1
|
Abil OZ, Liu S, Yeh YW, Wu Y, Sen Chaudhuri A, Li NS, Deng C, Xiang Z. A mucosal vaccine formulation against tuberculosis by exploiting the adjuvant activity of S100A4-A damage-associated molecular pattern molecule. Vaccine 2024; 42:126151. [PMID: 39089961 DOI: 10.1016/j.vaccine.2024.07.052] [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: 03/11/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains one of the top three causes of death. Currently, the only licensed vaccine against TB is the bacillus Calmette-Guerin (BCG), which lacks efficacy in preventing and controlling pulmonary TB in adults. We aimed to evaluate a nasal TB vaccine formulation composed of the Mtb-specific vaccine antigen ESAT-6, an Mtb-associated protein that can trigger protective immune responses, and S100A4, a recently characterized novel mucosal adjuvant. Mice were intranasally given recombinant ESAT-6 in the presence or absence of S100A4 as an adjuvant. We have provided experimental evidence demonstrating that S100A4 admixed to ESAT-6 could induce Mtb-specific adaptive immune responses after intranasal immunization. S100A4 remarkably augmented the levels of anti-ESAT-6 IgG in serum and IgA in mucosal sites, including lung exudates, bronchoalveolar lavage fluid (BALF) and nasal lavage. Furthermore, in both lung and spleen tissues, S100A4 strongly promoted ESAT-6-specific expansion of CD4 T cells. Both CD4 and CD8 T cells from these tissues expressed increased levels of IFN-γ, TNF-α, and IL-17, cytokines critical for antimicrobial activity. Antigen-reencounter-induced T cell proliferative responses, a key vaccine performance indicator, were augmented in the spleen of S100A4-adjuvanted mice. Furthermore, CD8 T cells from the spleen and lung tissues of these mice expressed higher levels of granzyme B upon antigen re-stimulation. S100A4-adjuvanted immunization may predict good mucosal protection against TB.
Collapse
Affiliation(s)
- Olifan Zewdie Abil
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Shuwei Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yu-Wen Yeh
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yuxuan Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Arka Sen Chaudhuri
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Nga Shan Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Chujun Deng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zou Xiang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
2
|
Passos BBS, Araújo-Pereira M, Vinhaes CL, Amaral EP, Andrade BB. The role of ESAT-6 in tuberculosis immunopathology. Front Immunol 2024; 15:1383098. [PMID: 38633252 PMCID: PMC11021698 DOI: 10.3389/fimmu.2024.1383098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Despite major global efforts to eliminate tuberculosis, which is caused by Mycobacterium tuberculosis (Mtb), this disease remains as a major plague of humanity. Several factors associated with the host and Mtb interaction favor the infection establishment and/or determine disease progression. The Early Secreted Antigenic Target 6 kDa (ESAT-6) is one of the most important and well-studied mycobacterial virulence factors. This molecule has been described to play an important role in the development of tuberculosis-associated pathology by subverting crucial components of the host immune responses. This review highlights the main effector mechanisms by which ESAT-6 modulates the immune system, directly impacting cell fate and disease progression.
Collapse
Affiliation(s)
- Beatriz B. S. Passos
- Curso de Medicina, Universidade Salvador, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
| | - Mariana Araújo-Pereira
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Caian L. Vinhaes
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Programa de Pós-Graduação em Medicina e Saúde Humana, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
- Departamento de Infectologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Bruno B. Andrade
- Curso de Medicina, Universidade Salvador, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Instituto de Pesquisa Clínica e Translacional, Faculdade Zarns, Clariens Educação, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Programa de Pós-Graduação em Medicina e Saúde Humana, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| |
Collapse
|
3
|
Hoseinpour R, Hasani A, Baradaran B, Abdolalizadeh J, Salehi R, Hasani A, Nabizadeh E, Yekani M, Hasani R, Kafil HS, Azizian K, Memar MY. Tuberculosis vaccine developments and efficient delivery systems: A comprehensive appraisal. Heliyon 2024; 10:e26193. [PMID: 38404880 PMCID: PMC10884459 DOI: 10.1016/j.heliyon.2024.e26193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Despite the widespread use of the Bacillus Calmette-Guérin (BCG) vaccine, Mycobacterium tuberculosis (MTB) continues to be a global burden. Vaccination has been proposed to prevent and treat tuberculosis (TB) infection, and several of them are in different phases of clinical trials. Though vaccine production is in progress but requires more attention. There are several TB vaccines in the trial phase, most of which are based on a combination of proteins/adjuvants or recombinant viral vectors used for selected MTB antigens. In this review, we attempted to discuss different types of TB vaccines based on the vaccine composition, the immune responses generated, and their clinical trial phases. Furthermore, we have briefly overviewed the effective delivery systems used for the TB vaccine and their effectiveness in different vaccines.
Collapse
Affiliation(s)
- Rasoul Hoseinpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Laboratory sciences and Microbiology, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Alka Hasani
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit, Sina Educational, Research, and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Hasani
- Department of Clinical Biochemistry and Applied Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Edris Nabizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Hossein Samadi Kafil
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Azizian
- Department of Microbiology, Faculty of Medicine, Kurdistan University of Medical Science, Sanandaj, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Fan X, Zhao X, Wang R, Li M, Luan X, Wang R, Wan K, Liu H. A novel multistage antigens ERA005f confer protection against Mycobacterium tuberculosis by driving Th-1 and Th-17 type T cell immune responses. Front Immunol 2023; 14:1276887. [PMID: 38022539 PMCID: PMC10662081 DOI: 10.3389/fimmu.2023.1276887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Tuberculosis (TB) is a major threat to human health. In 2021, TB was the second leading cause of death after COVID-19 among infectious diseases. The Bacillus Calmette-Guérin vaccine (BCG), the only licensed TB vaccine, is ineffective against adult TB. Therefore, there is an urgent need to develop new effective vaccines. Methods In this study, we developed a novel multistage subunit vaccine (ERA005f) comprising various proteins expressed in metabolic states, based on three immunodominant antigens (ESAT-6, Rv2628, and Ag85B). We utilized the E. coli prokaryotic expression system to express ERA005f and subsequently purified the protein using nickel affinity chromatography and anion exchange. Immunogenicity and protective efficacy of ERA005f and ERA005m were evaluated in BALB/c mice. Results ERA005f was consistently expressed as an inclusion body in a prokaryotic expression system, and a highly pure form of the protein was successfully obtained. Both ERA005f and ERA005m significantly improved IgG titers in the serum. In addition, mice immunized with ERA005f and ERA005m generated higher titers of antigen-specific IgG2a than the other groups. Elispot results showed that, compared with other groups, ERA005f increased the numbers of IFN-γ-secreting and IL-4-secreting T cells, especially the number of IFN-γ-secreting T cells. Meanwhile, ERA005f induced a higher number of IFN-γ+ T lymphocytes than ERA005m did. In addition, ERA005f improved the expression of cytokines, including IFN-γ, IL-12p70, TNF-α, IL-17, and GM-CSF and so on. Importantly, both ERA005f and ERA005m significantly inhibited the growth of Mtb. Conclusion The novel multistage antigen ERA005f elicited a strong antigen-specific humoral response and Th-1 and Th-17 cell-mediated immunity in mice. Meanwhile, it can effectively inhibit H37Rv growth in vitro, and represents a correlate of protection in vivo, indicating that ERA005f may exhibit excellent protective efficacy against Mycobacterium tuberculosis H37Rv infection. Our study suggests that ERA005f has the potential to be a promising multistage tuberculosis vaccine candidate.
Collapse
Affiliation(s)
- Xueting Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuqin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruibai Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Machao Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuli Luan
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Ruihuan Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kanglin Wan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haican Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
5
|
Duong VT, Skwarczynski M, Toth I. Towards the development of subunit vaccines against tuberculosis: The key role of adjuvant. Tuberculosis (Edinb) 2023; 139:102307. [PMID: 36706503 DOI: 10.1016/j.tube.2023.102307] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
According to the World Health Organization (WHO), tuberculosis (TB) is the leading cause of death triggered by a single infectious agent, worldwide. Bacillus Calmette-Guerin (BCG) is the only currently licensed anti-TB vaccine. However, other strategies, including modification of recombinant BCG vaccine, attenuated Mycobacterium tuberculosis (Mtb) mutant constructs, DNA and protein subunit vaccines, are under extensive investigation. As whole pathogen vaccines can trigger serious adverse reactions, most current strategies are focused on the development of safe anti-TB subunit vaccines; this is especially important given the rising TB infection rate in immunocompromised HIV patients. The whole Mtb genome has been mapped and major antigens have been identified; however, optimal vaccine delivery mode is still to be established. Isolated protein antigens are typically poorly immunogenic so adjuvants are required to induce strong and long-lasting immune responses. This article aims to review the developmental status of anti-TB subunit vaccine adjuvants.
Collapse
Affiliation(s)
- Viet Tram Duong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
6
|
Bhatt P, Sharma M, Prakash Sharma P, Rathi B, Sharma S. Mycobacterium tuberculosis dormancy regulon proteins Rv2627c and Rv2628 as Toll like receptor agonist and as potential adjuvant. Int Immunopharmacol 2022; 112:109238. [PMID: 36116151 DOI: 10.1016/j.intimp.2022.109238] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
During latency, DosR proteins of Mycobacterium tuberculosis (M.tb) get activated and help the bacterium to remain dormant. We have shown earlier that 2 such proteins Rv2627c and Rv2628 are immunogenic and induce a TH1 kind of immune response. In this study, through in-vitro experiments we have confirmed that Rv2627c and Rv2628 proteins act as protein Toll-Like Receptor (TLR) agonist-adjuvant. Rv2627c and Rv2628 stimulated THP-1 macrophages showed an increased expression of TLR2, TLR4 and co-stimulatory molecules CD40, CD80, CD86 and antigen presenting molecule HLA-DR. Further studies also found enhanced expression of downstream signaling molecules of TLR activation like MyD88, NF-κB-p65 and pro-inflammatory cytokines. Inhibition studies using TLR blocking antibodies decreased the expression of co-stimulatory molecules, MyD88, NF-κB-p65, and pro-inflammatory cytokines. Rv2627c and Rv2628 stimulation of HEK-TLR2 reporter cell line confirmed the interaction of these proteins with TLR2. Moreover, molecular docking and simulations of Rv2627c and Rv2628 proteins with TLR2 and TLR4 showed stable interactions. The adjuvant activity of Rv2628 was further validated by a protein adjuvanted with pre-clinically validated peptides as multi-epitope vaccine construct which showed good binding with TLR2 and TLR4 and activate dendritic cells and induce sustained pro-inflammatory cytokine response by C-ImmSim analysis. We propose that our vaccine construct will produce a better immune response than BCG and can be taken up as a post-exposure therapeutic subunit vaccine along with standard TB therapy. We also anticipate that our construct can be taken up as a protein adjuvant with other vaccine candidates as these can activate macrophages through TLR signaling.
Collapse
Affiliation(s)
- Parul Bhatt
- DSKC BioDiscovery Lab, Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| | - Monika Sharma
- DSKC BioDiscovery Lab, Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Prem Prakash Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Sadhna Sharma
- DSKC BioDiscovery Lab, Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| |
Collapse
|
7
|
A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int Immunopharmacol 2022; 109:108791. [PMID: 35487086 DOI: 10.1016/j.intimp.2022.108791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem despite widespread use of the Bacillus BCG vaccine. This situation is worsened by co-infection with HIV, and the development of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains. Thus, novel vaccine candidates and improved vaccination strategies are urgently needed in order to reduce the incidence of TB and even to eradicate TB by 2050. Over the last few decades, 23 novel TB vaccines have entered into clinical trials, more than 13 new vaccines have reached various stages of preclinical development, and more than 50 potential candidates are in the discovery stage as next-generation vaccines. Nevertheless, why has a century of attempts to introduce an effective TB vaccine failed? Who should be blamed -scientists, human response, or Mtb strategies? Literature review reveals that the elimination of latent or active Mtb infections in a given population seems to be an epigenetic process. With a better understanding of the connections between bacterial infections and gene expression conditions in epigenetic events, opportunities arise in designing protective vaccines or therapeutic agents, particularly as epigenetic processes can be reversed. Therefore, this review provides a brief overview of different approaches towards novel vaccination strategies and the mechanisms underlying these approaches.
Collapse
|
8
|
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a leading cause of mortality and morbidity due to a single infectious agent. Aerosol infection with Mtb can result in a range of responses from elimination, active, incipient, subclinical, and latent Mtb infections (LTBI), depending on the host's immune response and the dose and nature of infecting bacilli. Currently, BCG is the only vaccine approved to prevent TB. Although BCG confers protection against severe forms of childhood TB, its use in adults and those with comorbid conditions, such as HIV infection, is questionable. Novel vaccines, including recombinant BCG (rBCG), were developed to improve BCG's efficacy and use as an alternative to BCG in a vulnerable population. The first-generation rBCG vaccines had different Mtb antigens and were tested as a prime, prime-boost, or immunotherapeutic intervention. The novel vaccines target one or more of the following requirements, namely prevention of infection (POI), prevention of disease (POD), prevention of recurrence (POR), and therapeutic vaccines to treat a TB disease. Several vaccine candidates currently in development are classified into four primary categories: live attenuated whole-cell vaccine, inactivated whole-cell vaccine, adjuvanted protein subunit vaccine, and viral-vectored vaccine. Each vaccine's immunogenicity, safety, and efficacy are tested in preclinical animal models and further validated through various phases of clinical trials. This chapter summarizes the various TB vaccine candidates under different clinical trial stages and promises better protection against TB.
Collapse
Affiliation(s)
- Radha Gopalaswamy
- Department of Bacteriology, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamilnadu, India
| | - Selvakumar Subbian
- The Public Health Research Institute Center at New Jersey Medical School, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
9
|
He Y, Yu W, Xiao L, Shen L, Qi J, Hu T. Conjugation of Zika virus EDIII with CRM 197, 8-arm PEG and mannan for development of an effective Zika virus vaccine. Int J Biol Macromol 2021; 190:713-721. [PMID: 34474053 DOI: 10.1016/j.ijbiomac.2021.08.177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/15/2023]
Abstract
Zika virus (ZIKV) induces neurological and autoimmune complications such as microcephaly and Guillain-Barre syndrome. Effective vaccines are necessary to prevent the ZIKV infection. E protein of ZIKV is responsible for virus attachment, entry, and fusion. The domain III of E protein (EDIII) contains the neutralizing epitopes and is ideal to act as an antigen for ZIKV vaccine. However, EDIII is poorly immunogenic. CRM197 is a carrier protein and can activate T helper cells for EDIII. Mannan is a ligand of TLR-4 or TLR-2. Eight-arm PEG can link multiple EDIII molecules in one entity. In the present study, EDIII was covalently conjugated with CRM197, 8-arm PEG and mannan to improve the immunogenicity of EDIII. The conjugate (CRM-EDIII-PM) elicited high EDIII-specific antibody titers in the BALB/c mice. Th1-type cytokines (IFN-γ and IL-2) and Th2-type cytokines (IL-5 and IL-10) were secreted at a marked level. Thus, CRM-EDIII-PM could stimulate potent humoral and cellular immune response to EDIII. The serum exposure of CRM-EDIII-PM to the immune system was prolonged. Moreover, CRM-EDIII-PM did not lead to apparent toxicity to the organs. Therefore, CRM-EDIII-PM was expected as a promising vaccine candidate for its ability to induce strong immune responses.
Collapse
Affiliation(s)
- Yunxia He
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lucheng Xiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
10
|
Ritzau-Jost J, Hutloff A. T Cell/B Cell Interactions in the Establishment of Protective Immunity. Vaccines (Basel) 2021; 9:vaccines9101074. [PMID: 34696182 PMCID: PMC8536969 DOI: 10.3390/vaccines9101074] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Follicular helper T cells (Tfh) are the T cell subset providing help to B cells for the generation of high-affinity antibodies and are therefore of key interest for the development of vaccination strategies against infectious diseases. In this review, we will discuss how the generation of Tfh cells and their interaction with B cells in secondary lymphoid organs can be optimized for therapeutic purposes. We will summarize different T cell subsets including Tfh-like peripheral helper T cells (Tph) capable of providing B cell help. In particular, we will highlight the novel concept of T cell/B cell interaction in non-lymphoid tissues as an important element for the generation of protective antibodies directly at the site of pathogen invasion.
Collapse
|
11
|
Cho T, Khatchadourian C, Nguyen H, Dara Y, Jung S, Venketaraman V. A review of the BCG vaccine and other approaches toward tuberculosis eradication. Hum Vaccin Immunother 2021; 17:2454-2470. [PMID: 33769193 PMCID: PMC8475575 DOI: 10.1080/21645515.2021.1885280] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/29/2021] [Indexed: 02/02/2023] Open
Abstract
Despite aggressive eradication efforts, Tuberculosis (TB) remains a global health burden, one that disproportionally affects poorer, less developed nations. The only vaccine approved for TB, the Bacillus of Calmette and Guérin (BCG) vaccine remains controversial because it's stated efficacy has been cited as anywhere from 0 to 80%. Nevertheless, there have been exciting discoveries about the mechanism of action of the BCG vaccine that suggests it has a role in immunization schedules today. We review recent data suggesting the vaccine imparts protection against both tuberculosis and non-tuberculosis pathogens via a newly discovered immune system called trained immunity. BCG's efficacy also appears to be tied to its affect on granulocytes at the epigenetic and hematopoietic stem cell levels, which we discuss in this article at length. We also write about how the different strains of the BCG vaccine elicit different immune responses, suggesting that certain BCG strains are more immunogenic than others. Finally, our review delves into how the current vaccine is being reformulated to be more efficacious, and track the development of the next generation vaccines against TB.
Collapse
Affiliation(s)
- Thomas Cho
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | | | - Huy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Yash Dara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Shuna Jung
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
12
|
Qi J, He Y, Shen L, Yu W, Hu T. Conjugation of Hemoglobin and Mannan Markedly Improves the Immunogenicity of Domain III of the Zika Virus E Protein: Structural and Immunological Study. Bioconjug Chem 2021; 32:328-338. [PMID: 33522239 DOI: 10.1021/acs.bioconjchem.0c00700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Zika virus (ZIKV) leads to congenital microcephaly and anomalies and severe neurological diseases such as Guillain-Barre syndrome. Safe and effective vaccines are necessitated to deal with these severe health threats. As an ideal antigen, the domain III of the envelope protein (EDIII) of ZIKV can evoke potent neutralizing antibodies without any antibody-dependent enhancement (ADE) effect. However, EDIII necessitates to be formulated with an antigen delivery system or adjuvants to improve its immunogenicity. Hemoglobin (Hb) regulates inflammation, cytokine levels, and activate macrophage. Mannan is a polysaccharide of the fungal cell wall with an immunomodulatory activity. In this study, EDIII was conjugated with Hb and mannan, using the disulfide bond as the linker. Hb and mannan both functioned as the adjuvants. Conjugation of Hb and mannan acted as the delivery system for EDIII. The structure of EDIII was essentially maintained upon conjugation of Hb and mannan. The intracellular release of EDIII from the conjugate (HM-EDIII-2) was achieved by reduction of the glutathione-sensitive disulfide bond. As compared with EDIII, HM-EDIII-2 elicited high EDIII-specific IgG titers and high levels of Th1-type cytokines (IFN-γ and IL-2) and Th2-type cytokines (IL-5 and IL-10), along with no apparent toxicity to the organs. Moreover, the pharmacokinetic study revealed a prolonged serum exposure of HM-EDIII-2 to the immune cells. Thus, HM-EDIII-2 could boost a strong humoral and cellular immune response to EDIII. Our study was expected to provide the feasibility necessary to develop a robust and potentially safe ZIKV vaccine.
Collapse
Affiliation(s)
- Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yunxia He
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
13
|
Ferluga J, Yasmin H, Bhakta S, Kishore U. Vaccination Strategies Against Mycobacterium tuberculosis: BCG and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:217-240. [PMID: 34661897 DOI: 10.1007/978-3-030-67452-6_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Tuberculosis (TB) is a highly contagious disease caused by Mycobacterium tuberculosis (Mtb) and is the major cause of morbidity and mortality across the globe. The clinical outcome of TB infection and susceptibility varies among individuals and even among different populations, contributed by host genetic factors such as polymorphism in the human leukocyte antigen (HLA) alleles as well as in cytokine genes, nutritional differences between populations, immunometabolism, and other environmental factors. Till now, BCG is the only vaccine available to prevent TB but the protection rendered by BCG against pulmonary TB is not uniform. To deliver a vaccine which can give consistent protection against TB is a great challenge with rising burden of drug-resistant TB. Thus, expectations are quite high with new generation vaccines that will improve the efficiency of BCG without showing any discordance for all forms of TB, effective for individual of all ages in all parts of the world. In order to enhance or improve the efficacy of BCG, different strategies are being implemented by considering the immunogenicity of various Mtb virulence factors as well as of the recombinant strains, co-administration with adjuvants and use of appropriate vehicle for delivery. This chapter discusses several such pre-clinical attempts to boost BCG with subunit vaccines tested in murine models and also highlights various recombinant TB vaccines undergoing clinical trials. Promising candidates include new generation of live recombinant BCG (rBCG) vaccines, VPM1002, which are deleted in one or two virulence genes. They encode for the mycobacteria-infected macrophage-inhibitor proteins of host macrophage apoptosis and autophagy, key events in killing and eradication of Mtb. These vaccines are rBCG- ΔureC::hly HMR, and rBCG-ΔureC::hly ΔnuoG. The former vaccine has passed phase IIb in clinical trials involving South African infants and adults. Thus, with an aim of elimination of TB by 2050, all these cumulative efforts to develop a better TB vaccine possibly is new hope for positive outcomes.
Collapse
Affiliation(s)
- Janez Ferluga
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Sanjib Bhakta
- Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
14
|
Qi J, Yin Y, Yu W, Shen L, Xu J, Hu T. Conjugation of β-Glucan with the Hydrazone and Disulfide Linkers Markedly Improves the Immunogenicity of Zika Virus E Protein. Mol Pharm 2020; 17:1933-1944. [DOI: 10.1021/acs.molpharmaceut.0c00010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Ying Yin
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
15
|
Garcia-Vilanova A, Chan J, Torrelles JB. Underestimated Manipulative Roles of Mycobacterium tuberculosis Cell Envelope Glycolipids During Infection. Front Immunol 2019; 10:2909. [PMID: 31921168 PMCID: PMC6930167 DOI: 10.3389/fimmu.2019.02909] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
The Mycobacterium tuberculosis cell envelope has been evolving over time to make the bacterium transmissible and adaptable to the human host. In this context, the M. tuberculosis cell envelope contains a peripheral barrier full of lipids, some of them unique, which confer M. tuberculosis with a unique shield against the different host environments that the bacterium will encounter at the different stages of infection. This lipid barrier is mainly composed of glycolipids that can be characterized by three different subsets: trehalose-containing, mannose-containing, and 6-deoxy-pyranose-containing glycolipids. In this review, we explore the roles of these cell envelope glycolipids in M. tuberculosis virulence and pathogenesis, drug resistance, and further, how these glycolipids may dictate the M. tuberculosis cell envelope evolution from ancient to modern strains. Finally, we address how these M. tuberculosis cell envelope glycolipids are impacted by the host lung alveolar environment, their role in vaccination and masking host immunity, and subsequently the impact of these glycolipids in shaping how M. tuberculosis interacts with host cells, manipulating their immune response to favor the establishment of an infection.
Collapse
Affiliation(s)
- Andreu Garcia-Vilanova
- Population Health Program, TB Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - John Chan
- Department of Medicine (Infectious Diseases), Albert Einstein College of Medicine & Montefiore Medical Center, Bronx, NY, United States.,Department of Microbiology and Immunology, Albert Einstein College of Medicine & Montefiore Medical Center, Bronx, NY, United States
| | - Jordi B Torrelles
- Population Health Program, TB Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
16
|
Vasina DV, Kleymenov DA, Manuylov VA, Mazunina EP, Koptev EY, Tukhovskaya EA, Murashev AN, Gintsburg AL, Gushchin VA, Tkachuk AP. First-In-Human Trials of GamTBvac, a Recombinant Subunit Tuberculosis Vaccine Candidate: Safety and Immunogenicity Assessment. Vaccines (Basel) 2019; 7:vaccines7040166. [PMID: 31683812 PMCID: PMC6963980 DOI: 10.3390/vaccines7040166] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis is known to be the biggest global health problem, causing the most deaths by a single infectious agent. Vaccine-development efforts are extremely important. This paper represents the results of the first-in-human trial of recombinant subunit tuberculosis vaccine GamTBvac in a Phase I study. GamTBvac is a new BCG booster candidate vaccine containing dextran-binding domain modified Ag85a and ESAT6-CFP10 MTB antigens and CpG ODN adjuvant, formulated with dextrans. Safety and immunogenicity of GamTBvac were estimated in an open-label clinical trial on 60 Mycobacterium tuberculosis uninfected (MTB-uninfected) volunteers previously-vaccinated with Bacillus Calmette—Guérin vaccine (BCG). The candidate vaccine had an acceptable safety profile and was well-tolerated. Three different vaccine doses with a double-immunization scheme were assessed for immunogenicity and induced a significant increase in IFN-γ in-house IGRA response and IgG ELISA analysis. Among them, the half dose vaccine group (containing DBD-ESAT6-CFP10, 12.5 μg; DBD-Ag85a, 12.5 μg; CpG (ODN 2216), 75 μg; DEAE-Dextran 500 kDa, 250 μg; and Dextran 500 kDa, 5 mg) provided high, early and stable in time immune response specific to both protein antigen fusions and is proposed for the further studies.
Collapse
Affiliation(s)
- Daria V Vasina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Denis A Kleymenov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Victor A Manuylov
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena P Mazunina
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Egor Yu Koptev
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| | - Elena A Tukhovskaya
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Arkady N Murashev
- Branch of Shemyakin and Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Alexander L Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Infectology Department, I. M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Vladimir A Gushchin
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
- Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Artem P Tkachuk
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia.
| |
Collapse
|
17
|
Kuczkowska K, Øverland L, Rocha SDC, Eijsink VGH, Mathiesen G. Comparison of eight Lactobacillus species for delivery of surface-displayed mycobacterial antigen. Vaccine 2019; 37:6371-6379. [PMID: 31526620 DOI: 10.1016/j.vaccine.2019.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
Abstract
Lactobacillus spp. comprise a large group of Gram-positive lactic acid bacteria with varying physiological, ecological and immunomodulatory properties that are widely exploited by mankind, primarily in food production and as health-promoting probiotics. Recent years have shown increased interest in using lactobacilli for delivery of vaccines, mainly due to their ability to skew the immune system towards pro-inflammatory responses. We have compared the potential of eight Lactobacillus species, L. plantarum, L. brevis, L. curvatus, L. rhamnosus, L. sakei, L. gasseri, L. acidophilus and L. reuteri, as immunogenic carriers of the Ag85B-ESAT-6 antigen from Mycobacterium tuberculosis. Surface-display of the antigen was achieved in L. plantarum, L. brevis, L. gasseri and L. reuteri and these strains were further analyzed in terms of their in vitro and in vivo immunogenicity. All strains activated human dendritic cells in vitro. Immunization of mice using a homologous prime-boost regimen comprising a primary subcutaneous immunization followed by three intranasal boosters, led to slightly elevated IgG levels in serum in most strains, and, importantly, to significantly increased levels of antigen-specific mucosal IgA. Cellular immunity was assessed by studying antigen-specific T cell responses in splenocytes, which did not reveal proliferation as assessed by the expression of Ki67, but which showed clear antigen-specific IFN-γ and IL-17 responses for some of the groups. Taken together, the present results indicate that L. plantarum and L. brevis are the most promising carriers of TB vaccines.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway.
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Sergio D C Rocha
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| |
Collapse
|
18
|
Wahid AA, Doekhie A, Sartbaeva A, van den Elsen JMH. Ensilication Improves the Thermal Stability of the Tuberculosis Antigen Ag85b and an Sbi-Ag85b Vaccine Conjugate. Sci Rep 2019; 9:11409. [PMID: 31391509 PMCID: PMC6685958 DOI: 10.1038/s41598-019-47657-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/16/2019] [Indexed: 02/03/2023] Open
Abstract
There is an urgent need for the development of vaccine thermostabilisation methodologies as the maintenance of a continuous and reliable cold chain remains a major hurdle to the global distribution of safe and effective vaccines. Ensilication, a method that encases proteins in a resistant silica cage has been shown to physically prevent the thermal denaturation of a number of model proteins. In this study we investigate the utility of this promising approach in improving the thermal stability of antigens and vaccine conjugates highly relevant to the development of candidate tuberculosis vaccines, including antigen 85b conjugated with the Staphylococcus aureus-protein based adjuvant Sbi. Here we analyse the sensitivity of these constructs to thermal denaturation and demonstrate for the first time the benefits of ensilication in conferring these vaccine-relevant proteins with protection against temperature-induced loss of structure and function without the need for refrigeration. Our results reveal the potential of ensilication in facilitating the storage and transport of vaccines at ambient temperatures in the future and therefore in delivering life-saving vaccines globally, and in particular to remote areas of developing countries where disease rates are often highest.
Collapse
Affiliation(s)
- A A Wahid
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - A Doekhie
- Department of Chemistry, University of Bath, Bath, UK
| | - A Sartbaeva
- Department of Chemistry, University of Bath, Bath, UK.
| | | |
Collapse
|
19
|
Tran AC, Kim MY, Reljic R. Emerging Themes for the Role of Antibodies in Tuberculosis. Immune Netw 2019; 19:e24. [PMID: 31501712 PMCID: PMC6722270 DOI: 10.4110/in.2019.19.e24] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
The best way to debunk a scientific dogma is to throw irrefutable evidence at it. This is especially true if the dogma in question has been nurtured over many decades, as is the case with the apparent redundancy of antibodies (Abs) against intracellular pathogens. Although not fully compelling yet, that ‘hard core’ evidence is nevertheless now slowly beginning to emerge. This is true for several clinically relevant infections but none more so than Mycobacterium tuberculosis, the archetype intracellular pathogen that poses a great health challenge to the mankind. Here, prompted by a spate of recent high-profile reports on the effects of Abs in various experimental models of tuberculosis, we step back and take a critical look at the progress that has been made in the last 5 years and highlight some of the strengths and shortcomings of the presented evidence. We conclude that the tide of the opinion has begun to turn in favour of Abs but we also caution against overinterpreting the currently available limited evidence. For, until definitive evidence that can withstand even the most rigorous of experimental tests is produced, the dogma may yet survive. Or indeed, we may find that the truth is hidden somewhere in between the dogma and the unfulfilled scientific prophecy.
Collapse
Affiliation(s)
- Andy C Tran
- St George's, University of London, London SW17 0RE, UK
| | - Mi-Young Kim
- St George's, University of London, London SW17 0RE, UK.,Department of Molecular Biology and The Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 54896, Korea
| | - Rajko Reljic
- St George's, University of London, London SW17 0RE, UK
| |
Collapse
|
20
|
Diogo GR, Hart P, Copland A, Kim MY, Tran AC, Poerio N, Singh M, Paul MJ, Fraziano M, Reljic R. Immunization With Mycobacterium tuberculosis Antigens Encapsulated in Phosphatidylserine Liposomes Improves Protection Afforded by BCG. Front Immunol 2019; 10:1349. [PMID: 31293568 PMCID: PMC6598733 DOI: 10.3389/fimmu.2019.01349] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/28/2019] [Indexed: 11/13/2022] Open
Abstract
Liposomes have been long considered as a vaccine delivery system but this technology remains to be fully utilized. Here, we describe a novel liposome-based subunit vaccine formulation for tuberculosis (TB) based on phosphatidylserine encapsulating two prominent TB antigens, Ag85B, and ESAT-6. We show that the resulting liposomes (Lipo-AE) are stable upon storage and can be readily taken up by antigen presenting cells and that their antigenic cargo is delivered and processed within endosomal cell compartments. The Lipo-AE vaccine formulation combined with the PolyIC adjuvant induced a mixed Th1/Th17-Th2 immune response to Ag85B but only a weak response to ESAT-6. An immunization regimen based on systemic delivery followed by mucosal boost with Lipo-AE resulted in the accumulation of resident memory T cells in the lungs. Most importantly though, when Lipo-AE vaccine candidate was administered to BCG-immunized mice subsequently challenged with low dose aerosol Mycobacterium tuberculosis, we observed a significant reduction of the bacterial load in the lungs and spleen compared to BCG alone. We therefore conclude that the immunization with mycobacterial antigens delivered by phosphatidylserine based liposomes in combination with Poly:IC adjuvant may represent a novel BCG boosting vaccination strategy.
Collapse
Affiliation(s)
- Gil R Diogo
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Peter Hart
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Alastair Copland
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Mi-Young Kim
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Andy C Tran
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Noemi Poerio
- Dipartimento di Biologia, University of Rome Tor Vergata, Rome, Italy
| | | | - Matthew J Paul
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Maurizio Fraziano
- Dipartimento di Biologia, University of Rome Tor Vergata, Rome, Italy
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| |
Collapse
|
21
|
Malik A, Gupta M, Mani R, Bhatnagar R. Single-dose Ag85B-ESAT6-loaded poly(lactic- co-glycolic acid) nanoparticles confer protective immunity against tuberculosis. Int J Nanomedicine 2019; 14:3129-3143. [PMID: 31118627 PMCID: PMC6501725 DOI: 10.2147/ijn.s172391] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Bacillus Calmette-Guérin, the attenuated strain of Mycobacterium bovis, remains the only available vaccine against tuberculosis (TB). However, its ineffectiveness in adults against pulmonary TB and varied protective efficacy (0-80%) speak to an urgent need for the development of an improved and efficient TB vaccine. In this milieu, poly(lactic-co-glycolic acid) (PLGA), is a preferential candidate, due to such properties as biocompatibility, targeted delivery, sustained antigen release, and atoxic by-products. METHODS In this study, we formulated PLGA nanoparticles (NPs) encapsulating the bivalent H1 antigen, a fusion of Mycobacterium tuberculosis (Mtb) Ag85B and ESAT6 proteins, and investigated its role in immunomodulation and protection against Mtb challenge. Using the classical water-oil-water solvent-evaporation method, H1-NPs were prepared, with encapsulation efficiency of 86.1%±3.2%. These spherical NPs were ~244.4±32.6 nm in diameter, with a negatively charged surface (ζ-potential -4±0.6 mV). RESULTS Under physiological conditions, NPs degraded slowly and the encapsulated H1 antigen was released over a period of weeks. As a proof-of-concept vaccine candidate, H1 NPs were efficiently internalized by the THP-1 human macrophages. Six weeks after a single-dose vaccination, H1 NP-immunized C57BL/6J mice showed significant increase in the production of total serum IgG (P<0.0001) and its isotypes compared to H1 alone, IgG2a being the predominant one, followed by IgG1. Further, the cytokine-release profile of antigen-stimulated splenocyteculture supernatant indicated a strong TH1-biased immunoresponse in H1 NP-vaccinated mice, with ~6.03- and ~2.8-fold increase in IFNγ and TNFα cytokine levels, and ~twofold and 1.6 fold increase in IL4 and IL10 cytokines, respectively, compared to H1 alone-immunized mice. In protection studies, H1 NP-vaccinated mice displayed significant reductions in lung and spleen bacillary load (P<0.05) at 5-week post-Mtb H37Rv challenge and prolonged survival, with a mean survival time of 177 days, compared to H1 alone-vaccinated mice (mean survival time 80 days). CONCLUSION Altogether, our findings highlight the significance of the H1-PLGA nanoformulation in terms of providing long-term protection in mice with a single dose.
Collapse
Affiliation(s)
- Anshu Malik
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| | - Manish Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| | - Rajesh Mani
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India,
| |
Collapse
|
22
|
Sarmiento ME, Alvarez N, Chin KL, Bigi F, Tirado Y, García MA, Anis FZ, Norazmi MN, Acosta A. Tuberculosis vaccine candidates based on mycobacterial cell envelope components. Tuberculosis (Edinb) 2019; 115:26-41. [PMID: 30948174 DOI: 10.1016/j.tube.2019.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/12/2019] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
Even after decades searching for a new and more effective vaccine against tuberculosis, the scientific community is still pursuing this goal due to the complexity of its causative agent, Mycobacterium tuberculosis (Mtb). Mtb is a microorganism with a robust variety of survival mechanisms that allow it to remain in the host for years. The structure and nature of the Mtb envelope play a leading role in its resistance and survival. Mtb has a perfect machinery that allows it to modulate the immune response in its favor and to adapt to the host's environmental conditions in order to remain alive until the moment to reactivate its normal growing state. Mtb cell envelope protein, carbohydrate and lipid components have been the subject of interest for developing new vaccines because most of them are responsible for the pathogenicity and virulence of the bacteria. Many indirect evidences, mainly derived from the use of monoclonal antibodies, support the potential protective role of Mtb envelope components. Subunit and DNA vaccines, lipid extracts, liposomes and membrane vesicle formulations are some examples of technologies used, with encouraging results, to evaluate the potential of these antigens in the protective response against Mtb.
Collapse
Affiliation(s)
- M E Sarmiento
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia
| | - N Alvarez
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, USA
| | - K L Chin
- Department of Biomedical Sciences and Therapeutic, Faculty of Medicine and Health Sciences (FPSK), Universiti Malaysia Sabah (UMS), Sabah, Malaysia
| | - F Bigi
- Institute of Biotechnology, INTA, Buenos Aires, Argentina
| | - Y Tirado
- Finlay Institute of Vaccines, La Habana, Cuba
| | - M A García
- Finlay Institute of Vaccines, La Habana, Cuba
| | - F Z Anis
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia
| | - M N Norazmi
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia.
| | - A Acosta
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
23
|
Bekale RB, Du Plessis SM, Hsu NJ, Sharma JR, Sampson SL, Jacobs M, Meyer M, Morse GD, Dube A. Mycobacterium Tuberculosis and Interactions with the Host Immune System: Opportunities for Nanoparticle Based Immunotherapeutics and Vaccines. Pharm Res 2018; 36:8. [PMID: 30411187 PMCID: PMC6362825 DOI: 10.1007/s11095-018-2528-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023]
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis remains a deadly infectious disease. The thin pipeline of new drugs for TB, the ineffectiveness in adults of the only vaccine available, i.e. the Bacillus Calmette-Guerin vaccine, and increasing global antimicrobial resistance, has reinvigorated interest in immunotherapies. Nanoparticles (NPs) potentiate the effect of immune modulating compounds (IMC), enabling cell targeting, improved transfection of antigens, enhanced compound stability and provide opportunities for synergistic action, via delivery of multiple IMCs. In this review we describe work performed in the application of NPs towards achieving immune modulation for TB treatment and vaccination. Firstly, we present a comprehensive review of M. tuberculosis and how the bacterium modulates the host immune system. We find that current work suggest great promise of NP based immunotherapeutics as novel treatments and vaccination systems. There is need to intensify research efforts in this field, and rationally design novel NP immunotherapeutics based on current knowledge of the mycobacteriology and immune escape mechanisms employed by M. tuberculosis.
Collapse
Affiliation(s)
- Raymonde B Bekale
- Discipline of Pharmaceutics, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| | - Su-Mari Du Plessis
- NRF-DST Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nai-Jen Hsu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jyoti R Sharma
- National Health Laboratory Service, Johannesburg, South Africa
| | - Samantha L Sampson
- NRF-DST Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Johannesburg, South Africa
- Immunology of Infectious Disease Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Mervin Meyer
- DST/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Unit, Department of Biotechnology, University of the Western Cape (UWC), Cape Town, South Africa
| | - Gene D Morse
- AIDS Clinical Trials Group Pharmacology Specialty Laboratory, New York State Center of Excellence in Bioinformatics and Life Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Admire Dube
- Discipline of Pharmaceutics, School of Pharmacy, University of the Western Cape, Cape Town, South Africa.
| |
Collapse
|
24
|
Méndez-Samperio P. Development of tuberculosis vaccines in clinical trials: Current status. Scand J Immunol 2018; 88:e12710. [PMID: 30175850 DOI: 10.1111/sji.12710] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023]
Abstract
Tuberculosis (TB) is an important infectious disease worldwide. Currently, Bacillus Calmette-Guérin (BCG) remains the only TB vaccine licensed for human use. This TB vaccine is effective in protecting children against severe military TB but offers variable protective efficacy in adults. Therefore, new vaccines against TB are needed to overcome this serious disease. At present, around 14 TB vaccine candidates are in different phases of clinical trials. These TB vaccines in clinical evaluation can be classified into two groups including preventive pre- and post-exposure vaccines: subunit vaccines (attenuated viral vectors or adjuvanted fusion proteins), and whole-cell vaccines (genetically attenuated Mycobacterium tuberculosis (M. tb), recombinant BCG, killed M. tb or M. vaccae). Although, over the last two decades a great progress in the search for a more effective TB vaccine has been demonstrated there is still no replacement for the licensed BCG vaccine. This article summarizes the current status of TB vaccine development and identifies crucial gaps of research for the development of an effective TB vaccine in all age groups.
Collapse
|
25
|
Rhodes SJ, Guedj J, Fletcher HA, Lindenstrøm T, Scriba TJ, Evans TG, Knight GM, White RG. Using vaccine Immunostimulation/Immunodynamic modelling methods to inform vaccine dose decision-making. NPJ Vaccines 2018; 3:36. [PMID: 30245860 PMCID: PMC6141590 DOI: 10.1038/s41541-018-0075-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 06/30/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022] Open
Abstract
Unlike drug dose optimisation, mathematical modelling has not been applied to vaccine dose finding. We applied a novel Immunostimulation/Immunodynamic mathematical modelling framework to translate multi-dose TB vaccine immune responses from mice, to predict most immunogenic dose in humans. Data were previously collected on IFN-γ secreting CD4+ T cells over time for novel TB vaccines H56 and H1 adjuvanted with IC31 in mice (1 dose groups (0.1-1.5 and 15 μg H56 + IC31), 45 mice) and humans (1 dose (50 μg H56/H1 + IC31), 18 humans). A two-compartment mathematical model, describing the dynamics of the post-vaccination IFN-γ T cell response, was fitted to mouse and human data, separately, using nonlinear mixed effects methods. We used these fitted models and a vaccine dose allometric scaling assumption, to predict the most immunogenic human dose. Based on the changes in model parameters by mouse H56 + IC31 dose and by varying the H56 dose allometric scaling factor between mouse and humans, we established that, at a late time point (224 days) doses of 0.8-8 μg H56 + IC31 in humans may be the most immunogenic. A 0.8-8 μg of H-series TB vaccines in humans, may be as, or more, immunogenic, as larger doses. The Immunostimulation/Immunodynamic mathematical modelling framework is a novel, and potentially revolutionary tool, to predict most immunogenic vaccine doses, and accelerate vaccine development.
Collapse
Affiliation(s)
- Sophie J. Rhodes
- TB Modelling Group, CMMID, TB Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Jeremie Guedj
- IAME, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France
| | - Helen A. Fletcher
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | | - Gwenan M. Knight
- TB Modelling Group, CMMID, TB Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Richard G. White
- TB Modelling Group, CMMID, TB Centre, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
26
|
Coppola M, van den Eeden SJF, Robbins N, Wilson L, Franken KLMC, Adams LB, Gillis TP, Ottenhoff THM, Geluk A. Vaccines for Leprosy and Tuberculosis: Opportunities for Shared Research, Development, and Application. Front Immunol 2018. [PMID: 29535713 PMCID: PMC5834475 DOI: 10.3389/fimmu.2018.00308] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis (TB) and leprosy still represent significant public health challenges, especially in low- and lower middle-income countries. Both poverty-related mycobacterial diseases require better tools to improve disease control. For leprosy, there has been an increased emphasis on developing tools for improved detection of infection and early diagnosis of disease. For TB, there has been a similar emphasis on such diagnostic tests, while increased research efforts have also focused on the development of new vaccines. Bacille Calmette–Guérin (BCG), the only available TB vaccine, provides insufficient and inconsistent protection to pulmonary TB in adults. The impact of BCG on leprosy, however, is significant, and the introduction of new TB vaccines that might replace BCG could, therefore, have serious impact also on leprosy. Given the similarities in antigenic makeup between the pathogens Mycobacterium tuberculosis (Mtb) and M. leprae, it is well possible, however, that new TB vaccines could cross-protect against leprosy. New TB subunit vaccines currently evaluated in human phase I and II studies indeed often contain antigens with homologs in M. leprae. In this review, we discuss pre-clinical studies and clinical trials of subunit or whole mycobacterial vaccines for TB and leprosy and reflect on the development of vaccines that could provide protection against both diseases. Furthermore, we provide the first preclinical evidence of such cross-protection by Mtb antigen 85B (Ag85B)-early secretory antigenic target (ESAT6) fusion recombinant proteins in in vivo mouse models of Mtb and M. leprae infection. We propose that preclinical integration and harmonization of TB and leprosy research should be considered and included in global strategies with respect to cross-protective vaccine research and development.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Naoko Robbins
- The National Hansen's Disease Programs, Baton Rouge, LA, United States
| | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Linda B Adams
- The National Hansen's Disease Programs, Baton Rouge, LA, United States
| | - Tom P Gillis
- The National Hansen's Disease Programs, Baton Rouge, LA, United States
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
27
|
Hussein J, Zewdie M, Yamuah L, Bedru A, Abebe M, Dagnew AF, Chanyalew M, Yohannes AG, Ahmed J, Engers H, Doherty TM, Bang P, Kromann I, Hoff ST, Aseffa A. A phase I, open-label trial on the safety and immunogenicity of the adjuvanted tuberculosis subunit vaccine H1/IC31® in people living in a TB-endemic area. Trials 2018; 19:24. [PMID: 29321075 PMCID: PMC5764015 DOI: 10.1186/s13063-017-2354-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 11/27/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND H1/IC31® is a tuberculosis (TB) subunit vaccine candidate consisting of the fusion protein of Ag85B and ESAT-6 (H1) formulated with the IC31® adjuvant. Previous trials have reported on the H1/IC31® vaccine in M. tuberculosis (Mtb)-naïve, BCG-vaccinated and previously Mtb-infected individuals. In this trial, conducted between December 2008 and April 2010, the safety and immunogenicity of H1/IC31® was assessed in participants living in Ethiopia - a highly TB-endemic area. METHODS Healthy male participants aged 18-25 years were recruited into four groups. Participants in group 1 (N = 12) and group 2 (N = 12) were Tuberculin Skin Test (TST) negative and QuantiFERON-TB Gold in-tube test (QFT) negative (Mtb-naïve groups), participants in group 3 (N = 3) were TST positive and QFT negative (BCG group), and participants in group 4 (N = 12) were both TST and QFT positive (Mtb-infected group). H1 vaccine alone (group 1) or H1 formulated with the adjuvant IC31® (groups 2, 3 and 4) was administered intramuscularly on day 0 and day 56. Safety and immunogenicity parameters were evaluated for up to 32 weeks after day 0. RESULTS The H1/IC31®vaccine was safe and generally well tolerated. There was little difference among the four groups, with a tendency towards a higher incidence of adverse events in Mtb-infected compared to Mtb-naïve participants. Two serious adverse events were reported in the Mtb-infected group where a relationship to the vaccine could not be excluded. In both cases the participants recovered without sequelae within 72 h. Immunogenicity assays, evaluated in the 29 participants who received both vaccinations, showed a stronger response to TB antigens in the Mtb-naïve group vaccinated with the adjuvant. CONCLUSION The trial confirmed the need for an adjuvant for the vaccine to be immunogenic and highlighted the importance of early phase testing of a novel TB vaccine candidate in TB-endemic areas. TRIAL REGISTRATION ClinicalTrials.gov, ID: NCT01049282. Retrospectively registered on 14 January 2010.
Collapse
Affiliation(s)
- Jemal Hussein
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - Martha Zewdie
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia.
| | - Lawrence Yamuah
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - Ahmed Bedru
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia.,KNCV Tuberculosis foundation, Challenge TB project, Addis Ababa, Ethiopia
| | - Markos Abebe
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - Alemnew F Dagnew
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia.,GlaxoSmithKline Vaccines, Rockville, MD, USA
| | - Menberework Chanyalew
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - Asfawesen G Yohannes
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - Jemal Ahmed
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - Howard Engers
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| | - T Mark Doherty
- Statens Serum Institut (SSI), Artillerivej 5, 2300, Copenhagen, Denmark.,GlaxoSmithKline Vaccines, Wavre, Belgium
| | - Peter Bang
- Statens Serum Institut (SSI), Artillerivej 5, 2300, Copenhagen, Denmark
| | - Ingrid Kromann
- Statens Serum Institut (SSI), Artillerivej 5, 2300, Copenhagen, Denmark
| | - Søren T Hoff
- Statens Serum Institut (SSI), Artillerivej 5, 2300, Copenhagen, Denmark.,Present address: Novo Nordisk, Copenhagen, Denmark
| | - Abraham Aseffa
- Armauer Hansen Research Institute (AHRI), Jimma Road, PO Box 1005, Addis Ababa, Ethiopia
| |
Collapse
|
28
|
Lang R, Schick J. Review: Impact of Helminth Infection on Antimycobacterial Immunity-A Focus on the Macrophage. Front Immunol 2017; 8:1864. [PMID: 29312343 PMCID: PMC5743664 DOI: 10.3389/fimmu.2017.01864] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/08/2017] [Indexed: 12/16/2022] Open
Abstract
Successful immune control of Mycobacterium tuberculosis (MTB) requires robust CD4+ T cell responses, with IFNγs as the key cytokine promoting killing of intracellular mycobacteria by macrophages. By contrast, helminth infections typically direct the immune system toward a type 2 response, characterized by high levels of the cytokines IL-4 and IL-10, which can antagonize IFNγ production and its biological effects. In many countries with high burden of tuberculosis, helminth infections are endemic and have been associated with increased risk to develop tuberculosis or to inhibit vaccination-induced immunity. Mechanistically, regulation of the antimycobacterial immune response by helminths has been mostly been attributed to the T cell compartment. Here, we review the current status of the literature on the impact of helminths on vaccine-induced and natural immunity to MTB with a focus on the alterations enforced on the capacity of macrophages to function as sensors of mycobacteria and effector cells to control their replication.
Collapse
Affiliation(s)
- Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Judith Schick
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
29
|
Shala-Lawrence A, Beheshti S, Newman E, Tang M, Krylova SM, Leach M, Carpick B, Krylov SN. High-precision quantitation of a tuberculosis vaccine antigen with capillary-gel electrophoresis using an injection standard. Talanta 2017; 175:273-279. [DOI: 10.1016/j.talanta.2017.07.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 10/19/2022]
|
30
|
Karbalaei Zadeh Babaki M, Soleimanpour S, Rezaee SA. Antigen 85 complex as a powerful Mycobacterium tuberculosis immunogene: Biology, immune-pathogenicity, applications in diagnosis, and vaccine design. Microb Pathog 2017; 112:20-29. [PMID: 28942172 DOI: 10.1016/j.micpath.2017.08.040] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 01/24/2023]
Abstract
Mycobacterium tuberculosis (Mtb) is one of the most life-threatening mycobacterial species which is increasing the death rate due to emerging multi-drug resistant (MDR) strains. Concerned health authorities worldwide are interested in developing an effective vaccine to prevent the spread of Mtb. After years of research, including successful identification of many Mtb immunogenic molecules, effective therapeutic agents or a vaccine have yet to be found. However, among the identified Mtb immunogenes, antigen 85 (Ag85) complex (Ag85A, Ag85B, and Ag85C) is receiving attention from scientists as it allows bacteria to evade the host immune response by preventing formation of phagolysosomes for eradication of infection. Due to their importance, A85 molecules are being utilized as tools in diagnostic methods and in the construction of new vaccines, such as recombinant attenuated vaccines, DNA vaccines, and subunit vaccines. This paper represents a comprehensive review of studies on Mtb molecules examining pathogenicity, biochemistry, immunology, and the role of Mtb in therapeutic or vaccine research.
Collapse
Affiliation(s)
- Mohsen Karbalaei Zadeh Babaki
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Inflammation and Inflammatory Diseases Division, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Tkachuk AP, Gushchin VA, Potapov VD, Demidenko AV, Lunin VG, Gintsburg AL. Multi-subunit BCG booster vaccine GamTBvac: Assessment of immunogenicity and protective efficacy in murine and guinea pig TB models. PLoS One 2017; 12:e0176784. [PMID: 28453555 PMCID: PMC5409163 DOI: 10.1371/journal.pone.0176784] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/17/2017] [Indexed: 11/18/2022] Open
Abstract
New innovative vaccines are highly needed to combat the global threat posed by tuberculosis. Efficient components-antigens and adjuvants-are crucial for development of modern recombinant TB vaccines. This study describes a new vaccine (GamTBvac) consisting of two mycobacterial antigen fusions (Ag85A and ESAT6-CFP10)-with dextran-binding domain immobilized on dextran and mixed with an adjuvant consisting of DEAE-dextran core, and with CpG oligodeoxynucleotides (TLR9 agonists). GamTBvac and its components were assessed for immunogenicity and protective efficacy in GamTBvac-prime/boost and BCG-prime/ GamTBvac-boost in murine and guinea pig TB models. Results show that in both infectious models, GamTBvac has a strong immunogenicity and significant protective effect against Mycobacterium tuberculosis strain H37Rv under aerosol and intravenous challenges. GamTBvac showed a particularly strong protective effect as a BCG booster vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intravenous
- Aerosols
- Animals
- Antibodies, Bacterial/blood
- BCG Vaccine/immunology
- Cell Proliferation/physiology
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Female
- Guinea Pigs
- Immunization
- Immunization, Secondary
- Immunogenicity, Vaccine
- Lung/immunology
- Lymph Nodes/immunology
- Male
- Mice, Inbred C57BL
- Mycobacterium tuberculosis/immunology
- Spleen/immunology
- T-Lymphocytes/immunology
- Tuberculosis/immunology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- A. P. Tkachuk
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. A. Gushchin
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - V. D. Potapov
- State Research Center for Applied Microbiology and Biotechnology, Obolensk, Russia
| | - A. V. Demidenko
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. G. Lunin
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - A. L. Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
32
|
Novel vaccine potential of Rv3131, a DosR regulon-encoded putative nitroreductase, against hyper-virulent Mycobacterium tuberculosis strain K. Sci Rep 2017; 7:44151. [PMID: 28272457 PMCID: PMC5341159 DOI: 10.1038/srep44151] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/03/2017] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence indicates that latency-associated Mycobacterium tuberculosis (Mtb)-specific antigens from the dormancy survival regulator regulon (DosR) may be promising novel vaccine target antigens for the development of an improved tuberculosis vaccine. After transcriptional profiling of DosR-related genes in the hyper-virulent Beijing Mtb strain K and the reference Mtb strain H37Rv, we selected Rv3131, a hypothetical nitroreductase, as a vaccine antigen and evaluated its vaccine efficacy against Mtb K. Mtb K exhibited stable and constitutive up-regulation of rv3131 relative to Mtb H37Rv under three different growth conditions (at least 2-fold induction) including exponential growth in normal culture conditions, hypoxia, and inside macrophages. Mice immunised with Rv3131 formulated in GLA-SE, a well-defined TLR4 adjuvant, displayed enhanced Rv3131-specific IFN-γ and serum IgG2c responses along with effector/memory T cell expansion and remarkable generation of Rv3131-specific multifunctional CD4+ T cells co-producing TNF-α, IFN-γ and IL-2 in both spleen and lung. Following challenge with Mtb K, the Rv3131/GLA-SE-immunised group exhibited a significant reduction in bacterial number and less extensive lung inflammation accompanied by the obvious persistence of Rv3131-specific multifunctional CD4+ T cells. These results suggest that Rv3131 could be an excellent candidate for potential use in a multi-antigenic Mtb subunit vaccine, especially against Mtb Beijing strains.
Collapse
|
33
|
Norrby M, Vesikari T, Lindqvist L, Maeurer M, Ahmed R, Mahdavifar S, Bennett S, McClain JB, Shepherd BM, Li D, Hokey DA, Kromann I, Hoff ST, Andersen P, de Visser AW, Joosten SA, Ottenhoff THM, Andersson J, Brighenti S. Safety and immunogenicity of the novel H4:IC31 tuberculosis vaccine candidate in BCG-vaccinated adults: Two phase I dose escalation trials. Vaccine 2017; 35:1652-1661. [PMID: 28216183 DOI: 10.1016/j.vaccine.2017.01.055] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 12/28/2016] [Accepted: 01/20/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Novel vaccine strategies are required to provide protective immunity in tuberculosis (TB) and prevent development of active disease. We investigated the safety and immunogenicity of a novel TB vaccine candidate, H4:IC31 (AERAS-404) that is composed of a fusion protein of M. tuberculosis antigens Ag85B and TB10.4 combined with an IC31® adjuvant. METHODS BCG-vaccinated healthy subjects were immunized with various antigen (5, 15, 50, 150μg) and adjuvant (0, 100, 500nmol) doses of the H4:IC31 vaccine (n=106) or placebo (n=18) in two randomized, double-blind, placebo-controlled phase I studies conducted in a low TB endemic setting in Sweden and Finland. The subjects were followed for adverse events and CD4+ T cell responses. RESULTS H4:IC31 vaccination was well tolerated with a safety profile consisting of mostly mild to moderate self-limited injection site pain, myalgia, arthralgia, fever and post-vaccination inflammatory reaction at the screening tuberculin skin test injection site. The H4:IC31 vaccine elicited antigen-specific CD4+ T cell proliferation and cytokine production that persisted 18weeks after the last vaccination. CD4+ T cell expansion, IFN-γ production and multifunctional CD4+ Th1 responses were most prominent after two doses of H4:IC31 containing 5, 15, or 50μg of H4 in combination with the 500nmol IC31 adjuvant dose. CONCLUSIONS The novel TB vaccine candidate, H4:IC31, demonstrated an acceptable safety profile and was immunogenic, capable of triggering multifunctional CD4+ T cell responses in previously BCG-vaccinated healthy individuals. These dose-escalation trials provided evidence that the optimal antigen-adjuvant dose combinations are 5, 15, or 50μg of H4 and 500nmol of IC31. TRIAL REGISTRATION ClinicalTrials.gov, NCT02066428 and NCT02074956.
Collapse
Affiliation(s)
- Maria Norrby
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Timo Vesikari
- Vaccine Research Center, University of Tampere, Tampere, Finland
| | - Lars Lindqvist
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Markus Maeurer
- TIM, Department of Laboratory Medicine and CAST, Karolinska Institutet, Stockholm, Sweden
| | - Raija Ahmed
- TIM, Department of Laboratory Medicine and CAST, Karolinska Institutet, Stockholm, Sweden
| | - Shahnaz Mahdavifar
- TIM, Department of Laboratory Medicine and CAST, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | - Adriëtte W de Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Andersson
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Center for Infectious Medicine (CIM), Karolinska Institutet, Stockholm, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
Méndez-Samperio P. Global Efforts in the Development of Vaccines for Tuberculosis: Requirements for Improved Vaccines Against Mycobacterium tuberculosis. Scand J Immunol 2017; 84:204-10. [PMID: 27454335 DOI: 10.1111/sji.12465] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 07/13/2016] [Indexed: 11/28/2022]
Abstract
Currently, more than 9.0 million people develop acute pulmonary tuberculosis (TB) each year and about 1.5 million people worldwide die from this infection. Thus, developing vaccines to prevent active TB disease remains a priority. This article discusses recent progress in the development of new vaccines against TB and focusses on the main requirements for development of improved vaccines against Mycobacterium tuberculosis (M. tb). Over the last two decades, significant progress has been made in TB vaccine development, and some TB vaccine candidates have currently completed a phase III clinical trial. The potential public health benefits of these vaccines are possible, but it will need much more effort, including new global governance investment on this research. This investment would certainly be less than the annual global financial toll of TB treatment.
Collapse
Affiliation(s)
- P Méndez-Samperio
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN, CD México, México.
| |
Collapse
|
35
|
Triccas JA, Counoupas C. Novel vaccination approaches to prevent tuberculosis in children. Pneumonia (Nathan) 2016; 8:18. [PMID: 28702297 PMCID: PMC5471729 DOI: 10.1186/s41479-016-0020-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
Pediatric tuberculosis (TB) is an underappreciated problem and accounts for 10 % of all TB deaths worldwide. Children are highly susceptible to infection with Mycobacterium tuberculosis and interrupting TB spread would require the development of effective strategies to control TB transmission in pediatric populations. The current vaccine for TB, M. bovis Bacille Calmette-Guérin (BCG), can afford some level of protection against TB meningitis and severe forms of disseminated TB in children; however, its efficacy against pulmonary TB is variable and the vaccine does not afford life-long protective immunity. For these reasons there is considerable interest in the development of new vaccines to control TB in children. Multiple vaccine strategies are being assessed and include recombinant forms of the existing BCG vaccine, protein or viral candidates designed to boost BCG-induced immunity, or live attenuated forms of M. tuberculosis. A number of these candidates have entered clinical trials; however, no vaccine has shown improved protective efficacy compared to BCG in humans. The current challenge is to identify the most suitable candidates to progress from early to late stage clinical trials, in order to deliver a vaccine that can control and hopefully eliminate the global threat of TB.
Collapse
Affiliation(s)
- James A Triccas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Level 5, Charles Perkins Centre D17, Sydney, NSW 2006 Australia.,Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW Australia.,Sydney Medical School and the Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW Australia
| | - Claudio Counoupas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Level 5, Charles Perkins Centre D17, Sydney, NSW 2006 Australia.,Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW Australia
| |
Collapse
|
36
|
H1:IC31 vaccination is safe and induces long-lived TNF-α +IL-2 +CD4 T cell responses in M. tuberculosis infected and uninfected adolescents: A randomized trial. Vaccine 2016; 35:132-141. [PMID: 27866772 DOI: 10.1016/j.vaccine.2016.11.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/20/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Control of the tuberculosis epidemic requires a novel vaccine that is effective in preventing tuberculosis in adolescents, a key target population for vaccination against TB. METHODS Healthy adolescents, stratified by M. tuberculosis-infection status, were enrolled into this observer-blinded phase II clinical trial of the protein-subunit vaccine candidate, H1:IC31, comprising a fusion protein (H1) of Ag85B and ESAT-6, formulated with the IC31 adjuvant. Local and systemic adverse events and induced T cell responses were measured after one or two administrations of either 15μg or 50μg of the H1 protein. RESULTS Two hundred and forty participants were recruited and followed up for 224days. No notable safety events were observed regardless of H1 dose or vaccination schedule. H1:IC31 vaccination induced antigen-specific CD4 T cells, co-expressing IFN-γ, TNF-α and/or IL-2. H1:IC31 vaccination of M.tb-uninfected individuals preferentially drove the emergence of Ag85B and ESAT-6 specific TNF-α+IL-2+CD4 T cells, while H1:IC31 vaccination of M.tb-infected individuals resulted in the expansion of Ag85B-specific but not ESAT-6-specific TNF-α+IL-2+CD4 T cells. CONCLUSIONS H1:IC31 was safe and immunogenic in uninfected and M.tb-infected adolescents. Two administrations of the 15μg H1:IC31 dose induced the greatest magnitude immune response, and was considered optimal (South African National Clinical Trials Register, DoH-27-0612-3947; Pan African Clinical Trial Registry, PACTR201403000464306).
Collapse
|
37
|
Yu W, Hu T. Conjugation with an Inulin–Chitosan Adjuvant Markedly Improves the Immunogenicity of Mycobacterium tuberculosis CFP10-TB10.4 Fusion Protein. Mol Pharm 2016; 13:3626-3635. [DOI: 10.1021/acs.molpharmaceut.6b00138] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Weili Yu
- State
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Tao Hu
- State
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
38
|
Counoupas C, Pinto R, Nagalingam G, Hill-Cawthorne GA, Feng CG, Britton WJ, Triccas JA. Mycobacterium tuberculosis components expressed during chronic infection of the lung contribute to long-term control of pulmonary tuberculosis in mice. NPJ Vaccines 2016; 1:16012. [PMID: 29263854 PMCID: PMC5707878 DOI: 10.1038/npjvaccines.2016.12] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/27/2016] [Accepted: 07/05/2016] [Indexed: 11/09/2022] Open
Abstract
Tuberculosis (TB) remains a major cause of mortality and morbidity worldwide, yet current control strategies, including the existing BCG vaccine, have had little impact on disease control. The tubercle bacillus modifies protein expression to adapt to chronic infection of the host, and this can potentially be exploited to develop novel therapeutics. We identified the gene encoding the first step of the Mycobacterium tuberculosis sulphur assimilation pathway, cysD, as highly induced during chronic infection in the mouse lung, suggesting therapies based on CysD could be used to target infection. Vaccination with the composite vaccine CysVac2, a fusion of CysD and the immunogenic Ag85B of M. tuberculosis, resulted in the generation of multifunctional CD4+ T cells (interferon (IFN)-γ+TNF+IL-2+IL-17+) in the lung both pre- and post-aerosol challenge with M. tuberculosis. CysVac2 conferred significant protection against pulmonary M. tuberculosis challenge and was particularly effective at controlling late-stage infection, a property not shared by BCG. CysVac2 delivered as a booster following BCG vaccination afforded greater protection against M. tuberculosis challenge than BCG alone. The antigenic components of CysVac2 were conserved amongst M. tuberculosis strains, and protective efficacy afforded by CysVac2 was observed across varying murine MHC haplotypes. Strikingly, administration of CysVac2 to mice previously infected with M. tuberculosis reduced bacterial load and immunopathology in the lung compared with BCG-vaccinated mice. These results indicate that CysVac2 warrants further investigation to assess its potential to control pulmonary TB in humans.
Collapse
Affiliation(s)
- Claudio Counoupas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW, Australia
| | - Rachel Pinto
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW, Australia
| | - Gayathri Nagalingam
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW, Australia
| | - Grant A Hill-Cawthorne
- Sydney Medical School and The Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
- School of Public Health, University of Sydney, Sydney, NSW, Australia
| | - Carl G Feng
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW, Australia
- Sydney Medical School and The Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Warwick J Britton
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW, Australia
- Sydney Medical School and The Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
| | - James A Triccas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Tuberculosis Research Program, Centenary Institute, University of Sydney, Sydney, NSW, Australia
- Sydney Medical School and The Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
39
|
Méndez-Samperio P. Research progress in the field of immunotherapeutic vaccination in human TB and the way ahead. Immunotherapy 2016; 8:987-9. [DOI: 10.2217/imt-2016-0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Patricia Méndez-Samperio
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN. Prol. Carpio y Plan de Ayala, México, D.F. 11340 México
| |
Collapse
|
40
|
Cafaro A, Piccaro G, Altavilla G, Gigantino V, Matarese G, Olivieri E, Ferrantelli F, Ensoli B, Palma C. HIV-1 Tat protein vaccination in mice infected with Mycobacterium tuberculosis is safe, immunogenic and reduces bacterial lung pathology. BMC Infect Dis 2016; 16:442. [PMID: 27549342 PMCID: PMC4994248 DOI: 10.1186/s12879-016-1724-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 07/19/2016] [Indexed: 01/01/2023] Open
Abstract
Background The therapeutic HIV-1 Tat protein vaccine is in advanced clinical development. Tuberculosis, the main AIDS co-infection, is highly endemic in areas where AIDS prevention through vaccination is needed. However, safety and immunogenicity of Tat vaccination in the course of Mycobacterium tuberculosis (Mtb) infection is still unknown and it prevents the possibility to administer the vaccine to Mtb-infected individuals. We addressed the interplay and effects of Tat vaccination on Mtb infection in immunocompetent mice. Methods C57BL/6 mice were vaccinated or not with Bacillus Calmette-Guerin (BCG), the current tuberculosis vaccine, and after 5 weeks were infected with Mtb by intravenous route. The Tat protein was injected intradermally at 1, 2 and 4 weeks after Mtb challenge. Eight weeks after Mtb infection, all mice were sacrificed, and both the degree of pathology and immune responses to Mtb and Tat were evaluated. As additional control, some mice were either vaccinated or not with BCG, were not challenged with Mtb, but received the Tat protein. Statistical significances were evaluated by one-way or two-way ANOVA and Tukey’s multiple comparisons post-test. Results In the lungs of Mtb-infected mice, Tat-vaccine did not favour Mtb replication and indeed reduced both area of cellular infiltration and protein levels of Interferon-γ, Chemokine (C-C motif) ligand-4 and Interleukin-1β, pathological events triggered by Mtb-infection. Moreover, the protection against Mtb infection conferred by BCG remained good after Tat protein treatment. In spleen cells of Mtb-infected mice, Tat vaccination enhanced Mtb-specific Interferon-γ and Interleukin-17 responses, which may have a protective role. Of note, Mtb infection reduced, but did not suppress, the development of anti-Tat antibodies, required for Tat vaccine efficacy and the titer of anti-Tat IgG was potentiated by BCG vaccination in Mtb-free mice. In general, Tat treatment was well tolerated in both Mtb-infected and Mtb-free mice. Conclusions Tat protein vaccine, administered in Mtb-infected mice with a protocol resembling that used in the clinical trials, was safe, immunogenic, limited the lung Mtb-associated immunopathology and did not abrogate the protective efficacy of BCG. These data provide preliminary evidence for a safe use of Tat vaccine in people vaccinated with BCG and/or suffering from tuberculosis.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299 00161, Italy.
| | - Giovanni Piccaro
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299 00161, Italy
| | | | - Vincenzo Gigantino
- Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR) c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy.,Dipartimento di Patologia Fondazione G. Pascale IRCCS, Naples, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università di Salerno, Salerno, Italy.,MultiMedica IRCCS, Milan, Italy
| | - Erika Olivieri
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299 00161, Italy
| | - Flavia Ferrantelli
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299 00161, Italy
| | - Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299 00161, Italy
| | - Carla Palma
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299 00161, Italy.
| |
Collapse
|
41
|
Evans TG, Churchyard GJ, Penn-Nicholson A, Chen C, Gao X, Tait DR, Hatherill M. Epidemiologic studies and novel clinical research approaches that impact TB vaccine development. Tuberculosis (Edinb) 2016; 99 Suppl 1:S21-5. [DOI: 10.1016/j.tube.2016.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
42
|
Agger EM. Novel adjuvant formulations for delivery of anti-tuberculosis vaccine candidates. Adv Drug Deliv Rev 2016; 102:73-82. [PMID: 26596558 DOI: 10.1016/j.addr.2015.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 01/18/2023]
Abstract
There is an urgent need for a new and improved vaccine against tuberculosis for controlling this disease that continues to pose a global health threat. The current research strategy is to replace the present BCG vaccine or boost BCG-immunity with subunit vaccines such as viral vectored- or protein-based vaccines. The use of recombinant proteins holds a number of production advantages including ease of scalability, but requires an adjuvant inducing cell-mediated immune responses. A number of promising novel adjuvant formulations have recently been designed and show evidence of induction of cellular immune responses in humans. A common trait of effective TB adjuvants including those already in current clinical testing is a two-component approach combining a delivery system with an appropriate immunomodulator. This review summarizes the status of current TB adjuvant research with a focus on the division of labor between delivery systems and immunomodulators.
Collapse
Affiliation(s)
- Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark.
| |
Collapse
|
43
|
Boosting BCG-primed responses with a subunit Apa vaccine during the waning phase improves immunity and imparts protection against Mycobacterium tuberculosis. Sci Rep 2016; 6:25837. [PMID: 27173443 PMCID: PMC4865829 DOI: 10.1038/srep25837] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/15/2015] [Indexed: 11/08/2022] Open
Abstract
Heterologous prime-boosting has emerged as a powerful vaccination approach against tuberculosis. However, optimal timing to boost BCG-immunity using subunit vaccines remains unclear in clinical trials. Here, we followed the adhesin Apa-specific T-cell responses in BCG-primed mice and investigated its BCG-booster potential. The Apa-specific T-cell response peaked 32-52 weeks after parenteral or mucosal BCG-priming but waned significantly by 78 weeks. A subunit-Apa-boost during the contraction-phase of BCG-response had a greater effect on the magnitude and functional quality of specific cellular and humoral responses compared to a boost at the peak of BCG-response. The cellular response increased following mucosal BCG-prime-Apa-subunit-boost strategy compared to Apa-subunit-prime-BCG-boost approach. However, parenteral BCG-prime-Apa-subunit-boost by a homologous route was the most effective strategy in-terms of enhancing specific T-cell responses during waning in the lung and spleen. Two Apa-boosters markedly improved waning BCG-immunity and significantly reduced Mycobacterium tuberculosis burdens post-challenge. Our results highlight the challenges of optimization of prime-boost regimens in mice where BCG drives persistent immune-activation and suggest that boosting with a heterologous vaccine may be ideal once the specific persisting effector responses are contracted. Our results have important implications for design of prime-boost regimens against tuberculosis in humans.
Collapse
|
44
|
Prendergast KA, Counoupas C, Leotta L, Eto C, Bitter W, Winter N, Triccas JA. The Ag85B protein of the BCG vaccine facilitates macrophage uptake but is dispensable for protection against aerosol Mycobacterium tuberculosis infection. Vaccine 2016; 34:2608-15. [PMID: 27060378 DOI: 10.1016/j.vaccine.2016.03.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 02/09/2016] [Accepted: 03/28/2016] [Indexed: 10/22/2022]
Abstract
Defining the function and protective capacity of mycobacterial antigens is crucial for progression of tuberculosis (TB) vaccine candidates to clinical trials. The Ag85B protein is expressed by all pathogenic mycobacteria and is a component of multiple TB vaccines under evaluation in humans. In this report we examined the role of the BCG Ag85B protein in host cell interaction and vaccine-induced protection against virulent Mycobacterium tuberculosis infection. Ag85B was required for macrophage infection in vitro, as BCG deficient in Ag85B expression (BCG:(Δ85B)) was less able to infect RAW 264.7 macrophages compared to parental BCG, while an Ag85B-overexpressing BCG strain (BCG:(oex85B)) demonstrated improved uptake. A similar pattern was observed in vivo after intradermal delivery to mice, with significantly less BCG:(Δ85B) present in CD64(hi)CD11b(hi) macrophages compared to BCG or BCG:(oex85B). After vaccination of mice with BCG:(Δ85B) or parental BCG and subsequent aerosol M. tuberculosis challenge, similar numbers of activated CD4(+) and CD8(+) T cells were detected in the lungs of infected mice for both groups, suggesting the reduced macrophage uptake observed by BCG:(Δ85B) did not alter host immunity. Further, vaccination with both BCG:(Δ85B) and parental BCG resulted in a comparable reduction in pulmonary M. tuberculosis load. These data reveal an unappreciated role for Ag85B in the interaction of mycobacteria with host cells and indicates that single protective antigens are dispensable for protective immunity induced by BCG.
Collapse
Affiliation(s)
- Kelly A Prendergast
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, NSW, Australia; Mycobacterial Research Group, Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia
| | - Claudio Counoupas
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, NSW, Australia; Mycobacterial Research Group, Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia
| | - Lisa Leotta
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, NSW, Australia; Mycobacterial Research Group, Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia
| | - Carolina Eto
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, NSW, Australia
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, VU University Medical Center, Amsterdam, The Netherlands
| | - Nathalie Winter
- INRA, Université de Tours, UMR 1282, Infectiologie et Santé Publique, 37380 Nouzilly, France
| | - James A Triccas
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, NSW, Australia; Mycobacterial Research Group, Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia.
| |
Collapse
|
45
|
Pang Y, Zhao A, Cohen C, Kang W, Lu J, Wang G, Zhao Y, Zheng S. Current status of new tuberculosis vaccine in children. Hum Vaccin Immunother 2016; 12:960-70. [PMID: 27002369 DOI: 10.1080/21645515.2015.1120393] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Pediatric tuberculosis contributes significantly to the burden of TB disease worldwide. In order to achieve the goal of eliminating TB by 2050, an effective TB vaccine is urgently needed to prevent TB transmission in children. BCG vaccination can protect children from the severe types of TB such as TB meningitis and miliary TB, while its efficacy against pediatric pulmonary TB ranged from no protection to very high protection. In recent decades, multiple new vaccine candidates have been developed, and shown encouraging safety and immunogenicity in the preclinical experiments. However, the limited data on protective efficacy in infants evaluated by clinical trials has been disappointing, an example being MVA85A. To date, no vaccine has been shown to be clinically safer and more effective than the presently licensed BCG vaccine. Hence, before a new vaccine is developed with more promising efficacy, we must reconsider how to better use the current BCG vaccine to maximize its effectiveness in children.
Collapse
Affiliation(s)
- Yu Pang
- a Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University , Beijing , China.,b National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , China
| | - Aihua Zhao
- c National Institute for Food and Drug Control , Beijing , China
| | - Chad Cohen
- d McGill International TB Centre, Montreal , Quebec , Canada
| | - Wanli Kang
- a Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University , Beijing , China
| | - Jie Lu
- e Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University , Beijing , China
| | - Guozhi Wang
- c National Institute for Food and Drug Control , Beijing , China
| | - Yanlin Zhao
- b National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , China
| | - Suhua Zheng
- a Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University , Beijing , China
| |
Collapse
|
46
|
Son SJ, Harris PWR, Squire CJ, Baker EN, Brimble MA. Synthesis and structural insight into ESX-1 Substrate Protein C, an immunodominant Mycobacterium tuberculosis-secreted antigen. Biopolymers 2016; 106:267-74. [PMID: 26999334 DOI: 10.1002/bip.22838] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/18/2016] [Accepted: 03/16/2016] [Indexed: 11/08/2022]
Abstract
Tuberculosis, the second leading cause of death from a single infectious agent, is recognized as a major threat to human health due to a lack of practicable vaccines against the disease and the widespread occurrence of drug resistance. With a pressing need for a novel protein target as a platform for new vaccine development, ESX-1 Substrate Protein C (EspC) was recently identified as a novel Mycobacterium tuberculosis-secreted antigen that is as immunodominant as the two specific immunodiagnostic T-cell antigens, CFP-10 and ESAT-6. Here, we present the first chemical total synthesis, folding conditions, and circular dichroism data of EspC. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 267-274, 2016.
Collapse
Affiliation(s)
- Soo Jung Son
- School of Biological Sciences, The University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand
| | - Paul W R Harris
- School of Biological Sciences, The University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand.,School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, 1010, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand
| | - Chris J Squire
- School of Biological Sciences, The University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand
| | - Edward N Baker
- School of Biological Sciences, The University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand
| | - Margaret A Brimble
- School of Biological Sciences, The University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand.,School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, 1010, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 3a Symonds Street, Auckland, 1010, New Zealand
| |
Collapse
|
47
|
Aboutorabian S, Hakimi J, Boudet F, Montano S, Dookie A, Roque C, Ausar SF, Rahman N, Brookes RH. A high ratio of IC31(®) adjuvant to antigen is necessary for H4 TB vaccine immunomodulation. Hum Vaccin Immunother 2016; 11:1449-55. [PMID: 25997147 PMCID: PMC4514381 DOI: 10.1080/21645515.2015.1023970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
A tuberculosis (TB) vaccine consisting of a recombinant fusion protein (H4) and a novel TLR9 adjuvant (IC31) is in clinical development. To better understand the H4-IC31 ratio, we measured the binding capacity of IC31 for H4 protein and immunized mice with formulations that contained limiting to excess ratios of IC31 to H4. An immunomodulated H4-specific IFNγ response was only observed when IC31 was present in excess of H4. Since TLR expression is species-specific and the vaccine is intended to boost BCG-primed immunity, we questioned whether data in mice would translate to humans. To address this question, we used the fresh human Whole Blood (hWB) recovered from BCG-vaccinated subjects to screen H4-IC31 formulations. We found IC31 modulation in hWB to be quite distinct from the TLR4-Adjuvant. Unlike TLR4-Adjuvant, IC31 formulations did not induce the pro-inflammatory cytokine TNFα, but modulated a robust H4-specific IFNγ response after 12 d of culture. We then re-stimulated the fresh hWB of 5 BCG-primed subjects with formulations that had excess or limiting IC31 binding for H4 protein and again found that an immunomodulated H4-specific IFNγ response needed an excess of IC31. Finally, we monitored the zeta (ζ) potential of H4-IC31 formulations and found that the overall charge of H4-IC31 particles changes from negative to positive once IC31 is in greater than 9-fold excess. Using two diverse yet mutually supportive approaches, we confirm the need for an excess of IC31 adjuvant in H4 TB vaccine formulations and suggest surface potential may be an important factor.
Collapse
Key Words
- BCG, Bacillus Calmette-Guérin
- H4, TB, Antigen
- IC31 adjuvant modulation
- IC31®, Valneva Adjuvant consisting of KLK and ODN1a
- IFNγ, Interferon gamma
- KLK, Antimicrobial peptide H-KLKL5KLK-OH
- ODN1a, Oligodeoxynucleotide
- TB, Tuberculosis
- TLR, Toll-like Receptor
- TLR4, Toll-like Receptor 4
- TLR4-Adjuvant, TLR4A combined with an aluminum salt adjuvant
- TLR4A, TLR4 Agonist
- TLR9, Toll-like Receptor 9
- TNFα, Tumor Necrosis Factor alpha
- WBA
- functionality
- hWB, Human Whole Blood
- tuberculosis
- vaccine
Collapse
|
48
|
Tuberculosis vaccines--state of the art, and novel approaches to vaccine development. Int J Infect Dis 2016; 32:5-12. [PMID: 25809749 DOI: 10.1016/j.ijid.2014.11.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 11/21/2014] [Accepted: 11/25/2014] [Indexed: 11/20/2022] Open
Abstract
The quest for a vaccine that could have a major impact in reducing the current global burden of TB disease in humans continues to be extremely challenging. Significant gaps in our knowledge and understanding of the pathogenesis and immunology of tuberculosis continue to undermine efforts to break new ground, and traditional approaches to vaccine development have thus far met with limited success. Existing and novel candidate vaccines are being assessed in the context of their ability to impact the various stages that culminate in disease transmission and an increase in the global burden of disease. Innovative methods of vaccine administration and delivery have provided a fresh stimulus to the search for the elusive vaccine. Here we discuss the current status of preclinical vaccine development, providing insights into alternative approaches to vaccine delivery and promising candidate vaccines. The state of the art of clinical development also is reviewed.
Collapse
|
49
|
Fiore-Gartland A, Manso BA, Friedrich DP, Gabriel EE, Finak G, Moodie Z, Hertz T, De Rosa SC, Frahm N, Gilbert PB, McElrath MJ. Pooled-Peptide Epitope Mapping Strategies Are Efficient and Highly Sensitive: An Evaluation of Methods for Identifying Human T Cell Epitope Specificities in Large-Scale HIV Vaccine Efficacy Trials. PLoS One 2016; 11:e0147812. [PMID: 26863315 PMCID: PMC4749288 DOI: 10.1371/journal.pone.0147812] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/22/2015] [Indexed: 11/19/2022] Open
Abstract
The interferon gamma, enzyme-linked immunospot (IFN-γ ELISpot) assay is widely used to identify viral antigen-specific T cells is frequently employed to quantify T cell responses in HIV vaccine studies. It can be used to define T cell epitope specificities using panels of peptide antigens, but with sample and cost constraints there is a critical need to improve the efficiency of epitope mapping for large and variable pathogens. We evaluated two epitope mapping strategies, based on group testing, for their ability to identify vaccine-induced T-cells from participants in the Step HIV-1 vaccine efficacy trial, and compared the findings to an approach of assaying each peptide individually. The group testing strategies reduced the number of assays required by >7-fold without significantly altering the accuracy of T-cell breadth estimates. Assays of small pools containing 7–30 peptides were highly sensitive and effective at detecting single positive peptides as well as summating responses to multiple peptides. Also, assays with a single 15-mer peptide, containing an identified epitope, did not always elicit a response providing validation that 15-mer peptides are not optimal antigens for detecting CD8+ T cells. Our findings further validate pooling-based epitope mapping strategies, which are critical for characterizing vaccine-induced T-cell responses and more broadly for informing iterative vaccine design. We also show ways to improve their application with computational peptide:MHC binding predictors that can accurately identify the optimal epitope within a 15-mer peptide and within a pool of 15-mer peptides.
Collapse
Affiliation(s)
- Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
- * E-mail:
| | - Bryce A. Manso
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - David P. Friedrich
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Erin E. Gabriel
- Biostatistics Research Branch, National Institute of Allergy and Infectious Disease, Rockville, Maryland, 20852, United States of America
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Tomer Hertz
- Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion Institute of the Negev, Beer-Sheva, 84105, Israel
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| |
Collapse
|
50
|
A novel liposome adjuvant DPC mediates Mycobacterium tuberculosis subunit vaccine well to induce cell-mediated immunity and high protective efficacy in mice. Vaccine 2016; 34:1370-8. [PMID: 26845736 DOI: 10.1016/j.vaccine.2016.01.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 01/22/2016] [Indexed: 02/02/2023]
Abstract
Tuberculosis (TB) is a serious disease around the world, and protein based subunit vaccine is supposed to be a kind of promising novel vaccine against it. However, there is no effective adjuvant available in clinic to activate cell-mediated immune responses which is required for TB subunit vaccine. Therefore, it is imperative to develop new adjuvant. Here we reported an adjuvant composed of dimethyl dioctadecylammonium (DDA), Poly I:C and cholesterol (DPC for short). DDA can form a kind of cationic liposome with the ability to deliver and present antigen and can induce Th1 type cell-mediated immune response. Poly I:C, a ligand of TLR3 receptor, could attenuate the pathologic reaction induced by following Mycobacterium tuberculosis challenge. Cholesterol, which could enhance rigidity of lipid bilayer, is added to DDA and Poly I:C to improve the stability of the adjuvant. The particle size and Zeta-potential of DPC were analyzed in vitro. Furthermore, DPC was mixed with a TB fusion protein ESAT6-Ag85B-MPT64(190-198)-Mtb8.4-Rv2626c (LT70) to construct a subunit vaccine. The subunit vaccine-induced immune responses and protective efficacy against M. tuberculosis H37Rv infection in C57BL/6 mice were investigated. The results showed that the DPC adjuvant with particle size of 400 nm and zeta potential of 40 mV was in good stability. LT70 in the adjuvant of DPC generated strong antigen-specific humoral and cell-mediated immunity, and induced long-term higher protective efficacy against M. tuberculosis infection (5.41 ± 0.38log10CFU) than traditional vaccine Bacillus Calmette-Guerin (BCG) (6.01 ± 0.33log10CFU) and PBS control (6.53 ± 0.26log10CFU) at 30 weeks post-vaccination. In conclusion, DPC would be a promising vaccine adjuvant with the ability to stimulate Th1 type cell-mediated immunity, and could be used in TB subunit vaccine.
Collapse
|