1
|
Jeyanathan M, Afkhami S, Kang A, Xing Z. Viral-vectored respiratory mucosal vaccine strategies. Curr Opin Immunol 2023; 84:102370. [PMID: 37499279 DOI: 10.1016/j.coi.2023.102370] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023]
Abstract
Increasing global concerns of pandemic respiratory viruses highlight the importance of developing optimal vaccination strategies that encompass vaccine platform, delivery route, and regimens. The decades-long effort to develop vaccines to combat respiratory infections such as influenza, respiratory syncytial virus, and tuberculosis has met with challenges, including the inability of systemically administered vaccines to induce respiratory mucosal (RM) immunity. In this regard, ample preclinical and available clinical studies have demonstrated the superiority of RM vaccination to induce RM immunity over parenteral route of vaccination. A great stride has been made in developing vaccines for RM delivery against respiratory pathogens, including M. tuberculosis and SARS-CoV-2. In particular, inhaled aerosol delivery of adenoviral-vectored vaccines has shown significant promise.
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alisha Kang
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
2
|
Intranasal multivalent adenoviral-vectored vaccine protects against replicating and dormant M.tb in conventional and humanized mice. NPJ Vaccines 2023; 8:25. [PMID: 36823425 PMCID: PMC9948798 DOI: 10.1038/s41541-023-00623-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Viral-vectored vaccines are highly amenable for respiratory mucosal delivery as a means of inducing much-needed mucosal immunity at the point of pathogen entry. Unfortunately, current monovalent viral-vectored tuberculosis (TB) vaccine candidates have failed to demonstrate satisfactory clinical protective efficacy. As such, there is a need to develop next-generation viral-vectored TB vaccine strategies which incorporate both vaccine antigen design and delivery route. In this study, we have developed a trivalent chimpanzee adenoviral-vectored vaccine to provide protective immunity against pulmonary TB through targeting antigens linked to the three different growth phases (acute/chronic/dormancy) of Mycobacterium tuberculosis (M.tb) by expressing an acute replication-associated antigen, Ag85A, a chronically expressed virulence-associated antigen, TB10.4, and a dormancy/resuscitation-associated antigen, RpfB. Single-dose respiratory mucosal immunization with our trivalent vaccine induced robust, sustained tissue-resident multifunctional CD4+ and CD8+ T-cell responses within the lung tissues and airways, which were further quantitatively and qualitatively improved following boosting of subcutaneously BCG-primed hosts. Prophylactic and therapeutic immunization with this multivalent trivalent vaccine in conventional BALB/c mice provided significant protection against not only actively replicating M.tb bacilli but also dormant, non-replicating persisters. Importantly, when used as a booster, it also provided marked protection in the highly susceptible C3HeB/FeJ mice, and a single respiratory mucosal inoculation was capable of significant protection in a humanized mouse model. Our findings indicate the great potential of this next-generation TB vaccine strategy and support its further clinical development for both prophylactic and therapeutic applications.
Collapse
|
3
|
Hu Z, Lu SH, Lowrie DB, Fan XY. Research Advances for Virus-vectored Tuberculosis Vaccines and Latest Findings on Tuberculosis Vaccine Development. Front Immunol 2022; 13:895020. [PMID: 35812383 PMCID: PMC9259874 DOI: 10.3389/fimmu.2022.895020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by respiratory infection with Mycobacterium tuberculosis, remains a major global health threat. The only licensed TB vaccine, the one-hundred-year-old Bacille Calmette-Guérin has variable efficacy and often provides poor protection against adult pulmonary TB, the transmissible form of the disease. Thus, the lack of an optimal TB vaccine is one of the key barriers to TB control. Recently, the development of highly efficacious COVID-19 vaccines within one year accelerated the vaccine development process in human use, with the notable example of mRNA vaccines and adenovirus-vectored vaccines, and increased the public acceptance of the concept of the controlled human challenge model. In the TB vaccine field, recent progress also facilitated the deployment of an effective TB vaccine. In this review, we provide an update on the current virus-vectored TB vaccine pipeline and summarize the latest findings that might facilitate TB vaccine development. In detail, on the one hand, we provide a systematic literature review of the virus-vectored TB vaccines are in clinical trials, and other promising candidate vaccines at an earlier stage of development are being evaluated in preclinical animal models. These research sharply increase the likelihood of finding a more effective TB vaccine in the near future. On the other hand, we provide an update on the latest tools and concept that facilitating TB vaccine research development. We propose that a pre-requisite for successful development may be a better understanding of both the lung-resident memory T cell-mediated mucosal immunity and the trained immunity of phagocytic cells. Such knowledge could reveal novel targets and result in the innovative vaccine designs that may be needed for a quantum leap forward in vaccine efficacy. We also summarized the research on controlled human infection and ultra-low-dose aerosol infection murine models, which may provide more realistic assessments of vaccine utility at earlier stages. In addition, we believe that the success in the ongoing efforts to identify correlates of protection would be a game-changer for streamlining the triage of multiple next-generation TB vaccine candidates. Thus, with more advanced knowledge of TB vaccine research, we remain hopeful that a more effective TB vaccine will eventually be developed in the near future.
Collapse
Affiliation(s)
- Zhidong Hu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- *Correspondence: Zhidong Hu, ; Xiao-Yong Fan,
| | - Shui-Hua Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- National Medical Center for Infectious Diseases of China, Shenzhen Third People Hospital, South Science & Technology University, Shenzhen, China
| | - Douglas B. Lowrie
- National Medical Center for Infectious Diseases of China, Shenzhen Third People Hospital, South Science & Technology University, Shenzhen, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- *Correspondence: Zhidong Hu, ; Xiao-Yong Fan,
| |
Collapse
|
4
|
Enriquez AB, Izzo A, Miller SM, Stewart EL, Mahon RN, Frank DJ, Evans JT, Rengarajan J, Triccas JA. Advancing Adjuvants for Mycobacterium tuberculosis Therapeutics. Front Immunol 2021; 12:740117. [PMID: 34759923 PMCID: PMC8572789 DOI: 10.3389/fimmu.2021.740117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 01/15/2023] Open
Abstract
Tuberculosis (TB) remains one of the leading causes of death worldwide due to a single infectious disease agent. BCG, the only licensed vaccine against TB, offers limited protection against pulmonary disease in children and adults. TB vaccine research has recently been reinvigorated by new data suggesting alternative administration of BCG induces protection and a subunit/adjuvant vaccine that provides close to 50% protection. These results demonstrate the need for generating adjuvants in order to develop the next generation of TB vaccines. However, development of TB-targeted adjuvants is lacking. To help meet this need, NIAID convened a workshop in 2020 titled “Advancing Vaccine Adjuvants for Mycobacterium tuberculosis Therapeutics”. In this review, we present the four areas identified in the workshop as necessary for advancing TB adjuvants: 1) correlates of protective immunity, 2) targeting specific immune cells, 3) immune evasion mechanisms, and 4) animal models. We will discuss each of these four areas in detail and summarize what is known and what we can advance on in order to help develop more efficacious TB vaccines.
Collapse
Affiliation(s)
- Ana B Enriquez
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Angelo Izzo
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Erica L Stewart
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Robert N Mahon
- Division of AIDS, Columbus Technologies & Services Inc., Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel J Frank
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.,Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - James A Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Yang K, Kallies A. Tissue-specific differentiation of CD8 + resident memory T cells. Trends Immunol 2021; 42:876-890. [PMID: 34531111 DOI: 10.1016/j.it.2021.08.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022]
Abstract
CD8+ tissue-resident memory T (TRM) cells play crucial roles in defense against infections and cancer and have been implicated in autoimmune diseases such as psoriasis. In mice and humans, they exist in all nonlymphoid organs and share key characteristics across all tissues, including downregulation of tissue egress and lymph node homing pathways. However, recent studies demonstrate considerable heterogeneity across TRM cells lodged in different tissues - linked to the activity of tissue-specific molecules, including chemokines, cytokines, and transcription factors. Current work indicates that transforming growth factor (TGF)-β plays a major role in generating TRM heterogeneity at phenotypic and functional levels. Here, we review common and unique features of TRM cells in different tissues and discuss putative strategies aimed at harnessing TRM cells for site-specific protection against infectious and malignant diseases.
Collapse
Affiliation(s)
- Kun Yang
- Department of Dermatology, Beijing Hospital, National Center of Gerontology, Beijing, China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
6
|
Intramuscular Boosting with hIFN-Alpha 2b Enhances BCGphipps-Induced Protection in a Murine Model of Leprosy. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12030051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Host immunity to Mycobacterium leprae encompasses a spectrum of mechanisms that range from cellular immunity-driven protection to damage associated with humoral immunity as in type-2 leprosy reactions. Although type I interferons (IFNs) participate in eliminating intracellular pathogens, their contribution to the production of antibodies and CD3+ FOXP3+ regulatory T cells (Tregs) in BCG vaccine-mediated protection in leprosy is unknown. BCGphipps (BCGph) priming followed by intramuscular hIFN-α 2b boost significantly reduced lesion size and Mycobacterium lepraemurium growth in the skin. T follicular regulatory cells (TFR), a subset of Tregs induced by immunization or infection, reside in the germinal centers (GCs) and modulate antibody production. We found impaired Treg induction and improved GCs in draining lymph nodes of BCGph primed and hIFN-α 2b boosted mice. Moreover, these mice elicited significant amounts of IL-4 and IL-10 in serum. Thus, our results support the adjuvant properties of hIFN-α 2b in the context of BCGph priming to enhance protective immunity against skin leprosy.
Collapse
|
7
|
Chung H, Kim EA, Chang J. A "Prime and Deploy" Strategy for Universal Influenza Vaccine Targeting Nucleoprotein Induces Lung-Resident Memory CD8 T cells. Immune Netw 2021; 21:e28. [PMID: 34522441 PMCID: PMC8410988 DOI: 10.4110/in.2021.21.e28] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 01/31/2023] Open
Abstract
Lung-resident memory T cells (TRM) play an essential role in protecting against pulmonary virus infection. Parenteral administration of DNA vaccine is generally not sufficient to induce lung CD8 TRM cells. This study investigates whether intramuscularly administered DNA vaccine expressing the nucleoprotein (NP) induces lung TRM cells and protects against the influenza B virus. The results show that DNA vaccination poorly generates lung TRM cells and massive secondary effector CD8 T cells entering the lungs after challenge infection do not offer sufficient protection. Nonetheless, intranasal administration of non-replicating adenovirus vector expressing no Ag following priming DNA vaccination deploys NP-specific CD8 TRM cells in the lungs, which subsequently offers complete protection. This novel 'prime and deploy' strategy could be a promising regimen for a universal influenza vaccine targeting the conserved NP Ag.
Collapse
Affiliation(s)
- Haerynn Chung
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Eun-Ah Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
8
|
Martini V, Paudyal B, Chrun T, McNee A, Edmans M, Atangana Maze E, Clark B, Nunez A, Dolton G, Sewell A, Beverley P, MacLoughlin R, Townsend A, Tchilian E. Simultaneous Aerosol and Intramuscular Immunization with Influenza Vaccine Induces Powerful Protective Local T Cell and Systemic Antibody Immune Responses in Pigs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:652-663. [PMID: 33328212 PMCID: PMC7812058 DOI: 10.4049/jimmunol.2001086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 01/04/2023]
Abstract
A vaccine providing both powerful Ab and cross-reactive T cell immune responses against influenza viruses would be beneficial for both humans and pigs. In this study, we evaluated i.m., aerosol (Aer), and simultaneous systemic and respiratory immunization (SIM) by both routes in Babraham pigs, using the single cycle candidate influenza vaccine S-FLU. After prime and boost immunization, pigs were challenged with H1N1pdm09 virus. i.m.-immunized pigs generated a high titer of neutralizing Abs but poor T cell responses, whereas Aer induced powerful respiratory tract T cell responses but a low titer of Abs. SIM pigs combined high Ab titers and strong local T cell responses. SIM showed the most complete suppression of virus shedding and the greatest improvement in pathology. We conclude that SIM regimes for immunization against respiratory pathogens warrant further study.
Collapse
Affiliation(s)
- Veronica Martini
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom; .,Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Basu Paudyal
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Tiphany Chrun
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Adam McNee
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Matthew Edmans
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | | | - Beckie Clark
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Alejandro Nunez
- UK Animal and Plant Health Agency-Weybridge, New Haw, Addlestone KT15 3NB, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Andrew Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Peter Beverley
- National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom; and
| | | | - Alain Townsend
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Elma Tchilian
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom;
| |
Collapse
|
9
|
D'Agostino MR, Lai R, Afkhami S, Khera A, Yao Y, Vaseghi-Shanjani M, Zganiacz A, Jeyanathan M, Xing Z. Airway Macrophages Mediate Mucosal Vaccine-Induced Trained Innate Immunity against Mycobacterium tuberculosis in Early Stages of Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:2750-2762. [PMID: 32998983 DOI: 10.4049/jimmunol.2000532] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Abstract
Mycobacterium tuberculosis, the causative agent of pulmonary tuberculosis (TB), is responsible for millions of infections and deaths annually. Decades of TB vaccine development have focused on adaptive T cell immunity, whereas the importance of innate immune contributions toward vaccine efficacy has only recently been recognized. Airway macrophages (AwM) are the predominant host cell during early pulmonary M. tuberculosis infection and, therefore, represent attractive targets for vaccine-mediated immunity. We have demonstrated that respiratory mucosal immunization with a viral-vectored vaccine imprints AwM, conferring enhanced protection against heterologous bacterial challenge. However, it is unknown if innate immune memory also protects against M. tuberculosis In this study, by using a murine model, we detail whether respiratory mucosal TB vaccination profoundly alters the airway innate immune landscape associated with AwM prior to M. tuberculosis exposure and whether such AwM play a critical role in host defense against M. tuberculosis infection. Our study reveals an important role of AwM in innate immune protection in early stages of M. tuberculosis infection in the lung.
Collapse
Affiliation(s)
- Michael R D'Agostino
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Rocky Lai
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Amandeep Khera
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Yushi Yao
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Maryam Vaseghi-Shanjani
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada; and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
10
|
Toniolo SP, Afkhami S, D'Agostino MR, Lichty BD, Cranston ED, Xing Z, Thompson MR. Spray dried VSV-vectored vaccine is thermally stable and immunologically active in vivo. Sci Rep 2020; 10:13349. [PMID: 32770018 PMCID: PMC7414861 DOI: 10.1038/s41598-020-70325-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Effective vaccine delivery and coverage to rural and resource-poor countries is hindered by the dependence on cold chain storage. As such, developments of cold chain-free technologies are highly sought. Although spray dried adenoviral vectors have shown long term stability at ambient temperatures and relatively low humidity, it remains to be determined whether similar excipient formulations are applicable to other viral vectors. To address this, we have spray dried vesicular stomatitis virus (VSV)-vectors with a panel of well-characterized sugar excipients to determine the optimal formulation for vector stabilization. Upon reconstitution, we show that trehalose conferred superior stability of VSV both in vitro and in vivo. Importantly, following cold chain-free storage at elevated temperatures at 37 °C for 15 days, we show that a VSV-vectored vaccine retains its in vivo immunogenicity, whereas a liquid control completely lost its immune-stimulating ability. Our results provide foundational evidence that spray drying with properly tested excipients can stabilize viral vectors such as VSV, allowing them to be stored long-term at elevated temperatures without dependency on cold chain conditions.
Collapse
Affiliation(s)
- Steven P Toniolo
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Brian D Lichty
- McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Emily D Cranston
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada.,Department of Wood Science, The University of British Columbia, Vancouver, Canada.,Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, Canada
| | - Zhou Xing
- McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Michael R Thompson
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
11
|
Xing Z, Afkhami S, Bavananthasivam J, Fritz DK, D'Agostino MR, Vaseghi-Shanjani M, Yao Y, Jeyanathan M. Innate immune memory of tissue-resident macrophages and trained innate immunity: Re-vamping vaccine concept and strategies. J Leukoc Biol 2020; 108:825-834. [PMID: 32125045 DOI: 10.1002/jlb.4mr0220-446r] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023] Open
Abstract
In the past few years, our understanding of immunological memory has evolved remarkably due to a growing body of new knowledge in innate immune memory and immunity. Immunological memory now encompasses both innate and adaptive immune memory. The hypo-reactive and hyper-reactive types of innate immune memory lead to a suppressed and enhanced innate immune protective outcome, respectively. The latter is also named trained innate immunity (TII). The emerging information on innate immune memory has not only shed new light on the mechanisms of host defense but is also revolutionizing our long-held view of vaccination and vaccine strategies. Our current review will examine recent progress and knowledge gaps in innate immune memory with a focus on tissue-resident Mϕs, particularly lung Mϕs, and their relationship to local antimicrobial innate immunity. We will also discuss the impact of innate immune memory and TII on our understanding of vaccine concept and strategies and the significance of respiratory mucosal route of vaccination against respiratory pathogens.
Collapse
Affiliation(s)
- Zhou Xing
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jegarubee Bavananthasivam
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dominik K Fritz
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Maryam Vaseghi-Shanjani
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yushi Yao
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Current affiliation: Department of Immunology, Zhejiang University, Zhejiang, China
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
12
|
Haddadi S, Vaseghi-Shanjani M, Yao Y, Afkhami S, D'Agostino MR, Zganiacz A, Jeyanathan M, Xing Z. Mucosal-Pull Induction of Lung-Resident Memory CD8 T Cells in Parenteral TB Vaccine-Primed Hosts Requires Cognate Antigens and CD4 T Cells. Front Immunol 2019; 10:2075. [PMID: 31552032 PMCID: PMC6747041 DOI: 10.3389/fimmu.2019.02075] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022] Open
Abstract
Tissue-resident memory T cells (TRM) are critical to host defense at mucosal tissue sites. However, the parenteral route of immunization as the most commonly used immunization route in practice is not effective in inducing mucosal TRM cells particularly in the lung. While various respiratory mucosal (RM)-pull strategies are exploited to mobilize parenteral vaccine-primed T cells into the lung, whether such RM-pull strategies can all or differentially induce Ag-specific TRM cells in the lung remains unclear. Here, we have addressed this issue by using a parenteral TB vaccine-primed and RM-pull model. We show that both Ag-independent and Ag-dependent RM-pull strategies are able to mobilize Ag-specific CD8 T cells into the lung. However, only the RM-pull strategy with cognate antigens can induce robust Ag-specific CD8 TRM cells in the lung. We also show that the cognate Ag-based RM-pull strategy is the most effective in inducing TRM cells when carried out during the memory phase, as opposed to the effector phase, of T cell responses to parenteral prime vaccination. We further find that cognate Ag-induced CD4 T cells play an important role in the development of CD8 TRM cells in the lung. Our study holds implications in developing effective vaccine strategies against respiratory pathogens.
Collapse
Affiliation(s)
- Siamak Haddadi
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Maryam Vaseghi-Shanjani
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Yushi Yao
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Michael R D'Agostino
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Anna Zganiacz
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Mangalakumari Jeyanathan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Zhou Xing
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
13
|
Mucosal boosting of H56:CAF01 immunization promotes lung-localized T cells and an accelerated pulmonary response to Mycobacterium tuberculosis infection without enhancing vaccine protection. Mucosal Immunol 2019; 12:816-826. [PMID: 30760832 DOI: 10.1038/s41385-019-0145-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/20/2018] [Accepted: 01/23/2019] [Indexed: 02/04/2023]
Abstract
T cell-mediated protection against Mycobacterium tuberculosis (Mtb) is dependent upon the ability to localize within the site of pulmonary infection and directly interact with infected cells. In turn, vaccine strategies to improve rapid T cell targeting of Mtb-infected cells after pulmonary exposure are being actively pursued. Given parenterally, the subunit vaccine H56:CAF01 elicits polyfunctional CD4 T cells that localize to the lung parenchyma and confer durable protection. Here, we find that airway mucosal boosting of parenteral H56:CAF01 immunization greatly enhances the population of long-lived lung-resident T cells (Trm) and increases early vaccine T cell responses to pulmonary Mtb challenge in multiple mouse models. However, mucosal boosting does not alter the Th1/17 vaccine signature typical of H56:CAF01 and does not further improve durable control of pulmonary infection following aerosol Mtb-challenge. Additional mucosal boosting with H56:CAF01 further enhances the Trm response without further improving protection, while blocking the recruitment of non-Trm with FTY720-treatment failed to exposed Trm-mediated protection in mucosally boosting animals. These results demonstrate the limitations of maximizing lung-localized Trm in vaccine control of pulmonary Mtb infection, especially within an immunization protocol that is already optimized for the induction of mucosal-homing Th17 cells.
Collapse
|
14
|
Thakur A, Rodríguez-Rodríguez C, Saatchi K, Rose F, Esposito T, Nosrati Z, Andersen P, Christensen D, Häfeli UO, Foged C. Dual-Isotope SPECT/CT Imaging of the Tuberculosis Subunit Vaccine H56/CAF01: Induction of Strong Systemic and Mucosal IgA and T-Cell Responses in Mice Upon Subcutaneous Prime and Intrapulmonary Boost Immunization. Front Immunol 2018; 9:2825. [PMID: 30555488 PMCID: PMC6284049 DOI: 10.3389/fimmu.2018.02825] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
Pulmonary tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains a global pandemic, despite the widespread use of the parenteral live attenuated Bacillus Calmette–Guérin (BCG) vaccine during the past decades. Mucosal administration of next generation TB vaccines has great potential, but developing a safe and efficacious mucosal vaccine is challenging. Hence, understanding the in vivo biodistribution and pharmacokinetics of mucosal vaccines is essential for shaping the desired immune response and for optimal spatiotemporal targeting of the appropriate effector cells in the lungs. A subunit vaccine consisting of the fusion antigen H56 (Ag85B-ESAT-6-Rv2660) and the liposome-based cationic adjuvant formulation (CAF01) confers efficient protection in preclinical animal models. In this study, we devise a novel immunization strategy for the H56/CAF01 vaccine, which comply with the intrapulmonary (i.pulmon.) route of immunization. We also describe a novel dual-isotope (111In/67Ga) radiolabeling approach, which enables simultaneous non-invasive and longitudinal SPECT/CT imaging and quantification of H56 and CAF01 upon parenteral prime and/or i.pulmon. boost immunization. Our results demonstrate that the vaccine is distributed evenly in the lungs, and there are pronounced differences in the pharmacokinetics of H56 and CAF01. We provide convincing evidence that the H56/CAF01 vaccine is not only well-tolerated when administered to the respiratory tract, but it also induces strong lung mucosal and systemic IgA and polyfunctional Th1 and Th17 responses after parenteral prime and i.pulmon. boost immunization. The study furthermore evaluate the application of SPECT/CT imaging for the investigation of vaccine biodistribution after parenteral and i.pulmon. immunization of mice.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.,Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Fabrice Rose
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tullio Esposito
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Zeynab Nosrati
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Jeyanathan M, Yao Y, Afkhami S, Smaill F, Xing Z. New Tuberculosis Vaccine Strategies: Taking Aim at Un-Natural Immunity. Trends Immunol 2018; 39:419-433. [DOI: 10.1016/j.it.2018.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
|
16
|
Prevention of tuberculosis in rhesus macaques by a cytomegalovirus-based vaccine. Nat Med 2018; 24:130-143. [PMID: 29334373 DOI: 10.1038/nm.4473] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023]
Abstract
Despite widespread use of the bacille Calmette-Guérin (BCG) vaccine, tuberculosis (TB) remains a leading cause of global mortality from a single infectious agent (Mycobacterium tuberculosis or Mtb). Here, over two independent Mtb challenge studies, we demonstrate that subcutaneous vaccination of rhesus macaques (RMs) with rhesus cytomegalovirus vectors encoding Mtb antigen inserts (hereafter referred to as RhCMV/TB)-which elicit and maintain highly effector-differentiated, circulating and tissue-resident Mtb-specific CD4+ and CD8+ memory T cell responses-can reduce the overall (pulmonary and extrapulmonary) extent of Mtb infection and disease by 68%, as compared to that in unvaccinated controls, after intrabronchial challenge with the Erdman strain of Mtb at ∼1 year after the first vaccination. Fourteen of 34 RhCMV/TB-vaccinated RMs (41%) across both studies showed no TB disease by computed tomography scans or at necropsy after challenge (as compared to 0 of 17 unvaccinated controls), and ten of these RMs were Mtb-culture-negative for all tissues, an exceptional long-term vaccine effect in the RM challenge model with the Erdman strain of Mtb. These results suggest that complete vaccine-mediated immune control of highly pathogenic Mtb is possible if immune effector responses can intercept Mtb infection at its earliest stages.
Collapse
|
17
|
Haddadi S, Thanthrige-Don N, Afkhami S, Khera A, Jeyanathan M, Xing Z. Expression and role of VLA-1 in resident memory CD8 T cell responses to respiratory mucosal viral-vectored immunization against tuberculosis. Sci Rep 2017; 7:9525. [PMID: 28842633 PMCID: PMC5573413 DOI: 10.1038/s41598-017-09909-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/31/2017] [Indexed: 11/29/2022] Open
Abstract
Lung resident memory T cells (TRM) characterized by selective expression of mucosal integrins VLA-1 (α1β1) and CD103 (αEβ7) are generated following primary respiratory viral infections. Despite recent progress, the generation of lung TRM and the role of mucosal integrins following viral vector respiratory mucosal immunization still remains poorly understood. Here by using a replication-defective viral vector tuberculosis vaccine, we show that lung Ag-specific CD8 T cells express both VLA-1 and CD103 following respiratory mucosal immunization. However, VLA-1 and CD103 are acquired in differential tissue sites with the former acquired during T cell priming in the draining lymph nodes and the latter acquired after T cells entered the lung. Once in the lung, Ag-specific CD8 T cells continue to express VLA-1 at high levels through the effector/expansion, contraction, and memory phases of T cell responses. Using a functional VLA-1 blocking mAb, we show that VLA-1 is not required for trafficking of these cells to the lung, but it negatively regulates them in the contraction phase. Furthermore, VLA-1 plays a negligible role in the maintenance of these cells in the lung. Our study provides new information on vaccine-inducible lung TRM and shall help develop effective viral vector respiratory mucosal tuberculosis vaccination strategies.
Collapse
Affiliation(s)
- Siamak Haddadi
- McMaster Immunology Research Centre, Department of Pathology & Molecular Medicine, Hamilton, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Niroshan Thanthrige-Don
- McMaster Immunology Research Centre, Department of Pathology & Molecular Medicine, Hamilton, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, Department of Pathology & Molecular Medicine, Hamilton, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Amandeep Khera
- McMaster Immunology Research Centre, Department of Pathology & Molecular Medicine, Hamilton, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, Department of Pathology & Molecular Medicine, Hamilton, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, Department of Pathology & Molecular Medicine, Hamilton, Ontario, Canada. .,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
18
|
Jeyanathan M, Afkhami S, Khera A, Mandur T, Damjanovic D, Yao Y, Lai R, Haddadi S, Dvorkin-Gheva A, Jordana M, Kunkel SL, Xing Z. CXCR3 Signaling Is Required for Restricted Homing of Parenteral Tuberculosis Vaccine-Induced T Cells to Both the Lung Parenchyma and Airway. THE JOURNAL OF IMMUNOLOGY 2017; 199:2555-2569. [PMID: 28827285 DOI: 10.4049/jimmunol.1700382] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/24/2017] [Indexed: 01/19/2023]
Abstract
Although most novel tuberculosis (TB) vaccines are designed for delivery via the muscle or skin for enhanced protection in the lung, it has remained poorly understood whether systemic vaccine-induced memory T cells can readily home to the lung mucosa prior to and shortly after pathogen exposure. We have investigated this issue by using a model of parenteral TB immunization and intravascular immunostaining. We find that systemically induced memory T cells are restricted to the blood vessels in the lung, unable to populate either the lung parenchymal tissue or the airway under homeostatic conditions. We further find that after pulmonary TB infection, it still takes many days before such T cells can enter the lung parenchymal tissue and airway. We have identified the acquisition of CXCR3 expression by circulating T cells to be critical for their entry to these lung mucosal compartments. Our findings offer new insights into mucosal T cell biology and have important implications in vaccine strategies against pulmonary TB and other intracellular infections in the lung.
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Sam Afkhami
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Amandeep Khera
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Talveer Mandur
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Daniela Damjanovic
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Yushi Yao
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Rocky Lai
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Siamak Haddadi
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Anna Dvorkin-Gheva
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Manel Jordana
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| | - Steven L Kunkel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Zhou Xing
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada; .,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; and
| |
Collapse
|
19
|
Decreased Vector Gene Expression from E2b Gene-Deleted Adenovirus Serotype 5 Vaccines Intensifies Proinflammatory Immune Responses. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00061-17. [PMID: 28381403 DOI: 10.1128/cvi.00061-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/20/2017] [Indexed: 02/06/2023]
Abstract
Recombinant adenovirus serotype 5 (Ad5) vectors are promising vaccine candidates due to their intrinsic immunogenicity and potent transgene expression; however, widespread preexisting Ad5 immunity has been considered a developmental impediment to the use of traditional, or conventional, E1 and E3 gene-deleted Ad5 (Ad5[E1-]) vaccines. Even in the presence of anti-Ad5 immunity, recent murine and human studies have confirmed E2b gene-deleted Ad5 (Ad5[E1-,E2b-]) vaccines to be highly efficacious inducers of transgene-specific memory responses and significantly less toxic options than Ad5[E1-] vaccines. While these findings have been substantially confirmed, the molecular mechanisms underlying the different reactions to these vaccine platforms are unknown. Using cultures of human peripheral blood mononuclear cells (hPBMCs) derived from multiple human donors, we found that Ad5[E1-,E2b-] vaccines trigger higher levels of hPBMC proinflammatory cytokine secretion than Ad5[E1-] vaccines. Interestingly, these responses were generated regardless of the donors' preexisting anti-Ad5 humoral and cell-mediated immune response status. In vitro hPBMC infection with the Ad5[E1-,E2b-] vaccine also provoked greater Th1-dominant gene responses yet smaller amounts of Ad-derived gene expression than Ad5[E1-] vaccines. These results suggest that Ad5[E1-,E2b-] vaccines, in contrast to Ad5[E1-] vaccines, do not promote activities that suppress innate immune signaling, thereby allowing for improved vaccine efficacy and a superior safety profile independently of previous Ad5 immunity.
Collapse
|
20
|
Verreck FAW, Tchilian EZ, Vervenne RAW, Sombroek CC, Kondova I, Eissen OA, Sommandas V, van der Werff NM, Verschoor E, Braskamp G, Bakker J, Langermans JAM, Heidt PJ, Ottenhoff THM, van Kralingen KW, Thomas AW, Beverley PCL, Kocken CHM. Variable BCG efficacy in rhesus populations: Pulmonary BCG provides protection where standard intra-dermal vaccination fails. Tuberculosis (Edinb) 2017; 104:46-57. [PMID: 28454649 DOI: 10.1016/j.tube.2017.02.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 01/22/2023]
Abstract
M.bovis BCG vaccination against tuberculosis (TB) notoriously displays variable protective efficacy in different human populations. In non-human primate studies using rhesus macaques, despite efforts to standardise the model, we have also observed variable efficacy of BCG upon subsequent experimental M. tuberculosis challenge. In the present head-to-head study, we establish that the protective efficacy of standard parenteral BCG immunisation varies among different rhesus cohorts. This provides different dynamic ranges for evaluation of investigational vaccines, opportunities for identifying possible correlates of protective immunity and for determining why parenteral BCG immunisation sometimes fails. We also show that pulmonary mucosal BCG vaccination confers reduced local pathology and improves haematological and immunological parameters post-infection in animals that are not responsive to induction of protection by standard intra-dermal BCG. These results have important implications for pulmonary TB vaccination strategies in the future.
Collapse
Affiliation(s)
- Frank A W Verreck
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands.
| | - Elma Z Tchilian
- The Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, UK.
| | - Richard A W Vervenne
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Claudia C Sombroek
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Ivanela Kondova
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Okke A Eissen
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Vinod Sommandas
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Nicole M van der Werff
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Ernst Verschoor
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Gerco Braskamp
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Jaco Bakker
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Jan A M Langermans
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Peter J Heidt
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Centre (LUMC), Albinusdreef 2, 2333-ZA, Leiden, The Netherlands
| | - Klaas W van Kralingen
- Department of Pulmonology, Leiden University Medical Centre (LUMC), Albinusdreef 2, 2333-ZA, Leiden, The Netherlands
| | - Alan W Thomas
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| | - Peter C L Beverley
- The Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford, UK.
| | - Clemens H M Kocken
- Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288-GJ, Rijswijk, The Netherlands
| |
Collapse
|
21
|
Afkhami S, Yao Y, Xing Z. Methods and clinical development of adenovirus-vectored vaccines against mucosal pathogens. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16030. [PMID: 27162933 PMCID: PMC4847555 DOI: 10.1038/mtm.2016.30] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 03/27/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
Adenoviruses represent the most widely used viral-vectored platform for vaccine design, showing a great potential in the fight against intracellular infectious diseases to which either there is a lack of effective vaccines or the traditional vaccination strategy is suboptimal. The extensive understanding of the molecular biology of adenoviruses has made the new technologies and reagents available to efficient generation of adenoviral-vectored vaccines for both preclinical and clinical evaluation. The novel adenoviral vectors including nonhuman adenoviral vectors have emerged to be the further improved vectors for vaccine design. In this review, we discuss the latest adenoviral technologies and their utilization in vaccine development. We particularly focus on the application of adenoviral-vectored vaccines in mucosal immunization strategies against mucosal pathogens including Mycobacterium tuberculosis, flu virus, and human immunodeficiency virus.
Collapse
Affiliation(s)
- Sam Afkhami
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, Ontario, Canada
| | - Yushi Yao
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, Ontario, Canada
| | - Zhou Xing
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, Ontario, Canada
| |
Collapse
|
22
|
Lai R, Afkhami S, Haddadi S, Jeyanathan M, Xing Z. Mucosal immunity and novel tuberculosis vaccine strategies: route of immunisation-determined T-cell homing to restricted lung mucosal compartments. Eur Respir Rev 2016; 24:356-60. [PMID: 26028646 DOI: 10.1183/16000617.00002515] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Despite the use of bacille Calmette-Guérin (BCG) for almost a century, pulmonary tuberculosis (TB) continues to be a serious global health concern. Therefore, there has been a pressing need for the development of new booster vaccines to enhance existing BCG-induced immunity. Protection following mucosal intranasal immunisation with AdHu5Ag85A is associated with the localisation of antigen-specific T-cells to the lung airway. However, parenteral intramuscular immunisation is unable to provide protection despite the apparent presence of antigen-specific T-cells in the lung interstitium. Recent advances in intravascular staining have allowed us to reassess the previously established T-cell distribution profile and its relationship with the observed differential protection. Respiratory mucosal immunisation empowers T-cells to home to both the lung interstitium and the airway lumen, whereas intramuscular immunisation-activated T-cells are largely trapped within the pulmonary vasculature, unable to populate the lung interstitium and airway. Given the mounting evidence supporting the safety and enhanced efficacy of respiratory mucosal immunisation over the traditional parenteral immunisation route, a greater effort should be made to clinically develop respiratory mucosal-deliverable TB vaccines.
Collapse
Affiliation(s)
- Rocky Lai
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Dept of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Dept of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Siamak Haddadi
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Dept of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Dept of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Dept of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
23
|
Jeyanathan M, Thanthrige-Don N, Afkhami S, Lai R, Damjanovic D, Zganiacz A, Feng X, Yao XD, Rosenthal KL, Medina MF, Gauldie J, Ertl HC, Xing Z. Novel chimpanzee adenovirus-vectored respiratory mucosal tuberculosis vaccine: overcoming local anti-human adenovirus immunity for potent TB protection. Mucosal Immunol 2015; 8:1373-87. [PMID: 25872483 DOI: 10.1038/mi.2015.29] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 03/20/2015] [Indexed: 02/07/2023]
Abstract
Pulmonary tuberculosis (TB) remains to be a major global health problem despite many decades of parenteral use of Bacillus Calmette-Guérin (BCG) vaccine. Developing safe and effective respiratory mucosal TB vaccines represents a unique challenge. Over the past decade or so, the human serotype 5 adenovirus (AdHu5)-based TB vaccine has emerged as one of the most promising candidates based on a plethora of preclinical and early clinical studies. However, anti-AdHu5 immunity widely present in the lung of humans poses a serious gap and limitation to its real-world applications. In this study we have developed a novel chimpanzee adenovirus 68 (AdCh68)-vectored TB vaccine amenable to the respiratory route of vaccination. We have evaluated AdCh68-based TB vaccine for its safety, T-cell immunogenicity, and protective efficacy in relevant animal models of human pulmonary TB with or without parenteral BCG priming. We have also compared AdCh68-based TB vaccine with its AdHu5 counterpart in both naive animals and those with preexisting anti-AdHu5 immunity in the lung. We provide compelling evidence that AdCh68-based TB vaccine is not only safe when delivered to the respiratory tract but, importantly, is also superior to its AdHu5 counterpart in induction of T-cell responses and immune protection, and limiting lung immunopathology in the presence of preexisting anti-AdHu5 immunity in the lung. Our findings thus suggest AdCh68-based TB vaccine to be an ideal candidate for respiratory mucosal immunization, endorsing its further clinical development in humans.
Collapse
Affiliation(s)
- M Jeyanathan
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - N Thanthrige-Don
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - S Afkhami
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - R Lai
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - D Damjanovic
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - A Zganiacz
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - X Feng
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - X-D Yao
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - K L Rosenthal
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - M Fe Medina
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - J Gauldie
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - H C Ertl
- Department of Immunology, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Z Xing
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
24
|
Dalmia N, Klimstra WB, Mason C, Ramsay AJ. DNA-Launched Alphavirus Replicons Encoding a Fusion of Mycobacterial Antigens Acr and Ag85B Are Immunogenic and Protective in a Murine Model of TB Infection. PLoS One 2015; 10:e0136635. [PMID: 26317509 PMCID: PMC4552820 DOI: 10.1371/journal.pone.0136635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/06/2015] [Indexed: 11/18/2022] Open
Abstract
There is an urgent need for effective prophylactic measures against Mycobacterium tuberculosis (Mtb) infection, particularly given the highly variable efficacy of Bacille Calmette-Guerin (BCG), the only licensed vaccine against tuberculosis (TB). Most studies indicate that cell-mediated immune responses involving both CD4+ and CD8+ T cells are necessary for effective immunity against Mtb. Genetic vaccination induces humoral and cellular immune responses, including CD4+ and CD8+ T-cell responses, against a variety of bacterial, viral, parasitic and tumor antigens, and this strategy may therefore hold promise for the development of more effective TB vaccines. Novel formulations and delivery strategies to improve the immunogenicity of DNA-based vaccines have recently been evaluated, and have shown varying degrees of success. In the present study, we evaluated DNA-launched Venezuelan equine encephalitis replicons (Vrep) encoding a novel fusion of the mycobacterial antigens α-crystallin (Acr) and antigen 85B (Ag85B), termed Vrep-Acr/Ag85B, for their immunogenicity and protective efficacy in a murine model of pulmonary TB. Vrep-Acr/Ag85B generated antigen-specific CD4+ and CD8+ T cell responses that persisted for at least 10 wk post-immunization. Interestingly, parenterally administered Vrep-Acr/Ag85B also induced T cell responses in the lung tissues, the primary site of infection, and inhibited bacterial growth in both the lungs and spleens following aerosol challenge with Mtb. DNA-launched Vrep may, therefore, represent an effective approach to the development of gene-based vaccines against TB, particularly as components of heterologous prime-boost strategies or as BCG boosters.
Collapse
MESH Headings
- Acyltransferases/genetics
- Acyltransferases/immunology
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Disease Models, Animal
- Encephalitis Virus, Venezuelan Equine/genetics
- Encephalitis Virus, Venezuelan Equine/immunology
- Immunity, Cellular
- Immunity, Humoral
- Mice
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Replicon/immunology
- Tuberculosis Vaccines/genetics
- Tuberculosis Vaccines/immunology
- Tuberculosis, Pulmonary/genetics
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/pathology
- Tuberculosis, Pulmonary/prevention & control
- Vaccination
- alpha-Crystallins/genetics
- alpha-Crystallins/immunology
Collapse
Affiliation(s)
- Neha Dalmia
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
| | - William B. Klimstra
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Carol Mason
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Alistair J. Ramsay
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
25
|
Khera AK, Afkhami S, Lai R, Jeyanathan M, Zganiacz A, Mandur T, Hammill J, Damjanovic D, Xing Z. Role of B Cells in Mucosal Vaccine-Induced Protective CD8+ T Cell Immunity against Pulmonary Tuberculosis. THE JOURNAL OF IMMUNOLOGY 2015; 195:2900-7. [PMID: 26268652 DOI: 10.4049/jimmunol.1500981] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/20/2015] [Indexed: 12/11/2022]
Abstract
Emerging evidence suggests a role of B cells in host defense against primary pulmonary tuberculosis (TB). However, the role of B cells in TB vaccine-induced protective T cell immunity still remains unknown. Using a viral-vectored model TB vaccine and a number of experimental approaches, we have investigated the role of B cells in respiratory mucosal vaccine-induced T cell responses and protection against pulmonary TB. We found that respiratory mucosal vaccination activated Ag-specific B cell responses. Whereas respiratory mucosal vaccination elicited Ag-specific T cell responses in the airway and lung interstitium of genetic B cell-deficient (Jh(-/-) knockout [KO]) mice, the levels of airway T cell responses were lower than in wild-type hosts, which were associated with suboptimal protection against pulmonary Mycobacterium tuberculosis challenge. However, mucosal vaccination induced T cell responses in the airway and lung interstitium and protection in B cell-depleted wild-type mice to a similar extent as in B cell-competent hosts. Furthermore, by using an adoptive cell transfer approach, reconstitution of B cells in vaccinated Jh(-/-) KO mice did not enhance anti-TB protection. Moreover, respiratory mucosal vaccine-activated T cells alone were able to enhance anti-TB protection in SCID mice, and the transfer of vaccine-primed B cells alongside T cells did not further enhance such protection. Alternatively, adoptively transferring vaccine-primed T cells from Jh(-/-) KO mice into SCID mice only provided suboptimal protection. These data together suggest that B cells play a minimal role, and highlight a central role by T cells, in respiratory mucosal vaccine-induced protective immunity against M. tuberculosis.
Collapse
Affiliation(s)
- Amandeep K Khera
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Sam Afkhami
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Rocky Lai
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Mangalakumari Jeyanathan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Anna Zganiacz
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Talveer Mandur
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Joni Hammill
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Daniela Damjanovic
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Zhou Xing
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
26
|
Jeyanathan M, Shao Z, Yu X, Harkness R, Jiang R, Li J, Xing Z, Zhu T. AdHu5Ag85A Respiratory Mucosal Boost Immunization Enhances Protection against Pulmonary Tuberculosis in BCG-Primed Non-Human Primates. PLoS One 2015; 10:e0135009. [PMID: 26252520 PMCID: PMC4529167 DOI: 10.1371/journal.pone.0135009] [Citation(s) in RCA: 46] [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: 05/14/2015] [Accepted: 07/10/2015] [Indexed: 12/21/2022] Open
Abstract
Persisting high global tuberculosis (TB) morbidity and mortality and poor efficacy of BCG vaccine emphasizes an urgent need for developing effective novel boost vaccination strategies following parenteral BCG priming in humans. Most of the current lead TB vaccine candidates in the global pipeline were developed for parenteral route of immunization. Compelling evidence indicates respiratory mucosal delivery of vaccine to be the most effective way to induce robust local mucosal protective immunity against pulmonary TB. However, despite ample supporting evidence from various animal models, there has been a lack of evidence supporting the safety and protective efficacy of respiratory mucosal TB vaccination in non-human primates (NHP) and humans. By using a rhesus macaque TB model we have evaluated the safety and protective efficacy of a recombinant human serotype 5 adenovirus-based TB vaccine (AdHu5Ag85A) delivered via the respiratory mucosal route. We show that mucosal AdHu5Ag85A boost immunization was safe and well tolerated in parenteral BCG-primed rhesus macaques. A single AdHu5Ag85A mucosal boost immunization in BCG-primed rhesus macaques enhanced the antigen–specific T cell responses. Boost immunization significantly improved the survival and bacterial control following M.tb challenge. Furthermore, TB-related lung pathology and clinical outcomes were lessened in BCG-primed, mucosally boosted animals compared to control animals. Thus, for the first time we show that a single respiratory mucosal boost immunization with a novel TB vaccine enhances protection against pulmonary TB in parenteral BCG-primed NHP. Our study provides the evidence for the protective potential of AdHu5Ag85A as a respiratory mucosal boost TB vaccine for human application.
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zhongqi Shao
- Tianjin CanSino Biotechnology, Inc., Tianjin, China
| | - Xuefeng Yu
- Tianjin CanSino Biotechnology, Inc., Tianjin, China
| | | | - Rong Jiang
- Tianjin CanSino Biotechnology, Inc., Tianjin, China
| | - Junqiang Li
- Tianjin CanSino Biotechnology, Inc., Tianjin, China
| | - Zhou Xing
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- * E-mail: (ZX); (TZ)
| | - Tao Zhu
- Tianjin CanSino Biotechnology, Inc., Tianjin, China
- * E-mail: (ZX); (TZ)
| |
Collapse
|
27
|
Andersen P, Urdahl KB. TB vaccines; promoting rapid and durable protection in the lung. Curr Opin Immunol 2015; 35:55-62. [PMID: 26113434 DOI: 10.1016/j.coi.2015.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 06/04/2015] [Accepted: 06/04/2015] [Indexed: 12/19/2022]
Abstract
TB vaccine discovery has focused on IFN-γ both for the selection of antigens and vaccine delivery strategies. Recent breakthroughs in our understanding of the requirements for immunological memory and the expression of immunity to TB in the lung now provide a framework for reconsidering that strategy. We will discuss the status of the TB vaccine field, recent insights into the role of central memory cells and the potential of tissue-resident memory cells in vaccine promoted protection against TB.
Collapse
Affiliation(s)
- Peter Andersen
- Statens Serum Institut, Infectious Disease Immunology, 5 Artillerivej, DK-2300 Copenhagen S, Denmark.
| | - Kevin B Urdahl
- Center for Infectious Disease Research (Formerly Seattle Biomedical Research Institute), Seattle, WA 98109, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
28
|
Damjanovic D, Khera A, Afkhami S, Lai R, Zganiacz A, Jeyanathan M, Xing Z. Age at Mycobacterium bovis BCG Priming Has Limited Impact on Anti-Tuberculosis Immunity Boosted by Respiratory Mucosal AdHu5Ag85A Immunization in a Murine Model. PLoS One 2015; 10:e0131175. [PMID: 26098423 PMCID: PMC4476612 DOI: 10.1371/journal.pone.0131175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/31/2015] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB) remains a global pandemic despite the use of Bacillus Calmette-Guérin (BCG) vaccine, partly because BCG fails to effectively control adult pulmonary TB. The introduction of novel boost vaccines such as the human Adenovirus 5-vectored AdHu5Ag85A could improve and prolong the protective immunity of BCG immunization. Age at which BCG immunization is implemented varies greatly worldwide, and research is ongoing to discover the optimal stage during childhood to administer the vaccine, as well as when to boost the immune response with potential novel vaccines. Using a murine model of subcutaneous BCG immunization followed by intranasal AdHu5Ag85A boosting, we investigated the impact of age at BCG immunization on protective efficacy of BCG prime and AdHu5Ag85A boost immunization-mediated protection. Our results showed that age at parenteral BCG priming has limited impact on the efficacy of BCG prime-AdHu5Ag85A respiratory mucosal boost immunization-enhanced protection. However, when BCG immunization was delayed until the maturity of the immune system, longer sustained memory T cells were generated and resulted in enhanced boosting effect on T cells of AdHu5Ag85A respiratory mucosal immunization. Our findings hold implications for the design of new TB immunization protocols for humans.
Collapse
Affiliation(s)
- Daniela Damjanovic
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Amandeep Khera
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rocky Lai
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
29
|
Quest for correlates of protection against tuberculosis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:258-66. [PMID: 25589549 DOI: 10.1128/cvi.00721-14] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A major impediment to tuberculosis (TB) vaccine development is the lack of reliable correlates of immune protection or biomarkers that would predict vaccine efficacy. Gamma interferon (IFN-γ) produced by CD4(+) T cells and, recently, multifunctional CD4(+) T cells secreting IFN-γ, tumor necrosis factor (TNF), and interleukin-2 (IL-2) have been used in vaccine studies as a measurable immune parameter, reflecting activity of a vaccine and potentially predicting protection. However, accumulating experimental evidence suggests that host resistance against Mycobacterium tuberculosis infection is independent of IFN-γ and TNF secretion from CD4(+) T cells. Furthermore, the booster vaccine MVA85A, despite generating a high level of multifunctional CD4(+) T cell response in the host, failed to confer enhanced protection in vaccinated subjects. These findings suggest the need for identifying reliable correlates of protection to determine the efficacy of TB vaccine candidates. This article focuses on alternative pathways that mediate M. tuberculosis control and their potential for serving as markers of protection. The review also discusses the significance of investigating the natural human immune response to M. tuberculosis to identify the correlates of protection in vaccination.
Collapse
|
30
|
Abstract
Pulmonary TB remains a leading global health issue, but the current Bacille Calmette-Guérin (BCG) vaccine fails to control it effectively. Much effort has gone into developing safe and effective boost vaccine candidates for use after the BCG prime vaccination. To date, almost all the lead candidates are being evaluated clinically via a parenteral route. Abundant experimental evidence suggests that parenteral boosting with a virus-based vaccine is much less effective than respiratory mucosal boosting, because the former fails to activate a type of T cell capable of rapidly transmigrating into the airway luminal space in the early phase of the Mycobacterium tuberculosis infection. The next few years will determine whether parenteral boosting with some of the lead vaccine candidates, particularly the protein-based vaccines, improves protection in humans over that by BCG. Much effort is needed to develop respiratory mucosal boost vaccines and to identify the reliable immune protective correlates in humans.
Collapse
Affiliation(s)
- Zhou Xing
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Fiona Smaill
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
31
|
Kaveh DA, Garcia-Pelayo MC, Hogarth PJ. Persistent BCG bacilli perpetuate CD4 T effector memory and optimal protection against tuberculosis. Vaccine 2014; 32:6911-6918. [PMID: 25444816 DOI: 10.1016/j.vaccine.2014.10.041] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/17/2014] [Accepted: 10/25/2014] [Indexed: 10/24/2022]
Abstract
Tuberculosis (TB) remains one of the most important infectious diseases of man and animals, and the only available vaccine (BCG) requires urgent replacement or improvement. To facilitate this, the protective mechanisms induced by BCG require further understanding. As a live attenuated vaccine, persistence of BCG bacilli in the host may be a crucial mechanism. We have investigated the long term persistence of BCG following vaccination and the influence on the induced immune response and protection, using an established murine model. We sought to establish whether previously identified BCG-specific CD4 TEM cells represent genuine long-lived memory cells of a relatively high frequency, or are a consequence of continual priming by chronically persistent BCG vaccine bacilli. By clearing persistent bacilli, we have compared immune responses (spleen and lung CD4: cytokine producing T effector/TEM; TCR-specific) and BCG-induced protection, in the presence and absence of these persisting vaccine bacilli. Viable BCG bacilli persisted for at least 16 months post-vaccination, associated with specific CD4 T effector/TEM and tetramer-specific responses. Clearing these bacilli abrogated all BCG-specific CD4 T cells whilst only reducing protection by 1log10. BCG may induce two additive mechanisms of immunity: (i) dependant on the presence of viable bacilli and TEM; and (ii) independent of these factors. These data have crucial implications on the rational generation of replacement TB vaccines, and the interpretation of BCG induced immunity in animal models.
Collapse
Affiliation(s)
- Daryan A Kaveh
- Vaccine Immunology Team, Department of Bacteriology, Animal and Plant Health Agency (APHA) Addlestone KT15 3NB, Surrey, UK
| | - M C Garcia-Pelayo
- Vaccine Immunology Team, Department of Bacteriology, Animal and Plant Health Agency (APHA) Addlestone KT15 3NB, Surrey, UK
| | - Philip J Hogarth
- Vaccine Immunology Team, Department of Bacteriology, Animal and Plant Health Agency (APHA) Addlestone KT15 3NB, Surrey, UK.
| |
Collapse
|
32
|
Trivedi S, Jackson RJ, Ranasinghe C. Different HIV pox viral vector-based vaccines and adjuvants can induce unique antigen presenting cells that modulate CD8 T cell avidity. Virology 2014; 468-470:479-489. [PMID: 25261870 DOI: 10.1016/j.virol.2014.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/04/2014] [Accepted: 09/06/2014] [Indexed: 11/25/2022]
Abstract
The lung-derived dendritic cell (LDC) recruitment following intranasal (i.n.) vaccination of different poxviral vector-based vaccines/adjuvants were evaluated to decipher how these factors influenced CD8(+) T cell avidity. Compared to the standard i.n. recombinant fowlpox virus (FPV)-HIV vaccination, the FPV-HIV IL-13Rα2 or IL-4Rα antagonist adjuvanted vaccines that induced higher avidity CD8(+) T cells, also recruited significantly elevated MHCII(+) CD11c(+) CD11b(+) CD103(-) CD64(-) MAR-1(-) conventional DC (cDCs) to the lung mucosae (hierarchy: IL-4R antagonist>IL-13Rα2>unadjuvanted). In contrast, elevated CD11b(-) CD103(+) LDCs were detected in animals that received recombinant HIV vaccinia virus (rVV) or Modified Vaccinia Ankara virus (MVA) vector-based vaccines. Adoptive transfer studies indicated that CD11b(-) CD103(+) LDCs significantly dampened HIV-specific CD8(+) T cell avidity compared to CD11b(+) CD103(-) LDCs. Collectively; our observations revealed that rFPV vector prime and transient inhibition of IL-4/IL-13 at the vaccination site favoured the recruitment of unique LDCs, associated with the induction of high quality immunity.
Collapse
Affiliation(s)
- Shubhanshi Trivedi
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia.
| | - Ronald J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| |
Collapse
|
33
|
Yoshida O, Kimura S, Dou L, Matta B, Yokota S, Stolz D, Geller D, Thomson AW. DAP12 deficiency in liver allografts results in enhanced donor DC migration, augmented effector T cell responses and abrogation of transplant tolerance. Am J Transplant 2014; 14:1791-805. [PMID: 24935196 PMCID: PMC4107008 DOI: 10.1111/ajt.12757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/28/2014] [Accepted: 03/19/2014] [Indexed: 01/25/2023]
Abstract
Liver interstitial dendritic cells (DC) have been implicated in immune regulation and tolerance induction. We found that the transmembrane immuno-adaptor DNAX-activating protein of 12 kDa (DAP12) negatively regulated conventional liver myeloid (m) DC maturation and their in vivo migratory and T cell allostimulatory ability. Livers were transplanted from C57BL/6(H2(b) ) (B6) WT or DAP12(-/-) mice into WT C3H (H2(k) ) recipients. Donor mDC (H2-K(b+) CD11c(+) ) were quantified in spleens by flow cytometry. Anti-donor T cell reactivity was evaluated by ex vivo carboxyfluorescein diacetate succinimidyl ester-mixed leukocyte reaction and delayed-type hypersensitivity responses, while T effector and regulatory T cells were determined by flow analysis. A threefold to fourfold increase in donor-derived DC was detected in spleens of DAP12(-/-) liver recipients compared with those given WT grafts. Moreover, pro-inflammatory cytokine gene expression in the graft, interferon gamma (IFNγ) production by graft-infiltrating CD8(+) T cells and systemic levels of IFNγ were all elevated significantly in DAP12(-/-) liver recipients. DAP12(-/-) grafts also exhibited reduced incidences of CD4(+) Foxp3(+) cells and enhanced CD8(+) T cell IFNγ secretion in response to donor antigen challenge. Unlike WT grafts, DAP12(-/-) livers failed to induce tolerance and were rejected acutely. Thus, DAP12 expression in liver grafts regulates donor mDC migration to host lymphoid tissue, alloreactive T cell responses and transplant tolerance.
Collapse
Affiliation(s)
- O. Yoshida
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - S. Kimura
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Dou
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Hepatic Surgery Center, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - B.M. Matta
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - S. Yokota
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D.B. Stolz
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D.A. Geller
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA,Corresponding author: Angus W. Thomson PhD DSc Starzl Transplantation Institute University of Pittsburgh School of Medicine 200 Lothrop Street, W1540 BST Pittsburgh, PA 15261
| |
Collapse
|
34
|
Beverley PCL, Ruzsics Z, Hey A, Hutchings C, Boos S, Bolinger B, Marchi E, O'Hara G, Klenerman P, Koszinowski UH, Tchilian EZ. A novel murine cytomegalovirus vaccine vector protects against Mycobacterium tuberculosis. THE JOURNAL OF IMMUNOLOGY 2014; 193:2306-16. [PMID: 25070842 PMCID: PMC4134927 DOI: 10.4049/jimmunol.1302523] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis remains a global health problem so that a more effective vaccine than bacillus Calmette–Guérin is urgently needed. Cytomegaloviruses persist lifelong in vivo and induce powerful immune and increasing (“inflationary”) responses, making them attractive vaccine vectors. We have used an m1–m16-deleted recombinant murine CMV (MCMV) expressing Mycobacterium tuberculosis Ag 85A to show that infection of mice with this recombinant significantly reduces the mycobacterial load after challenge with M. tuberculosis, whereas control empty virus has a lesser effect. Both viruses induce immune responses to H-2d–restricted epitopes of MCMV pp89 and M18 Ags characteristic of infection with other MCMVs. A low frequency of 85A-specific memory cells could be revealed by in vivo or in vitro boosting or after challenge with M. tuberculosis. Kinetic analysis of M. tuberculosis growth in the lungs of CMV-infected mice shows early inhibition of M. tuberculosis growth abolished by treatment with NK-depleting anti–asialo ganglio-N-tetraosylceramide Ab. Microarray analysis of the lungs of naive and CMV-infected mice shows increased IL-21 mRNA in infected mice, whereas in vitro NK assays indicate increased levels of NK activity. These data indicate that activation of NK cells by MCMV provides early nonspecific protection against M. tuberculosis, potentiated by a weak 85A-specific T cell response, and they reinforce the view that the innate immune system plays an important role in both natural and vaccine-induced protection against M. tuberculosis.
Collapse
Affiliation(s)
- Peter C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Zsolt Ruzsics
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Ariann Hey
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Claire Hutchings
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Simone Boos
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Beatrice Bolinger
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Emanuele Marchi
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Geraldine O'Hara
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Ulrich H Koszinowski
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Elma Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| |
Collapse
|
35
|
Marinova D, Gonzalo-Asensio J, Aguilo N, Martin C. Recent developments in tuberculosis vaccines. Expert Rev Vaccines 2014; 12:1431-48. [PMID: 24195481 DOI: 10.1586/14760584.2013.856765] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Substantial efforts have been made over the past decade to develop vaccines against tuberculosis. We review recent developments in tuberculosis vaccines in the global portfolio, including those designed for use in a prophylactic setting, either alone or as boosts to Bacille Calmette-Guérin, and therapeutic vaccines designed to improve chemotherapy. While there is no doubt that progress is still being made, there are limitations to our animal model screening processes, which are further amplified by the lack of understanding of the immunological responses involved and the precise type of long-lived immunity that new vaccines need to induce. The challenge ahead is to optimize the planning for advanced clinical trials in poor endemic settings, which could be greatly facilitated by identifying correlates of protection.
Collapse
Affiliation(s)
- Dessislava Marinova
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009 Zaragoza, Spain
| | | | | | | |
Collapse
|
36
|
Lai R, Jeyanathan M, Shaler CR, Damjanovic D, Khera A, Horvath C, Ashkar AA, Xing Z. Restoration of innate immune activation accelerates Th1-cell priming and protection following pulmonary mycobacterial infection. Eur J Immunol 2014; 44:1375-86. [PMID: 24519467 DOI: 10.1002/eji.201344300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/12/2014] [Accepted: 02/06/2014] [Indexed: 02/06/2023]
Abstract
The immune mechanisms underlying delayed induction of Th1-type immunity in the lungs following pulmonary mycobacterial infection remain poorly understood. We have herein investigated the underlying immune mechanisms for such delayed responses and whether a selected innate immune-modulating strategy can accelerate Th1-type responses. We have found that, in the early stage of pulmonary infection with attenuated Mycobacterium tuberculosis (M.tb H37Ra), the levels of infection in the lung continue to increase logarithmically until days 14 and 21 postinfection in C57BL/6 mice. The activation of innate immune responses, particularly DCs, in the lung is delayed. This results in a delay in the subsequent downstream immune responses including the migration of antigen-bearing DCs to the draining lymph node (dLN), the Th1-cell priming in dLN, and the recruitment of Th1 cells to the lung. However, single lung mucosal exposure to the TLR agonist FimH postinfection is able to accelerate protective Th1-type immunity via facilitating DC migration to the lung and draining lymph nodes, enhancing DC antigen presentation and Th1-cell priming. These findings hold implications for the development of immunotherapeutic and vaccination strategies and suggest that enhancement of early innate immune activation is a viable option for improving Th1-type immunity against pulmonary mycobacterial diseases.
Collapse
Affiliation(s)
- Rocky Lai
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Beverley PCL, Sridhar S, Lalvani A, Tchilian EZ. Harnessing local and systemic immunity for vaccines against tuberculosis. Mucosal Immunol 2014; 7:20-6. [PMID: 24253104 DOI: 10.1038/mi.2013.99] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/09/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
Abstract
The lung is the portal of entry for Mycobacterium tuberculosis (Mtb) and animal experimental evidence indicates that local immune defense mechanisms are crucial for protective immunity. Immunization via the lower respiratory tract efficiently induces a dividing, activated, antigen-dependent, lung-resident, memory T-cell population, which is partly recoverable by bronchoalveolar lavage. These cells can inhibit the growth of Mtb in the lungs immediately after infection. Delivery of appropriate signals to the lung innate immune system is critical for induction of effective local immunity. In contrast after parenteral immunization, antigen-specific cells may be found in lung tissue but few are recoverable by lavage and inhibition of mycobacterial growth is delayed. Harnessing both local and systemic immunity can provide highly effective protection in animal models and the evidence suggests that taken in aggregate, multiple animal models may predict the success of novel vaccine strategies in humans.
Collapse
Affiliation(s)
- P C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S Sridhar
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - A Lalvani
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - E Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Tchilian E, Ahuja D, Hey A, Jiang S, Beverley P. Immunization with different formulations of Mycobacterium tuberculosis antigen 85A induces immune responses with different specificity and protective efficacy. Vaccine 2013; 31:4624-31. [PMID: 23896422 PMCID: PMC3898716 DOI: 10.1016/j.vaccine.2013.07.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/25/2013] [Accepted: 07/16/2013] [Indexed: 11/21/2022]
Abstract
Immunization intranasally with Mtb antigen 85A is more protective than parenterally. Three 85A vaccines platforms induce responses with differing epitope specificity. Responses to the CD8 85A70–78 but not the CD8 85A145–152 epitope are protective.
To test the relative efficacy of CD4 and CD8T cells in mediating protective immunity to Mycobacterium tuberculosis (Mtb), we compared three immunization regimes designed to induce preferentially each subset. BALB/c mice were immunized intranasally (i.n.) or parenterally with antigen 85A either in a recombinant Adenoviral vector (Ad85A), as recombinant protein (r85A) or as a set of overlapping 15mer peptides (p85A). For the first time we show that i.n. immunization with overlapping 85A synthetic peptides as well as Ad85A or r85A can provide protection against Mtb challenge. For all forms of the antigen, i.n. induces greater protection against Mtb challenge than parenteral immunization. Ad85A induces a predominantly CD8T cell response against the 85A70–78 epitope, r85A a CD4 response to 85A99–118 and p85A a balanced CD4/CD8 response to the CD4 85A99–118 and CD8 85A145–152 epitopes. Immune responses to CD4 85A99–118 and CD8 85A70–78 but not CD8 85A145–152 are protective. Although Ad85A induces a strong response to the protective CD8 85A70–78 epitope, we could not induce any response to this epitope by peptide immunization. These results show that although peptide immunization can induce protective immunity to Mtb challenge, it can also induce a response to a non-protective epitope in antigen 85A, indicating that the specificity of an immune response may be more important for protection against Mtb than its magnitude. These findings have important implications for the application of such vaccines in humans.
Collapse
Affiliation(s)
- Elma Tchilian
- University of Oxford, The Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK.
| | | | | | | | | |
Collapse
|