1
|
Konjengbam BD, Meitei HN, Pandey A, Haobam R. Goals and strategies in vaccine development against tuberculosis. Mol Immunol 2025; 183:56-71. [PMID: 40327952 DOI: 10.1016/j.molimm.2025.04.016] [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: 10/25/2023] [Revised: 04/16/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major health problem globally. The emergence of multi-drug-resistant TB and extensively drug-resistant TB has become a severe threat to TB control programs. Currently, the Bacille Calmette-Guerin (BCG) vaccine protects a child from disease dissemination efficiently, but its efficiency wanes in adults. Despite all the limitations of BCG and accelerated TB vaccine research, BCG remains the only approved vaccine available for TB. Anti-TB drug treatment has been successful in combating the disease, but it has various side effects and requires an extended drug treatment period. So, vaccination is the finest outlook that can surpass the above-mentioned limitations. Several vaccine candidates are in the pipeline, and the hope for a potential candidate to either boost the BCG vaccine or replace BCG is underway. This review discusses different approaches to TB vaccine development. It summarizes all the challenges and limitations in vaccine development, and its preclinical and clinical trials. Additionally, DNA vaccines and their vaccination techniques are also discussed. Furthermore, the immunoinformatics approach and nanomaterial-based vaccine delivery with practical and productive endpoints are also discussed. Lastly, the potential prospects are also suggested for further studies, which would help bring positive outcomes.
Collapse
Affiliation(s)
| | | | - Anupama Pandey
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India.
| |
Collapse
|
2
|
Wang J, Fan XY, Hu Z. Immune correlates of protection as a game changer in tuberculosis vaccine development. NPJ Vaccines 2024; 9:208. [PMID: 39478007 PMCID: PMC11526030 DOI: 10.1038/s41541-024-01004-w] [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: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
The absence of validated correlates of protection (CoPs) hampers the rational design and clinical development of new tuberculosis vaccines. In this review, we provide an overview of the potential CoPs in tuberculosis vaccine research. Major hindrances and potential opportunities are then discussed. Based on recent progress, it is reasonable to anticipate that success in the ongoing efforts to identify CoPs would be a game-changer in tuberculosis vaccine development.
Collapse
Affiliation(s)
- Jing Wang
- 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.
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
3
|
Colombatti Olivieri MA, Cuerda MX, Moyano RD, Gravisaco MJ, Pinedo MFA, Delgado FO, Calamante G, Mundo S, de la Paz Santangelo M, Romano MI, Alonso MN, Del Medico Zajac MP. Superior protection against paratuberculosis by a heterologous prime-boost immunization in a murine model. Vaccine 2024; 42:126055. [PMID: 38880691 DOI: 10.1016/j.vaccine.2024.06.022] [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: 07/14/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Vaccination is the best strategy to control Paratuberculosis (PTB), which is a significant disease in cattle and sheep. Previously we showed the humoral and cellular immune response induced by a novel vaccine candidate against PTB based on the Argentinian Mycobacterium avium subspecies paratuberculosis (Map) 6611 strain. To improve 6611 immunogenicity and efficacy, we evaluated this vaccine candidate in mice with two different adjuvants and a heterologous boost with a recombinant modified vaccinia Ankara virus (MVA) expressing the antigen 85A (MVA85A). We observed that boosting with MVA85A did not improve total IgG or specific isotypes in serum induced by one or two doses of 6611 formulated with incomplete Freund's adjuvant (IFA). However, when 6611 was formulated with ISA201 adjuvant, MVA85A boost enhanced the production of IFNγ, Th1/Th17 cytokines (IL-2, TNF, IL-17A) and IL-6, IL-4 and IL-10. Also, this group showed the highest levels of IgG2b and IgG3 isotypes, both important for better protection against Map infection in the murine model. Finally, the heterologous scheme elicited the highest levels of protection after Map challenge (lowest CFU count and liver lesion score). In conclusion, our results encourage further evaluation of 6611 strain + ISA201 prime and MVA85A boost in bovines.
Collapse
MESH Headings
- Animals
- Mycobacterium avium subsp. paratuberculosis/immunology
- Immunization, Secondary/methods
- Mice
- Paratuberculosis/prevention & control
- Paratuberculosis/immunology
- Immunoglobulin G/blood
- Cytokines/metabolism
- Female
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Adjuvants, Immunologic/administration & dosage
- Disease Models, Animal
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Mice, Inbred BALB C
- Vaccinia virus/immunology
- Vaccinia virus/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Immunity, Cellular/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Freund's Adjuvant/administration & dosage
- Freund's Adjuvant/immunology
Collapse
Affiliation(s)
| | - María Ximena Cuerda
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Roberto Damián Moyano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María José Gravisaco
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Fiorella Alvarado Pinedo
- Centro de Diagnóstico e Investigaciones Veterinarias (CEDIVE) de la Facultad de Ciencias Veterinarias - Universidad de La Plata, Chascomús, Buenos Aires 7130, Argentina
| | - Fernando Oscar Delgado
- Instituto de Patobiologia Veterinaria (IPV), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Gabriela Calamante
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Silvia Mundo
- Cátedra de Inmunología de la Facultad de Ciencias Veterinarias - Universidad de Buenos Aires, Ciudad de Buenos Aires 1427, Argentina
| | - María de la Paz Santangelo
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Isabel Romano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Natalia Alonso
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina.
| | - María Paula Del Medico Zajac
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| |
Collapse
|
4
|
Li F, Dang W, Du Y, Xu X, He P, Zhou Y, Zhu B. Tuberculosis Vaccines and T Cell Immune Memory. Vaccines (Basel) 2024; 12:483. [PMID: 38793734 PMCID: PMC11125691 DOI: 10.3390/vaccines12050483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis (TB) remains a major infectious disease partly due to the lack of an effective vaccine. Therefore, developing new and more effective TB vaccines is crucial for controlling TB. Mycobacterium tuberculosis (M. tuberculosis) usually parasitizes in macrophages; therefore, cell-mediated immunity plays an important role. The maintenance of memory T cells following M. tuberculosis infection or vaccination is a hallmark of immune protection. This review analyzes the development of memory T cells during M. tuberculosis infection and vaccine immunization, especially on immune memory induced by BCG and subunit vaccines. Furthermore, the factors affecting the development of memory T cells are discussed in detail. The understanding of the development of memory T cells should contribute to designing more effective TB vaccines and optimizing vaccination strategies.
Collapse
Affiliation(s)
- Fei Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Wenrui Dang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Yunjie Du
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Xiaonan Xu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Pu He
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Yuhe Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Bingdong Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
- College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
5
|
A prime-boost combination of a three-protein cocktail and multiepitopic MVA as a vaccine against Babesia bigemina elicits neutralizing antibodies and a Th1 cellular immune response in mice. Ticks Tick Borne Dis 2022; 13:101991. [DOI: 10.1016/j.ttbdis.2022.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022]
|
6
|
Hu Z, Lu SH, Lowrie DB, Fan XY. Research Advances for Virus-vectored Tuberculosis Vaccines and Latest Findings on Tuberculosis Vaccine Development. Front Immunol 2022; 13:895020. [PMID: 35812383 PMCID: PMC9259874 DOI: 10.3389/fimmu.2022.895020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by respiratory infection with Mycobacterium tuberculosis, remains a major global health threat. The only licensed TB vaccine, the one-hundred-year-old Bacille Calmette-Guérin has variable efficacy and often provides poor protection against adult pulmonary TB, the transmissible form of the disease. Thus, the lack of an optimal TB vaccine is one of the key barriers to TB control. Recently, the development of highly efficacious COVID-19 vaccines within one year accelerated the vaccine development process in human use, with the notable example of mRNA vaccines and adenovirus-vectored vaccines, and increased the public acceptance of the concept of the controlled human challenge model. In the TB vaccine field, recent progress also facilitated the deployment of an effective TB vaccine. In this review, we provide an update on the current virus-vectored TB vaccine pipeline and summarize the latest findings that might facilitate TB vaccine development. In detail, on the one hand, we provide a systematic literature review of the virus-vectored TB vaccines are in clinical trials, and other promising candidate vaccines at an earlier stage of development are being evaluated in preclinical animal models. These research sharply increase the likelihood of finding a more effective TB vaccine in the near future. On the other hand, we provide an update on the latest tools and concept that facilitating TB vaccine research development. We propose that a pre-requisite for successful development may be a better understanding of both the lung-resident memory T cell-mediated mucosal immunity and the trained immunity of phagocytic cells. Such knowledge could reveal novel targets and result in the innovative vaccine designs that may be needed for a quantum leap forward in vaccine efficacy. We also summarized the research on controlled human infection and ultra-low-dose aerosol infection murine models, which may provide more realistic assessments of vaccine utility at earlier stages. In addition, we believe that the success in the ongoing efforts to identify correlates of protection would be a game-changer for streamlining the triage of multiple next-generation TB vaccine candidates. Thus, with more advanced knowledge of TB vaccine research, we remain hopeful that a more effective TB vaccine will eventually be developed in the near future.
Collapse
Affiliation(s)
- Zhidong Hu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- *Correspondence: Zhidong Hu, ; Xiao-Yong Fan,
| | - Shui-Hua Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- National Medical Center for Infectious Diseases of China, Shenzhen Third People Hospital, South Science & Technology University, Shenzhen, China
| | - Douglas B. Lowrie
- National Medical Center for Infectious Diseases of China, Shenzhen Third People Hospital, South Science & Technology University, Shenzhen, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- *Correspondence: Zhidong Hu, ; Xiao-Yong Fan,
| |
Collapse
|
7
|
Ylösmäki E, Fusciello M, Martins B, Feola S, Hamdan F, Chiaro J, Ylösmäki L, Vaughan MJ, Viitala T, Kulkarni PS, Cerullo V. Novel personalized cancer vaccine platform based on Bacillus Calmette-Guèrin. J Immunother Cancer 2021; 9:jitc-2021-002707. [PMID: 34266884 PMCID: PMC8286790 DOI: 10.1136/jitc-2021-002707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 12/03/2022] Open
Abstract
Background Intratumoral BCG therapy, one of the earliest immunotherapies, can lead to infiltration of immune cells into a treated tumor. However, an increase in the number of BCG-induced tumor-specific T cells in the tumor microenvironment could lead to enhanced therapeutic effects. Methods Here, we have developed a novel cancer vaccine platform based on BCG that can broaden BCG-induced immune responses to include tumor antigens. By physically attaching tumor-specific peptides onto the mycobacterial outer membrane, we were able to induce strong systemic and intratumoral T cell-specific immune responses toward the attached tumor antigens. These therapeutic peptides can be efficiently attached to the mycobacterial outer membrane using a poly-lysine sequence N-terminally fused to the tumor-specific peptides. Results Using two mouse models of melanoma and a mouse model of colorectal cancer, we observed that the antitumor immune responses of BCG could be improved by coating the BCG with tumor-specific peptides. In addition, by combining this novel cancer vaccine platform with anti-programmed death 1 (anti-PD-1) immune checkpoint inhibitor (ICI) therapy, the number of responders to anti-PD-1 immunotherapy was markedly increased. Conclusions This study shows that intratumoral BCG immunotherapy can be improved by coating the bacteria with modified tumor-specific peptides. In addition, this improved BCG immunotherapy can be combined with ICI therapy to obtain enhanced tumor growth control. These results warrant clinical testing of this novel cancer vaccine platform.
Collapse
Affiliation(s)
- Erkko Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Manlio Fusciello
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Beatriz Martins
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Sara Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Firas Hamdan
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Jacopo Chiaro
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Leena Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Valo Therapeutics Oy, Helsinki, Finland
| | | | - Tapani Viitala
- Pharmaceutical Biophysics Research Group, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland .,TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,Department of Molecular Medicine and Medical Biotechnology and CEINGE, Naples University 24 Federico II, Naples, Italy
| |
Collapse
|
8
|
Prime Vaccination with Chitosan-Coated Phipps BCG and Boosting with CFP-PLGA against Tuberculosis in a Goat Model. Animals (Basel) 2021; 11:ani11041046. [PMID: 33917739 PMCID: PMC8068168 DOI: 10.3390/ani11041046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Bovine tuberculosis is a disease that affects cattle and other animal species worldwide and represents a risk to public health. Even though there is a vaccine that has been used to control tuberculosis in humans for almost 100 years, up to now, it has not been used in animals. The reason is that vaccination interferes with the tuberculin test, the current test to diagnose tuberculosis in the field, and shows an inconsistent efficacy in animals. Recent studies report that prime vaccinating with BCG and boosting with proteins vaccinations perform better. In addition, there are reports that some polymers increase the immune response against various infectious diseases; therefore, testing a vaccine formula with polymers sounds like a wise thing to do. In this study, we showed that priming with BCG and boosting with a culture filtrate protein, alone or in combination with a polymer, the number of animals with lesions, the number of lesions per animal, and the size of the lesions in vaccinated animals, compared with those not vaccinated or those vaccinated with BCG alone, are significantly reduced. Our results mean that a vaccination used as a complement of actual tuberculosis control programs in animal populations can be useful to reduce tuberculosis dissemination. Abstract Attempts to improve the immune response and efficacy of vaccines against tuberculosis in cattle, goats, and other animal species have been the focus of research in this field during the last two decades. Improving the vaccine efficacy is essential prior to running long-lasting and expensive field trials. Studies have shown that vaccine protocols utilizing boosting with proteins improve the vaccine efficacy. The use of polymers such as chitosan and PolyLactic-co-Glycolic Acid (PLGA) improves the immune response against different diseases by improving the interaction of antigens with the cellular immune system and modulating the host immune response. This study shows that the prime BCG vaccination, boosted with a culture filtrate protein (CFP), alone or in combination with chitosan and PolyLactic-co-Glycolic Acid (PLGA), have the potential to reduce tuberculosis (TB) dissemination by reducing the number of animals with lesions, the number of lesions per animal, and the size of the lesions in vaccinated animals, compared with those not vaccinated or those vaccinated with BCG alone. The vaccinated groups showed significantly higher Interferon-γ levels in the blood compared to the control, nonvaccinated group after vaccination, after boosting, and after the challenge with the wild-type Mycobacterium bovis strain.
Collapse
|
9
|
Srinivasan S, Conlan AJK, Easterling LA, Herrera C, Dandapat P, Veerasami M, Ameni G, Jindal N, Raj GD, Wood J, Juleff N, Bakker D, Vordermeier M, Kapur V. A Meta-Analysis of the Effect of Bacillus Calmette-Guérin Vaccination Against Bovine Tuberculosis: Is Perfect the Enemy of Good? Front Vet Sci 2021; 8:637580. [PMID: 33681334 PMCID: PMC7930010 DOI: 10.3389/fvets.2021.637580] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/27/2021] [Indexed: 01/10/2023] Open
Abstract
More than 50 million cattle are likely exposed to bovine tuberculosis (bTB) worldwide, highlighting an urgent need for bTB control strategies in low- and middle-income countries (LMICs) and other regions where the disease remains endemic and test-and-slaughter approaches are unfeasible. While Bacillus Calmette-Guérin (BCG) was first developed as a vaccine for use in cattle even before its widespread use in humans, its efficacy against bTB remains poorly understood. To address this important knowledge gap, we conducted a systematic review and meta-analysis to determine the direct efficacy of BCG against bTB challenge in cattle, and performed scenario analyses with transmission dynamic models incorporating direct and indirect vaccinal effects ("herd-immunity") to assess potential impact on herd level disease control. The analysis shows a relative risk of infection of 0.75 (95% CI: 0.68, 0.82) in 1,902 vaccinates as compared with 1,667 controls, corresponding to a direct vaccine efficacy of 25% (95% CI: 18, 32). Importantly, scenario analyses considering both direct and indirect effects suggest that disease prevalence could be driven down close to Officially TB-Free (OTF) status (<0.1%), if BCG were introduced in the next 10-year time period in low to moderate (<15%) prevalence settings, and that 50-95% of cumulative cases may be averted over the next 50 years even in high (20-40%) disease burden settings with immediate implementation of BCG vaccination. Taken together, the analyses suggest that BCG vaccination may help accelerate control of bTB in endemic settings, particularly with early implementation in the face of dairy intensification in regions that currently lack effective bTB control programs.
Collapse
Affiliation(s)
- Sreenidhi Srinivasan
- Department of Animal Science, The Pennsylvania State University, University Park, PA, United States
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Andrew J. K. Conlan
- Disease Dynamics Unit, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Laurel A. Easterling
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Christian Herrera
- Department of Animal Science, The Pennsylvania State University, University Park, PA, United States
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Premanshu Dandapat
- Indian Veterinary Research Institute, Eastern Regional Station, Kolkata, India
| | | | - Gobena Ameni
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Naresh Jindal
- Department of Veterinary Public Health and Epidemiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Gopal Dhinakar Raj
- Translational Research Platform for Veterinary Biological, Tamil Nadu University of Veterinary and Animal Sciences, Chennai, India
| | - James Wood
- Disease Dynamics Unit, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nick Juleff
- The Bill & Melinda Gates Foundation, Seattle, WA, United States
| | - Douwe Bakker
- Technical Consultant and Independent Researcher, Lelystad, Netherlands
| | - Martin Vordermeier
- Animal and Plant Health Agency, Addlestone, United Kingdom
- Centre for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, University of Aberystwyth, Aberystwyth, United Kingdom
| | - Vivek Kapur
- Department of Animal Science, The Pennsylvania State University, University Park, PA, United States
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
10
|
Wilkie M, Satti I, Minhinnick A, Harris S, Riste M, Ramon RL, Sheehan S, Thomas ZRM, Wright D, Stockdale L, Hamidi A, O'Shea MK, Dwivedi K, Behrens HM, Davenne T, Morton J, Vermaak S, Lawrie A, Moss P, McShane H. A phase I trial evaluating the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime - MVA85A boost in healthy UK adults. Vaccine 2019; 38:779-789. [PMID: 31735500 PMCID: PMC6985898 DOI: 10.1016/j.vaccine.2019.10.102] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 11/18/2022]
Abstract
Background This phase I trial evaluated the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime-MVA85A boost, previously demonstrated to be protective in animal studies, in healthy UK adults. Methods We enrolled 42 healthy, BCG-vaccinated adults into 4 groups: low dose Starter Group (n = 6; ChAdOx1 85A alone), high dose groups; Group A (n = 12; ChAdOx1 85A), Group B (n = 12; ChAdOx1 85A prime – MVA85A boost) or Group C (n = 12; ChAdOx1 85A – ChAdOx1 85A prime – MVA85A boost). Safety was determined by collection of solicited and unsolicited vaccine-related adverse events (AEs). Immunogenicity was measured by antigen-specific ex-vivo IFN-γ ELISpot, IgG serum ELISA, and antigen-specific intracellular IFN-γ, TNF-α, IL-2 and IL-17. Results AEs were mostly mild/moderate, with no Serious Adverse Events. ChAdOx1 85A induced Ag85A-specific ELISpot and intracellular cytokine CD4+ and CD8+ T cell responses, which were not boosted by a second dose, but were boosted with MVA85A. Polyfunctional CD4+ T cells (IFN-γ, TNF-α and IL-2) and IFN-γ+, TNF-α+ CD8+ T cells were induced by ChAdOx1 85A and boosted by MVA85A. ChAdOx1 85A induced serum Ag85A IgG responses which were boosted by MVA85A. Conclusion A ChAdOx1 85A prime – MVA85A boost is well tolerated and immunogenic in healthy UK adults.
Collapse
Affiliation(s)
- Morven Wilkie
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Michael Riste
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Sharon Sheehan
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Daniel Wright
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Lisa Stockdale
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ali Hamidi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Kritica Dwivedi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Tamara Davenne
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Joshua Morton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Alison Lawrie
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
11
|
Díez-Delgado I, Sevilla IA, Garrido JM, Romero B, Geijo MV, Dominguez L, Juste RA, Aranaz A, de la Fuente J, Gortazar C. Tuberculosis vaccination sequence effect on protection in wild boar. Comp Immunol Microbiol Infect Dis 2019; 66:101329. [PMID: 31437687 DOI: 10.1016/j.cimid.2019.101329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 10/26/2022]
Abstract
The Eurasian wild boar (Sus scrofa) is a reservoir for tuberculosis (TB) in which vaccination is a valuable tool for control. We evaluated the protection and immune response achieved by homologous and heterologous regimes administering BCG and heat-inactivated Mycobacterium bovis (IV). Twenty-one wild boar piglets were randomly allocated in five groups: Control, homologous BCG, homologous IV, heterologous IV-BCG, heterologous BCG-IV. Significant 67% and 66% total lesion score reductions were detected in homologous IV (IVx2) and heterologous IV-BCG groups when compared with Control group (F4,16 = 6.393, p = 0.003; Bonferroni Control vs IVx2 p = 0.026, Tukey Control vs IV-BCG p = 0.021). No significant differences were found for homologous BCG (although a 48% reduction in total lesion score was recorded) and BCG-IV (3% reduction). Heterologous regimes did not improve protection over homologous regimes in the wild boar model and showed variable results from no protection to similar protection as homologous regimes. Therefore, homologous regimes remain the best option to vaccinate wild boar against TB. Moreover, vaccine sequence dramatically influenced the outcome underlining the relevance of studying the effects of prior sensitization in the outcome of vaccination.
Collapse
Affiliation(s)
- Iratxe Díez-Delgado
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Avenida Puerta de Hierro s/n, 28040, Madrid, Spain; SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM), Ronda de Toledo 12, 13071, Ciudad Real, Spain.
| | - Iker A Sevilla
- NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Animal Health Department. Bizkaia Science and Technology Park 812L, 48160, Derio (Bizkaia), Spain
| | - Joseba M Garrido
- NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Animal Health Department. Bizkaia Science and Technology Park 812L, 48160, Derio (Bizkaia), Spain
| | - Beatriz Romero
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense de Madrid, Avenida Puerta de Hierro s/n, 28040, Madrid, Spain
| | - María V Geijo
- NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Animal Health Department. Bizkaia Science and Technology Park 812L, 48160, Derio (Bizkaia), Spain
| | - Lucas Dominguez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Avenida Puerta de Hierro s/n, 28040, Madrid, Spain; Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense de Madrid, Avenida Puerta de Hierro s/n, 28040, Madrid, Spain
| | - Ramón A Juste
- NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Animal Health Department. Bizkaia Science and Technology Park 812L, 48160, Derio (Bizkaia), Spain; Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Carretera de Oviedo s/n 13 P.O. Box, 33300, Villaviciosa, Asturias, Spain
| | - Alicia Aranaz
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Avenida Puerta de Hierro s/n, 28040, Madrid, Spain
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM), Ronda de Toledo 12, 13071, Ciudad Real, Spain; Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Christian Gortazar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM), Ronda de Toledo 12, 13071, Ciudad Real, Spain
| |
Collapse
|
12
|
Sun W, Singh AK. Plague vaccine: recent progress and prospects. NPJ Vaccines 2019; 4:11. [PMID: 30792905 PMCID: PMC6379378 DOI: 10.1038/s41541-019-0105-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/19/2018] [Indexed: 01/14/2023] Open
Abstract
Three great plague pandemics, resulting in nearly 200 million deaths in human history and usage as a biowarfare agent, have made Yersinia pestis as one of the most virulent human pathogens. In late 2017, a large plague outbreak raged in Madagascar attracted extensive attention and caused regional panics. The evolution of local outbreaks into a pandemic is a concern of the Centers for Disease Control and Prevention (CDC) in plague endemic regions. Until now, no licensed plague vaccine is available. Prophylactic vaccination counteracting this disease is certainly a primary choice for its long-term prevention. In this review, we summarize the latest advances in research and development of plague vaccines.
Collapse
Affiliation(s)
- Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208 USA
| | - Amit K. Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208 USA
| |
Collapse
|
13
|
Lesellier S. Immunological responses of European badgers (Meles Meles) to infection with Mycobacterium bovis. Comp Immunol Microbiol Infect Dis 2018; 61:9-15. [PMID: 30502833 DOI: 10.1016/j.cimid.2018.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 12/17/2022]
Abstract
Mycobacterium bovis is the main cause of bovine tuberculosis and its eradication is proving difficult in many countries because of wildlife reservoirs, including European badgers (Meles meles) in the UK Ireland. Following the development of badger specific immunological reagents, many studies have shown that some aspects of the cellular and serological immune responses of badgers to virulent M. bovis and the attenuated M. bovis BCG (Bacille of Calmette and Guérin) strain are similar to those seen in other animal hosts infected with M. bovis. However, badgers also appear to have developed specific immunological responses to M. bovis infection which may be associated with mild inflammatory responses. Badgers may therefore represent an interesting natural host for M. bovis that can provide a more thorough understanding of efficient immunological responses to tuberculosis.
Collapse
Affiliation(s)
- Sandrine Lesellier
- Animal and Plant Health Agency, Woodham Lane, New Haw, KT15 3NB, United Kingdom.
| |
Collapse
|
14
|
Abstract
In this article we present experimental Mycobacterium bovis infection models in domestic livestock species and how these models were applied to vaccine development, biomarker discovery, and the definition of specific antigens for the differential diagnosis of infected and vaccinated animals. In particular, we highlight synergies between human and bovine tuberculosis (TB) research approaches and data and propose that the application of bovine TB models could make a valuable contribution to human TB vaccine research and that close alignment of both research programs in a one health philosophy will lead to mutual and substantial benefits.
Collapse
|
15
|
Sendai Virus Mucosal Vaccination Establishes Lung-Resident Memory CD8 T Cell Immunity and Boosts BCG-Primed Protection against TB in Mice. Mol Ther 2017; 25:1222-1233. [PMID: 28342639 PMCID: PMC5417795 DOI: 10.1016/j.ymthe.2017.02.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/16/2017] [Accepted: 02/28/2017] [Indexed: 11/23/2022] Open
Abstract
Accumulating evidence has shown the protective role of CD8+ T cells in vaccine-induced immunity against Mycobacterium tuberculosis (Mtb) despite controversy over their role in natural immunity. However, the current vaccine BCG is unable to induce sufficient CD8+ T cell responses, especially in the lung. Sendai virus, a respiratory RNA virus, is here engineered firstly as a novel recombinant anti-TB vaccine (SeV85AB) that encodes Mtb immuno-dominant antigens, Ag85A and Ag85B. A single mucosal vaccination elicited potent antigen-specific T cell responses and a degree of protection against Mtb challenge similar to the effect of BCG in mice. Depletion of CD8+ T cells abrogated the protective immunity afforded by SeV85AB vaccination. Interestingly, only SeV85AB vaccination induced high levels of lung-resident memory CD8+ T (TRM) cells, and this led to a rapid and strong recall of antigen-specific CD8+ T cell responses against Mtb challenge infection. Furthermore, when used in a BCG prime-SeV85AB boost strategy, SeV85AB vaccine significantly enhanced protection above that seen after BCG vaccination alone. Our findings suggest that CD8+ TRM cells that arise in lungs responding to this mucosal vaccination might help to protect against TB, and SeV85AB holds notable promise to improve BCG’s protective efficacy in a prime-boost immunization regimen.
Collapse
|
16
|
Vordermeier HM, Jones GJ, Buddle BM, Hewinson RG, Villarreal-Ramos B. Bovine Tuberculosis in Cattle: Vaccines, DIVA Tests, and Host Biomarker Discovery. Annu Rev Anim Biosci 2016; 4:87-109. [PMID: 26884103 DOI: 10.1146/annurev-animal-021815-111311] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bovine tuberculosis remains a major economic and animal welfare concern worldwide. Cattle vaccination is being considered as part of control strategies. This approach, used alongside conventional control policies, also requires the development of vaccine-compatible diagnostic assays to distinguish vaccinated from infected animals (DIVA). We discuss progress made on optimizing the only potentially available vaccine, bacille Calmette Guérin (BCG), and on strategies to improve BCG efficacy. We also describe recent advances in DIVA development based on the detection of host cellular immune responses by blood-testing or skin-testing approaches. Finally, to accelerate vaccine development, definition of host biomarkers that provide meaningful stage-gating criteria to select vaccine candidates for further testing is highly desirable. Some progress has also been made in this area of research, and we summarize studies that defined either markers predicting vaccine success or markers that correlate with disease stage or severity.
Collapse
Affiliation(s)
- H Martin Vordermeier
- Animal and Plant Health Agency, Weybridge, Addlestone, Surrey KT15 3NB, United Kingdom;
| | - Gareth J Jones
- Animal and Plant Health Agency, Weybridge, Addlestone, Surrey KT15 3NB, United Kingdom;
| | - Bryce M Buddle
- AgResearch, Hopkirk Research Institute, Palmerston North 4442, New Zealand
| | - R Glyn Hewinson
- Animal and Plant Health Agency, Weybridge, Addlestone, Surrey KT15 3NB, United Kingdom;
| | | |
Collapse
|
17
|
Vordermeier HM, Jones GJ, Buddle BM, Hewinson RG. Development of immune-diagnostic reagents to diagnose bovine tuberculosis in cattle. Vet Immunol Immunopathol 2016; 181:10-14. [DOI: 10.1016/j.vetimm.2016.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/08/2016] [Indexed: 10/22/2022]
|
18
|
Kaveh DA, Garcia-Pelayo MC, Webb PR, Wooff EE, Bachy VS, Hogarth PJ. Parenteral adenoviral boost enhances BCG induced protection, but not long term survival in a murine model of bovine TB. Vaccine 2016; 34:4003-11. [PMID: 27317453 DOI: 10.1016/j.vaccine.2016.06.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/25/2016] [Accepted: 06/09/2016] [Indexed: 10/21/2022]
Abstract
Boosting BCG using heterologous prime-boost represents a promising strategy for improved tuberculosis (TB) vaccines, and adenovirus (Ad) delivery is established as an efficacious boosting vehicle. Although studies demonstrate that intranasal administration of Ad boost to BCG offers optimal protection, this is not currently possible in cattle. Using Ad vaccine expressing the mycobacterial antigen TB10.4 (BCG/Ad-TB10.4), we demonstrate, parenteral boost of BCG immunised mice to induce specific CD8(+) IFN-γ producing T cells via synergistic priming of new epitopes. This induces significant improvement in pulmonary protection against Mycobacterium bovis over that provided by BCG when assessed in a standard 4week challenge model. However, in a stringent, year-long survival study, BCG/Ad-TB10.4 did not improve outcome over BCG, which we suggest may be due to the lack of additional memory cells (IL-2(+)) induced by boosting. These data indicate BCG-prime/parenteral-Ad-TB10.4-boost to be a promising candidate, but also highlight the need for further understanding of the mechanisms of T cell priming and associated memory using Ad delivery systems. That we were able to generate significant improvement in pulmonary protection above BCG with parenteral, rather than mucosal administration of boost vaccine is critical; suggesting that the generation of effective mucosal immunity is possible, without the risks and challenges of mucosal administration, but that further work to specifically enhance sustained protective immunity is required.
Collapse
Affiliation(s)
- Daryan A Kaveh
- Department of Bacteriology, Animal & Plant Health Agency (APHA), Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom(1)
| | - M Carmen Garcia-Pelayo
- Department of Bacteriology, Animal & Plant Health Agency (APHA), Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom(1)
| | - Paul R Webb
- Department of Bacteriology, Animal & Plant Health Agency (APHA), Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom(1)
| | - Esen E Wooff
- Department of Bacteriology, Animal & Plant Health Agency (APHA), Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom(1)
| | - Véronique S Bachy
- Department of Bacteriology, Animal & Plant Health Agency (APHA), Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom(1)
| | - Philip J Hogarth
- Department of Bacteriology, Animal & Plant Health Agency (APHA), Woodham Lane, Addlestone, Surrey KT15 3NB, United Kingdom(1).
| |
Collapse
|
19
|
Graves AJ, Hokey DA. Tuberculosis vaccine development: Shifting focus amid increasing development challenges. Hum Vaccin Immunother 2016; 11:1910-6. [PMID: 26125249 PMCID: PMC4635864 DOI: 10.1080/21645515.2015.1040955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A new tuberculosis vaccine is needed to replace or enhance BCG, which induces variable protection against Mycobacterium tuberculosis pulmonary infections in adults. Development of new TB vaccine candidates is severely hampered by the lack of a correlate of immunity, unproven animal models, and limited funding opportunities. One candidate, MVA85A, recently failed to meet its efficacy endpoint goals despite promising early-phase trial data. As a result, some in the field believe we should now shift our focus away from product development and toward a research-oriented approach. Here, we outline our suggestions for this research-oriented strategy including diversification of the candidate pipeline, expanding measurements of immunity, improving pre-clinical animal models, and investing in combination pre-clinical/experimental medicine studies. As with any evolution, this change in strategy comes at a cost but may also represent an opportunity for advancing the field.
Collapse
|
20
|
Dean GS, Clifford D, Whelan AO, Tchilian EZ, Beverley PCL, Salguero FJ, Xing Z, Vordermeier HM, Villarreal-Ramos B. Protection Induced by Simultaneous Subcutaneous and Endobronchial Vaccination with BCG/BCG and BCG/Adenovirus Expressing Antigen 85A against Mycobacterium bovis in Cattle. PLoS One 2015; 10:e0142270. [PMID: 26544594 PMCID: PMC4636221 DOI: 10.1371/journal.pone.0142270] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/20/2015] [Indexed: 11/19/2022] Open
Abstract
The incidence of bovine tuberculosis (bTB) in the GB has been increasing since the 1980s. Immunisation, alongside current control measures, has been proposed as a sustainable measure to control bTB. Immunisation with Mycobacterium bovis bacillus Calmette-Guerin (BCG) has been shown to protect against bTB. Furthermore, much experimental data indicates that pulmonary local immunity is important for protection against respiratory infections including Mycobacterium tuberculosis and that pulmonary immunisation is highly effective. Here, we evaluated protection against M. bovis, the main causative agent of bTB, conferred by BCG delivered subcutaneously, endobronchially or by the new strategy of simultaneous immunisation by both routes. We also tested simultaneous subcutaneous immunisation with BCG and endobronchial delivery of a recombinant type 5 adenovirus expressing mycobacterial antigen 85A. There was significantly reduced visible pathology in animals receiving the simultaneous BCG/BCG or BCG/Ad85 treatment compared to naïve controls. Furthermore, there were significantly fewer advanced microscopic granulomata in animals receiving BCG/Ad85A compared to naive controls. Thus, combining local and systemic immunisation limits the development of pathology, which in turn could decrease bTB transmission.
Collapse
Affiliation(s)
- Gillian S. Dean
- TB Research Group, APHA Weybridge, Woodham Lane, New Haw, KT15 3NB, Surrey, United Kingdom
| | - Derek Clifford
- TB Research Group, APHA Weybridge, Woodham Lane, New Haw, KT15 3NB, Surrey, United Kingdom
| | - Adam O. Whelan
- TB Research Group, APHA Weybridge, Woodham Lane, New Haw, KT15 3NB, Surrey, United Kingdom
| | - Elma Z. Tchilian
- The Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, United Kingdom
| | - Peter C. L. Beverley
- The Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, United Kingdom
| | - Francisco J. Salguero
- TB Research Group, APHA Weybridge, Woodham Lane, New Haw, KT15 3NB, Surrey, United Kingdom
| | - Zhou Xing
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hans M. Vordermeier
- TB Research Group, APHA Weybridge, Woodham Lane, New Haw, KT15 3NB, Surrey, United Kingdom
| | | |
Collapse
|
21
|
Characterization of effector and memory T cell subsets in the immune response to bovine tuberculosis in cattle. PLoS One 2015; 10:e0122571. [PMID: 25879774 PMCID: PMC4400046 DOI: 10.1371/journal.pone.0122571] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/16/2015] [Indexed: 01/01/2023] Open
Abstract
Cultured IFN-γ ELISPOT assays are primarily a measure of central memory T cell (Tcm) responses with humans; however, this important subset of lymphocytes is poorly characterized in cattle. Vaccine-elicited cultured IFN-γ ELISPOT responses correlate with protection against bovine tuberculosis in cattle. However, whether this assay measures cattle Tcm responses or not is uncertain. The objective of the present study was to characterize the relative contribution of Tcm (CCR7+, CD62Lhi, CD45RO+), T effector memory (Tem, defined as: CCR7-, CD62Llow/int, CD45RO+), and T effector cells (CCR7-, CD62L-/low, CD45RO-), in the immune response to Mycobacterium bovis. Peripheral blood mononuclear cells (PBMC) from infected cattle were stimulated with a cocktail of M. bovis purified protein derivative, rTb10.4 and rAg85A for 13 days with periodic addition of fresh media and rIL-2. On day 13, cultured PBMC were re-stimulated with medium alone, rESAT-6:CFP10 or PPDb with fresh autologous adherent cells for antigen presentation. Cultured cells (13 days) or fresh PBMCs (ex vivo response) from the same calves were analyzed for IFN-γ production, proliferation, and CD4, CD45RO, CD62L, CD44, and CCR7 expression via flow cytometry after overnight stimulation. In response to mycobacterial antigens, ~75% of CD4+ IFN-γ+ cells in long-term cultures expressed a Tcm phenotype while less than 10% of the ex vivo response consisted of Tcm cells. Upon re-exposure to antigen, long-term cultured cells were highly proliferative, a distinctive characteristic of Tcm, and the predominant phenotype within the long-term cultures switched from Tcm to Tem. These findings suggest that proliferative responses of Tcm cells to some extent occurs simultaneously with reversion to effector phenotypes (mostly Tem). The present study characterizes Tcm cells of cattle and their participation in the response to M. bovis infection.
Collapse
|
22
|
Sánchez-Sampedro L, Perdiguero B, Mejías-Pérez E, García-Arriaza J, Di Pilato M, Esteban M. The evolution of poxvirus vaccines. Viruses 2015; 7:1726-803. [PMID: 25853483 PMCID: PMC4411676 DOI: 10.3390/v7041726] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
After Edward Jenner established human vaccination over 200 years ago, attenuated poxviruses became key players to contain the deadliest virus of its own family: Variola virus (VARV), the causative agent of smallpox. Cowpox virus (CPXV) and horsepox virus (HSPV) were extensively used to this end, passaged in cattle and humans until the appearance of vaccinia virus (VACV), which was used in the final campaigns aimed to eradicate the disease, an endeavor that was accomplished by the World Health Organization (WHO) in 1980. Ever since, naturally evolved strains used for vaccination were introduced into research laboratories where VACV and other poxviruses with improved safety profiles were generated. Recombinant DNA technology along with the DNA genome features of this virus family allowed the generation of vaccines against heterologous diseases, and the specific insertion and deletion of poxvirus genes generated an even broader spectrum of modified viruses with new properties that increase their immunogenicity and safety profile as vaccine vectors. In this review, we highlight the evolution of poxvirus vaccines, from first generation to the current status, pointing out how different vaccines have emerged and approaches that are being followed up in the development of more rational vaccines against a wide range of diseases.
Collapse
MESH Headings
- Animals
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Poxviridae/immunology
- Poxviridae/isolation & purification
- Smallpox/prevention & control
- Smallpox Vaccine/history
- Smallpox Vaccine/immunology
- Smallpox Vaccine/isolation & purification
- Vaccines, Attenuated/history
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/history
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
Collapse
Affiliation(s)
- Lucas Sánchez-Sampedro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Mauro Di Pilato
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| |
Collapse
|
23
|
Rahman MM, Noor M, Islam KM, Uddin MB, Hossain FMA, Zinnah MA, Mamun MA, Islam MR, Eo SK, Ashour HM. Molecular diagnosis of bovine tuberculosis in bovine and human samples: implications for zoonosis. Future Microbiol 2015; 10:527-35. [DOI: 10.2217/fmb.14.139] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Aim: To develop emerging diagnostic technique for bovine tuberculosis and to identify its potential risk factors. Materials & methods: Bacterial genomic DNA was isolated from bovine milk and human sputum samples and subjected to PCR using specific primer pairs. PCR results were validated using bacteriological cultures. Results: PCR amplification of the targeted DNA fragment of Mycobacterium bovis was successful in 12.33% (37/300) of the bovine samples. Interestingly, 500-bp DNA fragment was also amplified in 6.67% (6/90) of the sputum indicating the possibility of zoonotic transmission. Rearing of livestock in household, unpasteurized milk consumption and smoking were identified as potential risk factors. Conclusion: Results of the study may add value to bovine tuberculosis eradication campaigns to achieve the One Health initiative.
Collapse
Affiliation(s)
- Md Masudur Rahman
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
- Veterinary Research Institute, Hudcova 70, Brno 621 00, Czech Republic
| | - Monira Noor
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Kazi Mehetazul Islam
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Md Bashir Uddin
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ferdaus Mohd Altaf Hossain
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
- College of Veterinary Medicine & Bio-Safety Research Institute, Chonbuk National University, Jeonju 561–756, Republic of Korea
| | - Mohammad Ali Zinnah
- Faculty of Veterinary Medicine & Animal Science, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Mohammad Al Mamun
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Mohammad Rafiqul Islam
- Animal Health Research Division, Bangladesh Livestock Research Institute, Savar, Dhaka, Bangladesh
| | - Seong Kug Eo
- College of Veterinary Medicine & Bio-Safety Research Institute, Chonbuk National University, Jeonju 561–756, Republic of Korea
| | - Hossam M Ashour
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy & Health Sciences, Wayne State University, Detroit, MI 48202, USA
- Department of Microbiology & Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
24
|
Manjaly Thomas ZR, McShane H. Aerosol immunisation for TB: matching route of vaccination to route of infection. Trans R Soc Trop Med Hyg 2015; 109:175-81. [PMID: 25636950 PMCID: PMC4321022 DOI: 10.1093/trstmh/tru206] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 12/20/2022] Open
Abstract
TB remains a very significant global health burden. There is an urgent need for better tools for TB control, which include an effective vaccine. Bacillus Calmette-Guérin (BCG), the currently licensed vaccine, confers highly variable protection against pulmonary TB, the main source of TB transmission. Replacing BCG completely or boosting BCG with another vaccine are the two current strategies for TB vaccine development. Delivering a vaccine by aerosol represents a way to match the route of vaccination to the route of infection. This route of immunisation offers not only the scientific advantage of delivering the vaccine directly to the respiratory mucosa, but also practical and logistical advantages. This review summarises the state of current TB vaccine candidates in the pipeline, reviews current progress in aerosol administration of vaccines in general and evaluates the potential for TB vaccine candidates to be administered by the aerosol route.
Collapse
Affiliation(s)
| | - Helen McShane
- The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| |
Collapse
|
25
|
Leunda A, Baldo A, Goossens M, Huygen K, Herman P, Romano M. Novel GMO-Based Vaccines against Tuberculosis: State of the Art and Biosafety Considerations. Vaccines (Basel) 2014; 2:463-99. [PMID: 26344627 PMCID: PMC4494264 DOI: 10.3390/vaccines2020463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/24/2014] [Accepted: 05/06/2014] [Indexed: 12/13/2022] Open
Abstract
Novel efficient vaccines are needed to control tuberculosis (TB), a major cause of morbidity and mortality worldwide. Several TB vaccine candidates are currently in clinical and preclinical development. They fall into two categories, the one of candidates designed as a replacement of the Bacille Calmette Guérin (BCG) to be administered to infants and the one of sub-unit vaccines designed as booster vaccines. The latter are designed as vaccines that will be administered to individuals already vaccinated with BCG (or in the future with a BCG replacement vaccine). In this review we provide up to date information on novel tuberculosis (TB) vaccines in development focusing on the risk assessment of candidates composed of genetically modified organisms (GMO) which are currently evaluated in clinical trials. Indeed, these vaccines administered to volunteers raise biosafety concerns with respect to human health and the environment that need to be assessed and managed.
Collapse
Affiliation(s)
- Amaya Leunda
- Biosafety and Biotechnology Unit, Scientific Institute of Public Health, 14 Juliette Wytsman Street, Brussels 1050, Belgium.
| | - Aline Baldo
- Biosafety and Biotechnology Unit, Scientific Institute of Public Health, 14 Juliette Wytsman Street, Brussels 1050, Belgium.
| | - Martine Goossens
- Biosafety and Biotechnology Unit, Scientific Institute of Public Health, 14 Juliette Wytsman Street, Brussels 1050, Belgium.
| | - Kris Huygen
- Immunology Unit, Scientific Institute of Public Health, 642 Engeland Street, Brussels 1180, Belgium.
| | - Philippe Herman
- Biosafety and Biotechnology Unit, Scientific Institute of Public Health, 14 Juliette Wytsman Street, Brussels 1050, Belgium.
| | - Marta Romano
- Immunology Unit, Scientific Institute of Public Health, 642 Engeland Street, Brussels 1180, Belgium.
| |
Collapse
|
26
|
Dean G, Clifford D, Gilbert S, McShane H, Hewinson RG, Vordermeier HM, Villarreal-Ramos B. Effect of dose and route of immunisation on the immune response induced in cattle by heterologous Bacille Calmette-Guerin priming and recombinant adenoviral vector boosting. Vet Immunol Immunopathol 2014; 158:208-13. [PMID: 24581917 DOI: 10.1016/j.vetimm.2014.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 12/19/2013] [Accepted: 01/21/2014] [Indexed: 11/29/2022]
Abstract
BCG is used experimentally as a vaccine against tuberculosis (TB), induced by Mycobacterium bovis, in cattle (bTB). However, the efficacy of BCG is variable in humans, cattle and guinea pigs. An adenoviral vector expressing Antigen 85A (Ad5Ag85A) has enhanced protection against TB in mice when used in combination with BCG for prime-boost experiments. However, the route of immunisation affects the degree of protection seen. This work examines the immunogenicity of a new vectored vaccine (Ad5-TBF) that expresses Ag85A, Rv0287, Rv0288 and Rv0251c to explore the effects of dose of adenoviral boost and route of inoculation on immunogenicity. We found that 2×10(9) infectious units (iu) delivered intradermally conferred the most consistent and strongest responses of the different regimes tested.
Collapse
Affiliation(s)
- G Dean
- Bovine TB, AHVLA-Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK
| | - D Clifford
- Bovine TB, AHVLA-Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK
| | - S Gilbert
- The Jenner Institute Old Road Campus Research Building Oxford University, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - H McShane
- The Jenner Institute Old Road Campus Research Building Oxford University, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - R G Hewinson
- Bovine TB, AHVLA-Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK
| | - H M Vordermeier
- Bovine TB, AHVLA-Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK
| | | |
Collapse
|
27
|
Vordermeier M, Jones GJ, Whelan AO. DIVA reagents for bovine tuberculosis vaccines in cattle. Expert Rev Vaccines 2014; 10:1083-91. [DOI: 10.1586/erv.11.22] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Skinner MA, Laidlaw SM, Eldaghayes I, Kaiser P, Cottingham MG. Fowlpox virus as a recombinant vaccine vector for use in mammals and poultry. Expert Rev Vaccines 2014; 4:63-76. [PMID: 15757474 DOI: 10.1586/14760584.4.1.63] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Live vaccines against fowlpox virus, which causes moderate pathology in poultry and is the type species of the Avipoxvirus genus, were developed in the 1920s. Development of recombinant fowlpox virus vector vaccines began in the 1980s, for use not only in poultry, but also in mammals including humans. In common with other avipoxviruses, such as canarypox virus, fowlpox virus enters mammalian cells and expresses proteins, but replicates abortively. The use of fowlpox virus as a safe vehicle for expression of foreign antigens and host immunomodulators, is being evaluated in numerous clinical trials of vaccines against cancer, malaria, tuberculosis and AIDS, notably in heterologous prime-boost regimens. In this article, technical approaches to, and issues surrounding, the use of fowlpox virus as a recombinant vaccine vector in poultry and mammals are reviewed.
Collapse
Affiliation(s)
- Michael A Skinner
- Institute for Animal Health, Compton, Newbury, Berkshire, RG20 7NN, UK.
| | | | | | | | | |
Collapse
|
29
|
Beverley PCL, Sridhar S, Lalvani A, Tchilian EZ. Harnessing local and systemic immunity for vaccines against tuberculosis. Mucosal Immunol 2014; 7:20-6. [PMID: 24253104 DOI: 10.1038/mi.2013.99] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/09/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
Abstract
The lung is the portal of entry for Mycobacterium tuberculosis (Mtb) and animal experimental evidence indicates that local immune defense mechanisms are crucial for protective immunity. Immunization via the lower respiratory tract efficiently induces a dividing, activated, antigen-dependent, lung-resident, memory T-cell population, which is partly recoverable by bronchoalveolar lavage. These cells can inhibit the growth of Mtb in the lungs immediately after infection. Delivery of appropriate signals to the lung innate immune system is critical for induction of effective local immunity. In contrast after parenteral immunization, antigen-specific cells may be found in lung tissue but few are recoverable by lavage and inhibition of mycobacterial growth is delayed. Harnessing both local and systemic immunity can provide highly effective protection in animal models and the evidence suggests that taken in aggregate, multiple animal models may predict the success of novel vaccine strategies in humans.
Collapse
Affiliation(s)
- P C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S Sridhar
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - A Lalvani
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - E Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Gómez CE, Perdiguero B, García-Arriaza J, Esteban M. Clinical applications of attenuated MVA poxvirus strain. Expert Rev Vaccines 2013; 12:1395-416. [PMID: 24168097 DOI: 10.1586/14760584.2013.845531] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The highly attenuated poxvirus strain modified vaccinia virus Ankara (MVA) has reached maturity as a vector delivery system and as a vaccine candidate against a broad spectrum of diseases. This has been largely recognized from research on virus-host cell interactions and immunological studies in pre-clinical and clinical trials. This review addresses the studies of MVA vectors used in phase I/II clinical trials, with the aim to provide the main findings obtained on their behavior when tested against relevant human diseases and cancer and also highlights the strategies currently implemented to improve the MVA immunogenicity. The authors assess that MVA vectors are progressing as strong vaccine candidates either alone or when administered in combination with other vectors.
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | | | | |
Collapse
|
31
|
Canto Alarcon GJ, Rubio Venegas Y, Bojorquez Narvaez L, Pizano Martínez OE, García Casanova L, Sosa Gallegos S, Nava Vargas A, Olvera Ramírez AM, Milian Suazo F. Efficacy of a vaccine formula against tuberculosis in cattle. PLoS One 2013; 8:e76418. [PMID: 24204624 PMCID: PMC3799756 DOI: 10.1371/journal.pone.0076418] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 08/26/2013] [Indexed: 11/18/2022] Open
Abstract
"Test-and-slaughter" has been successful in industrialized countries to control and eradicate tuberculosis from cattle; however, this strategy is too expensive for developing nations, where the prevalence is especially high. Vaccination with the Calmette-Guérin (BCG) strain has been shown to protect against the development of lesions in vaccinated animals: mouse, cattle and wildlife species. In this study, the immune response and the pathology of vaccinated (BCG-prime and BCG prime-CFP-boosted) and unvaccinated (controls) calves were evaluated under experimental settings. A 10(6) CFU dose of the BCG strain was inoculated subcutaneously on the neck to two groups of ten animas each. Thirty days after vaccination, one of the vaccinated groups was boosted with an M. bovis culture filtrate protein (CFP). Three months after vaccination, the three groups of animals were challenged with 5×10(5) CFU via intranasal by aerosol with a field strain of M. bovis. The immune response was monitored throughout the study. Protection was assessed based on immune response (IFN-g release) prechallenge, presence of visible lesions in lymph nodes and lungs at slaughter, and presence of bacilli in lymph nodes and lung samples in histological analysis. Vaccinated cattle, either with the BCG alone or with BCG and boosted with CFP showed higher IFN-g response, fewer lesions, and fewer bacilli per lesion than unvaccinated controls after challenge. Animals with low levels of IFN-g postvaccine-prechallenge showed more lesions than animals with high levels. Results from this study support the argument that vaccination could be incorporated into control programs to reduce the incidence of TB in cattle in countries with high prevalence.
Collapse
Affiliation(s)
| | - Yezenia Rubio Venegas
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, Qro., México
| | | | - Oscar E. Pizano Martínez
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal, INIFAP, Ajuchitlán, Qro., México
| | - Leticia García Casanova
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal, INIFAP, Ajuchitlán, Qro., México
| | - Susana Sosa Gallegos
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, Qro., México
| | - Alejandro Nava Vargas
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal, INIFAP, Ajuchitlán, Qro., México
| | | | - Feliciano Milian Suazo
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, Qro., México
| |
Collapse
|
32
|
Lu M, Xia ZY, Bao L. Enhancement of antimycobacterial Th1-cell responses by a Mycobacterium bovis BCG prime-protein boost vaccination strategy. Cell Immunol 2013; 285:111-7. [PMID: 24177251 DOI: 10.1016/j.cellimm.2013.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/07/2013] [Accepted: 10/01/2013] [Indexed: 02/04/2023]
Abstract
Tuberculosis is a major global health problem, and the only available vaccine Bacille Calmette-Guérin (BCG) is not sufficiently effective against the disease. It is extremely urgent to develop novel vaccine approaches. Previous research demonstrated that there were several Regions of Difference (RD1-16) between the substrains of BCG and Mycobacterium tuberculosis or Mycobacterium bovis. The ORFs Rv1769 and Rv1772 are located in the RD14 deletions and have not been major targets of study. However, some studies have demonstrated that the two genes (Rv1769 and Rv1772) are excellent T cell antigens, which might induce an immune response. What kind of role these ORFs might play in anti-mycobacterial immunity, however, is still unknown. In our research we used the BCG prime-protein boost strategy to immunize BALB/c mice and evaluated its immunogenicity. Our data suggest that our novel BCG-P+PRO69 vaccine could elicit the most long-lasting and strongest Th1 type cellular immune responses. This response is characterized by a strong antibody response, the proliferation rate of splenocytes, a high percentage of CD4+ and CD8+ T cells and high levels of IFN-γ in antigen-stimulated splenocyte cultures. These results indicate that prime-boost is a potent strategy and the protein of gene Rv1769 is a potential antigen or subunit vaccine to TB for further study.
Collapse
Affiliation(s)
- Miao Lu
- Laboratory of Infection and Immunity, School of Basic Medical Science, West China Center of Medical Sciences, Sichuan University, Chengdu, China
| | | | | |
Collapse
|
33
|
Dalmia N, Ramsay AJ. Prime-boost approaches to tuberculosis vaccine development. Expert Rev Vaccines 2013; 11:1221-33. [PMID: 23176655 DOI: 10.1586/erv.12.94] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Four individuals die from active TB disease each minute, while at least 2 billion are latently infected and at risk for disease reactivation. BCG, the only licensed TB vaccine, is effective in preventing childhood forms of TB; however its poor efficacy in adults, emerging drug-resistant TB strains and tedious chemotherapy regimes, warrant the development of novel prophylactic measures. Designing safe and effective vaccines against TB will require novel approaches on several levels, including the administration of rationally selected mycobacterial antigens in efficient delivery vehicles via optimal immunization routes. Given the primary site of disease manifestation in the lungs, development of mucosal immunization strategies to generate protective immune responses both locally, and in the circulation, may be important for effective TB prophylaxis. This review focuses on prime-boost immunization strategies currently under investigation and highlights the potential of mucosal delivery and rational vaccine design based on systems biology.
Collapse
Affiliation(s)
- Neha Dalmia
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, 1901 Perdido Street, New Orleans, LA 70112, USA
| | | |
Collapse
|
34
|
Marassi CD, Medeiros L, Figueiredo E, Fonseca LS, Duarte R, Paschoalin V, Oelemann WM, Lilenbaum W. A multidisciplinary approach to diagnose naturally occurring bovine tuberculosis in Brazil. PESQUISA VETERINÁRIA BRASILEIRA 2013. [DOI: 10.1590/s0100-736x2013000100004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A herd infected naturally with tuberculosis was investigated by different diagnostic methods. Ninety days after a screening test that identified 21 cows as skin test positive, a Comparative Intradermal Tuberculin Test (CITT) was performed in those 21 cows and in 29 other randomly selected skin test negative cows. Milk samples and nasal swabs were collected prior to the CITT for bacteriological culture and PCR, while blood samples were collected for IFN release and antibody responses to MPB70 and MPB83, at three time points post tuberculin injection. Animals positive by CITT were slaughtered and disease confirmation undertaken. Based on the Kappa test, IFN was comparable to the standard tests (culture, PCR and CITT) at all three sampling points. Results from both antibody ELISAs were similar but were not comparable to the standard tests. T-test analysis of the CITT, IFN and ELISAs demonstrated that their performances were not correlated. There is increasing recognition that individually, available diagnostic tests do not detect all infected cattle. Therefore, a comprehensive strategy for the diagnosis of bovine TB should include test results for the detection of both cellular and humoral immune responses where there may be animals at different stages of infection.
Collapse
|
35
|
Whelan A, Court P, Xing Z, Clifford D, Hogarth PJ, Vordermeier M, Villarreal-Ramos B. Immunogenicity comparison of the intradermal or endobronchial boosting of BCG vaccinates with Ad5-85A. Vaccine 2012; 30:6294-300. [PMID: 22885013 DOI: 10.1016/j.vaccine.2012.07.086] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/10/2012] [Accepted: 07/31/2012] [Indexed: 11/18/2022]
Abstract
Experiments in small animal models have indicated that intranasal vaccination confers a greater degree of protection against TB than other routes such as intradermal (i.d.) or intramuscular. In this work, using a prime-boost vaccination strategy, we have compared in cattle vaccinated with BCG as a priming vaccine the boosting capabilities of Ad5-85A delivered either via the endobronchial (e.b.) or i.d. route. We show that Ad5-85A delivered through either route induced comparable peripheral blood antigen specific responses, and that both i.d. and e.b. routes induced bronchioalveolar lavage cells (BALC) that produced antigen-specific IFNgamma. We also show that, regardless of the route of boosting, the kinetics of peripheral blood and BALC responses, as assessed by antigen specific IFNgamma production, are different with systemic responses being detectable earlier than mucosal responses. These results contribute to our understanding on how different vaccination strategies may affect different compartments of the immune response and in turn to the development of safer and more effective vaccines.
Collapse
Affiliation(s)
- Adam Whelan
- TB Research Group, Animal Health and Veterinary Laboratories Agency, Weybridge, Surrey KT 15 3NB, UK
| | | | | | | | | | | | | |
Collapse
|
36
|
Opportunities for improved serodiagnosis of human tuberculosis, bovine tuberculosis, and paratuberculosis. Vet Med Int 2012; 2012:674238. [PMID: 22720192 PMCID: PMC3375143 DOI: 10.1155/2012/674238] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 04/02/2012] [Indexed: 11/21/2022] Open
Abstract
Mycobacterial infections—tuberculosis (TB), bovine tuberculosis (bTB), and Johne's disease (JD)—are major infectious diseases of both human and animals. Methods presently in use for diagnosis of mycobacterial infections include bacterial culture, nucleic acid amplification, tuberculin skin test, interferon-γ assay, and serology. Serological tests have several advantages over other methods, including short turn-around time, relatively simple procedures, and low cost. However, current serodiagnostic methods for TB, bTB and JD exhibit low sensitivity and/or specificity. Recent studies that have aimed to develop improved serodiagnostic tests have mostly focused on identifying useful species-specific protein antigens. A review of recent attempts to improve diagnostic test performance indicates that the use of multiple antigens can improve the accuracy of serodiagnosis of these mycobacterial diseases. Mycobacteria also produce a variety of species-specific nonprotein molecules; however, only a few such molecules (e.g., cord factor and lipoarabinomannan) have so far been evaluated for their effectiveness as diagnostic antigens. For TB and bTB, there has been recent progress in developing laboratory-free diagnostic methods. New technologies such as microfluidics and “Lab-on-Chip” are examples of promising new technologies that can underpin development of laboratory-free diagnostic devices for these mycobacterial infections.
Collapse
|
37
|
Guzman E, Cubillos-Zapata C, Cottingham MG, Gilbert SC, Prentice H, Charleston B, Hope JC. Modified vaccinia virus Ankara-based vaccine vectors induce apoptosis in dendritic cells draining from the skin via both the extrinsic and intrinsic caspase pathways, preventing efficient antigen presentation. J Virol 2012; 86:5452-66. [PMID: 22419811 PMCID: PMC3347273 DOI: 10.1128/jvi.00264-12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/05/2012] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC) are potent antigen-presenting cells and central to the induction of immune responses following infection or vaccination. The collection of DC migrating from peripheral tissues by cannulation of the afferent lymphatic vessels provides DC which can be used directly ex vivo without extensive in vitro manipulations. We have previously used bovine migrating DC to show that recombinant human adenovirus 5 vectors efficiently transduce afferent lymph migrating DEC-205(+) CD11c(+) CD8(-) DC (ALDC). We have also shown that recombinant modified vaccinia virus Ankara (MVA) infects ALDC in vitro, causing downregulation of costimulatory molecules, apoptosis, and cell death. We now show that in the bovine system, modified vaccinia virus Ankara-induced apoptosis in DC draining from the skin occurs soon after virus binding via the caspase 8 pathway and is not associated with viral gene expression. We also show that after virus entry, the caspase 9 pathway cascade is initiated. The magnitude of T cell responses to mycobacterial antigen 85A (Ag85A) expressed by recombinant MVA-infected ALDC is increased by blocking caspase-induced apoptosis. Apoptotic bodies generated by recombinant MVA (rMVA)-Ag85A-infected ALDC and containing Ag85A were phagocytosed by noninfected migrating ALDC expressing SIRPα via actin-dependent phagocytosis, and these ALDC in turn presented antigen. However, the addition of fresh ALDC to MVA-infected cultures did not improve on the magnitude of the T cell responses; in contrast, these noninfected DC showed downregulation of major histocompatibility complex class II (MHC-II), CD40, CD80, and CD86. We also observed that MVA-infected ALDC promoted migration of DEC-205(+) SIRPα(+) CD21(+) DC as well as CD4(+) and CD8(+) T cells independently of caspase activation. These in vitro studies show that induction of apoptosis in DC by MVA vectors is detrimental to the subsequent induction of T cell responses.
Collapse
Affiliation(s)
- E Guzman
- Institute for Animal Health, Compton, Newbury, Berkshire, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
38
|
Cayabyab MJ, Kashino SS, Campos-Neto A. Robust immune response elicited by a novel and unique Mycobacterium tuberculosis protein using an optimized DNA/protein heterologous prime/boost protocol. Immunology 2012; 135:216-25. [PMID: 22043824 DOI: 10.1111/j.1365-2567.2011.03525.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An efficacious tuberculosis (TB) vaccine will probably need to induce both CD4 and CD8 T-cell responses specific to a protective Mycobacterium tuberculosis antigen(s). To achieve this broad cellular immune response we tested a heterologous DNA/protein combination vaccine strategy. We used a purified recombinant protein preparation of a unique M. tuberculosis antigen (rMT1721) found in the urine of TB patients, an optimized plasmid DNA expressing this protein (DNA-MT1721), and a Toll-like receptor 4 agonist adjuvant. We found that priming mice with DNA-MT1721 and subsequently boosting with rMT1721 elicited high titres of specific IgG1 and IgG2a antibodies as well as high magnitude and polyfunctional CD4(+) T-cell responses. However, no detectable CD8(+) T-cell response was observed using this regimen of immunization. In contrast, both CD4(+) and CD8(+) T-cell responses were detected after a prime/boost vaccination regimen using rMT1721 as the priming antigen and DNA-MT1721 as the boosting immunogen. These findings support the exploration of heterologous DNA/protein immunization strategies in vaccine development against TB and possibly other infectious diseases.
Collapse
Affiliation(s)
- Mark J Cayabyab
- Global Infectious Disease Research Center, The Forsyth Institute, Cambridge, MA, USA
| | | | | |
Collapse
|
39
|
Vordermeier M, Whelan AO. ELISPOT assays to enumerate bovine IFN-γ-secreting cells for the development of novel vaccines against bovine tuberculosis. Methods Mol Biol 2012; 792:219-27. [PMID: 21956513 DOI: 10.1007/978-1-61779-325-7_17] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Enumeration of antigen-specific cells after vaccination is one of the prime immunological parameters determined when developing vaccines. Due to their exquisite sensitivity (limits of detection can be below 1/100,000 cells), ELISPOT assays are therefore an important tool in vaccine development programs. This is particularly the case for vaccines against diseases that require protective cell-mediated immunity, such as tuberculosis. This chapter describes ELISPOT assays detecting bovine IFN-γ.
Collapse
Affiliation(s)
- Martin Vordermeier
- Department of Bacteriology, TB Research Group, Veterinary Laboratories Agency, WeybridgeAddlestone, Surrey, UK.
| | | |
Collapse
|
40
|
Luo Y, Jiang W, Da Z, Wang B, Hu L, Zhang Y, An R, Yu H, Sun H, Tang K, Tang Z, Wang Y, Jing T, Zhu B. Subunit Vaccine Candidate AMM Down-Regulated the Regulatory T Cells and Enhanced the Protective Immunity of BCG on a Suitable Schedule. Scand J Immunol 2012; 75:293-300. [DOI: 10.1111/j.1365-3083.2011.02666.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Improved skin test for differential diagnosis of bovine tuberculosis by the addition of Rv3020c-derived peptides. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:620-2. [PMID: 22301696 DOI: 10.1128/cvi.00024-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A peptide cocktail derived from the mycobacterial antigens ESAT-6, CFP-10, and Rv3615c allowed differentiation between Mycobacterium bovis-infected and M. bovis bacillus Calmette-Guérin (BCG)-vaccinated cattle when used as a skin test reagent for a "DIVA" test (i.e., a test capable of differentiating infected and uninfected vaccinated animals). Addition of the antigen Rv3020c improves the diagnostic sensitivity without compromising specificity in the face of BCG or Johne's disease vaccination.
Collapse
|
42
|
Cosgrove MK, Campa H, Schmitt SM, Marks DR, Wilson AS, O'Brien DJ. Live-trapping and bovine tuberculosis testing of free-ranging white-tailed deer for targeted removal. WILDLIFE RESEARCH 2012. [DOI: 10.1071/wr11147] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Context
Significant efforts have been made in Michigan, USA, to reduce the prevalence of bovine tuberculosis (TB) in free-ranging white-tailed deer (Odocoileus virginianus) over the past 15 years. Since 2002, however, prevalence has changed little, prompting the need for new control strategies.
Aims
In January–March of 2007 and 2008, a trap–test–cull project was conducted on an 11 000-ha property in the north-eastern Lower Peninsula of Michigan. The objectives were to assess the feasibility of live-trapping and testing white-tailed deer for TB as a means for targeted removal and estimate the cost of this effort.
Methods
Live-trapped deer were ear-tagged and a blood sample was drawn for use with the CervidTB STAT-PAK (commonly called Rapid Test) for TB diagnosis in the field. Deer testing negative were released, whereas deer testing positive were euthanised to confirm blood-test results via bacterial culture.
Key results
In all, 762 (741 with known sex and age) individual deer were captured and tested for TB. Adults comprised 59% (437 of 741) of the captures. Eight (1.8%) adults were positive on the blood test; six of eight (1.4% of adults) were confirmed TB positive via bacterial culture. Estimated TB prevalence in the present study was 2.5% (adjusted for Rapid Test sensitivity of 56%), being lower than what would be expected on the basis of routine hunter-harvest surveillance for this site which has yielded prevalence rates from 3.4% to 4.8%. Results demonstrated the ability to trap and test a substantial number of deer given high deer densities (16–20 deer per km2), availability of traps and abundant workers. The 2-year project cost a total of ~US$228 000, or US$38 000 per culture-positive animal.
Conclusions
Because of the cost and effort involved, a project such as the present one applied to Michigan’s larger TB-management area (148 018 ha) is not feasible.
Implications
If the efficiency and effectiveness of a trap–test–cull project could be improved by vaccinating test-negative animals, should a vaccine be approved for use in free-ranging white-tailed deer, a trap–test–cull project applied on a scale similar to the present study may prove beneficial by possibly reducing disease transmission, in addition to removing TB-positive animals.
Collapse
|
43
|
West NP, Thomson SA, Triccas JA, Medveczky CJ, Ramshaw IA, Britton WJ. Delivery of a multivalent scrambled antigen vaccine induces broad spectrum immunity and protection against tuberculosis. Vaccine 2011; 29:7759-65. [PMID: 21846485 DOI: 10.1016/j.vaccine.2011.07.109] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/21/2011] [Accepted: 07/25/2011] [Indexed: 12/23/2022]
Abstract
The development of effective anti-Tuberculosis (TB) vaccines is an important step towards improved control of TB in high burden countries. Subunit vaccines are advantageous in terms of safety, particularly in the context of high rates of HIV co-infection, but they must contain sufficient Mycobacterium tuberculosis antigens to stimulate immunity in genetically diverse human populations. We have used a novel approach to develop a synthetic scrambled antigen vaccine (TB-SAVINE), comprised of overlapping, recombined peptides from four M. tuberculosis proteins, Ag85B, ESAT-6, PstS3 and Mpt83, each of which is immunogenic and protective against experimental TB. This polyvalent TB-SAVINE construct stimulated CD4 and CD8T cell responses against the individual proteins and M. tuberculosis in C57BL/6 and Balb/c mice, when delivered as DNA, Fowl Pox Virus or Vaccinia Virus vaccines. In addition, the DNA-TBS vaccine induced protective immunity against pulmonary M. tuberculosis infection in C57BL/6 mice. Co-immunization of Balb/c mice with virally expressed TBS and HIV1-SAVINE vaccine stimulated strong T cell responses to both the M. tuberculosis and HIV proteins, indicating no effects of antigenic competition. Further development of this TB-SAVINE vaccine expressing components from multiple M. tuberculosis proteins may prove an effective vaccine candidate against TB, which could potentially form part of a safe, combined preventative strategy together with HIV immunisations.
Collapse
Affiliation(s)
- Nicholas P West
- Mycobacterial Research Program, Centenary Institute, NSW, 2042, Australia.
| | | | | | | | | | | |
Collapse
|
44
|
Jones GJ, Bagaini F, Hewinson RG, Vordermeier HM. The use of binding-prediction models to identify M. bovis-specific antigenic peptides for screening assays in bovine tuberculosis. Vet Immunol Immunopathol 2011; 141:239-45. [DOI: 10.1016/j.vetimm.2011.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 02/08/2011] [Accepted: 03/02/2011] [Indexed: 11/28/2022]
|
45
|
Immunogenicity and protective efficacy against murine tuberculosis of a prime-boost regimen with BCG and a DNA vaccine expressing ESAT-6 and Ag85A fusion protein. Clin Dev Immunol 2011; 2011:617892. [PMID: 21461375 PMCID: PMC3065234 DOI: 10.1155/2011/617892] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Heterologous prime-boost regimens utilizing BCG as a prime vaccine probably represent the best hope for the development of novel tuberculosis (TB) vaccines. In this study, we examined the immunogenicity and protective efficacy of DNA vaccine (pcD685A) expressing the fusion protein of Ag85A and ESAT-6 (r685A) and its booster effects in BCG-immunized mice. The recombinant r685A fusion protein stimulated higher level of antigen-specific IFN-γ release in tuberculin skin test- (TST-) positive healthy household contacts of active pulmonary TB patients than that in TST-negative population. Vaccination of C57BL/6 mice with pcD685A resulted in significant protection against challenge with virulent Mycobacterium tuberculosis H37Rv when compared with the control group. Most importantly, pcD685A could act as a BCG booster and amplify Th1-type cell-mediated immunity in the lung of BCG-vaccinated mice as shown the increased expression of IFN-γ. The most significant reduction in bacterial load of both spleen and lung was obtained in mice vaccinated with BCG prime and pcD685A DNA booster when compared with BCG or pcD685A alone. Thus, our study indicates that pcD685A may be an efficient booster vaccine against TB with a strong ability to enhance prior BCG immunity.
Collapse
|
46
|
Buddle BM, Wedlock DN, Denis M, Vordermeier HM, Hewinson RG. Update on vaccination of cattle and wildlife populations against tuberculosis. Vet Microbiol 2011; 151:14-22. [PMID: 21420804 DOI: 10.1016/j.vetmic.2011.02.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this review, the status of vaccination strategies to reduce bovine tuberculosis of cattle and wildlife reservoirs of the disease is discussed, with a focus on recent developments. Recent work in vaccines to protect humans against tuberculosis has been followed by a similar surge of interest in developing vaccines against bovine tuberculosis. The human vaccine, bacille Calmette-Guérin (BCG) affords protection against tuberculosis in cattle, but this protection is variable. In addition, vaccination with BCG compromises control strategies based on skin testing animals. In general, no single vaccine approach has shown itself to be significantly superior to BCG alone, however, vaccine combinations of BCG and vaccinating moiety such as adjuvanted subunit, virus vectored or DNA vaccines have been shown to induce protection superior to that achieved by BCG alone. Vaccinating wildlife species against tuberculosis is also an area which has been subjected to scrutiny. Recent work has focused on vaccinating wildlife orally, via the use of BCG formulated in baits consumed by these species. Results from trials in a number of animal species indicate that oral BCG vaccination can reduce disease severity following experimental challenge with Mycobacterium bovis and in a recent field trial, oral BCG vaccination was shown to prevent infection of wild possums following natural exposure to M. bovis. In conclusion, recent studies in cattle and wildlife have demonstrated the practicality and effectiveness of vaccinating animals against tuberculosis and provide much impetus for future use of vaccines.
Collapse
Affiliation(s)
- Bryce M Buddle
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand.
| | | | | | | | | |
Collapse
|
47
|
Mycobacterium bovis antigens for the differential diagnosis of vaccinated and infected cattle. Vet Microbiol 2011; 151:8-13. [PMID: 21411245 DOI: 10.1016/j.vetmic.2011.02.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The urgency for new and improved cattle vaccines and diagnostic reagents for Bovine tuberculosis (TB) has made their development a research priority in Great Britain (GB). Significant progress has been made to develop specific antigens that allow the differentiation of BCG vaccinated and Mycobacterium bovis infected cattle (DIVA test). This has been greatly facilitated by the completion of the genome sequences of M. tuberculosis, M. bovis and BCG Pasteur and the subsequent application of comparative genome and transcriptome analysis to define DIVA antigens that complemented the prototype DIVA antigens ESAT-6 and CFP-10 by increasing their test sensitivity. Finally, we present an up-date of our current approaches in this area.
Collapse
|
48
|
Identification of surrogates and correlates of protection in protective immunity against Mycobacterium bovis infection induced in neonatal calves by vaccination with M. bovis BCG Pasteur and M. bovis BCG Danish. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:373-9. [PMID: 21228141 DOI: 10.1128/cvi.00543-10] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Vaccination of neonatal calves with Mycobacterium bovis bacillus Calmette-Guérin (BCG) induces a significant degree of protection against infection with virulent M. bovis, the causative agent of bovine tuberculosis (bTB). We compared two strains of BCG, Pasteur and Danish, in order to confirm that the current European human vaccine strain (BCG Danish) induced protective immunity in calves, and we assessed immune responses to determine correlates of protection that could assist future vaccine evaluation in cattle. Both vaccine strains induced antigen (purified protein derivate [PPD])-specific gamma interferon (IFN-γ) in whole-blood cultures. These responses were not significantly different for BCG Pasteur and BCG Danish and peaked at week 2 to 4 postvaccination. Vaccination with either BCG Danish or BCG Pasteur induced significant protection against bTB, with reductions in both lesion score and bacteriological burden evident in both groups of vaccinated calves compared with nonvaccinated control calves. Measurement of IFN-γ-expressing T lymphocytes postvaccination and postchallenge revealed both correlates and surrogates of protective efficacy. The frequency of central memory T lymphocytes present at 12 weeks postvaccination (at the time of M. bovis challenge) correlated significantly with protection. Conversely, the number of IFN-γ-expressing effector T cells present after M. bovis challenge was correlated with disease. These results demonstrate that vaccination of neonatal calves with either BCG Pasteur or BCG Danish induces protective immune responses against TB. In addition, we show that measurement of antigen-specific T lymphocyte populations may provide a reliable means for identifying protective vaccine candidates.
Collapse
|
49
|
Medeiros LDS, Marassi CD, Figueiredo EES, Lilenbaum W. Potential application of new diagnostic methods for controlling bovine tuberculosis in Brazil. Braz J Microbiol 2010; 41:531-41. [PMID: 24031527 PMCID: PMC3768653 DOI: 10.1590/s1517-83822010005000002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 08/20/2009] [Accepted: 02/18/2010] [Indexed: 11/22/2022] Open
Abstract
Bovine tuberculosis, a chronic infection in cattle caused by Mycobacterium bovis, remains an economic and public health problem for several countries. Due to its economic impact on international trade, contagious nature, and implications for human health, global programs to eradicate the disease were implemented worldwide. Those programs are based on slaughtering PPD-reactive animals. Despite the National Programs in Brazil, complete eradication has not been achieved, and the disease remains, albeit at a lower prevalence. The central purpose of this review is to address diagnostic tests for tuberculosis. Considering the course of the infection in cattle, at least two tests, ideally complementary to one another, may be necessary for an adequate diagnosis: the first based on the cellular response, and the second capable of identifying anergic animals by detection of specific anti-M.bovis antibodies.
Collapse
|
50
|
Sharpe SA, McShane H, Dennis MJ, Basaraba RJ, Gleeson F, Hall G, McIntyre A, Gooch K, Clark S, Beveridge NER, Nuth E, White A, Marriott A, Dowall S, Hill AVS, Williams A, Marsh PD. Establishment of an aerosol challenge model of tuberculosis in rhesus macaques and an evaluation of endpoints for vaccine testing. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1170-82. [PMID: 20534795 PMCID: PMC2916246 DOI: 10.1128/cvi.00079-10] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 04/06/2010] [Accepted: 06/01/2010] [Indexed: 01/19/2023]
Abstract
The establishment of an aerosol challenge model in nonhuman primates (NHPs) for the testing of vaccines against Mycobacterium tuberculosis would assist the global effort to optimize novel vaccination strategies. The endpoints used in preclinical challenge studies to identify measures of disease burden need to be accurate and sensitive enough to distinguish subtle differences and benefits afforded by different tuberculosis (TB) vaccine regimens when group sizes are inevitably small. This study sought to assess clinical and nonclinical endpoints as potentially sensitive measures of disease burden in a challenge study with rhesus macaques by using a new protocol of aerosol administration of M. tuberculosis. Immunological and clinical readouts were assessed for utility in vaccine evaluation studies. This is the first example of TB vaccine evaluation with rhesus macaques where long-term survival was one of the primary endpoints. However, we found that in NHP vaccine efficacy studies with maximum group sizes of six animals, survival did not provide a valuable endpoint. Two approaches used in human clinical trials for the evaluation of the gamma interferon (IFN-gamma) response to vaccination (enzyme-linked immunospot [ELISpot] assay and enzyme-linked immunosorbent assay [ELISA]) were included in this study. The IFN-gamma profiles induced following vaccination were found not to correlate with protection, nor did the level of purified protein derivative (PPD)-specific proliferation. The only readout to reliably distinguish vaccinated and unvaccinated NHPs was the determination of lung lesion burden using magnetic resonance (MR) imaging combined with stereology at the end of the study. Therefore, the currently proposed key markers were not shown to correlate with protection, and only imaging offered a potentially reliable correlate.
Collapse
Affiliation(s)
- S A Sharpe
- Health Protection Agency, Centre for Emergency Preparedness and Response, Porton Down, Salisbury, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|