1
|
Paul MJ, Hudda MT, Pallett S, Groppelli E, Boariu E, Finardi NF, Wake R, Sofat N, Biddle K, Koushesh S, Dwyer-Hemmings L, Cook R, Ma JKC. Mucosal immune responses to SARS-CoV-2 infection and COVID-19 vaccination. Vaccine 2025; 56:127175. [PMID: 40311214 DOI: 10.1016/j.vaccine.2025.127175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/20/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
SARS-CoV-2 continues to circulate in the community. We hypothesise that mucosal immunity is required to prevent continuing viral acquisition and transmission. OBJECTIVES To determine whether SARS-CoV-2 infection or vaccination elicits specific neutralising antibodies in saliva, and to assess the longevity of protection. METHODS Initially, 111 COVID-19 convalescent participants were recruited, 11-369 days after diagnosis. Saliva and blood samples were assayed for antibodies specific for Spike protein, Receptor Binding Domain and Nucleoprotein. In a second cohort, 123 participants were recruited. Saliva and serum antibodies to the same antigens were assayed before and after their first and second COVID-19 vaccinations, with 150 day follow up. RESULTS Natural infection induces and boosts IgA and IgG in oral fluid and serum; vaccination does not induce or boost specific saliva IgA; IgG can be found in saliva after vaccination, but only when serum IgG concentrations are high; IgA is important for SARS-CoV-2 neutralisation activity by oral fluid, but there can also be contributions from serum IgG and other factors. CONCLUSIONS New COVID-19 vaccines should target both systemic and mucosal immunity, to establish a first line of immune defence at the mucosal barrier. This would benefit vulnerable patient populations and may help to eradicate SARS-CoV-2 circulation.
Collapse
Affiliation(s)
- Mathew J Paul
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Mohammed T Hudda
- Department of Population Health, Dasman Institute, Jasim Mohamad Al Bahar St, Kuwait City, Kuwait.
| | - Scott Pallett
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; Centre of Defence Pathology, Royal Centre of Defence Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, B15 2WB, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Elisabetta Groppelli
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Eugenia Boariu
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Nicole Falci Finardi
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Rachel Wake
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Nidhi Sofat
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Kathryn Biddle
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| | - Soraya Koushesh
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Louis Dwyer-Hemmings
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK
| | - Richard Cook
- Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, Floor 22, Guy's Tower, Guy's Hospital, Great Maze Pond, London, SE1 1UL, UK.
| | - Julian K-C Ma
- Institute for Infection and Immunity, City St. George's University of London, Cranmer Terrace, London SW17 0RE, UK; St. George's University Hospitals NHS Foundation Trust, Blackshaw Road Tooting, London SW17 0QT, UK.
| |
Collapse
|
2
|
Zhang Y, Wu Y, Zhang MQ, Rao H, Zhang Z, He X, Liang Y, Guo R, Yuan Y, Sun J, Duyvesteyn HME, Fry EE, Stuart DI, Zhao J, Pan X, Liu SL, Zhao J, Huo J. An RBD-Fc mucosal vaccine provides variant-proof protection against SARS-CoV-2 in mice and hamsters. NPJ Vaccines 2025; 10:100. [PMID: 40383816 PMCID: PMC12086204 DOI: 10.1038/s41541-025-01155-4] [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: 02/08/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are effective against severe disease and death, but do not prevent viral infections, probably due to the limited mucosal immunity induced by intramuscular administration of the vaccine. Fusion of SARS-CoV-2 subunit immunogens with a human IgG Fc backbone can be used as a mucosal vaccine but its effectiveness in delivery in animal models, and its immunogenicity and the vaccine-induced protection against viral infections requires further studies. Here we investigate a bivalent RBD-Fc vaccine that includes the spike receptor-binding domains (RBDs) of the ancestral and BQ.1.1 variant of SARS-CoV-2. Ex vivo fluorescent imaging demonstrates that this vaccine can be effectively delivered to the lungs of mice through intranasal administration, with enhancement of retention in the nasal cavity and lung parenchyma. In mice, the vaccine elicited potent and broad-spectrum antibody responses against different variants including KP.3 which could persist for at least 3 months after booster. Importantly, it was able to induce RBD-specific mucosal IgA responses. Further, heterologous intranasal immunisation with adeno-vectored Chadv1 and RBD-Fc elicited both potent neutralising antibody and T cell responses. Immunised BALB/c and K18-hACE2-transgenic mice were also protected against viral challenge of XBB.1 and viral transmission was effectively limited in hamsters through intranasal immunisation. This work thus demonstrates the potential of RBD-Fc antigens as mucosal vaccines for prevention of breakthrough infections and onward transmission. Moreover, Fc-fusion proteins can be used as an effective mucosal vaccine strategy which can be used either alone or in combination with other vaccine technology to constitute heterologous immunisations, enabling strong protection against SARS-CoV-2 and other respiratory viruses.
Collapse
Affiliation(s)
- Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Bio-island, Guangzhou, China
| | - Yan Wu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Beijing, China
| | - Meng-Qian Zhang
- State Key Laboratory of Medicinal Chemical Biology, TianjinKey Laboratory of Biosensing and Molecular Recognition,Frontiers Science Centre for New Organic Matter, ResearchCentre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, PR China
| | - Haiyue Rao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangyue He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yiwen Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Raoqing Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaochang Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Helen M E Duyvesteyn
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - Elizabeth E Fry
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
| | - David I Stuart
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- Diamond Light Source Ltd, Harwell Science & Innovation Campus, Didcot, UK
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Bio-island, Guangzhou, China
| | - XiaoYan Pan
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Beijing, China.
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, TianjinKey Laboratory of Biosensing and Molecular Recognition,Frontiers Science Centre for New Organic Matter, ResearchCentre for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, PR China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Bio-island, Guangzhou, China.
| | - Jiandong Huo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Bio-island, Guangzhou, China.
| |
Collapse
|
3
|
Saade C, Bruel T, Vrignaud LL, Killian M, Drouillard A, Barateau V, Espi M, Mariano N, Mignon C, Bruyère L, Khoryati L, Bolland WH, Schwartz O, Lina B, Valette M, Thaunat O, Fassier JB, Pozzetto B, Paul S, Walzer T, Trouillet-Assant S. BA.1 breakthrough infection elicits distinct antibody and memory B cell responses in vaccinated-only versus hybrid immunity individuals. iScience 2025; 28:111962. [PMID: 40224022 PMCID: PMC11987676 DOI: 10.1016/j.isci.2025.111962] [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: 07/22/2024] [Revised: 01/13/2025] [Accepted: 02/03/2025] [Indexed: 04/15/2025] Open
Abstract
Immune memory is influenced by the frequency and type of antigenic challenges. Here, we performed a cross-sectional comparison of immune parameters following a BA.1 breakthrough infection in individuals with prior hybrid immunity (conferred by infection and vaccination) versus those solely vaccinated in a cohort of health care workers in Lyon, France. The results showed higher levels of serum anti-receptor binding domain (RBD) antibodies and neutralizing antibodies against BA.1 post-infection in the vaccine-only group. Individuals in this group also showed a decrease in memory B cells against the ancestral strain but an increase in those specific and cross-reactive to BA.1, suggesting a more limited immune imprinting. Conversely, hybrid immunity prevents the decrease in antibody dependent cellular cytotoxicity (ADCC) response, possibly by limiting IgG4 class-switching and enhanced anti-N responses post-infection. This highlights that BA.1 breakthrough infection induces different immune responses depending on prior history of vaccination and infection, which should be considered for further vaccination guidelines.
Collapse
Affiliation(s)
- Carla Saade
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Civils Hospices of Lyon, Lyon Sud Hospital, 69310 Pierre-Bénite, France
| | - Timothée Bruel
- Antiviral Activities of Antibodies group, Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Lou-Léna Vrignaud
- Antiviral Activities of Antibodies group, Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
| | - Martin Killian
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Annabelle Drouillard
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Véronique Barateau
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Maxime Espi
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Department of nephrology and hemodialysis, Hôpital Lyon Sud, Hospices civils de Lyon, Lyon, France
| | | | | | - Lily Bruyère
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Liliane Khoryati
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - William Henry Bolland
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, Paris, France
| | - Bruno Lina
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Centre National de Référence des virus des infections respiratoires dont la grippe, Laboratoire de Virologie, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- GenEPII Sequencing Platform, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Martine Valette
- Centre National de Référence des virus des infections respiratoires dont la grippe, Laboratoire de Virologie, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Olivier Thaunat
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Department of Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Jean-Baptiste Fassier
- Occupational Health and Medicine Department, Hospices Civils de Lyon, Université Claude Bernard Lyon1, Ifsttar, UMRESTTE, UMR T_9405, Lyon University, Avenue Rockefeller, Lyon, France
| | - Bruno Pozzetto
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Department of Microbiology, CHU Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Immunology laboratory, CIC1408, CHU Saint-Etienne, Saint-Etienne, France
| | - Thierry Walzer
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Sophie Trouillet-Assant
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Civils Hospices of Lyon, Lyon Sud Hospital, 69310 Pierre-Bénite, France
| |
Collapse
|
4
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [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] [Indexed: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
5
|
Mwape RK, Barday MA, van der Zalm MM, Verhagen LM. Overview of mucosal immunity and respiratory infections in children: a focus on Africa. Curr Opin Pediatr 2025; 37:137-144. [PMID: 39907513 PMCID: PMC11888837 DOI: 10.1097/mop.0000000000001438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
PURPOSE OF REVIEW Given the substantial burden of respiratory tract infections (RTIs) on global paediatric health, enhancing our understanding of mucosal immunity can help us advance mucosal biomarkers for diagnosis, prognosis and possible interventions in order to improve health outcomes. This review highlights the critical role of mucosal immunity in paediatric RTIs and recent advances in mucosal interventions, which offer promising strategies to improve outcomes. RECENT FINDINGS The significant burden of paediatric RTIs and growing interest in mucosal immunity advanced our understanding of the role of the respiratory mucosal immune system in protective immunity against RTIs. Studies show that sub-Saharan Africa is disproportionately affected by paediatric RTIs with poverty-associated factors such as human immunodeficiency virus (HIV) and malnutrition as risk factors. Emerging evidence highlights the important role of the respiratory microbiome and mucosal innate and adaptive immune responses in protective immunity against RTIs. SUMMARY The growing interest in mucosal immunity in RTIs has not only advanced our understanding of the overall immune responses in RTIs but also created opportunities to improve RTI care through translation of knowledge from these studies into diagnostics, therapeutics, and vaccines.
Collapse
Affiliation(s)
| | - Mish-Al Barday
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Marieke M. van der Zalm
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lilly M. Verhagen
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Paediatric Infectious Diseases and Immunology, Radboud Community for Infectious Diseases, Amalia Children's Hospital, Radboud University Medical Center
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Kesuma S, Raras TYM, Winarsih S, Shimosato T, Yurina V. Lactococcus lactis as an Effective Mucosal Vaccination Carrier: a Systematic Literature Review. J Microbiol Biotechnol 2025; 35:e2411036. [PMID: 40081887 PMCID: PMC11925755 DOI: 10.4014/jmb.2411.11036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/30/2024] [Accepted: 01/12/2025] [Indexed: 03/16/2025]
Abstract
Lactococcus lactis has potential as a mucosal vaccine delivery system. L. lactis can express antigens from bacteria or viruses, which are tightly controlled using nisin. Although L. lactis-based vaccine shows great promise, no product is ready for human use. Several studies have been conducted to develop L. lactis-based vaccine, and the efficacy of these vaccines has been evaluated in many scientific articles. This paper aims to review key aspects of current knowledge on the promising characteristics of L. lactis and to suggest its implications for vaccine design. Articles were obtained online using inclusion and exclusion criteria through Harzing's Publish or Perish. The article assessment used the Joanna Briggs Institute critical appraisal checklist for quasi-experimental studies. The efficacy evaluation of 24 articles showed that L. lactis-based vaccine can induce IgA and IgG as humoral immune responses; T CD4, T CD8, and B cells as cellular immune responses; and various proinflammatory cytokines such as IFN-γ, TNF-α, IL-2, IL-4, IL-8, IL-10, IL-12, IL-17. L. lactis is suitable as a vector carrier for oral or nasal mucosal vaccines targeting bacterial and viral infections. The development of L. lactis as a vaccine delivery system is promising.
Collapse
Affiliation(s)
- Suryanata Kesuma
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Brawijaya, East Java 65145, Indonesia
- Departement of Medical Laboratory Technology, Poltekkes Kemenkes Kalimantan Timur, Samarinda, East Borneo
| | - Tri Yudani Mardining Raras
- Departement of Biochemistry and Molecular Biology, Medical Faculty, Universitas Brawijaya, Malang 65145, Indonesia
| | - Sri Winarsih
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, Malang 65145, Indonesia
| | - Takeshi Shimosato
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, Malang 65145, Indonesia
- Institute for Aqua Regeneration, Shinshu University, Nagano, Japan
| | - Valentina Yurina
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, Malang 65145, Indonesia
| |
Collapse
|
7
|
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
|
8
|
Beeker L, Obadia T, Bloch E, Garcia L, Le Fol M, Charmet T, Arowas L, Artus R, Cheny O, Cheval D, Dahoumane Y, Delhaye M, Ergen D, Essaidani M, Fanaud C, Fernandes Pellerin S, Jolly N, Laude H, Roux E, Samson M, Sangari L, Ungeheuer MN, Vacant S, Zayoud A, Donnadieu F, Pelleau S, Galmiche S, Fontanet A, White M. Correlates of Protection Against Symptomatic COVID-19: The CORSER 5 Case-Control Study. Open Forum Infect Dis 2025; 12:ofaf006. [PMID: 39872812 PMCID: PMC11770277 DOI: 10.1093/ofid/ofaf006] [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: 06/24/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Background Establishing correlates of protection often requires large cohorts. A rapid and adaptable case-control study design can be used to identify antibody correlates of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in serum and saliva. Methods We designed a case-control study to compare antibody levels between cases of SARS-CoV-2 infection within 5 days of symptom onset and uninfected controls. Controls were matched on age, number of coronavirus disease 2019 vaccine doses, time since last dose, and past episodes of infection. We quantified anti-SARS-CoV-2 and seasonal coronavirus immunoglobulin (Ig) G in serum and saliva at inclusion, 1 month, and 6 months. Results We included 90 cases and 62 controls between February and September 2022. A boost and decay pattern of serum antibodies was observed in cases at 1 and 6 months, respectively, but not in controls. Anti-SARS-CoV-2 antibody levels were significantly higher in controls at inclusion both in serum (particularly antinucleocapsid IgG: 4.14 times higher compared with cases; 95% CI, 2.46-6.96) and saliva (particularly antispike for Delta variant IgG: 4.89 times higher compared with cases; 95% CI, 2.91-9.89). Saliva antibodies generally outperformed serum antibodies for case/control differentiation. Conclusions In this case-control study, we provided evidence of correlates of protection of anti-SARS-CoV-2 IgG in saliva and serum, with saliva antibodies often outperforming serum. The finding that antibodies in saliva are a better correlate of protection than antibodies in serum may inform vaccine development by highlighting the importance of robust induction of mucosal immune responses. This study design may be used during future epidemics for the prompt assessment of correlates of protection.
Collapse
Affiliation(s)
- Léopold Beeker
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
- Emerging Diseases Epidemiology Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Thomas Obadia
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
- Hub de Bioinformatique et Biostatistique, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Emma Bloch
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Laura Garcia
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Manon Le Fol
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Tiffany Charmet
- Emerging Diseases Epidemiology Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Laurence Arowas
- Plateforme d’Investigation Clinique (ICAReB-Clin), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Rémy Artus
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Olivia Cheny
- Pôle de Coordination de la Recherche Clinique (PC-RC), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Dorian Cheval
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Yanis Dahoumane
- Pôle de Coordination de la Recherche Clinique (PC-RC), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Maurine Delhaye
- Plateforme d’Investigation Clinique (ICAReB-Clin), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Delal Ergen
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Mariem Essaidani
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Christine Fanaud
- Plateforme d’Investigation Clinique (ICAReB-Clin), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Sandrine Fernandes Pellerin
- Pôle de Coordination de la Recherche Clinique (PC-RC), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Nathalie Jolly
- Pôle de Coordination de la Recherche Clinique (PC-RC), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Hélène Laude
- Plateforme d’Investigation Clinique (ICAReB-Clin), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Emmanuel Roux
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Marine Samson
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Linda Sangari
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Marie-Noëlle Ungeheuer
- Plateforme d’Investigation Clinique (ICAReB-Clin), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Sophie Vacant
- Biobanque de Collections de Ressources Biologiques d’Origine Humaine (ICAReB-Biobank), Center for Translational Science, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Ayla Zayoud
- Plateforme d’Investigation Clinique (ICAReB-Clin), Institut Pasteur, Université Paris-Cité, Paris, France
| | - Françoise Donnadieu
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Stéphane Pelleau
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Simon Galmiche
- Emerging Diseases Epidemiology Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Arnaud Fontanet
- Emerging Diseases Epidemiology Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
- PACRI, Risques Infectieux et Emergents, Conservatoire National des Arts et Métiers, Paris, France
| | - Michael White
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Université Paris-Cité, Paris, France
| |
Collapse
|
9
|
Fricke C, Ulrich L, Kochmann J, Gergen J, Kovacikova K, Roth N, Beer J, Schnepf D, Mettenleiter TC, Rauch S, Petsch B, Hoffmann D, Beer M, Corleis B, Dorhoi A. mRNA vaccine-induced IgG mediates nasal SARS-CoV-2 clearance in mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102360. [PMID: 39524696 PMCID: PMC11550364 DOI: 10.1016/j.omtn.2024.102360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Coronavirus disease 2019 (COVID-19) mRNA vaccines that have contributed to controlling the SARS-CoV-2 pandemic induce specific serum antibodies, which correlate with protection. However, the neutralizing capacity of antibodies for emerging SARS-CoV-2 variants is altered. Suboptimal antibody responses are observed in patients with humoral immunodeficiency diseases, ongoing B cell depletion therapy, and aging. Common experimental mouse models with altered B cell compartments, such as B cell depletion or deficiency, do not fully recapitulate scenarios of declining or suboptimal antibody levels as observed in humans. We report on SARS-CoV-2 immunity in a transgenic mouse model with restricted virus-specific antibodies. Vaccination of C57BL/6-Tg(IghelMD4)4Ccg/J mice with unmodified or N1mΨ-modified mRNA encoding for ancestral spike (S) protein and subsequent challenge with mouse-adapted SARS-CoV-2 provided insights into antibody-independent immunity and the impact of antibody titers on mucosal immunity. Protection against fatal disease was independent of seroconversion following mRNA vaccination, suggesting that virus-specific T cells can compensate for suboptimal antibody levels. In contrast, mRNA-induced IgG in the nasal conchae limited the local viral load and disease progression. Our results indicate that parenteral mRNA immunization can elicit nasal IgG antibodies that effectively suppress local viral replication, highlighting the potential of vaccines in controlling SARS-CoV-2 transmission and epidemiology.
Collapse
Affiliation(s)
- Charlie Fricke
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Lorenz Ulrich
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Jana Kochmann
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | | | | | | | - Julius Beer
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
| | | | | | | | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, 17489 Greifswald, Germany
| |
Collapse
|
10
|
Laghlali G, Wiest MJ, Karadag D, Warang P, O'Konek JJ, Chang LA, Park SC, Yan V, Farazuddin M, Janczak KW, García-Sastre A, Baker JR, Wong PT, Schotsaert M. Enhanced mucosal SARS-CoV-2 immunity after heterologous intramuscular mRNA prime/intranasal protein boost vaccination with a combination adjuvant. Mol Ther 2024; 32:4448-4466. [PMID: 39489918 PMCID: PMC11638833 DOI: 10.1016/j.ymthe.2024.10.016] [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: 03/28/2024] [Revised: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Current COVID-19 mRNA vaccines delivered intramuscularly (IM) induce effective systemic immunity, but with suboptimal immunity at mucosal sites, limiting their ability to impart sterilizing immunity. There is strong interest in rerouting immune responses induced in the periphery by parenteral vaccination to the portal entry site of respiratory viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), by mucosal vaccination. We previously demonstrated the combination adjuvant, NE/IVT, consisting of a nanoemulsion (NE) and an RNA-based RIG-I agonist (IVT) induces potent systemic and mucosal immune responses in protein-based SARS-CoV-2 vaccines administered intranasally (IN). Herein, we demonstrate priming IM with mRNA followed by heterologous IN boosting with NE/IVT adjuvanted recombinant antigen induces strong mucosal and systemic antibody responses and enhances antigen-specific T cell responses in mucosa-draining lymph nodes compared to IM/IM and IN/IN prime/boost regimens. While all regimens induced cross-neutralizing antibodies against divergent variants and sterilizing immunity in the lungs of challenged mice, mucosal vaccination, either as homologous prime/boost or heterologous IN boost after IM mRNA prime, was required to impart sterilizing immunity in the upper respiratory tract. Our data demonstrate the benefit of hybrid regimens whereby strong immune responses primed via IM vaccination are rerouted by IN vaccination to mucosal sites to provide optimal protection against SARS-CoV-2.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- Mice
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19/prevention & control
- COVID-19/immunology
- Administration, Intranasal
- Immunity, Mucosal
- Antibodies, Viral/immunology
- Injections, Intramuscular
- Female
- Immunization, Secondary
- Humans
- Antibodies, Neutralizing/immunology
- Adjuvants, Immunologic/administration & dosage
- mRNA Vaccines/immunology
- Vaccination/methods
- Adjuvants, Vaccine/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
Collapse
Affiliation(s)
- Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Matthew J Wiest
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dilara Karadag
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica J O'Konek
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seok-Chan Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vivian Yan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohammad Farazuddin
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Katarzyna W Janczak
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James R Baker
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pamela T Wong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
11
|
Tang J, Sun J. From blood to mucosa. Sci Transl Med 2024; 16:eads6271. [PMID: 39441906 DOI: 10.1126/scitranslmed.ads6271] [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: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Current COVID-19 vaccines induce suboptimal respiratory mucosal immunity even after mRNA boosters (Declercq et al. and Lasrado et al., this issue).
Collapse
Affiliation(s)
- Jinyi Tang
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
12
|
Lasrado N, Rowe M, McMahan K, Hachmann NP, Miller J, Jacob-Dolan C, Liu J, Verrette B, Gotthardt KA, Ty DM, Pereira J, Mazurek CR, Hoyt A, Collier ARY, Barouch DH. SARS-CoV-2 XBB.1.5 mRNA booster vaccination elicits limited mucosal immunity. Sci Transl Med 2024; 16:eadp8920. [PMID: 39441905 PMCID: PMC11542980 DOI: 10.1126/scitranslmed.adp8920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/05/2024] [Indexed: 10/25/2024]
Abstract
Current COVID-19 vaccines provide robust protection against severe disease but minimal protection against acquisition of infection. Intramuscularly administered COVID-19 vaccines induce robust serum neutralizing antibodies (NAbs), but their ability to boost mucosal immune responses remains to be determined. In this study, we show that the XBB.1.5 messenger RNA (mRNA) boosters result in increased serum neutralization to multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in humans, including the dominant circulating variant JN.1. In contrast, we found that the XBB.1.5 mRNA booster did not augment mucosal NAbs or mucosal IgA responses, although acute SARS-CoV-2 XBB infection substantially increased mucosal antibody responses. These data demonstrate that current XBB.1.5 mRNA boosters substantially enhance peripheral antibody responses but do not robustly increase mucosal antibody responses. Our data highlight a separation between the peripheral and mucosal immune systems in humans and emphasize the importance of developing next-generation vaccines to augment mucosal immunity to protect against respiratory virus infections.
Collapse
Affiliation(s)
- Ninaad Lasrado
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Marjorie Rowe
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Katherine McMahan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Nicole P. Hachmann
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Jessica Miller
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Catherine Jacob-Dolan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Jinyan Liu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Brookelynne Verrette
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Kristin A. Gotthardt
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Darren M. Ty
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Juliana Pereira
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Camille R. Mazurek
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Amelia Hoyt
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Ai-ris Y. Collier
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Dan H. Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
13
|
Suthar MS. Durability of immune responses to SARS-CoV-2 infection and vaccination. Semin Immunol 2024; 73:101884. [PMID: 38861769 PMCID: PMC11490408 DOI: 10.1016/j.smim.2024.101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024]
Abstract
Infection with SARS-CoV-2 in humans has caused a pandemic of unprecedented dimensions. SARS-CoV-2 is primarily transmitted through respiratory droplets and targets ciliated epithelial cells in the nasal cavity, trachea, and lungs by utilizing the cellular receptor angiotensin-converting enzyme 2 (ACE2). The innate immune response, including type I and III interferons, inflammatory cytokines (IL-6, TNF-α, IL-1β), innate immune cells (monocytes, DCs, neutrophils, natural killer cells), antibodies (IgG, sIgA, neutralizing antibodies), and adaptive immune cells (B cells, CD8+ and CD4+ T cells) play pivotal roles in mitigating COVID-19 disease. Broad and durable B-cell- and T-cell immunity elicited by infection and vaccination is essential for protection against severe disease, hospitalization and death. However, the emergence of SARS-CoV-2 variants that evade neutralizing antibodies continue to jeopardize vaccine efficacy. In this review, we highlight our understanding the infection- and vaccine-mediated humoral, B and T cell responses, the durability of the immune responses, and how variants continue to threaten the efficacy of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, USA; Emory Center of Excellence of Influenza Research and Response (CEIRR), Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Laghlali G, Wiest MJ, Karadag D, Warang P, O'Konek JJ, Chang LA, Park S, Farazuddin M, Landers JJ, Janczak KW, García-Sastre A, Baker JR, Wong PT, Schotsaert M. Enhanced mucosal B- and T-cell responses against SARS-CoV-2 after heterologous intramuscular mRNA prime/intranasal protein boost vaccination with a combination adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587260. [PMID: 38586014 PMCID: PMC10996704 DOI: 10.1101/2024.03.28.587260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Current COVID-19 mRNA vaccines delivered intramuscularly (IM) induce effective systemic immunity, but with suboptimal immunity at mucosal sites, limiting their ability to impart sterilizing immunity. There is strong interest in rerouting immune responses induced in the periphery by parenteral vaccination to the portal entry site of respiratory viruses, such as SARS-CoV-2, by mucosal vaccination. We previously demonstrated the combination adjuvant, NE/IVT, consisting of a nanoemulsion (NE) and an RNA-based RIG-I agonist (IVT) induces potent systemic and mucosal immune responses in protein-based SARS-CoV-2 vaccines administered intranasally (IN). Herein, we demonstrate priming IM with mRNA followed by heterologous IN boosting with NE/IVT adjuvanted recombinant antigen induces strong mucosal and systemic antibody responses and enhances antigen-specific T cell responses in mucosa-draining lymph nodes compared to IM/IM and IN/IN prime/boost regimens. While all regimens induced cross-neutralizing antibodies against divergent variants and sterilizing immunity in the lungs of challenged mice, mucosal vaccination, either as homologous prime/boost or heterologous IN boost after IM mRNA prime was required to impart sterilizing immunity in the upper respiratory tract. Our data demonstrate the benefit of hybrid regimens whereby strong immune responses primed via IM vaccination are rerouted by IN vaccination to mucosal sites to provide optimal protection to SARS-CoV-2.
Collapse
|