1
|
Mehanna N, Pradhan A, Kaur R, Kontopoulos T, Rosati B, Carlson D, Cheung NKV, Xu H, Bean J, Hsu KC, Le Luduec JB, Vorkas CK. CD8α marks a Mycobacterium tuberculosis-reactive human NK cell population with high activation potential. Sci Rep 2025; 15:15095. [PMID: 40301594 PMCID: PMC12041513 DOI: 10.1038/s41598-025-98367-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
Natural Killer (NK) cells can recognize and kill Mycobacterium tuberculosis (Mtb)-infected cells in vitro, however their role after natural human exposure has not been well-studied. To identify Mtb-responsive NK cell populations, we analyzed the peripheral blood of healthy household contacts of active Tuberculosis (TB) cases and source community donors in an endemic region of Port-au-Prince, Haiti by flow cytometry. We observed higher CD8α expression on NK cells in putative resistors (Interferon γ release assay negative; IGRA- contacts) with a loss of CD8α surface expression during household-associated exposure and active TB disease. In vitro assays and CITE-seq analysis of CD8α+ NK cells demonstrated enhanced maturity, cytotoxic gene expression, and response to cytokine stimulation relative to CD8α- NK cells. CD8α+ NK cells also displayed dynamic surface expression dependent on MHC class I in contrast to conventional CD8+ T cells. Together, these results support a specialized role for CD8α+ NK cell populations during Mtb infection correlating with disease resistance.
Collapse
Affiliation(s)
- Nezar Mehanna
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Atul Pradhan
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Rimanpreet Kaur
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Theodota Kontopoulos
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Barbara Rosati
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
| | - David Carlson
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - James Bean
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Katharine C Hsu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jean-Benoit Le Luduec
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Charles Kyriakos Vorkas
- Renaissance School of Medicine at Stony Brook University, 101 Nicolls Road, Health Sciences Center 15060-I, Stony Brook, NY, 11794, USA.
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794, USA.
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794, USA.
- Division of Infectious Diseases, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
2
|
Shi Z, Zhou L, Wang X, Zhang Z, Kong L, Zhang Y. Immunogenic profiling of Mycobacterium tuberculosis Rv1513 reveals its ability to switch on Th1 based immunity. Arch Microbiol 2024; 206:352. [PMID: 39012499 DOI: 10.1007/s00203-024-04033-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/05/2024] [Indexed: 07/17/2024]
Abstract
Tuberculosis (TB) is one of the infectious diseases caused by the pathogen Mycobacterium tuberculosis that continuously threatens the global human health. Bacillus Calmette-Guérin (BCG) vaccine is the only vaccine that has been used clinically to prevent tuberculosis in recent centuries, but its limitations in preventing latent infection and reactivation of tuberculosis do not provide full protection. In this study, we selected the membrane-associated antigen Rv1513 of Mycobacterium. In order to achieve stable expression and function of the target gene, the prokaryotic expression recombinant vector pET30b-Rv1513 was constructed and expressed and purified its protein. Detection of IFN- γ levels in the peripheral blood of TB patients stimulated by whole blood interferon release assay (WBIA) and multi-microsphere flow immunofluorescence luminescence (MFCIA) revealed that the induced production of cytokines, such as IFN-γ and IL-6, was significantly higher than that in the healthy group. Rv1513 combined with adjuvant DMT (adjuvant system liposomes containing dimethyldioctadecylammonium bromide (DDA), monophospholipid A (MPL), and trehalose-660-dibenzoic acid (TDB)) was used to detect serum specific antibodies, cytokine secretion from splenic suprasplenic cell supernatants, and multifunctional T-cell levels in splenocytes in immunised mice. The levels of IFN-γ, TNF-α, and IL-2 secreted by mouse splenocytes were found in the Rv1513+DMT group and the BCG+Rv1513+DMT group. The serum levels of IgG and its subclasses and the number of IFN-γ+T cells, TNF-α+T and IFN-γ+TNF-α+T cells in the induced CD4+/CD8+T cells in mice were significantly higher than those in the BCG group, and the highest levels were found in the BCG+Rv1513+DMT group. These findings suggest that Rv1513/DMT may serve as a potential subunit vaccine candidate that may be effective as a booster vaccine after the first BCG vaccination.
Collapse
Affiliation(s)
- Zilun Shi
- Department of Cancer, Hospital of Huainan Eastern Hospital Group, Huainan, 232035, China
| | - Lili Zhou
- Department of Cancer, Hospital of Huainan Eastern Hospital Group, Huainan, 232035, China
| | - Xiaochun Wang
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China.
| | - Zian Zhang
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - LingYun Kong
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Yanpeng Zhang
- Department of Cosmetology, College of Medicine, Huainan Union University, Huainan, 232001, China
| |
Collapse
|
3
|
Yang Y, Chen YZ, Xia T. Optimizing antigen selection for the development of tuberculosis vaccines. CELL INSIGHT 2024; 3:100163. [PMID: 38572176 PMCID: PMC10987857 DOI: 10.1016/j.cellin.2024.100163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
Tuberculosis (TB) remains a prevalent global infectious disease caused by genetically closely related tubercle bacilli in Mycobacterium tuberculosis complex (MTBC). For a century, the Bacillus Calmette-Guérin (BCG) vaccine has been the primary preventive measure against TB. While it effectively protects against extrapulmonary forms of pediatric TB, it lacks consistent efficacy in providing protection against pulmonary TB in adults. Consequently, the exploration and development of novel TB vaccines, capable of providing broad protection to populations, have consistently constituted a prominent area of interest in medical research. This article presents a concise overview of the novel TB vaccines currently undergoing clinical trials, discussing their classification, protective efficacy, immunogenicity, advantages, and limitations. In vaccine development, the careful selection of antigens that can induce strong and diverse specific immune responses is essential. Therefore, we have summarized the molecular characteristics, biological function, immunogenicity, and relevant studies associated with the chosen antigens for TB vaccines. These insights gained from vaccines and immunogenic proteins will inform the development of novel mycobacterial vaccines, particularly mRNA vaccines, for effective TB control.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yi-Zhen Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Tian Xia
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| |
Collapse
|
4
|
Mehanna N, Pradhan A, Kaur R, Kontopoulos T, Rosati B, Carlson D, Cheung NK, Xu H, Bean J, Hsu K, Le Luduec JB, Vorkas CK. Loss of circulating CD8α + NK cells during human Mycobacterium tuberculosis infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.588542. [PMID: 38659858 PMCID: PMC11042275 DOI: 10.1101/2024.04.16.588542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Natural Killer (NK) cells can recognize and kill Mtb-infected cells in vitro, however their role after natural human exposure has not been well-studied. To identify Mtb-responsive NK cell populations, we analyzed the peripheral blood of healthy household contacts of active Tuberculosis (TB) cases and source community donors in an endemic region of Port-au-Prince, Haiti by flow cytometry. We observed higher CD8α expression on NK cells in putative resistors (IGRA- contacts) with a progressive loss of these circulating cells during household-associated latent infection and disease. In vitro assays and CITE-seq analysis of CD8α+ NK cells demonstrated enhanced maturity, cytotoxic gene expression, and response to cytokine stimulation relative to CD8α- NK cells. CD8α+ NK cells also displayed dynamic surface expression dependent on MHC I in contrast to conventional CD8+ T cells. Together, these results support a specialized role for CD8α+ NK cell populations during Mtb infection correlating with disease resistance.
Collapse
Affiliation(s)
- Nezar Mehanna
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Atul Pradhan
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Rimanpreet Kaur
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Theodota Kontopoulos
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Barbara Rosati
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - David Carlson
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
| | - Nai-Kong Cheung
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Hong Xu
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - James Bean
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Katherine Hsu
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Jean-Benoit Le Luduec
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Charles Kyriakos Vorkas
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, 11794
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, 11794
| |
Collapse
|
5
|
Zhang Z, Xu L, Wang X, Kong L, Shi Z, Zhong Q, Xu Y, Wang J. Construction and expression of Mycobacterium tuberculosis fusion protein SHR3 and its immunogenicity analysis in combination with various adjuvants. Tuberculosis (Edinb) 2024; 145:102480. [PMID: 38278100 DOI: 10.1016/j.tube.2024.102480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/01/2024] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
Tuberculosis (TB) today remains the leading cause of global deaths due to infectious bacterial pathogens. The Bacillus Calmette-Guérin (BCG) vaccine is the only vaccine clinically used to prevent TB. However, its limitations in preventing latent infection and TB reactivation mean that it does not provide comprehensive protection. In this study, we successfully constructed and expressed the multistage fusion protein, SHR3, and used whole blood IFN-γ release assay (WBIA) with flow cytometry to detect antigen specificity, further confirmed by enzyme-linked immunosorbent assay (ELISA). SHR3 and its subfractional proteins stimulated the level of IFN-γ production by lymphocytes from M. tb-infected patients, inducing the production of single-positive and double-positive CD4+ and CD8+ T cells with IFN-γ and IL-2, at levels significantly higher than those of healthy controls. The fusion protein and complex adjuvant group (SHR3/DMT) induced mice to produce significantly higher levels of IgG antibodies and their subclasses, with IgG2a/IgG1 results showing a convergent Th1-type response; mice in the BCG + SHR3/DMT group induced secretion of the highest levels of IL-2, and TNF-α, irrespective of stimulation with purified protein derivative or SHR3. These findings suggest that SHR3/DMT could be a potential subunit vaccine candidate that may serve as an effective booster vaccine after BCG primary immunization.
Collapse
Affiliation(s)
- Zian Zhang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Lifa Xu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China.
| | - Xiaochun Wang
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China.
| | - LingYun Kong
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Zilun Shi
- Department of Clinical Laboratory, Affiliated Cancer Hospital, Anhui University of Science and Technology, Huainan, 232001, China
| | - Qiangsen Zhong
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Yun Xu
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Jianghong Wang
- Department of Pathogen Biology, School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| |
Collapse
|
6
|
Kim HD, Choi H, Abekura F, Park J, Cho SH, Lee YC, Kim CH. Up-regulation of inflammatory reactions by MPT32, a secreted protein of Mycobacterium tuberculosis in RAW264.7 macrophages. J Cell Biochem 2023; 124:1423-1434. [PMID: 37642132 DOI: 10.1002/jcb.30456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023]
Abstract
Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb) and is still one of the global health burdens. The occurrence of various cases and multidrug resistance confirm that TB has not been completely conquered. For these reasons, the present research has been conducted to explore TB vaccine and drug candidate possibility using Mtb-secreted proteins. Among these proteins, MPT32 is known to have antigenicity and immunogenicity. There has not been a report on the host immune responses and regulation in macrophage cells. The present study was conducted with MPT32 in RAW 264.7 murine macrophage cells that control immune responses by sensing pathogen invasion and environmental change. We have found that MPT32 could activate lipopolysaccharide (LPS)-induced gene expression of metalloproteinase-9 (MMP-9) and inflammation in RAW 264.7 cells. After treating cells with MPT32, the increase in pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β (IL-1β) and IL-6, was observed. In addition, activated macrophages expressed inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) to generate various inflammatory mediator molecules, such as nitric oxide (NO). The increase in iNOS and COX-2 levels, which are up-regulators of MMP-9 expression, was also confirmed. The biochemical events are involved in the downstream of activated MAPK signaling and translocation of NF-κ B transcription factor. The present results prove the immunomodulatory effect of MPT32 in the RAW 264.7 murine macrophage cells. it claims the possibility of a TB vaccination and drug candidate using MPT32, contributing to the prevention of TB.
Collapse
Affiliation(s)
- Hee-Do Kim
- Department of Biological Science, SungKyunkwan University, Suwon, Republic of Korea
| | - Hyunju Choi
- Department of Biological Science, SungKyunkwan University, Suwon, Republic of Korea
| | - Fukushi Abekura
- Department of Biological Science, SungKyunkwan University, Suwon, Republic of Korea
| | - Junyoung Park
- Department of Biological Science, SungKyunkwan University, Suwon, Republic of Korea
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Seung-Hak Cho
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, South Korea
| | - Cheorl-Ho Kim
- Department of Biological Science, SungKyunkwan University, Suwon, Republic of Korea
- Samsung Advanced Institute for Health Science and Technology (SAIHST), Suwon, Republic of Korea
| |
Collapse
|
7
|
Paliwal D, Thom M, Hussein A, Ravishankar D, Wilkes A, Charleston B, Jones IM. Towards Reverse Vaccinology for Bovine TB: High Throughput Expression of Full Length Recombinant Mycobacterium bovis Proteins. Front Mol Biosci 2022; 9:889667. [PMID: 36032666 PMCID: PMC9402895 DOI: 10.3389/fmolb.2022.889667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis caused by Mycobacterium bovis, is a significant global pathogen causing economic loss in livestock and zoonotic TB in man. Several vaccine approaches are in development including reverse vaccinology which uses an unbiased approach to select open reading frames (ORF) of potential vaccine candidates, produce them as recombinant proteins and assesses their immunogenicity by direct immunization. To provide feasibility data for this approach we have cloned and expressed 123 ORFs from the M. bovis genome, using a mixture of E. coli and insect cell expression. We used a concatenated open reading frames design to reduce the number of clones required and single chain fusion proteins for protein pairs known to interact, such as the members of the PPE-PE family. Over 60% of clones showed soluble expression in one or the other host and most allowed rapid purification of the tagged bTB protein from the host cell background. The catalogue of recombinant proteins represents a resource that may be suitable for test immunisations in the development of an effective bTB vaccine.
Collapse
Affiliation(s)
- Deepa Paliwal
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Areej Hussein
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Alex Wilkes
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Ian M. Jones
- School of Biological Sciences, University of Reading, Reading, United Kingdom
- *Correspondence: Ian M. Jones,
| |
Collapse
|
8
|
Characterization of the Protective Immune Responses Conferred by Recombinant BCG Overexpressing Components of Mycobacterium tuberculosis Sec Protein Export System. Vaccines (Basel) 2022; 10:vaccines10060945. [PMID: 35746553 PMCID: PMC9229301 DOI: 10.3390/vaccines10060945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 11/29/2022] Open
Abstract
Mycobacterium bovis Bacillus Calmette-Guérin (BCG) is the only approved vaccine against tuberculosis (TB). However, its efficacy in preventing pulmonary TB in adults is limited. Despite its variable efficacy, BCG offers a number of unique and beneficial characteristics, which make it suitable as a vaccine vehicle to express recombinant molecules. In Mycobacterium tuberculosis, the general Sec pathway is an essential cellular process, and it is responsible for exporting the majority of proteins across the cytoplasmic membrane, including potent immune-protective antigens, such as members of the antigen 85 (Ag85) complex. We engineered BCG to overexpress the M. tuberculosis SecDFG proteins in order to improve the efficiency of the Sec-dependent export system and, thus, enhance the secretion of immunogenic proteins. BCGSecDFG displayed increased intracellular survival within macrophages in vitro and greater persistence in the lymphoid organs of vaccinated mice than parental BCG. In addition, vaccination with BCGSecDFG generated higher numbers of IFN-γ-secreting T cells in response to secreted mycobacterial antigens compared to BCG, particularly members of the Ag85 complex. Furthermore, vaccination with BCGSecDFG significantly reduced the bacterial load in the lungs and spleens of M. tuberculosis-infected mice, which was comparable to the protection afforded by parental BCG. Therefore, the modification of protein secretion in BCG can improve antigen-specific immunogenicity.
Collapse
|
9
|
Therapeutic Effect of Subunit Vaccine AEC/BC02 on Mycobacterium tuberculosis Post-Chemotherapy Relapse Using a Latent Infection Murine Model. Vaccines (Basel) 2022; 10:vaccines10050825. [PMID: 35632581 PMCID: PMC9145927 DOI: 10.3390/vaccines10050825] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/21/2022] [Indexed: 11/17/2022] Open
Abstract
Tuberculosis (TB), caused by the human pathogen Mycobacterium tuberculosis (Mtb), is an infectious disease that presents a major threat to human health. Bacillus Calmette-Guérin (BCG), the only licensed TB vaccine, is ineffective against latent TB infection, necessitating the development of further TB drugs or therapeutic vaccines. Herein, we evaluated the therapeutic effect of a novel subunit vaccine AEC/BC02 after chemotherapy in a spontaneous Mtb relapse model. Immunotherapy followed 4 weeks of treatment with isoniazid and rifapentine, and bacterial loads in organs, pathological changes, and adaptive immune characteristics were investigated. The results showed slowly increased bacterial loads in the spleen and lungs of mice inoculated with AEC/BC02 with significantly lower loads than those of the control groups. Pathological scores for the liver, spleen, and lungs decreased accordingly. Moreover, AEC/BC02 induced antigen-specific IFN-γ-secreting or IL-2-secreting cellular immune responses, which decreased with the number of immunizations and times. Obvious Ag85b- and EC-specific IgG were observed in mice following the treatment with AEC/BC02, indicating a significant Th1-biased response. Taken together, these data suggest that AEC/BC02 immunotherapy post-chemotherapy may shorten future TB treatment.
Collapse
|
10
|
A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int Immunopharmacol 2022; 109:108791. [PMID: 35487086 DOI: 10.1016/j.intimp.2022.108791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem despite widespread use of the Bacillus BCG vaccine. This situation is worsened by co-infection with HIV, and the development of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains. Thus, novel vaccine candidates and improved vaccination strategies are urgently needed in order to reduce the incidence of TB and even to eradicate TB by 2050. Over the last few decades, 23 novel TB vaccines have entered into clinical trials, more than 13 new vaccines have reached various stages of preclinical development, and more than 50 potential candidates are in the discovery stage as next-generation vaccines. Nevertheless, why has a century of attempts to introduce an effective TB vaccine failed? Who should be blamed -scientists, human response, or Mtb strategies? Literature review reveals that the elimination of latent or active Mtb infections in a given population seems to be an epigenetic process. With a better understanding of the connections between bacterial infections and gene expression conditions in epigenetic events, opportunities arise in designing protective vaccines or therapeutic agents, particularly as epigenetic processes can be reversed. Therefore, this review provides a brief overview of different approaches towards novel vaccination strategies and the mechanisms underlying these approaches.
Collapse
|
11
|
Belnoue E, Vogelzang A, Nieuwenhuizen NE, Krzyzaniak MA, Darbre S, Kreutzfeldt M, Wagner I, Merkler D, Lambert PH, Kaufmann SHE, Siegrist CA, Pinschewer DD. Replication-Deficient Lymphocytic Choriomeningitis Virus-Vectored Vaccine Candidate for the Induction of T Cell Immunity against Mycobacterium tuberculosis. Int J Mol Sci 2022; 23:2700. [PMID: 35269842 PMCID: PMC8911050 DOI: 10.3390/ijms23052700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 11/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) represents a major burden to global health, and refined vaccines are needed. Replication-deficient lymphocytic choriomeningitis virus (rLCMV)-based vaccine vectors against cytomegalovirus have proven safe for human use and elicited robust T cell responses in a large proportion of vaccine recipients. Here, we developed an rLCMV vaccine expressing the Mtb antigens TB10.4 and Ag85B. In mice, rLCMV elicited high frequencies of polyfunctional Mtb-specific CD8 and CD4 T cell responses. CD8 but not CD4 T cells were efficiently boosted upon vector re-vaccination. High-frequency responses were also observed in neonatally vaccinated mice, and co-administration of rLCMV with Expanded Program of Immunization (EPI) vaccines did not result in substantial reciprocal interference. Importantly, rLCMV immunization significantly reduced the lung Mtb burden upon aerosol challenge, resulting in improved lung ventilation. Protection was associated with increased CD8 T cell recruitment but reduced CD4 T cell infiltration upon Mtb challenge. When combining rLCMV with BCG vaccination in a heterologous prime-boost regimen, responses to the rLCMV-encoded Mtb antigens were further augmented, but protection was not significantly different from rLCMV or BCG vaccination alone. This work suggests that rLCMV may show utility for neonatal and/or adult vaccination efforts against pulmonary tuberculosis.
Collapse
Affiliation(s)
- Elodie Belnoue
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Alexis Vogelzang
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; (A.V.); (N.E.N.); (S.H.E.K.)
| | - Natalie E. Nieuwenhuizen
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; (A.V.); (N.E.N.); (S.H.E.K.)
| | - Magdalena A. Krzyzaniak
- Division of Experimental Virology, Department of Biomedicine, University of Basel, 4003 Basel, Switzerland;
| | - Stephanie Darbre
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva 4, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva 4, Switzerland
| | - Paul-Henri Lambert
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; (A.V.); (N.E.N.); (S.H.E.K.)
| | - Claire-Anne Siegrist
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Daniel D. Pinschewer
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; (E.B.); (S.D.); (M.K.); (I.W.); (D.M.); (P.-H.L.); (C.-A.S.)
- W.H.O. Collaborating Centre for Vaccine Immunology, University of Geneva, 1211 Geneva 4, Switzerland
- Division of Experimental Virology, Department of Biomedicine, University of Basel, 4003 Basel, Switzerland;
| |
Collapse
|
12
|
Junqueira-Kipnis AP, de Castro Souza C, de Oliveira Carvalho AC, de Oliveira FM, Almeida VP, de Paula AR, Celes MR, Kipnis A. Protease-Based Subunit Vaccine in Mice Boosts BCG Protection against Mycobacterium tuberculosis. Vaccines (Basel) 2022; 10:vaccines10020306. [PMID: 35214766 PMCID: PMC8877678 DOI: 10.3390/vaccines10020306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
The significant number of people with latent and active tuberculosis infection requires further efforts to develop new vaccines or improve the Bacillus Calmette-Guérin (BCG), which is the only approved vaccine against this disease. In this study, we developed a recombinant fusion protein (PEPf) containing high-density immunodominant epitope sequences from Rv0125, Rv2467, and Rv2672 Mycobacterium tuberculosis (Mtb) proteases that proved immunogenic and used it to develop a recombinant BCG vaccine expressing the fusion protein. After challenging using Mtb, a specific immune response was recalled, resulting in a reduced lung bacterial load with similar protective capabilities to BCG. Thus BCG PEPf failed to increase the protection conferred by BCG. The PEPf was combined with Advax4 adjuvant and tested as a subunit vaccine using a prime-boost strategy. PEPf + Advax4 significantly improved protection after Mtb challenge, with a reduction in bacterial load in the lungs. Our results confirm that Mtb proteases can be used to develop vaccines against tuberculosis and that the use of the recombinant PEPf subunit protein following a prime-boost regimen is a promising strategy to improve BCG immunity.
Collapse
|
13
|
Golichenari B, Yari S, Tasbiti AH, Behravan J, Vaziri F, Ghazvini K. First conjugation directed traverse of gene cassettes harboring α1,3GT from fast-growing Mycobacterium smegmatis mc2 155 to slow-growing pathogen Mycobacterium tuberculosis H37Rv, presumably opening up new scopes in tuberculosis treatment. Enzyme Microb Technol 2022; 156:110003. [DOI: 10.1016/j.enzmictec.2022.110003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/02/2022] [Accepted: 01/31/2022] [Indexed: 11/03/2022]
|
14
|
Keating T, Lethbridge S, Allnutt JC, Hendon-Dunn CL, Thomas SR, Alderwick LJ, Taylor SC, Bacon J. Mycobacterium tuberculosis modifies cell wall carbohydrates during biofilm growth with a concomitant reduction in complement activation. ACTA ACUST UNITED AC 2021; 7:100065. [PMID: 34778603 PMCID: PMC8577165 DOI: 10.1016/j.tcsw.2021.100065] [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: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 11/26/2022]
Abstract
The development of new vaccines for TB needs to be underpinned by an understanding of both the molecular and cellular mechanisms of host-pathogen interactions and how the immune response can be modulated to achieve protection from disease. Complement orchestrates many aspects of the innate and adaptive immune responses. However, little is known about the contribution of the complement pathways during TB disease, particularly with respect to mycobacterial phenotype. Extracellular communities (biofilms) of M. tuberculosis are found in the acellular rim of granulomas, during disease, and these are likely to be present in post-primary TB episodes, in necrotic lesions. Our study aimed to determine which mycobacterial cell wall components were altered during biofilm growth and how these cell wall alterations modified the complement response. We have shown that M. tuberculosis biofilms modified their cell wall carbohydrates and elicited reduced classical and lectin pathway activation. Consistent with this finding was the reduction of C3b/iC3b deposition on biofilm cell wall carbohydrate extracts. Here, we have highlighted the role of cell wall carbohydrate alterations during biofilm growth of M. tuberculosis and subsequent modulation of complement activation.
Collapse
Affiliation(s)
- Thomas Keating
- TB Discovery Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom.,School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Samuel Lethbridge
- TB Discovery Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom
| | - Jon C Allnutt
- TB Discovery Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom
| | - Charlotte L Hendon-Dunn
- TB Discovery Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom
| | - Stephen R Thomas
- Pathogen Immunology Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom
| | - Luke J Alderwick
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stephen C Taylor
- Pathogen Immunology Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom
| | - Joanna Bacon
- TB Discovery Group, National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, United Kingdom
| |
Collapse
|
15
|
PE_PGRS33, an Important Virulence Factor of Mycobacterium tuberculosis and Potential Target of Host Humoral Immune Response. Cells 2021; 10:cells10010161. [PMID: 33467487 PMCID: PMC7830552 DOI: 10.3390/cells10010161] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/04/2021] [Accepted: 01/12/2021] [Indexed: 01/16/2023] Open
Abstract
PE_PGRS proteins are surface antigens of Mycobacterium tuberculosis (Mtb) and a few other pathogenic mycobacteria. The PE_PGRS33 protein is among the most studied PE_PGRSs. It is known that the PE domain of PE_PGRS33 is required for the protein translocation through the mycobacterial cell wall, where the PGRS domain remains available for interaction with host receptors. Interaction with Toll like receptor 2 (TLR2) promotes secretion of inflammatory chemokines and cytokines, which are key in the immunopathogenesis of tuberculosis (TB). In this review, we briefly address some key challenges in the development of a TB vaccine and attempt to provide a rationale for the development of new vaccines aimed at fostering a humoral response against Mtb. Using PE_PGRS33 as a model for a surface-exposed antigen, we exploit the availability of current structural data using homology modeling to gather insights on the PGRS domain features. Our study suggests that the PGRS domain of PE_PGRS33 exposes four PGII sandwiches on the outer surface, which, we propose, are directly involved through their loops in the interactions with the host receptors and, as such, are promising targets for a vaccination strategy aimed at inducing a humoral response.
Collapse
|
16
|
Van Der Walt M, Keddy KH. The Tuberculosis-Depression Syndemic and Evolution of Pharmaceutical Therapeutics: From Ancient Times to the Future. Front Psychiatry 2021; 12:617751. [PMID: 34140898 PMCID: PMC8203803 DOI: 10.3389/fpsyt.2021.617751] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/15/2021] [Indexed: 01/08/2023] Open
Abstract
The interplay between tuberculosis and depression has been problematic since the humoralists. Over the centuries similarities in disease management have transpired. With the advent of isoniazid chemotherapy, transformation of tuberculosis patients from morbidly depressive to euphoric was noted. Isoniazid was thereafter widely prescribed for depression: hepatotoxicity ending its use as an antidepressant in 1961. Isoniazid monotherapy led to the emergence of drug resistant tuberculosis, stimulating new drug development. Vastly increased investment into antidepressants ensued thereafter while investment in new drugs for tuberculosis lagged. In the 21st century, both diseases independently contribute significantly to global disease burdens: renewed convergence and the resultant syndemic is detrimental to both patient groups. Ending the global tuberculosis epidemic and decreasing the burden of depression and will require multidisciplinary, patient-centered approaches that consider this combined co-morbidity. The emerging era of big data for health, digital interventions and novel and repurposed compounds promise new ways to treat both diseases and manage the syndemic, but absence of clinical structures to support these innovations may derail the treatment programs for both. New policies are urgently required optimizing use of the current advances in healthcare available in the digital era, to ensure that patient-centered care takes cognizance of both diseases.
Collapse
Affiliation(s)
- Martie Van Der Walt
- Tuberculosis Platform, South African Medical Research Council, Pretoria, South Africa
| | - Karen H Keddy
- Tuberculosis Platform, South African Medical Research Council, Pretoria, South Africa
| |
Collapse
|
17
|
Differential Immunogenicity and Protective Efficacy Elicited by MTO- and DMT-Adjuvanted CMFO Subunit Vaccines against Mycobacterium tuberculosis Infection. J Immunol Res 2020; 2020:2083793. [PMID: 32953889 PMCID: PMC7487112 DOI: 10.1155/2020/2083793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/01/2020] [Indexed: 12/03/2022] Open
Abstract
Tuberculosis (TB) remains a major and global problem of public health. An effective TB subunit vaccine is urgently needed. Proper selection of the delivery system for the vaccine is crucial for inducing an appropriate immune response tailored to control the target pathogen. In this study, we compared the immunogenicity and protective efficacy of CMFO subunit vaccines against primary progressive TB in two different adjuvant systems: the MTO oil-in-water (O/W) emulsion composed of monophosphoryl lipid A (MPL), trehalose-6,60-dibehenate (TDB), and oil in water emulsion MF59 and the DMT liposome containing dimethyldioctadecylammonium bromide (DDA), monophosphoryl lipid A (MPL), and trehalose-6,60-dibehenate (TDB). Our results demonstrated that the DMT-adjuvanted CMFO could confer more significant protection against M. tuberculosis infection than the CMFO/MTO did in mice. In particular, the adjuvant DMT showed a stronger ability than the O/W emulsion to adjuvant CMFO subunit vaccine and enhanced protection, attributed to elicit Th1-biased responses, strong Th1/Th17 cytokine responses, and IFN-γ+ or IL-2+ T cell responses. Therefore, our findings demonstrate that the liposome delivery system shows more effectiveness to adjuvant TB subunit vaccine than O/W emulsion and highlight the importance of adjuvant formulation for the better efficacy of a protein vaccine.
Collapse
|
18
|
Abstract
Mycobacterium tuberculosis remains the leading cause of death attributed to a single infectious organism. Bacillus Calmette-Guerin (BCG), the standard vaccine against M. tuberculosis, is thought to prevent only 5% of all vaccine-preventable deaths due to tuberculosis, thus an alternative vaccine is required. One of the principal barriers to vaccine development against M. tuberculosis is the complexity of the immune response to infection, with uncertainty as to what constitutes an immunological correlate of protection. In this paper, we seek to give an overview of the immunology of M. tuberculosis infection, and by doing so, investigate possible targets of vaccine development. This encompasses the innate, adaptive, mucosal and humoral immune systems. Though MVA85A did not improve protection compared with BCG alone in a large-scale clinical trial, the correlates of protection this has revealed, in addition to promising results from candidate such as VPM1002, M72/ASO1E and H56:IC31 point to a brighter future in the field of TB vaccine development.
Collapse
Affiliation(s)
- Benedict Brazier
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| |
Collapse
|
19
|
Vasina DV, Kleymenov DA, Manuylov VA, Mazunina EP, Koptev EY, Tukhovskaya EA, Murashev AN, Gintsburg AL, Gushchin VA, Tkachuk AP. First-In-Human Trials of GamTBvac, a Recombinant Subunit Tuberculosis Vaccine Candidate: Safety and Immunogenicity Assessment. Vaccines (Basel) 2019; 7:vaccines7040166. [PMID: 31683812 PMCID: PMC6963980 DOI: 10.3390/vaccines7040166] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis is known to be the biggest global health problem, causing the most deaths by a single infectious agent. Vaccine-development efforts are extremely important. This paper represents the results of the first-in-human trial of recombinant subunit tuberculosis vaccine GamTBvac in a Phase I study. GamTBvac is a new BCG booster candidate vaccine containing dextran-binding domain modified Ag85a and ESAT6-CFP10 MTB antigens and CpG ODN adjuvant, formulated with dextrans. Safety and immunogenicity of GamTBvac were estimated in an open-label clinical trial on 60 Mycobacterium tuberculosis uninfected (MTB-uninfected) volunteers previously-vaccinated with Bacillus Calmette—Guérin vaccine (BCG). The candidate vaccine had an acceptable safety profile and was well-tolerated. Three different vaccine doses with a double-immunization scheme were assessed for immunogenicity and induced a significant increase in IFN-γ in-house IGRA response and IgG ELISA analysis. Among them, the half dose vaccine group (containing DBD-ESAT6-CFP10, 12.5 μg; DBD-Ag85a, 12.5 μg; CpG (ODN 2216), 75 μg; DEAE-Dextran 500 kDa, 250 μg; and Dextran 500 kDa, 5 mg) provided high, early and stable in time immune response specific to both protein antigen fusions and is proposed for the further studies.
Collapse
Affiliation(s)
- Daria V Vasina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Denis A Kleymenov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Victor A Manuylov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena P Mazunina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Egor Yu Koptev
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena A Tukhovskaya
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Arkady N Murashev
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Alexander L Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Infectology Department, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Vladimir A Gushchin
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Artem P Tkachuk
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| |
Collapse
|
20
|
Multiplexed Quantitation of Intraphagocyte Mycobacterium tuberculosis Secreted Protein Effectors. Cell Rep 2019; 23:1072-1084. [PMID: 29694886 PMCID: PMC5946722 DOI: 10.1016/j.celrep.2018.03.125] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/16/2018] [Accepted: 03/26/2018] [Indexed: 01/21/2023] Open
Abstract
The pathogenic potential of Mycobacterium tuberculosis largely depends on ESX secretion systems exporting members of the multigenic Esx, Esp, and PE/PPE protein families. To study the secretion and regulation patterns of these proteins while circumventing immune cross-reactions due to their extensive sequence homologies, we developed an approach that relies on the recognition of their MHC class II epitopes by highly discriminative T cell receptors (TCRs) of a panel of T cell hybridomas. The latter were engineered so that each expresses a unique fluorescent reporter linked to specific antigen recognition. The resulting polychromatic and multiplexed imaging assay enabled us to measure the secretion of mycobacterial effectors inside infected host cells. We applied this novel technology to a large panel of mutants, clinical isolates, and host-cell types to explore the host-mycobacteria interplay and its impact on the intracellular bacterial secretome, which also revealed the unexpected capacity of phagocytes from lung granuloma to present mycobacterial antigens via MHC class II. T cell hybridomas detect individual mycobacterial proteins without cross-reactivity Detection of mycobacterial proteins by T cells allows visualization of their cellular topography Measurement of intraphagocyte mycobacterial proteins can be performed with T cells A multiplexed assay of mycobacterial protein quantitation has numerous applications
Collapse
|
21
|
Kuczkowska K, Øverland L, Rocha SDC, Eijsink VGH, Mathiesen G. Comparison of eight Lactobacillus species for delivery of surface-displayed mycobacterial antigen. Vaccine 2019; 37:6371-6379. [PMID: 31526620 DOI: 10.1016/j.vaccine.2019.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
Abstract
Lactobacillus spp. comprise a large group of Gram-positive lactic acid bacteria with varying physiological, ecological and immunomodulatory properties that are widely exploited by mankind, primarily in food production and as health-promoting probiotics. Recent years have shown increased interest in using lactobacilli for delivery of vaccines, mainly due to their ability to skew the immune system towards pro-inflammatory responses. We have compared the potential of eight Lactobacillus species, L. plantarum, L. brevis, L. curvatus, L. rhamnosus, L. sakei, L. gasseri, L. acidophilus and L. reuteri, as immunogenic carriers of the Ag85B-ESAT-6 antigen from Mycobacterium tuberculosis. Surface-display of the antigen was achieved in L. plantarum, L. brevis, L. gasseri and L. reuteri and these strains were further analyzed in terms of their in vitro and in vivo immunogenicity. All strains activated human dendritic cells in vitro. Immunization of mice using a homologous prime-boost regimen comprising a primary subcutaneous immunization followed by three intranasal boosters, led to slightly elevated IgG levels in serum in most strains, and, importantly, to significantly increased levels of antigen-specific mucosal IgA. Cellular immunity was assessed by studying antigen-specific T cell responses in splenocytes, which did not reveal proliferation as assessed by the expression of Ki67, but which showed clear antigen-specific IFN-γ and IL-17 responses for some of the groups. Taken together, the present results indicate that L. plantarum and L. brevis are the most promising carriers of TB vaccines.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway.
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Sergio D C Rocha
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| |
Collapse
|
22
|
Pooran A, Davids M, Nel A, Shoko A, Blackburn J, Dheda K. IL-4 subverts mycobacterial containment in Mycobacterium tuberculosis-infected human macrophages. Eur Respir J 2019; 54:13993003.02242-2018. [PMID: 31097521 DOI: 10.1183/13993003.02242-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/07/2019] [Indexed: 12/17/2022]
Abstract
Protective immunity against Mycobacterium tuberculosis is poorly understood. The role of interleukin (IL)-4, the archetypal T-helper type 2 (Th2) cytokine, in the immunopathogenesis of human tuberculosis remains unclear.Blood and/or bronchoalveolar lavage fluid (BAL) were obtained from participants with pulmonary tuberculosis (TB) (n=23) and presumed latent TB infection (LTBI) (n=22). Messenger RNA expression levels of interferon (IFN)-γ, IL-4 and its splice variant IL-4δ2 were determined by real-time PCR. The effect of human recombinant (hr)IL-4 on mycobacterial survival/containment (CFU·mL-1) was evaluated in M. tuberculosis-infected macrophages co-cultured with mycobacterial antigen-primed effector T-cells. Regulatory T-cell (Treg) and Th1 cytokine levels were evaluated using flow cytometry.In blood, but not BAL, IL-4 mRNA levels (p=0.02) and the IL-4/IFN-γ ratio (p=0.01) was higher in TB versus LTBI. hrIL-4 reduced mycobacterial containment in infected macrophages (p<0.008) in a dose-dependent manner and was associated with an increase in Tregs (p<0.001), but decreased CD4+Th1 cytokine levels (CD4+IFN-γ+ p<0.001; CD4+TNFα+ p=0.01). Blocking IL-4 significantly neutralised mycobacterial containment (p=0.03), CD4+IFNγ+ levels (p=0.03) and Treg expression (p=0.03).IL-4 can subvert mycobacterial containment in human macrophages, probably via perturbations in Treg and Th1-linked pathways. These data may have implications for the design of effective TB vaccines and host-directed therapies.
Collapse
Affiliation(s)
- Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Dept of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Malika Davids
- Centre for Lung Infection and Immunity, Division of Pulmonology, Dept of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Andrew Nel
- Dept of Integrative Biomedical Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Aubrey Shoko
- Centre for Proteomics and Genomics Research, Cape Town, South Africa
| | - Jonathan Blackburn
- Dept of Integrative Biomedical Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Dept of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa .,Faculty of Infectious and Tropical Diseases, Dept of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
23
|
Kuczkowska K, Copland A, Øverland L, Mathiesen G, Tran AC, Paul MJ, Eijsink VGH, Reljic R. Inactivated Lactobacillus plantarum Carrying a Surface-Displayed Ag85B-ESAT-6 Fusion Antigen as a Booster Vaccine Against Mycobacterium tuberculosis Infection. Front Immunol 2019; 10:1588. [PMID: 31354727 PMCID: PMC6632704 DOI: 10.3389/fimmu.2019.01588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022] Open
Abstract
Vaccination is considered the most effective strategy for controlling tuberculosis (TB). The existing vaccine, the Bacille Calmette-Guérin (BCG), although partially protective, has a number of limitations. Therefore, there is a need for developing new TB vaccines and several strategies are currently exploited including the use of viral and bacterial delivery vectors. We have previously shown that Lactobacillus plantarum (Lp) producing Ag85B and ESAT-6 antigens fused to a dendritic cell-targeting peptide (referred to as Lp_DC) induced specific immune responses in mice. Here, we analyzed the ability of two Lp-based vaccines, Lp_DC and Lp_HBD (in which the DC-binding peptide was replaced by an HBD-domain directing the antigen to non-phagocytic cells) to activate antigen-presenting cells, induce specific immunity and protect mice from Mycobacterium tuberculosis infection. We tested two strategies: (i) Lp as BCG boosting vaccine (a heterologous regimen comprising parenteral BCG immunization followed by intranasal Lp boost), and (ii) Lp as primary vaccine (a homologous regimen including subcutaneous priming followed by intranasal boost). The results showed that both Lp constructs applied as a BCG boost induced specific cellular immunity, manifested in T cell proliferation, antigen-specific IFN-γ responses and multifunctional T cells phenotypes. More importantly, intranasal boost with Lp_DC or Lp_HBD enhanced protection offered by BCG, as shown by reduced M. tuberculosis counts in lungs. These findings suggest that Lp constructs could be developed as a potential mucosal vaccine platform against mycobacterial infections.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Alastair Copland
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom.,College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Andy C Tran
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Mathew J Paul
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| |
Collapse
|
24
|
Lyadova I, Nikitina I. Cell Differentiation Degree as a Factor Determining the Role for Different T-Helper Populations in Tuberculosis Protection. Front Immunol 2019; 10:972. [PMID: 31134070 PMCID: PMC6517507 DOI: 10.3389/fimmu.2019.00972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/16/2019] [Indexed: 12/15/2022] Open
Abstract
Efficient tuberculosis (TB) control depends on early TB prediction and prevention. Solution to these tasks requires knowledge of TB protection correlates (TB CoPs), i.e., laboratory markers that are mechanistically involved in the protection and which allow to determine how well an individual is protected against TB or how efficient the candidate TB vaccine is. The search for TB CoPs has been largely focused on different T-helper populations, however, the data are controversial, and no reliable CoPs are still known. Here we discuss the role of different T-helper populations in TB protection focusing predominantly on Th17, “non-classical” Th1 (Th1*) and “classical” Th1 (cTh1) populations. We analyze how these populations differ besides their effector activity and suggest the hypothesis that: (i) links the protective potential of Th17, Th1*, and cTh1 to their differentiation degree and plasticity; (ii) implies different roles of these populations in response to vaccination, latent TB infection (LTBI), and active TB. One of the clinically relevant outcomes of this hypothesis is that over-stimulating T cells during vaccination and biasing T cell response toward the preferential generation of Th1 are not beneficial. The review sheds new light on the problem of TB CoPs and will help develop better strategies for TB control.
Collapse
Affiliation(s)
- Irina Lyadova
- Laboratory of Cellular and Molecular Mechanisms of Histogenesis, Koltsov Institute of Developmental Biology, Moscow, Russia.,Laboratory of Biotechnology, Department of Immunology, Central Tuberculosis Research Institute, Moscow, Russia
| | - Irina Nikitina
- Laboratory of Cellular and Molecular Mechanisms of Histogenesis, Koltsov Institute of Developmental Biology, Moscow, Russia.,Laboratory of Biotechnology, Department of Immunology, Central Tuberculosis Research Institute, Moscow, Russia
| |
Collapse
|
25
|
Prabowo SA, Painter H, Zelmer A, Smith SG, Seifert K, Amat M, Cardona PJ, Fletcher HA. RUTI Vaccination Enhances Inhibition of Mycobacterial Growth ex vivo and Induces a Shift of Monocyte Phenotype in Mice. Front Immunol 2019; 10:894. [PMID: 31114572 PMCID: PMC6503078 DOI: 10.3389/fimmu.2019.00894] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/08/2019] [Indexed: 12/17/2022] Open
Abstract
Tuberculosis (TB) is a major global health problem and there is a dire need for an improved treatment. A strategy to combine vaccination with drug treatment, termed therapeutic vaccination, is expected to provide benefit in shortening treatment duration and augmenting treatment success rate. RUTI candidate vaccine has been specifically developed as a therapeutic vaccine for TB. The vaccine is shown to reduce bacillary load when administered after chemotherapy in murine and guinea pig models, and is also immunogenic when given to healthy adults and individuals with latent TB. In the absence of a validated correlate of vaccine-induced protection for TB vaccine testing, mycobacterial growth inhibition assay (MGIA) has been developed as a comprehensive tool to evaluate vaccine potency ex vivo. In this study, we investigated the potential of RUTI vaccine to control mycobacterial growth ex vivo and demonstrated the capacity of MGIA to aid the identification of essential immune mechanism. We found an association between the peak response of vaccine-induced growth inhibition and a shift in monocyte phenotype following RUTI vaccination in healthy mice. The vaccination significantly increased the frequency of non-classical Ly6C− monocytes in the spleen after two doses of RUTI. Furthermore, mRNA expressions of Ly6C−-related transcripts (Nr4a1, Itgax, Pparg, Bcl2) were upregulated at the peak vaccine response. This is the first time the impact of RUTI has been assessed on monocyte phenotype. Given that non-classical Ly6C− monocytes are considered to play an anti-inflammatory role, our findings in conjunction with previous studies have demonstrated that RUTI could induce a balanced immune response, promoting an effective cell-mediated response whilst at the same time limiting excessive inflammation. On the other hand, the impact of RUTI on non-classical monocytes could also reflect its impact on trained innate immunity which warrants further investigation. In summary, we have demonstrated a novel mechanism of action of the RUTI vaccine, which suggests the importance of a balanced M1/M2 monocyte function in controlling mycobacterial infection. The MGIA could be used as a screening tool for therapeutic TB vaccine candidates and may aid the development of therapeutic vaccination regimens for TB in the near future.
Collapse
Affiliation(s)
- Satria A Prabowo
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Hannah Painter
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Andrea Zelmer
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Steven G Smith
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Karin Seifert
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Pere-Joan Cardona
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona (UAB), Badalona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Helen A Fletcher
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
26
|
Sarkar I, Garg R, van Drunen Littel-van den Hurk S. Selection of adjuvants for vaccines targeting specific pathogens. Expert Rev Vaccines 2019; 18:505-521. [PMID: 31009255 PMCID: PMC7103699 DOI: 10.1080/14760584.2019.1604231] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Adjuvants form an integral component in most of the inactivated and subunit vaccine formulations. Careful and proper selection of adjuvants helps in promoting appropriate immune responses against target pathogens at both innate and adaptive levels such that protective immunity can be elicited. Areas covered: Herein, we describe the recent progress in our understanding of the mode of action of adjuvants that are licensed for use in human vaccines or in clinical or pre-clinical stages at both innate and adaptive levels. Different pathogens have distinct characteristics, which require the host to mount an appropriate immune response against them. Adjuvants can be selected to elicit a tailor-made immune response to specific pathogens based on their unique properties. Identification of biomarkers of adjuvanticity for several candidate vaccines using omics-based technologies can unravel the mechanism of action of modern and experimental adjuvants. Expert opinion: Adjuvant technology has been revolutionized over the last two decades. In-depth understanding of the role of adjuvants in activating the innate immune system, combined with systems vaccinology approaches, have led to the development of next-generation, novel adjuvants that can be used in vaccines against challenging pathogens and in specific target populations.
Collapse
Affiliation(s)
- Indranil Sarkar
- a VIDO-InterVac , University of Saskatchewan , Saskatoon , Canada.,b Microbiology and Immunology , University of Saskatchewan , Saskatoon , Canada
| | - Ravendra Garg
- a VIDO-InterVac , University of Saskatchewan , Saskatoon , Canada
| | | |
Collapse
|
27
|
Prabowo SA, Zelmer A, Stockdale L, Ojha U, Smith SG, Seifert K, Fletcher HA. Historical BCG vaccination combined with drug treatment enhances inhibition of mycobacterial growth ex vivo in human peripheral blood cells. Sci Rep 2019; 9:4842. [PMID: 30890730 PMCID: PMC6425030 DOI: 10.1038/s41598-019-41008-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 02/20/2019] [Indexed: 12/24/2022] Open
Abstract
Tuberculosis (TB) is a leading infectious cause of death globally. Drug treatment and vaccination, in particular with Bacillus Calmette-Guérin (BCG), remain the main strategies to control TB. With the emergence of drug resistance, it has been proposed that a combination of TB vaccination with pharmacological treatment may provide a greater therapeutic value. We implemented an ex vivo mycobacterial growth inhibition assay (MGIA) to discriminate vaccine responses in historically BCG-vaccinated human volunteers and to assess the contribution of vaccine-mediated immune response towards the killing effect of mycobacteria in the presence of the antibiotics isoniazid (INH) and rifampicin (RIF), in an attempt to develop the assay as a screening tool for therapeutic TB vaccines. BCG vaccination significantly enhanced the ability of INH to control mycobacterial growth ex vivo. The BCG-vaccinated group displayed a higher production of IFN-γ and IP-10 when peripheral blood mononuclear cells (PBMC) were co-cultured with INH, with a similar trend during co-culture with RIF. A higher frequency of IFN-γ+ and TNF-α+ CD3- CD4- CD8- cells was observed, suggesting the contribution of Natural Killer (NK) cells in the combined effect between BCG vaccination and INH. Taken together, our data indicate the efficacy of INH can be augmented following historical BCG vaccination, which support findings from previous observational and animal studies.
Collapse
Affiliation(s)
- Satria A Prabowo
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Andrea Zelmer
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Lisa Stockdale
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Utkarsh Ojha
- Faculty of Medicine, Imperial College School of Medicine, Imperial College London, London, UK
| | - Steven G Smith
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Karin Seifert
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Helen A Fletcher
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
- Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
28
|
Burggraaf MJ, Ates LS, Speer A, van der Kuij K, Kuijl C, Bitter W. Optimization of secretion and surface localization of heterologous OVA protein in mycobacteria by using LipY as a carrier. Microb Cell Fact 2019; 18:44. [PMID: 30841891 PMCID: PMC6402100 DOI: 10.1186/s12934-019-1093-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/25/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mycobacterium bovis Bacille Calmette-Guérin (BCG) is not only used as a vaccine against tuberculosis but also protects against leprosy and is used as part of bladder cancer treatment to induce a protective immune response. However, protection by BCG vaccination is not optimal. To improve vaccine efficacy, recombinant BCG expressing heterologous antigens has been put forward to elicit antigen-specific cellular and humoral responses. Cell surface localized or secreted antigens induce better immune responses than their cytosolic counterparts. Optimizing secretion of heterologous proteins or protein fragments holds therefore unexplored potential for improving the efficacy of recombinant BCG vaccine candidates. Secretion of heterologous antigens requires crossing the mycobacterial inner and outer membrane. Mycobacteria have specialized ESX or type VII secretion systems that enable translocation of proteins across both membranes. Probing this secretion system could therefore be a valid approach to surface localize heterologous antigens. RESULTS We show that ESX-5 substrate LipY, a lipase, can be used as a carrier for heterologous secretion of an ovalbumin fragment (OVA). LipY contains a PE domain and a lipase domain, separated by a linker region. This linker domain is processed upon secretion. Fusion of the PE and linker domains of LipY to OVA enabled ESX-5-dependent secretion of the fusion construct LipY-OVA in M. marinum, albeit with low efficiency. Subsequent random mutagenesis of LipY-OVA and screening for increased secretion resulted in mutants with improved heterologous secretion. Detailed analysis identified two mutations in OVA that improved secretion, i.e. an L280P mutation and a protein-extending frameshift mutation. Finally, deletion of the linker domain of LipY enhanced secretion of LipY-OVA, although this mutation also reduced surface association. Further analysis in wild type LipY showed that the linker domain is required for surface association. CONCLUSION We show that the ESX-5 system can be used for heterologous secretion. Furthermore, minor mutations in the substrate can enhance secretion. Especially the C-terminal region seems to be important for this. The linker domain of LipY is involved in surface association. These findings show that non-biased screening approaches aid in optimization of heterologous secretion, which can contribute to heterologous vaccine development.
Collapse
Affiliation(s)
- Maroeska J Burggraaf
- Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Louis S Ates
- Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands
| | - Alexander Speer
- Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Kim van der Kuij
- Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Coen Kuijl
- Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Wilbert Bitter
- Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands. .,Molecular Microbiology, Vrije Universiteit Amsterdam, de Boelelaan 1105, Amsterdam, Netherlands.
| |
Collapse
|
29
|
Ruhwald M, Andersen PL, Schrager L. Towards a new vaccine for tuberculosis. Tuberculosis (Edinb) 2018. [DOI: 10.1183/2312508x.10022417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
|
31
|
AlMatar M, Makky EA, AlMandeal H, Eker E, Kayar B, Var I, Köksal F. Does the Development of Vaccines Advance Solutions for Tuberculosis? Curr Mol Pharmacol 2018; 12:83-104. [PMID: 30474542 DOI: 10.2174/1874467212666181126151948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/06/2018] [Accepted: 10/17/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mycobacterium tuberculosis (Mtb) is considered as one of the most efficacious human pathogens. The global mortality rate of TB stands at approximately 2 million, while about 8 to 10 million active new cases are documented yearly. It is, therefore, a priority to develop vaccines that will prevent active TB. The vaccines currently used for the management of TB can only proffer a certain level of protection against meningitis, TB, and other forms of disseminated TB in children; however, their effectiveness against pulmonary TB varies and cannot provide life-long protective immunity. Based on these reasons, more efforts are channeled towards the development of new TB vaccines. During the development of TB vaccines, a major challenge has always been the lack of diversity in both the antigens contained in TB vaccines and the immune responses of the TB sufferers. Current efforts are channeled on widening both the range of antigens selection and the range of immune response elicited by the vaccines. The past two decades witnessed a significant progress in the development of TB vaccines; some of the discovered TB vaccines have recently even completed the third phase (phase III) of a clinical trial. OBJECTIVE The objectives of this article are to discuss the recent progress in the development of new vaccines against TB; to provide an insight on the mechanism of vaccine-mediated specific immune response stimulation, and to debate on the interaction between vaccines and global interventions to end TB.
Collapse
Affiliation(s)
- Manaf AlMatar
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitusu) Cukurova University, Adana, Turkey
| | - Essam A Makky
- Department of Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), Kuantan, Malaysia
| | - Husam AlMandeal
- Freiburg Universität, Moltkestraße 90, 76133 karlsruhe Augenklinik, Germany
| | - Emel Eker
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Begüm Kayar
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Işıl Var
- Department of Food Engineering, Agricultural Faculty, Cukurova University, Adana, Turkey
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
32
|
Warimwe GM, Purushotham J, Perry BD, Hill AV, Gilbert SC, Dungu B, Charleston B. Tackling human and animal health threats through innovative vaccinology in Africa. AAS Open Res 2018; 1:18. [PMID: 32259020 PMCID: PMC7118973 DOI: 10.12688/aasopenres.12877.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2018] [Indexed: 07/27/2023] Open
Abstract
Africa bears the brunt of many of the world's most devastating human and animal infectious diseases, a good number of which have no licensed or effective vaccines available. The continent's potential to generate novel interventions against these global health threats is however largely untapped. Strengthening Africa's vaccine research and development (R&D) sector could accelerate discovery, development and deployment of effective countermeasures against locally prevalent infectious diseases, many of which are neglected and have the capacity to spread to new geographical settings. Here, we review Africa's human and veterinary vaccine R&D sectors and identify key areas that should be prioritized for investment, and synergies that could be exploited from Africa's veterinary vaccine industry, which is surprisingly strong and has close parallels with human vaccine R&D.
Collapse
Affiliation(s)
- George M. Warimwe
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- The Pirbright Institute, Woking, GU24 0NF, UK
| | | | - Brian D. Perry
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | | | | | | | | |
Collapse
|
33
|
Warimwe GM, Purushotham J, Perry BD, Hill AVS, Gilbert SC, Dungu B, Charleston B. Tackling human and animal health threats through innovative vaccinology in Africa. AAS Open Res 2018; 1:18. [PMID: 32259020 PMCID: PMC7118973 DOI: 10.12688/aasopenres.12877.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2018] [Indexed: 11/20/2022] Open
Abstract
Africa bears the brunt of many of the world's most devastating human and animal infectious diseases, a good number of which have no licensed or effective vaccines available. The continent's potential to generate novel interventions against these global health threats is however largely untapped. Strengthening Africa's vaccine research and development (R&D) sector could accelerate discovery, development and deployment of effective countermeasures against locally prevalent infectious diseases, many of which are neglected and have the capacity to spread to new geographical settings. Here, we review Africa's human and veterinary vaccine R&D sectors and identify key areas that should be prioritized for investment, and synergies that could be exploited from Africa's veterinary vaccine industry, which is surprisingly strong and has close parallels with human vaccine R&D.
Collapse
Affiliation(s)
- George M Warimwe
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.,The Pirbright Institute, Woking, GU24 0NF, UK
| | | | - Brian D Perry
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | | | | |
Collapse
|
34
|
Kilpeläinen A, Maya-Hoyos M, Saubí N, Soto CY, Joseph Munne J. Advances and challenges in recombinant Mycobacterium bovis BCG-based HIV vaccine development: lessons learned. Expert Rev Vaccines 2018; 17:1005-1020. [PMID: 30300040 DOI: 10.1080/14760584.2018.1534588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Human Immunodeficiency Virus/Acquired Immune Deficiency Syndrome, tuberculosis, and malaria are responsible for most human deaths produced by infectious diseases worldwide. Vaccination against HIV requires generation of memory T cells and neutralizing antibodies, mucosal immunity, and stimulation of an innate immune responses. In this context, the use of Mycobacterium bovis bacillus Calmette-Guérin (BCG) as a live vaccine vehicle is a promising approach for T-cell induction. AREAS COVERED In this review, we provide a comprehensive summary of the literature regarding immunogenicity studies in animal models performed since 2005. Furthermore, we provide expert commentary and 5-year view on how the development of potential recombinant BCG-based HIV vaccines involves careful selection of the HIV antigen, expression vectors, promoters, BCG strain, preclinical animal models, influence of preexisting immunity, and safety issues, for the rational design of recombinant BCG:HIV vaccines to prevent HIV transmission in the general population. EXPERT COMMENTARY The three critical issues to be considered when developing a rBCG:HIV vaccine are codon optimization, antigen localization, and plasmid stability in vivo. The use of integrative expression vectors are likely to improve the mycobacterial vaccine stability and immunogenicity to develop not only recombinant BCG-based vaccines expressing second generation of HIV-1 immunogens but also other major pediatric pathogens to prime protective responses shortly following birth.
Collapse
Affiliation(s)
- Athina Kilpeläinen
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Milena Maya-Hoyos
- b Chemistry Department, Faculty of Sciences , Universidad Nacional de Colombia, Ciudad Universitaria , Bogotá , Colombia
| | - Narcís Saubí
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Carlos Y Soto
- b Chemistry Department, Faculty of Sciences , Universidad Nacional de Colombia, Ciudad Universitaria , Bogotá , Colombia
| | - Joan Joseph Munne
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| |
Collapse
|
35
|
Gupta T, LaGatta M, Helms S, Pavlicek RL, Owino SO, Sakamoto K, Nagy T, Harvey SB, Papania M, Ledden S, Schultz KT, McCombs C, Quinn FD, Karls RK. Evaluation of a temperature-restricted, mucosal tuberculosis vaccine in guinea pigs. Tuberculosis (Edinb) 2018; 113:179-188. [PMID: 30514501 DOI: 10.1016/j.tube.2018.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 12/13/2022]
Abstract
Tuberculosis (TB) is currently the leading cause of death in humans by a single infectious agent, Mycobacterium tuberculosis. The Bacillus Calmette-Guérin (BCG) vaccine prevents pulmonary TB with variable efficacy, but can cause life-threatening systemic infection in HIV-infected infants. In this study, TBvac85, a derivative of Mycobacterium shottsii expressing M. tuberculosis Antigen 85B, was examined as a safer alternative to BCG. Intranasal vaccination of guinea pigs with TBvac85, a naturally temperature-restricted species, resulted in serum Ag85B-specific IgG antibodies. Delivery of the vaccine by this route also induced protection equivalent to intradermal BCG based on organ bacterial burdens and lung pathology six weeks after aerosol challenge with M. tuberculosis strain Erdman. These results support the potential of TBvac85 as the basis of an effective TB vaccine. Next-generation derivatives expressing multiple M. tuberculosis immunogens are in development.
Collapse
Affiliation(s)
- Tuhina Gupta
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Monica LaGatta
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Pathens, Inc., Athens, GA, USA
| | - Shelly Helms
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Rebecca L Pavlicek
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Simon O Owino
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Pathens, Inc., Athens, GA, USA
| | - Kaori Sakamoto
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Stephen B Harvey
- Animal Resources Program, University of Georgia, Athens, GA, USA; Department of Population Heath, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Mark Papania
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stephanie Ledden
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | | | | - Frederick D Quinn
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Pathens, Inc., Athens, GA, USA
| | - Russell K Karls
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Pathens, Inc., Athens, GA, USA.
| |
Collapse
|
36
|
Kimuda SG, Biraro IA, Bagaya BS, Raynes JG, Cose S. Characterising antibody avidity in individuals of varied Mycobacterium tuberculosis infection status using surface plasmon resonance. PLoS One 2018; 13:e0205102. [PMID: 30312318 PMCID: PMC6185725 DOI: 10.1371/journal.pone.0205102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/19/2018] [Indexed: 12/20/2022] Open
Abstract
There is increasing evidence supporting a role for antibodies in protection against tuberculosis (TB), with functional antibodies being described in the latent state of TB infection. Antibody avidity is an important determinant of antibody-mediated protection. This study characterised the avidity of antibodies against Ag85A, an immunodominant Mycobacterium tuberculosis (M.tb) antigen and constituent of several anti-TB vaccine candidates, in individuals of varied M.tb infection status. Avidity of Ag85A specific antibodies was measured in 30 uninfected controls, 34 individuals with latent TB infection (LTBI) and 75 active pulmonary TB (APTB) cases, employing the more commonly used chaotrope-based dissociation assays, and surface plasmon resonance (SPR). Chaotrope-based assays indicated that APTB was associated with a higher antibody avidity index compared to uninfected controls [adjusted geometric mean ratio (GMR): 1.641, 95% confidence interval (CI): 1.153, 2.337, p = 0.006, q = 0.018] and to individuals with LTBI [adjusted GMR: 1.604, 95% CI: 1.282, 2.006, p < 0.001, q <0.001]. SPR assays showed that APTB was associated with slower dissociation rates, an indication of higher avidity, compared to uninfected controls (adjusted GMR: 0.796, 95% CI: 0.681, 0.932, p = 0.004, q = 0.012) and there was also weak evidence of more avid antibodies in the LTBI compared to the uninfected controls (adjusted GMR: 0.871, 95% CI: 0.763, 0.994, p = 0.041, q = 0.123). We found no statistically significant differences in anti-Ag85A antibody avidity between the APTB and LTBI groups. This study shows that antibodies of increased avidity are generated against a principle vaccine antigen in M.tb infected individuals. It would be important to determine whether TB vaccines are able to elicit a similar response. Additionally, more research is needed to determine whether antibody avidity is important in protection against infection and disease.
Collapse
Affiliation(s)
- Simon G. Kimuda
- Department of Medical Microbiology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Immunomodulation and Vaccines Programme, Medical Research Council/ Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Irene Andia Biraro
- Immunomodulation and Vaccines Programme, Medical Research Council/ Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Internal Medicine, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Bernard S. Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - John G. Raynes
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Stephen Cose
- Department of Medical Microbiology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Immunomodulation and Vaccines Programme, Medical Research Council/ Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
MARTINI M, BESOZZI G, BARBERIS I. The never-ending story of the fight against tuberculosis: from Koch's bacillus to global control programs. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2018; 59:E241-E247. [PMID: 30397682 PMCID: PMC6196368 DOI: 10.15167/2421-4248/jpmh2018.59.3.1051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/29/2018] [Indexed: 11/24/2022]
Abstract
Tuberculosis (TB) is one of the oldest diseases known to affect humanity, and is still a major public health problem. It is caused by the bacillus Mycobacterium tuberculosis (MT), isolated in 1882 by Robert Koch. Until the 1950s, X rays were used as a cheap method of diagnostic screening together with the tuberculin skin sensitivity test. In the diagnosis and treatment of TB, an important role was also played by surgery. The late Nineteenth century saw the introduction of the tuberculosis sanatorium, which proved to be one of the first useful measures against TB. Subsequently, Albert Calmette and Camille Guérin used a non-virulent MT strain to produce a live attenuated vaccine. In the 1980s and 1990s, the incidence of tuberculosis surged as a major opportunistic infection in people with HIV infection and AIDS; for this reason, a combined strategy based on improving drug treatment, diagnostic instruments and prevention was needed.
Collapse
Affiliation(s)
- M. MARTINI
- University of Genoa, Department of Health Sciences, Section of Medical History and Ethics, Genoa, Italy
- UNESCO CHAIR Anthropology of Health - Biosphere and Healing System, University of Genoa, Italy
| | - G. BESOZZI
- Centro di Formazione TB Italia Onlus
- Istituto Villa Marelli, Milano
| | - I. BARBERIS
- University of Genoa, Department of Health Sciences, Section of Medical History and Ethics, Genoa, Italy
- * Correspondence: Ilaria Barberis, University of Genoa, Department of Health Sciences, Section of Medical History and Ethics, largo R. Benzi 10 Pad 3, 16132 Genoa, Italy - Tel./Fax +39 010 353 85 02 - E-mail:
| |
Collapse
|
38
|
Huggins MA, Jameson SC, Hamilton SE. Embracing microbial exposure in mouse research. J Leukoc Biol 2018; 105:73-79. [PMID: 30260516 DOI: 10.1002/jlb.4ri0718-273r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 01/06/2023] Open
Abstract
Research using mouse models have contributed essential knowledge toward our current understanding of how the human immune system functions. One key difference between humans and typical laboratory mice, however, is exposure to pathogens in their respective environments. Several recent studies have highlighted that these microbial encounters shape the development and functional status of the immune system. For humans, such numerous and unavoidable encounters with viruses, bacteria, and parasites may be a defining factor in generating a healthy and robust immune system, poised to respond to new infections and to vaccination. Additionally, the commensal organisms that make up the host microbiome also change with environment and impact the immune response. Hence, there is a pressing need to generate more faithful mouse models that reflect the natural state of the human immune system. This review explores the use of new experimental mouse models designed to better understand how host-microbial interactions shape the immune response. By embracing these technologies to complement traditional mouse models, researchers can remove a significant barrier that has long separated murine and human immunologists.
Collapse
Affiliation(s)
- Mathew A Huggins
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara E Hamilton
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
39
|
Hiza H, Fenner L, Hella J, Kuchaka D, Sasamalo M, Blauenfeldt T, Kibiki G, Kavishe RA, Mhimbira F, Ruhwald M. Boosting effect of IL-7 in interferon gamma release assays to diagnose Mycobacterium tuberculosis infection. PLoS One 2018; 13:e0202525. [PMID: 30157233 PMCID: PMC6114790 DOI: 10.1371/journal.pone.0202525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/03/2018] [Indexed: 11/18/2022] Open
Abstract
Background A quarter of the world’s population is estimated to be infected with Myobacterium tuberculosis (Mtb). Infection is detected by immune response to M. tuberculosis antigens using either tuberculin skin test (TST) and interferon gamma release (IGRA’s), tests which have low sensitivity in immunocompromised. IL-7 is an important cytokine for T-cell function with potential to augment cytokine release in in-vitro assays. This study aimed to determine whether the addition of IL-7 in interferon-gamma release assays (IGRAs) improves its diagnostic performance of Mtb infection. Methods 44 cases with confirmed TB and 45 household contacts without TB were recruited and 1ml of blood was stimulated in two separate IGRA’s tube set: one set of standard Quantiferon TB gold tubes mitogen, TB antigen and TB Nil; one set of customized Quantiferon TB gold tubes with added IL-7. Following IFN-γ and IP-10 release was determined using ELISA. Results We found that the addition of IL-7 led to significantly higher release of IFN-γ in individuals with active TB from 4.2IU/ml (IQR 1.4–6.9IU/ml) to 5.1IU/ml (IQR 1.5–8.1IU/ml, p = 0.0057), and we found an indication of a lower release of both IFN-γ and IP-10 in participants with negative tests. Conclusions In TB cases addition of IL-7 in IGRA tubes augments IFN-γ but not IP-10 release, and seems to lower the response in controls. Whether IL-7 boosted IGRA holds potential over standard IGRA needs to be confirmed in larger studies in high and low TB incidence countries.
Collapse
Affiliation(s)
- Hellen Hiza
- Ifakara Health Institute, Bagamoyo, Tanzania
| | - Lukas Fenner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Jerry Hella
- Ifakara Health Institute, Bagamoyo, Tanzania
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Davis Kuchaka
- Kilimanjaro Clinical Research Institute, Kilimanjaro, Tanzania
| | - Mohamed Sasamalo
- Ifakara Health Institute, Bagamoyo, Tanzania
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Thomas Blauenfeldt
- Statens Serum Institut, Center for Vaccine Research, Copenhagen, Denmark
| | - Gibson Kibiki
- Kilimanjaro Clinical Research Institute, Kilimanjaro, Tanzania
- East African Health Research Commission, Bujumbura, Burundi
| | - Reginald A Kavishe
- Kilimanjaro Clinical Research Institute, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical University College, Tumaini University, Kilimanjaro, Tanzania
| | | | - Morten Ruhwald
- Statens Serum Institut, Center for Vaccine Research, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
40
|
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: 7] [Impact Index Per Article: 1.0] [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
|
41
|
de Paula Oliveira Santos B, Trentini MM, Machado RB, Rúbia Nunes Celes M, Kipnis A, Petrovsky N, Junqueira-Kipnis AP. Advax4 delta inulin combination adjuvant together with ECMX, a fusion construct of four protective mTB antigens, induces a potent Th1 immune response and protects mice against Mycobacterium tuberculosis infection. Hum Vaccin Immunother 2018; 13:2967-2976. [PMID: 28937879 DOI: 10.1080/21645515.2017.1368598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Tuberculosis (TB) remains a main public health concern and 10.4 million new cases occurred in 2015 around the world. BCG is the only approved vaccine against TB, but has variable efficacy and new vaccines are needed. We developed two new mTB vaccine candidates based on the recombinant fusion proteins, rCMX and rECMX formulated with Advax4, a new combination adjuvant combining delta inulin, CpG oligonucleotide and murabutide. BALB/c mice were immunized three times intramuscularly with these vaccine formulations. Injection of Advax4 alone increased the percentage of lymphatic endothelial cells and activated macrophages (F480/CD11b+) in the draining lymph nodes consistent with a chemotactic adjuvant effect. Advax4+CMX and Advax4+ECMX induced the highest levels of IgG1 and IgG2a antibodies against rCMX and rECMX, respectively. Immunized mice challenged with Mycobacterium tuberculosis (Mtb) had increased vaccine-specific Th1 responses in the lungs together with reduced Mtb - associated alveolar damage, although only the Advax4+ECMX vaccine demonstrated significant reduction of lung bacterial load. This study confirmed Advax4+ECMX as a potential TB vaccine candidate, with potential for further optimization and clinical development.
Collapse
Affiliation(s)
- Bruno de Paula Oliveira Santos
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Monalisa Martins Trentini
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Renato Beilner Machado
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Mara Rúbia Nunes Celes
- b Laboratory of Pathology, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - André Kipnis
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| | - Nikolai Petrovsky
- c Flinders University and Vaxine Pty Ltd, Flinders Medical Center , Adelaide , Australia
| | - Ana Paula Junqueira-Kipnis
- a Laboratory of Immunopathology of Infectious Diseases, Department of Microbiology, Immunology, Parasitology, and Pathology, Tropical Institute of Pathology and Public Health , Federal University of Goiás , Goiás , Brazil
| |
Collapse
|
42
|
Ates LS, Sayes F, Frigui W, Ummels R, Damen MPM, Bottai D, Behr MA, van Heijst JWJ, Bitter W, Majlessi L, Brosch R. RD5-mediated lack of PE_PGRS and PPE-MPTR export in BCG vaccine strains results in strong reduction of antigenic repertoire but little impact on protection. PLoS Pathog 2018; 14:e1007139. [PMID: 29912964 PMCID: PMC6023246 DOI: 10.1371/journal.ppat.1007139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/28/2018] [Accepted: 06/05/2018] [Indexed: 01/03/2023] Open
Abstract
Tuberculosis is the deadliest infectious disease worldwide. Although the BCG vaccine is widely used, it does not efficiently protect against pulmonary tuberculosis and an improved tuberculosis vaccine is therefore urgently needed. Mycobacterium tuberculosis uses different ESX/Type VII secretion (T7S) systems to transport proteins important for virulence and host immune responses. We recently reported that secretion of T7S substrates belonging to the mycobacteria-specific Pro-Glu (PE) and Pro-Pro-Glu (PPE) proteins of the PGRS (polymorphic GC-rich sequences) and MPTR (major polymorphic tandem repeat) subfamilies required both a functional ESX-5 system and a functional PPE38/71 protein for secretion. Inactivation of ppe38/71 and the resulting loss of PE_PGRS/PPE-MPTR secretion were linked to increased virulence of M. tuberculosis strains. Here, we show that a predicted total of 89 PE_PGRS/PPE-MPTR surface proteins are not exported by certain animal-adapted strains of the M. tuberculosis complex including M. bovis. This Δppe38/71-associated secretion defect therefore also occurs in the M. bovis-derived tuberculosis vaccine BCG and could be partially restored by introduction of the M. tuberculosis ppe38-locus. Epitope mapping of the PPE-MPTR protein PPE10, further allowed us to monitor T-cell responses in splenocytes from BCG/M. tuberculosis immunized mice, confirming the dependence of PPE10-specific immune-induction on ESX-5/PPE38-mediated secretion. Restoration of PE_PGRS/PPE-MPTR secretion in recombinant BCG neither altered global antigenic presentation or activation of innate immune cells, nor protective efficacy in two different mouse vaccination-infection models. This unexpected finding stimulates a reassessment of the immunomodulatory properties of PE_PGRS/PPE-MPTR proteins, some of which are contained in vaccine formulations currently in clinical evaluation. One of the major findings of the pioneering Mycobacterium tuberculosis H37Rv genome sequencing project was the identification of the highly abundant PE and PPE proteins, named after their N-terminal motifs Pro–Glu (PE) or Pro–Pro–Glu (PPE). Within the 20 years of research since then, many claims were made that PE/PPE proteins, including the two large subgroups encoded by repetitive sequences with very high GC content (PE_PGRS and PPE-MPTR families), are exported to the bacterial surface or beyond, and show broad immunomodulatory impact on host-pathogen interaction. We thus screened strains from different branches of the M. tuberculosis complex, including the attenuated Mycobacterium bovis BCG vaccine strains, for their capacity to export PE_PGRS/PPE-MPTR proteins. Strikingly, we found that BCG strains were unable to export the plethora of PE_PGRS/PPE-MPTR proteins due to the absence of the region of difference RD5, which in M. tuberculosis encodes PPE38, required for PE_PGRS/PPE-MPTR export. Surprisingly, the restoration of PE_PGRS/PPE-MPTR export by genetic complementation in recombinant BCG did not result in immunomodulatory changes or altered protection in mouse models. Our results thus put into perspective the numerous reports on virulence-associated immunomodulatory impact of individual PE_PGRS and PPE-MPTR proteins and open novel questions on their biological function(s).
Collapse
Affiliation(s)
- Louis S. Ates
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR3525, Paris, France
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- * E-mail: (LSA); (RB)
| | - Fadel Sayes
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR3525, Paris, France
| | - Wafa Frigui
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR3525, Paris, France
| | - Roy Ummels
- Department of Medical Microbiology and Infection Prevention, VU University Medical Center, Amsterdam, the Netherlands
| | - Merel P. M. Damen
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR3525, Paris, France
- Section Molecular Microbiology, Amsterdam Institute of Molecules, Medicine & Systems, Vrije Universiteit, Amsterdam, the Netherlands
| | - Daria Bottai
- University of Pisa, Department of Biology, Pisa, Italy
| | - Marcel A. Behr
- McGill International TB Centre, Infectious Diseases and Immunity in Global Health Program at the McGill University Health Centre Research Institute, Montreal, Canada
| | - Jeroen W. J. van Heijst
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Prevention, VU University Medical Center, Amsterdam, the Netherlands
- Section Molecular Microbiology, Amsterdam Institute of Molecules, Medicine & Systems, Vrije Universiteit, Amsterdam, the Netherlands
| | - Laleh Majlessi
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR3525, Paris, France
| | - Roland Brosch
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR3525, Paris, France
- * E-mail: (LSA); (RB)
| |
Collapse
|
43
|
Larsen SE, Baldwin SL, Orr MT, Reese VA, Pecor T, Granger B, Dubois Cauwelaert N, Podell BK, Coler RN. Enhanced Anti- Mycobacterium tuberculosis Immunity over Time with Combined Drug and Immunotherapy Treatment. Vaccines (Basel) 2018; 6:vaccines6020030. [PMID: 29795025 PMCID: PMC6027321 DOI: 10.3390/vaccines6020030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/27/2022] Open
Abstract
It is estimated that one third of the world’s population is infected with Mycobacterium tuberculosis (Mtb). This astounding statistic, in combination with costly and lengthy treatment regimens make the development of therapeutic vaccines paramount for controlling the global burden of tuberculosis. Unlike prophylactic vaccination, therapeutic immunization relies on the natural pulmonary infection with Mtb as the mucosal prime that directs boost responses back to the lung. The purpose of this work was to determine the protection and safety profile over time following therapeutic administration of our lead Mtb vaccine candidate, ID93 with a synthetic TLR4 agonist (glucopyranosyl lipid adjuvant in a stable emulsion (GLA-SE)), in combination with rifampicin, isoniazid, and pyrazinamide (RHZ) drug treatment. We assessed the host inflammatory immune responses and lung pathology 7–22 weeks post infection, and determined the therapeutic efficacy of combined treatment by enumeration of the bacterial load and survival in the SWR/J mouse model. We show that drug treatment alone, or with immunotherapy, tempered the inflammatory responses measured in brochoalveolar lavage fluid and plasma compared to untreated cohorts. RHZ combined with therapeutic immunizations significantly enhanced TH1-type cytokine responses in the lung over time, corresponding to decreased pulmonary pathology evidenced by a significant decrease in the percentage of lung lesions and destructive lung inflammation. These data suggest that bacterial burden assessment alone may miss important correlates of lung architecture that directly contribute to therapeutic vaccine efficacy in the preclinical mouse model. We also confirmed our previous finding that in combination with antibiotics therapeutic immunizations provide an additive survival advantage. Moreover, therapeutic immunizations with ID93/GLA-SE induced differential T cell immune responses over the course of infection that correlated with periods of enhanced bacterial control over that of drug treatment alone. Here we advance the immunotherapy model and investigate reliable correlates of protection and Mtb control.
Collapse
Affiliation(s)
- Sasha E Larsen
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| | - Susan L Baldwin
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
| | - Mark T Orr
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| | - Valerie A Reese
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
| | - Tiffany Pecor
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
| | - Brian Granger
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
| | | | - Brendan K Podell
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA 98102, USA.
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
- PAI Life Sciences Inc., Seattle, WA 98102, USA.
| |
Collapse
|
44
|
Kesik‐Brodacka M. Progress in biopharmaceutical development. Biotechnol Appl Biochem 2018; 65:306-322. [PMID: 28972297 PMCID: PMC6749944 DOI: 10.1002/bab.1617] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
Since its introduction in 1982, biopharmaceutical drugs have revolutionized the treatment of a broad spectrum of diseases and are increasingly used in nearly all branches of medicine. In recent years, the biopharmaceuticals market has developed much faster than the market for all drugs and is believed to have great potential for further dynamic growth because of the tremendous demand for these drugs. Biobetters, which contain altered active pharmaceutical ingredients with enhanced efficacy, will play an important role in the development of biopharmaceuticals. Another significant group of biopharmaceuticals are biosimilars. Their introduction in the European Union and, recently, the Unites States markets will reduce the costs of biopharmaceutical treatment. This review highlights recent progress in the field of biopharmaceutical development and issues concerning the registration of innovative biopharmaceuticals and biosimilars. The leading class of biopharmaceuticals, the current biopharmaceuticals market, and forecasts are also discussed.
Collapse
|
45
|
Immunological and physical evaluation of the multistage tuberculosis subunit vaccine candidate H56/CAF01 formulated as a spray-dried powder. Vaccine 2018; 36:3331-3339. [PMID: 29699790 DOI: 10.1016/j.vaccine.2018.04.055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/16/2018] [Accepted: 04/19/2018] [Indexed: 01/11/2023]
Abstract
Liquid vaccine dosage forms have limited stability and require refrigeration during their manufacture, distribution and storage. In contrast, solid vaccine dosage forms, produced by for example spray drying, offer improved storage stability and reduced dependence on cold-chain facilities. This is advantageous for mass immunization campaigns for global public health threats, e.g., tuberculosis (TB), and offers cheaper vaccine distribution. The multistage subunit vaccine antigen H56, which is a fusion protein of the Mycobacterium tuberculosis (Mtb) antigens Ag85B, ESAT-6, and Rv2660, has been shown to confer protective efficacy against active TB before and after Mtb exposure in preclinical models, and it is currently undergoing clinical phase 2a testing. In several studies, including a recent study comparing multiple clinically relevant vaccine adjuvants, the T helper type 1 (Th1)/Th17-inducing adjuvant CAF01 was the most efficacious adjuvant for H56 to stimulate protective immunity against Mtb. With the long-term goal of designing a thermostable and self-administrable dry powder vaccine based on H56 and CAF01 for inhalation, we compared H56 spray-dried with CAF01 with the non-spray-dried H56/CAF01 vaccine with respect to their ability to induce systemic Th1, Th17 and humoral responses after subcutaneous immunization. Here we show that spray drying of the H56/CAF01 vaccine results in preserved antigenic epitope recognition and adjuvant activity of CAF01, and the spray-dried, reconstituted vaccine induces antigen-specific Th1, Th17 and humoral immune responses, which are comparable to those stimulated by the non-spray-dried H56/CAF01 vaccine. In addition, the spray-dried and reconstituted H56/CAF01 vaccine promotes similar polyfunctional CD4+ T-cell responses as the non-spray-dried vaccine. Thus, our study provides proof-of-concept that spray drying of the subunit vaccine H56/CAF01 preserves vaccine-induced humoral and cell-mediated immune responses. These results support our ongoing efforts to develop a thermostable, dry powder-based TB vaccine.
Collapse
|
46
|
Kwon BE, Ahn JH, Min S, Kim H, Seo J, Yeo SG, Ko HJ. Development of New Preventive and Therapeutic Vaccines for Tuberculosis. Immune Netw 2018; 18:e17. [PMID: 29732235 PMCID: PMC5928416 DOI: 10.4110/in.2018.18.e17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/19/2018] [Accepted: 03/06/2018] [Indexed: 01/08/2023] Open
Abstract
Tuberculosis (TB) is a contagious disease that has been responsible for the death of one billion people in the last 200 years. Until now, the only vaccine approved for the prevention of TB is Bacillus Calmette-Guérin (BCG), which is prepared by attenuating Mycobacterium bovis. However, one of the limitations of BCG is that its preventive effect against pulmonary TB varies from person to person. Therefore, there arises a need for a new TB vaccine to replace or supplement BCG. In this review, we have summarized the findings of current clinical trials on preventive and therapeutic TB vaccine candidates. In addition, we have discussed a novel vaccination approach using the cell-based vaccine presenting early secretory antigenic target-6 (ESAT-6), which is a potent immunogenic antigen. The role of ESAT-6 in hosts has also been described.
Collapse
Affiliation(s)
- Bo-Eun Kwon
- Laboratory of Microbiology and Immunology, Kangwon National University, College of Pharmacy, Chuncheon 24341, Korea
| | - Jae-Hee Ahn
- Laboratory of Microbiology and Immunology, Kangwon National University, College of Pharmacy, Chuncheon 24341, Korea
| | - Seunghwan Min
- Laboratory of Microbiology and Immunology, Kangwon National University, College of Pharmacy, Chuncheon 24341, Korea
| | - Hyeongseop Kim
- Laboratory of Microbiology and Immunology, Kangwon National University, College of Pharmacy, Chuncheon 24341, Korea
| | - Jungheun Seo
- Laboratory of Microbiology and Immunology, Kangwon National University, College of Pharmacy, Chuncheon 24341, Korea
| | - Sang-Gu Yeo
- Division of Vaccine Research, Korea National Research Institute of Health, Korea Centers for Disease Control and Prevention, Cheongju 28159, Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, Kangwon National University, College of Pharmacy, Chuncheon 24341, Korea
| |
Collapse
|
47
|
Ashhurst AS, Parumasivam T, Chan JGY, Lin LCW, Flórido M, West NP, Chan HK, Britton WJ. PLGA particulate subunit tuberculosis vaccines promote humoral and Th17 responses but do not enhance control of Mycobacterium tuberculosis infection. PLoS One 2018; 13:e0194620. [PMID: 29554138 PMCID: PMC5858788 DOI: 10.1371/journal.pone.0194620] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/06/2018] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis places a staggering burden on human health globally. The new World Health Organisation End-TB Strategy has highlighted the urgent need for more effective TB vaccines to improve control of the disease. Protein-based subunit vaccines offer potential as safe and effective generators of protective immunity, and the use of particulate vaccine formulation and delivery by the pulmonary route may enhance local immunogenicity. In this study, novel particulate subunit vaccines were developed utilising biodegradable poly(lactic-co-glycolic acid) (PLGA) slow-release particles as carriers for the Mycobacterium tuberculosis lipoprotein MPT83, together with the adjuvants trehalose-dibehenate (TDB) or Monophosphoryl lipid A (MPL). Following delivery by the pulmonary or subcutaneous routes, the immunogenicity and protective efficacy of these vaccines were assessed in a murine model of M. tuberculosis infection. When delivered peripherally, these vaccines induced modest, antigen-specific Th1 and Th17 responses, but strong anti-MPT83 antibody responses. Mucosal delivery of the PLGA(MPT83) vaccine, with or without TDB, increased antigen-specific Th17 responses in the lungs, however, PLGA-encapsulated vaccines did not provide protection against M. tuberculosis challenge. By contrast, peripheral delivery of DDA liposomes containing MPT83 and TDB or MPL, stimulated both Th1 and Th17 responses and generated protection against M. tuberculosis challenge. Therefore, PLGA-formulated vaccines primarily stimulate strong humoral immunity, or Th17 responses if used mucosally, and may be a suitable carrier for vaccines against extracellular pathogens. This study emphasises the critical nature of the vaccine carrier, adjuvant and route of delivery for optimising vaccine efficacy against TB.
Collapse
Affiliation(s)
- Anneliese S. Ashhurst
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | | | - John Gar Yan Chan
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | - Leon C. W. Lin
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Manuela Flórido
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Biosciences and Australian Infectious Disease Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Hak-Kim Chan
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | - Warwick J. Britton
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
- Discipline of Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
48
|
Nieuwenhuizen NE, Kaufmann SHE. Next-Generation Vaccines Based on Bacille Calmette-Guérin. Front Immunol 2018; 9:121. [PMID: 29459859 PMCID: PMC5807593 DOI: 10.3389/fimmu.2018.00121] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/15/2018] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by the intracellular bacterium Mycobacterium tuberculosis (Mtb), remains a major health threat. A live, attenuated mycobacterium known as Bacille Calmette-Guérin (BCG), derived from the causative agent of cattle TB, Mycobacterium bovis, has been in clinical use as a vaccine for 90 years. The current incidence of TB demonstrates that BCG fails to protect sufficiently against pulmonary TB, the major disease manifestation and source of dissemination. The protective efficacy of BCG is on average 50% but varies substantially with geographical location and is poorer in those with previous exposure to mycobacteria. BCG can also cause adverse reactions in immunocompromised individuals. However, BCG has contributed to reduced infant TB mortality by protecting against extrapulmonary TB. In addition, BCG has been associated with reduced general childhood mortality by stimulating immune responses. In order to improve the efficacy of BCG, two major strategies have been employed. The first involves the development of recombinant live mycobacterial vaccines with improved efficacy and safety. The second strategy is to boost BCG with subunit vaccines containing Mtb antigens. This article reviews recombinant BCG strains that have been tested against TB in animal models. This includes BCG strains that have been engineered to induce increased immune responses by the insertion of genes for Mtb antigens, mammalian cytokines, or host resistance factors, the insertion of bacterial toxin-derived adjuvants, and the manipulation of bacterial genes in order to increase antigen presentation and immune activation. Subunit vaccines for boosting BCG are also briefly discussed.
Collapse
|
49
|
Karbalaei Zadeh Babaki M, Soleimanpour S, Rezaee SA. Antigen 85 complex as a powerful Mycobacterium tuberculosis immunogene: Biology, immune-pathogenicity, applications in diagnosis, and vaccine design. Microb Pathog 2017; 112:20-29. [PMID: 28942172 DOI: 10.1016/j.micpath.2017.08.040] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 01/24/2023]
Abstract
Mycobacterium tuberculosis (Mtb) is one of the most life-threatening mycobacterial species which is increasing the death rate due to emerging multi-drug resistant (MDR) strains. Concerned health authorities worldwide are interested in developing an effective vaccine to prevent the spread of Mtb. After years of research, including successful identification of many Mtb immunogenic molecules, effective therapeutic agents or a vaccine have yet to be found. However, among the identified Mtb immunogenes, antigen 85 (Ag85) complex (Ag85A, Ag85B, and Ag85C) is receiving attention from scientists as it allows bacteria to evade the host immune response by preventing formation of phagolysosomes for eradication of infection. Due to their importance, A85 molecules are being utilized as tools in diagnostic methods and in the construction of new vaccines, such as recombinant attenuated vaccines, DNA vaccines, and subunit vaccines. This paper represents a comprehensive review of studies on Mtb molecules examining pathogenicity, biochemistry, immunology, and the role of Mtb in therapeutic or vaccine research.
Collapse
Affiliation(s)
- Mohsen Karbalaei Zadeh Babaki
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Inflammation and Inflammatory Diseases Division, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
50
|
Myllymäki H, Niskanen M, Oksanen KE, Sherwood E, Ahava M, Parikka M, Rämet M. Identification of novel antigen candidates for a tuberculosis vaccine in the adult zebrafish (Danio rerio). PLoS One 2017; 12:e0181942. [PMID: 28742838 PMCID: PMC5526617 DOI: 10.1371/journal.pone.0181942] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/10/2017] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) remains a major global health challenge and the development of a better vaccine takes center stage in fighting the disease. For this purpose, animal models that are capable of replicating the course of the disease and are suitable for the early-stage screening of vaccine candidates are needed. A Mycobacterium marinum infection in adult zebrafish resembles human TB. Here, we present a pre-clinical screen for a DNA-based tuberculosis vaccine in the adult zebrafish using an M. marinum infection model. We tested 15 antigens representing different types of mycobacterial proteins, including the Resuscitation Promoting factors (Rpf), PE/PPE protein family members, other membrane proteins and metabolic enzymes. The antigens were expressed as GFP fusion proteins, facilitating the validation of their expression in vivo. The efficiency of the antigens was tested against a low-dose intraperitoneal M. marinum infection (≈ 40 colony forming units), which mimics a primary M. tuberculosis infection. While none of the antigens was able to completely prevent a mycobacterial infection, four of them, namely RpfE, PE5_1, PE31 and cdh, led to significantly reduced bacterial burdens at four weeks post infection. Immunization with RpfE also improved the survival of the fish against a high-dose intraperitoneal injection with M. marinum (≈ 10.000 colony forming units), resembling the disseminated form of the disease. This study shows that the M. marinum infection model in adult zebrafish is suitable for the pre-clinical screening of tuberculosis vaccines and presents RpfE as a potential antigen candidate for further studies.
Collapse
Affiliation(s)
- Henna Myllymäki
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- * E-mail:
| | - Mirja Niskanen
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Kaisa Ester Oksanen
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Eleanor Sherwood
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Maarit Ahava
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Mataleena Parikka
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Oral and Maxillofacial Unit, Tampere University Hospital, Tampere, Finland
| | - Mika Rämet
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|