1
|
Xu G, Li Y, Lu G, Xie D. Tissue-resident memory T cells in urinary tract diseases. Front Immunol 2025; 16:1535930. [PMID: 40066439 PMCID: PMC11891219 DOI: 10.3389/fimmu.2025.1535930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are a specialized subset of memory T cells that permanently reside in non-lymphoid tissues, providing localized and long-lasting immune protection. In the urinary tract, TRM cells play critical roles in defending against infections, mediating tumor immunity, and influencing the pathogenesis of chronic inflammatory diseases. Their therapeutic potential is immense, with promising avenues for vaccine development, enhanced cancer immunotherapy, and targeted treatments for chronic inflammation. However, challenges remain in harnessing their protective roles while minimizing their pathological effects, particularly in immunosuppressive or inflammatory microenvironments. This review explores the diverse roles of TRM cells in urinary tract diseases, including infections, cancer, and chronic inflammation, and discusses therapeutic strategies and future directions for leveraging TRM cells to improve clinical outcomes. By advancing our understanding of TRM cell biology, we can develop innovative interventions that balance their immune-protective and regulatory functions.
Collapse
Affiliation(s)
- Guofeng Xu
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuying Li
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Respiratory Critical Care, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guanting Lu
- Laboratory of Translational Medicine Research, Deyang People’s Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Daoyuan Xie
- Laboratory of Translational Medicine Research, Deyang People’s Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| |
Collapse
|
2
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
3
|
Wu L, Xu W, Jiang H, Yang M, Cun D. Respiratory delivered vaccines: Current status and perspectives in rational formulation design. Acta Pharm Sin B 2024; 14:5132-5160. [PMID: 39807330 PMCID: PMC11725141 DOI: 10.1016/j.apsb.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 08/18/2024] [Indexed: 01/16/2025] Open
Abstract
The respiratory tract is susceptible to various infections and can be affected by many serious diseases. Vaccination is one of the most promising ways that prevent infectious diseases and treatment of some diseases such as malignancy. Direct delivery of vaccines to the respiratory tract could mimic the natural process of infection and shorten the delivery path, therefore unique mucosal immunity at the first line might be induced and the efficiency of delivery can be high. Despite considerable attempts at the development of respiratory vaccines, the rational formulation design still warrants attention, i.e., how the formulation composition, particle properties, formulation type (liquid or solid), and devices would influence the immune outcome. This article reviews the recent advances in the formulation design and development of respiratory vaccines. The focus is on the state of the art of delivering antigenic compounds through the respiratory tract, overcoming the pulmonary bio-barriers, enhancing delivery efficiencies of respiratory vaccines as well as maintaining the stability of vaccines during storage and use. The choice of devices and the influence of deposition sites on vaccine efficiencies were also reviewed.
Collapse
Affiliation(s)
- Lan Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Wenwen Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Huiyang Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- School of Food and Drug, Shenzhen Polytechnic University, China, Shenzhen 518055, China
| |
Collapse
|
4
|
Mosmann TR, McMichael AJ, LeVert A, McCauley JW, Almond JW. Opportunities and challenges for T cell-based influenza vaccines. Nat Rev Immunol 2024; 24:736-752. [PMID: 38698082 DOI: 10.1038/s41577-024-01030-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2024] [Indexed: 05/05/2024]
Abstract
Vaccination remains our main defence against influenza, which causes substantial annual mortality and poses a serious pandemic threat. Influenza virus evades immunity by rapidly changing its surface antigens but, even when the vaccine is well matched to the current circulating virus strains, influenza vaccines are not as effective as many other vaccines. Influenza vaccine development has traditionally focused on the induction of protective antibodies, but there is mounting evidence that T cell responses are also protective against influenza. Thus, future vaccines designed to promote both broad T cell effector functions and antibodies may provide enhanced protection. As we discuss, such vaccines present several challenges that require new strategic and economic considerations. Vaccine-induced T cells relevant to protection may reside in the lungs or lymphoid tissues, requiring more invasive assays to assess the immunogenicity of vaccine candidates. T cell functions may contain and resolve infection rather than completely prevent infection and early illness, requiring vaccine effectiveness to be assessed based on the prevention of severe disease and death rather than symptomatic infection. It can be complex and costly to measure T cell responses and infrequent clinical outcomes, and thus innovations in clinical trial design are needed for economic reasons. Nevertheless, the goal of more effective influenza vaccines justifies renewed and intensive efforts.
Collapse
Affiliation(s)
- Tim R Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| | - Andrew J McMichael
- Centre for Immuno-Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | | | | | - Jeffrey W Almond
- The Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Macedo BG, Masuda MY, Borges da Silva H. Location versus ID: what matters to lung-resident memory T cells? Front Immunol 2024; 15:1355910. [PMID: 38375476 PMCID: PMC10875077 DOI: 10.3389/fimmu.2024.1355910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
Tissue-resident memory T cells (TRM cells) are vital for the promotion of barrier immunity. The lung, a tissue constantly exposed to foreign pathogenic or non-pathogenic antigens, is not devoid of these cells. Lung TRM cells have been considered major players in either the protection against respiratory viral infections or the pathogenesis of lung allergies. Establishment of lung TRM cells rely on intrinsic and extrinsic factors. Among the extrinsic regulators of lung TRM cells, the magnitude of the impact of factors such as the route of antigen entry or the antigen natural tropism for the lung is not entirely clear. In this perspective, we provide a summary of the literature covering this subject and present some preliminary results on this potential dichotomy between antigen location versus antigen type. Finally, we propose a hypothesis to synthesize the potential contributions of these two variables for lung TRM cell development.
Collapse
|
6
|
Mittra S, Harding SM, Kaech SM. Memory T Cells in the Immunoprevention of Cancer: A Switch from Therapeutic to Prophylactic Approaches. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:907-916. [PMID: 37669503 PMCID: PMC10491418 DOI: 10.4049/jimmunol.2300049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/24/2023] [Indexed: 09/07/2023]
Abstract
Cancer immunoprevention, the engagement of the immune system to prevent cancer, is largely overshadowed by therapeutic approaches to treating cancer after detection. Vaccines or, alternatively, the utilization of genetically engineered memory T cells could be methods of engaging and creating cancer-specific T cells with superb memory, lenient activation requirements, potent antitumor cytotoxicity, tumor surveillance, and resilience against immunosuppressive factors in the tumor microenvironment. In this review we analyze memory T cell subtypes based on their potential utility in cancer immunoprevention with regard to longevity, localization, activation requirements, and efficacy in fighting cancers. A particular focus is on how both tissue-resident memory T cells and stem memory T cells could be promising subtypes for engaging in immunoprevention.
Collapse
Affiliation(s)
- Siddhesh Mittra
- University of Toronto Schools, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shane M. Harding
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Departments of Radiation Oncology and Immunology, University of Toronto; Toronto, Canada
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
7
|
Longet S, Paul S. Pivotal role of tissue-resident memory lymphocytes in the control of mucosal infections: can mucosal vaccination induce protective tissue-resident memory T and B cells? Front Immunol 2023; 14:1216402. [PMID: 37753095 PMCID: PMC10518612 DOI: 10.3389/fimmu.2023.1216402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Affiliation(s)
- Stephanie Longet
- Centre International de Recherche en Infectiologie, Team Groupe sur l'immunité des muqueuses et agents pathogènes (GIMAP), Université Jean Monnet, Université Claude Bernard Lyon, Inserm, Saint-Etienne, France
| | - Stephane Paul
- Centre International de Recherche en Infectiologie, Team Groupe sur l'immunité des muqueuses et agents pathogènes (GIMAP), Université Jean Monnet, Université Claude Bernard Lyon, Inserm, Saint-Etienne, France
- Centre d'investigation clinique (CIC) 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
8
|
Suberi A, Grun MK, Mao T, Israelow B, Reschke M, Grundler J, Akhtar L, Lee T, Shin K, Piotrowski-Daspit AS, Homer RJ, Iwasaki A, Suh HW, Saltzman WM. Polymer nanoparticles deliver mRNA to the lung for mucosal vaccination. Sci Transl Med 2023; 15:eabq0603. [PMID: 37585505 PMCID: PMC11137749 DOI: 10.1126/scitranslmed.abq0603] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/26/2023] [Indexed: 08/18/2023]
Abstract
An inhalable platform for messenger RNA (mRNA) therapeutics would enable minimally invasive and lung-targeted delivery for a host of pulmonary diseases. Development of lung-targeted mRNA therapeutics has been limited by poor transfection efficiency and risk of vehicle-induced pathology. Here, we report an inhalable polymer-based vehicle for delivery of therapeutic mRNAs to the lung. We optimized biodegradable poly(amine-co-ester) (PACE) polyplexes for mRNA delivery using end-group modifications and polyethylene glycol. These polyplexes achieved high transfection of mRNA throughout the lung, particularly in epithelial and antigen-presenting cells. We applied this technology to develop a mucosal vaccine for severe acute respiratory syndrome coronavirus 2 and found that intranasal vaccination with spike protein-encoding mRNA polyplexes induced potent cellular and humoral adaptive immunity and protected susceptible mice from lethal viral challenge. Together, these results demonstrate the translational potential of PACE polyplexes for therapeutic delivery of mRNA to the lungs.
Collapse
Affiliation(s)
- Alexandra Suberi
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Molly K Grun
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06511, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Melanie Reschke
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Julian Grundler
- Department of Chemistry, Yale University, New Haven, CT 06511, USA
| | - Laiba Akhtar
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06511, USA
| | - Teresa Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | | | - Robert J Homer
- Department of Pathology, Yale University School of Medicine, CT 06510, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06511, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
9
|
Humphries DC, O’Connor RA, Stewart HL, Quinn TM, Gaughan EE, Mills B, Williams GO, Stone JM, Finlayson K, Chabaud-Riou M, Boudet F, Dhaliwal K, Pavot V. Specific in situ immuno-imaging of pulmonary-resident memory lymphocytes in human lungs. Front Immunol 2023; 14:1100161. [PMID: 36845117 PMCID: PMC9951616 DOI: 10.3389/fimmu.2023.1100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Pulmonary-resident memory T cells (TRM) and B cells (BRM) orchestrate protective immunity to reinfection with respiratory pathogens. Developing methods for the in situ detection of these populations would benefit both research and clinical settings. Methods To address this need, we developed a novel in situ immunolabelling approach combined with clinic-ready fibre-based optical endomicroscopy (OEM) to detect canonical markers of lymphocyte tissue residency in situ in human lungs undergoing ex vivo lung ventilation (EVLV). Results Initially, cells from human lung digests (confirmed to contain TRM/BRM populations using flow cytometry) were stained with CD69 and CD103/CD20 fluorescent antibodies and imaged in vitro using KronoScan, demonstrating it's ability to detect antibody labelled cells. We next instilled these pre-labelled cells into human lungs undergoing EVLV and confirmed they could still be visualised using both fluorescence intensity and lifetime imaging against background lung architecture. Finally, we instilled fluorescent CD69 and CD103/CD20 antibodies directly into the lung and were able to detect TRM/BRM following in situ labelling within seconds of direct intra-alveolar delivery of microdoses of fluorescently labelled antibodies. Discussion In situ, no wash, immunolabelling with intra-alveolar OEM imaging is a novel methodology with the potential to expand the experimental utility of EVLV and pre-clinical models.
Collapse
Affiliation(s)
- Duncan C. Humphries
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom,Research & Development, Sanofi, Marcy L’Etoile, France
| | - Richard A. O’Connor
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hazel L. Stewart
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tom M. Quinn
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Erin E. Gaughan
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Beth Mills
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gareth O.S. Williams
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - James M. Stone
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom,Centre for Photonic and Physics, Bath University, Bath, United Kingdom
| | - Keith Finlayson
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Kevin Dhaliwal
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom,*Correspondence: Kevin Dhaliwal, ; Vincent Pavot,
| | - Vincent Pavot
- Research & Development, Sanofi, Marcy L’Etoile, France,*Correspondence: Kevin Dhaliwal, ; Vincent Pavot,
| |
Collapse
|
10
|
Pellegrini JM, Gorvel JP, Mémet S. Immunosuppressive Mechanisms in Brucellosis in Light of Chronic Bacterial Diseases. Microorganisms 2022; 10:1260. [PMID: 35888979 PMCID: PMC9324529 DOI: 10.3390/microorganisms10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Brucellosis is considered one of the major zoonoses worldwide, constituting a critical livestock and human health concern with a huge socio-economic burden. Brucella genus, its etiologic agent, is composed of intracellular bacteria that have evolved a prodigious ability to elude and shape host immunity to establish chronic infection. Brucella's intracellular lifestyle and pathogen-associated molecular patterns, such as its specific lipopolysaccharide (LPS), are key factors for hiding and hampering recognition by the immune system. Here, we will review the current knowledge of evading and immunosuppressive mechanisms elicited by Brucella species to persist stealthily in their hosts, such as those triggered by their LPS and cyclic β-1,2-d-glucan or involved in neutrophil and monocyte avoidance, antigen presentation impairment, the modulation of T cell responses and immunometabolism. Attractive strategies exploited by other successful chronic pathogenic bacteria, including Mycobacteria, Salmonella, and Chlamydia, will be also discussed, with a special emphasis on the mechanisms operating in brucellosis, such as granuloma formation, pyroptosis, and manipulation of type I and III IFNs, B cells, innate lymphoid cells, and host lipids. A better understanding of these stratagems is essential to fighting bacterial chronic infections and designing innovative treatments and vaccines.
Collapse
|
11
|
Files MA, Naqvi KF, Saito TB, Clover TM, Rudra JS, Endsley JJ. Self-adjuvanting nanovaccines boost lung-resident CD4 + T cell immune responses in BCG-primed mice. NPJ Vaccines 2022; 7:48. [PMID: 35474079 PMCID: PMC9043212 DOI: 10.1038/s41541-022-00466-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/21/2022] [Indexed: 01/04/2023] Open
Abstract
Heterologous vaccine regimens could extend waning protection in the global population immunized with Mycobacterium bovis Bacille Calmette-Guerin (BCG). We demonstrate that pulmonary delivery of peptide nanofibers (PNFs) bearing an Ag85B CD4+ T cell epitope increased the frequency of antigen-specific T cells in BCG-primed mice, including heterogenous populations with tissue resident memory (Trm) and effector memory (Tem) phenotype, and functional cytokine recall. Adoptive transfer of dendritic cells pulsed with Ag85B-bearing PNFs further expanded the frequency and functional repertoire of memory CD4+ T cells. Transcriptomic analysis suggested that the adjuvanticity of peptide nanofibers is, in part, due to the release of damage-associated molecular patterns. A single boost with monovalent Ag85B PNF in BCG-primed mice did not reduce lung bacterial burden compared to BCG alone following aerosol Mtb challenge. These findings support the need for novel BCG booster strategies that activate pools of Trm cells with potentially diverse localization, trafficking, and immune function.
Collapse
Grants
- R01 AI130278 NIAID NIH HHS
- R21 AI115302 NIAID NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- Predoctoral Fellowship, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas 77555
- Predoctoral Fellowship, James W. McLaughlin Endowment, University of Texas Medical Branch, Galveston, Texas, 77555
- Washington University McKelvey School of Engineering, Department of Biomedical Engineering Commitment Funds (12-360-94361J)
Collapse
Affiliation(s)
- Megan A Files
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kubra F Naqvi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tais B Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Tara M Clover
- Comprehensive Industrial Hygiene Laboratory (CIHL), Navy Environmental and Preventive Medicine Unit TWO (NEPMU-2), Department of the Navy, Norfolk, VA, 23551, USA
| | - Jai S Rudra
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
12
|
Garland KM, Sheehy TL, Wilson JT. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem Rev 2022; 122:5977-6039. [PMID: 35107989 PMCID: PMC8994686 DOI: 10.1021/acs.chemrev.1c00750] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The stimulator of interferon genes (STING) cellular signaling pathway is a promising target for cancer immunotherapy. Activation of the intracellular STING protein triggers the production of a multifaceted array of immunostimulatory molecules, which, in the proper context, can drive dendritic cell maturation, antitumor macrophage polarization, T cell priming and activation, natural killer cell activation, vascular reprogramming, and/or cancer cell death, resulting in immune-mediated tumor elimination and generation of antitumor immune memory. Accordingly, there is a significant amount of ongoing preclinical and clinical research toward further understanding the role of the STING pathway in cancer immune surveillance as well as the development of modulators of the pathway as a strategy to stimulate antitumor immunity. Yet, the efficacy of STING pathway agonists is limited by many drug delivery and pharmacological challenges. Depending on the class of STING agonist and the desired administration route, these may include poor drug stability, immunocellular toxicity, immune-related adverse events, limited tumor or lymph node targeting and/or retention, low cellular uptake and intracellular delivery, and a complex dependence on the magnitude and kinetics of STING signaling. This review provides a concise summary of the STING pathway, highlighting recent biological developments, immunological consequences, and implications for drug delivery. This review also offers a critical analysis of an expanding arsenal of chemical strategies that are being employed to enhance the efficacy, safety, and/or clinical utility of STING pathway agonists and lastly draws attention to several opportunities for therapeutic advancements.
Collapse
Affiliation(s)
- Kyle M Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - Taylor L Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
| |
Collapse
|
13
|
Suberi A, Grun MK, Mao T, Israelow B, Reschke M, Grundler J, Akhtar L, Lee T, Shin K, Piotrowski-Daspit AS, Homer RJ, Iwasaki A, Suh HW, Saltzman WM. Inhalable polymer nanoparticles for versatile mRNA delivery and mucosal vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.22.485401. [PMID: 35350207 PMCID: PMC8963702 DOI: 10.1101/2022.03.22.485401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
An inhalable platform for mRNA therapeutics would enable minimally invasive and lung targeted delivery for a host of pulmonary diseases. Development of lung targeted mRNA therapeutics has been limited by poor transfection efficiency and risk of vehicle-induced pathology. Here we report an inhalable polymer-based vehicle for delivery of therapeutic mRNAs to the lung. We optimized biodegradable poly(amine-co-ester) polyplexes for mRNA delivery using end group modifications and polyethylene glycol. Our polyplexes achieved high transfection of mRNA throughout the lung, particularly in epithelial and antigen-presenting cells. We applied this technology to develop a mucosal vaccine for SARS-CoV-2. Intranasal vaccination with spike protein mRNA polyplexes induced potent cellular and humoral adaptive immunity and protected K18-hACE2 mice from lethal viral challenge. One-sentence summary Inhaled polymer nanoparticles (NPs) achieve high mRNA expression in the lung and induce protective immunity against SARS-CoV-2.
Collapse
|
14
|
McFall-Boegeman H, Huang X. Mechanisms of cellular and humoral immunity through the lens of VLP-based vaccines. Expert Rev Vaccines 2022; 21:453-469. [PMID: 35023430 PMCID: PMC8960355 DOI: 10.1080/14760584.2022.2029415] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Vaccination can be effective defense against many infectious agents and the corresponding diseases. Discoveries elucidating the mechanisms of the immune system have given hopes to developing vaccines against diseases recalcitrant to current treatment/prevention strategies. One such finding is the ability of immunogenic biological nanoparticles to powerfully boost the immunogenicity of poorer antigens conjugated to them with virus-like particle (VLP)-based vaccines as a key example. VLPs take advantage of the well-defined molecular structures associated with sub-unit vaccines and the immunostimulatory nature of conjugate vaccines. AREAS COVERED In this review, we will discuss how advances in understanding the immune system can inform VLP-based vaccine design and how VLP-based vaccines have uncovered underlying mechanisms in the immune system. EXPERT OPINION As our understanding of mechanisms underlying the immune system increases, that knowledge should inform our vaccine design. Testing of proof-of-concept vaccines in the lab should seek to elucidate the underlying mechanisms of immune responses. The integration of these approaches will allow for VLP-based vaccines to live up to their promise as a powerful plug-and-play platform for next generation vaccine development.
Collapse
Affiliation(s)
- Hunter McFall-Boegeman
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
15
|
Zheng MZM, Wakim LM. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022; 15:379-388. [PMID: 34671115 PMCID: PMC8526531 DOI: 10.1038/s41385-021-00461-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/04/2023]
Abstract
Owing to their capacity to rapidly spread across the population, airborne pathogens represent a significant risk to global health. Indeed, several of the past major global pandemics have been instigated by respiratory pathogens. A greater understanding of the immune cells tasked with protecting the airways from infection will allow for the development of strategies that curb the spread and impact of these airborne diseases. A specific subset of memory T-cell resident in both the upper and lower respiratory tract, termed tissue-resident memory (Trm), have been shown to play an instrumental role in local immune responses against a wide breadth of both viral and bacterial infections. In this review, we discuss factors that influence respiratory tract Trm development, longevity, and immune surveillance and explore vaccination regimes that harness these cells, such approaches represent exciting new strategies that may be utilized to tackle the next global pandemic.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
16
|
12-month SARS-CoV-2 antibody persistency in a Tyrolean COVID-19 cohort. Wien Klin Wochenschr 2021; 133:1265-1271. [PMID: 34812944 PMCID: PMC8609251 DOI: 10.1007/s00508-021-01985-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/07/2021] [Indexed: 11/16/2022]
Abstract
Background Short-term antibody response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been shown previously. The further development remains to be determined. Methods We prospectively followed 29 coronavirus disease 2019 cases, mean age 44 ± 13.2 years. Except for one participant in whom rheumatoid arthritis existed, all other cases were previously healthy. We determined anti-viral binding antibodies at 2–10 weeks, 3 months, 6 months, and 12 months after disease onset as well as neutralizing antibodies (NAb) against wild type at 6 and 12 months and the B.1.1.7 and B.1.351 variants at month 12. Three binding antibody assays were used, targeting the nucleocapsid protein (NCP), the S1 subunit of the spike protein, and the receptor binding domain (RBD). Results Antibodies to the RBD persisted for 12 months in all cases with increasing concentrations, whereas antibodies to S1 dropped below cut-off point in 7 participants and NCP antibodies were above cut-off point in only 5 subjects at month 12. The NAb against wild type were detected in all but 2 samples at 12 months of follow-up but clearly less frequently when targeting the variants. In 5 participants who were vaccinated against COVID-19 there was a strong increase of antibodies against S1 and RBD as well as an increase of NAb titres against wild type and the variants. Conclusion There was a persisting antibody response against SARS-CoV‑2 up to 12 months after COVID-19 with declining concentrations except for RBD and a strong increase of all antibody concentrations after vaccination. Supplementary Information The online version of this article (10.1007/s00508-021-01985-x) contains supplementary material, which is available to authorized users.
Collapse
|
17
|
Humphries DC, O’Connor RA, Larocque D, Chabaud-Riou M, Dhaliwal K, Pavot V. Pulmonary-Resident Memory Lymphocytes: Pivotal Orchestrators of Local Immunity Against Respiratory Infections. Front Immunol 2021; 12:738955. [PMID: 34603321 PMCID: PMC8485048 DOI: 10.3389/fimmu.2021.738955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence that lung-resident memory T and B cells play a critical role in protecting against respiratory reinfection. With a unique transcriptional and phenotypic profile, resident memory lymphocytes are maintained in a quiescent state, constantly surveying the lung for microbial intruders. Upon reactivation with cognate antigen, these cells provide rapid effector function to enhance immunity and prevent infection. Immunization strategies designed to induce their formation, alongside novel techniques enabling their detection, have the potential to accelerate and transform vaccine development. Despite most data originating from murine studies, this review will discuss recent insights into the generation, maintenance and characterisation of pulmonary resident memory lymphocytes in the context of respiratory infection and vaccination using recent findings from human and non-human primate studies.
Collapse
Affiliation(s)
- Duncan C. Humphries
- Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, United Kingdom
- Sanofi Pasteur, R&D, Marcy l’Etoile, Lyon, France
| | - Richard A. O’Connor
- Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Kevin Dhaliwal
- Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|