1
|
Li Y, Warren JA, Poston TB, Clutton G, Shaw FR, Conrad SZ, Xu Y, Zheng X, Yount KS, O’Connell CM, Wiesenfeld HC, Darville T, Goonetilleke N. Low-Frequency, Sustained CD4 T-Cell Responses Chlamydia trachomatis in Women: Predominant Targeting of Chlamydial Proteaselike Activity Factor (CPAF). J Infect Dis 2025; 231:e385-e395. [PMID: 39250505 PMCID: PMC11841636 DOI: 10.1093/infdis/jiae443] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Chlamydia trachomatis (CT) is a globally prevalent sexually transmitted infection that can result in pelvic inflammatory disease, ectopic pregnancy, and infertility in women. Currently, there is no prophylactic vaccine. METHODS This study examined T-cell immunity in a cohort of women recently infected with CT. Participants were screened against peptides spanning 33 of 894 possible CT proteins, either ex vivo or using short-term cell lines. CT-specific T cells were characterized by interferon (IFN) γ enzyme-linked immunospot (ELISPOT) assay and flow cytometry. RESULTS Ex vivo CT-specific T cells were rarely detected; however, in vitro expanded CT-specific T cells were detected by IFN-γ ELISPOT in 90% (27 of 30) of participants. Notably, >50% of participants had T-cell responses targeting chlamydial proteaselike activity factor (CPAF). T-cell epitopes were dispersed across the CPAF protein. Flow cytometric analysis of short-term cell lines found that CT-specific cells, mainly CD4, produced IFN-γ and tumor necrosis factor (TNF) α and were sustained over 12 months. Ex vivo analysis suggested that CT-specific T cells mostly exhibited a central memory phenotype. CONCLUSIONS Our results indicate that CT infection elicits low-frequency, persistent CD4 T-cell responses in most women and that the secreted protein, CPAF, is an immunoprevalent CT antigen. Altogether, these data support development and testing of CT vaccines that enhance CD4 T cells against CPAF.
Collapse
Affiliation(s)
- Yanli Li
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joanna A Warren
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Taylor B Poston
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Genevieve Clutton
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Fiona R Shaw
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shayla Z Conrad
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yinyan Xu
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xiaojing Zheng
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kacy S Yount
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Catherine M O’Connell
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Harold C Wiesenfeld
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Pittsburgh School of Medicine, Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Toni Darville
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nilu Goonetilleke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Londt R, Semple L, Esmail A, Pooran A, Meldau R, Davids M, Dheda K, Tomasicchio M. Autologous Human Dendritic Cells from XDR-TB Patients Polarize a Th1 Response Which Is Bactericidal to Mycobacterium tuberculosis. Microorganisms 2025; 13:345. [PMID: 40005712 PMCID: PMC11857998 DOI: 10.3390/microorganisms13020345] [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: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Extensively drug-resistant tuberculosis (XDR-TB) is a public health concern as drug resistance is outpacing the drug development pipeline. Alternative immunotherapeutic approaches are needed. Peripheral blood mononuclear cells (PBMCs) were isolated from pre-XDR/XDR-TB (n = 25) patients and LTBI (n = 18) participants. Thereafter, monocytic-derived dendritic cells (mo-DCs) were co-cultured with M. tb antigens, with/without a maturation cocktail (interferon-γ, interferon-α, CD40L, IL-1β, and TLR3 and TLR7/8 agonists). Two peptide pools were evaluated: (i) an ECAT peptide pool (ESAT6, CFP10, Ag85B, and TB10.4 peptides) and (ii) a PE/PPE peptide pool. Sonicated lysate of the M. tb HN878 strain served as a control. Mo-DCs were assessed for DC maturation markers, Th1 cytokines, and the ability of the DC-primed PBMCs to restrict the growth of M. tb-infected monocyte-derived macrophages. In pre-XDR/XDR-TB, mo-DCs matured with M. tb antigens (ECAT or PE/PPE peptide pool, or HN878 lysate) + cocktail, compared to mo-DCs matured with M. tb antigens only, showed higher upregulation of co-stimulatory molecules and IL-12p70 (p < 0.001 for both comparisons). The matured mo-DCs had enhanced antigen-specific CD8+ T-cell responses to ESAT-6 (p = 0.05) and Ag85B (p = 0.03). Containment was higher with mo-DCs matured with the PE/PPE peptide pool cocktail versus mo-DCs matured with the PE/PPE peptide pool (p = 0.0002). Mo-DCs matured with the PE/PPE peptide pool + cocktail achieved better containment than the ECAT peptide pool + cocktail [50%, (IQR:39-75) versus 46%, (IQR:15-62); p = 0.02]. In patients with pre-XDR/XDR-TB, an effector response primed by mo-DCs matured with an ECAT or PE/PPE peptide pool + cocktail was capable of restricting the growth of M. tb in vitro.
Collapse
Affiliation(s)
- Rolanda Londt
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| | - Lynn Semple
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| | - Aliasgar Esmail
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| | - Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| | - Richard Meldau
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| | - Malika Davids
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Michele Tomasicchio
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town 7925, South Africa
- South Africa MRC Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
3
|
Li F, Dang W, Du Y, Xu X, He P, Zhou Y, Zhu B. Tuberculosis Vaccines and T Cell Immune Memory. Vaccines (Basel) 2024; 12:483. [PMID: 38793734 PMCID: PMC11125691 DOI: 10.3390/vaccines12050483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis (TB) remains a major infectious disease partly due to the lack of an effective vaccine. Therefore, developing new and more effective TB vaccines is crucial for controlling TB. Mycobacterium tuberculosis (M. tuberculosis) usually parasitizes in macrophages; therefore, cell-mediated immunity plays an important role. The maintenance of memory T cells following M. tuberculosis infection or vaccination is a hallmark of immune protection. This review analyzes the development of memory T cells during M. tuberculosis infection and vaccine immunization, especially on immune memory induced by BCG and subunit vaccines. Furthermore, the factors affecting the development of memory T cells are discussed in detail. The understanding of the development of memory T cells should contribute to designing more effective TB vaccines and optimizing vaccination strategies.
Collapse
Affiliation(s)
- Fei Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Wenrui Dang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Yunjie Du
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Xiaonan Xu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Pu He
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Yuhe Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Bingdong Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
- College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
4
|
Du J, Su Y, Wang R, Dong E, Cao Y, Zhao W, Gong W. Research progress on specific and non-specific immune effects of BCG and the possibility of BCG protection against COVID-19. Front Immunol 2023; 14:1118378. [PMID: 36798128 PMCID: PMC9927227 DOI: 10.3389/fimmu.2023.1118378] [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: 12/07/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Bacille Calmette-Guérin (BCG) is the only approved vaccine for tuberculosis (TB) prevention worldwide. BCG has an excellent protective effect on miliary tuberculosis and tuberculous meningitis in children or infants. Interestingly, a growing number of studies have shown that BCG vaccination can induce nonspecific and specific immunity to fight against other respiratory disease pathogens, including SARS-CoV-2. The continuous emergence of variants of SARS-CoV-2 makes the protective efficiency of COVID-19-specific vaccines an unprecedented challenge. Therefore, it has been hypothesized that BCG-induced trained immunity might protect against COVID-19 infection. This study comprehensively described BCG-induced nonspecific and specific immunity and the mechanism of trained immunity. In addition, this study also reviewed the research on BCG revaccination to prevent TB, the impact of BCG on other non-tuberculous diseases, and the clinical trials of BCG to prevent COVID-19 infection. These data will provide new evidence to confirm the hypotheses mentioned above.
Collapse
Affiliation(s)
- Jingli Du
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Yue Su
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Ruilan Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Enjun Dong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Yan Cao
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Wenjuan Zhao
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Negi K, Bhaskar A, Dwivedi VP. Progressive Host-Directed Strategies to Potentiate BCG Vaccination Against Tuberculosis. Front Immunol 2022; 13:944183. [PMID: 35967410 PMCID: PMC9365942 DOI: 10.3389/fimmu.2022.944183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
The pursuit to improve the TB control program comprising one approved vaccine, M. bovis Bacille Calmette-Guerin (BCG) has directed researchers to explore progressive approaches to halt the eternal TB pandemic. Mycobacterium tuberculosis (M.tb) was first identified as the causative agent of TB in 1882 by Dr. Robert Koch. However, TB has plagued living beings since ancient times and continues to endure as an eternal scourge ravaging even with existing chemoprophylaxis and preventive therapy. We have scientifically come a long way since then, but despite accessibility to the standard antimycobacterial antibiotics and prophylactic vaccine, almost one-fourth of humankind is infected latently with M.tb. Existing therapeutics fail to control TB, due to the upsurge of drug-resistant strains and increasing incidents of co-infections in immune-compromised individuals. Unresponsiveness to established antibiotics leaves patients with no therapeutic possibilities. Hence the search for an efficacious TB immunization strategy is a global health priority. Researchers are paving the course for efficient vaccination strategies with the radically advanced operation of core principles of protective immune responses against M.tb. In this review; we have reassessed the progression of the TB vaccination program comprising BCG immunization in children and potential stratagems to reinforce BCG-induced protection in adults.
Collapse
Affiliation(s)
| | | | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
6
|
Balle C, Armistead B, Kiravu A, Song X, Happel AU, Hoffmann AA, Kanaan SB, Nelson JL, Gray CM, Jaspan HB, Harrington WE. Factors influencing maternal microchimerism throughout infancy and its impact on infant T cell immunity. J Clin Invest 2022; 132:e148826. [PMID: 35550376 PMCID: PMC9246390 DOI: 10.1172/jci148826] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Determinants of the acquisition and maintenance of maternal microchimerism (MMc) during infancy and the impact of MMc on infant immune responses are unknown. We examined factors that influence MMc detection and level across infancy and the effect of MMc on T cell responses to bacillus Calmette-Guérin (BCG) vaccination in a cohort of HIV-exposed, uninfected and HIV-unexposed infants in South Africa. MMc was measured in whole blood from 58 infants using a panel of quantitative PCR assays at day 1, and 7, 15, and 36 weeks of life. Infants received BCG at birth, and selected whole blood samples from infancy were stimulated in vitro with BCG and assessed for polyfunctional CD4+ T cell responses. MMc was present in most infants across infancy, with levels ranging from 0 to 1,193/100,000 genomic equivalents and was positively impacted by absence of maternal HIV, maternal and infant HLA compatibility, infant female sex, and exclusive breastfeeding. Initiation of maternal antiretroviral therapy prior to pregnancy partially restored MMc level in HIV-exposed, uninfected infants. Birth MMc was associated with an improved polyfunctional CD4+ T cell response to BCG. These data emphasize that both maternal and infant factors influence the level of MMc, which may subsequently affect infant T cell responses.
Collapse
Affiliation(s)
- Christina Balle
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Blair Armistead
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Agano Kiravu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Xiaochang Song
- Seattle Children’s Research Institute, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Anna-Ursula Happel
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Angela A. Hoffmann
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Sami B. Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - J. Lee Nelson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Clive M. Gray
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Heather B. Jaspan
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics and
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Whitney E. Harrington
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics and
| |
Collapse
|
7
|
Soto JA, Gálvez NMS, Andrade CA, Ramírez MA, Riedel CA, Kalergis AM, Bueno SM. BCG vaccination induces cross-protective immunity against pathogenic microorganisms. Trends Immunol 2022; 43:322-335. [PMID: 35074254 DOI: 10.1016/j.it.2021.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 01/03/2023]
Abstract
Bacillus Calmette-Guérin (BCG) is an attenuated Mycobacterium bovis strain used as a vaccine to prevent Mycobacterium tuberculosis (M. tb) infection. Its ability to potentiate the immune response induced by other vaccines and to promote nonspecific immunomodulatory effects has been described. These effects can be triggered by epigenetic reprogramming and metabolic shifts on innate immune cells, a phenomenon known as trained immunity. The induction of trained immunity may contribute to explain why BCG vaccination effectively decreases disease symptoms caused by pathogens different from M. tb. This article explains the importance of BCG immunization and the possible mechanisms associated with the induction of trained immunity, which might be used as a strategy for rapid activation of the immune system against unrelated pathogens.
Collapse
Affiliation(s)
- Jorge A Soto
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Nicolás M S Gálvez
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Catalina A Andrade
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Mario A Ramírez
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Claudia A Riedel
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile.
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Santiago 8330025, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile.
| |
Collapse
|
8
|
Silva CS, Sundling C, Folkesson E, Fröberg G, Nobrega C, Canto-Gomes J, Chambers BJ, Lakshmikanth T, Brodin P, Bruchfeld J, Nigou J, Correia-Neves M, Källenius G. High Dimensional Immune Profiling Reveals Different Response Patterns in Active and Latent Tuberculosis Following Stimulation With Mycobacterial Glycolipids. Front Immunol 2021; 12:727300. [PMID: 34887849 PMCID: PMC8650708 DOI: 10.3389/fimmu.2021.727300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Upon infection with Mycobacterium tuberculosis (Mtb) the host immune response might clear the bacteria, control its growth leading to latent tuberculosis (LTB), or fail to control its growth resulting in active TB (ATB). There is however no clear understanding of the features underlying a more or less effective response. Mtb glycolipids are abundant in the bacterial cell envelope and modulate the immune response to Mtb, but the patterns of response to glycolipids are still underexplored. To identify the CD45+ leukocyte activation landscape induced by Mtb glycolipids in peripheral blood of ATB and LTB, we performed a detailed assessment of the immune response of PBMCs to the Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic precursor phosphatidyl-inositol mannoside (PIM), and purified-protein derivate (PPD). At 24 h of stimulation, cell profiling and secretome analysis was done using mass cytometry and high-multiplex immunoassay. PIM induced a diverse cytokine response, mainly affecting antigen-presenting cells to produce both pro-inflammatory and anti-inflammatory cytokines, but not IFN-γ, contrasting with PPD that was a strong inducer of IFN-γ. The effect of PIM on the antigen-presenting cells was partly TLR2-dependent. Expansion of monocyte subsets in response to PIM or LAM was reduced primarily in LTB as compared to healthy controls, suggesting a hyporesponsive/tolerance pattern derived from Mtb infection.
Collapse
Affiliation(s)
- Carolina S Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Elin Folkesson
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gabrielle Fröberg
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Claudia Nobrega
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - João Canto-Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Benedict J Chambers
- Center for Infectious Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Judith Bruchfeld
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal.,Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Tangie E, Walters A, Hsu NJ, Fisher M, Magez S, Jacobs M, Keeton R. BCG-mediated protection against M. tuberculosis is sustained post-malaria infection independent of parasite virulence. Immunology 2021; 165:219-233. [PMID: 34775598 DOI: 10.1111/imm.13431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB) and malaria remain serious threats to global health. Bacillus Calmette-Guerin (BCG), the only licensed vaccine against TB protects against severe disseminated forms of TB in infants but shows poor efficacy against pulmonary TB in adults. Co-infections have been reported as one of the factors implicated in vaccine inefficacy. Given the geographical overlap of malaria and TB in areas where BCG vaccination is routinely administered, we hypothesized that virulence-dependent co-infection with Plasmodium species could alter the BCG-specific immune responses thus resulting in failure to protect against Mycobacterium tuberculosis. We compared virulent Plasmodium berghei and non-virulent Plasmodium chabaudi, their effects on B cells, effector and memory T cells, and the outcome on BCG-induced efficacy against M. tuberculosis infection. We demonstrate that malaria co-infection modulates both B- and T-cell immune responses but does not significantly alter the ability of the BCG vaccine to inhibit the growth of M. tuberculosis irrespective of parasite virulence. This malaria-driven immune regulation may have serious consequences in the early clinical trials of novel vaccines, which rely on vaccine-specific T-cell responses to screen novel vaccines for progression to the more costly vaccine efficacy trials.
Collapse
Affiliation(s)
- Emily Tangie
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| | - Avril Walters
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| | - Nai-Jen Hsu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| | - Michelle Fisher
- South African Tuberculosis Vaccine Initiative, University of Cape Town, Observatory, South Africa
| | - Stefan Magez
- Laboratory for Cellular and Molecular Immunology (CMIM), Department of Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Gent, Belgium.,Laboratory for Biomedical Research, Department of Molecular Biotechnology, Environment Technology and Food Technology, Ghent University Global Campus, Incheon, Korea
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa.,National Health Laboratory Service, Cape Town, South Africa.,Infectious Disease Research Unit, University of Cape Town, Observatory, South Africa
| | - Roanne Keeton
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Observatory, South Africa
| |
Collapse
|
10
|
Ritter K, Behrends J, Erdmann H, Rousseau J, Hölscher A, Volz J, Prinz I, Lindenstrøm T, Hölscher C. Interleukin-23 instructs protective multifunctional CD4 T cell responses after immunization with the Mycobacterium tuberculosis subunit vaccine H1 DDA/TDB independently of interleukin-17A. J Mol Med (Berl) 2021; 99:1585-1602. [PMID: 34351501 PMCID: PMC8541990 DOI: 10.1007/s00109-021-02100-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 01/01/2023]
Abstract
Interleukin (IL)-17A-producing T helper (Th)17 cells are increasingly being acknowledged to be associated with protective immunity to Mycobacterium tuberculosis (Mtb). Subunit vaccines potently promote protective immune responses against Mtb infection that correlate with an expansion of IL-23-dependent Th17 cells. Previous studies revealed that after vaccination, IL-23 is required for protection against challenge with Mtb but the underlying IL-23-dependent-and possibly IL-17A-mediated-mechanisms remain elusive. Therefore, we here analyzed the early outcome of Mtb infection in C57BL/6, IL-23p19-deficient (-/-), and IL-17A-/- mice after vaccination with the subunit vaccine H1-DDA/TDB to investigate the role of the IL-23-Th17 immune axis for the instruction of vaccine-induced protection. While in IL-23p19-/- mice the protective effect was reduced, protection after vaccination was maintained in IL-17A-/- animals for the course of infection of 6 weeks, indicating that after vaccination with H1-DDA/TDB early protection against Mtb is-although dependent on IL-23-not mediated by IL-17A. In contrast, IL-17A deficiency appears to have an impact on maintaining long-term protection. In fact, IL-23 instructed the vaccine-induced memory immunity in the lung, in particular the sustained expansion of tumor necrosis factor (TNF)+IL-2+ multifunctional T cells, independently of IL-17A. Altogether, a targeted induction of IL-23 during vaccination against Mtb might improve the magnitude and quality of vaccine-induced memory immune responses. KEY MESSAGES: After subunit Mtb vaccination with H1-DDA/TDB, IL-23 but not IL-17A contributes to vaccine-induced early protection against infection with Mtb. IL-17F does not compensate for IL-17A deficiency in terms of H1-DDA/TDB-induced protection against Mtb infection. IL 23 promotes the H1-DDA/TDB-induced accumulation of effector memory T cells independently of IL 17A. IL-23 arbitrates the induction of H1-specific IFN-γ-TNF+IL-2+ double-positive multifunctional CD4 T cells after subunit Mtb vaccination in an IL-17A-independent manner.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Jochen Behrends
- Fluorescence Cytometry Core Unit, Research Center Borstel, Borstel, Germany
| | - Hanna Erdmann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Jasmin Rousseau
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | | | - Johanna Volz
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Center for Molecular Neurobiology Hamburg, Eppendorf University Medical Center, Hamburg, Germany
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | |
Collapse
|
11
|
T-Cell Cytokine Response in Salmonella Typhimurium-Vaccinated versus Infected Pigs. Vaccines (Basel) 2021; 9:vaccines9080845. [PMID: 34451970 PMCID: PMC8402558 DOI: 10.3390/vaccines9080845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 11/16/2022] Open
Abstract
Vaccination with the live attenuated vaccine Salmoporc is an effective measure to control Salmonella Typhimurium (STM) in affected swine populations. However, the cellular immune response evoked by the Salmoporc vaccine including differences in vaccinated pigs versus non-vaccinated pigs upon STM infection have not been characterized yet. To investigate this, tissue-derived porcine lymphocytes from different treatment groups (vaccination-only, vaccination and infection, infection-only, untreated controls) were stimulated in vitro with heat-inactivated STM and abundances of IFN-γ, TNF-α and/or IL-17A-producing T-cell subsets were compared across organs and treatment groups. Overall, our results show the induction of a strong CD4+ T-cell response after STM infection, both locally and systemically. Low-level induction of STM-specific cytokine-producing CD4+ T cells, notably for the IFN-γ/TNF-α co-producing phenotype, was detected after vaccination-only. Numerous significant contrasts in cytokine-producing T-cell phenotypes were observed after infection in vaccinated and infected versus infected-only animals. These results suggest that vaccine-induced STM-specific cytokine-producing CD4+ T cells contribute to local immunity in the gut and may limit the spread of STM to lymph nodes and systemic organs. Hence, our study provides insights into the underlying immune mechanisms that account for the efficacy of the Salmoporc vaccine.
Collapse
|
12
|
Khakhum N, Bharaj P, Walker DH, Torres AG, Endsley JJ. Antigen-specific antibody and polyfunctional T cells generated by respiratory immunization with protective Burkholderia ΔtonB Δhcp1 live attenuated vaccines. NPJ Vaccines 2021; 6:72. [PMID: 33986290 PMCID: PMC8119421 DOI: 10.1038/s41541-021-00333-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/18/2021] [Indexed: 11/09/2022] Open
Abstract
Melioidosis, caused by Burkholderia pseudomallei (Bpm), lacks a vaccine. We identify the immune correlates of protection induced by B. mallei ΔtonB Δhcp1 (CLH001) and Bpm ΔtonB Δhcp1 (PBK001) vaccines against inhalational melioidosis. Mucosal immunization with either vaccine generates Bpm-specific IgM and IgG (IgG2b/c > IgG1 > IgG3) antibodies in sera and lungs, and lung IgA antibodies. Sera confers complement-independent bactericidal activity and macrophages opsonophagocytic uptake but is insufficient in passive transfer experiments to provide significant protection. Both vaccines elicit memory Th1 and Th17 CD4+ T-cell responses in lung and spleen after Bpm antigen-specific recall. The PBK001 vaccine is superior in generating respiratory IgA post-boost, anamnestic IgG at challenge, T-cell recall to specific antigen, and development of diverse polyfunctional memory T-cell pools. Analysis of lung histology suggests that potent polyfunctional T-cell memory and/or IL-17 signatures generated with PBK001 vaccination may be associated with moderate lung inflammation post vaccination.
Collapse
Affiliation(s)
- Nittaya Khakhum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Preeti Bharaj
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - David H Walker
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alfredo G Torres
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
13
|
Fatma F, Tripathi DK, Srivastava M, Srivastava KK, Arora A. Immunological characterization of chimeras of high specificity antigens from Mycobacterium tuberculosis H37Rv. Tuberculosis (Edinb) 2021; 127:102054. [PMID: 33550109 DOI: 10.1016/j.tube.2021.102054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
Abstract
Tuberculosis remains a serious global health problem. BCG is the only prophylactic TB vaccine and it shows variable protective efficacy. Chimeric protein subunit vaccines hold great potential as stand-alone vaccines or heterologous BCG prime boosters. We have designed a protein chimera, PP31, by combining Mtb ESAT-6 family antigen Rv1198 and MoCo biosynthesis family antigen Rv3111. Further, PP31 was extended by addition of latency antigen Rv1813c to yield PP43. Immunization of BALB/c mice with PP31 or PP43 with FIA adjuvant elicited strong humoral immune response. Restimulation of splenocytes of the immunized mice lead to significant proliferation of lymphocytes, secretion of cytokines IFN-γ, TNF, IL-2 of the Th1 class, IL-17A of the Th17 class, and IL-6. PP31 and PP43 also induced intracellular cytokine expression (IFN-γ, TNF, and IL-2) from both CD4+-CD44high and CD8+-CD44high T-cells. Antigen-specific IFN-γ+/IL-2+ double positive CD4+ T-cells were significantly higher in case of PP43 than PP31-immunized mice and control group. PP43 showed protection equivalent to heat-inactivated BCG in response to challenge of the immunized mice with Mtb H37Ra. Based on its immunogenicity and protective efficacy, PP43 appears to be a potential candidate for further development as a subunit vaccine against TB.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/immunology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytokines/metabolism
- Epitopes
- Female
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Immunogenicity, Vaccine
- Lymphocyte Activation/drug effects
- Lymphocyte Subsets/drug effects
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Mice, Inbred BALB C
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/immunology
- Tuberculosis/blood
- Tuberculosis/immunology
- Tuberculosis/microbiology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/administration & dosage
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Mice
Collapse
Affiliation(s)
- Farheen Fatma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Dinesh K Tripathi
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Mrigank Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Kishore K Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
| | - Ashish Arora
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
14
|
Morelli MP, Del Medico Zajac MP, Pellegrini JM, Amiano NO, Tateosian NL, Calamante G, Gherardi MM, García VE. IL-12 DNA Displays Efficient Adjuvant Effects Improving Immunogenicity of Ag85A in DNA Prime/MVA Boost Immunizations. Front Cell Infect Microbiol 2020; 10:581812. [PMID: 33072631 PMCID: PMC7538621 DOI: 10.3389/fcimb.2020.581812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/18/2020] [Indexed: 01/26/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) infection is one of the leading causes of death worldwide. The Modified Vaccinia Ankara (MVA) vaccine vector expressing the mycobacterial antigen 85A (MVA85A) was demonstrated to be safe, although it did not improve BCG efficacy, denoting the need to search for improved tuberculosis vaccines. In this work, we investigated the effect of IL-12 DNA -as an adjuvant- on an Ag85A DNA prime/MVA85A boost vaccination regimen. We evaluated the immune response profile elicited in mice and the protection conferred against intratracheal Mtb H37Rv challenge. We observed that the immunization scheme including DNA-A85A+DNA-IL-12/MVA85A induced a strong IFN-γ production to Ag85A in vitro, with a significant expansion of IFN-γ+CD4+ and IFN-γ+CD8+ anti-Ag85A lymphocytes. Furthermore, we also detected a significant increase in the proportion of specific CD8+CD107+ T cells against Ag85A. Additionally, inclusion of IL-12 DNA in the DNA-A85A/MVA85A vaccine scheme induced a marked augment in anti-Ag85A IgG levels. Interestingly, after 30 days of infection with Mtb H37Rv, DNA-A85A+DNA-IL-12/MVA85A vaccinated mice displayed a significant reduction in lung bacterial burden. Together, our findings suggest that IL-12 DNA might be useful as a molecular adjuvant in an Ag85A DNA/MVA prime-boost vaccine against Mtb infection.
Collapse
Affiliation(s)
- María Paula Morelli
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Paula Del Medico Zajac
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Joaquín Miguel Pellegrini
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nicolás Oscar Amiano
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nancy Liliana Tateosian
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela Calamante
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - María Magdalena Gherardi
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Verónica Edith García
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
15
|
Li Z, Zheng C, Terreni M, Tanzi L, Sollogoub M, Zhang Y. Novel Vaccine Candidates against Tuberculosis. Curr Med Chem 2020; 27:5095-5118. [PMID: 30474525 DOI: 10.2174/0929867326666181126112124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Ranking above AIDS, Tuberculosis (TB) is the ninth leading cause of death affecting and
killing many individuals every year. Drugs’ efficacy is limited by a series of problems such as Multi-
Drug Resistance (MDR) and Extensively-Drug Resistance (XDR). Meanwhile, the only licensed vaccine
BCG (Bacillus Calmette-Guérin) existing for over 90 years is not effective enough. Consequently,
it is essential to develop novel vaccines for TB prevention and immunotherapy. This paper
provides an overall review of the TB prevalence, immune system response against TB and recent
progress of TB vaccine research and development. Several vaccines in clinical trials are described as
well as LAM-based candidates.
Collapse
Affiliation(s)
- Zhihao Li
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Changping Zheng
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Marco Terreni
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Lisa Tanzi
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Matthieu Sollogoub
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| | - Yongmin Zhang
- Sorbonne Universite, CNRS, Institut Parisien de Chimie Moleculaire (UMR 8232), 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
16
|
Li J, Zhao A, Tang J, Wang G, Shi Y, Zhan L, Qin C. Tuberculosis vaccine development: from classic to clinical candidates. Eur J Clin Microbiol Infect Dis 2020; 39:1405-1425. [PMID: 32060754 PMCID: PMC7223099 DOI: 10.1007/s10096-020-03843-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Bacillus Calmette-Guérin (BCG) has been in use for nearly 100 years and is the only licensed TB vaccine. While BCG provides protection against disseminated TB in infants, its protection against adult pulmonary tuberculosis (PTB) is variable. To achieve the ambitious goal of eradicating TB worldwide by 2050, there is an urgent need to develop novel TB vaccines. Currently, there are more than a dozen novel TB vaccines including prophylactic and therapeutic at different stages of clinical research. This literature review provides an overview of the clinical status of candidate TB vaccines and discusses the challenges and future development trends of novel TB vaccine research in combination with the efficacy of evaluation of TB vaccines, provides insight for the development of safer and more efficient vaccines, and may inspire new ideas for the prevention of TB.
Collapse
Affiliation(s)
- Junli Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Aihua Zhao
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Jun Tang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Guozhi Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Yanan Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Lingjun Zhan
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| |
Collapse
|
17
|
Imai N, Tawara I, Yamane M, Muraoka D, Shiku H, Ikeda H. CD4 + T cells support polyfunctionality of cytotoxic CD8 + T cells with memory potential in immunological control of tumor. Cancer Sci 2020; 111:1958-1968. [PMID: 32304127 PMCID: PMC7293103 DOI: 10.1111/cas.14420] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022] Open
Abstract
Polyfunctionality/multifunctionality of effector T cells at the single cell level has been shown as an important parameter to predict the quality of T cell response and immunological control of infectious disease and malignancy. However, the fate of polyfunctional CD8+ CTLs and the factors that control the polyfunctionality of T cells remain largely unknown. Here we show that the acquisition of polyfunctionality on the initial stimulation is a sensitive immune correlate of CTL survival and memory formation. CD8+ T cells with high polyfunctionality, assessed with γ‐interferon and tumor necrosis factor‐α production and surface mobilization of the degranulation marker CD107a, showed enhanced Bcl‐2 expression, low apoptosis, and increased CD127highKLRG1low memory precursor phenotype. Consistent with these observations, CD8+ T cells were found to acquire high frequency of cells with polyfunctionality when stimulated in conditions known to enhance memory formation, such as the presence of CD4+ T cells, interleukin (IL)‐2, or IL‐21. Utilizing T‐cell receptor (TCR) transgenic mouse‐derived CD8+ T cells that express a TCR specific for a tumor‐derived neoantigen, we showed that polyfunctional tumor‐specific CTLs generated in the presence of CD4+ T cells showed long persistence in vivo and induced enhanced tumor regression when adoptively transferred into mice with progressing tumor. Acquisition of polyfunctionality thus impacts CTL survival and memory formation associated with immunological control of tumor.
Collapse
Affiliation(s)
- Naoko Imai
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Makiko Yamane
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Daisuke Muraoka
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroaki Ikeda
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
18
|
Cell-Mediated Immunity Generated in Response to a Purified Inactivated Vaccine for Dengue Virus Type 1. mSphere 2020; 5:5/1/e00671-19. [PMID: 31969476 PMCID: PMC6977178 DOI: 10.1128/msphere.00671-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Dengue is a tropical disease transmitted by mosquitoes, and nearly half of the world’s population lives in areas where individuals are at risk of infection. Several vaccines for dengue are in development, including one which was recently licensed in several countries, although its utility is limited to people who have already been infected with one of the four dengue viruses. One major hurdle to understanding whether a dengue vaccine will work for everyone—before exposure—is the necessity of knowing which marker can be measured in the blood to signal that the individual has protective immunity. This report describes an approach measuring multiple different parts of immunity in order to characterize which signals one candidate vaccine imparted to a small number of human volunteers. This approach was designed to be able to be applied to any dengue vaccine study so that the data can be compared and used to inform future vaccine design and/or optimization strategies. Dengue is the most prevalent arboviral disease afflicting humans, and a vaccine appears to be the most rational means of control. Dengue vaccine development is in a critical phase, with the first vaccine licensed in some countries where dengue is endemic but demonstrating insufficient efficacy in immunologically naive populations. Since virus-neutralizing antibodies do not invariably correlate with vaccine efficacy, other markers that may predict protection, including cell-mediated immunity, are urgently needed. Previously, the Walter Reed Army Institute of Research developed a monovalent purified inactivated virus (PIV) vaccine candidate against dengue virus serotype 1 (DENV-1) adjuvanted with alum. The PIV vaccine was safe and immunogenic in a phase I dose escalation trial in healthy, flavivirus-naive adults in the United States. From that trial, peripheral blood mononuclear cells obtained at various time points pre- and postvaccination were used to measure DENV-1-specific T cell responses. After vaccination, a predominant CD4+ T cell-mediated response to peptide pools covering the DENV-1 structural proteins was observed. Over half (13/20) of the subjects produced interleukin-2 (IL-2) in response to DENV peptides, and the majority (17/20) demonstrated peptide-specific CD4+ T cell proliferation. In addition, analysis of postvaccination cell culture supernatants demonstrated an increased rate of production of cytokines, including gamma interferon (IFN-γ), IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Overall, the vaccine was found to have elicited DENV-specific CD4+ T cell responses as measured by enzyme-linked immunosorbent spot (ELISpot), intracellular cytokine staining (ICS), lymphocyte proliferation, and cytokine production assays. Thus, together with antibody readouts, the use of a multifaceted measurement of cell-mediated immune responses after vaccination is a useful strategy for more comprehensively characterizing immunity generated by dengue vaccines. IMPORTANCE Dengue is a tropical disease transmitted by mosquitoes, and nearly half of the world’s population lives in areas where individuals are at risk of infection. Several vaccines for dengue are in development, including one which was recently licensed in several countries, although its utility is limited to people who have already been infected with one of the four dengue viruses. One major hurdle to understanding whether a dengue vaccine will work for everyone—before exposure—is the necessity of knowing which marker can be measured in the blood to signal that the individual has protective immunity. This report describes an approach measuring multiple different parts of immunity in order to characterize which signals one candidate vaccine imparted to a small number of human volunteers. This approach was designed to be able to be applied to any dengue vaccine study so that the data can be compared and used to inform future vaccine design and/or optimization strategies.
Collapse
|
19
|
Wilkie M, Satti I, Minhinnick A, Harris S, Riste M, Ramon RL, Sheehan S, Thomas ZRM, Wright D, Stockdale L, Hamidi A, O'Shea MK, Dwivedi K, Behrens HM, Davenne T, Morton J, Vermaak S, Lawrie A, Moss P, McShane H. A phase I trial evaluating the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime - MVA85A boost in healthy UK adults. Vaccine 2019; 38:779-789. [PMID: 31735500 PMCID: PMC6985898 DOI: 10.1016/j.vaccine.2019.10.102] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 11/18/2022]
Abstract
Background This phase I trial evaluated the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime-MVA85A boost, previously demonstrated to be protective in animal studies, in healthy UK adults. Methods We enrolled 42 healthy, BCG-vaccinated adults into 4 groups: low dose Starter Group (n = 6; ChAdOx1 85A alone), high dose groups; Group A (n = 12; ChAdOx1 85A), Group B (n = 12; ChAdOx1 85A prime – MVA85A boost) or Group C (n = 12; ChAdOx1 85A – ChAdOx1 85A prime – MVA85A boost). Safety was determined by collection of solicited and unsolicited vaccine-related adverse events (AEs). Immunogenicity was measured by antigen-specific ex-vivo IFN-γ ELISpot, IgG serum ELISA, and antigen-specific intracellular IFN-γ, TNF-α, IL-2 and IL-17. Results AEs were mostly mild/moderate, with no Serious Adverse Events. ChAdOx1 85A induced Ag85A-specific ELISpot and intracellular cytokine CD4+ and CD8+ T cell responses, which were not boosted by a second dose, but were boosted with MVA85A. Polyfunctional CD4+ T cells (IFN-γ, TNF-α and IL-2) and IFN-γ+, TNF-α+ CD8+ T cells were induced by ChAdOx1 85A and boosted by MVA85A. ChAdOx1 85A induced serum Ag85A IgG responses which were boosted by MVA85A. Conclusion A ChAdOx1 85A prime – MVA85A boost is well tolerated and immunogenic in healthy UK adults.
Collapse
Affiliation(s)
- Morven Wilkie
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Michael Riste
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Sharon Sheehan
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Daniel Wright
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Lisa Stockdale
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ali Hamidi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Kritica Dwivedi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Tamara Davenne
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Joshua Morton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Alison Lawrie
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
20
|
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
|
21
|
Kashangura R, Jullien S, Garner P, Johnson S, Cochrane Infectious Diseases Group. MVA85A vaccine to enhance BCG for preventing tuberculosis. Cochrane Database Syst Rev 2019; 4:CD012915. [PMID: 31038197 PMCID: PMC6488980 DOI: 10.1002/14651858.cd012915.pub2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Tuberculosis causes more deaths than any other infectious disease globally. Bacillus Calmette-Guérin (BCG) is the only available vaccine, but protection is incomplete and variable. The modified Vaccinia Ankara virus expressing antigen 85A (MVA85A) is a viral vector vaccine produced to prevent tuberculosis. OBJECTIVES To assess and summarize the effects of the MVA85A vaccine boosting BCG in humans. SEARCH METHODS We searched the Cochrane Infectious Diseases Group Specialized Register; Central Register of Controlled Trials (CENTRAL); MEDLINE (PubMed); Embase (Ovid); and four other databases. We searched the WHO ICTRP and ClinicalTrials.gov. All searches were run up to 10 May 2018. SELECTION CRITERIA We evaluated randomized controlled trials of MVA85A vaccine given with BCG in people regardless of age or HIV status. DATA COLLECTION AND ANALYSIS Two review authors independently assessed the eligibility and risk of bias of trials, and extracted and analyzed data. The primary outcome was active tuberculosis disease. We summarized dichotomous outcomes using risk ratios (RR) and risk differences (RD), with 95% confidence intervals (CI). Where appropriate, we combined data in meta-analyses. Where meta-analysis was inappropriate, we summarized results narratively. MAIN RESULTS The search identified six studies relating to four Phase 2 randomized controlled trials enrolling 3838 participants. Funding was by government bodies, charities, and philanthropic donors. Five studies included infants, one of them infants born to HIV-positive mothers. One study included adults living with HIV. All trials included authors from Oxford University who led the laboratory development of the vaccine. Participants received intradermal MVA85A after BCG in some studies, and before selective deferred BCG in HIV-exposed infants.The largest trial in 2797 African children was well conducted with low risk of bias for most parameters. Risk of bias was uncertain for selective reporting because there were no precise case definition endpoints for active tuberculosis published prior to the trial analysis.MVA85A added to BCG compared to BCG alone probably has no effect on the risk of developing microbiologically confirmed tuberculosis (RR 0.97, 95% CI 0.58 to 1.62; 3439 participants, 2 trials; moderate-certainty evidence), or the risk of starting on tuberculosis treatment (RR 1.10, 95% CI 0.92 to 1.33; 3687 participants, 3 trials; moderate-certainty evidence). MVA85A probably has no effect on the risk of developing latent tuberculosis (RR 1.01, 95% CI 0.85 to 1.21; 3831 participants, 4 trials; moderate-certainty evidence). Vaccinating people with MVA85A in addition to BCG did not cause life-threatening serious adverse effects (RD 0.00, 95% CI -0.00 to 0.00; 3692 participants, 3 trials; high-certainty evidence). Vaccination with MVA85A is probably associated with an increased risk of local skin adverse effects (3187 participants, 3 trials; moderate-certainty evidence), but not systemic adverse effect related to vaccination (144 participants, 1 trial; low-certainty evidence). This safety profile is consistent with Phase 1 studies which outlined a transient, superficial reaction local to the injection site and mild short-lived symptoms such as malaise and fever. AUTHORS' CONCLUSIONS MVA85A delivered by intradermal injection in addition to BCG is safe but not effective in reducing the risk of developing tuberculosis.
Collapse
Affiliation(s)
| | - Sophie Jullien
- Jigme Dorji Wangchuck National Referral HospitalThimphuBhutan
| | - Paul Garner
- Liverpool School of Tropical MedicineDepartment of Clinical SciencesPembroke PlaceLiverpoolMerseysideUKL3 5QA
| | - Samuel Johnson
- Liverpool School of Tropical MedicineDepartment of Clinical SciencesPembroke PlaceLiverpoolMerseysideUKL3 5QA
| | | |
Collapse
|
22
|
Cicchese JM, Evans S, Hult C, Joslyn LR, Wessler T, Millar JA, Marino S, Cilfone NA, Mattila JT, Linderman JJ, Kirschner DE. Dynamic balance of pro- and anti-inflammatory signals controls disease and limits pathology. Immunol Rev 2018; 285:147-167. [PMID: 30129209 PMCID: PMC6292442 DOI: 10.1111/imr.12671] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune responses to pathogens are complex and not well understood in many diseases, and this is especially true for infections by persistent pathogens. One mechanism that allows for long-term control of infection while also preventing an over-zealous inflammatory response from causing extensive tissue damage is for the immune system to balance pro- and anti-inflammatory cells and signals. This balance is dynamic and the immune system responds to cues from both host and pathogen, maintaining a steady state across multiple scales through continuous feedback. Identifying the signals, cells, cytokines, and other immune response factors that mediate this balance over time has been difficult using traditional research strategies. Computational modeling studies based on data from traditional systems can identify how this balance contributes to immunity. Here we provide evidence from both experimental and mathematical/computational studies to support the concept of a dynamic balance operating during persistent and other infection scenarios. We focus mainly on tuberculosis, currently the leading cause of death due to infectious disease in the world, and also provide evidence for other infections. A better understanding of the dynamically balanced immune response can help shape treatment strategies that utilize both drugs and host-directed therapies.
Collapse
Affiliation(s)
- Joseph M. Cicchese
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Evans
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Caitlin Hult
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Louis R. Joslyn
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Timothy Wessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jess A. Millar
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Simeone Marino
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicholas A. Cilfone
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joshua T. Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Identification of Mycobacterial Ribosomal Proteins as Targets for CD4 + T Cells That Enhance Protective Immunity in Tuberculosis. Infect Immun 2018; 86:IAI.00009-18. [PMID: 29891545 DOI: 10.1128/iai.00009-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/08/2018] [Indexed: 12/25/2022] Open
Abstract
Mycobacterium tuberculosis remains a threat to global health, and a more efficacious vaccine is needed to prevent disease caused by M. tuberculosis We previously reported that the mycobacterial ribosome is a major target of CD4+ T cells in mice immunized with a genetically modified Mycobacterium smegmatis strain (IKEPLUS) but not in mice immunized with Mycobacterium bovis BCG. Two specific ribosomal proteins, RplJ and RpsA, were identified as cross-reactive targets of M. tuberculosis, but the breadth of the CD4+ T cell response to M. tuberculosis ribosomes was not determined. In the present study, a library of M. tuberculosis ribosomal proteins and in silico-predicted peptide libraries were used to screen CD4+ T cell responses in IKEPLUS-immunized mice. This identified 24 out of 57 M. tuberculosis ribosomal proteins distributed over both large and small ribosome subunits as specific CD4+ T cell targets. Although BCG did not induce detectable responses against ribosomal proteins or peptide epitopes, the M. tuberculosis ribosomal protein RplJ produced a robust and multifunctional Th1-like CD4+ T cell population when administered as a booster vaccine to previously BCG-primed mice. Boosting of BCG-primed immunity with the M. tuberculosis RplJ protein led to significantly reduced lung pathology compared to that in BCG-immunized animals and reductions in the bacterial burdens in the mediastinal lymph node compared to those in naive and standard BCG-vaccinated mice. These results identify the mycobacterial ribosome as a potential source of cryptic or subdominant antigenic targets of protective CD4+ T cell responses and suggest that supplementing BCG with ribosomal antigens may enhance protective vaccination against M. tuberculosis.
Collapse
|
24
|
Arfa N, Allami A. The effect of fish oil on two-step tuberculin test in hospitalized patients: A randomized controlled trial. THE JOURNAL OF QAZVIN UNIVERSITY OF MEDICAL SCIENCES 2018. [DOI: 10.29252/qums.21.6.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
25
|
Kashangura R, Jullien S, Garner P, Young T, Johnson S. MVA85A vaccine to enhance BCG for preventing tuberculosis. Hippokratia 2018. [DOI: 10.1002/14651858.cd012915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
| | - Sophie Jullien
- Jigme Dorji Wangchuck National Referral Hospital; Thimphu Bhutan
| | - Paul Garner
- Liverpool School of Tropical Medicine; Department of Clinical Sciences; Pembroke Place Liverpool Merseyside UK L3 5QA
| | - Taryn Young
- Stellenbosch University; Centre for Evidence-based Health Care, Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences; PO Box 241 Cape Town South Africa 8000
- South African Medical Research Council; Cochrane South Africa; PO Box 19070 Tygerberg Cape Town South Africa 7505
| | - Samuel Johnson
- Liverpool School of Tropical Medicine; Department of Clinical Sciences; Pembroke Place Liverpool Merseyside UK L3 5QA
| |
Collapse
|
26
|
Bizzell E, Sia JK, Quezada M, Enriquez A, Georgieva M, Rengarajan J. Deletion of BCG Hip1 protease enhances dendritic cell and CD4 T cell responses. J Leukoc Biol 2017; 103:739-748. [PMID: 29345365 DOI: 10.1002/jlb.4a0917-363rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/29/2017] [Accepted: 12/02/2017] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) play a key role in the generation of CD4 T cell responses to pathogens. Mycobacterium tuberculosis (Mtb) harbors immune evasion mechanisms that impair DC responses and prevent optimal CD4 T cell immunity. The vaccine strain Mycobacterium bovis Bacille Calmette-Guérin (BCG) shares many of the immune evasion proteins utilized by Mtb, but the role of these proteins in DC and T cell responses elicited by BCG is poorly understood. We previously reported that the Mtb serine protease, Hip1, promotes sub-optimal DC responses during infection. Here, we tested the hypothesis that BCG Hip1 modulates DC functions and prevents optimal antigen-specific CD4 T cell responses that limit the immunogenicity of BCG. We generated a strain of BCG lacking hip1 (BCGΔhip1) and show that it has superior capacity to induce DC maturation and cytokine production compared with the parental BCG. Furthermore, BCGΔhip1-infected DCs were more effective at driving the production of IFN-γ and IL-17 from antigen-specific CD4 T cells in vitro. Mucosal transfer of BCGΔhip1-infected DCs into mouse lungs induced robust CD4 T cell activation in vivo and generated antigen-specific polyfunctional CD4 T cell responses in the lungs. Importantly, BCGΔhip1-infected DCs enhanced control of pulmonary bacterial burden following Mtb aerosol challenge compared with the transfer of BCG-infected DCs. These results reveal that BCG employs Hip1 to impair DC activation, leading to attenuated lung CD4 T cell responses with limited capacity to control Mtb burden after challenge.
Collapse
Affiliation(s)
- Erica Bizzell
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | | | - Melanie Quezada
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | - Ana Enriquez
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | - Maria Georgieva
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.,Current affiliation: Maria Georgieva, Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
27
|
Schussek S, Trieu A, Apte SH, Sidney J, Sette A, Doolan DL. Novel Plasmodium antigens identified via genome-based antibody screen induce protection associated with polyfunctional T cell responses. Sci Rep 2017; 7:15053. [PMID: 29118376 PMCID: PMC5678182 DOI: 10.1038/s41598-017-15354-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
The development of vaccines against complex intracellular pathogens, such as Plasmodium spp., where protection is likely mediated by cellular immune responses, has proven elusive. The availability of whole genome, proteome and transcriptome data has the potential to advance rational vaccine development but yet there are no licensed vaccines against malaria based on antigens identified from genomic data. Here, we show that the Plasmodium yoelii orthologs of four Plasmodium falciparum proteins identified by an antibody-based genome-wide screening strategy induce a high degree of sterile infection-blocking protection against sporozoite challenge in a stringent rodent malaria model. Protection increased in multi-antigen formulations. Importantly, protection was highly correlated with the induction of multifunctional triple-positive T cells expressing high amounts of IFN-γ, IL-2 and TNF. These data demonstrate that antigens identified by serological screening are targets of multifunctional cellular immune responses that correlate with protection. Our results provide experimental validation for the concept of rational vaccine design from genomic sequence data.
Collapse
Affiliation(s)
- Sophie Schussek
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia.,University of Queensland, School of Medicine, Herston, QLD 4006, Australia
| | - Angela Trieu
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia
| | - Simon H Apte
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia
| | - John Sidney
- La Jolla Institute of Allergy and Immunology, San Diego, CA, 92121, USA
| | - Alessandro Sette
- La Jolla Institute of Allergy and Immunology, San Diego, CA, 92121, USA
| | - Denise L Doolan
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia. .,Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4879, Australia.
| |
Collapse
|
28
|
Lewinsohn DA, Lewinsohn DM, Scriba TJ. Polyfunctional CD4 + T Cells As Targets for Tuberculosis Vaccination. Front Immunol 2017; 8:1262. [PMID: 29051764 PMCID: PMC5633696 DOI: 10.3389/fimmu.2017.01262] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/21/2017] [Indexed: 01/14/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a leading cause of morbidity and mortality worldwide, despite the widespread use of the only licensed vaccine, Bacille Calmette Guerin (BCG). Eradication of TB will require a more effective vaccine, yet evaluation of new vaccine candidates is hampered by lack of defined correlates of protection. Animal and human studies of intracellular pathogens have extensively evaluated polyfunctional CD4+ T cells producing multiple pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-2) as a possible correlate of protection from infection and disease. In this study, we review the published literature that evaluates whether or not BCG and/or novel TB vaccine candidates induce polyfunctional CD4+ T cells and if these T cell responses correlate with vaccine-mediated protection. Ample evidence suggests that BCG and several novel vaccine candidates evaluated in animal models and humans induce polyfunctional CD4+ T cells. However, while a number of studies utilizing the mouse TB model support that polyfunctional CD4+ T cells are associated with vaccine-induced protection, other studies in mouse and human infants demonstrate no correlation between these T cell responses and protection. We conclude that induction of polyfunctional CD4+ T cells is certainly not sufficient and may not even be necessary to mediate protection and suggest that other functional attributes, such as additional effector functions, T cell differentiation state, tissue homing potential, or long-term survival capacity of the T cell may be equally or more important to promote protection. Thus, a correlate of protection for TB vaccine development remains elusive. Future studies should address polyfunctional CD4+ T cells within the context of more comprehensive immunological signatures of protection that include other functions and phenotypes of T cells as well as the full spectrum of immune cells and mediators that participate in the immune response against Mtb.
Collapse
Affiliation(s)
- Deborah A Lewinsohn
- Division of Infectious Disease, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - David M Lewinsohn
- Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, OR, United States.,Department of Medicine, VA Portland Health Care System, Portland, OR, United States
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM) and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
29
|
Wajja A, Kizito D, Nassanga B, Nalwoga A, Kabagenyi J, Kimuda S, Galiwango R, Mutonyi G, Vermaak S, Satti I, Verweij J, Tukahebwa E, Cose S, Levin J, Kaleebu P, Elliott AM, McShane H. The effect of current Schistosoma mansoni infection on the immunogenicity of a candidate TB vaccine, MVA85A, in BCG-vaccinated adolescents: An open-label trial. PLoS Negl Trop Dis 2017; 11:e0005440. [PMID: 28472067 PMCID: PMC5417418 DOI: 10.1371/journal.pntd.0005440] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/27/2017] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Helminth infection may affect vaccine immunogenicity and efficacy. Adolescents, a target population for tuberculosis booster vaccines, often have a high helminth burden. We investigated effects of Schistosoma mansoni (Sm) on the immunogenicity and safety of MVA85A, a model candidate tuberculosis vaccine, in BCG-vaccinated Ugandan adolescents. METHODS In this phase II open label trial we enrolled 36 healthy, previously BCG-vaccinated adolescents, 18 with no helminth infection detected, 18 with Sm only. The primary outcome was immunogenicity measured by Ag85A-specific interferon gamma ELISpot assay. Tuberculosis and schistosome-specific responses were also assessed by whole-blood stimulation and multiplex cytokine assay, and by antibody ELISAs. RESULTS Ag85A-specific cellular responses increased significantly following immunisation but with no differences between the two groups. Sm infection was associated with higher pre-immunisation Ag85A-specific IgG4 but with no change in antibody levels following immunisation. There were no serious adverse events. Most reactogenicity events were of mild or moderate severity and resolved quickly. CONCLUSIONS The significant Ag85A-specific T cell responses and lack of difference between Sm-infected and uninfected participants is encouraging for tuberculosis vaccine development. The implications of pre-existing Ag85A-specific IgG4 antibodies for protective immunity against tuberculosis among those infected with Sm are not known. MVA85A was safe in this population. TRIAL REGISTRATION ClinicalTrials.gov NCT02178748.
Collapse
Affiliation(s)
- Anne Wajja
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Dennison Kizito
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Beatrice Nassanga
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Angela Nalwoga
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Joyce Kabagenyi
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Simon Kimuda
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Ronald Galiwango
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Gertrude Mutonyi
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Samantha Vermaak
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jaco Verweij
- Laboratory for Medical Microbiology and Immunology & Laboratory for Clinical Pathology, St. Elisabeth Hospital, Tilburg, The Netherlands
| | | | - Stephen Cose
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jonathan Levin
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Pontiano Kaleebu
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Alison M. Elliott
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Identification of Mycobacterial RplJ/L10 and RpsA/S1 Proteins as Novel Targets for CD4 + T Cells. Infect Immun 2017; 85:IAI.01023-16. [PMID: 28115505 PMCID: PMC5364311 DOI: 10.1128/iai.01023-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Tuberculosis (TB) due to Mycobacterium tuberculosis remains a major global infectious disease problem, and a more efficacious vaccine is urgently needed for the control and prevention of disease caused by this organism. We previously reported that a genetically modified strain of Mycobacterium smegmatis called IKEPLUS is a promising TB vaccine candidate. Since protective immunity induced by IKEPLUS is dependent on antigen-specific CD4+ T cell memory, we hypothesized that the specificity of the CD4+ T cell response was a critical feature of this protection. Using in vitro assays of interferon gamma production (enzyme-linked immunosorbent spot [ELISPOT] assays) by splenocytes from IKEPLUS-immunized C57BL/6J mice, we identified an immunogenic peptide within the mycobacterial ribosomal large subunit protein RplJ, encoded by the Rv0651 gene. In a complementary approach, we generated major histocompatibility complex (MHC) class II-restricted T cell hybridomas from IKEPLUS-immunized mice. Screening of these T cell hybridomas against IKEPLUS and ribosomes enriched from IKEPLUS suggested that the CD4+ T cell response in IKEPLUS-immunized mice was dominated by the recognition of multiple components of the mycobacterial ribosome. Importantly, CD4+ T cells specific for mycobacterial ribosomes accumulate to significant levels in the lungs of IKEPLUS-immunized mice following aerosol challenge with virulent M. tuberculosis, consistent with a role for these T cells in protective host immunity in TB. The identification of CD4+ T cell responses to defined ribosomal protein epitopes expands the range of antigenic targets for adaptive immune responses to M. tuberculosis and may help to inform the design of more effective vaccines against tuberculosis.
Collapse
|
31
|
Adenovirally-Induced Polyfunctional T Cells Do Not Necessarily Recognize the Infected Target: Lessons from a Phase I Trial of the AERAS-402 Vaccine. Sci Rep 2016; 6:36355. [PMID: 27805026 PMCID: PMC5141283 DOI: 10.1038/srep36355] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 10/13/2016] [Indexed: 11/08/2022] Open
Abstract
The development of a vaccine for Mycobacterium tuberculosis (Mtb) has been impeded by the absence of correlates of protective immunity. One correlate would be the ability of cells induced by vaccination to recognize the Mtb-infected cell. AERAS-402 is a replication-deficient serotype 35 adenovirus containing DNA expressing a fusion protein of Mtb antigens 85A, 85B and TB10.4. We undertook a phase I double-blind, randomized placebo controlled trial of vaccination with AERAS-402 following BCG. Analysis of the vaccine-induced immune response revealed strong antigen-specific polyfunctional CD4+ and CD8+ T cell responses. However, analysis of the vaccine-induced CD8+ T cells revealed that in many instances these cells did not recognize the Mtb-infected cell. Our findings highlight the measurement of vaccine-induced, polyfunctional T cells may not reflect the extent or degree to which these cells are capable of identifying the Mtb-infected cell and correspondingly, the value of detailed experimental medicine studies early in vaccine development.
Collapse
|
32
|
Mycobacterium indicus pranii as a booster vaccine enhances BCG induced immunity and confers higher protection in animal models of tuberculosis. Tuberculosis (Edinb) 2016; 101:164-173. [PMID: 27865389 DOI: 10.1016/j.tube.2016.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 11/20/2022]
Abstract
BCG, the only approved vaccine protects against severe form of childhood tuberculosis but its protective efficacy wanes in adolescence. BCG has reduced the incidence of infant TB considerably in endemic areas; therefore prime-boost strategy is the most realistic measure for control of tuberculosis in near future. Mycobacterium indicus pranii (MIP) shares significant antigenic repertoire with Mtb and BCG and has been shown to impart significant protection in animal models of tuberculosis. In this study, MIP was given as a booster to BCG vaccine which enhanced the BCG mediated immune response, resulting in higher protection. MIP booster via aerosol route was found to be more effective in protection than subcutaneous route of booster immunization. Pro-inflammatory cytokines like IFN-γ, IL-12 and IL-17 were induced at higher level in infected lungs of 'BCG-MIP' group both at mRNA expression level and in secretory form when compared with 'only BCG' group. BCG-MIP groups had increased frequency of multifunctional T cells with high MFI for IFN-γ and TNF-α in Mtb infected mice. Our data demonstrate for the first time, potential application of MIP as a booster to BCG vaccine for efficient protection against tuberculosis. This could be very cost effective strategy for efficient control of tuberculosis.
Collapse
|
33
|
Carrère-Kremer S, Rubbo PA, Pisoni A, Bendriss S, Marin G, Peries M, Bolloré K, Terru D, Godreuil S, Bourdin A, Van de Perre P, Tuaillon E. High IFN-γ Release and Impaired Capacity of Multi-Cytokine Secretion in IGRA Supernatants Are Associated with Active Tuberculosis. PLoS One 2016; 11:e0162137. [PMID: 27603919 PMCID: PMC5014470 DOI: 10.1371/journal.pone.0162137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022] Open
Abstract
Interferon gamma (IFN-γ) release assays (IGRAs) detect Mycobacterium tuberculosis (Mtb) infection regardless of the active (ATB) or latent (LTBI) forms of tuberculosis (TB). In this study, Mtb-specific T cell response against region of deletion 1 (RD1) antigens were explored by a microbead multiplex assay performed in T-SPOT TB assay (T-SPOT) supernatants from 35 patients with ATB and 115 patients with LTBI. T-SPOT is positive when over 7 IFN-γ secreting cells (SC)/250 000 peripheral blood mononuclear cells (PBMC) are enumerated. However, over 100 IFN-γ SC /250 000 PBMC were more frequently observed in the ATB group compared to the LTBI group. By contrast, lower cytokine concentrations and lower cytokine productions relative to IFN-γ secretion were observed for IL 4, IL-12, TNF-α, GM-CSF, Eotaxin and IFN-α when compared to LTBI. Thus, high IFN-γ release and low cytokine secretions in relation with IFN-γ production appeared as signatures of ATB, corroborating that multicytokine Mtb-specific response against RD1 antigens reflects host capacity to contain TB reactivation. In this way, testing cytokine profile in IGRA supernatants would be helpful to improve ATB screening strategy including immunologic tests.
Collapse
Affiliation(s)
- Séverine Carrère-Kremer
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France.,University Montpellier, Montpellier, France
| | - Pierre-Alain Rubbo
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France.,University Montpellier, Montpellier, France
| | - Amandine Pisoni
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France.,CHRU Montpellier, Departments of Bacteriology-Virology, Montpellier, France
| | - Sophie Bendriss
- CHRU Montpellier, Departments of Bacteriology-Virology, Montpellier, France
| | - Grégory Marin
- CHRU Montpellier, Department of Medical Information, Montpellier, France
| | - Marianne Peries
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France
| | - Karine Bolloré
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France.,University Montpellier, Montpellier, France
| | - Dominique Terru
- CHRU Montpellier, Departments of Bacteriology-Virology, Montpellier, France
| | - Sylvain Godreuil
- CHRU Montpellier, Departments of Bacteriology-Virology, Montpellier, France
| | - Arnaud Bourdin
- CHRU Montpellier, Department of Pneumology, Montpellier, France
| | - Philippe Van de Perre
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France.,University Montpellier, Montpellier, France.,CHRU Montpellier, Departments of Bacteriology-Virology, Montpellier, France
| | - Edouard Tuaillon
- UMR1058 INSERM/University Montpellier/EFS, Montpellier, France.,University Montpellier, Montpellier, France.,CHRU Montpellier, Departments of Bacteriology-Virology, Montpellier, France
| |
Collapse
|
34
|
Roberts LM, Crane DD, Wehrly TD, Fletcher JR, Jones BD, Bosio CM. Inclusion of Epitopes That Expand High-Avidity CD4+ T Cells Transforms Subprotective Vaccines to Efficacious Immunogens against Virulent Francisella tularensis. THE JOURNAL OF IMMUNOLOGY 2016; 197:2738-47. [PMID: 27543611 DOI: 10.4049/jimmunol.1600879] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/21/2016] [Indexed: 12/24/2022]
Abstract
T cells are the immunological cornerstone in host defense against infections by intracellular bacterial pathogens, such as virulent Francisella tularensis spp. tularensis (Ftt). The general paucity of novel vaccines for Ftt during the past 60 y can, in part, be attributed to the poor understanding of immune parameters required to survive infection. Thus, we developed a strategy utilizing classical immunological tools to elucidate requirements for effective adaptive immune responses directed against Ftt. Following generation of various Francisella strains expressing well-characterized lymphocytic choriomeningitis virus epitopes, we found that survival correlated with persistence of Ag-specific CD4(+) T cells. Function of these cells was confirmed in their ability to more effectively control Ftt replication in vitro. The importance of understanding the Ag-specific response was underscored by our observation that inclusion of an epitope that elicits high-avidity CD4(+) T cells converted a poorly protective vaccine to one that engenders 100% protection. Taken together, these data suggest that improved efficacy of current tularemia vaccine platforms will require targeting appropriate Ag-specific CD4(+) T cell responses and that elucidation of Francisella epitopes that elicit high-avidity CD4(+) T cell responses, specifically in humans, will be required for successful vaccine development.
Collapse
Affiliation(s)
- Lydia M Roberts
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| | - Deborah D Crane
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| | - Tara D Wehrly
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| | - Joshua R Fletcher
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Bradley D Jones
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and
| |
Collapse
|
35
|
A Subset of Protective γ9δ2 T Cells Is Activated by Novel Mycobacterial Glycolipid Components. Infect Immun 2016; 84:2449-62. [PMID: 27297390 PMCID: PMC4995917 DOI: 10.1128/iai.01322-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 06/07/2016] [Indexed: 01/28/2023] Open
Abstract
γ9δ2 T cells provide a natural bridge between innate and adaptive immunity, rapidly and potently respond to pathogen infection in mucosal tissues, and are prominently induced by both tuberculosis (TB) infection and bacillus Calmette Guérin (BCG) vaccination. Mycobacterium-expanded γ9δ2 T cells represent only a subset of the phosphoantigen {isopentenyl pyrophosphate [IPP] and (E)-4-hydroxy-3-methyl-but-2-enylpyrophosphate [HMBPP]}-responsive γ9δ2 T cells, expressing an oligoclonal set of T cell receptor (TCR) sequences which more efficiently recognize and inhibit intracellular Mycobacterium tuberculosis infection. Based on this premise, we have been searching for M. tuberculosis antigens specifically capable of inducing a unique subset of mycobacterium-protective γ9δ2 T cells. Our screening strategy includes the identification of M. tuberculosis fractions that expand γ9δ2 T cells with biological functions capable of inhibiting intracellular mycobacterial replication. Chemical treatments of M. tuberculosis whole-cell lysates (MtbWL) ruled out protein, nucleic acid, and nonpolar lipids as the M. tuberculosis antigens inducing protective γ9δ2 T cells. Mild acid hydrolysis, which transforms complex carbohydrate to monomeric residues, abrogated the specific activity of M. tuberculosis whole-cell lysates, suggesting that a polysaccharide was required for biological activity. Extraction of MtbWL with chloroform-methanol-water (10:10:3) resulted in a polar lipid fraction with highly enriched specific activity; this activity was further enriched by silica gel chromatography. A combination of mass spectrometry and nuclear magnetic resonance analysis of bioactive fractions indicated that 6-O-methylglucose-containing lipopolysaccharides (mGLP) are predominant components present in this active fraction. These results have important implications for the development of new immunotherapeutic approaches for prevention and treatment of TB.
Collapse
|
36
|
A novel dendritic cell-targeted lentiviral vector, encoding Ag85A-ESAT6 fusion gene of Mycobacterium tuberculosis, could elicit potent cell-mediated immune responses in mice. Mol Immunol 2016; 75:101-11. [DOI: 10.1016/j.molimm.2016.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/08/2016] [Accepted: 04/28/2016] [Indexed: 01/13/2023]
|
37
|
Graves AJ, Hokey DA. Tuberculosis vaccine development: Shifting focus amid increasing development challenges. Hum Vaccin Immunother 2016; 11:1910-6. [PMID: 26125249 PMCID: PMC4635864 DOI: 10.1080/21645515.2015.1040955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A new tuberculosis vaccine is needed to replace or enhance BCG, which induces variable protection against Mycobacterium tuberculosis pulmonary infections in adults. Development of new TB vaccine candidates is severely hampered by the lack of a correlate of immunity, unproven animal models, and limited funding opportunities. One candidate, MVA85A, recently failed to meet its efficacy endpoint goals despite promising early-phase trial data. As a result, some in the field believe we should now shift our focus away from product development and toward a research-oriented approach. Here, we outline our suggestions for this research-oriented strategy including diversification of the candidate pipeline, expanding measurements of immunity, improving pre-clinical animal models, and investing in combination pre-clinical/experimental medicine studies. As with any evolution, this change in strategy comes at a cost but may also represent an opportunity for advancing the field.
Collapse
|
38
|
Satchidanandam V, Kumar N, Biswas S, Jumani RS, Jain C, Rani R, Aggarwal B, Singh J, Kotnur MR, Sridharan A. The Secreted Protein Rv1860 of Mycobacterium tuberculosis Stimulates Human Polyfunctional CD8+ T Cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:282-93. [PMID: 26843486 PMCID: PMC4820513 DOI: 10.1128/cvi.00554-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/22/2016] [Indexed: 12/21/2022]
Abstract
We previously reported that Rv1860 protein from Mycobacterium tuberculosis stimulated CD4(+)and CD8(+)T cells secreting gamma interferon (IFN-γ) in healthy purified protein derivative (PPD)-positive individuals and protected guinea pigs immunized with a DNA vaccine and a recombinant poxvirus expressing Rv1860 from a challenge with virulent M. tuberculosis We now show Rv1860-specific polyfunctional T (PFT) cell responses in the blood of healthy latently M. tuberculosis-infected individuals dominated by CD8(+) T cells, using a panel of 32 overlapping peptides spanning the length of Rv1860. Multiple subsets of CD8(+) PFT cells were significantly more numerous in healthy latently infected volunteers (HV) than in tuberculosis (TB) patients (PAT). The responses of peripheral blood mononuclear cells (PBMC) from PAT to the peptides of Rv1860 were dominated by tumor necrosis factor alpha (TNF-α) and interleukin-10 (IL-10) secretions, the former coming predominantly from non-T cell sources. Notably, the pattern of the T cell response to Rv1860 was distinctly different from those of the widely studied M. tuberculosis antigens ESAT-6, CFP-10, Ag85A, and Ag85B, which elicited CD4(+) T cell-dominated responses as previously reported in other cohorts. We further identified a peptide spanning amino acids 21 to 39 of the Rv1860 protein with the potential to distinguish latent TB infection from disease due to its ability to stimulate differential cytokine signatures in HV and PAT. We suggest that a TB vaccine carrying these and other CD8(+) T-cell-stimulating antigens has the potential to prevent progression of latent M. tuberculosis infection to TB disease.
Collapse
Affiliation(s)
- Vijaya Satchidanandam
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Naveen Kumar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Sunetra Biswas
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Rajiv S Jumani
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Chandni Jain
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Rajni Rani
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, India
| | - Bharti Aggarwal
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, India
| | - Jaya Singh
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, India
| | - Mohan Rao Kotnur
- Department of Chest Medicine, M. S. Ramiah Hospital, Bangalore, Karnataka, India
| | - Anand Sridharan
- National Tuberculosis Institute, Bangalore, Karnataka, India
| |
Collapse
|
39
|
Tuberculosis vaccines--state of the art, and novel approaches to vaccine development. Int J Infect Dis 2016; 32:5-12. [PMID: 25809749 DOI: 10.1016/j.ijid.2014.11.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 11/21/2014] [Accepted: 11/25/2014] [Indexed: 11/20/2022] Open
Abstract
The quest for a vaccine that could have a major impact in reducing the current global burden of TB disease in humans continues to be extremely challenging. Significant gaps in our knowledge and understanding of the pathogenesis and immunology of tuberculosis continue to undermine efforts to break new ground, and traditional approaches to vaccine development have thus far met with limited success. Existing and novel candidate vaccines are being assessed in the context of their ability to impact the various stages that culminate in disease transmission and an increase in the global burden of disease. Innovative methods of vaccine administration and delivery have provided a fresh stimulus to the search for the elusive vaccine. Here we discuss the current status of preclinical vaccine development, providing insights into alternative approaches to vaccine delivery and promising candidate vaccines. The state of the art of clinical development also is reviewed.
Collapse
|
40
|
Mycobacterium tuberculosis-specific and MHC class I-restricted CD8+ T-cells exhibit a stem cell precursor-like phenotype in patients with active pulmonary tuberculosis. Int J Infect Dis 2016; 32:13-22. [PMID: 25809750 DOI: 10.1016/j.ijid.2014.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/06/2014] [Indexed: 02/03/2023] Open
Abstract
The nature and longevity of the T-cell response directed against Mycobacterium tuberculosis (MTB) are important for effective pathogen containment. We analyzed ex vivo the nature of MTB antigen-specific T-cell responses directed against the MTB secreted antigens Rv0288, Rv1886c, Rv3875, the antigens Rv2958c, Rv2957, and Rv0447c (intracellular, non-secreted enzymes) in blood from Korean patients with active tuberculosis (TB). MTB-specific T-cell function was defined by intracellular cytokine production (interleukin (IL)-2, interferon gamma, tumour necrosis factor alpha, and IL-17) and by multimer-guided (HLA-A*02:01 and HLA-A*24:02) analysis of epitope-specific CD8+ T-cells, along with phenotypic markers (CD45RA and CCR7), CD107a, a marker for degranulation, and CD127 co-staining for T-cell differentiation and homing. Cytokine production analysis underestimated the frequencies of MTB antigen-specific T-cells defined by major histocompatibility complex (MHC) class I-peptide multimer analysis. We showed that MTB antigen-specific CD8+ T-cells exhibit a distinct marker profile associated with the nature of the MTB antigens, i.e., Rv0288, Rv1886c, and Rv3875-reactive T-cells clustered in the precursor T-cell compartment, whereas Rv2958c, Rv2957, and Rv0447c-reactive T-cells were associated with the terminally differentiated T-cell phenotype, in the patient cohort. Rv0288, Rv1886c, and Rv3875-specific CD8+ T-cells were significantly enriched for CD107a+ T-cells in HLA-A*02:01 (p<0.0001) and HLA-A*24:02 (p=0.0018) positive individuals, as compared to Rv2958c, Rv2957, and Rv0447c antigens. CD127 (IL-7 receptor)-expressing T-cells were enriched in HLA-A*02:01-positive individuals for the Rv0288, Rv1886c, and Rv3875 specificities (p=0.03). A high proportion of antigen-specific T-cells showed a precursor-like phenotype (CD45RA+CCR7+) and expressed the stem cell-associated markers CD95 and c-kit. These data show that MTB-specific T-cells can express stem cell-like features; this is associated with the nature of the MTB antigen and the genetic background of the individual.
Collapse
|
41
|
Abstract
While much progress has been made in the fight against the scourge of tuberculosis (TB), we are still some way from reaching the ambitious targets of eliminating it as a global public health problem by the mid twenty-first century. A new and effective vaccine that protects against pulmonary TB disease will be an essential element of any control strategy. Over a dozen vaccines are currently in development, but recent efficacy trial data from one of the most advanced candidates have been disappointing. Limitations of current preclinical animal models exist, together with a lack of a complete understanding of host immunity to TB or robust correlates of disease risk and protection. Therefore, in the context of such obstacles, we discuss the lessons identified from recent efficacy trials, current concepts of biomarkers and correlates of protection, the potential of innovative clinical models such as human challenge and conducting trials in high-incidence settings to evaluate TB vaccines in humans, and the use of systems vaccinology and novel technologies including transcriptomics and metabolomics, that may facilitate their utility.
Collapse
Affiliation(s)
| | - Helen McShane
- a The Jenner Institute, University of Oxford , Oxford , UK
| |
Collapse
|
42
|
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
|
43
|
Ravimohan S, Tamuhla N, Nfanyana K, Steenhoff AP, Letlhogile R, Frank I, MacGregor RR, Gross R, Weissman D, Bisson GP. Robust Reconstitution of Tuberculosis-Specific Polyfunctional CD4+ T-Cell Responses and Rising Systemic Interleukin 6 in Paradoxical Tuberculosis-Associated Immune Reconstitution Inflammatory Syndrome. Clin Infect Dis 2015; 62:795-803. [PMID: 26611774 DOI: 10.1093/cid/civ978] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/17/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The immunopathogenesis of paradoxical tuberculosis-associated immune reconstitution inflammatory syndrome (TB-IRIS) remains unclear. We determined the association between pathogen-specific T-cell responses and development of paradoxical TB-IRIS on antiretroviral therapy (ART). METHODS This study was nested within a prospective cohort study of HIV-infected patients with active pulmonary tuberculosis and baseline CD4 counts ≤125 cells/µL initiating ART. T-cell immune activation (CD38, HLA-DR, and PD-1 expression), phenotype, and polyfunctional pathogen-specific cellular immune responses prior to and 4 weeks after ART initiation were determined by flow cytometry. Patients with TB-IRIS were compared to non-IRIS controls using χ(2) and rank-sum tests and logistic regression. RESULTS TB-IRIS patients and controls had similar CD4 counts, levels of T-cell-associated immune activation, frequencies of T-cell memory subsets, and frequencies of interferon gamma (IFN-γ(+))/interleukin 2 (IL-2(+))/tumor necrosis factor alpha (TNF-α(+)) CD4(+) T-cells prior to ART initiation. After ART initiation, cellular immune activation and T-cell subsets also were similar in TB-IRIS patients and controls. In contrast, TB-IRIS patients had significantly greater early increases in the frequency of tuberculosis-specific polyfunctional IFN-γ(+)/IL-2(+)/TNF-α(+) CD4(+) T-cells on ART (P = .02); each quartile increase in the percentage of these cells was independently associated with a 2.8-fold increased risk of TB-IRIS (95% confidence interval, 1.1 to 7.5-fold). In a secondary analysis, patients with TB-IRIS had rapid, concomitant increases in tuberculosis-specific adaptive immune responses and interleukin 6 (IL-6) levels, whereas controls with similarly rapid increases in cellular immune function had IL-6 levels that tended to decrease on ART. CONCLUSIONS Rapid expansion of tuberculosis-specific polyfunctional CD4(+) T-cell responses, likely linked to increases in IL-6, is associated with development of paradoxical TB-IRIS.
Collapse
Affiliation(s)
- Shruthi Ravimohan
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia.,Botswana-UPenn Partnership, Gaborone
| | | | | | - Andrew P Steenhoff
- Botswana-UPenn Partnership, Gaborone.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania.,Children's Hospital of Philadelphia
| | | | - Ian Frank
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Rob Roy MacGregor
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Robert Gross
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia.,Botswana-UPenn Partnership, Gaborone.,Department of Biostatistics and Epidemiology, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Drew Weissman
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia.,Botswana-UPenn Partnership, Gaborone
| | - Gregory P Bisson
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine at the University of Pennsylvania, Philadelphia.,Botswana-UPenn Partnership, Gaborone.,Department of Biostatistics and Epidemiology, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
44
|
Ronacher K, Joosten SA, van Crevel R, Dockrell HM, Walzl G, Ottenhoff THM. Acquired immunodeficiencies and tuberculosis: focus on HIV/AIDS and diabetes mellitus. Immunol Rev 2015; 264:121-37. [PMID: 25703556 DOI: 10.1111/imr.12257] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The spread of human immunodeficiency virus (HIV) infection within Africa led to marked increases in numbers of cases of tuberculosis (TB), and although the epidemic peaked in 2006, there were still 1.8 million new cases in 2013, with 29.2 million prevalent cases. Half of all TB cases in Africa are in those with HIV co-infection. A brief review of the well-documented main immunological mechanisms of HIV-associated increased susceptibility to TB is presented. However, a new threat is facing TB control, which presents itself in the form of a rapid increase in the number of people living with type II diabetes mellitus (T2DM), particularly in areas that are already hardest hit by the TB epidemic. T2DM increases susceptibility to TB threefold, and the TB burden attributable to T2DM is 15%. This review addresses the much smaller body of research information available on T2DM-TB, compared to HIV-TB comorbidity. We discuss the altered clinical presentation of TB in the context of T2DM comorbidity, changes in innate and adaptive immune responses, including lymphocyte subsets and T-cell phenotypes, the effect of treatment of the different comorbidities, changes in biomarker expression and genetic predisposition to the respective morbidities, and other factors affecting the comorbidity. Although significant gains have been made in improving our understanding of the underlying mechanisms of T2DM-associated increased susceptibility, knowledge gaps still exist that require urgent attention.
Collapse
Affiliation(s)
- Katharina Ronacher
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and MRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | | | | | | | | |
Collapse
|
45
|
Stervbo U, Bozzetti C, Baron U, Jürchott K, Meier S, Mälzer JN, Nienen M, Olek S, Rachwalik D, Schulz AR, Neumann A, Babel N, Grützkau A, Thiel A. Effects of aging on human leukocytes (part II): immunophenotyping of adaptive immune B and T cell subsets. AGE (DORDRECHT, NETHERLANDS) 2015; 37:93. [PMID: 26324156 PMCID: PMC5005833 DOI: 10.1007/s11357-015-9829-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 08/07/2015] [Indexed: 06/04/2023]
Abstract
Immunosenescence results from a continuous deterioration of immune responses resulting in a decreased response to vaccines. A well-described age-related alteration of the immune system is the decrease of de novo generation of T and B cells. In addition, the accumulation of memory cells and loss of diversity in antigen specificities resulting from a lifetime of exposure to pathogens has also been described. However, the effect of aging on subsets of γδTCR(+) T cells and Tregs has been poorly described, and the efficacy of the recall response to common persistent infections in the elderly remains obscure. Here, we investigated alterations in the subpopulations of the B and T cells among 24 healthy young (aged 19-30) and 26 healthy elderly (aged 53-67) individuals. The analysis was performed by flow cytometry using freshly collected peripheral blood. γδTCR(+) T cells were overall decreased, while CD4(+)CD8(-) cells among γδTCR(+) T cells were increased in the elderly. Helios(+)Foxp3(+) and Helios(-)Foxp3(+) Treg cells were unaffected with age. Recent thymic emigrants, based on CD31 expression, were decreased among the Helios(+)Foxp3(+), but not the Helios(-)Foxp3(+) cell populations. We observed a decrease in Adenovirus-specific CD4(+) and CD8(+) T cells and an increase in CMV-specific CD4(+) T cells in the elderly. Similarly, INFγ(+)TNFα(+) double-positive cells were decreased among activated T cells after Adenovirus stimulation but increased after CMV stimulation. The data presented here indicate that γδTCR(+) T cells might stabilize B cells, and functional senescence might dominate at higher ages than those studied here.
Collapse
Affiliation(s)
- Ulrik Stervbo
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
- Deutsches Rheuma-Forschungszentrum – a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
- Marienhospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Cecilia Bozzetti
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Udo Baron
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Karsten Jürchott
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sarah Meier
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Julia Nora Mälzer
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Mikalai Nienen
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
- Marienhospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Sven Olek
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Dominika Rachwalik
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Axel Ronald Schulz
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Avidan Neumann
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
- Marienhospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Andreas Grützkau
- Deutsches Rheuma-Forschungszentrum – a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Thiel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
46
|
Smith SG, Smits K, Joosten SA, van Meijgaarden KE, Satti I, Fletcher HA, Caccamo N, Dieli F, Mascart F, McShane H, Dockrell HM, Ottenhoff THM, TBVI TB Biomarker Working Group. Intracellular Cytokine Staining and Flow Cytometry: Considerations for Application in Clinical Trials of Novel Tuberculosis Vaccines. PLoS One 2015; 10:e0138042. [PMID: 26367374 PMCID: PMC4569436 DOI: 10.1371/journal.pone.0138042] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/24/2015] [Indexed: 01/24/2023] Open
Abstract
Intracellular cytokine staining combined with flow cytometry is one of a number of assays designed to assess T-cell immune responses. It has the specific advantage of enabling the simultaneous assessment of multiple phenotypic, differentiation and functional parameters pertaining to responding T-cells, most notably, the expression of multiple effector cytokines. These attributes make the technique particularly suitable for the assessment of T-cell immune responses induced by novel tuberculosis vaccines in clinical trials. However, depending upon the particular nature of a given vaccine and trial setting, there are approaches that may be taken at different stages of the assay that are more suitable than other alternatives. In this paper, the Tuberculosis Vaccine Initiative (TBVI) TB Biomarker Working group reports on efforts to assess the conditions that will determine when particular assay approaches should be employed. We have found that choices relating to the use of fresh whole blood or peripheral blood mononuclear cells (PBMC) and frozen PBMC; use of serum-containing or serum-free medium; length of stimulation period and use of co-stimulatory antibodies can all affect the sensitivity of intracellular cytokine assays. In the case of sample material, frozen PBMC, despite some loss of sensitivity, may be more advantageous for batch analysis. We also recommend that for multi-site studies, common antibody panels, gating strategies and analysis approaches should be employed for better comparability.
Collapse
Affiliation(s)
- Steven G. Smith
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
- * E-mail:
| | - Kaatje Smits
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Iman Satti
- The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, United Kingdom
| | - Helen A. Fletcher
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
- The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, United Kingdom
| | - Nadia Caccamo
- Biomedical Research Centre, Università di Palermo, Palermo, Italy, Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Francesco Dieli
- Biomedical Research Centre, Università di Palermo, Palermo, Italy, Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Francoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
- Immunobiology Clinic, Hôpital Erasme, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Helen McShane
- The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, United Kingdom
| | - Hazel M. Dockrell
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
47
|
Correlates of Vaccine-Induced Protection against Mycobacterium tuberculosis Revealed in Comparative Analyses of Lymphocyte Populations. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:1096-108. [PMID: 26269537 DOI: 10.1128/cvi.00301-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/09/2015] [Indexed: 12/16/2022]
Abstract
A critical hindrance to the development of a novel vaccine against Mycobacterium tuberculosis is a lack of understanding of protective correlates of immunity and of host factors involved in a successful adaptive immune response. Studies from our group and others have used a mouse-based in vitro model system to assess correlates of protection. Here, using this coculture system and a panel of whole-cell vaccines with varied efficacy, we developed a comprehensive approach to understand correlates of protection. We compared the gene and protein expression profiles of vaccine-generated immune peripheral blood lymphocytes (PBLs) to the profiles found in immune splenocytes. PBLs not only represent a clinically relevant cell population, but comparing the expression in these populations gave insight into compartmentally specific mechanisms of protection. Additionally, we performed a direct comparison of host responses induced when immune cells were cocultured with either the vaccine strain Mycobacterium bovis BCG or virulent M. tuberculosis. These comparisons revealed host-specific and bacterium-specific factors involved in protection against virulent M. tuberculosis. Most significantly, we identified a set of 13 core molecules induced in the most protective vaccines under all of the conditions tested. Further validation of this panel of mediators as a predictor of vaccine efficacy will facilitate vaccine development, and determining how each promotes adaptive immunity will advance our understanding of antimycobacterial immune responses.
Collapse
|
48
|
Kallert S, Zenk SF, Walther P, Grieshober M, Weil T, Stenger S. Liposomal delivery of lipoarabinomannan triggers Mycobacterium tuberculosis specific T-cells. Tuberculosis (Edinb) 2015; 95:452-62. [PMID: 26043674 DOI: 10.1016/j.tube.2015.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 12/20/2022]
Abstract
Lipoarabinomannan (LAM) is a major cell wall component of Mycobacterium tuberculosis (Mtb). LAM specific human T-lymphocytes release interferon-γ (IFNγ) and have antimicrobial activity against intracellular Mtb suggesting that they contribute to protection. Therefore the induction of LAM-specific memory T-cells is an attractive approach for the design of a new vaccine against tuberculosis. A prerequisite for the activation of LAM-specific T-cells is the efficient uptake and transport of the glycolipid antigen to the CD1 antigen presenting machinery. Based on the hydrophobicity of LAM we hypothesized that packaging of LAM into liposomes will support the activation of T-lymphocytes. We prepared liposomes containing phosphatidylcholine, cholesterol, stearylated octaarginine and LAM via thin layer hydration method (LIPLAM). Flow cytometry analysis using fluorescently labelled LIPLAM showed an efficient uptake by antigen presenting cells. LAM delivered via liposomes was biologically active as demonstrated by the down-regulation of peroxisome proliferator activated receptor gamma (PPARγ) protein expression. Importantly, LIPLAM induced higher IFNγ production by primary human T-lymphocytes than purified LAM (2-16 times) or empty liposomes. These results suggest that the delivery of mycobacterial glycolipids via liposomes is a promising approach to promote the induction of M. tuberculosis specific T-cell responses.
Collapse
Affiliation(s)
- Stephanie Kallert
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | - Sebastian F Zenk
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | - Paul Walther
- Central Unit Electron Microscopy, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Mark Grieshober
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | - Tanja Weil
- Institute for Organic Chemistry III/Macromolecular Chemistry, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
49
|
Measuring Cellular Immunity to Influenza: Methods of Detection, Applications and Challenges. Vaccines (Basel) 2015; 3:293-319. [PMID: 26343189 PMCID: PMC4494351 DOI: 10.3390/vaccines3020293] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022] Open
Abstract
Influenza A virus is a respiratory pathogen which causes both seasonal epidemics and occasional pandemics; infection continues to be a significant cause of mortality worldwide. Current influenza vaccines principally stimulate humoral immune responses that are largely directed towards the variant surface antigens of influenza. Vaccination can result in an effective, albeit strain-specific antibody response and there is a need for vaccines that can provide superior, long-lasting immunity to influenza. Vaccination approaches targeting conserved viral antigens have the potential to provide broadly cross-reactive, heterosubtypic immunity to diverse influenza viruses. However, the field lacks consensus on the correlates of protection for cellular immunity in reducing severe influenza infection, transmission or disease outcome. Furthermore, unlike serological methods such as the standardized haemagglutination inhibition assay, there remains a large degree of variation in both the types of assays and method of reporting cellular outputs. T-cell directed immunity has long been known to play a role in ameliorating the severity and/or duration of influenza infection, but the precise phenotype, magnitude and longevity of the requisite protective response is unclear. In order to progress the development of universal influenza vaccines, it is critical to standardize assays across sites to facilitate direct comparisons between clinical trials.
Collapse
|
50
|
Tchakoute CT, Hesseling AC, Kidzeru EB, Gamieldien H, Passmore JAS, Jones CE, Gray CM, Sodora DL, Jaspan HB. Delaying BCG vaccination until 8 weeks of age results in robust BCG-specific T-cell responses in HIV-exposed infants. J Infect Dis 2015; 211:338-46. [PMID: 25108027 PMCID: PMC4318913 DOI: 10.1093/infdis/jiu434] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/14/2014] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND BCG vaccination prevents disseminated tuberculosis in children, but it is contraindicated for persons with human immunodeficiency virus (HIV) infection because it can result in severe disease in this population. In tuberculosis-endemic regions, BCG vaccine is administered soon after birth, before in utero and peripartum HIV infection is excluded. We therefore assessed the immunogenicity of BCG vaccine in HIV-exposed infants who received BCG at birth or at 8 weeks of age. METHODS HIV-exposed, uninfected infants were randomly assigned to receive BCG vaccination at birth (the early vaccination arm) or 8 weeks of age (the delayed vaccination arm). BCG-specific proliferative and intracellular cytokine responses were assessed in 28 infants per arm at 6, 8, and 14 weeks of life. RESULTS There was no difference in BCG-specific T-cell proliferation between the study arms 6 weeks after vaccination. However, at 14 weeks of age, the frequency of interferon γ-expressing CD4(+) T cells and multifunctional BCG-specific responses in the delayed vaccinated arm were significantly higher than those in the early vaccination arm (P = .021 and P = .011, respectively). CONCLUSIONS The immunogenicity of BCG vaccination in HIV-exposed, uninfected infants is not compromised when delayed until 8 weeks of age and results in robust BCG-specific T-cell responses at 14 weeks of age. These findings support further evaluation of this modified BCG vaccination strategy for HIV-exposed infants. CLINICAL TRIALS REGISTRATION NCT02062580.
Collapse
Affiliation(s)
| | - Anneke C. Hesseling
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town
| | | | | | - Jo-Ann S. Passmore
- Division of Medical Virology
- National Health Laboratory Services, South Africa
| | - Christine E. Jones
- Institute of Infectious Disease and Molecular Medicine, Department of Clinical Laboratory Sciences, University of Cape Town
- Paediatric Infectious Diseases Research Group, St George's, University of London, United Kingdom
| | - Clive M. Gray
- Division of Immunology
- National Health Laboratory Services, South Africa
| | | | - Heather B. Jaspan
- Division of Immunology
- Seattle Biomedical Research Institute, Washington
| |
Collapse
|