1
|
Kim H, Shin SJ. Immunogenicity and vaccine potential of clinical isolate Mycobacterium kansasii strain against Mycobacterium tuberculosis infection. Microbiol Spectr 2024; 12:e0081924. [PMID: 38980025 PMCID: PMC11302008 DOI: 10.1128/spectrum.00819-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
Mycobacterium kansasii is a bacterium included in non-tuberculous mycobacteria (NTM) that can cause lung disease. It shares a significant number of antigens with Mycobacterium tuberculosis (Mtb), suggesting that it has the potential to be used as a tuberculosis (TB) vaccine. Therefore, we subcutaneously vaccinated mice with reference strain, M. kansasii-ATCC12478 [M. kansasii-American Type Culture Collection (ATCC)], and clinically isolated strain, M. kansasii-SM-1 to evaluate potential as a TB vaccine by comparing with bacille Calmette-Guerin (BCG) vaccine. Ten weeks after vaccination, we evaluated immunogenicity of M. kansasii-ATCC and M. kansasii-SM-1, and M. kansasii-SM-1 immunization induces potent Mtb antigen-specific IFN-γ-producing CD4+ T cells than M. kansasii-ATCC. Upon Mtb infection, M. kansasii-SM-1 provided better protection than M. kansasii-ATCC, which was comparable to the efficacy of BCG. These results showed that the clinical strain M. kansasii-SM-1, which exhibits an enhanced Mtb antigen-specific Th1 response, shows greater vaccine efficacy compared to M. kansasii-ATCC. In this study, we demonstrated that vaccine efficacy can vary depending on the strain of M. kansasii and that its efficacy can be comparable to BCG. This suggests that M. kansasii has the potential to be a live TB vaccine candidate.IMPORTANCEMycobacterium kansasii, a non-tuberculous mycobacteria (NTM) species causing lung disease, shares key antigens with Mycobacterium tuberculosis (Mtb), indicating its potential for TB vaccine development. Subcutaneous vaccination of mice with M. kansasii strains reference strain M. kansasii-ATCC12478 [(M. kansasii-American Type Culture Collection (ATCC)] and clinically isolated strain M. kansasii-SM-1 revealed differences in immunogenicity. M. kansasii-SM-1 induced a robust Mtb antigen-specific IFN-γ-producing CD4+ T cell response compared to M. kansasii-ATCC. Additionally, M. kansasii-SM-1 conferred better protection against Mtb infection than M. kansasii-ATCC, which is comparable to bacille Calmette-Guerin (BCG). These findings underscore the variable vaccine efficacy among M. kansasii strains, with M. kansasii-SM-1 exhibiting promising potential as a live TB vaccine candidate, suggesting its comparative effectiveness to BCG.
Collapse
Affiliation(s)
- Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
Chen Z, Zhang Y, Wu J, Xu J, Hu Z, Fan XY. A multistage protein subunit vaccine as BCG-booster confers protection against Mycobacterium tuberculosis infection in murine models. Int Immunopharmacol 2024; 139:112811. [PMID: 39068754 DOI: 10.1016/j.intimp.2024.112811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
The eradication of tuberculosis remains a global challenge. Despite being the only licensed vaccine, Bacillus Calmette-Guérin (BCG) confers limited protective efficacy in adults and individuals with latent tuberculosis infections (LTBI). There is an urgent need to develop novel vaccines that can enhance the protective effect of BCG. Protein subunit vaccines have garnered significant research interest due to their safety and plasticity. Based on previous studies, we selected three antigens associated with LTBI (Rv2028c, Rv2029c, Rv3126c) and fused them with an immunodominant antigen Ag85A, resulting in the construction of a multistage protein subunit vaccine named A986. We evaluated the protective effect of recombinant protein A986 adjuvanted with MPL/QS21 as a booster vaccine for BCG against Mycobacterium tuberculosis (Mtb) infection in mice. The A986 + MPL/QS21 induced the secretion of antigen-specific Th1 (IL-2+, IFN-γ+ and TNF-α+) and Th17 (IL-17A+) cytokines in CD4+ and CD8+ T cells within the lung and spleen of mice, while also increased the frequency of central memory and effector memory T cells. Additionally, it also induced the enhanced production of IgG antibodies. Compared to BCG alone, A986 + MPL/QS21 boosting significantly augmented the proliferation of antigen-specific multifunctional T cells and effectively reduced bacterial load in infected mice. Taken together, A986 + MPL/QS21 formulation induced robust antigen-specific immune responses and provided enhanced protection against Mtb infection as a booster of BCG vaccine.
Collapse
Affiliation(s)
- Zhenyan Chen
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Ying Zhang
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Juan Wu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Jinchuan Xu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China.
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China.
| |
Collapse
|
3
|
Cohen SB, Plumlee CR, Engels L, Mai D, Murray TA, Jahn AN, Alexander B, Delahaye JL, Cross LM, Maciag K, Schrader S, Durga K, Gold ES, Aderem A, Gerner MY, Gern BH, Diercks AH, Urdahl KB. Host and pathogen genetic diversity shape vaccine-mediated protection to Mycobacterium tuberculosis. Front Immunol 2024; 15:1427846. [PMID: 39007152 PMCID: PMC11239334 DOI: 10.3389/fimmu.2024.1427846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
To investigate how host and pathogen diversity govern immunity against Mycobacterium tuberculosis (Mtb), we performed a large-scale screen of vaccine-mediated protection against aerosol Mtb infection using three inbred mouse strains [C57BL/6 (B6), C3HeB/FeJ (C3H), Balb/c x 129/SvJ (C129F1)] and three Mtb strains (H37Rv, CDC1551, SA161) representing two lineages and distinct virulence properties. We compared three protective modalities, all of which involve inoculation with live mycobacteria: Bacillus Calmette-Guérin (BCG), the only approved TB vaccine, delivered either subcutaneously or intravenously, and concomitant Mtb infection (CoMtb), a model of pre-existing immunity in which a low-level Mtb infection is established in the cervical lymph node following intradermal inoculation. We examined lung bacterial burdens at early (Day 28) and late (Day 98) time points after aerosol Mtb challenge and histopathology at Day 98. We observed substantial heterogeneity in the reduction of bacterial load afforded by these modalities at Day 28 across the combinations and noted a strong positive correlation between bacterial burden in unvaccinated mice and the degree of protection afforded by vaccination. Although we observed variation in the degree of reduction in bacterial burdens across the nine mouse/bacterium strain combinations, virtually all protective modalities performed similarly for a given strain-strain combination. We also noted dramatic variation in histopathology changes driven by both host and bacterial genetic backgrounds. Vaccination improved pathology scores for all infections except CDC1551. However, the most dramatic impact of vaccination on lesion development occurred for the C3H-SA161 combination, where vaccination entirely abrogated the development of the large necrotic lesions that arise in unvaccinated mice. In conclusion, we find that substantial TB heterogeneity can be recapitulated by introducing variability in both host and bacterial genetics, resulting in changes in vaccine-mediated protection as measured both by bacterial burden as well as histopathology. These differences can be harnessed in future studies to identify immune correlates of vaccine efficacy.
Collapse
Affiliation(s)
- Sara B. Cohen
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Courtney R. Plumlee
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Lindsay Engels
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Dat Mai
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Tara A. Murray
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Ana N. Jahn
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Bridget Alexander
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Jared L. Delahaye
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Lauren M. Cross
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Karolina Maciag
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Medicine, Division of Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Sam Schrader
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Kaitlin Durga
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Elizabeth S. Gold
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Alan Aderem
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Michael Y. Gerner
- Department of Immunology, University of Washington, Seattle, WA, United States
| | - Benjamin H. Gern
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Alan H. Diercks
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Kevin B. Urdahl
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
4
|
Fan X, Zhao X, Wang R, Li M, Luan X, Wang R, Wan K, Liu H. A novel multistage antigens ERA005f confer protection against Mycobacterium tuberculosis by driving Th-1 and Th-17 type T cell immune responses. Front Immunol 2023; 14:1276887. [PMID: 38022539 PMCID: PMC10662081 DOI: 10.3389/fimmu.2023.1276887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Tuberculosis (TB) is a major threat to human health. In 2021, TB was the second leading cause of death after COVID-19 among infectious diseases. The Bacillus Calmette-Guérin vaccine (BCG), the only licensed TB vaccine, is ineffective against adult TB. Therefore, there is an urgent need to develop new effective vaccines. Methods In this study, we developed a novel multistage subunit vaccine (ERA005f) comprising various proteins expressed in metabolic states, based on three immunodominant antigens (ESAT-6, Rv2628, and Ag85B). We utilized the E. coli prokaryotic expression system to express ERA005f and subsequently purified the protein using nickel affinity chromatography and anion exchange. Immunogenicity and protective efficacy of ERA005f and ERA005m were evaluated in BALB/c mice. Results ERA005f was consistently expressed as an inclusion body in a prokaryotic expression system, and a highly pure form of the protein was successfully obtained. Both ERA005f and ERA005m significantly improved IgG titers in the serum. In addition, mice immunized with ERA005f and ERA005m generated higher titers of antigen-specific IgG2a than the other groups. Elispot results showed that, compared with other groups, ERA005f increased the numbers of IFN-γ-secreting and IL-4-secreting T cells, especially the number of IFN-γ-secreting T cells. Meanwhile, ERA005f induced a higher number of IFN-γ+ T lymphocytes than ERA005m did. In addition, ERA005f improved the expression of cytokines, including IFN-γ, IL-12p70, TNF-α, IL-17, and GM-CSF and so on. Importantly, both ERA005f and ERA005m significantly inhibited the growth of Mtb. Conclusion The novel multistage antigen ERA005f elicited a strong antigen-specific humoral response and Th-1 and Th-17 cell-mediated immunity in mice. Meanwhile, it can effectively inhibit H37Rv growth in vitro, and represents a correlate of protection in vivo, indicating that ERA005f may exhibit excellent protective efficacy against Mycobacterium tuberculosis H37Rv infection. Our study suggests that ERA005f has the potential to be a promising multistage tuberculosis vaccine candidate.
Collapse
Affiliation(s)
- Xueting Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuqin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruibai Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Machao Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuli Luan
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Ruihuan Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kanglin Wan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haican Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
5
|
Plumlee CR, Barrett HW, Shao DE, Lien KA, Cross LM, Cohen SB, Edlefsen PT, Urdahl KB. Assessing vaccine-mediated protection in an ultra-low dose Mycobacterium tuberculosis murine model. PLoS Pathog 2023; 19:e1011825. [PMID: 38011264 PMCID: PMC10703413 DOI: 10.1371/journal.ppat.1011825] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/07/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Despite widespread immunization with Bacille-Calmette-Guérin (BCG), the only currently licensed tuberculosis (TB) vaccine, TB remains a leading cause of mortality globally. There are many TB vaccine candidates in the developmental pipeline, but the lack of a robust animal model to assess vaccine efficacy has hindered our ability to prioritize candidates for human clinical trials. Here we use a murine ultra-low dose (ULD) Mycobacterium tuberculosis (Mtb) challenge model to assess protection conferred by BCG vaccination. We show that BCG confers a reduction in lung bacterial burdens that is more durable than that observed after conventional dose challenge, curbs Mtb dissemination to the contralateral lung, and, in a small percentage of mice, prevents detectable infection. These findings are consistent with the ability of human BCG vaccination to mediate protection, particularly against disseminated disease, in specific human populations and clinical settings. Overall, our findings demonstrate that the ultra-low dose Mtb infection model can measure distinct parameters of immune protection that cannot be assessed in conventional dose murine infection models and could provide an improved platform for TB vaccine testing.
Collapse
Affiliation(s)
- Courtney R. Plumlee
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Holly W. Barrett
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Global Health, Seattle, Washington, United States of America
| | - Danica E. Shao
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Katie A. Lien
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Lauren M. Cross
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Sara B. Cohen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Paul T. Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Kevin B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
- University of Washington, Dept. of Pediatrics, Seattle, Washington, United States of America
| |
Collapse
|
6
|
Jalbert E, Liu C, Mave V, Lang N, Kagal A, Valvi C, Paradkar M, Gupte N, Lokhande R, Bharadwaj R, Kulkarni V, Gupta A, Weinberg A. Comparative immune responses to Mycobacterium tuberculosis in people with latent infection or sterilizing protection. iScience 2023; 26:107425. [PMID: 37564701 PMCID: PMC10410524 DOI: 10.1016/j.isci.2023.107425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 03/22/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
There is great need for vaccines against tuberculosis (TB) more efficacious than the licensed BCG. Our goal was to identify new vaccine benchmarks by identifying immune responses that distinguish individuals able to eradicate the infection (TB-resisters) from individuals with latent infection (LTBI-participants). TB-resisters had higher frequencies of circulating CD8+ glucose monomycolate (GMM)+ Granzyme-B+ T cells than LTBI-participants and higher proportions of polyfunctional conventional and nonconventional T cells expressing Granzyme-B and/or PD-1 after ex vivo M. tuberculosis stimulation of blood mononuclear cells. LTBI-participants had higher expression of activation markers and cytokines, including IL10, and IFNγ. An exploratory analysis of BCG-recipients with minimal exposure to TB showed absence of CD8+GMM+Granzyme-B+ T cells, lower or equal proportions of Granzyme-B+PD-1+ polyfunctional T cells than TB-resisters and higher or equal than LTBI-participants. In conclusion, high Granzyme-B+PD-1+ T cell responses to M. tuberculosis and, possibly, of CD8+GMM+Granzyme-B+ T cells may be desirable for new TB vaccines.
Collapse
Affiliation(s)
- Emilie Jalbert
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Cuining Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Vidya Mave
- Byramjee Jeejeebhoy Government Medical College- Johns Hopkins University Clinical Research Site (BJGMC-JHU CRS), Pune, Maharashtra, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, Maharashtra, India
- School of Medicine, Center for Clinical Global Health Education (CCGHE), Johns Hopkins University, Baltimore, MD, USA
| | - Nancy Lang
- Department of Pediatrics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Anju Kagal
- Department of Microbiology, Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospital, Pune, Maharashtra, India
| | - Chhaya Valvi
- Department of Pediatrics, Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospital, Pune, Maharashtra, India
| | - Mandar Paradkar
- Byramjee Jeejeebhoy Government Medical College- Johns Hopkins University Clinical Research Site (BJGMC-JHU CRS), Pune, Maharashtra, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, Maharashtra, India
- School of Medicine, Center for Clinical Global Health Education (CCGHE), Johns Hopkins University, Baltimore, MD, USA
| | - Nikhil Gupte
- Byramjee Jeejeebhoy Government Medical College- Johns Hopkins University Clinical Research Site (BJGMC-JHU CRS), Pune, Maharashtra, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, Maharashtra, India
- School of Medicine, Center for Clinical Global Health Education (CCGHE), Johns Hopkins University, Baltimore, MD, USA
| | - Rahul Lokhande
- Department of Pulmonary Medicine, Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospital, Pune, Maharashtra, India
| | - Renu Bharadwaj
- Department of Microbiology, Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospital, Pune, Maharashtra, India
| | - Vandana Kulkarni
- Byramjee Jeejeebhoy Government Medical College- Johns Hopkins University Clinical Research Site (BJGMC-JHU CRS), Pune, Maharashtra, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, Maharashtra, India
- School of Medicine, Center for Clinical Global Health Education (CCGHE), Johns Hopkins University, Baltimore, MD, USA
| | - Amita Gupta
- Johns Hopkins Center for Infectious Diseases in India, Pune, Maharashtra, India
- School of Medicine, Center for Clinical Global Health Education (CCGHE), Johns Hopkins University, Baltimore, MD, USA
| | - Adriana Weinberg
- Departments of Pediatrics, Medicine and Pathology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
7
|
Plumlee C, Barrett H, Shao D, Lien K, Cross L, Cohen S, Edlefsen P, Urdahl K. Assessing vaccine-mediated protection in an ultra-low dose Mycobacterium tuberculosis murine model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533820. [PMID: 36993415 PMCID: PMC10055404 DOI: 10.1101/2023.03.22.533820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Despite widespread immunization with Bacille-Calmette-Guerin (BCG), the only currently licensed tuberculosis (TB) vaccine, TB remains a leading cause of mortality globally. There are many TB vaccine candidates in the developmental pipeline, but the lack of a robust animal model to assess vaccine efficacy has hindered our ability to prioritize candidates for human clinical trials. Here we use a murine ultra-low dose (ULD) Mycobacterium tuberculosis (Mtb) challenge model to assess protection conferred by BCG vaccination. We show that BCGconfers a reduction in lung bacterial burdens that is more durable than that observed afterconventional dose challenge, curbs Mtb dissemination to the contralateral lung, and, in a smallpercentage of mice, prevents detectable infection. These findings are consistent with the ability of human BCG vaccination to mediate protection, particularly against disseminated disease, in specific human populations and clinical settings. Overall, our findings demonstrate that the ultra-low dose Mtb infection model can measure distinct parameters of immune protection that cannot be assessed in conventional dose murine infection models and could provide an improved platform for TB vaccine testing.
Collapse
Affiliation(s)
- C.R. Plumlee
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - H.W. Barrett
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
- University of Washington, Dept. of Global Health, Seattle, WA, 98109, USA
| | - D.E. Shao
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, 98109, USA
| | - K.A. Lien
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - L.M. Cross
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - S.B. Cohen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - P.T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, 98109, USA
| | - K.B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
- University of Washington, Dept. of Immunology, Seattle, WA, 98109, USA
- University of Washington, Dept. of Pediatrics, Seattle, WA, 98109, USA
- Lead Contact
| |
Collapse
|
8
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
9
|
Elkington P, Polak ME, Reichmann MT, Leslie A. Understanding the tuberculosis granuloma: the matrix revolutions. Trends Mol Med 2022; 28:143-154. [PMID: 34922835 PMCID: PMC8673590 DOI: 10.1016/j.molmed.2021.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023]
Abstract
Mycobacterium tuberculosis (Mtb) causes the human disease tuberculosis (TB) and remains the top global infectious pandemic after coronavirus disease 2019 (COVID-19). Furthermore, TB has killed many more humans than any other pathogen, after prolonged coevolution to optimise its pathogenic strategies. Full understanding of fundamental disease processes in humans is necessary to successfully combat this highly successful pathogen. While the importance of immunodeficiency has been long recognised, biologic therapies and unbiased approaches are providing unprecedented insights into the intricacy of the host-pathogen interaction. The nature of a protective response is more complex than previously hypothesised. Here, we integrate recent evidence from human studies and unbiased approaches to consider how Mtb causes human TB and highlight the recurring theme of extracellular matrix (ECM) turnover.
Collapse
Affiliation(s)
- Paul Elkington
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| | - Marta E Polak
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Michaela T Reichmann
- NIHR Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Alasdair Leslie
- Department of Infection and Immunity, University College London, London, UK; Africa Health Research Institute, KwaZulu-Natal, South Africa
| |
Collapse
|
10
|
Plumlee CR, Duffy FJ, Gern BH, Delahaye JL, Cohen SB, Stoltzfus CR, Rustad TR, Hansen SG, Axthelm MK, Picker LJ, Aitchison JD, Sherman DR, Ganusov VV, Gerner MY, Zak DE, Urdahl KB. Ultra-low Dose Aerosol Infection of Mice with Mycobacterium tuberculosis More Closely Models Human Tuberculosis. Cell Host Microbe 2021; 29:68-82.e5. [PMID: 33142108 PMCID: PMC7854984 DOI: 10.1016/j.chom.2020.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/21/2020] [Accepted: 09/25/2020] [Indexed: 02/02/2023]
Abstract
Tuberculosis (TB) is a heterogeneous disease manifesting in a subset of individuals infected with aerosolized Mycobacterium tuberculosis (Mtb). Unlike human TB, murine infection results in uniformly high lung bacterial burdens and poorly organized granulomas. To develop a TB model that more closely resembles human disease, we infected mice with an ultra-low dose (ULD) of between 1-3 founding bacteria, reflecting a physiologic inoculum. ULD-infected mice exhibited highly heterogeneous bacterial burdens, well-circumscribed granulomas that shared features with human granulomas, and prolonged Mtb containment with unilateral pulmonary infection in some mice. We identified blood RNA signatures in mice infected with an ULD or a conventional Mtb dose (50-100 CFU) that correlated with lung bacterial burdens and predicted Mtb infection outcomes across species, including risk of progression to active TB in humans. Overall, these findings highlight the potential of the murine TB model and show that ULD infection recapitulates key features of human TB.
Collapse
Affiliation(s)
- Courtney R Plumlee
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Fergal J Duffy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Benjamin H Gern
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Jared L Delahaye
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Sara B Cohen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Caleb R Stoltzfus
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Tige R Rustad
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - David R Sherman
- Department of Microbiology, University of Washington, Seattle, WA 98109, USA
| | - Vitaly V Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Michael Y Gerner
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Daniel E Zak
- Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Kevin B Urdahl
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98109, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
11
|
Tezera LB, Mansour S, Elkington P. Reconsidering the Optimal Immune Response to Mycobacterium tuberculosis. Am J Respir Crit Care Med 2020; 201:407-413. [PMID: 31657633 DOI: 10.1164/rccm.201908-1506pp] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Liku B Tezera
- National Institute for Health Research Biomedical Research Centre, School of Clinical and Experimental Sciences and
| | - Salah Mansour
- National Institute for Health Research Biomedical Research Centre, School of Clinical and Experimental Sciences and.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul Elkington
- National Institute for Health Research Biomedical Research Centre, School of Clinical and Experimental Sciences and.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
12
|
Kim H, Kwon KW, Park J, Kang H, Lee Y, Sohn EJ, Hwang I, Eum SY, Shin SJ. Plant-Produced N-glycosylated Ag85A Exhibits Enhanced Vaccine Efficacy Against Mycobacterium tuberculosis HN878 Through Balanced Multifunctional Th1 T Cell Immunity. Vaccines (Basel) 2020; 8:vaccines8020189. [PMID: 32325740 PMCID: PMC7349862 DOI: 10.3390/vaccines8020189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 11/16/2022] Open
Abstract
Tuberculosis (TB) is one of the deadliest infectious diseases worldwide and is caused by Mycobacterium tuberculosis (Mtb). An effective vaccine to prevent TB is considered the most cost-effective measure for controlling this disease. Many different vaccine antigen (Ag) candidates, including well-known and newly identified Ags, have been evaluated in clinical and preclinical studies. In this study, we took advantage of a plant system of protein expression using Nicotiana benthamiana to produce N-glycosylated antigen 85A (G-Ag85A), which is one of the most well-characterized vaccine Ag candidates in the field of TB vaccines, and compared its immunogenicity and vaccine efficacy with those of nonglycosylated Ag85A (NG-Ag85A) produced with an Escherichia coli system. Notably, G-Ag85A induced a more robust IFN-γ response than NG-Ag85A, which indicated that G-Ag85A is well recognized by the host immune system during Mtb infection. We subsequently compared the vaccine potential of G-Ag85A and NG-Ag85A by evaluating their immunological features and substantial protection efficacies. Interestingly, G-Ag85A yielded moderately enhanced long-term protective efficacy, as measured in terms of bacterial burden and lung inflammation. Strikingly, G-Ag85A-immunized mice showed a more balanced proportion of multifunctional Th1-biased immune responses with sustained IFN-γ response than did NG-Ag85A-immunized mice. Collectively, plant-derived G-Ag85A could induce protective and balanced Th1 responses and confer long-term protection against a hypervirulent Mtb Beijing strain infection, which indicated that plant-produced G-Ag85A might provide an excellent example for the production of an Mtb subunit vaccine Ag and could be an effective platform for the development of anti-TB vaccines.
Collapse
Affiliation(s)
- Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
| | - Jaehun Park
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
| | - Hyangju Kang
- BioApplications Inc., Pohang 37668, Korea; (H.K.); (Y.L.); (E.-J.S.)
| | - Yongjik Lee
- BioApplications Inc., Pohang 37668, Korea; (H.K.); (Y.L.); (E.-J.S.)
| | - Eun-Ju Sohn
- BioApplications Inc., Pohang 37668, Korea; (H.K.); (Y.L.); (E.-J.S.)
- School of Interdisciplinary Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Inhwan Hwang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea;
| | - Seok-Yong Eum
- Division of Immunopathology and Cellular Immunology, International Tuberculosis Research Center, Changwon 51755, Korea;
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea; (H.K.); (K.W.K.); (J.P.)
- Correspondence:
| |
Collapse
|
13
|
Vasina DV, Kleymenov DA, Manuylov VA, Mazunina EP, Koptev EY, Tukhovskaya EA, Murashev AN, Gintsburg AL, Gushchin VA, Tkachuk AP. First-In-Human Trials of GamTBvac, a Recombinant Subunit Tuberculosis Vaccine Candidate: Safety and Immunogenicity Assessment. Vaccines (Basel) 2019; 7:vaccines7040166. [PMID: 31683812 PMCID: PMC6963980 DOI: 10.3390/vaccines7040166] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis is known to be the biggest global health problem, causing the most deaths by a single infectious agent. Vaccine-development efforts are extremely important. This paper represents the results of the first-in-human trial of recombinant subunit tuberculosis vaccine GamTBvac in a Phase I study. GamTBvac is a new BCG booster candidate vaccine containing dextran-binding domain modified Ag85a and ESAT6-CFP10 MTB antigens and CpG ODN adjuvant, formulated with dextrans. Safety and immunogenicity of GamTBvac were estimated in an open-label clinical trial on 60 Mycobacterium tuberculosis uninfected (MTB-uninfected) volunteers previously-vaccinated with Bacillus Calmette—Guérin vaccine (BCG). The candidate vaccine had an acceptable safety profile and was well-tolerated. Three different vaccine doses with a double-immunization scheme were assessed for immunogenicity and induced a significant increase in IFN-γ in-house IGRA response and IgG ELISA analysis. Among them, the half dose vaccine group (containing DBD-ESAT6-CFP10, 12.5 μg; DBD-Ag85a, 12.5 μg; CpG (ODN 2216), 75 μg; DEAE-Dextran 500 kDa, 250 μg; and Dextran 500 kDa, 5 mg) provided high, early and stable in time immune response specific to both protein antigen fusions and is proposed for the further studies.
Collapse
Affiliation(s)
- Daria V Vasina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Denis A Kleymenov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Victor A Manuylov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena P Mazunina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Egor Yu Koptev
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena A Tukhovskaya
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Arkady N Murashev
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Alexander L Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Infectology Department, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Vladimir A Gushchin
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Artem P Tkachuk
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| |
Collapse
|
14
|
Abstract
Are antibodies important for protection against tuberculosis? The jury has been out for more than 100 years. B cell depletion in experimental Mycobacterium tuberculosis infection failed to identify a major role for these cells in immunity to tuberculosis. However, recent identification of naturally occurring antibodies in humans that are protective during M. tuberculosis infection has reignited the debate. Here, we discuss the evidence for a protective role for antibodies in tuberculosis and consider the feasibility of designing novel tuberculosis vaccines targeting humoral immunity.
Collapse
Affiliation(s)
- Hao Li
- Centre for Global Health and Infectious Diseases, Collaborative Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, Tsinghua University School of Medicine, Beijing, China
| | - Babak Javid
- Centre for Global Health and Infectious Diseases, Collaborative Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, Tsinghua University School of Medicine, Beijing, China.
| |
Collapse
|
15
|
Counoupas C, Triccas JA, Britton WJ. Deciphering protective immunity against tuberculosis: implications for vaccine development. Expert Rev Vaccines 2019; 18:353-364. [PMID: 30793629 DOI: 10.1080/14760584.2019.1585246] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The development of more effective tuberculosis (TB) vaccines is essential for the global control of TB. Recently, there have been major advances in the field, but an important hindrance remains the lack of correlates of protection against TB. This requires each vaccine candidate to undergo clinical efficacy trials based on data from animal protection studies, but the results from animal models do not necessarily predict efficacy in humans. AREAS COVERED In this review we summarize our current knowledge of immune mechanisms that may contribute to protective immunity against TB following vaccination and relate these to protective efficacy in animal models and recent clinical trials. Although some initial trials did not reproduce protection against TB in humans, recent trials have demonstrated promising efficacy for three vaccine approaches. EXPERT OPINION Although CD4+ T lymphocytes are essential for protection against TB, there is no clear correlation between conventional CD4+ or CD8+ T cell responses and protective efficacy of TB vaccines. Recent attention has focused on other immune responses, including donor unrestricted T cells, B lymphocytes, and antibodies. Prospective studies on samples from vaccinated individuals protected in recent trials will allow evaluation of these alternative immune mechanisms as potential correlates of protection.
Collapse
Affiliation(s)
- Claudio Counoupas
- a Tuberculosis Research Program Centenary Institute , The University of Sydney , Camperdown , NSW , Australia.,b The University of Sydney , Central Clinical School Faculty of Medicine and Health , Sydney , NSW , Australia
| | - James A Triccas
- a Tuberculosis Research Program Centenary Institute , The University of Sydney , Camperdown , NSW , Australia
| | - Warwick J Britton
- a Tuberculosis Research Program Centenary Institute , The University of Sydney , Camperdown , NSW , Australia.,b The University of Sydney , Central Clinical School Faculty of Medicine and Health , Sydney , NSW , Australia
| |
Collapse
|
16
|
Basu Roy R, Whittaker E, Seddon JA, Kampmann B. Tuberculosis susceptibility and protection in children. THE LANCET. INFECTIOUS DISEASES 2019; 19:e96-e108. [PMID: 30322790 PMCID: PMC6464092 DOI: 10.1016/s1473-3099(18)30157-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 01/27/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022]
Abstract
Children represent both a clinically important population susceptible to tuberculosis and a key group in whom to study intrinsic and vaccine-induced mechanisms of protection. After exposure to Mycobacterium tuberculosis, children aged under 5 years are at high risk of progressing first to tuberculosis infection, then to tuberculosis disease and possibly disseminated forms of tuberculosis, with accompanying high risks of morbidity and mortality. Children aged 5-10 years are somewhat protected, until risk increases again in adolescence. Furthermore, neonatal BCG programmes show the clearest proven benefit of vaccination against tuberculosis. Case-control comparisons from key cohorts, which recruited more than 15 000 children and adolescents in total, have identified that the ratio of monocytes to lymphocytes, activated CD4 T cell count, and a blood RNA signature could be correlates of risk for developing tuberculosis. Further studies of protected and susceptible populations are necessary to guide development of novel tuberculosis vaccines that could facilitate the achievement of WHO's goal to eliminate deaths from tuberculosis in childhood.
Collapse
Affiliation(s)
- Robindra Basu Roy
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK; Vaccines and Immunity Theme MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Elizabeth Whittaker
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK
| | - James A Seddon
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK
| | - Beate Kampmann
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, UK; Vaccines and Immunity Theme MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia.
| |
Collapse
|
17
|
Kim WS, Kim H, Kwon KW, Cho SN, Shin SJ. Immunogenicity and Vaccine Potential of InsB, an ESAT-6-Like Antigen Identified in the Highly Virulent Mycobacterium tuberculosis Beijing K Strain. Front Microbiol 2019; 10:220. [PMID: 30809214 PMCID: PMC6379281 DOI: 10.3389/fmicb.2019.00220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/28/2019] [Indexed: 12/13/2022] Open
Abstract
Our group recently identified InsB, an ESAT-6-like antigen belonging to the Mtb9.9 subfamily within the Esx family, in the Mycobacterium tuberculosis Korean Beijing strain (Mtb K) via a comparative genomic analysis with that of the reference Mtb H37Rv and characterized its immunogenicity and its induced immune response in patients with tuberculosis (TB). However, the vaccine potential of InsB has not been fully elucidated. In the present study, InsB was evaluated as a subunit vaccine in comparison with the most well-known ESAT-6 against the hypervirulent Mtb K. Mice immunized with InsB/MPL-DDA exhibited an antigen-specific IFN-γ response along with antigen-specific effector/memory T cell expansion in the lungs and spleen upon antigen restimulation. In addition, InsB immunization markedly induced multifunctional Th1-type CD4+ T cells coexpressing TNF-α, IL-2, and IFN-γ in the lungs following Mtb K challenge. Finally, we found that InsB immunization conferred long-term protection against Mtb K comparable to that conferred by ESAT-6 immunization, as evidenced by a similar level of CFU reduction in the lung and spleen and reduced lung inflammation. These results suggest that InsB may be an excellent vaccine antigen component for developing a multiantigenic Mtb subunit vaccine by generating Th1-biased memory T cells with a multifunctional capacity and may confer durable protection against the highly virulent Mtb K.
Collapse
Affiliation(s)
- Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, South Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang-Nae Cho
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
18
|
Basu Roy R, Sambou B, Uhía I, Roetynck S, Robertson BD, Kampmann B. An Auto-luminescent Fluorescent BCG Whole Blood Assay to Enable Evaluation of Paediatric Mycobacterial Responses Using Minimal Blood Volumes. Front Pediatr 2019; 7:151. [PMID: 31114771 PMCID: PMC6503113 DOI: 10.3389/fped.2019.00151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 04/01/2019] [Indexed: 12/15/2022] Open
Abstract
Introduction: Understanding protective human immunity against mycobacteria is critical to developing and evaluating new vaccines against tuberculosis. Children are the most susceptible population to infection, disease, and death from tuberculosis, but also have the strongest evidence of BCG-inducible protection. Limited amounts of blood can be obtained for research purposes in paediatrics and therefore there is a need for high-yield, low-volume, human immunology assays. Methods: We transformed BCG Danish with plasmids encoding luciferase full operon derived from Photorhabdus luminescens together with Green Fluorescent Protein and antibiotic selection markers. We characterised the luminescent and fluorescent properties of this recombinant BCG strain (BCG-GFP-LuxFO) using a luminometer and flow cytometry and developed a paediatric whole blood in vitro infection model. Results: Luminescence of BCG-GFP-LuxFO correlated with optical density (Spearman Rank Correlation coefficient r = 0.985, p < 0.0001) and colony forming units (CFUs) in liquid culture medium (r = 0.971, p < 0.0001). Fluorescence of BCG-GFP-LuxFO in paediatric whole blood was confirmed by flow cytometry in granulocytes and monocytes 1 h following infection. Luminescence of BCG-GFP-LuxFO in whole blood corresponded with CFUs (r = 0.7123, p < 0.0001). Conclusion: The BCG-GFP-LuxFO assay requires 225 μL whole blood per sample, from which serial luminescence measurements can be obtained, together with biochemical analysis of supernatants and cellular assay applications using its fluorescent properties. This offers the opportunity to study human-mycobacterial interactions using multiple experimental modalities with only minimal blood volumes. It is therefore a valuable method for investigating paediatric immunity to tuberculosis.
Collapse
Affiliation(s)
- Robindra Basu Roy
- Department of Paediatrics, Centre for International Child Health, Imperial College London, London, United Kingdom.,Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine (LSHTM), Banjul, The Gambia
| | - Basil Sambou
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine (LSHTM), Banjul, The Gambia
| | - Iria Uhía
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Sophie Roetynck
- Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine (LSHTM), Banjul, The Gambia
| | - Brian D Robertson
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Beate Kampmann
- Department of Paediatrics, Centre for International Child Health, Imperial College London, London, United Kingdom.,Vaccines & Immunity Theme, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine (LSHTM), Banjul, The Gambia.,Faculty of Infectious and Tropical Diseases, The Vaccine Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
19
|
Sousa J, Saraiva M. Paradigm changing evidence that alter tuberculosis perception and detection: Focus on latency. INFECTION GENETICS AND EVOLUTION 2018; 72:78-85. [PMID: 30576838 DOI: 10.1016/j.meegid.2018.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/12/2018] [Accepted: 12/15/2018] [Indexed: 12/23/2022]
Abstract
Tuberculosis remains a devastating disease to Mankind, ranking as the ninth cause of death worldwide. Eliminating tuberculosis as proven much more difficult than once anticipated. In addition to the delay in diagnosis and drug resistance problems that compromise the efficacy of treatment, the enormous reservoir of latently infected individuals continuously feeds the epidemics. However, targeting latency with prophylactic antibiotic administration is not possible at the populational level. Together, these issues call for a better understanding of latency, as well as for a more precise identification of individuals at high risk of reactivation. For this, recent paradigm changing evidence need to be taken into account, most notably, the existence of a tuberculosis spectrum; the genetic diversity of both humans and tuberculosis-causing bacteria; and the changes in the human population that interfere with tuberculosis. Here we discuss latency in the light of these variables and how that understanding can move forward tuberculosis research and elimination.
Collapse
Affiliation(s)
- Jeremy Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
20
|
Webster GR, van Dolleweerd C, Guerra T, Stelter S, Hofmann S, Kim M, Teh AY, Diogo GR, Copland A, Paul MJ, Hart P, Reljic R, Ma JK. A polymeric immunoglobulin-antigen fusion protein strategy for enhancing vaccine immunogenicity. PLANT BIOTECHNOLOGY JOURNAL 2018; 16:1983-1996. [PMID: 29682888 PMCID: PMC6230950 DOI: 10.1111/pbi.12932] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 05/06/2023]
Abstract
In this study, a strategy based on polymeric immunoglobulin G scaffolds (PIGS) was used to produce a vaccine candidate for Mycobacterium tuberculosis. A genetic fusion construct comprising genes encoding the mycobacterial Ag85B antigen, an immunoglobulin γ-chain fragment and the tailpiece from immunoglobulin μ chain was engineered. Expression was attempted in Chinese Hamster Ovary (CHO) cells and in Nicotiana benthamiana. The recombinant protein assembled into polymeric structures (TB-PIGS) in N. benthamiana, similar in size to polymeric IgM. These complexes were subsequently shown to bind to the complement protein C1q and FcγRs with increased affinity. Modification of the N-glycans linked to TB-PIGS by removal of xylose and fucose residues that are normally found in plant glycosylated proteins also resulted in increased affinity for low-affinity FcγRs. Immunization studies in mice indicated that TB-PIGS are highly immunogenic with and without adjuvant. However, they did not improve protective efficacy in mice against challenge with M. tuberculosis compared to conventional vaccination with BCG, suggesting that additional or alternative antigens may be needed to protect against this disease. Nevertheless, these results establish a novel platform for producing polymeric antigen-IgG γ-chain molecules with inherent functional characteristics that are desirable in vaccines.
Collapse
Affiliation(s)
- Gina R. Webster
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | | | - Thais Guerra
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Szymon Stelter
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Sven Hofmann
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Mi‐Young Kim
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Audrey Y‐H. Teh
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Gil Reynolds Diogo
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Alastair Copland
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Mathew J. Paul
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Peter Hart
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Rajko Reljic
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Julian K‐C. Ma
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| |
Collapse
|
21
|
Coler RN, Day TA, Ellis R, Piazza FM, Beckmann AM, Vergara J, Rolf T, Lu L, Alter G, Hokey D, Jayashankar L, Walker R, Snowden MA, Evans T, Ginsberg A, Reed SG. The TLR-4 agonist adjuvant, GLA-SE, improves magnitude and quality of immune responses elicited by the ID93 tuberculosis vaccine: first-in-human trial. NPJ Vaccines 2018; 3:34. [PMID: 30210819 PMCID: PMC6123489 DOI: 10.1038/s41541-018-0057-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/20/2018] [Accepted: 04/27/2018] [Indexed: 02/06/2023] Open
Abstract
Tuberculosis (TB) is the leading cause of infectious death worldwide. Development of improved TB vaccines that boost or replace BCG is a major global health goal. ID93 + GLA-SE is a fusion protein TB vaccine candidate combined with the Toll-like Receptor 4 agonist adjuvant, GLA-SE. We conducted a phase 1, randomized, double-blind, dose-escalation clinical trial to evaluate two dose levels of the ID93 antigen, administered intramuscularly alone or in combination with two dose levels of the GLA-SE adjuvant, in 60 BCG-naive, QuantiFERON-negative, healthy adults in the US (ClinicalTrials.gov identifier: NCT01599897). When administered as 3 injections, 28 days apart, all dose levels of ID93 alone and ID93 + GLA-SE demonstrated an acceptable safety profile. All regimens elicited vaccine-specific humoral and cellular responses. Compared with ID93 alone, vaccination with ID93 + GLA-SE elicited higher titers of ID93-specific antibodies, a preferential increase in IgG1 and IgG3 subclasses, and a multifaceted Fc-mediated effector function response. The addition of GLA-SE also enhanced the magnitude and polyfunctional cytokine profile of CD4+ T cells. The data demonstrate an acceptable safety profile and indicate that the GLA-SE adjuvant drives a functional humoral and T-helper 1 type cellular response. A tuberculosis vaccine containing an immunity-potentiating agent stimulated strong immune responses in a first-in-human trial. Tuberculosis (TB) is the world’s foremost cause of infectious disease deaths, yet lacks an effective vaccine for adult humans. Rhea Coler, of the Infectious Disease Research Institute, Seattle, and a team from the United States and South Africa, tested their prophylactic on 60 healthy US adults. The vaccine consisted of ID93, a fusion of TB therapeutic target proteins, and GLA-SE—a supplement to boost immune responses. The candidate proved safe in all participants, with mild-to-moderate adverse effects, and provoked promising immune responses. The formulation was significantly more effective with GLA-SE than without. Further studies will elucidate the therapeutic benefit of this formulation and its ability to combat the pathogenicity of TB.
Collapse
Affiliation(s)
- Rhea N Coler
- 1Infectious Disease Research Institute, Seattle, WA 98102 USA.,2Department of Global Health, University of Washington, Seattle, WA 98195 USA.,3PAI Life Sciences, Seattle, WA 98102 USA
| | - Tracey A Day
- 1Infectious Disease Research Institute, Seattle, WA 98102 USA
| | | | - Franco M Piazza
- 1Infectious Disease Research Institute, Seattle, WA 98102 USA
| | | | - Julie Vergara
- 1Infectious Disease Research Institute, Seattle, WA 98102 USA
| | - Tom Rolf
- 1Infectious Disease Research Institute, Seattle, WA 98102 USA
| | - Lenette Lu
- 5Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Boston, MA 02139 USA
| | - Galit Alter
- 5Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Boston, MA 02139 USA
| | | | | | | | | | | | | | - Steven G Reed
- 1Infectious Disease Research Institute, Seattle, WA 98102 USA
| | | |
Collapse
|
22
|
Mycobacterium tuberculosis Protein Rv3841 Activates Dendritic Cells and Contributes to a T Helper 1 Immune Response. J Immunol Res 2018; 2018:3525302. [PMID: 29736404 PMCID: PMC5875036 DOI: 10.1155/2018/3525302] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/02/2018] [Accepted: 01/10/2018] [Indexed: 12/15/2022] Open
Abstract
The attenuated vaccine Mycobacterium bovis BCG (Bacille Calmette Guerin) has limited protective efficacy against TB. The development of more effective TB vaccines has focused on the mycobacterial antigens that cause strong T helper 1 (Th1) responses. Mtb protein Rv3841 (bacterioferritin B; BfrB) is known to play a crucial role in the growth of Mtb. Nonetheless, it is unclear whether Rv3841 can induce protective immunity against Mtb. Here, we studied the action of Rv3841 in maturation of dendritic cells (DCs) and its engagement in the development of T-cell immunity. We found that Rv3841 functionally activated DCs by upregulating costimulatory molecules and increased secretion of proinflammatory cytokines. Activation of DCs by Rv3841 was mediated by Toll-like receptor 4 (TLR4), followed by triggering of mitogen-activated protein kinase and nuclear factor-κB signaling pathways. In addition, Rv3841-matured DCs effectively proliferated and polarized Th1 immune response of naïve CD4+ and CD8+ T-cells. Moreover, Rv3841 specifically caused the expansion of CD4+CD44highCD62Llow T-cells from Mtb-infected mice; besides, the T-cells activated by Rv3841-matured DCs inhibited intracellular mycobacterial growth. Our data suggest that Rv3841 induces DC maturation and protective immune responses, a finding that may provide candidate of effective TB vaccines.
Collapse
|
23
|
Whatney WE, Gandhi NR, Lindestam Arlehamn CS, Nizam A, Wu H, Quezada MJ, Campbell A, Allana S, Kabongo MM, Khayumbi J, Muchiri B, Ongalo J, Tonui J, Sasser LE, Fergus TJ, Ouma GS, Ouma SG, Beck AA, Mulligan MJ, Oladele A, Kaushal D, Cain KP, Waller L, Blumberg HM, Altman JD, Ernst JD, Rengarajan J, Day CL. A High Throughput Whole Blood Assay for Analysis of Multiple Antigen-Specific T Cell Responses in Human Mycobacterium tuberculosis Infection. THE JOURNAL OF IMMUNOLOGY 2018. [PMID: 29540577 DOI: 10.4049/jimmunol.1701737] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Antigen-specific CD4 and CD8 T cells are important components of the immune response to Mycobacterium tuberculosis, yet little information is currently known regarding how the breadth, specificity, phenotype, and function of M. tuberculosis-specific T cells correlate with M. tuberculosis infection outcome in humans. To facilitate evaluation of human M. tuberculosis-specific T cell responses targeting multiple different Ags, we sought to develop a high throughput and reproducible T cell response spectrum assay requiring low blood sample volumes. We describe here the optimization and standardization of a microtiter plate-based, diluted whole blood stimulation assay utilizing overlapping peptide pools corresponding to a functionally diverse panel of 60 M. tuberculosis Ags. Using IFN-γ production as a readout of Ag specificity, the assay can be conducted using 50 μl of blood per test condition and can be expanded to accommodate additional Ags. We evaluated the intra- and interassay variability, and implemented testing of the assay in diverse cohorts of M. tuberculosis-unexposed healthy adults, foreign-born adults with latent M. tuberculosis infection residing in the United States, and tuberculosis household contacts with latent M. tuberculosis infection in a tuberculosis-endemic setting in Kenya. The M. tuberculosis-specific T cell response spectrum assay further enhances the immunological toolkit available for evaluating M. tuberculosis-specific T cell responses across different states of M. tuberculosis infection, and can be readily implemented in resource-limited settings. Moreover, application of the assay to longitudinal cohorts will facilitate evaluation of treatment- or vaccine-induced changes in the breadth and specificity of Ag-specific T cell responses, as well as identification of M. tuberculosis-specific T cell responses associated with M. tuberculosis infection outcomes.
Collapse
Affiliation(s)
- Wendy E Whatney
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329
| | - Neel R Gandhi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | | | - Azhar Nizam
- Department of Biostatistics, Rollins School of Public Health, Emory University, Atlanta, GA 30322
| | - Hao Wu
- Department of Biostatistics, Rollins School of Public Health, Emory University, Atlanta, GA 30322
| | - Melanie J Quezada
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329
| | - Angela Campbell
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322
| | - Salim Allana
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322
| | - Mbuyi Madeleine Kabongo
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322
| | - Jeremiah Khayumbi
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Benson Muchiri
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Joshua Ongalo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Joan Tonui
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Loren E Sasser
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329
| | - Tawania J Fergus
- Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Gregory Sadat Ouma
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Samuel Gurrion Ouma
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Allison A Beck
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Mark J Mulligan
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | | | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433.,Department of Microbiology and Immunology, Tulane Health Sciences Center, New Orleans, LA 70112
| | - Kevin P Cain
- Division of Global HIV and Tuberculosis, U.S. Centers for Disease Control and Prevention, Kisumu 40100, Kenya; and
| | - Lance Waller
- Department of Biostatistics, Rollins School of Public Health, Emory University, Atlanta, GA 30322
| | - Henry M Blumberg
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - John D Altman
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329.,Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Joel D Ernst
- Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329; .,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Cheryl L Day
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329; .,Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | | |
Collapse
|
24
|
Steigler P, Daniels NJ, McCulloch TR, Ryder BM, Sandford SK, Kirman JR. BCG vaccination drives accumulation and effector function of innate lymphoid cells in murine lungs. Immunol Cell Biol 2018; 96:379-389. [PMID: 29363172 DOI: 10.1111/imcb.12007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/30/2017] [Accepted: 01/03/2018] [Indexed: 12/11/2022]
Abstract
The tuberculosis (TB) vaccine bacille Calmette-Guérin (BCG) prevents disseminated childhood TB; however, it fails to protect against the more prevalent pulmonary TB. Limited understanding of the immune response to Mycobacterium tuberculosis, the causative agent of TB, has hindered development of improved vaccines. Although memory CD4 T cells are considered the main mediators of protection against TB, recent studies suggest there are other key subsets that contribute to antimycobacterial immunity. To that end, innate cells may be involved in the protective response. In this study, we investigated the primary response of innate lymphoid cells (ILCs) to BCG exposure. Using a murine model, we showed that ILCs increased in number in the lungs and lymph nodes in response to BCG vaccination. Additionally, there was significant production of the antimycobacterial cytokine IFN-γ by ILCs. As ILCs are located at mucosal sites, it was investigated whether mucosal vaccination (intranasal) stimulated an enhanced response compared to the traditional vaccination approach (intradermal or subcutaneous). Indeed, in response to intranasal vaccination, the number of ILCs, and IFN-γ production in NK cells and ILC1s in the lungs and lymph nodes, were higher than that provoked through intradermal or subcutaneous vaccination. This work provides the first evidence that BCG vaccination activates ILCs, paving the way for future research to elucidate the protective potential of ILCs against mycobacterial infection. Additionally, the finding that lung ILCs respond rigorously to mucosal vaccination may have implications for the delivery of novel TB vaccines.
Collapse
Affiliation(s)
- Pia Steigler
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Naomi J Daniels
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Tim R McCulloch
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.,The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Brin M Ryder
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Sarah K Sandford
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Joanna R Kirman
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
25
|
Chancellor A, Tocheva AS, Cave-Ayland C, Tezera L, White A, Al Dulayymi JR, Bridgeman JS, Tews I, Wilson S, Lissin NM, Tebruegge M, Marshall B, Sharpe S, Elliott T, Skylaris CK, Essex JW, Baird MS, Gadola S, Elkington P, Mansour S. CD1b-restricted GEM T cell responses are modulated by Mycobacterium tuberculosis mycolic acid meromycolate chains. Proc Natl Acad Sci U S A 2017; 114:E10956-E10964. [PMID: 29158404 PMCID: PMC5754766 DOI: 10.1073/pnas.1708252114] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, remains a major human pandemic. Germline-encoded mycolyl lipid-reactive (GEM) T cells are donor-unrestricted and recognize CD1b-presented mycobacterial mycolates. However, the molecular requirements governing mycolate antigenicity for the GEM T cell receptor (TCR) remain poorly understood. Here, we demonstrate CD1b expression in TB granulomas and reveal a central role for meromycolate chains in influencing GEM-TCR activity. Meromycolate fine structure influences T cell responses in TB-exposed individuals, and meromycolate alterations modulate functional responses by GEM-TCRs. Computational simulations suggest that meromycolate chain dynamics regulate mycolate head group movement, thereby modulating GEM-TCR activity. Our findings have significant implications for the design of future vaccines that target GEM T cells.
Collapse
Affiliation(s)
- Andrew Chancellor
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Public Health England, National Infections Service, Porton Down, Salisbury SP4 0JQ, United Kingdom
| | - Anna S Tocheva
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Chris Cave-Ayland
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Liku Tezera
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Andrew White
- Public Health England, National Infections Service, Porton Down, Salisbury SP4 0JQ, United Kingdom
| | - Juma'a R Al Dulayymi
- School of Chemistry, Bangor University, Bangor, Gwynedd LL57 2UW, United Kingdom
| | | | - Ivo Tews
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Susan Wilson
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Histochemistry Unit, University of Southampton, Southampton SO16 6YD, United Kingdom
| | | | - Marc Tebruegge
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton SO17 1BJ, United Kingdom
- Global Health Research Institute, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Department of Paediatrics, Faculty of Medicine, University of Melbourne, 3052 Parkville, Australia
- Department of Paediatric Infectious Diseases & Immunology, Evelina London Children's Hospital, Guy's and St. Thomas' NHS Foundation Trust, London SE1 7EH, United Kingdom
| | - Ben Marshall
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton SO17 1BJ, United Kingdom
| | - Sally Sharpe
- Public Health England, National Infections Service, Porton Down, Salisbury SP4 0JQ, United Kingdom
| | - Tim Elliott
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Chris-Kriton Skylaris
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Jonathan W Essex
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Mark S Baird
- School of Chemistry, Bangor University, Bangor, Gwynedd LL57 2UW, United Kingdom
| | - Stephan Gadola
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Paul Elkington
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton SO17 1BJ, United Kingdom
- Global Health Research Institute, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Salah Mansour
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom;
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
26
|
Ahmad F, Zubair S, Gupta P, Gupta UD, Patel R, Owais M. Evaluation of Aggregated Ag85B Antigen for Its Biophysical Properties, Immunogenicity, and Vaccination Potential in a Murine Model of Tuberculosis Infection. Front Immunol 2017; 8:1608. [PMID: 29230211 PMCID: PMC5711810 DOI: 10.3389/fimmu.2017.01608] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 11/07/2017] [Indexed: 11/13/2022] Open
Abstract
Protein aggregates have been reported to act as a reservoir that can release biologically active, native form of precursor protein. Keeping this fact into consideration, it is tempting to exploit protein aggregate-based antigen delivery system as a functional vaccine to expand desirable immunological response in the host. Herein, we explored the capacity of aggregated Ag85B of Mycobacterium tuberculosis (Mtb) to act as a prophylactic vaccine system that releases the precursor antigen in slow and sustained manner. Being particulate system with exposed hydrophobic residues, aggregated Ag85B is likely to be avidly taken up by both phagocytosis as well as fusion with plasma membrane of antigen presenting cells, leading to its direct delivery to their cytosol. Its unique ability to access cytosol of target cells is further evident from the fact that immunization with aggregated Ag85B led to the induction of Th1-dominant immune response along with upregulated expression of qualitatively superior polyfunctional T cells in the mice. Antibodies generated following immunization with aggregated antigen recognized both native and monomeric Ag85B released from protein aggregate. The implicated immunization strategy offers protection at par to that of established BCG vaccine with desirable central and effector memory responses against subsequent Mtb aerosol challenge. The study highlights the potential of aggregated Ag85B as promising antigen delivery system and paves the way to design better prophylactic regimes against various intracellular pathogens including Mtb.
Collapse
Affiliation(s)
- Faraz Ahmad
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Swaleha Zubair
- Department of Computer Science, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Pushpa Gupta
- BSL-3 Experimental Animal Facility, Department of Animal Experimentation, National JALMA Institute for Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | - Umesh Datta Gupta
- BSL-3 Experimental Animal Facility, Department of Animal Experimentation, National JALMA Institute for Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | - Rakesh Patel
- Department of Immunology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | - Mohammad Owais
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
27
|
Mishra SK, Tripathi G, Kishore N, Singh RK, Singh A, Tiwari VK. Drug development against tuberculosis: Impact of alkaloids. Eur J Med Chem 2017. [DOI: 10.1016/j.ejmech.2017.06.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
28
|
Fonseca KL, Rodrigues PNS, Olsson IAS, Saraiva M. Experimental study of tuberculosis: From animal models to complex cell systems and organoids. PLoS Pathog 2017; 13:e1006421. [PMID: 28817682 PMCID: PMC5560521 DOI: 10.1371/journal.ppat.1006421] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is a devastating disease to mankind that has killed more people than any other infectious disease. Despite many efforts and successes from the scientific and health communities, the prospect of TB elimination remains distant. On the one hand, sustainable public health programs with affordable and broad implementation of anti-TB measures are needed. On the other hand, achieving TB elimination requires critical advances in three areas: vaccination, diagnosis, and treatment. It is also well accepted that succeeding in advancing these areas requires a deeper knowledge of host—pathogen interactions during infection, and for that, better experimental models are needed. Here, we review the potential and limitations of different experimental approaches used in TB research, focusing on animal and human-based cell culture models. We highlight the most recent advances in developing in vitro 3D models and introduce the potential of lung organoids as a new tool to study Mycobacterium tuberculosis infection. Tuberculosis (TB) is the number 1 killer in the world due to a bacterial infection. The study of this disease through clinical and epidemiological data and through the use of different experimental models has provided important knowledge on the role of the immune response generated during infection. This is critical for the development of novel vaccines and therapeutic strategies. However, in spite of the advances made, it is well accepted that better models are needed to study TB. This review discusses the different models used to study TB, highlighting the advantages and disadvantages of the available animal and cellular models and introducing recently developed state-of-the-art approaches based on human-based cell culture systems. These new advances are integrated in a road map for future study of TB, converging for the potential of lung organoids in TB research.
Collapse
Affiliation(s)
- Kaori L. Fonseca
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Pedro N. S. Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - I. Anna S. Olsson
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
29
|
van Zyl-Smit RN, Esmail A, Bateman ME, Dawson R, Goldin J, van Rikxoort E, Douoguih M, Pau MG, Sadoff JC, McClain JB, Snowden MA, Benko J, Hokey DA, Rutkowski KT, Graves A, Shepherd B, Ishmukhamedov S, Kagina BMN, Abel B, Hanekom WA, Scriba TJ, Bateman ED. Safety and Immunogenicity of Adenovirus 35 Tuberculosis Vaccine Candidate in Adults with Active or Previous Tuberculosis. A Randomized Trial. Am J Respir Crit Care Med 2017; 195:1171-1180. [PMID: 28060545 DOI: 10.1164/rccm.201603-0654oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Administration of tuberculosis (TB) vaccines in participants with previous or current pulmonary TB may have the potential for causing harmful postvaccination immunologic (Koch-type) reactions. OBJECTIVES To assess the safety and immunogenicity of three dose levels of the AERAS-402 live, replication-deficient adenovirus 35-vectored TB candidate vaccine, containing three mycobacterial antigens, in individuals with current or previous pulmonary TB. METHODS We performed a phase II randomized, placebo-controlled, double-blinded dose-escalation study in an HIV-negative adult South African cohort (n = 72) with active pulmonary TB (on treatment for 1-4 mo) or pulmonary TB treated at least 12 months before study entry and considered cured. Safety endpoints included clinical assessment, flow volume curves, diffusing capacity of the lung for carbon monoxide, pulse oximetry, chest radiograph, and high-resolution thoracic computerized tomography scans. Cytokine expression by CD4 and CD8 T cells, after stimulation with Ag85A, Ag85B, and TB10.4 peptide pools, was examined by intracellular cytokine staining. MEASUREMENTS AND MAIN RESULTS No apparent temporal or dose-related changes in clinical status (specifically acute, Koch phenomenon-like reactions), lung function, or radiology attributable to vaccine were observed. Injection site reactions were mild or moderate. Hematuria (by dipstick only) occurred in 25 (41%) of 61 AERAS-402 recipients and 3 (27%) of 11 placebo recipients, although no gross hematuria was reported. AERAS-402 induced robust CD8+ and moderate CD4+ T-cell responses, mainly to Ag85B in both vaccine groups. CONCLUSIONS Administration of the AERAS-402 candidate TB vaccine to participants with current or previous pulmonary TB induced a robust immune response and is not associated with clinically significant pulmonary complications. Clinical trial registered with www.clinicaltrials.gov (NCT 02414828) and in the South African National Clinical Trials Register ( www.sanctr.gov.za DOH 27-0808-2060).
Collapse
Affiliation(s)
- Richard N van Zyl-Smit
- 1 University of Cape Town Lung Institute, Division of Pulmonology, Department of Medicine
| | - Aliasgar Esmail
- 1 University of Cape Town Lung Institute, Division of Pulmonology, Department of Medicine
| | - Mary E Bateman
- 1 University of Cape Town Lung Institute, Division of Pulmonology, Department of Medicine
| | - Rodney Dawson
- 1 University of Cape Town Lung Institute, Division of Pulmonology, Department of Medicine
| | | | - Eva van Rikxoort
- 3 Department of Radiology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Macaya Douoguih
- 4 Crucell Holland B.V., a Janssen Pharmaceutical company of Johnson & Johnson, Leiden, the Netherlands
| | - Maria Grazia Pau
- 4 Crucell Holland B.V., a Janssen Pharmaceutical company of Johnson & Johnson, Leiden, the Netherlands
| | - Jerald C Sadoff
- 4 Crucell Holland B.V., a Janssen Pharmaceutical company of Johnson & Johnson, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | - Benjamin M N Kagina
- 6 South African TB Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, and.,7 Vaccines for Africa Initiative, Division of Medical Microbiology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Brian Abel
- 6 South African TB Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, and.,8 Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Willem A Hanekom
- 6 South African TB Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, and
| | - Thomas J Scriba
- 6 South African TB Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, and
| | - Eric D Bateman
- 1 University of Cape Town Lung Institute, Division of Pulmonology, Department of Medicine
| |
Collapse
|
30
|
Sun X, Yu W, Pang Q, Hu T. Conjugation Reaction with 8-Arm PEG Markedly Improves the Immunogenicity of Mycobacterium tuberculosis CFP10-TB10.4 Fusion Protein. Bioconjug Chem 2017; 28:1658-1668. [DOI: 10.1021/acs.bioconjchem.7b00131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Xiaowei Sun
- College
of Animal Science, Shanxi Agricultural University, Taigu 030801, Shanxi Province, China
- State
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Weili Yu
- State
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Quanhai Pang
- College
of Animal Science, Shanxi Agricultural University, Taigu 030801, Shanxi Province, China
| | - Tao Hu
- State
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
31
|
Tkachuk AP, Gushchin VA, Potapov VD, Demidenko AV, Lunin VG, Gintsburg AL. Multi-subunit BCG booster vaccine GamTBvac: Assessment of immunogenicity and protective efficacy in murine and guinea pig TB models. PLoS One 2017; 12:e0176784. [PMID: 28453555 PMCID: PMC5409163 DOI: 10.1371/journal.pone.0176784] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/17/2017] [Indexed: 11/18/2022] Open
Abstract
New innovative vaccines are highly needed to combat the global threat posed by tuberculosis. Efficient components-antigens and adjuvants-are crucial for development of modern recombinant TB vaccines. This study describes a new vaccine (GamTBvac) consisting of two mycobacterial antigen fusions (Ag85A and ESAT6-CFP10)-with dextran-binding domain immobilized on dextran and mixed with an adjuvant consisting of DEAE-dextran core, and with CpG oligodeoxynucleotides (TLR9 agonists). GamTBvac and its components were assessed for immunogenicity and protective efficacy in GamTBvac-prime/boost and BCG-prime/ GamTBvac-boost in murine and guinea pig TB models. Results show that in both infectious models, GamTBvac has a strong immunogenicity and significant protective effect against Mycobacterium tuberculosis strain H37Rv under aerosol and intravenous challenges. GamTBvac showed a particularly strong protective effect as a BCG booster vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intravenous
- Aerosols
- Animals
- Antibodies, Bacterial/blood
- BCG Vaccine/immunology
- Cell Proliferation/physiology
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Female
- Guinea Pigs
- Immunization
- Immunization, Secondary
- Immunogenicity, Vaccine
- Lung/immunology
- Lymph Nodes/immunology
- Male
- Mice, Inbred C57BL
- Mycobacterium tuberculosis/immunology
- Spleen/immunology
- T-Lymphocytes/immunology
- Tuberculosis/immunology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- A. P. Tkachuk
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. A. Gushchin
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - V. D. Potapov
- State Research Center for Applied Microbiology and Biotechnology, Obolensk, Russia
| | - A. V. Demidenko
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. G. Lunin
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - A. L. Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
32
|
Latently and uninfected healthcare workers exposed to TB make protective antibodies against Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2017; 114:5023-5028. [PMID: 28438994 DOI: 10.1073/pnas.1611776114] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The role of Igs in natural protection against infection by Mycobacterium tuberculosis (Mtb), the causative agent of TB, is controversial. Although passive immunization with mAbs generated against mycobacterial antigens has shown protective efficacy in murine models of infection, studies in B cell-depleted animals only showed modest phenotypes. We do not know if humans make protective antibody responses. Here, we investigated whether healthcare workers in a Beijing TB hospital-who, although exposed to suprainfectious doses of pathogenic Mtb, remain healthy-make antibody responses that are effective in protecting against infection by Mtb. We tested antibodies isolated from 48 healthcare workers and compared these with 12 patients with active TB. We found that antibodies from 7 of 48 healthcare workers but none from active TB patients showed moderate protection against Mtb in an aerosol mouse challenge model. Intriguingly, three of seven healthcare workers who made protective antibody responses had no evidence of prior TB infection by IFN-γ release assay. There was also good correlation between protection observed in vivo and neutralization of Mtb in an in vitro human whole-blood assay. Antibodies mediating protection were directed against the surface of Mtb and depended on both immune complexes and CD4+ T cells for efficacy. Our results indicate that certain individuals make protective antibodies against Mtb and challenge paradigms about the nature of an effective immune response to TB.
Collapse
|
33
|
Protective efficacy of a lipid antigen vaccine in a guinea pig model of tuberculosis. Vaccine 2017; 35:1395-1402. [DOI: 10.1016/j.vaccine.2017.01.079] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/12/2017] [Accepted: 01/30/2017] [Indexed: 01/25/2023]
|
34
|
Tezera LB, Bielecka MK, Chancellor A, Reichmann MT, Shammari BA, Brace P, Batty A, Tocheva A, Jogai S, Marshall BG, Tebruegge M, Jayasinghe SN, Mansour S, Elkington PT. Dissection of the host-pathogen interaction in human tuberculosis using a bioengineered 3-dimensional model. eLife 2017; 6. [PMID: 28063256 PMCID: PMC5238961 DOI: 10.7554/elife.21283] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Cell biology differs between traditional cell culture and 3-dimensional (3-D) systems, and is modulated by the extracellular matrix. Experimentation in 3-D presents challenges, especially with virulent pathogens. Mycobacterium tuberculosis (Mtb) kills more humans than any other infection and is characterised by a spatially organised immune response and extracellular matrix remodelling. We developed a 3-D system incorporating virulent mycobacteria, primary human blood mononuclear cells and collagen–alginate matrix to dissect the host-pathogen interaction. Infection in 3-D led to greater cellular survival and permitted longitudinal analysis over 21 days. Key features of human tuberculosis develop, and extracellular matrix integrity favours the host over the pathogen. We optimised multiparameter readouts to study emerging therapeutic interventions: cytokine supplementation, host-directed therapy and immunoaugmentation. Each intervention modulates the host-pathogen interaction, but has both beneficial and harmful effects. This methodology has wide applicability to investigate infectious, inflammatory and neoplastic diseases and develop novel drug regimes and vaccination approaches. DOI:http://dx.doi.org/10.7554/eLife.21283.001
Collapse
Affiliation(s)
- Liku B Tezera
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Magdalena K Bielecka
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew Chancellor
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Michaela T Reichmann
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Basim Al Shammari
- King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, Department of Infectious Diseases, MNGHA, Riyadh, Saudi Arabia
| | - Patience Brace
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Alex Batty
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Annie Tocheva
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Sanjay Jogai
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ben G Marshall
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Marc Tebruegge
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Suwan N Jayasinghe
- BioPhysics Group, UCL Institute of Biomedical Engineering, UCL Centre for Stem Cells and Regenerative Medicine and UCL Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Salah Mansour
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul T Elkington
- NIHR Respiratory Biomedical Research Unit, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
35
|
Wowk PF, Franco LH, Fonseca DMD, Paula MO, Vianna ÉDSO, Wendling AP, Augusto VM, Elói-Santos SM, Teixeira-Carvalho A, Silva FDC, Vinhas SA, Martins-Filho OA, Palaci M, Silva CL, Bonato VLD. Mycobacterial Hsp65 antigen upregulates the cellular immune response of healthy individuals compared with tuberculosis patients. Hum Vaccin Immunother 2017; 13:1040-1050. [PMID: 28059670 DOI: 10.1080/21645515.2016.1264547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previously we showed that 65-kDa Mycobacterium leprae heat shock protein (Hsp65) is a target for the development of a tuberculosis vaccine. Here we evaluated peripheral blood mononuclear cells (PBMC) from healthy individuals or tuberculosis patients stimulated with two forms of Hsp65 antigen, recombinant DNA that encodes Hsp65 (DNA-HSP65) or recombinant Hsp65 protein (rHsp65) in attempting to mimic a prophylactic or therapeutic study in vitro, respectively. Proliferation and cytokine-producing CD4+ or CD8+ cell were assessed by flow cytometry. The CD4+ cell proliferation from healthy individuals was stimulated by DNA-HSP65 and rHsp65, while CD8+ cell proliferation from healthy individuals or tuberculosis patients was stimulated by rHSP65. DNA-HSP65 did not improve the frequency of IFN-gamma+ cells from healthy individuals or tuberculosis patients. Furthermore, we found an increase in the frequency of IL-10-producing cells in both groups. These findings show that Hsp65 antigen activates human lymphocytes and plays an immune regulatory role that should be addressed as an additional antigen for the development of antigen-combined therapies.
Collapse
Affiliation(s)
- Pryscilla Fanini Wowk
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Luís Henrique Franco
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Denise Morais da Fonseca
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Marina Oliveira Paula
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | | | - Ana Paula Wendling
- c Laboratory of Biomarkers for Diagnosis and Monitoring , René Rachou Research Center , Fiocruz, Belo Horizonte , Minas Gerais , Brazil
| | | | | | - Andréa Teixeira-Carvalho
- c Laboratory of Biomarkers for Diagnosis and Monitoring , René Rachou Research Center , Fiocruz, Belo Horizonte , Minas Gerais , Brazil
| | - Flávia Dias Coelho Silva
- e Mycobacteriology Laboratory, Nucleus of Infectious Diseases , Federal University of Espírito Santo , Vitória , Espírito Santos , Brazil
| | - Solange Alves Vinhas
- e Mycobacteriology Laboratory, Nucleus of Infectious Diseases , Federal University of Espírito Santo , Vitória , Espírito Santos , Brazil
| | - Olindo Assis Martins-Filho
- c Laboratory of Biomarkers for Diagnosis and Monitoring , René Rachou Research Center , Fiocruz, Belo Horizonte , Minas Gerais , Brazil
| | - Moisés Palaci
- e Mycobacteriology Laboratory, Nucleus of Infectious Diseases , Federal University of Espírito Santo , Vitória , Espírito Santos , Brazil
| | - Célio Lopes Silva
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Vânia Luiza Deperon Bonato
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| |
Collapse
|
36
|
Fletcher HA, Schrager L. TB vaccine development and the End TB Strategy: importance and current status. Trans R Soc Trop Med Hyg 2017; 110:212-8. [PMID: 27076508 PMCID: PMC4830404 DOI: 10.1093/trstmh/trw016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/17/2016] [Indexed: 01/12/2023] Open
Abstract
TB is now the leading, global cause of death due to a single infectious microbe. To achieve the End TB vision of reducing TB by 90% by 2035 we will need new interventions. The objectives of this manuscript are to summarize the status of the clinical TB vaccine pipeline; to assess the challenges facing the TB development field; and to discuss some of the key strategies being embraced by the field to overcome these challenges. Currently, 8 of the 13 vaccines in clinical development are subunit vaccines; 6 of these contain or express either Ag85A or Ag85B proteins. A major challenge to TB vaccine development is the lack of diversity in both the antigens included in TB vaccines, and the immune responses elicited by TB vaccine candidates. Both will need to be expanded to maximise the potential for developing a successful candidate by 2025. Current research efforts are focused on broadening both antigen selection and the range of vaccine-mediated immune responses. Previous and ongoing TB vaccine efficacy trials have built capacity, generated high quality data on TB incidence and prevalence, and provided insight into immune correlates of risk of TB disease. These gains will enable the design of better TB vaccines and, importantly, move these vaccines into efficacy trials more rapidly and at a lower cost than was possible for previous TB vaccine candidates.
Collapse
Affiliation(s)
- Helen A Fletcher
- Immunology and Infection Department, London School of Hygiene & Tropical Medicine, UK, W1CE7HT
| | | |
Collapse
|
37
|
Abstract
New and effective tuberculosis (TB) vaccines are urgently needed to control pulmonary TB, and in particular to prevent the spread of drug-resistant strains of Mycobacterium tuberculosis. These drug-resistant strains can range from those resistant to first-line drugs to those that are almost impossible to treat. To develop new and effective vaccines for HIV and malaria has been difficult and it is proving to be just as challenging for TB. TB is a complicated disease with a spectrum from apparently controlled latent infection to active clinical disease and so different types of preventive or post-exposure vaccine may be needed. Identifying the most promising vaccine candidates to move into clinical trials is difficult, as we lack biomarker signatures that can predict protective efficacy. There is a risk that the failure of the MVA-85A vaccine to show efficacy when given to previously BCG-vaccinated South African infants will impact on the resources available for the development and trials of other candidate TB vaccines. Continued support for the development of new TB vaccines should remain a priority as an effective vaccine would bring huge public health benefits.
Collapse
|
38
|
Booty MG, Barreira-Silva P, Carpenter SM, Nunes-Alves C, Jacques MK, Stowell BL, Jayaraman P, Beamer G, Behar SM. IL-21 signaling is essential for optimal host resistance against Mycobacterium tuberculosis infection. Sci Rep 2016; 6:36720. [PMID: 27819295 PMCID: PMC5098191 DOI: 10.1038/srep36720] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 10/19/2016] [Indexed: 12/28/2022] Open
Abstract
IL-21 is produced predominantly by activated CD4+ T cells and has pleiotropic effects on immunity via the IL-21 receptor (IL-21R), a member of the common gamma chain (γc) cytokine receptor family. We show that IL-21 signaling plays a crucial role in T cell responses during Mycobacterium tuberculosis infection by augmenting CD8+ T cell priming, promoting T cell accumulation in the lungs, and enhancing T cell cytokine production. In the absence of IL-21 signaling, more CD4+ and CD8+ T cells in chronically infected mice express the T cell inhibitory molecules PD-1 and TIM-3. We correlate these immune alterations with increased susceptibility of IL-21R−/− mice, which have increased lung bacterial burden and earlier mortality compared to WT mice. Finally, to causally link the immune defects with host susceptibility, we use an adoptive transfer model to show that IL-21R−/− T cells transfer less protection than WT T cells. These results prove that IL-21 signaling has an intrinsic role in promoting the protective capacity of T cells. Thus, the net effect of IL-21 signaling is to enhance host resistance to M. tuberculosis. These data position IL-21 as a candidate biomarker of resistance to tuberculosis.
Collapse
Affiliation(s)
- Matthew G Booty
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US.,Program in Immunology, Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts 02115, US
| | - Palmira Barreira-Silva
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, Portuguese Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Stephen M Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US
| | - Cláudio Nunes-Alves
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US
| | - Miye K Jacques
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US
| | - Britni L Stowell
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US
| | - Pushpa Jayaraman
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US
| | - Gillian Beamer
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, Grafton, MA 01536, US
| | - Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, US
| |
Collapse
|
39
|
Lindestam Arlehamn CS, McKinney DM, Carpenter C, Paul S, Rozot V, Makgotlho E, Gregg Y, van Rooyen M, Ernst JD, Hatherill M, Hanekom WA, Peters B, Scriba TJ, Sette A. A Quantitative Analysis of Complexity of Human Pathogen-Specific CD4 T Cell Responses in Healthy M. tuberculosis Infected South Africans. PLoS Pathog 2016; 12:e1005760. [PMID: 27409590 PMCID: PMC4943605 DOI: 10.1371/journal.ppat.1005760] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/18/2016] [Indexed: 12/13/2022] Open
Abstract
We performed a quantitative analysis of the HLA restriction, antigen and epitope specificity of human pathogen specific responses in healthy individuals infected with M. tuberculosis (Mtb), in a South African cohort as a test case. The results estimate the breadth of T cell responses for the first time in the context of an infection and human population setting. We determined the epitope repertoire of eleven representative Mtb antigens and a large panel of previously defined Mtb epitopes. We estimated that our analytic methods detected 50-75% of the total response in a cohort of 63 individuals. As expected, responses were highly heterogeneous, with responses to a total of 125 epitopes detected. The 66 top epitopes provided 80% coverage of the responses identified in our study. Using a panel of 48 HLA class II-transfected antigen-presenting cells, we determined HLA class II restrictions for 278 epitope/donor recognition events (36% of the total). The majority of epitopes were restricted by multiple HLA alleles, and 380 different epitope/HLA combinations comprised less than 30% of the estimated Mtb-specific response. Our results underline the complexity of human T cell responses at a population level. Efforts to capture and characterize this broad and highly HLA promiscuous Mtb-specific T cell epitope repertoire will require significant peptide multiplexing efforts. We show that a comprehensive "megapool" of Mtb peptides captured a large fraction of the Mtb-specific T cells and can be used to characterize this response.
Collapse
Affiliation(s)
- Cecilia S. Lindestam Arlehamn
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, La Jolla, California, United States of America
| | - Denise M. McKinney
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, La Jolla, California, United States of America
| | - Chelsea Carpenter
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, La Jolla, California, United States of America
| | - Sinu Paul
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, La Jolla, California, United States of America
| | - Virginie Rozot
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Edward Makgotlho
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Yolande Gregg
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Michele van Rooyen
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Joel D. Ernst
- Department of Medicine, Division of Infectious Diseases, New York University School of Medicine, New York, New York, United States of America
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Willem A. Hanekom
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, La Jolla, California, United States of America
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, La Jolla, California, United States of America
| |
Collapse
|
40
|
García-Basteiro AL, Ruhwald M, Lange C. Design of tuberculosis vaccine trials under financial constraints. Expert Rev Vaccines 2016; 15:799-801. [PMID: 27078056 DOI: 10.1080/14760584.2016.1178067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Alberto L García-Basteiro
- a Centro de Investigação em Saúde de Manhiça (CISM) , Maputo , Mozambique.,b ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB) , Hospital Clínic - Universitat de Barcelona , Barcelona , Spain.,c Amsterdam Institute for Global Health and Development (AIGHD) , Amsterdam , The Netherlands
| | - Morten Ruhwald
- d Human immunology, Dept. of infectious disease immunology , Statens Serum Institut , Copenhagen , Denmark
| | - Christoph Lange
- e Division of Clinical Infectious Diseases , Research Center Borstel , Borstel , Germany.,f German Center for Infection Research , Clinical Tuberculosis Center , Borstel , Germany.,g International Health/Infectious Diseases , University of Lübeck , Lübeck , Germany.,h Department of Medicine , Karolinska Institute , Stockholm , Sweden.,i Department of Internal Medicine , University of Namibia School of Medicine , Windhoek , Namibia
| |
Collapse
|
41
|
Ziraldo C, Gong C, Kirschner DE, Linderman JJ. Strategic Priming with Multiple Antigens can Yield Memory Cell Phenotypes Optimized for Infection with Mycobacterium tuberculosis: A Computational Study. Front Microbiol 2016; 6:1477. [PMID: 26779136 PMCID: PMC4701940 DOI: 10.3389/fmicb.2015.01477] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/08/2015] [Indexed: 12/16/2022] Open
Abstract
Lack of an effective vaccine results in 9 million new cases of tuberculosis (TB) every year and 1.8 million deaths worldwide. Although many infants are vaccinated at birth with BCG (an attenuated M. bovis), this does not prevent infection or development of TB after childhood. Immune responses necessary for prevention of infection or disease are still unknown, making development of effective vaccines against TB challenging. Several new vaccines are ready for human clinical trials, but these trials are difficult and expensive; especially challenging is determining the appropriate cellular response necessary for protection. The magnitude of an immune response is likely key to generating a successful vaccine. Characteristics such as numbers of central memory (CM) and effector memory (EM) T cells responsive to a diverse set of epitopes are also correlated with protection. Promising vaccines against TB contain mycobacterial subunit antigens (Ag) present during both active and latent infection. We hypothesize that protection against different key immunodominant antigens could require a vaccine that produces different levels of EM and CM for each Ag-specific memory population. We created a computational model to explore EM and CM values, and their ratio, within what we term Memory Design Space. Our model captures events involved in T cell priming within lymph nodes and tracks their circulation through blood to peripheral tissues. We used the model to test whether multiple Ag-specific memory cell populations could be generated with distinct locations within Memory Design Space at a specific time point post vaccination. Boosting can further shift memory populations to memory cell ratios unreachable by initial priming events. By strategically varying antigen load, properties of cellular interactions within the LN, and delivery parameters (e.g., number of boosts) of multi-subunit vaccines, we can generate multiple Ag-specific memory populations that cover a wide range of Memory Design Space. Given a set of desired characteristics for Ag-specific memory populations, we can use our model as a tool to predict vaccine formulations that will generate those populations.
Collapse
Affiliation(s)
- Cordelia Ziraldo
- Department of Chemical Engineering, University of Michigan, Ann ArborMI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann ArborMI, USA
| | - Chang Gong
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann ArborMI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann ArborMI, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor MI, USA
| | - Jennifer J Linderman
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI, USA
| |
Collapse
|
42
|
Hunter RL. Tuberculosis as a three-act play: A new paradigm for the pathogenesis of pulmonary tuberculosis. Tuberculosis (Edinb) 2016; 97:8-17. [PMID: 26980490 DOI: 10.1016/j.tube.2015.11.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 11/22/2015] [Accepted: 11/29/2015] [Indexed: 01/08/2023]
Abstract
Lack of access to human tissues with untreated tuberculosis (TB) has forced generations of researchers to use animal models and to adopt a paradigm that granulomas are the characteristic lesion of both primary and post primary TB. An extended search of studies of human lung tissues failed to find any reports that support this paradigm. We found scores of publications from gross pathology in 1804 through high resolution CT scans in 2015 that identify obstructive lobular pneumonia, not granulomas, as the characteristic lesion of developing post-primary TB. This paper reviews this literature together with other relevant observations to formulate a new paradigm of TB with three distinct stages: a three-act play. First, primary TB, a war of attrition, begins with infection that spreads via lymphatics and blood stream before inducing systemic immunity that contains and controls the organisms within granulomas. Second, post-primary TB, a sneak attack, develops during latent TB as an asymptomatic obstructive lobular pneumonia in persons with effective systemic immunity. It is a paucibacillary process with no granulomas that spreads via bronchi and accumulates mycobacterial antigens and host lipids for 1-2 years before suddenly undergoing caseous necrosis. Third, the fallout, is responsible for nearly all clinical post primary disease. It begins with caseous necrotic pneumonia that is either retained to become the focus of fibrocaseous disease or is coughed out to leave a cavity. This three-stage paradigm suggests testable hypotheses and plausible answers to long standing questions of immunity to TB.
Collapse
Affiliation(s)
- Robert L Hunter
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center at Houston, MSB 2.136, 6431 Fannin, Houston, TX 77030, USA.
| |
Collapse
|
43
|
Abstract
A brief history of vaccination is presented since the Jenner's observation, through the first golden age of vaccinology (from Pasteur's era to 1938), the second golden age (from 1940 to 1970), until the current period. In the first golden age, live, such as Bacille Calmette Guérin (BCG), and yellow fever, inactivated, such as typhoid, cholera, plague, and influenza, and subunit vaccines, such as tetanus and diphtheria toxoids, have been developed. In the second golden age, the cell culture technology enabled polio, measles, mumps, and rubella vaccines be developed. In the era of modern vaccines, in addition to the conjugate polysaccharide, hepatitis A, oral typhoid, and varicella vaccines, the advent of molecular biology enabled to develop hepatitis B, acellular pertussis, papillomavirus, and rotavirus recombinant vaccines. Great successes have been achieved in the fight against infectious diseases, including the smallpox global eradication, the nearly disappearance of polio, the control of tetanus, diphtheria, measles, rubella, yellow fever, and rabies. However, much work should still be done for improving old vaccines, such as BCG, anthrax, smallpox, plague, or for developing effective vaccines against old or emerging infectious threats, such as human-immunodeficiency-virus, malaria, hepatitis C, dengue, respiratory-syncytial-virus, cytomegalovirus, multiresistant bacteria, Clostridium difficile, Ebola virus. In addition to search for innovative and effective vaccines and global infant coverage, even risk categories should adequately be protected. Despite patients under immunosuppressive therapy are globally increasing, their vaccine coverage is lower than recommended, even in developed and affluent countries.
Collapse
Affiliation(s)
| | - Simonetta Salemi
- c S. Andrea University Hospital , Via di Grottarossa Rome, Italy
| | - Raffaele D'Amelio
- b Sapienza University of Rome , Department of Clinical and Molecular Medicine , Via di Grottarossa Rome, Italy.,c S. Andrea University Hospital , Via di Grottarossa Rome, Italy
| |
Collapse
|
44
|
Nathan C. What can immunology contribute to the control of the world's leading cause of death from bacterial infection? Immunol Rev 2015; 264:2-5. [PMID: 25703548 DOI: 10.1111/imr.12277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Video podcast available Go to www.immunologicalreviews.com to watch an interview with Guest Editor Carl Nathan.
Collapse
Affiliation(s)
- Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
45
|
Abstract
The world is in need of more effective approaches to controlling tuberculosis. The development of improved control strategies has been hampered by deficiencies in the tools available for detecting Mycobacterium tuberculosis and defining the dynamic consequences of the interaction of M. tuberculosis with its human host. Key needs include a highly sensitive, specific nonsputum diagnostic; biomarkers predictive of responses to therapy; correlates of risk for disease development; and host response-independent markers of M. tuberculosis infection. Tools able to sensitively detect and quantify total body M. tuberculosis burden might well be transformative across many needed use cases. Here, we review the current state of the field, paying particular attention to needed changes in experimental paradigms that would facilitate the discovery, validation, and development of such tools.
Collapse
Affiliation(s)
- Jennifer L Gardiner
- Discovery and Translational Sciences, Global Health, Bill & Melinda Gates Foundation, Seattle, WA 98102
| | - Christopher L Karp
- Discovery and Translational Sciences, Global Health, Bill & Melinda Gates Foundation, Seattle, WA 98102
| |
Collapse
|
46
|
Carpenter C, Sidney J, Kolla R, Nayak K, Tomiyama H, Tomiyama C, Padilla OA, Rozot V, Ahamed SF, Ponte C, Rolla V, Antas PR, Chandele A, Kenneth J, Laxmi S, Makgotlho E, Vanini V, Ippolito G, Kazanova AS, Panteleev AV, Hanekom W, Mayanja-Kizza H, Lewinsohn D, Saito M, McElrath MJ, Boom WH, Goletti D, Gilman R, Lyadova IV, Scriba TJ, Kallas EG, Murali-Krishna K, Sette A, Lindestam Arlehamn CS. A side-by-side comparison of T cell reactivity to fifty-nine Mycobacterium tuberculosis antigens in diverse populations from five continents. Tuberculosis (Edinb) 2015; 95:713-721. [PMID: 26277695 DOI: 10.1016/j.tube.2015.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
We compared T cell recognition of 59 prevalently recognized Mycobacterium tuberculosis (MTB) antigens in individuals latently infected with MTB (LTBI), and uninfected individuals with previous BCG vaccination, from nine locations and populations with different HLA distribution, MTB exposure rates, and standards of TB care. This comparison revealed similar response magnitudes in diverse LTBI and BCG-vaccinated cohorts and significant correlation between responses in LTBIs from the USA and other locations. Many antigens were uniformly recognized, suggesting suitability for inclusion in vaccines targeting diverse populations. Several antigens were similarly immunodominant in LTBI and BCG cohorts, suggesting applicability for vaccines aimed at boosting BCG responses. The panel of MTB antigens will be valuable for characterizing MTB-specific CD4 T cell responses irrespective of ethnicity, infecting MTB strains and BCG vaccination status. Our results illustrate how a comparative analysis can provide insight into the relative immunogenicity of existing and novel vaccine candidates in LTBIs.
Collapse
Affiliation(s)
- Chelsea Carpenter
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, 9420 Athena Circle, La Jolla, 92037, USA
| | - John Sidney
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, 9420 Athena Circle, La Jolla, 92037, USA
| | - Ravi Kolla
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, 9420 Athena Circle, La Jolla, 92037, USA
| | - Kaustuv Nayak
- ICGEB-Emory Vaccine Centre, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Helena Tomiyama
- Division of Clinical Immunology and Allergy, University of São Paulo Medical School, São Paulo, Brazil
| | - Claudia Tomiyama
- Division of Clinical Immunology and Allergy, University of São Paulo Medical School, São Paulo, Brazil
| | - Oscar A Padilla
- Division of Clinical Immunology and Allergy, University of São Paulo Medical School, São Paulo, Brazil
| | - Virginie Rozot
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Department of Pediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Syed F Ahamed
- Division of Infectious Diseases, St. John's Research Institute, St. John's National Academy of Sciences, Sarjapur Road, Koramangala 2 Block, Bangaluru, Karnataka, 560034, India
| | - Carlos Ponte
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| | - Valeria Rolla
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| | - Paulo R Antas
- Clinical Immunology Laboratory, Oswaldo Cruz Institute-Fiocruz, Rio de Janeiro, Brazil
| | - Anmol Chandele
- ICGEB-Emory Vaccine Centre, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - John Kenneth
- Division of Infectious Diseases, St. John's Research Institute, St. John's National Academy of Sciences, Sarjapur Road, Koramangala 2 Block, Bangaluru, Karnataka, 560034, India
| | - Seetha Laxmi
- Department of Transfusion Medicine and Immunohematology, St. John's Medical College Hospital, St. John's National Academy of Health Sciences, Bangaluru, 560034, India
| | - Edward Makgotlho
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Department of Pediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Valentina Vanini
- Translational Research Unit, Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases, Rome, Italy
| | - Giuseppe Ippolito
- Translational Research Unit, Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases, Rome, Italy
| | - Alexandra S Kazanova
- Department of Immunology, Federal State Budgetary Scientific Institution "Central Tuberculosis Research Institute", Moscow, Russia
| | - Alexander V Panteleev
- Department of Immunology, Federal State Budgetary Scientific Institution "Central Tuberculosis Research Institute", Moscow, Russia
| | - Willem Hanekom
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Department of Pediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Harriet Mayanja-Kizza
- Department of Medicine, College of Health Sciences, Faculty of Medicine, Makerere University, Kampala, Uganda
| | | | - Mayuko Saito
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, 21205, USA; Universidad Peruana Caytano Hereida, Lima, Peru; Department of Virology, Tohoku University, Sendai, Japan
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, 98109, USA
| | - W Henry Boom
- Tuberculosis Research Unit, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, 44106, USA
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases, Rome, Italy
| | - Robert Gilman
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, 21205, USA; Universidad Peruana Caytano Hereida, Lima, Peru
| | - Irina V Lyadova
- Department of Immunology, Federal State Budgetary Scientific Institution "Central Tuberculosis Research Institute", Moscow, Russia
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Department of Pediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Esper G Kallas
- Division of Clinical Immunology and Allergy, University of São Paulo Medical School, São Paulo, Brazil
| | - Kaja Murali-Krishna
- ICGEB-Emory Vaccine Centre, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India; Emory Vaccine Center, 1501 Clifton Road, Atlanta, GA, 30329, USA; Department of Pediatrics, Emory University, 1760 Haygood Drive, Atlanta, GA, 30322, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, 9420 Athena Circle, La Jolla, 92037, USA
| | - Cecilia S Lindestam Arlehamn
- La Jolla Institute for Allergy and Immunology, Department of Vaccine Discovery, 9420 Athena Circle, La Jolla, 92037, USA.
| |
Collapse
|