberculosis challenge whereas the ESAT-6(15-29) epitope was barely recognized during infection. Delivery of the viral construct in cationic liposomes switched the immune response to a protective one dominated by CD4 T cells targeted to the Ag85B(241-255) epitope. These data demonstrate that the nature of the T cell response to a vaccine antigen is more important than its magnitude with respect to protective efficacy and that vaccine-mediated changes in immunodominance can result in T cell responses of limited relevance during the natural infection.
Collapse
MESH Headings
- Acyltransferases/genetics
- Acyltransferases/immunology
- Acyltransferases/metabolism
- Adenoviridae/genetics
- Adenoviridae/immunology
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Cell Line
- Epitope Mapping
- Female
- Immunologic Memory/genetics
- Mice
- Mice, Inbred C57BL
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Tuberculosis/immunology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/genetics
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Thomas Bennekov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
40
|
Hogan LH, Co DO, Karman J, Heninger E, Suresh M, Sandor M. Virally activated CD8 T cells home to Mycobacterium bovis BCG-induced granulomas but enhance antimycobacterial protection only in immunodeficient mice. Infect Immun 2006; 75:1154-66. [PMID: 17178783 PMCID: PMC1828579 DOI: 10.1128/iai.00943-06] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The effect of secondary infections on CD4 T-cell-regulated chronic granulomatous inflammation is not well understood. Here, we have investigated the effect of an acute viral infection on the cellular composition and bacterial protection in Mycobacterium bovis strain bacille Calmette-Guérin (BCG)-induced granulomas using an immunocompetent and a partially immunodeficient murine model. Acute lymphocytic choriomeningitis virus (LCMV) coinfection of C57BL/6 mice led to substantial accumulation of gamma interferon (IFN-gamma)-producing LCMV-specific T cells in liver granulomas and increased local IFN-gamma. Despite traffic of activated T cells that resulted in a CD8 T-cell-dominated granuloma, the BCG liver organ load was unaltered from control levels. In OT-1 T-cell-receptor (TCR) transgenic mice, ovalbumin (OVA) immunization or LCMV coinfection of BCG-infected mice induced CD8 T-cell-dominated granulomas containing large numbers of non-BCG-specific activated T cells. The higher baseline BCG organ load in this CD8 TCR transgenic animal allowed us to demonstrate that OVA immunization and LCMV coinfection increased anti-BCG protection. The bacterial load remained substantially higher than in mice with a more complete TCR repertoire. Overall, the present study suggests that peripherally activated CD8 T cells can be recruited to chronic inflammatory sites, but their contribution to protective immunity is limited to conditions of underlying immunodeficiency.
Collapse
Affiliation(s)
- Laura H Hogan
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, 1300 University Ave., Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
41
|
Sada-Ovalle I, Torre-Bouscoulet L, Valdez-Vázquez R, Martínez-Cairo S, Zenteno E, Lascurain R. Characterization of a cytotoxic CD57+ T cell subset from patients with pulmonary tuberculosis. Clin Immunol 2006; 121:314-23. [PMID: 17035093 DOI: 10.1016/j.clim.2006.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 07/28/2006] [Accepted: 08/18/2006] [Indexed: 11/26/2022]
Abstract
We investigated the proportion, phenotype, and cytotoxicity of CD8+CD57+ and CD57- T cells in peripheral blood from 20 tuberculosis (TB)-patients and 20 healthy tuberculin skin test-positive donors. Our results showed an increase in CD8+CD57+ T cells from TB-patients as compared with those from age-matched healthy donors (p<0.0001). CD8+CD57+ T cells from TB-patients expressed CD69, perforin, granzyme-A, and a CD28-CD62L-CD161- phenotype without recognition for the alpha-galactosylceramide-CD1d complex. This cell subset also expressed TNF-alpha and IFN-gamma, under phorbol-myristate-acetate/ionomycin stimulation. Interestingly, the cytotoxicity against autologous monocytes was higher in CD57- cells from TB-patients and donors than their CD57+ counterparts, in the presence of Mycobacterium tuberculosis H37Rv culture filtrate. However, only CD8+CD57+ T cells from TB-patients exhibited spontaneous cytotoxicity against monocytes in the absence of antigen. Our results suggest that CD8+CD57+ T cells are a subset of effector cells that could be helpful to evaluate the cell-mediated immune response to M. tuberculosis.
Collapse
Affiliation(s)
- I Sada-Ovalle
- Departamento de Bioquímica, Instituto Nacional de Enfermedades, Respiratorias, Mexico
| | | | | | | | | | | |
Collapse
|
42
|
Derrick SC, Evering TH, Sambandamurthy VK, Jalapathy KV, Hsu T, Chen B, Chen M, Russell RG, Junqueira-Kipnis AP, Orme IM, Porcelli SA, Jacobs WR, Morris SL. Characterization of the protective T-cell response generated in CD4-deficient mice by a live attenuated Mycobacterium tuberculosis vaccine. Immunology 2006; 120:192-206. [PMID: 17076705 PMCID: PMC2265854 DOI: 10.1111/j.1365-2567.2006.02491.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The global epidemic of tuberculosis, fuelled by acquired immune-deficiency syndrome, necessitates the development of a safe and effective vaccine. We have constructed a DeltaRD1DeltapanCD mutant of Mycobacterium tuberculosis (mc(2)6030) that undergoes limited replication and is severely attenuated in immunocompromised mice, yet induces significant protection against tuberculosis in wild-type mice and even in mice that completely lack CD4(+) T cells as a result of targeted disruption of their CD4 genes (CD4(-/-) mice). Ex vivo studies of T cells from mc(2)6030-immunized mice showed that these immune cells responded to protein antigens of M. tuberculosis in a major histocompatibility complex (MHC) class II-restricted manner. Antibody depletion experiments showed that antituberculosis protective responses in the lung were not diminished by removal of CD8(+), T-cell receptor gammadelta (TCR-gammadelta(+)) and NK1.1(+) T cells from vaccinated CD4(-/-) mice before challenge, implying that the observed recall and immune effector functions resulting from vaccination of CD4(-/-) mice with mc(2)6030 were attributable to a population of CD4(-) CD8(-) (double-negative) TCR-alphabeta(+), TCR-gammadelta(-), NK1.1(-) T cells. Transfer of highly enriched double-negative TCR-alphabeta(+) T cells from mc(2)6030-immunized CD4(-/-) mice into naive CD4(-/-) mice resulted in significant protection against an aerosol tuberculosis challenge. Enriched pulmonary double-negative T cells transcribed significantly more interferon-gamma and interleukin-2 mRNA than double-negative T cells from naive mice after a tuberculous challenge. These results confirmed previous findings on the potential for a subset of MHC class II-restricted T cells to develop and function without expression of CD4 and suggest novel vaccination strategies to assist in the control of tuberculosis in human immunodeficiency virus-infected humans who have chronic depletion of their CD4(+) T cells.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Gene Expression Regulation/immunology
- Immunity, Cellular
- Immunocompromised Host
- Interferon-gamma/biosynthesis
- Interleukin-2/biosynthesis
- Interleukin-2/genetics
- Lymphocyte Activation
- Mice
- Mice, Inbred Strains
- Mycobacterium tuberculosis/immunology
- RNA, Messenger/genetics
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Tuberculosis Vaccines/immunology
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/pathology
- Tuberculosis, Pulmonary/prevention & control
- Vaccination/methods
- Vaccines, Attenuated/immunology
Collapse
Affiliation(s)
- Steven C Derrick
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cai H, Yu DH, Hu XD, Li SX, Zhu YX. A combined DNA vaccine-prime, BCG-boost strategy results in better protection against Mycobacterium bovis challenge. DNA Cell Biol 2006; 25:438-47. [PMID: 16907641 DOI: 10.1089/dna.2006.25.438] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In this study, we demonstrated that calves vaccinated with a combined DNA vaccine encoding Ag85B, MPT- 64, and MPT-83 antigens from the Mycobacterium tuberculosis for the priming and subsequently boosting with BCG prior to experimental challenge with virulent Mycobacterium bovis (M. bovis) resulted in improved immune responses over immunizing. Vaccination with the combined DNA/BCG induced higher levels of antigen- specific gamma interferon (IFN-gamma) in whole-blood cultures 4 weeks after final vaccination and the level of antigen-specific IFN-gamma in response to Ag85, MPT-64, and MPT-83 were still higher 4 weeks after challenge when compared to the combined DNA group. There was a significant bias toward induction of CD4+ T cells rather than CD8+ T cells responses, and the mean percentage of CD4+ T cells was increased about 2.6-fold in peripheral blood mononuclear cells (PBMC) cultures in DNA prime-BCG boost vaccination when compared to the nonvaccinated group. In addition, DNA prime-BCG boost vaccination resulted in stronger humoral immune responses, and the levels of the specific antibodies to three antigens were increased two- to 32- fold when compared to the combined DNA group. Vaccination with the combined DNA/BCG induced a high level of protection against an intratracheal challenge with virulent M. bovis, based on a significant enhancement of six pathological and microbiological parameters of protection compared to the nonvaccinated group. Finally, the combined DNA/BCG increased the protective efficacy by more than 10-100-fold as measured by reduced CFU counts in the lungs from calves challenged with M. bovis compared to the combined DNA and BCG groups. These results suggest that use of the prime-boost strategy offers better protection against bovine tuberculosis than does the combined DNA vaccines and BCG.
Collapse
Affiliation(s)
- H Cai
- National Laboratory of Protein Engineering and Plant Genetic Engineering, Peking University, Beijing, People's Republic of China.
| | | | | | | | | |
Collapse
|
44
|
Sambandamurthy VK, Derrick SC, Hsu T, Chen B, Larsen MH, Jalapathy KV, Chen M, Kim J, Porcelli SA, Chan J, Morris SL, Jacobs WR. Mycobacterium tuberculosis ΔRD1 ΔpanCD: A safe and limited replicating mutant strain that protects immunocompetent and immunocompromised mice against experimental tuberculosis. Vaccine 2006; 24:6309-20. [PMID: 16860907 DOI: 10.1016/j.vaccine.2006.05.097] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Revised: 05/12/2006] [Accepted: 05/22/2006] [Indexed: 11/28/2022]
Abstract
The global epidemic of tuberculosis (TB), fueled by the growing HIV pandemic, warrants the development of a safe and effective vaccine against TB. We report the construction and characterization of an unlinked double deletion mutant of Mycobacterium tuberculosis H37Rv that deletes both the primary attenuating mutation of BCG (DeltaRD1) and two genes required for the synthesis of pantothenate (DeltapanCD). The M. tuberculosis DeltaRD1 DeltapanCD (mc(2)6030) mutant undergoes limited replication in mice, and yet is both significantly safer than BCG in immunocompromised mice and also safe in guinea pigs. Additionally, the mc(2)6030 strain does not reactivate in a mouse chemo-immunosuppression model. Importantly, long-lived protective immune responses following immunization with the mc(2)6030 strain prolong the survival of wild type mice, and CD4-deficient mice against an aerosol challenge with virulent M. tuberculosis. Given its overall safety and effectiveness, the mc(2)6030 live attenuated strain should be considered as a human vaccine candidate for protecting both healthy and HIV-infected individuals against TB.
Collapse
Affiliation(s)
- Vasan K Sambandamurthy
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Tuberculosis (TB), an ancient human scourge, is a growing health problem in the developing world. Approximately two million deaths each year are caused by TB, which is the leading cause of death in HIV-infected individuals. Clearly, an improved TB vaccine is desperately needed. Heterologous prime-boost regimens probably represent the best hope for an improved vaccine regimen to prevent TB. This first generation of new vaccines might also complement drug treatment regimens and be effective against reactivation of TB from the latent state, which would significantly enhance their usefulness.
Collapse
Affiliation(s)
- Yasir A W Skeiky
- Aeras Global TB Vaccine Foundation, 1405 Research Blvd, Rockville, Maryland 20850, USA.
| | | |
Collapse
|
46
|
Selection of novel TB vaccine candidates and their evaluation as DNA vaccines against aerosol challenge. Vaccine 2006; 24:6340-50. [PMID: 16781800 DOI: 10.1016/j.vaccine.2006.05.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 05/09/2006] [Accepted: 05/15/2006] [Indexed: 10/24/2022]
Abstract
Putative TB vaccine candidates were selected from lists of genes induced in response to in vivo-like stimuli, such as low oxygen and carbon starvation or growth in macrophages, and tested as plasmid DNA vaccines for their ability to protect against Mycobacterium tuberculosis challenge in a guinea pig aerosol infection model. This vaccination method was chosen as it induces the Th1 cell-mediated immune response required against intracellular pathogens such as M. tuberculosis. Protection was assessed in the guinea pig model in terms of mycobacteria present in the lungs at 30 days post-challenge. Protection achieved by the novel candidates was compared to BCG (positive control) and saline (negative control). Four vaccines encoding for proteins such as PE and PPE proteins, a zinc metalloprotease and an acyltransferase, gave a level of protection that was statistically better than saline in the lungs. These findings have enabled us to focus on a sub-set of vaccine candidates for further evaluation using additional vaccination strategies.
Collapse
|
47
|
Vipond J, Clark SO, Hatch GJ, Vipond R, Marie Agger E, Tree JA, Williams A, Marsh PD. Re-formulation of selected DNA vaccine candidates and their evaluation as protein vaccines using a guinea pig aerosol infection model of tuberculosis. Tuberculosis (Edinb) 2006; 86:218-24. [PMID: 16520093 DOI: 10.1016/j.tube.2006.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Accepted: 01/20/2006] [Indexed: 10/24/2022]
Abstract
A selection of previously identified protective Mycobacterium tuberculosis DNA vaccines were re-formulated as proteins and administered with a Th1-inducing adjuvant to help stimulate the relevant immune responses necessary for protection. All three candidate-vaccines conferred high levels of antigen-specific cellular and humoral responses, as indicated by lymphocyte proliferation and serum IgG levels. Protective efficacy was also assessed in comparison with the current vaccine, BCG (the 'gold-standard' against which new vaccines are tested), and a saline (negative) control. One candidate (Rv1806-1807) induced protection in the guinea pig aerosol infection model 30 days post-challenge on the basis of reducing the bacterial burden of M. tuberculosis in the lungs.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial/immunology
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Epitopes
- Escherichia coli/genetics
- Female
- Genes, Bacterial
- Genetic Vectors
- Guinea Pigs
- Immunity, Cellular
- Immunoglobulin G/blood
- Lung/microbiology
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Mycobacterium tuberculosis/isolation & purification
- Th1 Cells/immunology
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/immunology
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/prevention & control
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Julia Vipond
- Research Division, Health Protection Agency, Salisbury, UK.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Lowrie DB. DNA vaccines for therapy of tuberculosis: Where are we now? Vaccine 2006; 24:1983-9. [PMID: 16316711 DOI: 10.1016/j.vaccine.2005.11.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Revised: 10/24/2005] [Accepted: 11/04/2005] [Indexed: 10/25/2022]
Abstract
DNA vaccines that were being investigated in mice for prophylactic use against tuberculosis were soon found also to be surprisingly effective as treatment against established infection. The immune system was stimulated to kill the bacteria, even including the persistent latent bacteria that are otherwise refractory to the immune system and antibacterial chemotherapeutic drugs alike. Subsequent results from a range of laboratories using diverse DNA vaccines in diverse murine models of infection have been very varied, ranging from enhanced pathology, through negligible effects, to major additive benefit from combined vaccine and chemotherapy. This review summarises the data and assesses future prospects.
Collapse
Affiliation(s)
- Douglas B Lowrie
- National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK.
| |
Collapse
|
49
|
Abstract
There are more cases of tuberculosis in the world today than at any other time in history. The global epidemic has generated intense interest into the immunological mechanisms that control infection. Although CD4+ T cells play a critical role in host immunity to Mycobacterium tuberculosis, there is considerable interest in understanding the role of other T cell subsets in preventing disease development following infection. CD8+ T cells are required for optimum host defense following M. tuberculosis infection, which has led to investigation into how this protective effect is mediated. A critical review of recent literature regarding the role of CD8+ T cells in protective immunity to M. tuberculosis infection is now required to address the strengths and weaknesses of these studies. In this article, we evaluate the evidence that CD8+ T cells are critical in immunity to M. tuberculosis infection. We discuss the specific mycobacterial proteins that are recognized by CD8+ T cells elicited during infection. Finally, we examine the effector mechanisms of CD8+ T cells generated during infection and synthesize recent studies to consider the protective roles that these T cells serve in vivo.
Collapse
Affiliation(s)
- Joshua S M Woodworth
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
50
|
Orme IM. Preclinical testing of new vaccines for tuberculosis: A comprehensive review. Vaccine 2006; 24:2-19. [PMID: 16139397 DOI: 10.1016/j.vaccine.2005.07.078] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Accepted: 07/22/2005] [Indexed: 10/25/2022]
Abstract
The past decade has seen an explosive increase in the development of potential new tuberculosis vaccine candidates, as well as the establishment of at least two testing centers. Various animal models, but particularly the mouse and guinea pig models, have provided a lot information about how new vaccines can reduce disease progression and how this influences the pathology of the disease, but there is still much to learn at the immunological level, particularly in terms of the nature of the T cell response that is needed to confer long lived resistance. Several categories of vaccine candidates have been tried to date, and there are at least five individual vaccines moving towards clinical evaluation. There are still areas of the field that are poorly developed however. These include the fact that we have no models of post- exposure vaccination, or any models of latent disease. In addition, no standardized models of safety/toxicology exist as yet, which will be needed before extensive clinical development of the new vaccines.
Collapse
Affiliation(s)
- Ian M Orme
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Lake Street, Fort Collins, CO 80523, USA.
| |
Collapse
|