1
|
Borges ÁH, Russell M, Tait D, Scriba TJ, Nemes E, Skallerup P, van Brakel E, Cabibbe AM, Cirillo DM, Leuvennink-Steyn M, Rutkowski KT, Wood GK, Thierry-Carstensen B, Tingskov PN, Meldgaard EC, Kristiansen MP, Søndergaard RE, Hansen CH, Follmann F, Jensen CG, Gela A, Ntinginya NE, Ruhwald M, Shenje J, White L, Innes C, Selepe P, Ngaraguza B, Holmgren C, Collings T, Andersen P, Dawson R, Churchyard G, Sabi I, Diacon AH, Mortensen R, Hatherill M, POR TB study group. Immunogenicity, safety, and efficacy of the vaccine H56:IC31 in reducing the rate of tuberculosis disease recurrence in HIV-negative adults successfully treated for drug-susceptible pulmonary tuberculosis: a double-blind, randomised, placebo-controlled, phase 2b trial. THE LANCET. INFECTIOUS DISEASES 2025; 25:751-763. [PMID: 40056922 DOI: 10.1016/s1473-3099(24)00814-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 06/22/2025]
Abstract
BACKGROUND People with tuberculosis who complete treatment remain at risk of recurrent disease. The vaccine H56:IC31 has been shown to be safe and immunogenic in phase 1 and 2 studies, but whether it can reduce the risk of tuberculosis recurrence is unknown. METHODS In a double-blind, randomised, placebo-controlled, phase 2b trial in South Africa (five clinical trial sites) and Tanzania (one clinical trial site), we enrolled participants aged 18-60 years, without HIV, who had completed more than 5 months (22 weeks) of treatment for drug-susceptible pulmonary tuberculosis. During trial screening (≤7 days after starting treatment), two sputum samples were obtained and frozen for later comparison to recurrent isolates by whole-genome sequencing (WGS). Eligible participants were randomly assigned (1:1; block size of four) to receive two intramuscular doses in the deltoid, 56 days apart, of H56:IC31 or placebo. After the first dose of H56:IC31 or placebo, participants were followed up until study day 421 (1 year after the second dose) and checked at each visit for tuberculosis signs and symptoms. If tuberculosis was suspected, two sputum samples were obtained: one sample was tested by automated molecular test (Xpert MTB/RIF Ultra) and sent for liquid culture; and the other sample was stored frozen for later analysis by whole-genome sequencing (WGS). At the last visit (day 421), two sputum samples were obtained from all sputum-productive participants, regardless of symptoms, to detect cases of asymptomatic tuberculosis. The primary endpoint was culture-confirmed recurrent pulmonary tuberculosis (due to relapse with the same strain, reinfection by a different strain, or indeterminate) occuring during the period starting at day 70 (14 days after the second dose) and ending on day 421 (1 year after the second dose). Vaccine efficacy against recurrent tuberculosis was derived from Cox proportional hazards models. Secondary endpoints included vaccine efficacy to prevent tuberculosis relapse or reinfection independently, as differentiated by WGS, and safety and immunogenicity outcomes (H56-specific CD4 T-cell responses and humoral anti-H56 IgG responses). Primary analysis of vaccine efficacy was based on modified intention-to-treat (mITT), in all randomly assigned participants except those with tuberculosis disease recurrence or who withdrew before day 70 (or 14 days after the second dose for those who received both doses). Safety was assessed in all randomly assigned participants who received at least one dose of vaccine or placebo. The trial was registered with ClinicalTrials.gov, NCT03512249, and is complete. FINDINGS 831 participants (mean age 34·7 years [SD 11·1]; 229 [28%] female and 602 [72%] male; 549 [66%] Black) were enrolled from Jan 31, 2019, to Jan 20, 2022; 415 participants were randomly assigned to receive H56:IC31 and 416 to receive placebo. Follow-up was completed by March 20, 2023 (mean follow-up duration 410·1 days [SD 82·8]). In the primary mITT analysis, recurrent tuberculosis occurred in 23 of 400 participants in the H56:IC31 group (12 relapses, eight reinfections, and three indeterminate); and in 14 of 406 in the placebo group (six relapses, seven reinfections, and one indeterminate). Vaccine efficacy for prevention of recurrence was -73·8% (95% CI -246·9 to 9·8; p=0·10). Vaccine efficacy for prevention of relapse was -116·1% (-522·2 to 16·3; p=0·11) and for prevention of reinfection was -21·1% (-245·3 to 56·5; p=0·71). 2 weeks after the planned second dose, H56:IC31 had significantly increased the frequencies of H56-specific CD4 T cells expressing interferon-γ, tumour necrosis factor, interleukin (IL)-2, or IL-17 in vaccinees (median percentage of CD4 T cells, 0·35% [IQR 0·19 to 0·57]) compared with placebo (0·11% [0·09 to 0·23]; p<0·0001). H56-specific IgG responses were significantly higher in H56:IC31 recipients (median arbitrary units per mL, 6·84 [IQR 1·64 to 32·8]) than in placebo recipients (1·94 [1·05 to 3·86]; p<0·0001). A greater proportion of H56:IC31 recipients had mild-to-moderate injection site reactions than placebo recipients (165 [40%] of 415 vs 78 [19%] of 416). No treatment-related serious adverse events were reported. Two participants who received H56:IC31 and six who received placebo died. INTERPRETATION Vaccination with H56:IC31 at treatment completion for pulmonary tuberculosis did not reduce the risk of recurrent disease. H56:IC31 was well tolerated and immunogenic but might have increased the risk of relapses by endogenous strains. FUNDING The European and Developing Countries Clinical Trials Partnership (EDCTP2) supported by the EU (grant number RIA2016V-1631, POR TB consortium). Additional funding to support completion of the trial was provided by the Statens Serum Institut, Aurum Institute, and the South African Tuberculosis Vaccine Initiative.
Collapse
Affiliation(s)
- Álvaro H Borges
- Department of Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.
| | | | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Per Skallerup
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Andrea M Cabibbe
- Emerging Bacterial Pathogens Unit, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS Ospedale San Raffaele, Milano, Italy
| | | | | | - Grith K Wood
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Birgit Thierry-Carstensen
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Pernille N Tingskov
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Emilie C Meldgaard
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Max P Kristiansen
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Christian H Hansen
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark; MRC International Statistics and Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK
| | - Frank Follmann
- Department of Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Charlotte G Jensen
- Department of Vaccine Development, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anele Gela
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nyanda E Ntinginya
- Mbeya Medical Research Center, National Institute for Medical Research, Mbeya, Tanzania
| | - Morten Ruhwald
- Foundation for Innovative New Diagnostics, Geneva, Switzerland
| | - Justin Shenje
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Lisa White
- Centre for Tuberculosis Research Innovation, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Craig Innes
- The Aurum Institute, Johannesburg, South Africa
| | | | - Beatrice Ngaraguza
- Mbeya Medical Research Center, National Institute for Medical Research, Mbeya, Tanzania
| | | | | | - Peter Andersen
- Novo Nordisk Foundation, Hellerup, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Rodney Dawson
- Centre for Tuberculosis Research Innovation, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Gavin Churchyard
- The Aurum Institute, Johannesburg, South Africa; School of Public Health, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa; Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Issa Sabi
- Mbeya Medical Research Center, National Institute for Medical Research, Mbeya, Tanzania
| | | | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
Collaborators
Justin Shenje, Danelle Van As, Angelique Luabeya-Kanykany, Michele Tameris, Simon Mendelsohn, Nicolette Tredoux, Hennie Geldenhuys, Alessandro Companie, Sivuyile Buhlungu, Sonia Stryers, Miriam Moses, Veronica Baartman, Cynthia Gwintsa, Denis Arendsen, Habibullah Valley, Haydn Africa, Marcia Steyn, Simbarashe Mabwe, Johanna Van Rooyen, Angelique Mouton, Fajwa Opperman, Julia Noble, Thelma Leopeng, Humphrey Mulenga, Carmen Segelaar, Elizabeth Beyers, Denise Van der Westhuizen, Terence Esterhuizen, Asma Toefy, Mzwandile Erasmus, Yolundi Cloete, Lungisa Jaxa, Constance Schreuder, Onke Nombida, Rodney Raphela, Nicole Bilek, Cynthia Ontong, Ilse Davids, Margareth Erasmus, Christel Petersen, Rose Ockhius, Bongiwe Vasana, Mamosa Moleleki-Mabala, Sandisiwe Mangali, Hlengiwe Nkambule, Alida Carstens, Suzette Visagie, Phumzile Langafa, Elmien Verster, Rachel Oelofse, Rodney Dawson, Linda White, Frederico di Marco, Issa Sabi, Nyanda Elias Ntinginya, Beatrice Ngaraguza, Julieth Lalashowi, Ombeni Chimbe, Bariki Mtafya, Godlove Chaula, Emmanuel Sichone, Lwitiho Sudi, Abisai Kisinda, Revocatus Kunambi, Bahati Myombe, Simeon Mwanyonga, Cyprian Mtweve, Dereck Tait, Elana van Brakel, Maureen Lambrick, Fay Swanson, Kathryn Rutkowski, Devin Hunt, Anja van der Westhuizen, Heather Siefers, Leonie Coetzee, Mildie Leuvennink-Steyn, Deidre Albertyn, Marisa Russell, Viola-Marie Raubenheimer, Arrie van der Merwe, Aaliya Gangat, Adriane Kock, Amanda van Aswegen, Amukelani Dolly Nyathi, Aneesa Lakhi, Audrey Dlamini, Bertha Pei, Blossom Makhubalo, Candice Eyre, Craig Innes, Moeti Godfrey Serake, Pearl Selepe, Shirley Malefo-Grootboom, Elizabeth Barbara Janse van Rensburg, Hildah Ledwaba, Immaculate Mabasa, Juanita Market, Ken Clarke, Lawrence Ntoahae, Lebogang Isaac Tswaile, Letlhogonolo Seabela, Lezaan-Marie Erasmus, Lindiwe Nhlangulela, Mantai Makhetha, Marelize Collignon, Marietjie Zietsman, Maryna Nel, Mmanare Maria Tlhapi, Molly Majola, Moogo Fikizolo, Morongoenyane Josephine Motsiri, Mpho Makoanyane, Ndlela Israel Kunene, Nhlamulo Ndlovu, Nishanee Arjun, Nkosinathi Mabuza, Nondumiso Langa, Nontsikelelo Mbipha, Palesa Mosweu, Pearl Nomsa Sanyaka, Richard Nteleki, Samuel Mompati, Serame Sylvester Modipa, Tedrius Ntshauba, Thelma Goliath, Thompho Mugwena, Tshegofatso Mabe, Victor Rameetse, Vincent Tshikovhi, Welseh Phindile Galaive, Álvaro H Borges, Rasmus Mortensen, Per Skallerup, Birgit Thierry-Carstensen, Frank Follmann, Charlotte G Jensen, Pernille N Tingskov, Emilie C Meldgaard, Max P Kristiansen, Rebecca Bach Dohn, Rikke Dahl Bartholomæussen, Alida van Deventer, Hesti-Mari van Huyssteen, Carmen Kleinhans, Julia Sims, Monde Patientia, Thirumani Govender, Priscilla Samaai,
Collapse
|
2
|
Larsen SE, Rais M, Reese VA, Ferede D, Pecor T, Kaur S, Nag D, Smytheman T, Gray SA, Carter D, Baldwin SL, Coler RN. Characterizing TLR4 agonist EmT4™ as an anti-Mycobacterium tuberculosis vaccine adjuvant. Immunohorizons 2025; 9:vlaf014. [PMID: 40285479 PMCID: PMC12032397 DOI: 10.1093/immhor/vlaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Abstract
Tuberculosis (TB) is again the deadliest infectious disease globally, and more efficacious vaccines are needed to reduce this mortality. Successful subunit TB vaccines need antigens and adjuvants that are immunogenic, inexpensive, and accessible. Here we evaluated the potential of synthetically produced Monophosphoryl lipid A (SyMLP), a TLR4-agonist, formulated in an oil-in-water emulsion (EmT4™) in combination with selected fusion proteins, to drive an effective vaccine-mediated immunogenic response in C57BL/6 mice against Mycobacterium tuberculosis (M.tb) HN878 and H37Rv challenge. We first observed that EmT4™ enhances activation of C57BL/6 bone-marrow derived macrophages and dendritic cells measured by CD40, CD86, and MHCII expression by flow cytometry. EmT4™ did not induce safety signals in a scaled tolerability study. In immunogenicity studies, mice immunized 3 times 3 weeks apart with ID93 antigen + EmT4™ produced a significantly higher magnitude of circulating proinflammatory cytokines and ID93-specific immunoglobulin G (IgG) antibodies pre- and post-challenge with M.tb than saline control animals. Ex vivo ID93 restimulated splenocytes and lung cells elicited significant polyfunctional CD4+ T-helper 1 responses. Importantly, ID93 + EmT4™ immunizations significantly reduced bacterial burden in C57BL/6 mice 4 weeks post-challenge. Interestingly, EmT4™ paired with a next generation protein fusion ID91 also afforded prophylactic protection against M.tb HN878 challenge in both young (6 to 8 wk) and aged (20 mo) immunocompromised Beige mice. These protection and immunogenicity findings suggest that synthetically derived EmT4™ adjuvant is not only suitable to help backfill the preclinical TB vaccine candidate pipeline but is also suitable for the needs of the global community.
Collapse
Affiliation(s)
- Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Valerie A Reese
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Debora Ferede
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Suhavi Kaur
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Deepika Nag
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Thomas Smytheman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Sean A Gray
- PAI Life Sciences Inc, Seattle, WA, United States
| | | | - Susan L Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
3
|
Zhang D, Wang D, Jiang S, Wan Y, Zhao Y, Dong W, Li X, Fu L, Zhang W. Prediction of a novel synthetic peptide vaccine against tuberculosis and validation of its immunogenicity. Int Immunopharmacol 2025; 153:114531. [PMID: 40154176 DOI: 10.1016/j.intimp.2025.114531] [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: 06/25/2024] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Tuberculosis (TB) is an infectious disease transmitted through the respiratory system that affects people worldwide. Bacillus Calmette-Guérin (BCG), the only approved TB vaccine, has been shown to have highly variable protective efficacy in different populations and is ineffective at protecting adults. Therefore, the development of more effective TB vaccines is vital. METHODS Three dominant antigens (ESAT6, CFP10, and MPT64) from the region of difference were selected for this study. Their physicochemical properties, spatial structures, and immune responses were evaluated using bioinformatics screening of dominant T cell and B cell epitopes. Three vaccine constructs were developed. After selecting the appropriate linkers, their physicochemical properties, spatial structures, and immune responses of the vaccines were evaluated, and molecular dynamics simulations were performed to test their ability to bind to major histocompatibility complex (MHC) receptors within 100 ns. This process aimed to create highly antigenic vaccine constructs capable of eliciting an immune response. The effects of the vaccine constructs on the host immune response were assessed using enzyme-linked immunosorbent assays, flow cytometry, and hematoxylin and eosin staining. RESULTS A novel peptide vaccine, designated ECM-64, was developed by screening six immunodominant peptides from three antigens and constructing independent T-epitope and B-epitope vaccines. Compared weith BCG-immunized mice, ECM-64-immunized mice exhibited a substantial augmentation in Th1/Th2 cytokine secretion and CD3+CD4+T and CD3+CD8-T lymphocyte counts. ECM-64-specific IgG and IgG1 antibodies were produced after immunization. The immunoinformatics findings were largely consistent with those obtained from the analysis of immunized mice. CONCLUSION ECM-64 is a promising multipeptide TB vaccine with the advantage of inducing high levels of Th1/Th2 cytokines, antibodies, and CD3+CD4+T and CD3+CD8-T lymphocytes in mice. This study also provides preliminary evidence that bioinformatic methods can be used to screen for dominant epitopes. These findings lay the groundwork for the development of peptide-based TB vaccines.
Collapse
Affiliation(s)
- Dongdong Zhang
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Donghui Wang
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Shuo Jiang
- College of Food Science and Engineering, Jilin Agricultural University,Changchun 130118, China
| | - Yue Wan
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Yukun Zhao
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Weiqi Dong
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Xiaodi Li
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Lu Fu
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China.
| | - Wenhui Zhang
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
4
|
Thong PM, Wong YH, Kornfeld H, Goletti D, Ong CWM. Immune dysregulation of diabetes in tuberculosis. Semin Immunol 2025; 78:101959. [PMID: 40267700 DOI: 10.1016/j.smim.2025.101959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
The rising prevalence of diabetes mellitus (DM) is undermining global efforts to eliminate tuberculosis (TB). Most studies found that patients with pulmonary TB and DM have more cavitary lung lesions, higher mycobacterial burden on the lungs, longer periods of infectiousness, and worse outcomes. Both human and animal studies indicate that TB-DM is associated with impaired innate and adaptive immune responses, resulting in delayed bacterial clearance. Similar observations have been noted in other infections, such as those caused by Klebsiella pneumoniae, where DM contributes to increased susceptibility and worse outcomes due to compromised immune functions including defective phagocytosis and impaired early immune cell recruitment. This review delves into the mechanisms of immune dysfunction in TB-DM, exploring how DM increases TB susceptibility and severity. By elucidating these complex interactions, this review aims to offer insights into more effective strategies for managing and improving outcomes for patients with this challenging comorbidity.
Collapse
Affiliation(s)
- Pei Min Thong
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yi Hao Wong
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hardy Kornfeld
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology, National Institute for infectious diseases-IRCCS L. Spallanzani, Rome, Italy.
| | - Catherine W M Ong
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore; Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore.
| |
Collapse
|
5
|
Seo H, Kim BJ, Oh J, Jung S, Lee JY, Kim BJ. Protection against tuberculosis by vaccination of secreted chorismate mutase (Rv1885c) combined with a hepatitis B virus (HBV)-derived peptide, Poly6, and alum adjuvants. Vaccine 2025; 47:126710. [PMID: 39787795 DOI: 10.1016/j.vaccine.2025.126710] [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: 08/09/2024] [Revised: 12/10/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Tuberculosis (TB) remains a significant global health issue due to the limited efficacy of the Bacillus Calmette-Guérin (BCG) vaccine, highlighting the need for the development of an improved TB vaccine. In this study, we created a novel TB subunit vaccine consisting of TB-secreted chorismate mutase (TBCM) (Rv1885c) and a hepatitis B virus (HBV)-derived peptide (Poly6), which elicits Type I interferon responses, both with and without an alum adjuvant. We evaluated the immunogenicity, protective efficacy, and therapeutic efficacy of this vaccine candidate in an in vivo mouse model. Our results revealed that subcutaneous vaccination with TBCM combined with Poly6 induced stronger antigen-specific humoral and cell-mediated immune responses than TBCM alone or TBCM combined with alum adjuvant. Furthermore, adding the alum adjuvant to TBCM combined with Poly6 significantly enhanced antigen-specific immune responses. Subcutaneous vaccination with TBCM combined with both Poly6 and alum adjuvants effectively protected mice against Mycobacterium tuberculosis (Mtb) K strain infection, demonstrating its potential as a TB vaccine. Additionally, this vaccine platform exhibited therapeutic potential by significantly reducing bacterial loads and lung inflammation in mice previously infected with the virulent Mtb K strain. In conclusion, our findings suggest that TBCM is highly immunogenic in mice and that TBCM combined with both Poly6 and alum adjuvants represents a promising vaccine platform for TB prevention and treatment.
Collapse
Affiliation(s)
- Hyejun Seo
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Seoul National University Medical Research Center (SNUMRC), Seoul 03080, Republic of Korea
| | - Byung-Jun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Jaehun Oh
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sangkwon Jung
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Young Lee
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Seoul National University Medical Research Center (SNUMRC), Seoul 03080, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Liver Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
6
|
Zhao G, Sathkumara HD, Miranda-Hernandez S, Seifert J, Valencia-Hernandez AM, Puri M, Huang W, Toth I, Daly N, Skwarczynski M, Kupz A. A Modular Self-Assembling and Self-Adjuvanting Multiepitope Peptide Nanoparticle Vaccine Platform to Improve the Efficacy and Immunogenicity of BCG. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406874. [PMID: 39757706 DOI: 10.1002/smll.202406874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/29/2024] [Indexed: 01/07/2025]
Abstract
After more than a century since its initial development, Bacille Calmette-Guérin (BCG) remains the only licensed vaccine against tuberculosis (TB). Subunit boosters are considered a viable strategy to enhance BCG efficacy, which often wanes in adolescence. While many studies on booster subunit vaccines have concentrated on recombinant proteins, here we developed a novel modular peptide-based subunit vaccine platform that is flexible, cold-chain independent and customizable to diverse circumstances and populations. Each individual peptide building block consists of a linear arrangement comprising a 15-leucine self-assembly inducer moiety, a Mycobacterium tuberculosis (Mtb) target epitope and an human leukocyte antigen E (HLA-E) binding moiety, with each moiety separated by a triple lysine spacer. The building blocks, in any combination, are able to form a multiepitope nanoparticle. Six Mtb epitopes were selected to produce the self-assembling and self-adjuvating peptide-based TB nano-vaccine candidate PNx6. In vivo vaccination-challenge experiments demonstrated that subcutaneous boost of parenteral BCG immunization with PNx6 significantly enhanced its immunogenicity and improved its protective efficacy in a murine model of TB by more than 5-fold. This study presents evidence that purely amphiphilic peptides self-assemble into self-adjuvating nanoparticles with appropriate size and morphology for TB vaccination with great potential for a multitude of other diseases.
Collapse
Affiliation(s)
- Guangzu Zhao
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Harindra D Sathkumara
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Socorro Miranda-Hernandez
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Julia Seifert
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Ana Maria Valencia-Hernandez
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Munish Puri
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Wenbin Huang
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Istvan Toth
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Norelle Daly
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Mariusz Skwarczynski
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Andreas Kupz
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| |
Collapse
|
7
|
Gurmessa SK, Choi HG, Back YW, Jiang Z, Pham TA, Choi S, Kim HJ. Novel fusion protein REA induces robust prime protection against tuberculosis in mice. NPJ Vaccines 2025; 10:20. [PMID: 39890787 PMCID: PMC11785989 DOI: 10.1038/s41541-025-01077-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025] Open
Abstract
While many novel candidates for tuberculosis vaccines are presently undergoing pre-clinical or clinical trials, none of them have been able to eliminate infection entirely. In this study, we engineered a potent chimeric protein vaccine candidate, Rv2299cD2D3-ESAT-6-Ag85B (REA), which induced Th1 and Th17 responses via dendritic cell maturation. REA-activated macrophages operated the killing mechanisms of Mycobacterium tuberculosis (MTB), such as phagosomal maturation and phagolysosome fusion, through the (PI3K)-p38 MAPK-Ca2+-NADPH oxidase pathway. Dendritic cells and macrophages activated by REA elicited synergistic anti-mycobacterial responses. Notably, REA-immunized mice suppressed MTB growth to undetectable levels at 16 weeks post-infection, which was supported by gross and pathologic findings and acid-fast staining of the lung tissues, and maintained antigen-specific multifunctional IFN-γ+IL-2+TNF-α CD4+ T and long-lasting T cells producing cytokines in the tissues. Our findings suggest that REA is an outstanding prime prophylactic vaccine candidate against tuberculosis.
Collapse
Affiliation(s)
- Sintayehu Kebede Gurmessa
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Han-Gyu Choi
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yong Woo Back
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Zongyou Jiang
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Thuy An Pham
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seunga Choi
- R&D Center, Myco-Rapha Inc., Daejeon, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
8
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
9
|
Tait D, Diacon A, Borges ÁH, van Brakel E, Hokey D, Rutkowski KT, Hunt DJ, Russell M, Andersen PL, Kromann I, Ruhwald M, Churchyard G, Dawson R. Safety and Immunogenicity of the H56:IC31 Tuberculosis Vaccine Candidate in Adults Successfully Treated for Drug-Susceptible Pulmonary Tuberculosis: A Phase 1 Randomized Trial. J Infect Dis 2024; 230:1262-1270. [PMID: 38557639 DOI: 10.1093/infdis/jiae170] [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: 11/14/2023] [Revised: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND H56:IC31 is a candidate vaccine against tuberculosis (TB) with the potential to reduce TB recurrence rate. It is thus important for future clinical trials to demonstrate safety and immunogenicity of H56:IC31 in individuals treated for TB. METHODS Twenty-two adults confirmed to be Mycobacterium tuberculosis negative (by 2 GeneXpert tests or 2 sputum cultures) after 4-5 months of TB treatment, and not more than 28 days after completion of TB treatment, were randomized to receive 2 doses of H56:IC31 (5 mg H56:500 nmol IC31; n = 16) or placebo (n = 6) 56 days apart. Participants were followed for 420 days for safety and immunogenicity. RESULTS H56:IC31 vaccination was associated with an acceptable safety profile, consisting mostly of mild self-limited injection site reactions. No serious adverse events or vaccine-related severe adverse events were reported. H56:IC31 induced a CD4+ T-cell response for Ag85B and ESAT-6, with ESAT-6 being immunodominant, which persisted through 6 months after the last vaccination. There was some evidence of CD8+ T-cell responses for both Ag85B and ESAT-6, but to a lesser extent than CD4+ responses. CONCLUSIONS H56:IC31 was associated with an acceptable safety profile, and induced a predominant CD4+ T-cell response, in adults recently treated for drug-susceptible, uncomplicated pulmonary TB. CLINICAL TRIALS REGISTRATION NCT02375698.
Collapse
Affiliation(s)
| | | | - Álvaro H Borges
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | | | | | | | | | - Peter L Andersen
- Novo Nordisk Foundation, Hellerup
- Department of Immunology and Microbiology, University of Copenhagen
| | - Ingrid Kromann
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Morten Ruhwald
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
- Foundation of Innovative New Diagnostics, Geneva, Switzerland
| | - Gavin Churchyard
- Aurum Institute
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Rodney Dawson
- Centre for Tuberculosis Research Innovation, University of Cape Town Lung Institute, Cape Town, South Africa
| |
Collapse
|
10
|
Chen Z, Zhang Y, Wu J, Xu J, Hu Z, Fan XY. A multistage protein subunit vaccine as BCG-booster confers protection against Mycobacterium tuberculosis infection in murine models. Int Immunopharmacol 2024; 139:112811. [PMID: 39068754 DOI: 10.1016/j.intimp.2024.112811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
The eradication of tuberculosis remains a global challenge. Despite being the only licensed vaccine, Bacillus Calmette-Guérin (BCG) confers limited protective efficacy in adults and individuals with latent tuberculosis infections (LTBI). There is an urgent need to develop novel vaccines that can enhance the protective effect of BCG. Protein subunit vaccines have garnered significant research interest due to their safety and plasticity. Based on previous studies, we selected three antigens associated with LTBI (Rv2028c, Rv2029c, Rv3126c) and fused them with an immunodominant antigen Ag85A, resulting in the construction of a multistage protein subunit vaccine named A986. We evaluated the protective effect of recombinant protein A986 adjuvanted with MPL/QS21 as a booster vaccine for BCG against Mycobacterium tuberculosis (Mtb) infection in mice. The A986 + MPL/QS21 induced the secretion of antigen-specific Th1 (IL-2+, IFN-γ+ and TNF-α+) and Th17 (IL-17A+) cytokines in CD4+ and CD8+ T cells within the lung and spleen of mice, while also increased the frequency of central memory and effector memory T cells. Additionally, it also induced the enhanced production of IgG antibodies. Compared to BCG alone, A986 + MPL/QS21 boosting significantly augmented the proliferation of antigen-specific multifunctional T cells and effectively reduced bacterial load in infected mice. Taken together, A986 + MPL/QS21 formulation induced robust antigen-specific immune responses and provided enhanced protection against Mtb infection as a booster of BCG vaccine.
Collapse
Affiliation(s)
- Zhenyan Chen
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China
| | - Ying Zhang
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Juan Wu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Jinchuan Xu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China.
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 201508, China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital & The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518112, Guangdong Province, China.
| |
Collapse
|
11
|
Sethi G, Varghese RP, Lakra AK, Nayak SS, Krishna R, Hwang JH. Immunoinformatics and structural aided approach to develop multi-epitope based subunit vaccine against Mycobacterium tuberculosis. Sci Rep 2024; 14:15923. [PMID: 38987613 PMCID: PMC11237054 DOI: 10.1038/s41598-024-66858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024] Open
Abstract
Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis (Mtb), which is one of the prominent reasons for the death of millions worldwide. The bacterium has a substantially higher mortality rate than other bacterial diseases, and the rapid rise of drug-resistant strains only makes the situation more concerning. Currently, the only licensed vaccine BCG (Bacillus Calmette-Guérin) is ineffective in preventing adult pulmonary tuberculosis prophylaxis and latent tuberculosis re-activation. Therefore, there is a pressing need to find novel and safe vaccines that provide robust immune defense and have various applications. Vaccines that combine epitopes from multiple candidate proteins have been shown to boost immunity against Mtb infection. This study applies an immunoinformatic strategy to generate an adequate multi-epitope immunization against Mtb employing five antigenic proteins. Potential B-cell, cytotoxic T lymphocyte, and helper T lymphocyte epitopes were speculated from the intended proteins and coupled with 50 s ribosomal L7/L12 adjuvant, and the vaccine was constructed. The vaccine's physicochemical profile demonstrates antigenic, soluble, and non-allergic. In the meantime, docking, molecular dynamics simulations, and essential dynamics analysis revealed that the multi-epitope vaccine structure interacted strongly with Toll-like receptors (TLR2 and TLR3). MM-PBSA analysis was performed to ascertain the system's intermolecular binding free energies accurately. The immune simulation was applied to the vaccine to forecast its immunogenic profile. Finally, in silico cloning was used to validate the vaccine's efficacy. The immunoinformatics analysis suggests the multi-epitope vaccine could induce specific immune responses, making it a potential candidate against Mtb. However, validation through the in-vivo study of the developed vaccine is essential to assess its efficacy and immunogenicity profile, which will assure active protection against Mtb.
Collapse
Affiliation(s)
- Guneswar Sethi
- Department of Predictive Toxicology, Korea Institute of Toxicology (KIT), Daejeon, Republic of Korea
- Animal Model Research Group, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea
| | | | - Avinash Kant Lakra
- Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | | | - Ramadas Krishna
- Department of Bioinformatics, Pondicherry University, Puducherry, 605014, India.
| | - Jeong Ho Hwang
- Animal Model Research Group, Korea Institute of Toxicology, 30 Baehak 1-gil, Jeonguep, Jeollabuk-do, 56212, Republic of Korea.
| |
Collapse
|
12
|
Liu Y, Li H, Dai D, He J, Liang Z. Gene Regulatory Mechanism of Mycobacterium Tuberculosis during Dormancy. Curr Issues Mol Biol 2024; 46:5825-5844. [PMID: 38921019 PMCID: PMC11203133 DOI: 10.3390/cimb46060348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) complex, is a zoonotic disease that remains one of the leading causes of death worldwide. Latent tuberculosis infection reactivation is a challenging obstacle to eradicating TB globally. Understanding the gene regulatory network of Mtb during dormancy is important. This review discusses up-to-date information about TB gene regulatory networks during dormancy, focusing on the regulation of lipid and energy metabolism, dormancy survival regulator (DosR), White B-like (Wbl) family, Toxin-Antitoxin (TA) systems, sigma factors, and MprAB. We outline the progress in vaccine and drug development associated with Mtb dormancy.
Collapse
Affiliation(s)
- Yiduo Liu
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Han Li
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Dejia Dai
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| |
Collapse
|
13
|
Passos BBS, Araújo-Pereira M, Vinhaes CL, Amaral EP, Andrade BB. The role of ESAT-6 in tuberculosis immunopathology. Front Immunol 2024; 15:1383098. [PMID: 38633252 PMCID: PMC11021698 DOI: 10.3389/fimmu.2024.1383098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Despite major global efforts to eliminate tuberculosis, which is caused by Mycobacterium tuberculosis (Mtb), this disease remains as a major plague of humanity. Several factors associated with the host and Mtb interaction favor the infection establishment and/or determine disease progression. The Early Secreted Antigenic Target 6 kDa (ESAT-6) is one of the most important and well-studied mycobacterial virulence factors. This molecule has been described to play an important role in the development of tuberculosis-associated pathology by subverting crucial components of the host immune responses. This review highlights the main effector mechanisms by which ESAT-6 modulates the immune system, directly impacting cell fate and disease progression.
Collapse
Affiliation(s)
- Beatriz B. S. Passos
- Curso de Medicina, Universidade Salvador, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
| | - Mariana Araújo-Pereira
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Caian L. Vinhaes
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Programa de Pós-Graduação em Medicina e Saúde Humana, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
- Departamento de Infectologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Bruno B. Andrade
- Curso de Medicina, Universidade Salvador, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Programa de Pós-Graduação em Medicina e Saúde Humana, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| |
Collapse
|
14
|
Kwon KW, Choi HG, Choi HH, Choi E, Kim H, Kim HJ, Shin SJ. Immunogenicity and protective efficacy of RipA, a peptidoglycan hydrolase, against Mycobacterium tuberculosis Beijing outbreak strains. Vaccine 2024; 42:1941-1952. [PMID: 38368223 DOI: 10.1016/j.vaccine.2024.02.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/11/2023] [Accepted: 02/11/2024] [Indexed: 02/19/2024]
Abstract
Given that individuals with latent tuberculosis (TB) infection represent the major reservoir of TB infection, latency-associated antigens may be promising options for development of improved multi-antigenic TB subunit vaccine. Thus, we selected RipA, a peptidoglycan hydrolase required for efficient cell division of Mycobacterium tuberculosis (Mtb), as vaccine candidate. We found that RipA elicited activation of dendritic cells (DCs) by induction of phenotypic maturation, increased production of inflammatory cytokines, and prompt stimulation of MAPK and NF-κB signaling pathways. In addition, RipA-treated DCs promoted Th1-polarzied immune responses of naïve CD4+ T cells with increased proliferation and activated T cells from Mtb-infected mice, which conferred enhanced control of mycobacterial growth inside macrophages. Moreover, mice immunized with RipA formulated in GLA-SE adjuvant displayed remarkable generation of Ag-specific polyfunctional CD4+ T cells in both lung and spleen. Following an either conventional or ultra-low dose aerosol challenges with 2 Mtb Beijing clinical strains, RipA/GLA-SE-immunization was not inferior to BCG by mediating protection as single Ag. Collectively, our findings highlighted that RipA could be a novel candidate as a component of multi-antigenic TB subunit vaccines.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea; Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, South Korea; Department of Microbiology, College of Medicine, Gyeongsang National University, Jinju 52727, South Korea
| | - Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Hong-Hee Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Eunsol Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hagyu Kim
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea; Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, South Korea.
| |
Collapse
|
15
|
Kwon KW, Choi HG, Kim KS, Park SA, Kim HJ, Shin SJ. BCG-booster vaccination with HSP90-ESAT-6-HspX-RipA multivalent subunit vaccine confers durable protection against hypervirulent Mtb in mice. NPJ Vaccines 2024; 9:55. [PMID: 38459038 PMCID: PMC10923817 DOI: 10.1038/s41541-024-00847-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/21/2024] [Indexed: 03/10/2024] Open
Abstract
The quest for effective and enhanced multiantigenic tuberculosis (TB) subunit vaccine necessitates the induction of a protective pathogen-specific immune response while circumventing detrimental inflammation within the lung milieu. In line with this goal, we engineered a modified iteration of the quadrivalent vaccine, namely HSP90-ESAT-6-HspX-RipA (HEHR), which was coupled with the TLR4 adjuvant, CIA09A. The ensuing formulation was subjected to comprehensive assessment to gauge its protective efficacy against the hypervirulent Mycobacterium tuberculosis (Mtb) Haarlem clinical strain M2, following a BCG-prime boost regimen. Regardless of vaccination route, both intramuscular and subcutaneous administration with the HEHR vaccine exhibited remarkable protective efficacy in significantly reducing the Mtb bacterial burden and pulmonary inflammation. This underscores its notably superior protective potential compared to the BCG vaccine alone or a former prototype, the HSP90-E6 subunit vaccine. In addition, this superior protective efficacy was confirmed when testing a tag-free version of the HEHR vaccine. Furthermore, the protective immune determinant, represented by durable antigen-specific CD4+IFN-γ+IL-17A+ T-cells expressing a CXCR3+KLRG1- cell surface phenotype in the lung, was robustly induced in HEHR-boosted mice at 12 weeks post-challenge. Collectively, our data suggest that the BCG-prime HEHR boost vaccine regimen conferred improved and long-term protection against hypervirulent Mtb strain with robust antigen-specific Th1/Th17 responses.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
- Department of Microbiology, College of Medicine, Gyeongsang National University, Jinju, 52727, South Korea
| | - Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | | | - Shin Ae Park
- R&D Center, EyeGene Inc., Goyang, 10551, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea.
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
16
|
Hoseinpour R, Hasani A, Baradaran B, Abdolalizadeh J, Salehi R, Hasani A, Nabizadeh E, Yekani M, Hasani R, Kafil HS, Azizian K, Memar MY. Tuberculosis vaccine developments and efficient delivery systems: A comprehensive appraisal. Heliyon 2024; 10:e26193. [PMID: 38404880 PMCID: PMC10884459 DOI: 10.1016/j.heliyon.2024.e26193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Despite the widespread use of the Bacillus Calmette-Guérin (BCG) vaccine, Mycobacterium tuberculosis (MTB) continues to be a global burden. Vaccination has been proposed to prevent and treat tuberculosis (TB) infection, and several of them are in different phases of clinical trials. Though vaccine production is in progress but requires more attention. There are several TB vaccines in the trial phase, most of which are based on a combination of proteins/adjuvants or recombinant viral vectors used for selected MTB antigens. In this review, we attempted to discuss different types of TB vaccines based on the vaccine composition, the immune responses generated, and their clinical trial phases. Furthermore, we have briefly overviewed the effective delivery systems used for the TB vaccine and their effectiveness in different vaccines.
Collapse
Affiliation(s)
- Rasoul Hoseinpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Laboratory sciences and Microbiology, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Alka Hasani
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit, Sina Educational, Research, and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Hasani
- Department of Clinical Biochemistry and Applied Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Edris Nabizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Hossein Samadi Kafil
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Azizian
- Department of Microbiology, Faculty of Medicine, Kurdistan University of Medical Science, Sanandaj, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Alonso-Rodríguez N, Vianello E, van Veen S, Jenum S, Tonby K, van Riessen R, Lai X, Mortensen R, Ottenhoff THM, Dyrhol-Riise AM. Whole blood RNA signatures in tuberculosis patients receiving H56:IC31 vaccine as adjunctive therapy. Front Immunol 2024; 15:1350593. [PMID: 38433842 PMCID: PMC10904528 DOI: 10.3389/fimmu.2024.1350593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Therapeutic vaccination in tuberculosis (TB) represents a Host Directed Therapy strategy which enhances immune responses in order to improve clinical outcomes and shorten TB treatment. Previously, we have shown that the subunit H56:IC31 vaccine induced both humoral and cellular immune responses when administered to TB patients adjunctive to standard TB treatment (TBCOX2 study, NCT02503839). Here we present the longitudinal whole blood gene expression patterns in H56:IC31 vaccinated TB patients compared to controls receiving standard TB treatment only. Methods The H56:IC31 group (N=11) and Control group (N=7) underwent first-line TB treatment for 182 days. The H56:IC31 group received 5 micrograms of the H56:IC31 vaccine (Statens Serum Institut; SSI, Valneva Austria GmbH) intramuscularly at day 84 and day 140. Total RNA was extracted from whole blood samples collected in PAXgene tubes on days 0, 84, 98, 140, 154, 182 and 238. The expression level of 183 immune-related genes was measured by high-throughput microfluidic qPCR (Biomark HD system, Standard BioTools). Results The targeted gene expression profiling unveiled the upregulation of modules such as interferon (IFN) signalling genes, pattern recognition receptors and small nucleotide guanosine triphosphate (GTP)-ases in the vaccinated group compared to controls two weeks after administration of the first H56:IC31 vaccine. Additionally, the longitudinal analysis of the Adolescent Cohort Study-Correlation of Risk (ACS-COR) signature showed a progressive downregulation in both study arms towards the end of TB treatment, in congruence with reported treatment responses and clinical improvements. Still, two months after the end of TB treatment, vaccinated patients, and especially those developing both cellular and humoral vaccine responses, showed a lower expression of the ACS-COR genes compared to controls. Discussion Our data report gene expression patterns following H56:IC31 vaccination which might be interpreted as a lower risk of relapse in therapeutically vaccinated patients. Further studies are needed to conclude if these gene expression patterns could be used as prognostic biosignatures for therapeutic TB vaccine responses.
Collapse
Affiliation(s)
| | - Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Rosalie van Riessen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Xiaoran Lai
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rasmus Mortensen
- Deptartment of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Jackson S, McShane H. Challenges in Developing a Controlled Human Tuberculosis Challenge Model. Curr Top Microbiol Immunol 2024; 445:229-255. [PMID: 35332386 DOI: 10.1007/82_2022_252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Controlled human infection models (CHIMs) have provided pivotal scientific advancements, contributing to the licensure of new vaccines for many pathogens. Despite being one of the world's oldest known pathogens, there are still significant gaps in our knowledge surrounding the immunobiology of Mycobacterium tuberculosis (M. tb). Furthermore, the only licensed vaccine, BCG, is a century old and demonstrates limited efficacy in adults from endemic areas. Despite good global uptake of BCG, tuberculosis (TB) remains a silent epidemic killing 1.4 million in 2019 (WHO, Global tuberculosis report 2020). A mycobacterial CHIM could expedite the development pipeline of novel TB vaccines and provide critical understanding on the immune response to TB. However, developing a CHIM for such a complex organism is a challenging process. The first hurdle to address is which challenge agent to use, as it would not be ethical to use virulent M. tb. This chapter describes the current progress and outstanding issues in the development of a TB CHIM. Previous and current human studies include both aerosol and intradermal models using either BCG or purified protein derivative (PPD) as a surrogate agent. Future work investigating the use of attenuated M. tb is underway.
Collapse
Affiliation(s)
- Susan Jackson
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, Oxford University, Oxford, UK
| | - Helen McShane
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, Oxford University, Oxford, UK.
| |
Collapse
|
19
|
Zhuang L, Yang L, Li L, Ye Z, Gong W. Mycobacterium tuberculosis: immune response, biomarkers, and therapeutic intervention. MedComm (Beijing) 2024; 5:e419. [PMID: 38188605 PMCID: PMC10771061 DOI: 10.1002/mco2.419] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 01/09/2024] Open
Abstract
Although tuberculosis (TB) is an infectious disease, the progression of the disease following Mycobacterium tuberculosis (MTB) infection is closely associated with the host's immune response. In this review, a comprehensive analysis of TB prevention, diagnosis, and treatment was conducted from an immunological perspective. First, we delved into the host's immune response mechanisms against MTB infection as well as the immune evasion mechanisms of the bacteria. Addressing the challenges currently faced in TB diagnosis and treatment, we also emphasized the importance of protein, genetic, and immunological biomarkers, aiming to provide new insights for early and personalized diagnosis and treatment of TB. Building upon this foundation, we further discussed intervention strategies involving chemical and immunological treatments for the increasingly critical issue of drug-resistant TB and other forms of TB. Finally, we summarized TB prevention, diagnosis, and treatment challenges and put forward future perspectives. Overall, these findings provide valuable insights into the immunological aspects of TB and offer new directions toward achieving the WHO's goal of eradicating TB by 2035.
Collapse
Affiliation(s)
- Li Zhuang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and TreatmentSenior Department of Tuberculosis, the Eighth Medical Center of PLA General HospitalBeijingChina
- Senior Department of TuberculosisHebei North UniversityZhangjiakouHebeiChina
| | - Ling Yang
- Senior Department of TuberculosisHebei North UniversityZhangjiakouHebeiChina
| | - Linsheng Li
- Senior Department of TuberculosisHebei North UniversityZhangjiakouHebeiChina
| | - Zhaoyang Ye
- Senior Department of TuberculosisHebei North UniversityZhangjiakouHebeiChina
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and TreatmentSenior Department of Tuberculosis, the Eighth Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
20
|
Zhou F, Zhang D. Recent advance in the development of tuberculosis vaccines in clinical trials and virus-like particle-based vaccine candidates. Front Immunol 2023; 14:1238649. [PMID: 38022657 PMCID: PMC10652786 DOI: 10.3389/fimmu.2023.1238649] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Tuberculosis (TB) remains a serious public health threat around the world. An effective vaccine is urgently required for cost-effective, long-term control of TB. However, the only licensed vaccine Bacillus Calmette-Guerin (BCG) is limited to prevent TB for its highly variable efficacy. Substantial progress has been made in research and development (R&D) of TB vaccines in the past decades, and a dozen vaccine candidates, including live attenuated mycobacterial vaccines, killed mycobacterial vaccines, adjuvanted subunit vaccines, viral vector vaccines, and messenger RNA (mRNA) vaccines were developed in clinical trials to date. Nevertheless, many challenges to the successful authorization for the use and deployment of an effective tuberculosis vaccine remain. Therefore, it is still necessary and urgent to continue exploring new vaccine construction approaches. Virus-like particles (VLPs) present excellent prospects in the field of vaccine development because of their helpful immunological features such as being safe templates without containing viral nucleic acid, repetitive surface geometry, conformational epitopes similar to natural viruses, and enhancing both innate and adaptive immune responses. The marketization process of VLP vaccines has never stopped despite VLP vaccines face several shortcomings such as their complex and slow development process and high production cost, and several VLP-based vaccines, including vaccines against Human papillomavirus (HPV), Hepatitis B Virus (HBV) and malaria, are successfully licensed for use at the market. In this review, we provide an update on the current progress regarding the development of TB vaccines in clinical trials and seek to give an overview of VLP-based TB vaccine candidates.
Collapse
Affiliation(s)
- Fangbin Zhou
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| | - Dongmei Zhang
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Niu H, Cao Q, Zhang T, Du Y, He P, Jiao L, Wang B, Zhu B, Hu L, Zhang Y. Construction and evaluation of a novel multi-antigenic Mycobacterium tuberculosis subunit vaccine candidate BfrB-GrpE/DPC. Int Immunopharmacol 2023; 124:111060. [PMID: 37862738 DOI: 10.1016/j.intimp.2023.111060] [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: 07/16/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/22/2023]
Abstract
Tuberculosis poses a significant threat to human health due to the lack of an effective vaccine. Although promising progress has been made in the development of tuberculosis vaccines, new vaccines that broaden the antigenic repertoire need to be developed to eradicate this illness. In this study, we used Mycobacterium tuberculosis ferritin BfrB and heat-shock protein GrpE to construct a novel multi-antigenic fusion protein, BfrB-GrpE (BG). BG protein was stably overexpressed in the soluble form in Escherichia coli at a high yield and purified via sequential salt fractionation and hydrophobic chromatography. Purified BG was emulsified in an adjuvant containing N, N'-dimethyl-N, N'-dioctadecylammonium bromide, polyinosinic-polycytidylic acid, and cholesterol (DPC) to construct the BG/DPC vaccine, which stimulated strong cellular and humoral immune responses in mice. Moreover, combination of BG with our previously developed vaccine, Mtb10.4-HspX (MH), containing antigens from both the proliferating and dormant stages, significantly reduced the bacterial counts in the lungs and spleens of M. tuberculosis-infected mice. Importantly, mice that received BG + MH/DPC after M. tuberculosis H37Rv infection survived slightly better (100% survival) than those that received the BCG vaccine (80% survival), although the difference was not statistically significant. Our findings can aid in the selection of antigens and optimization of vaccination regimens to improve the efficacy of tuberculosis vaccines.
Collapse
Affiliation(s)
- Hongxia Niu
- School of Basic Medical Sciences & Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China; School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qianqian Cao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Tingting Zhang
- Lanzhou Institute of Biological Products Co., Ltd., Lanzhou, China
| | - Yunjie Du
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Pu He
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lei Jiao
- Lanzhou Institute of Biological Products Co., Ltd., Lanzhou, China
| | - Bingxiang Wang
- Lanzhou Institute of Biological Products Co., Ltd., Lanzhou, China
| | - Bingdong Zhu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lina Hu
- Lanzhou Institute of Biological Products Co., Ltd., Lanzhou, China.
| | - Ying Zhang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
22
|
Zhou F, Zhang D. Nano-Sized Chimeric Human Papillomavirus-16 L1 Virus-like Particles Displaying Mycobacterium tuberculosis Antigen Ag85B Enhance Ag85B-Specific Immune Responses in Female C57BL/c Mice. Viruses 2023; 15:2123. [PMID: 37896900 PMCID: PMC10612075 DOI: 10.3390/v15102123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Bacillus Calmette-Guerin (BCG), the only current vaccine against tuberculosis (TB) that is licensed in clinics, successfully protects infants and young children against several TB types, such as TB meningitis and miliary TB, but it is ineffective in protecting adolescents and adults against pulmonary TB. Thus, it is a matter of the utmost urgency to develop an improved and efficient TB vaccine. In this milieu, virus-like particles (VLPs) exhibit excellent characteristics in the field of vaccine development due to their numerous characteristics, including but not limited to their good safety without the risk of infection, their ability to mimic the size and structure of original viruses, and their ability to display foreign antigens on their surface to enhance the immune response. In this study, the HPV16 L1 capsid protein (HPV16L1) acted as a structural vaccine scaffold, and the extracellular domain of Ag85B was selected as the M. tb immunogen and inserted into the FG loop of the HPV16 L1 protein to construct chimeric HPV16L1/Ag85B VLPs. The chimeric HPV16L1/Ag85B VLPs were produced via the Pichia pastoris expression system and purified via discontinuous Optiprep density gradient centrifugation. The humoral and T cell-mediated immune response induced by the chimeric HPV16L1/Ag85B VLP was studied in female C57BL/c mice. We demonstrated that the insertion of the extracellular domain of Ag85B into the FG loop of HPV16L1 did not affect the in vitro stability and self-assembly of the chimeric HPV16L1/Ag85B VLPs. Importantly, it did not interfere with the immunogenicity of Ag85B. We observed that the chimeric HPV16L1/Ag85B VLPs induced higher Ag85B-specific antibody responses and elicited significant Ag85B-specific T cell immune responses in female C57BL/c mice compared with recombinant Ag85B. Our findings provide new insights into the development of novel chimeric HPV16L1/TB VLP-based vaccine platforms for controlling TB infection, which are urgently required in low-income and developing countries.
Collapse
Affiliation(s)
- Fangbin Zhou
- Department of Tropical Diseases, Naval Medical University, Shanghai 200433, China
| | - Dongmei Zhang
- Department of Tropical Diseases, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
23
|
Zhang Y, Xu JC, Hu ZD, Fan XY. Advances in protein subunit vaccines against tuberculosis. Front Immunol 2023; 14:1238586. [PMID: 37654500 PMCID: PMC10465801 DOI: 10.3389/fimmu.2023.1238586] [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: 06/12/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023] Open
Abstract
Tuberculosis (TB), also known as the "White Plague", is caused by Mycobacterium tuberculosis (Mtb). Before the COVID-19 epidemic, TB had the highest mortality rate of any single infectious disease. Vaccination is considered one of the most effective strategies for controlling TB. Despite the limitations of the Bacille Calmette-Guérin (BCG) vaccine in terms of protection against TB among adults, it is currently the only licensed TB vaccine. Recently, with the evolution of bioinformatics and structural biology techniques to screen and optimize protective antigens of Mtb, the tremendous potential of protein subunit vaccines is being exploited. Multistage subunit vaccines obtained by fusing immunodominant antigens from different stages of TB infection are being used both to prevent and to treat TB. Additionally, the development of novel adjuvants is compensating for weaknesses of immunogenicity, which is conducive to the flourishing of subunit vaccines. With advances in the development of animal models, preclinical vaccine protection assessments are becoming increasingly accurate. This review summarizes progress in the research of protein subunit TB vaccines during the past decades to facilitate the further optimization of protein subunit vaccines that may eradicate TB.
Collapse
Affiliation(s)
- Ying Zhang
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jin-chuan Xu
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Zhi-dong Hu
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai, China
| | - Xiao-yong Fan
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
25
|
Zhuang L, Ye Z, Li L, Yang L, Gong W. Next-Generation TB Vaccines: Progress, Challenges, and Prospects. Vaccines (Basel) 2023; 11:1304. [PMID: 37631874 PMCID: PMC10457792 DOI: 10.3390/vaccines11081304] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), is a prevalent global infectious disease and a leading cause of mortality worldwide. Currently, the only available vaccine for TB prevention is Bacillus Calmette-Guérin (BCG). However, BCG demonstrates limited efficacy, particularly in adults. Efforts to develop effective TB vaccines have been ongoing for nearly a century. In this review, we have examined the current obstacles in TB vaccine research and emphasized the significance of understanding the interaction mechanism between MTB and hosts in order to provide new avenues for research and establish a solid foundation for the development of novel vaccines. We have also assessed various TB vaccine candidates, including inactivated vaccines, attenuated live vaccines, subunit vaccines, viral vector vaccines, DNA vaccines, and the emerging mRNA vaccines as well as virus-like particle (VLP)-based vaccines, which are currently in preclinical stages or clinical trials. Furthermore, we have discussed the challenges and opportunities associated with developing different types of TB vaccines and outlined future directions for TB vaccine research, aiming to expedite the development of effective vaccines. This comprehensive review offers a summary of the progress made in the field of novel TB vaccines.
Collapse
Affiliation(s)
- Li Zhuang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
- Hebei North University, Zhangjiakou 075000, China
| | - Zhaoyang Ye
- Hebei North University, Zhangjiakou 075000, China
| | - Linsheng Li
- Hebei North University, Zhangjiakou 075000, China
| | - Ling Yang
- Hebei North University, Zhangjiakou 075000, China
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| |
Collapse
|
26
|
Wang R, Fan X, Jiang Y, Li G, Li M, Zhao X, Luan X, Deng Y, Chen Z, Liu H, Wan K. Immunogenicity and efficacy analyses of EPC002, ECA006, and EPCP009 protein subunit combinations as tuberculosis vaccine candidates. Vaccine 2023:S0264-410X(23)00385-7. [PMID: 37225573 DOI: 10.1016/j.vaccine.2023.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/13/2023] [Accepted: 04/03/2023] [Indexed: 05/26/2023]
Abstract
Tuberculosis (TB) is the leading cause of death from infectious diseases worldwide, and developing a new TB vaccine is a priority for TB control. Combining multiple immunodominant antigens to form a novel multicomponent vaccine with broad-spectrum antigens to induce protective immune responses is a trend in TB vaccine development. In this study, we used T-cell epitope-rich protein subunits to construct three antigenic combinations: EPC002, ECA006, and EPCP009. Fusion expression of purified protein EPC002f (CFP-10-linker-ESAT-6-linker-nPPE18), ECA006f (CFP-10-linker-ESAT-6-linker-Ag85B), and EPCP009f (CFP-10-linker-ESAT-6-linker-nPPE18-linker-nPstS1) and recombinant purified protein mixtures EPC002m (mix of CFP-10, ESAT-6, and nPPE18), ECA006m (mix of CFP-10, ESAT-6, and Ag85B), and EPCP009m (mix of CFP-10, ESAT-6, nPPE18, and nPstS1) were used as antigens, formulated with alum adjuvant, and the immunogenicity and efficacy were analyzed using immunity experiments with BALB/c mice. All protein-immunized groups elicited higher levels of humoral immunity, including IgG and IgG1. The IgG2a/IgG1 ratio of the EPCP009m-immunized group was the highest, followed by that of the EPCP009f-immunized group, which was significantly higher than the ratios of the other four groups. The multiplex microsphere-based cytokine immunoassay revealed that EPCP009f and EPCP009m induced the production of a wider range of cytokines than EPC002f, EPC002m, ECA006f, and ECA006m, which included Th1-type (IL-2, IFN-γ, TNF-α), Th2-type (IL-4, IL-6, IL-10), Th17-type (IL-17), and other proinflammatory cytokines (GM-CSF, IL-12). The enzyme-linked immunospot assays demonstrated that the EPCP009f- and EPCP009m-immunized groups had significantly higher amounts of IFN-γ than the other four groups. The in vitro mycobacterial growth inhibition assay demonstrated that EPCP009m inhibited Mycobacterium tuberculosis (Mtb) growth most strongly, followed by EPCP009f, which was significantly better than that of the other four vaccine candidates. These results indicated that EPCP009m containing four immunodominant antigens exhibited better immunogenicity and Mtb growth inhibition in vitro and may be a promising candidate vaccine for the control of TB.
Collapse
Affiliation(s)
- Ruihuan Wang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xueting Fan
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yi Jiang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guilian Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Machao Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuqin Zhao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuli Luan
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yunli Deng
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; Community Health Management Service Center, Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Zixin Chen
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Infection Control, Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Haican Liu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Kanglin Wan
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
27
|
Biswas B, Kumar Misra T, Ray D, Majumder T, Kanti Bandyopadhyay T, Kumar Bhowmick T. Current Therapeutic Delivery Approaches Using Nanocarriers for the Treatment of Tuberculosis Disease. Int J Pharm 2023; 640:123018. [PMID: 37149113 DOI: 10.1016/j.ijpharm.2023.123018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/04/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023]
Abstract
Tuberculosis is a major health issue globally and a leading cause of death due to the infective microorganism Mycobacterium tuberculosis. Treatment of drug resistance tuberculosis requires longer treatment with multiple daily doses of drugs. Unfortunately, these drugs are often associated with poor patient compliance. In this situation, a need has been felt for the less toxic, shorter, and more effective treatment of the infected tuberculosis patients. Current research to develop novel anti-tubercular drugs shows hope for better management of the disease. Research on drug targeting and precise delivery of the old anti-tubercular drugs with the help of nanotechnology is promising for effective treatment. This review has discussed the status currently available treatments for tuberculosis patients infected with Mycobacterium alone or in comorbid conditions like diabetes, HIV and cancer. This review also highlighted the challenges in the current treatment and research on the novel anti-tubercular drugs to prevent multi-drug-resistant tuberculosis. It presents the research highlights on the targeted delivery of anti-tubercular drugs using different nanocarriers for preventing multi-drug resistant tuberculosis. Report has shown the importance and development of the research on nanocarriers mediated anti-tubercular delivery of the drugs to overcome the current challenges in tuberculosis treatment.
Collapse
Affiliation(s)
- Bhabatush Biswas
- Department of Bioengineering, National Institute of Technology Agartala, West Tripura - 799046, India
| | - Tarun Kumar Misra
- Department of Chemistry, National Institute of Technology Agartala, West Tripura - 799046, India
| | - Debasish Ray
- Agartala Govt. Medical College, Agartala, 799006, Tripura - 799006, India
| | - Tapan Majumder
- Agartala Govt. Medical College, Agartala, 799006, Tripura - 799006, India
| | - Tarun Kanti Bandyopadhyay
- Department of Bioengineering, National Institute of Technology Agartala, West Tripura - 799046, India
| | - Tridib Kumar Bhowmick
- Department of Bioengineering, National Institute of Technology Agartala, West Tripura - 799046, India.
| |
Collapse
|
28
|
Hanthamrongwit J, Aruvornlop P, Saelee C, Wanta N, Poneksawat P, Soe PT, Kyaw SP, Khaenam P, Warit S, Valentini D, Mahasirimongkol S, Dhepakson P, Soonthornchartrawat S, Chootong P, Leepiyasakulchai C. Peptide microarray-based identification of dormancy-associated Mycobacterium tuberculosis antigens inducing immune responses among latent tuberculosis infection individuals in Thailand. Sci Rep 2023; 13:6978. [PMID: 37117690 PMCID: PMC10141872 DOI: 10.1038/s41598-023-34307-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/27/2023] [Indexed: 04/30/2023] Open
Abstract
Multi-stage tuberculosis (TB) vaccines composed of active- and dormancy-associated antigens are promising to trigger the immune protection against all TB stages. However, scientists are still in quest of the suitable vaccine candidates. In this study, we identified the potential targets for this vaccine in a high TB burden country, Thailand. Peptide microarray was applied to gauge IgA and IgG antibodies specific to 16,730 linear epitopes of 52 dormancy-associated Mycobacterium tuberculosis (M. tb) proteins in three study groups: active tuberculosis (ATB), latent tuberculosis infection (LTBI) and endemic healthy control (EHC). Preferential IgA recognition against epitopes of dormancy-associated proteins was identified in LTBI group. Validation of these findings revealed that LTBI subjects exhibited the greater levels of Rv2659c- and Rv1738-specific IgA than those of household contacts, but less than did ATB subjects. Frequencies of IFNγ-producing CD4+ and CD8+ T cells induced by proteins Rv2659c and Rv1738 were higher in LTBI than ATB individuals. The results indicated that LTBI group in a high TB burden country demonstrated cell-mediated immune response to proteins Rv2659c and Rv1738 stronger than those of ATB. These immune responses likely contribute to natural protection against dormant M. tb and might be potential targets for a multi-stage TB vaccine.
Collapse
Affiliation(s)
- Jariya Hanthamrongwit
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Panicha Aruvornlop
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Chutiphon Saelee
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Nattiya Wanta
- Center of Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Passarun Poneksawat
- Center of Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Phyu Thwe Soe
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
- Department of Medical Laboratory Technology, University of Medical Technology, Mandalay, 05071, Myanmar
| | - Soe Paing Kyaw
- Clinical Pathology Laboratory, (1000) Bedded General Hospital, Nay Pyi Taw, 15011, Myanmar
| | - Prasong Khaenam
- Center of Standardization and Product Validation, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Saradee Warit
- Industrial Tuberculosis Team, Industrial Medical Molecular Biotechnology Research Group, BIOTEC, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, 12120, Thailand
| | - Davide Valentini
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Surakameth Mahasirimongkol
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Panadda Dhepakson
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Sakulrat Soonthornchartrawat
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Chaniya Leepiyasakulchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
29
|
Wilson L, Gracie L, Kidy F, Thomas GN, Nirantharakumar K, Greenfield S, Manaseki-Holland S, Ward DJ, Gooden TE. Safety and efficacy of tuberculosis vaccine candidates in low- and middle-income countries: a systematic review of randomised controlled clinical trials. BMC Infect Dis 2023; 23:120. [PMID: 36829123 PMCID: PMC9951834 DOI: 10.1186/s12879-023-08092-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Tuberculosis (TB) remains a leading cause of death worldwide, with 98% of cases occurring in low- and middle-income countries (LMICs). The only vaccine licenced for the prevention of TB has limited protection for adolescents, adults and vulnerable populations. A safe and effective vaccine for all populations at risk is imperative to achieve global elimination of TB. We aimed to systematically review the efficacy and safety of TB vaccine candidates in late-phase clinical trials conducted in LMICs. METHODS Medline, Embase, CENTRAL, PubMed, Clinicaltrials.gov and Greylit.org were searched in June 2021 to identify phase 2 or later clinical randomised controlled trials that report the efficacy or safety (adverse events) of TB vaccine candidates with participants of any age living in an LMIC. TB vaccine candidates listed in the 2020 WHO Global TB Report were eligible for inclusion aside from BCG revaccination. Trials were excluded if all participants had active TB at baseline. Two reviewers independently assessed papers for eligibility, and for bias and quality using the Risk of Bias 2 tool and GRADE guidelines, respectively. We report efficacy rates and frequencies of adverse events from each included trial where available and qualitatively synthesise the findings. RESULTS Thirteen papers representing eleven trials met our inclusion criteria. Seven vaccine candidates were reviewed across seven countries: M72/AS01, RUTI, VPM1002, H56:IC31, MTBVAC, DAR-901 and ID93 + GLA-SE. Two trials reported on efficacy: an efficacy rate of 54% (95% CI 11.5, 76.2) was reported for M72/AS01 in adults with latent TB and 3% (95% CI -13.9, 17.7) for DAR-901 in healthy adolescents. However, the latter trial was underpowered. All vaccine candidates had comparable occurrences of adverse events between treatment arms and demonstrated acceptable safety profiles; though, RUTI resulted in one serious complication in a person living with HIV. M72/AS01 was the only vaccine considered safe across a diverse group of people including people living with HIV or latent TB and healthy infants and adolescents. CONCLUSION Further efficacy trials for M72/AS01 are warranted to include additional populations at risk where safety has been demonstrated. Further safety trials are needed for the remaining vaccine candidates to confirm safety in vulnerable populations.
Collapse
Affiliation(s)
- Lydia Wilson
- grid.439591.30000 0004 0399 2770Homerton University Hospital, Homerton Row, London, UK
| | - Lara Gracie
- grid.6572.60000 0004 1936 7486Institute of Medical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Farah Kidy
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| | - G. Neil Thomas
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| | - Krishnarajah Nirantharakumar
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| | - Sheila Greenfield
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| | - Semira Manaseki-Holland
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| | - Derek J. Ward
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| | - Tiffany E. Gooden
- grid.6572.60000 0004 1936 7486Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
| |
Collapse
|
30
|
Romano M, Squeglia F, Kramarska E, Barra G, Choi HG, Kim HJ, Ruggiero A, Berisio R. A Structural View at Vaccine Development against M. tuberculosis. Cells 2023; 12:317. [PMID: 36672252 PMCID: PMC9857197 DOI: 10.3390/cells12020317] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Tuberculosis (TB) is still the leading global cause of death from an infectious bacterial agent. Limiting tuberculosis epidemic spread is therefore an urgent global public health priority. As stated by the WHO, to stop the spread of the disease we need a new vaccine, with better coverage than the current Mycobacterium bovis BCG vaccine. This vaccine was first used in 1921 and, since then, there are still no new licensed tuberculosis vaccines. However, there is extremely active research in the field, with a steep acceleration in the past decades, due to the advance of technologies and more rational vaccine design strategies. This review aims to gather latest updates in vaccine development in the various clinical phases and to underline the contribution of Structural Vaccinology (SV) to the development of safer and effective antigens. In particular, SV and the development of vaccine adjuvants is making the use of subunit vaccines, which are the safest albeit the less antigenic ones, an achievable goal. Indeed, subunit vaccines overcome safety concerns but need to be rationally re-engineered to enhance their immunostimulating effects. The larger availability of antigen structural information as well as a better understanding of the complex host immune response to TB infection is a strong premise for a further acceleration of TB vaccine development.
Collapse
Affiliation(s)
- Maria Romano
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Flavia Squeglia
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Eliza Kramarska
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Giovanni Barra
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Alessia Ruggiero
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, IBB, CNR, 80131 Naples, Italy
| |
Collapse
|
31
|
Rawat BS, Kumar D, Soni V, Rosenn EH. Therapeutic Potentials of Immunometabolomic Modulations Induced by Tuberculosis Vaccination. Vaccines (Basel) 2022; 10:vaccines10122127. [PMID: 36560537 PMCID: PMC9781011 DOI: 10.3390/vaccines10122127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolomics is emerging as a promising tool to understand the effect of immunometabolism for the development of novel host-directed alternative therapies. Immunometabolism can modulate both innate and adaptive immunity in response to pathogens and vaccinations. For instance, infections can affect lipid and amino acid metabolism while vaccines can trigger bile acid and carbohydrate pathways. Metabolomics as a vaccinomics tool, can provide a broader picture of vaccine-induced biochemical changes and pave a path to potentiate the vaccine efficacy. Its integration with other systems biology tools or treatment modes can enhance the cure, response rate, and control over the emergence of drug-resistant strains. Mycobacterium tuberculosis (Mtb) infection can remodel the host metabolism for its survival, while there are many biochemical pathways that the host adjusts to combat the infection. Similarly, the anti-TB vaccine, Bacillus Calmette-Guerin (BCG), was also found to affect the host metabolic pathways thus modulating immune responses. In this review, we highlight the metabolomic schema of the anti-TB vaccine and its therapeutic applications. Rewiring of immune metabolism upon BCG vaccination induces different signaling pathways which lead to epigenetic modifications underlying trained immunity. Metabolic pathways such as glycolysis, central carbon metabolism, and cholesterol synthesis play an important role in these aspects of immunity. Trained immunity and its applications are increasing day by day and it can be used to develop the next generation of vaccines to treat various other infections and orphan diseases. Our goal is to provide fresh insight into this direction and connect various dots to develop a conceptual framework.
Collapse
Affiliation(s)
- Bhupendra Singh Rawat
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Deepak Kumar
- Department of Zoology, University of Rajasthan, Jaipur 302004, Rajasthan, India
| | - Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Correspondence:
| | - Eric H. Rosenn
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
32
|
Bouzeyen R, Javid B. Therapeutic Vaccines for Tuberculosis: An Overview. Front Immunol 2022; 13:878471. [PMID: 35812462 PMCID: PMC9263712 DOI: 10.3389/fimmu.2022.878471] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis is the world’s deadliest bacterial infection, resulting in more than 1.4 million deaths annually. The emergence of drug-resistance to first-line antibiotic therapy poses a threat to successful treatment, and novel therapeutic options are required, particularly for drug-resistant tuberculosis. One modality emerging for TB treatment is therapeutic vaccination. As opposed to preventative vaccination – the aim of which is to prevent getting infected by M. tuberculosis or developing active tuberculosis, the purpose of therapeutic vaccination is as adjunctive treatment of TB or to prevent relapse following cure. Several candidate therapeutic vaccines, using killed whole-cell or live attenuated mycobacteria, mycobacterial fragments and viral vectored vaccines are in current clinical trials. Other modes of passive immunization, including monoclonal antibodies directed against M. tuberculosis antigens are in various pre-clinical stages of development. Here, we will discuss these various therapeutics and their proposed mechanisms of action. Although the full clinical utility of therapeutic vaccination for the treatment of tuberculosis is yet to be established, they hold potential as useful adjunct therapies.
Collapse
|
33
|
Lu Y, Ning H, Kang J, Bai G, Zhou L, Kang Y, Wu Z, Tian M, Zhao J, Ma Y, Bai Y. Cyclic-di-AMP Phosphodiesterase Elicits Protective Immune Responses Against Mycobacterium tuberculosis H37Ra Infection in Mice. Front Cell Infect Microbiol 2022; 12:871135. [PMID: 35811674 PMCID: PMC9256937 DOI: 10.3389/fcimb.2022.871135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Many antigens from Mycobacterium tuberculosis (M. tuberculosis) have been demonstrated as strong immunogens and proved to have application potential as vaccine candidate antigens. Cyclic di-AMP (c-di-AMP) as a bacterial second messenger regulates various bacterial processes as well as the host immune responses. Rv2837c, the c-di-AMP phosphodiesterase (CnpB), was found to be relative to virulence of M. tuberculosis and interference with host innate immune response. In this study, recombinant CnpB was administered subcutaneously to mice. We found that CnpB had strong immunogenicity and induced high levels of humoral response and lung mucosal immunity after M. tuberculosis intranasally infection. CnpB immunization stimulated splenocyte proliferation and the increasing number of activated NK cells but had little effects on Th1/Th2 cellular immune responses in spleens. However, CnpB induced significant Th1/Th2 cellular immune responses with a decreased number of T and B cells in the lungs, and significantly recruits of CD4+ and CD8+ T cells after M. tuberculosis attenuated strain H37Ra infection. Besides, we first reported that CnpB could stimulate IFN-β expression transitorily and inhibit the autophagy of macrophages in vitro. In mice intranasally infection model, CnpB immunization alleviated pathological changes and reduced M. tuberculosis H37Ra loads in the lungs. Thus, our results suggested that CnpB interferes with host innate and adaptive immune responses and confers protection against M. tuberculosis respiratory infection, which should be considered in vaccine development as well as a drug target.
Collapse
Affiliation(s)
- Yanzhi Lu
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Huanhuan Ning
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Jian Kang
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Lei Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital, Air Force Medical University, Xi’an, China
| | - Yali Kang
- Department of Physiology, Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Zhengfeng Wu
- Student Brigade, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Maolin Tian
- Student Brigade, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Junhao Zhao
- Student Brigade, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Yueyun Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, Air Force Medical University, Xi’an, China
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, China
- *Correspondence: Yinlan Bai, ; Yueyun Ma,
| | - Yinlan Bai
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
- *Correspondence: Yinlan Bai, ; Yueyun Ma,
| |
Collapse
|
34
|
Khan Z, Ualiyeva D, Amissah OB, Sapkota S, Hameed HMA, Zhang T. Insight Into Novel Anti-tuberculosis Vaccines by Using Immunoinformatics Approaches. Front Microbiol 2022; 13:866873. [PMID: 35722321 PMCID: PMC9201507 DOI: 10.3389/fmicb.2022.866873] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/16/2022] [Indexed: 01/26/2023] Open
Abstract
Tuberculosis (TB), an infectious disease, has been a leading cause of morbidity and mortality for decades. The causative agent of TB is the Mycobacterium tuberculosis (Mtb) which can infects various parts of the body, mainly the lungs in pulmonary TB cases. Mycobacterium bovis Bacillus Calmette–Guerin (BCG) is the only approved vaccine for TB, but its efficiency to combat pulmonary TB is limited. Multidrug-resistant (MDR) TB and extensive drug-resistant (XDR) TB requires the evolution of more potent vaccines. Therefore, this research aims to generate a universal TB subunit vaccine using advanced immunoinformatics techniques. In generating a novel multiepitope subunit vaccine, we selected the conserved and experimentally confirmed antigens Rv0058, Rv0101, and Rv3343. After a rigorous evaluation, the top candidates from predicted Helper T-lymphocytes (HTL), Cytotoxic T-lymphocytes (CTL), and B-cell epitopes were considered potential vaccine candidates. Immunogenicity was enhanced by the addition of an adjuvant to the ultimate construct of the vaccine. B-cell epitopes predictions guaranteed the eventual induction of a humoral response. Thereafter, dynamics simulations and molecular docking validated the vaccine-receptor complex’s stability and high affinity for the immune receptor TLR-3. Also, immune simulations revealed the significantly elevated levels of immunoglobulins such as IgM, cytokines such as interleukin-2, helper T (Th) cells, and cytotoxic T-cell populations. These results agreed with the actual inflammatory response and showed rapid antigen clearance after manifold exposure. Finally, the E. coli K12 strain was confirmed via in-silico cloning for quality expression. Nevertheless, in vivo experiments should be performed to validate the safety of the proposed vaccine and its inherent ability to prevent TB infection.
Collapse
Affiliation(s)
- Zafran Khan
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou, China.,China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Daniya Ualiyeva
- University of Chinese Academy of Sciences, Beijing, China.,Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Obed Boadi Amissah
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sanjeep Sapkota
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou, China.,China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - H M Adnan Hameed
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou, China.,China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou, China.,China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| |
Collapse
|
35
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
36
|
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a leading cause of mortality and morbidity due to a single infectious agent. Aerosol infection with Mtb can result in a range of responses from elimination, active, incipient, subclinical, and latent Mtb infections (LTBI), depending on the host's immune response and the dose and nature of infecting bacilli. Currently, BCG is the only vaccine approved to prevent TB. Although BCG confers protection against severe forms of childhood TB, its use in adults and those with comorbid conditions, such as HIV infection, is questionable. Novel vaccines, including recombinant BCG (rBCG), were developed to improve BCG's efficacy and use as an alternative to BCG in a vulnerable population. The first-generation rBCG vaccines had different Mtb antigens and were tested as a prime, prime-boost, or immunotherapeutic intervention. The novel vaccines target one or more of the following requirements, namely prevention of infection (POI), prevention of disease (POD), prevention of recurrence (POR), and therapeutic vaccines to treat a TB disease. Several vaccine candidates currently in development are classified into four primary categories: live attenuated whole-cell vaccine, inactivated whole-cell vaccine, adjuvanted protein subunit vaccine, and viral-vectored vaccine. Each vaccine's immunogenicity, safety, and efficacy are tested in preclinical animal models and further validated through various phases of clinical trials. This chapter summarizes the various TB vaccine candidates under different clinical trial stages and promises better protection against TB.
Collapse
Affiliation(s)
- Radha Gopalaswamy
- Department of Bacteriology, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamilnadu, India
| | - Selvakumar Subbian
- The Public Health Research Institute Center at New Jersey Medical School, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
37
|
Suliman S, Pelzer PT, Shaku M, Rozot V, Mendelsohn SC. Meeting report: Virtual Global Forum on Tuberculosis Vaccines, 20-22 April 2021. Vaccine 2021; 39:7223-7229. [PMID: 34538522 PMCID: PMC8441545 DOI: 10.1016/j.vaccine.2021.08.094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/26/2021] [Indexed: 12/28/2022]
Abstract
The Global Forum on Tuberculosis (TB) Vaccines was held virtually from 20 to 22 April 2021, marking its 20th anniversary. The Global Forum on TB Vaccines is the world's largest gathering of stakeholders striving to develop new vaccines to prevent TB. The program included more than 60 speakers in 11 scientific sessions, panel discussions, and workshops. It provided an overview of the state of the field, and an opportunity to share the latest research findings, as well as new and innovative approaches to TB vaccine research and development (R&D). This year, it was held against the backdrop of the COVID-19 pandemic and convened researchers, developers, funders, and other stakeholders remotely to discuss opportunities and challenges for TB vaccine R&D in these unprecedented times.
Collapse
Affiliation(s)
- Sara Suliman
- Stop TB Partnership Working Group on New TB Vaccines, New York, NY, USA; Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Puck T Pelzer
- Stop TB Partnership Working Group on New TB Vaccines, New York, NY, USA; KNCV Tuberculosis Foundation, The Hague, the Netherlands
| | - Moagi Shaku
- Stop TB Partnership Working Group on New TB Vaccines, New York, NY, USA
| | - Virginie Rozot
- Stop TB Partnership Working Group on New TB Vaccines, New York, NY, USA; South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, Wernher and Beit South Building, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Simon C Mendelsohn
- Stop TB Partnership Working Group on New TB Vaccines, New York, NY, USA; South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, Wernher and Beit South Building, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| |
Collapse
|
38
|
Qu M, Zhou X, Li H. BCG vaccination strategies against tuberculosis: updates and perspectives. Hum Vaccin Immunother 2021; 17:5284-5295. [PMID: 34856853 DOI: 10.1080/21645515.2021.2007711] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Bacillus Calmette-Guérin (BCG) is the only licensed vaccine against tuberculosis (TB). However, BCG has variable efficacy and cannot completely prevent TB infection and transmission. Therefore, the worldwide prevalence of TB calls for urgent development of a more effective TB vaccine. In the absence of other approved vaccines, it is also necessary to improve the efficacy of BCG itself. Intravenous (IV) BCG administration and BCG revaccination strategies have recently shown promising results for clinical usage. Therefore, it is necessary for us to revisit the BCG vaccination strategies and summarize the current research updates related to BCG vaccination. This literature review provides an updated overview and perspectives of the immunization strategies against TB using BCG, which may inspire the following research on TB vaccine development.
Collapse
Affiliation(s)
- Mengjin Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, China Agricultural University, Beijing, China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
39
|
Joshi H, Kandari D, Bhatnagar R. Insights into the molecular determinants involved in Mycobacterium tuberculosis persistence and their therapeutic implications. Virulence 2021; 12:2721-2749. [PMID: 34637683 PMCID: PMC8565819 DOI: 10.1080/21505594.2021.1990660] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
The establishment of persistent infections and the reactivation of persistent bacteria to active bacilli are the two hurdles in effective tuberculosis treatment. Mycobacterium tuberculosis, an etiologic tuberculosis agent, adapts to numerous antibiotics and resists the host immune system causing a disease of public health concern. Extensive research has been employed to combat this disease due to its sheer ability to persist in the host system, undetected, waiting for the opportunity to declare itself. Persisters are a bacterial subpopulation that possesses transient tolerance to high doses of antibiotics. There are certain inherent mechanisms that facilitate the persister cell formation in Mycobacterium tuberculosis, some of those had been characterized in the past namely, stringent response, transcriptional regulators, energy production pathways, lipid metabolism, cell wall remodeling enzymes, phosphate metabolism, and proteasome protein degradation. This article reviews the recent advancements made in various in vitro persistence models that assist to unravel the mechanisms involved in the persister cell formation and to hunt for the possible preventive or treatment measures. To tackle the persister population the immunodominant proteins that express specifically at the latent phase of infection can be used for diagnosis to distinguish between the active and latent tuberculosis, as well as to select potential drug or vaccine candidates. In addition, we discuss the genes engaged in the persistence to get more insights into resuscitation and persister cell formation. The in-depth understanding of persistent cells of mycobacteria can certainly unravel novel ways to target the pathogen and tackle its persistence.
Collapse
Affiliation(s)
- Hemant Joshi
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Divya Kandari
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Amity University of Rajasthan, Jaipur, Rajasthan, India
| |
Collapse
|
40
|
Jenum S, Tonby K, Rueegg CS, Rühwald M, Kristiansen MP, Bang P, Olsen IC, Sellæg K, Røstad K, Mustafa T, Taskén K, Kvale D, Mortensen R, Dyrhol-Riise AM. A Phase I/II randomized trial of H56:IC31 vaccination and adjunctive cyclooxygenase-2-inhibitor treatment in tuberculosis patients. Nat Commun 2021; 12:6774. [PMID: 34811370 PMCID: PMC8608791 DOI: 10.1038/s41467-021-27029-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022] Open
Abstract
Host-directed-therapy strategies are warranted to fight tuberculosis. Here we assess the safety and immunogenicity of adjunctive vaccination with the H56:IC31 candidate and cyclooxygenase-2-inhibitor treatment (etoricoxib) in pulmonary and extra-pulmonary tuberculosis patients in a randomized open-label phase I/II clinical trial (TBCOX2, NCT02503839). A total of 222 patients were screened, 51 enrolled and randomized; 13 in the etoricoxib-group, 14 in the H56:IC31-group, 12 in the etoricoxib+H56:IC31-group and 12 controls. Three Serious Adverse Events were reported in the etoricoxib-groups; two urticarial rash and one possible disease progression, no Serious Adverse Events were vaccine related. H56:IC31 induces robust expansion of antigen-specific T-cells analyzed by fluorospot and flow cytometry, and higher proportion of seroconversions. Etoricoxib reduced H56:IC31-induced T-cell responses. Here, we show the first clinical data that H56:IC31 vaccination is safe and immunogenic in tuberculosis patients, supporting further studies of H56:IC31 as a host-directed-therapy strategy. Although etoricoxib appears safe, our data do not support therapy with adjunctive cyclooxygenase-2-inhibitors. Modulating the host immune response during tuberculosis is an emerging and critical advance in the therapeutic approach. Here the authors present data from a first-in-human phase I/II randomised trial on the safety and immunogenicity of adjuvant therapy of the H56:IC31 vaccine and cyclooxygenase-2 inhibitors in patients with tuberculosis.
Collapse
Affiliation(s)
- Synne Jenum
- Department of Infectious diseases, Oslo University Hospital, P.O box 4952, Nydalen, N-0424, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious diseases, Oslo University Hospital, P.O box 4952, Nydalen, N-0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, P.O box 1171, Blindern, N-0318, Oslo, Norway
| | - Corina S Rueegg
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, P.O box 4950, Nydalen, N-0424, Oslo, Norway
| | - Morten Rühwald
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut Artillerivej 5, 2300, Copenhagen, Denmark.,Foundation of Innovative New Diagnostics (FIND), the global alliance for diagnostics, Chemin des Mines 9, 1201, Geneva, Switzerland
| | - Max P Kristiansen
- Center for Vaccine Research, Vaccine Development, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Peter Bang
- Center for Vaccine Research, Vaccine Development, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Inge Christoffer Olsen
- Department of Research Support for Clinical Trials,, Oslo University Hospital, P.O box 4950, Nydalen, N-0424, Oslo, Norway
| | - Kjersti Sellæg
- Department of Infectious diseases, Oslo University Hospital, P.O box 4952, Nydalen, N-0424, Oslo, Norway
| | - Kjerstin Røstad
- Department of Infectious diseases, Oslo University Hospital, P.O box 4952, Nydalen, N-0424, Oslo, Norway
| | - Tehmina Mustafa
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, P.O box 7804, N-5020, Bergen, Norway.,Department of Thoracic Medicine, Haukeland University Hospital, P.O box 1400, N-5021, Bergen, Norway
| | - Kjetil Taskén
- Institute of Clinical Medicine, University of Oslo, P.O box 1171, Blindern, N-0318, Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Dag Kvale
- Department of Infectious diseases, Oslo University Hospital, P.O box 4952, Nydalen, N-0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, P.O box 1171, Blindern, N-0318, Oslo, Norway
| | - Rasmus Mortensen
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut Artillerivej 5, 2300, Copenhagen, Denmark
| | - Anne Ma Dyrhol-Riise
- Department of Infectious diseases, Oslo University Hospital, P.O box 4952, Nydalen, N-0424, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, P.O box 1171, Blindern, N-0318, Oslo, Norway.
| |
Collapse
|
41
|
Albutti A. An integrated computational framework to design a multi-epitopes vaccine against Mycobacterium tuberculosis. Sci Rep 2021; 11:21929. [PMID: 34753983 PMCID: PMC8578660 DOI: 10.1038/s41598-021-01283-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Tuberculosis (TB) is a highly contagious disease that mostly affects the lungs and is caused by a bacterial pathogen, Mycobacterium tuberculosis. The associated mortality rate of TB is much higher compared to any other disease and the situation is more worrisome by the rapid emergence of drug resistant strains. Bacillus Calmette-Guerin (BCG) is the only licensed attenuated vaccine available for use in humans however, many countries have stopped its use as it fails to confer protective immunity. Therefore, urgent efforts are required to identify new and safe vaccine candidates that are not only provide high immune protection but also have broad spectrum applicability. Considering this, herein, I performed an extensive computational vaccine analysis to investigate 200 complete sequenced genomes of M. tuberculosis to identify core vaccine candidates that harbor safe, antigenic, non-toxic, and non-allergic epitopes. To overcome literature reported limitations of epitope-based vaccines, I carried out additional analysis by designing a multi-epitopes vaccine to achieve maximum protective immunity as well as to make experimental follow up studies easy by selecting a vaccine that can be easily analyzed because of its favorable physiochemical profile. Based on these analyses, I identified two potential vaccine proteins that fulfill all required vaccine properties. These two vaccine proteins are diacylglycerol acyltransferase and ESAT-6-like protein. Epitopes: DSGGYNANS from diacylglycerol acyltransferase and AGVQYSRAD, ADEEQQQAL, and VSRADEEQQ from ESAT-6-like protein were found to cover all necessary parameters and thus used in a multi-epitope vaccine construct. The designed vaccine is depicting a high binding affinity for different immune receptors and shows stable dynamics and rigorous van der Waals and electrostatic binding energies. The vaccine also simulates profound primary, secondary, tertiary immunoglobulin production as well as high interleukins and interferons count. In summary, the designed vaccine is ideal to be evaluated experimentally to decipher its real biological efficacy in controlling drug resistant infections of M. tuberculosis.
Collapse
Affiliation(s)
- Aqel Albutti
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.
| |
Collapse
|
42
|
Ong GH, Lian BSX, Kawasaki T, Kawai T. Exploration of Pattern Recognition Receptor Agonists as Candidate Adjuvants. Front Cell Infect Microbiol 2021; 11:745016. [PMID: 34692565 PMCID: PMC8526852 DOI: 10.3389/fcimb.2021.745016] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Adjuvants are used to maximize the potency of vaccines by enhancing immune reactions. Components of adjuvants include pathogen-associated molecular patterns (PAMPs) and damage-associate molecular patterns (DAMPs) that are agonists for innate immune receptors. Innate immune responses are usually activated when pathogen recognition receptors (PRRs) recognize PAMPs derived from invading pathogens or DAMPs released by host cells upon tissue damage. Activation of innate immunity by PRR agonists in adjuvants activates acquired immune responses, which is crucial to enhance immune reactions against the targeted pathogen. For example, agonists for Toll-like receptors have yielded promising results as adjuvants, which target PRR as adjuvant candidates. However, a comprehensive understanding of the type of immunological reaction against agonists for PRRs is essential to ensure the safety and reliability of vaccine adjuvants. This review provides an overview of the current progress in development of PRR agonists as vaccine adjuvants, the molecular mechanisms that underlie activation of immune responses, and the enhancement of vaccine efficacy by these potential adjuvant candidates.
Collapse
Affiliation(s)
- Guang Han Ong
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Benedict Shi Xiang Lian
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Takumi Kawasaki
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| |
Collapse
|
43
|
Mustafa AS. Adjuvants and Antigen-Delivery Systems for Subunit Vaccines against Tuberculosis. Vaccines (Basel) 2021; 9:vaccines9090972. [PMID: 34579209 PMCID: PMC8472090 DOI: 10.3390/vaccines9090972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
The only licensed vaccine against tuberculosis is BCG. However, BCG has failed to provide consistent protection against tuberculosis, especially pulmonary disease in adults. Furthermore, the use of BCG is contraindicated in immunocompromised subjects. The research towards the development of new vaccines against TB includes the use of Mycobacterium tuberculosis antigens as subunit vaccines. Such vaccines may be used either alone or in the prime-boost model in BCG-vaccinated people. However, the antigens for subunit vaccines require adjuvants and/or delivery systems to induce appropriate and protective immune responses against tuberculosis and other diseases. Articles published in this Special Issue have studied the pathogenesis of BCG in children and the use of BCG and recombinant BCG as potential vaccines against asthma. Furthermore, the use of different adjuvants and delivery systems in inducing the protective immune responses after immunization with subunit vaccines has been described.
Collapse
Affiliation(s)
- Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| |
Collapse
|
44
|
Cho T, Khatchadourian C, Nguyen H, Dara Y, Jung S, Venketaraman V. A review of the BCG vaccine and other approaches toward tuberculosis eradication. Hum Vaccin Immunother 2021; 17:2454-2470. [PMID: 33769193 PMCID: PMC8475575 DOI: 10.1080/21645515.2021.1885280] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/29/2021] [Indexed: 02/02/2023] Open
Abstract
Despite aggressive eradication efforts, Tuberculosis (TB) remains a global health burden, one that disproportionally affects poorer, less developed nations. The only vaccine approved for TB, the Bacillus of Calmette and Guérin (BCG) vaccine remains controversial because it's stated efficacy has been cited as anywhere from 0 to 80%. Nevertheless, there have been exciting discoveries about the mechanism of action of the BCG vaccine that suggests it has a role in immunization schedules today. We review recent data suggesting the vaccine imparts protection against both tuberculosis and non-tuberculosis pathogens via a newly discovered immune system called trained immunity. BCG's efficacy also appears to be tied to its affect on granulocytes at the epigenetic and hematopoietic stem cell levels, which we discuss in this article at length. We also write about how the different strains of the BCG vaccine elicit different immune responses, suggesting that certain BCG strains are more immunogenic than others. Finally, our review delves into how the current vaccine is being reformulated to be more efficacious, and track the development of the next generation vaccines against TB.
Collapse
Affiliation(s)
- Thomas Cho
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | | | - Huy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Yash Dara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Shuna Jung
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
45
|
Bibi S, Ullah I, Zhu B, Adnan M, Liaqat R, Kong WB, Niu S. In silico analysis of epitope-based vaccine candidate against tuberculosis using reverse vaccinology. Sci Rep 2021; 11:1249. [PMID: 33441913 PMCID: PMC7807040 DOI: 10.1038/s41598-020-80899-6] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/29/2020] [Indexed: 01/29/2023] Open
Abstract
Tuberculosis (TB) kills more individuals in the world than any other disease, and a threat made direr by the coverage of drug-resistant strains of Mycobacterium tuberculosis (Mtb). Bacillus Calmette-Guérin (BCG) is the single TB vaccine licensed for use in human beings and effectively protects infants and children against severe military and meningeal TB. We applied advanced computational techniques to develop a universal TB vaccine. In the current study, we select the very conserved, experimentally confirmed Mtb antigens, including Rv2608, Rv2684, Rv3804c (Ag85A), and Rv0125 (Mtb32A) to design a novel multi-epitope subunit vaccine. By using the Immune Epitopes Database (IEDB), we predicted different B-cell and T-cell epitopes. An adjuvant (Griselimycin) was also added to vaccine construct to improve its immunogenicity. Bioinformatics tools were used to predict, refined, and validate the 3D structure and then docked with toll-like-receptor (TLR-3) using different servers. The constructed vaccine was used for further processing based on allergenicity, antigenicity, solubility, different physiochemical properties, and molecular docking scores. The in silico immune simulation results showed significant response for immune cells. For successful expression of the vaccine in E. coli, in-silico cloning and codon optimization were performed. This research also sets out a good signal for the design of a peptide-based tuberculosis vaccine. In conclusion, our findings show that the known multi-epitope vaccine may activate humoral and cellular immune responses and maybe a possible tuberculosis vaccine candidate. Therefore, more experimental validations should be exposed to it.
Collapse
Affiliation(s)
- Shaheen Bibi
- College of Life Science, Northwest Normal University, Lanzhou, 730070, Gansu, China
- Lanzhou Center for Tuberculosis Research and Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, 730000, China
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Inayat Ullah
- Lanzhou Center for Tuberculosis Research and Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, 730000, China
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Bingdong Zhu
- Lanzhou Center for Tuberculosis Research and Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, 730000, China
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Muhammad Adnan
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, 99 Lincheng west Road, Guanshan Lake District, Guiyang, 550081, Guizhou, China
| | - Romana Liaqat
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Wei-Bao Kong
- College of Life Science, Northwest Normal University, Lanzhou, 730070, Gansu, China
| | - Shiquan Niu
- College of Life Science, Northwest Normal University, Lanzhou, 730070, Gansu, China.
| |
Collapse
|
46
|
Immunological Characterization of Proteins Expressed by Genes Located in Mycobacterium tuberculosis-Specific Genomic Regions Encoding the ESAT6-like Proteins. Vaccines (Basel) 2021; 9:vaccines9010027. [PMID: 33430286 PMCID: PMC7825740 DOI: 10.3390/vaccines9010027] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
The 6 kDa early secreted antigen target (ESAT6) is a low molecular weight and highly immunogenic protein of Mycobacterium tuberculosis with relevance in the diagnosis of tuberculosis and subunit vaccine development. The gene encoding the ESAT6 protein is located in the M. tuberculosis-specific genomic region known as the region of difference (RD)1. There are 11 M. tuberculosis-specific RDs absent in all of the vaccine strains of BCG, and three of them (RD1, RD7, and RD9) encode immunodominant proteins. Each of these RDs has genes for a pair of ESAT6-like proteins. The immunological characterizations of all the possible proteins encoded by genes in RD1, RD7 and RD9 have shown that, besides ESAT-6 like proteins, several other proteins are major antigens useful for the development of subunit vaccines to substitute or supplement BCG. Furthermore, some of these proteins may replace the purified protein derivative of M. tuberculosis in the specific diagnosis of tuberculosis by using interferon-gamma release assays and/or tuberculin-type skin tests. At least three subunit vaccine candidates containing ESAT6-like proteins as antigen components of multimeric proteins have shown efficacy in phase 1 and phase II clinical trials in humans.
Collapse
|
47
|
Franco AR, Peri F. Developing New Anti-Tuberculosis Vaccines: Focus on Adjuvants. Cells 2021; 10:cells10010078. [PMID: 33466444 PMCID: PMC7824815 DOI: 10.3390/cells10010078] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb) that sits in the top 10 leading causes of death in the world today and is the current leading cause of death among infectious diseases. Although there is a licensed vaccine against TB, the Mycobacterium bovis bacilli Calmette–Guérin (BCG) vaccine, it has several limitations, namely its high variability of efficacy in the population and low protection against pulmonary tuberculosis. New vaccines for TB are needed. The World Health Organization (WHO) considers the development and implementation of new TB vaccines to be a priority. Subunit vaccines are promising candidates since they can overcome safety concerns and optimize antigen targeting. Nevertheless, these vaccines need adjuvants in their formulation in order to increase immunogenicity, decrease the needed antigen dose, ensure a targeted delivery and optimize the antigens delivery and interaction with the immune cells. This review aims to focus on adjuvants being used in new formulations of TB vaccines, namely candidates already in clinical trials and others in preclinical development. Although no correlates of protection are defined, most research lines in the field of TB vaccination focus on T-helper 1 (Th1) type of response, namely polyfunctional CD4+ cells expressing simultaneously IFN-γ, TNF-α, and IL-2 cytokines, and also Th17 responses. Accordingly, most of the adjuvants reviewed here are able to promote such responses. In the future, it might be advantageous to consider a wider array of immune parameters to better understand the role of adjuvants in TB immunity and establish correlates of protection.
Collapse
|
48
|
Bellini C, Horváti K. Recent Advances in the Development of Protein- and Peptide-Based Subunit Vaccines against Tuberculosis. Cells 2020; 9:cells9122673. [PMID: 33333744 PMCID: PMC7765234 DOI: 10.3390/cells9122673] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The World Health Organization (WHO) herald of the “End TB Strategy” has defined goals and targets for tuberculosis prevention, care, and control to end the global tuberculosis endemic. The emergence of drug resistance and the relative dreadful consequences in treatment outcome has led to increased awareness on immunization against Mycobacterium tuberculosis (Mtb). However, the proven limited efficacy of Bacillus Calmette-Guérin (BCG), the only licensed vaccine against Mtb, has highlighted the need for alternative vaccines. In this review, we seek to give an overview of Mtb infection and failure of BCG to control it. Afterward, we focus on the protein- and peptide-based subunit vaccine subtype, examining the advantages and drawbacks of using this design approach. Finally, we explore the features of subunit vaccine candidates currently in pre-clinical and clinical evaluation, including the antigen repertoire, the exploited adjuvanted delivery systems, as well as the spawned immune response.
Collapse
Affiliation(s)
- Chiara Bellini
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary;
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Kata Horváti
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary
- Correspondence:
| |
Collapse
|
49
|
Adeagbo BA, Akinlalu AO, Phan T, Guderian J, Boukes G, Willenburg E, Fenner C, Bolaji OO, Fox CB. Controlled Covalent Conjugation of a Tuberculosis Subunit Antigen (ID93) to Liposome Improved In Vitro Th1-Type Cytokine Recall Responses in Human Whole Blood. ACS OMEGA 2020; 5:31306-31313. [PMID: 33324841 PMCID: PMC7726955 DOI: 10.1021/acsomega.0c04774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/05/2020] [Indexed: 06/12/2023]
Abstract
Tuberculosis (TB) remains a foremost poverty-related disease with a high rate of mortality despite global immunization with Bacille Calmette-Guérin (BCG). Several adjuvanted recombinant proteins are in clinical development for TB to protect against the disease in infants and adults. Nevertheless, simple mixing of adjuvants with antigens may not be optimal for enhancing the immune response due to poor association. Hence, co-delivery of adjuvants with antigens has been advocated for improved immune response. This report, therefore, presents a strategy of using chemical conjugation to co-deliver an adjuvanted recombinant protein TB vaccine (ID93 + GLA-LSQ). Chemical conjugation involving glutaraldehyde (GA) or 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC) was used to associate the antigen (ID93) to the modified liposome (mGLA-LSQ). The physicochemical stability of the formulations was evaluated using high-performance liquid chromatography (HPLC) (adjuvant content), dynamic light scattering (DLS, particle size analysis), and sodium dodecyl sulfate-polyacrylamide gel (SDS) electrophoresis (protein analysis). The bioactivity was assessed by cytokine stimulation using fresh whole blood from 10 healthy donors. The conjugates of ID93 + mGLA_LSQ maintained liposomal and protein integrity with the two protein chemistries. The GLA and QS21 content of the vaccine were also stable for 3 months. However, only the glutaraldehyde conjugates provoked significant secretion of interleukin-2 (210.4 ± 11.45 vs 166.7 ± 9.15; p = 0.0059), interferon-gamma (210.5 ± 14.79 vs 144.1 ± 4.997; p = 0.0011), and tumor necrosis factor alpha (2075 ± 46.8 vs 1456 ± 144.8; p = 0.0082) compared to simple mixing. Conjugation of recombinant protein (ID93) to the liposome (mGLA_LSQ) through chemical conjugation resulted in a stable vaccine formulation, which could facilitate co-delivery of the subunit vaccine to promote a robust immune response.
Collapse
Affiliation(s)
- Babatunde Ayodeji Adeagbo
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, NG 220280, Nigeria
- Infectious
Disease Research Institute, 1616 Eastlake Avenue East Suite 400, Seattle, Washington 98102, United States
| | - Akintunde Oluseto Akinlalu
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, NG 220280, Nigeria
| | - Tony Phan
- Infectious
Disease Research Institute, 1616 Eastlake Avenue East Suite 400, Seattle, Washington 98102, United States
| | - Jeff Guderian
- Infectious
Disease Research Institute, 1616 Eastlake Avenue East Suite 400, Seattle, Washington 98102, United States
| | - Gerhardt Boukes
- Afrigen
Biologics (Pty) Limited, South Africa Medical
Research Council Medicina Campus Francie van Zijl Drive, Cape Town, ZA 7500, South Africa
| | - Elize Willenburg
- Afrigen
Biologics (Pty) Limited, South Africa Medical
Research Council Medicina Campus Francie van Zijl Drive, Cape Town, ZA 7500, South Africa
| | - Caryn Fenner
- Afrigen
Biologics (Pty) Limited, South Africa Medical
Research Council Medicina Campus Francie van Zijl Drive, Cape Town, ZA 7500, South Africa
| | - Oluseye Oladotun Bolaji
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, NG 220280, Nigeria
| | - Christopher B. Fox
- Infectious
Disease Research Institute, 1616 Eastlake Avenue East Suite 400, Seattle, Washington 98102, United States
- Department
of Global Health, University of Washington, 3980 15th Ave NE, Seattle, Washington 98195, United States
| |
Collapse
|
50
|
Safar HA, Mustafa AS, McHugh TD. COVID-19 vaccine development: What lessons can we learn from TB? Ann Clin Microbiol Antimicrob 2020; 19:56. [PMID: 33256750 PMCID: PMC7702199 DOI: 10.1186/s12941-020-00402-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
At the time of writing, the SARS-CoV-2 virus has infected more than 49 million people causing more than 1.2 million deaths worldwide since its emergence from Wuhan, China in December 2019. Vaccine development against SARS-CoV-2 has drawn the global attention in order to stop the spread of the virus, with more than 10 vaccines being tested in phase III clinical trials, as of November 2020. However, critical to vaccine development is consideration of the immunological response elicited as well as biological features of the vaccine and both need to be evaluated thoroughly. Tuberculosis is also a major infectious respiratory disease of worldwide prevalence and the vaccine development for tuberculosis has been ongoing for decades. In this review, we highlight some of the common features, challenges and complications in tuberculosis vaccine development, which may also be relevant for, and inform, COVID-19 vaccine development.
Collapse
Affiliation(s)
- Hussain A Safar
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK.
| | - Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Timothy D McHugh
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|