1
|
Merolle M, Striepen B, Hunter CA. Parasite and host immune factors that impact the development of a mucosal vaccine for Cryptosporidium. Mucosal Immunol 2025:S1933-0219(25)00049-2. [PMID: 40379259 DOI: 10.1016/j.mucimm.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/28/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025]
Abstract
The parasite Cryptosporidium is a leading cause of diarrhea and death in malnourished children and immunocompromised individuals and an important enteric pathogen of livestock. A mucosal vaccine to mitigate clinical disease and decrease transmission would address the public health impact of this organism, but current options are limited. The development of a rational strategy for vaccination requires an appreciation of the parasite lifecycle, how Cryptosporidium interacts with its host cell (the enterocyte), and the immune mechanisms that act locally to control this organism. Here we review current knowledge of the adaptive immune mechanisms that mediate resistance to Cryptosporidium, their relevance to vaccine design, and how recent advances in parasite genetics inform vaccine development.
Collapse
Affiliation(s)
- Maria Merolle
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, PA 19104, United States
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, PA 19104, United States
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, PA 19104, United States.
| |
Collapse
|
2
|
Counoupas C, Chan E, Pino P, Armitano J, Johansen MD, Smith LJ, Ashley CL, Estapé E, Troyon J, Alca S, Miemczyk S, Hansbro NG, Scandurra G, Britton WJ, Courant T, Dubois PM, Collin N, Mohan VK, Hansbro PM, Wurm MJ, Wurm FM, Steain M, Triccas JA. An adjuvanted chimeric spike antigen boosts lung-resident memory T-cells and induces pan-sarbecovirus protective immunity. NPJ Vaccines 2025; 10:89. [PMID: 40341541 PMCID: PMC12062434 DOI: 10.1038/s41541-025-01144-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
Next-generation vaccines are essential to address the evolving nature of SARS-CoV-2 and to protect against emerging pandemic threats from other coronaviruses. These vaccines should elicit broad protection, provide long-lasting immunity and ensure equitable access for all populations. In this study, we developed a panel of chimeric, full-length spike antigens incorporating mutations from previous, circulating and predicted SARS-CoV-2 variants. The lead candidate (CoVEXS5) was produced through a high-yield production process in stable CHO cells achieving >95% purity, demonstrated long-term stability and elicited broadly cross-reactive neutralising antibodies when delivered to mice in a squalene emulsion adjuvant (Sepivac SWE™). In both mice and hamsters, CoVEXS5 immunisation reduced clinical disease signs, lung inflammation and organ viral titres after SARS-CoV-2 infection, including following challenge with the highly immunoevasive Omicron XBB.1.5 subvariant. In mice previously primed with a licenced mRNA vaccine (Comirnaty XBB.1.5, termed mRNA-XBB), CoVEXS5 boosting significantly increased neutralising antibody (nAb) levels against viruses from three sarbecoviruses clades. Boosting with CoVEXS5 via systemic delivery elicited CD4+ lung-resident memory T cells, typically associated with mucosal immunisation strategies, which were not detected following mRNA-XBB boosting. Vaccination of hamsters with CoVEXS5 conferred significant protection against weight loss after SARS-CoV-1 challenge, compared to mRNA-XBB immunisation, that correlated with anti-SARS-CoV-1 nAbs in the sera of vaccinated animals. These findings highlight the potential of a chimeric spike antigen, formulated in an open-access adjuvant, as a next-generation vaccine candidate to enhance cross-protection against emerging sarbecoviruses in vaccinated populations globally.
Collapse
Affiliation(s)
- Claudio Counoupas
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Elizabeth Chan
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | | | | | - Matt D Johansen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Lachlan J Smith
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Caroline L Ashley
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | | | | | - Sibel Alca
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Stefan Miemczyk
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Gabriella Scandurra
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Warwick J Britton
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Thomas Courant
- Vaccine Formulation Institute, Plan-Les-Ouates, Switzerland
| | | | - Nicolas Collin
- Vaccine Formulation Institute, Plan-Les-Ouates, Switzerland
| | - V Krishna Mohan
- Bharat Biotech International Limited, Genome Valley, Hyderabad, Telangana, India
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | | | - Florian M Wurm
- ExcellGene SA, Monthey, Switzerland
- Life Science Faculty, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Megan Steain
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Charles Perkin Centre, The University of Sydney, Camperdown, NSW, Australia
| | - James A Triccas
- Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Charles Perkin Centre, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
3
|
Lukeman H, Al-Wassiti H, Fabb SA, Lim L, Wang T, Britton WJ, Steain M, Pouton CW, Triccas JA, Counoupas C. An LNP-mRNA vaccine modulates innate cell trafficking and promotes polyfunctional Th1 CD4 + T cell responses to enhance BCG-induced protective immunity against Mycobacterium tuberculosis. EBioMedicine 2025; 113:105599. [PMID: 39955975 PMCID: PMC11871481 DOI: 10.1016/j.ebiom.2025.105599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Mycobacterium tuberculosis remains the largest infectious cause of mortality worldwide, even with over a century of widespread administration of the only licenced tuberculosis (TB) vaccine, Bacillus Calmette-Guérin (BCG). mRNA technology remains an underexplored approach for combating chronic bacterial infections such as TB. METHODS We have developed a lipid nanoparticle (LNP)-mRNA vaccine, termed mRNACV2, encoding for the M. tuberculosis CysVac2 fusion protein, which we have previously formulated as an adjuvanted subunit vaccine. This LNP-mRNA vaccine was administered intramuscularly to female C57BL/6 mice as a standalone vaccine or as booster to BCG to assess immunogenicity and efficacy of the construct. FINDINGS Vaccination with mRNACV2 induced high frequencies of polyfunctional, antigen-specific Th1 CD4+ T cells in the blood and lungs, which was associated with the rapid recruitment of both innate and adaptive immune cells to lymph nodes draining the site of immunisation. mRNACV2 vaccination also provided significant pulmonary protection in M. tuberculosis-infected mice, reducing bacterial load and inflammatory infiltration in the lungs. Importantly, mRNACV2 enhanced immune responses and long-term protection when used to boost BCG-primed mice. INTERPRETATION These findings of a protective LNP-mRNA vaccine for TB highlight the potential of the LNP-mRNA platform for TB control and support further research to facilitate translation to humans. FUNDING This work was supported by the NHMRC Centre of Research Excellence in Tuberculosis Control to JAT and WJB (APP1153493), and MRFF mRNA Clinical Trial Enabling Infrastructure grant to CWP and HAW (MRFCTI000006).
Collapse
Affiliation(s)
- Hannah Lukeman
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Hareth Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Trixie Wang
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Warwick J Britton
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Megan Steain
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - James A Triccas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Claudio Counoupas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
4
|
Walsh JML, Miao VN, Owings AH, Tang Y, Bromley JD, Kazer SW, Kimler K, Asare C, Ziegler CGK, Ibrahim S, Jivanjee T, George M, Navia AW, Drake RS, Parker A, Billingsley BC, Dotherow P, Tarugu S, Kota SK, Laird H, Wichman TG, Davis YT, Dhaliwal NS, Pride Y, Guo Y, Senitko M, Harvey J, Bates JT, Diamond G, Garrett MR, Robinson DA, Frame IJ, Lyons JJ, Robinson TO, Shalek AK, Horwitz BH, Glover SC, Ordovas-Montanes J. Variants and vaccines impact nasal immunity over three waves of SARS-CoV-2. Nat Immunol 2025; 26:294-307. [PMID: 39833605 DOI: 10.1038/s41590-024-02052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025]
Abstract
Viral variant and host vaccination status impact infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), yet how these factors shift cellular responses in the human nasal mucosa remains uncharacterized. We performed single-cell RNA sequencing (scRNA-seq) on nasopharyngeal swabs from vaccinated and unvaccinated adults with acute Delta and Omicron SARS-CoV-2 infections and integrated with data from acute infections with ancestral SARS-CoV-2. Patients with Delta and Omicron exhibited greater similarity in nasal cell composition driven by myeloid, T cell and SARS-CoV-2hi cell subsets, which was distinct from that of ancestral cases. Delta-infected samples had a marked increase in viral RNA, and a subset of PER2+EGR1+GDF15+ epithelial cells was enriched in SARS-CoV-2 RNA+ cells in all variants. Prior vaccination was associated with increased frequency and activation of nasal macrophages. Expression of interferon-stimulated genes negatively correlated with coronavirus disease 2019 (COVID-19) severity in patients with ancestral and Delta but not Omicron variants. Our study defines nasal cell responses and signatures of disease severity across SARS-CoV-2 variants and vaccination.
Collapse
Affiliation(s)
- Jaclyn M L Walsh
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vincent N Miao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
| | - Anna H Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ying Tang
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Joshua D Bromley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel W Kazer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Kyle Kimler
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Chelsea Asare
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carly G K Ziegler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
| | - Samira Ibrahim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tasneem Jivanjee
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Micayla George
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew W Navia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Riley S Drake
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Parker
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Paul Dotherow
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Spurthi Tarugu
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sai K Kota
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hannah Laird
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - T Grant Wichman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yesenia T Davis
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Neha S Dhaliwal
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yilianys Pride
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yanglin Guo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michal Senitko
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jessie Harvey
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - John T Bates
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gill Diamond
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Michael R Garrett
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - D Ashley Robinson
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - I J Frame
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jonathan J Lyons
- Division of Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Tanya O Robinson
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alex K Shalek
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bruce H Horwitz
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sarah C Glover
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University, New Orleans, LA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
5
|
Brandi R, Paganelli A, D’Amelio R, Giuliani P, Lista F, Salemi S, Paganelli R. mRNA Vaccines Against COVID-19 as Trailblazers for Other Human Infectious Diseases. Vaccines (Basel) 2024; 12:1418. [PMID: 39772079 PMCID: PMC11680146 DOI: 10.3390/vaccines12121418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
mRNA vaccines represent a milestone in the history of vaccinology, because they are safe, very effective, quick and cost-effective to produce, easy to adapt should the antigen vary, and able to induce humoral and cellular immunity. METHODS To date, only two COVID-19 mRNA and one RSV vaccines have been approved. However, several mRNA vaccines are currently under development for the prevention of human viral (influenza, human immunodeficiency virus [HIV], Epstein-Barr virus, cytomegalovirus, Zika, respiratory syncytial virus, metapneumovirus/parainfluenza 3, Chikungunya, Nipah, rabies, varicella zoster virus, and herpes simplex virus 1 and 2), bacterial (tuberculosis), and parasitic (malaria) diseases. RESULTS RNA viruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV)-2, HIV, and influenza, are characterized by high variability, thus creating the need to rapidly adapt the vaccines to the circulating viral strain, a task that mRNA vaccines can easily accomplish; however, the speed of variability may be higher than the time needed for a vaccine to be adapted. mRNA vaccines, using lipid nanoparticles as the delivery system, may act as adjuvants, thus powerfully stimulating innate as well as adaptive immunity, both humoral, which is rapidly waning, and cell-mediated, which is highly persistent. Safety profiles were satisfactory, considering that only a slight increase in prognostically favorable anaphylactic reactions in young females and myopericarditis in young males has been observed. CONCLUSIONS The COVID-19 pandemic determined a shift in the use of RNA: after having been used in medicine as micro-RNAs and tumor vaccines, the new era of anti-infectious mRNA vaccines has begun, which is currently in great development, to either improve already available, but unsatisfactory, vaccines or develop protective vaccines against infectious agents for which no preventative tools have been realized yet.
Collapse
Affiliation(s)
- Rossella Brandi
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | | | | | - Paolo Giuliani
- Poliambulatorio Montezemolo, Ente Sanitario Militare del Ministero Della Difesa Presso la Corte dei Conti, 00195 Rome, Italy;
| | - Florigio Lista
- Istituto di Science Biomediche della Difesa, Stato Maggiore Della Difesa, 00184 Rome, Italy; (R.B.); (F.L.)
| | - Simonetta Salemi
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria S. Andrea, 00189 Rome, Italy
| | - Roberto Paganelli
- Internal Medicine, Faculty of Medicine and Surgery, Unicamillus, International School of Medicine, 00131 Rome, Italy
| |
Collapse
|
6
|
Vorobyeva DA, Potashnikova DM, Maryukhnich EV, Rusakovich GI, Tvorogova AV, Kalinskaya AI, Pinegina NV, Kovyrshina AV, Dolzhikova IV, Postnikov AB, Rozov FN, Sotnikova TN, Kanner DY, Logunov DY, Gintsburg AL, Vasilieva EJ, Margolis LB. Cytokine production in an ex vivo model of SARS-CoV-2 lung infection. Front Immunol 2024; 15:1448515. [PMID: 39497823 PMCID: PMC11532052 DOI: 10.3389/fimmu.2024.1448515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction The mechanisms of the SARS-CoV-2-triggered complex alterations in immune cell activation and production of cytokines in lung tissue remain poorly understood, in part because of the limited use of adequate tissue models that simulate the structure and cell composition of the lung in vivo. We developed a novel ex vivo model of SARS-CoV-2 infection of lung explants, that maintains the intact tissue composition and the viral load for up to 7-10 days. Using this model, we studied cytokine production during SARS-CoV-2 infection. Materials and methods Lung tissue was monitored for viability and cell composition using flow cytometry and histological analysis. SARS-CoV-2 infection was verified immunohistochemically, viral loads in tissue and culture medium were monitored by qPCR. A panel of 41 cytokines was measured in culture medium using xMAP technology. Results The explant lung tissue was viable and maintained viral infection that influenced the cytokine production. Elevated concentrations of G-CSF, GM-CSF, GRO-a, IFN-g, IL-6, IL-8, IP-10, MCP-3, MIP-1a, PDGF-AA, and VEGF, and decreased IL-1RA concentration were observed in infected tissue compared to non-infected tissue. Discussion Our results generally reflect the data obtained in COVID-19 patients. GRO-a, IFN-g, IL-6, IL-8, MCP-1, MCP-3, and RANTES correlated with the viral load, forming a distinct pro-inflammatory cluster. Thus, our lung ex vivo model faithfully reproduces some aspects of cytokine alterations in COVID-19 patients at an early disease stage, making the investigation of SARS-CoV-2 infection mechanisms more accessible and providing a potential platform for antiviral drug testing.
Collapse
Affiliation(s)
- Daria A. Vorobyeva
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Daria M. Potashnikova
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Elena V. Maryukhnich
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - George I. Rusakovich
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna V. Tvorogova
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna I. Kalinskaya
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Natalia V. Pinegina
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna V. Kovyrshina
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Inna V. Dolzhikova
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Tatiana N. Sotnikova
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | | | - Denis Yu. Logunov
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L. Gintsburg
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena J. Vasilieva
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Leonid B. Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
7
|
Notarbartolo S. T-Cell Immune Responses to SARS-CoV-2 Infection and Vaccination. Vaccines (Basel) 2024; 12:1126. [PMID: 39460293 PMCID: PMC11511197 DOI: 10.3390/vaccines12101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The innate and adaptive immune systems collaborate to detect SARS-CoV-2 infection, minimize the viral spread, and kill infected cells, ultimately leading to the resolution of the infection. The adaptive immune system develops a memory of previous encounters with the virus, providing enhanced responses when rechallenged by the same pathogen. Such immunological memory is the basis of vaccine function. Here, we review the current knowledge on the immune response to SARS-CoV-2 infection and vaccination, focusing on the pivotal role of T cells in establishing protective immunity against the virus. After providing an overview of the immune response to SARS-CoV-2 infection, we describe the main features of SARS-CoV-2-specific CD4+ and CD8+ T cells, including cross-reactive T cells, generated in patients with different degrees of COVID-19 severity, and of Spike-specific CD4+ and CD8+ T cells induced by vaccines. Finally, we discuss T-cell responses to SARS-CoV-2 variants and hybrid immunity and conclude by highlighting possible strategies to improve the efficacy of COVID-19 vaccination.
Collapse
Affiliation(s)
- Samuele Notarbartolo
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
8
|
Verheul MK, Kaczorowska J, Hofstee MI, Schepp RM, Smits GP, Wessels Beljaars D, Kuijer M, Schuin W, Middelhof I, Wong D, van Hagen CCE, Vos ERA, Nicolaie MA, de Melker HE, van Binnendijk RS, van der Klis FRM, den Hartog G. Protective mucosal SARS-CoV-2 antibodies in the majority of the general population in the Netherlands. Mucosal Immunol 2024; 17:554-564. [PMID: 38553008 DOI: 10.1016/j.mucimm.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024]
Abstract
Antibodies to SARS-CoV-2 on the mucosal surfaces of the respiratory tract are understood to contribute to protection against SARS-CoV-2 infection. We aimed to describe the prevalence, levels, and functionality of mucosal antibodies in the general Dutch population. Nasal samples were collected from 778 randomly selected participants, 1-90 years of age, nested within the nationwide prospective SARS-CoV-2 PIENTER corona serosurvey in the Netherlands. Spike-specific immunoglobulin (Ig)G was detected in the nasal samples of 94.6% (in case of the wild-type S1 variant) and 94.9% (Omicron BA.1) of the individuals, whereas 44.2% and 62.7% of the individuals were positive for wild-type and Omicron BA.1 S1 IgA, respectively. The lowest prevalence of mucosal antibodies was observed in children under 12 years of age. The prevalence and levels of IgA and IgG were higher in individuals with a history of SARS-CoV-2 infection. Mucosal antibodies inhibited the binding of Wuhan, Delta, and Omicron BA.1 receptor binding domain to human angiotensin-converting enzyme 2 in 94.4%, 95.4%, and 92.6% of the participants, respectively. Higher levels of mucosal antibodies were associated with a lower risk of future infection.
Collapse
Affiliation(s)
- Marije K Verheul
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Joanna Kaczorowska
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Marloes I Hofstee
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Rutger M Schepp
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Gaby P Smits
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Dewi Wessels Beljaars
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Marjan Kuijer
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Wendy Schuin
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Irene Middelhof
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Denise Wong
- Centre for Infectious Diseases, Epidemiology and Surveillance, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Cheyenne C E van Hagen
- Centre for Infectious Diseases, Epidemiology and Surveillance, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Eric R A Vos
- Centre for Infectious Diseases, Epidemiology and Surveillance, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - M Alina Nicolaie
- Department of Statistics, Data Science and Modelling, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Hester E de Melker
- Centre for Infectious Diseases, Epidemiology and Surveillance, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Robert S van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Fiona R M van der Klis
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Gerco den Hartog
- Centre for Immunology of Infectious Diseases and Vaccines, Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands; Laboratory of Medical Immunology, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW The complex nature and distribution of the HIV reservoir in tissue of people with HIV remains one of the major obstacles to achieve the elimination of HIV persistence. Challenges include the tissue-specific states of latency and viral persistence, which translates into high levels of reservoir heterogeneity. Moreover, the best strategies to reach and eliminate these reservoirs may differ based on the intrinsic characteristics of the cellular and anatomical reservoir to reach. RECENT FINDINGS While major focus has been undertaken for lymphoid tissues and follicular T helper cells, evidence of viral persistence in HIV and non-HIV antigen-specific CD4 + T cells and macrophages resident in multiple tissues providing long-term protection presents new challenges in the quest for an HIV cure. Considering the microenvironments where these cellular reservoirs persist opens new venues for the delivery of drugs and immunotherapies to target these niches. New tools, such as single-cell RNA sequencing, CRISPR screenings, mRNA technology or tissue organoids are quickly developing and providing detailed information about the complex nature of the tissue reservoirs. SUMMARY Targeting persistence in tissue reservoirs represents a complex but essential step towards achieving HIV cure. Combinatorial strategies, particularly during the early phases of infection to impact initial reservoirs, capable of reaching and reactivating multiple long-lived reservoirs in the body may lead the path.
Collapse
Affiliation(s)
- Daan K J Pieren
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | | |
Collapse
|
10
|
Lavelle EC, Genescà M. Editorial overview: The march of mucosal vaccines. Curr Opin Immunol 2024; 86:102408. [PMID: 38128438 DOI: 10.1016/j.coi.2023.102408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Affiliation(s)
- E C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| |
Collapse
|
11
|
Noh HE, Rha MS. Mucosal Immunity against SARS-CoV-2 in the Respiratory Tract. Pathogens 2024; 13:113. [PMID: 38392851 PMCID: PMC10892713 DOI: 10.3390/pathogens13020113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
The respiratory tract, the first-line defense, is constantly exposed to inhaled allergens, pollutants, and pathogens such as respiratory viruses. Emerging evidence has demonstrated that the coordination of innate and adaptive immune responses in the respiratory tract plays a crucial role in the protection against invading respiratory pathogens. Therefore, a better understanding of mucosal immunity in the airways is critical for the development of novel therapeutics and next-generation vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory viruses. Since the coronavirus disease 2019 pandemic, our knowledge of mucosal immune responses in the airways has expanded. In this review, we describe the latest knowledge regarding the key components of the mucosal immune system in the respiratory tract. In addition, we summarize the host immune responses in the upper and lower airways following SARS-CoV-2 infection and vaccination, and discuss the impact of allergic airway inflammation on mucosal immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Hae-Eun Noh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Min-Seok Rha
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
12
|
Proß V, Sattler A, Lukassen S, Tóth L, Thole LML, Siegle J, Stahl C, He A, Damm G, Seehofer D, Götz C, Bayerl C, Jäger P, Macke A, Eggeling S, Kirzinger B, Mayr T, Herbst H, Beyer K, Laue D, Krönke J, Braune J, Rosseck F, Kittner B, Friedersdorff F, Hubatsch M, Weinberger S, Lachmann N, Hofmann VM, Schrezenmeier E, Ludwig C, Schrezenmeier H, Jechow K, Conrad C, Kotsch K. SARS-CoV-2 mRNA vaccination-induced immunological memory in human nonlymphoid and lymphoid tissues. J Clin Invest 2023; 133:e171797. [PMID: 37815874 PMCID: PMC10721158 DOI: 10.1172/jci171797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Tissue-resident lymphocytes provide organ-adapted protection against invading pathogens. Whereas their biology has been examined in great detail in various infection models, their generation and functionality in response to vaccination have not been comprehensively analyzed in humans. We therefore studied SARS-CoV-2 mRNA vaccine-specific T cells in surgery specimens of kidney, liver, lung, bone marrow, and spleen compared with paired blood samples from largely virus-naive individuals. As opposed to lymphoid tissues, nonlymphoid organs harbored significantly elevated frequencies of spike-specific CD4+ T cells compared with blood showing hallmarks of tissue residency and an expanded memory pool. Organ-derived CD4+ T cells further exhibited increased polyfunctionality over those detected in blood. Single-cell RNA-Seq together with T cell receptor repertoire analysis indicated that the clonotype rather than organ origin is a major determinant of transcriptomic state in vaccine-specific CD4+ T cells. In summary, our data demonstrate that SARS-CoV-2 vaccination entails acquisition of tissue memory and residency features in organs distant from the inoculation site, thereby contributing to our understanding of how local tissue protection might be accomplished.
Collapse
Affiliation(s)
- Vanessa Proß
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Arne Sattler
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sören Lukassen
- Center of Digital Health, Berlin Institute of Health and Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura Tóth
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Linda Marie Laura Thole
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Janine Siegle
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carolin Stahl
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - An He
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Christina Götz
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Christian Bayerl
- Department of Radiology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Pia Jäger
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | | | | | | | - Hermann Herbst
- Department of Pathology, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Katharina Beyer
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dominik Laue
- Department of Traumatology and Reconstructive Surgery, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Krönke
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan Braune
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Rosseck
- Institute of Pathology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Beatrice Kittner
- Department of Urology, Evangelisches Krankenhaus Königin Elisabeth Herzberge, Berlin, Germany
| | - Frank Friedersdorff
- Department of Urology, Evangelisches Krankenhaus Königin Elisabeth Herzberge, Berlin, Germany
| | - Mandy Hubatsch
- Department of Urology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sarah Weinberger
- Department of Urology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nils Lachmann
- Institute of Transfusion Medicine, Berlin Institute of Health, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Veit Maria Hofmann
- Department of Otolaryngology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Carolin Ludwig
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, Ulm, Germany
| | - Katharina Jechow
- Center of Digital Health, Berlin Institute of Health and Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Conrad
- Center of Digital Health, Berlin Institute of Health and Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katja Kotsch
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
Kingstad-Bakke B, Cleven T, Bussan H, Yount BL, Uraki R, Iwatsuki-Horimoto K, Koga M, Yamamoto S, Yotsuyanagi H, Park H, Mishra JS, Kumar S, Baric RS, Halfmann PJ, Kawaoka Y, Suresh M. Airway surveillance and lung viral control by memory T cells induced by COVID-19 mRNA vaccine. JCI Insight 2023; 8:e172510. [PMID: 37796612 PMCID: PMC10721330 DOI: 10.1172/jci.insight.172510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
Although SARS-CoV-2 evolution seeds a continuous stream of antibody-evasive viral variants, COVID-19 mRNA vaccines provide robust protection against severe disease and hospitalization. Here, we asked whether mRNA vaccine-induced memory T cells limit lung SARS-CoV-2 replication and severe disease. We show that mice and humans receiving booster BioNTech mRNA vaccine developed potent CD8 T cell responses and showed similar kinetics of expansion and contraction of granzyme B/perforin-expressing effector CD8 T cells. Both monovalent and bivalent mRNA vaccines elicited strong expansion of a heterogeneous pool of terminal effectors and memory precursor effector CD8 T cells in spleen, inguinal and mediastinal lymph nodes, pulmonary vasculature, and most surprisingly in the airways, suggestive of systemic and regional surveillance. Furthermore, we document that: (a) CD8 T cell memory persists in multiple tissues for > 200 days; (b) following challenge with pathogenic SARS-CoV-2, circulating memory CD8 T cells rapidly extravasate to the lungs and promote expeditious viral clearance, by mechanisms that require CD4 T cell help; and (c) adoptively transferred splenic memory CD8 T cells traffic to the airways and promote lung SARS-CoV-2 clearance. These findings provide insights into the critical role of memory T cells in preventing severe lung disease following breakthrough infections with antibody-evasive SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thomas Cleven
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hailey Bussan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Boyd L. Yount
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryuta Uraki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | | | - Michiko Koga
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shinya Yamamoto
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hongtae Park
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jay S. Mishra
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter J. Halfmann
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Wang L, Nicols A, Turtle L, Richter A, Duncan CJA, Dunachie SJ, Klenerman P, Payne RP. T cell immune memory after covid-19 and vaccination. BMJ MEDICINE 2023; 2:e000468. [PMID: 38027416 PMCID: PMC10668147 DOI: 10.1136/bmjmed-2022-000468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
The T cell memory response is a crucial component of adaptive immunity responsible for limiting or preventing viral reinfection. T cell memory after infection with the SARS-CoV-2 virus or vaccination is broad, and spans multiple viral proteins and epitopes, about 20 in each individual. So far the T cell memory response is long lasting and provides a high level of cross reactivity and hence resistance to viral escape by variants of the SARS-CoV-2 virus, such as the omicron variant. All current vaccine regimens tested produce robust T cell memory responses, and heterologous regimens will probably enhance protective responses through increased breadth. T cell memory could have a major role in protecting against severe covid-19 disease through rapid viral clearance and early presentation of epitopes, and the presence of cross reactive T cells might enhance this protection. T cell memory is likely to provide ongoing protection against admission to hospital and death, and the development of a pan-coronovirus vaccine might future proof against new pandemic strains.
Collapse
Affiliation(s)
- Lulu Wang
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Alex Nicols
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Alex Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Christopher JA Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- Department of Infection and Tropical Medicine, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Susanna J Dunachie
- NDM Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University Faculty of Science, Bangkok, Thailand
| | - Paul Klenerman
- Oxford University Hospitals NHS Foundation Trust, Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, Oxfordshire, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
Mitsi E, Diniz MO, Reiné J, Collins AM, Robinson RE, Hyder-Wright A, Farrar M, Liatsikos K, Hamilton J, Onyema O, Urban BC, Solórzano C, Belij-Rammerstorfer S, Sheehan E, Lambe T, Draper SJ, Weiskopf D, Sette A, Maini MK, Ferreira DM. Respiratory mucosal immune memory to SARS-CoV-2 after infection and vaccination. Nat Commun 2023; 14:6815. [PMID: 37884506 PMCID: PMC10603102 DOI: 10.1038/s41467-023-42433-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Respiratory mucosal immunity induced by vaccination is vital for protection from coronavirus infection in animal models. In humans, the capacity of peripheral vaccination to generate sustained immunity in the lung mucosa, and how this is influenced by prior SARS-CoV-2 infection, is unknown. Here we show using bronchoalveolar lavage samples that donors with history of both infection and vaccination have more airway mucosal SARS-CoV-2 antibodies and memory B cells than those only vaccinated. Infection also induces populations of airway spike-specific memory CD4+ and CD8+ T cells that are not expanded by vaccination alone. Airway mucosal T cells induced by infection have a distinct hierarchy of antigen specificity compared to the periphery. Spike-specific T cells persist in the lung mucosa for 7 months after the last immunising event. Thus, peripheral vaccination alone does not appear to induce durable lung mucosal immunity against SARS-CoV-2, supporting an argument for the need for vaccines targeting the airways.
Collapse
Affiliation(s)
- Elena Mitsi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Mariana O Diniz
- Division of Infection and Immunity and Institute of Immunity and Transplantation, UCL, London, UK
| | - Jesús Reiné
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Andrea M Collins
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Ryan E Robinson
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Angela Hyder-Wright
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Madlen Farrar
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Josh Hamilton
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Onyia Onyema
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Britta C Urban
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Carla Solórzano
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Emma Sheehan
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, USA
| | - Mala K Maini
- Division of Infection and Immunity and Institute of Immunity and Transplantation, UCL, London, UK
| | - Daniela M Ferreira
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
16
|
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
|