1
|
Mura M, Trignol A, Le Dault E, Tournier JN. Lessons for medical countermeasure development from unforeseen outbreaks. Emerg Microbes Infect 2025; 14:2471035. [PMID: 39976365 PMCID: PMC11894751 DOI: 10.1080/22221751.2025.2471035] [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: 11/06/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/21/2025]
Abstract
The unanticipated emergence of the COVID-19 pandemic and the rapid spread of the mpox epidemic in 2022 and 2024 brought unforeseen challenges to public health. While distinct in nature, these outbreaks share some similarities and offer valuable insights into responding to novel virus dissemination in vulnerable populations. In light of these two experiences, we aim to discern the prioritization of medical countermeasures (MCM) among antivirals, antibodies, and vaccines. Comparative analysis of MCMs reveals that while antivirals serve essential roles as therapeutic tools, monoclonal antibodies can be used for both prevention and treatment, and vaccines remain of paramount importance for controlling epidemics as mass or targeted prophylaxis. Variability in production processes, administration methods, logistics, and costs distinguish these countermeasures. Vaccines, by inducing long-lasting immunity and ideally promoting herd effects, exhibit substantial advantages over other options. To enhance future pandemic readiness, proactive measures must include ready-to-use vaccine platforms with regulatory approval and manufacturing capacities, as well as prototype vaccines for representative pathogens and preexisting protocols to evaluate their efficacies and side effects. The comparison underscores the challenges of social acceptance and equity, particularly in vaccine production and distribution. As the world faces unknown agents, the three major types of MCMs do not have equal and symmetrical effects in terms of epidemic control. Thus, a vaccine-oriented strategy with a community-centered approach, proves essential for effective pandemic preparedness, encouraging continued innovation in vaccinology.
Collapse
Affiliation(s)
- Marie Mura
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
| | - Aurélie Trignol
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
| | - Erwan Le Dault
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
- Department of Infectious Diseases and Tropical Medicine, Laveran Military Teaching Hospital, Marseille, France
| | - Jean-Nicolas Tournier
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
- École du Val-de-Grâce, Paris, France
| |
Collapse
|
2
|
Wachter BT, Xu Q, Shi L, Burbelo PD, Myint-Hpu K, Schwartzberg PL, Rehman MT, Dewar RL, Boswell KL, Koup RA, Oguz C, Imberti L, Bellusci L, Pourhashemi S, Khurana S, Manthiram K, Notarangelo LD, Delmonte OM. Prior SARS-CoV-2 infection affects adaptive immune responses to Omicron BA.4/BA.5 mRNA booster. J Allergy Clin Immunol 2025; 155:2038-2051. [PMID: 40044048 PMCID: PMC12145247 DOI: 10.1016/j.jaci.2025.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Bivalent coronavirus disease 2019 (COVID) mRNA vaccines encoding Wuhan-1 and Omicron BA.4/BA.5 spike proteins (S) can prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, but the quality of adaptive immune responses and the importance of hybrid immunity are not well documented. OBJECTIVES Adaptive immune responses to the bivalent vaccine were studied in 40 healthy participants with (COVID+) or without (COVID-) history of SARS-CoV-2 infection. METHODS We analyzed anti-nucleocapsid protein and anti-S IgG titers and surrogate virus neutralization capacity against variants of concern and assessed SARS-CoV-2-specific B- and T-cell responses by high-dimensional spectral flow cytometry, intracellular cytokine staining assay on stimulation with SARS-CoV-2 peptides, and TRB and IGH repertoire analysis. RESULTS The COVID+ group had higher anti-S IgG levels before and after boost and higher neutralization activity against BA.4/BA.5 than the COVID- group. Spike antibody levels positively correlated with neutralizing activity against Omicron variants of concern in all participants. For variants of concern, lowest neutralization capacity was against XBB.1.5. At baseline, the proportion of S1+RBD+ B cells was higher in COVID+ than in COVID- subjects, but an increase of these cells after boost was detected only in the COVID- group. Consistent with natural infection, COVID+ subjects had a higher frequency of IgA+CXCR3+S1+RBD+ B cells at baseline than COVID- subjects. CD4+ memory T-cell responses and breath of class II epitope SARS-CoV-2-specific clonotypes were increased after boost only in COVID- participants. CONCLUSIONS The bivalent vaccine induces robust adaptive immune responses against the Omicron variant. Prior SARS-CoV-2 infection provides increased protection, but optimal timing of booster administration after natural infection should be defined to maximize benefits.
Collapse
Affiliation(s)
- Brianna T Wachter
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Qin Xu
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, Md
| | - Lihong Shi
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, Md
| | - Peter D Burbelo
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Md
| | - Kathy Myint-Hpu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | | | | | - Robin L Dewar
- Virus Isolation and Serology Laboratory, Frederick National Laboratory, Frederick, Md
| | - Kristin L Boswell
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, Md
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, Md
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, NIAID, NIH, Bethesda, Md
| | - Luisa Imberti
- Spedali Civili di Brescia, Universita' Degli Studi di Brescia, Brescia, Italy
| | - Lorenza Bellusci
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Md
| | - Sara Pourhashemi
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Md
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Md
| | | | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md.
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md.
| |
Collapse
|
3
|
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
|
4
|
Baum HE, Santopaolo M, Francis O, Milodowski EJ, Entwistle K, Oliver E, Hitchings B, Diamond D, Thomas AC, Mitchell RE, Kibble M, Gupta K, Di Bartolo N, Klenerman P, Brown A, Morales-Aza B, Oliver J, Berger I, Toye AM, Finn A, Goenka A, Davidson AD, Ring S, Molloy L, Lewcock M, Northstone K, Roth F, Timpson NJ, Wooldridge L, Halliday A, Rivino L. Hybrid B- and T-Cell Immunity Associates With Protection Against Breakthrough Infection After Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination in Avon Longitudinal Study of Parents and Children (ALSPAC) Participants. J Infect Dis 2025:jiaf246. [PMID: 40392230 DOI: 10.1093/infdis/jiaf246] [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: 01/06/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Immunological memory to vaccination and viral infection involves the coordinated action of B and T cells; thus, integrated analysis of these 2 components is critical for understanding their respective contributions to protection against breakthrough infections (BIs) after vaccination. METHODS We investigated cellular and humoral immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and/or vaccination in 300 adult participants from the Avon Longitudinal Study of Parents and Children (ALSPAC). Participants were grouped by those with (cases) and without (controls) a history of SARS-CoV-2 infection. To provide a quantitative correlate for protection against BI in the 8-month period after the study, Youden index thresholds were calculated for all immune measures analyzed. RESULTS The magnitude of antibody and T-cell responses following the second vaccine dose was associated with protection against BI in participants with a history of SARS-CoV-2 infection (cases), but not in infection-naive controls. Over 8 months of follow-up, 2 threshold combinations provided the best performance for protection against BI in cases: (i) anti-spike immunoglobulin G (IgG) (≥666.4 binding antibody units [BAU]/mL) combined with anti-nucleocapsid pan-immunoglobulin (pan-Ig) (≥0.1332 BAU/mL) and (ii) spike 1-specific T cells (≥195.6 spot-forming units/106 peripheral blood mononuclear cells) combined with anti-N pan-Ig (≥0.1332 BAU/mL). Both combinations offered 100% specificity for detecting cases without BI, with sensitivities of 83.3% and 72.2%, respectively. CONCLUSIONS Collectively, these results suggest that hybrid B- and T-cell immunity offers superior protection from BI after coronavirus disease 2019 (COVID-19) vaccination, and this finding has implications for designing next-generation COVID-19 vaccines that are capable of eliciting immunity to a broader repertoire of SARS-CoV-2 proteins.
Collapse
Affiliation(s)
- Holly E Baum
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Marianna Santopaolo
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Ore Francis
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Emily J Milodowski
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Katrina Entwistle
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Elizabeth Oliver
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Benjamin Hitchings
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Divya Diamond
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Amy C Thomas
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ruth E Mitchell
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Milla Kibble
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Kapil Gupta
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Natalie Di Bartolo
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Paul Klenerman
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Begonia Morales-Aza
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Jennifer Oliver
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Imre Berger
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Ash M Toye
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Adam Finn
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Paediatric Immunology and Infectious Diseases, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Anu Goenka
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Paediatric Immunology and Infectious Diseases, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Andrew D Davidson
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Susan Ring
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Lynn Molloy
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Melanie Lewcock
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kate Northstone
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Firona Roth
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Nicholas J Timpson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Linda Wooldridge
- Bristol Veterinary School, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Alice Halliday
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Vaccine Centre, University of Bristol, Bristol, United Kingdom
| | - Laura Rivino
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
5
|
Waki K, Tani H, Kawahara E, Saga Y, Shimada T, Yamazaki E, Koike S, Morinaga Y, Isobe M, Kurosawa N. Comprehensive analysis of nasal IgA antibodies induced by intranasal administration of the SARS-CoV-2 spike protein. eLife 2025; 12:RP88387. [PMID: 40338637 PMCID: PMC12061477 DOI: 10.7554/elife.88387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
Intranasal vaccination is an attractive strategy for preventing COVID-19 disease as it stimulates the production of multimeric secretory immunoglobulin A (IgA), the predominant antibody isotype in the mucosal immune system, at the target site of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry. Currently, intranasal vaccine efficacy is evaluated based on the measurement of polyclonal antibody titers in nasal lavage fluid. However, how individual multimeric secretory IgA protects the mucosa from SARS-CoV-2 infection remains to be elucidated. To understand the precise contribution and molecular nature of multimeric secretory IgA induced by intranasal vaccines, we developed 99 monoclonal IgA clones from nasal mucosa and 114 monoclonal IgA or IgG clones from nonmucosal tissues of mice that were intranasally immunized with the SARS-CoV-2 spike protein. The nonmucosal IgA clones exhibited shared origins and common and unique somatic mutations with the related nasal IgA clones, indicating that the antigen-specific plasma cells in the nonmucosal tissues originated from B cells stimulated at the nasal mucosa. Comparing the spike protein binding reactivity, angiotensin-converting enzyme-2-blocking, and in vitro SARS-CoV-2 virus neutralization of monomeric and multimeric secretory IgA pairs recognizing different epitopes showed that even non-neutralizing monomeric IgAs, which represent 70% of the nasal IgA repertoire, can protect against SARS-CoV-2 infection when expressed as multimeric secretory IgAs. We also demonstrated that the intranasal administration of multimeric secretory IgA delivered as prophylaxis in the hamster model reduced infection-induced weight loss. Our investigation is the first to demonstrate the function of nasal IgA at the monoclonal level, showing that nasal immunization can provide effective immunity against SARS-CoV-2 by inducing multimeric secretory IgAs at the target site of the virus infection.
Collapse
Affiliation(s)
- Kentarou Waki
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Education, University of ToyamaToyamaJapan
| | - Hideki Tani
- Department of Virology, Toyama Institute of HealthToyamaJapan
| | - Eigo Kawahara
- Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical SciencesToyamaJapan
- Center for Advanced Antibody Drug Development, University of ToyamaToyamaJapan
| | - Yumiko Saga
- Department of Virology, Toyama Institute of HealthToyamaJapan
| | | | - Emiko Yamazaki
- Department of Virology, Toyama Institute of HealthToyamaJapan
| | - Seiichi Koike
- Laboratory of Molecular and Cellular Biology, Graduate School of Innovative Life Science, University of ToyamaToyamaJapan
| | - Yoshitomo Morinaga
- Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical SciencesToyamaJapan
- Center for Advanced Antibody Drug Development, University of ToyamaToyamaJapan
| | - Masaharu Isobe
- Center for Advanced Antibody Drug Development, University of ToyamaToyamaJapan
- Laboratory of Molecular and Cellular Biology, Graduate School of Innovative Life Science, University of ToyamaToyamaJapan
| | - Nobuyuki Kurosawa
- Center for Advanced Antibody Drug Development, University of ToyamaToyamaJapan
- Laboratory of Molecular and Cellular Biology, Graduate School of Innovative Life Science, University of ToyamaToyamaJapan
| |
Collapse
|
6
|
Breznik JA, Cowbrough B, Bilaver L, Dushoff M, Stacey HD, Ang J, Clare R, Kennedy A, Costa AP, Nazy I, Loeb M, Verschoor CP, Bramson J, Miller MS, Bowdish DME. Minimal Impact of Prior Common Cold Coronavirus Exposure on Immune Responses to Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination or Infection Risk in Older Adults in Congregate Care. Open Forum Infect Dis 2025; 12:ofaf178. [PMID: 40313478 PMCID: PMC12044602 DOI: 10.1093/ofid/ofaf178] [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: 11/26/2024] [Accepted: 03/19/2025] [Indexed: 05/03/2025] Open
Abstract
Background Common cold coronaviruses were a frequent cause of respiratory infections in older adults living in congregate care homes before the coronavirus disease 2019 pandemic, which may influence immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination and infection. We investigated humoral and cellular immune responses to prior common cold coronaviruses and SARS-CoV-2, how they are affected by SARS-CoV-2 vaccination and infection, and their associations with Omicron BA.1 SARS-CoV-2 infections in residents of long-term care and retirement homes. Methods In SARS-CoV-2 infection-naive residents with 3 monovalent messenger RNA SARS-CoV-2 vaccinations, we measured serum anti-receptor binding domain (RBD) immunoglobulin (Ig) G and IgA antibody titers against SARS-CoV-2 and common cold human coronavirus (HCoV) NL63, HCoV-OC43, and HCoV-229E; ancestral and Omicron BA.1 neutralizing antibodies; and CD4+ and CD8+ T-cell activation responses to membrane, nucleocapsid, and spike proteins. We examined the relationships of common cold coronavirus and SARS-CoV-2 humoral immune responses, whether antibody and T-cell responses changed after SARS-CoV-2 messenger RNA vaccination or infection, and their associations with Omicron BA.1 infection. Results Anti-RBD IgG HCoV-OC43 titers were positively correlated with SARS-CoV-2 anti-RBD IgG and neutralizing antibody titers. Common cold coronavirus anti-RBD IgA titers, but not anti-RBD IgG titers, increased after SARS-CoV-2 vaccination or infection, and many residents had cross-reactive T cells. Common cold coronavirus humoral immunity was similar in residents without and those with subsequent Omicron BA.1 infection. Conclusions Despite frequent exposure, and associations of common cold coronavirus and vaccine-induced SARS-CoV-2 humoral immunity, preexisting common cold coronavirus immunity was not associated with Omicron BA.1 infection in residents of long-term care and retirement communities.
Collapse
Affiliation(s)
- Jessica A Breznik
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
| | - Braeden Cowbrough
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lucas Bilaver
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Miriam Dushoff
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Hannah D Stacey
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jann Ang
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Rumi Clare
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Allison Kennedy
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Andrew P Costa
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mark Loeb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Chris P Verschoor
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Health Sciences North Research Institute, Health Sciences North, Sudbury, Ontario, Canada
| | - Jonathan Bramson
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Matthew S Miller
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Dawn M E Bowdish
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Saha A, Ghosh Roy S, Dwivedi R, Tripathi P, Kumar K, Nambiar SM, Pathak R. Beyond the Pandemic Era: Recent Advances and Efficacy of SARS-CoV-2 Vaccines Against Emerging Variants of Concern. Vaccines (Basel) 2025; 13:424. [PMID: 40333293 PMCID: PMC12031379 DOI: 10.3390/vaccines13040424] [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: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccination has been instrumental in curbing the transmission of SARS-CoV-2 and mitigating the severity of clinical manifestations associated with COVID-19. Numerous COVID-19 vaccines have been developed to this effect, including BioNTech-Pfizer and Moderna's mRNA vaccines, as well as adenovirus vector-based vaccines such as Oxford-AstraZeneca. However, the emergence of new variants and subvariants of SARS-CoV-2, characterized by enhanced transmissibility and immune evasion, poses significant challenges to the efficacy of current vaccination strategies. In this review, we aim to comprehensively outline the landscape of emerging SARS-CoV-2 variants of concern (VOCs) and sub-lineages that have recently surfaced in the post-pandemic years. We assess the effectiveness of existing vaccines, including their booster doses, against these emerging variants and subvariants, such as BA.2-derived sub-lineages, XBB sub-lineages, and BA.2.86 (Pirola). Furthermore, we discuss the latest advancements in vaccine technology, including multivalent and pan-coronavirus approaches, along with the development of several next-generation coronavirus vaccines, such as exosome-based, virus-like particle (VLP), mucosal, and nanomaterial-based vaccines. Finally, we highlight the key challenges and critical areas for future research to address the evolving threat of SARS-CoV-2 subvariants and to develop strategies for combating the emergence of new viral threats, thereby improving preparedness for future pandemics.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Sounak Ghosh Roy
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD 20910, USA;
| | - Richa Dwivedi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Kamal Kumar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA;
| | - Shashank Manohar Nambiar
- Division of Hepatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
8
|
Maltseva M, Galipeau Y, McCluskie P, Castonguay N, Cooper CL, Langlois MA. Systemic and Mucosal Antibody Responses to SARS-CoV-2 Variant-Specific Prime-and-Boost and Prime-and-Spike Vaccination: A Comparison of Intramuscular and Intranasal Bivalent Vaccine Administration in a Murine Model. Vaccines (Basel) 2025; 13:351. [PMID: 40333249 PMCID: PMC12031244 DOI: 10.3390/vaccines13040351] [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: 02/28/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 05/09/2025] Open
Abstract
Background: The rapid genetic evolution of SARS-CoV-2 has led to the emergence of immune-evading, highly transmissible variants of concern (VOCs). This prompts the need for next-generation vaccines that elicit robust mucosal immunity in the airways to directly curb viral infection. Objective: Here, we investigate the impact of heterologous variant prime-boost regimens on humoral responses, focusing on intramuscular (IM) and intranasal (IN) routes of administration. Using a murine model, we assessed the immunogenicity of unadjuvanted protein boosts with Wu-1, Omicron BA.4/5, or Wu-1 + BA.4/5 spike antigens following monovalent or bivalent IM priming with mRNA-LNP vaccines. Results: IM priming induced strong systemic total and neutralizing antibody responses that were further enhanced by IN boosts with BA.4/5. IN boosting achieved the broadest serum neutralization across all VOCs tested. Notably, bivalent mRNA-LNP IM priming induced robust, cross-variant serum neutralizing antibody production, independent of subsequent IN boost combinations. Conclusions: Our findings highlight the benefit of including distinct antigenic variants in the prime vaccination followed by a variant-tailored IN boost to elicit both systemic and mucosal variant-specific responses that are potentially capable of reducing SARS-CoV-2 transmission.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Pauline McCluskie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nicolas Castonguay
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Curtis L. Cooper
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
9
|
Fant P, Laurent S, Desert P, Combadière B, Palazzi X, Choudhary S, Gervais F, Broudic K, Rossi R, Gauthier BE. Proceedings of the 2023 Annual Scientific Meeting of the French Society of Toxicologic Pathology (SFPT) on Preclinical Development and Therapeutic Applications of mRNA-Based Technologies. Toxicol Pathol 2025:1926233251326089. [PMID: 40110665 DOI: 10.1177/01926233251326089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The 2023 annual scientific meeting of the French Society of Toxicologic Pathology (Société Française de Pathologie Toxicologique, SFPT), entitled "mRNA-based technologies: preclinical development and therapeutic applications," was held in Lyon (France) on May 25 to 26, 2023. The aim of the meeting was to discuss the biology, immunology, and preclinical development of messenger RNA (mRNA)-based vaccines and therapeutics, including immuno-oncology and rare diseases, as well as the regulatory aspect of the COVID-19 vaccines and an overview of the principles and applications of in situ hybridization techniques. This article presents the summary of five lectures along with selected figures, tables, and key literature references on this topic.
Collapse
Affiliation(s)
- Pierluigi Fant
- Charles River Laboratories Safety Assessment, Saint Germain-Nuelles, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Korosec CS, Conway JM, Matveev VA, Ostrowski M, Heffernan JM, Ghaemi MS. Machine Learning Reveals Distinct Immunogenic Signatures of Th1 Imprinting in ART-Treated Individuals with HIV Following Repeated SARS-CoV-2 Vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643769. [PMID: 40166325 PMCID: PMC11956973 DOI: 10.1101/2025.03.18.643769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The human immune system is intrinsically variable and remarkably diverse across a population. The immune response to antigens is driven by a complex interplay of time-dependent interdependencies across components of the immune system. After repeated vaccination, the humoral and cellular arms of the immune response display highly heterogeneous dynamics, further complicating the attribution of a phenotypic outcome to specific immune system components. We employ a random forest (RF) approach to classify informative differences in immunogenicity between older people living with HIV (PLWH) on ART and an age-matched control group who received up to five SARS-CoV-2 vaccinations over 104 weeks. RFs identify immunological variables of importance, interpreted as evidence for Th1 imprinting, and suggest novel distinguishing immune features, such as saliva-based antibody screening, as promising diagnostic features towards classifying responses (whereas serum IgG is not). Additionally, we implement supervised and unsupervised Machine Learning methods to produce physiologically accurate synthetic datasets that conform to the statistical distribution of the original immunological data, thus enabling further data-driven hypothesis testing and model validation. Our results highlight the effectiveness of RFs in utilizing informative immune feature interdependencies for classification tasks and suggests broad impacts of ML applications for personalized vaccination strategies among high-risk populations.
Collapse
|
11
|
Samaan P, Korosec CS, Budylowski P, Chau SLL, Pasculescu A, Qi F, Delgado-Brand M, Tursun TR, Mailhot G, Dayam RM, Arnold CR, Langlois MA, Mendoza J, Morningstar T, Law R, Mihelic E, Sheikh-Mohamed S, Cao EY, Paul N, Patel A, de Launay KQ, Boyd JM, Takaoka A, Colwill K, Matveev V, Yue FY, McGeer A, Straus S, Gingras AC, Heffernen JM, Ostrowski M. mRNA vaccine-induced SARS-CoV-2 spike-specific IFN-γ and IL-2 T-cell responses are predictive of serological neutralization and are transiently enhanced by pre-existing cross-reactive immunity. J Virol 2025; 99:e0168524. [PMID: 39887249 PMCID: PMC11915849 DOI: 10.1128/jvi.01685-24] [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: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
The contributions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells to vaccine efficacy and durability are unclear. We investigated relationships between mRNA vaccine-induced spike-specific interferon- gamma (IFN-γ) and interleukin-2 (IL-2) T-cell responses and neutralizing antibody development in long-term care home staff doubly vaccinated with BNT162b2 or mRNA-1273. The impacts of pre-existing cross-reactive T-cell immunity on cellular and humoral responses to vaccination were additionally assessed. Mathematical modeling of the kinetics of spike-specific IFN-γ and IL-2 T-cell responses over 6 months post-second dose was bifurcated into recipients who exhibited gradual increases with doubling times of 155 and 167 days or decreases with half-lives of 165 and 132 days, respectively. Differences in kinetics did not correlate with clinical phenotypes. Serological anti-spike IgG, anti-receptor binding domain (RBD) IgG, anti-spike IgA, and anti-RBD IgA antibody levels otherwise decayed in all participants with half-lives of 63, 57, 79, and 46 days, respectively, alongside waning neutralizing capacity (t1/2 = 408 days). Spike-specific T-cell responses induced at 2-6 weeks positively correlated with live viral neutralization at 6 months post-second dose, especially in hybrid immune individuals. Participants with pre-existing cross-reactive T-cell immunity to SARS-CoV-2 exhibited greater spike-specific T-cell responses, reduced anti-RBD IgA antibody levels, and a trending increase in neutralization at 2-6 weeks post-second dose. Non-spike-specific T-cells predominantly targeted SARS-CoV-2 non-structural protein at 6 months post-second dose in cross-reactive participants. mRNA vaccination was lastly shown to induce off-target T-cell responses against unrelated antigens. In summary, vaccine-induced spike-specific T-cell immunity appeared to influence serological neutralizing capacity, with only a modest effect induced by pre-existing cross-reactivity. IMPORTANCE Our findings provide valuable insights into the potential contributions of mRNA vaccine-induced spike-specific T-cell responses to the durability of neutralizing antibody levels in both uninfected and hybrid immune recipients. Our study additionally sheds light on the precise impacts of pre-existing cross-reactive T-cell immunity to severe acute respiratory syndrome coronavirus 2 on the magnitude and kinetics of cellular and humoral responses to vaccination. Accordingly, our data will help optimize the development of next-generation T cell-based coronavirus vaccines and vaccine regimens to maximize efficacy and durability.
Collapse
Affiliation(s)
- Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Chapin S. Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, Ontario, Canada
- Center for Disease Modelling, Mathematics and Statistics, York University, Toronto, Ontario, Canada
| | - Patrick Budylowski
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Serena L. L. Chau
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Pasculescu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Freda Qi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | | | - Tulunay R. Tursun
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Geneviève Mailhot
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Roya Monica Dayam
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Corey R. Arnold
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Justin Mendoza
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Ryan Law
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Erik Mihelic
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Eric Yixiao Cao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nimitha Paul
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Anjali Patel
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | | | - Jamie M. Boyd
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Alyson Takaoka
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Vitaliy Matveev
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Feng Yun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - Sharon Straus
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jane M. Heffernen
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, Toronto, Ontario, Canada
- Center for Disease Modelling, Mathematics and Statistics, York University, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada
- Keenan Research Center for Biomedical Science, St Michael's Hospital Keenan, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Liu B, Gu Y, Ou Y, Liu L, Wang W, Zhou J, Wang Y, Du Y, Xie J, Liu Y, Zhang R, Zuo Q, Wang B. Protection conferred by mucosal novel bivalent Klebsiella pneumoniae vaccine immunization associates with presence of lung CD4 + T RM. Microbes Infect 2025:105483. [PMID: 40081566 DOI: 10.1016/j.micinf.2025.105483] [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: 10/31/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Klebsiella pneumoniae is the principal cause of hospital-acquired infection with a high morbidity and mortality in immunocompromised individuals, yet no vaccine is approved. Here, we developed a novel bivalent subunit vaccine for the prevention of K. pneumoniae infection based on the outer membrane protein GlnH and the fimbriae protein FimA. The survival rate of immunized mice was significantly increased compared to that of unimmunized mice, while the bacterial burden, weight loss, and lung pathology were drastically reduced. Furthermore, vaccine-elicited CD4+ TRM cells were observed in lung tissues and appeared to play a critical role in vaccine efficacy. Transcriptomic analysis of total lung tissues from mice treated by FTY720 (S1PR1 inhibitor that blocks lymphocyte egress from secondary lymphoid structures) showed that cell activation, cytokine secretion and enhancement of the killing ability of neutrophils were related to the mechanism of protection against K. pneumoniae infection. These findings indicate that GlnH and FimA are effective candidate bivalent vaccine to fight K. pneumoniae infection.
Collapse
Affiliation(s)
- BiXia Liu
- College of Medicine, Southwest Jiaotong University, Chengdu, 610083, PR China
| | - YaRu Gu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 401320, PR China
| | - YangXue Ou
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - LuXuan Liu
- Affiliated Dongfeng Hospital, Hubei University of Medicine, Shiyan, 442000, PR China
| | - WenHao Wang
- College of Pharmacy, Henan University, Kaifeng, 475001, PR China
| | - JinRui Zhou
- College of Medicine, Southwest Jiaotong University, Chengdu, 610083, PR China
| | - Ying Wang
- 953rd Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, Shigatse, 857000, PR China
| | - YeXiang Du
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Jing Xie
- Clinical Laboratory, Chengdu Military General Hospital, Chengdu, 610083, PR China
| | - Yuan Liu
- Clinical Laboratory, Chengdu Military General Hospital, Chengdu, 610083, PR China
| | - Rui Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610083, PR China; Clinical Laboratory, Chengdu Military General Hospital, Chengdu, 610083, PR China.
| | - QianFei Zuo
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China.
| | - Bin Wang
- Department of Respiratory Medicine, Second Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
13
|
Liu Y, Meng F, Feng W, Chen Z, Xing H, Zheng A. Oral DNA Vaccine Utilizing the Yeast Cell Wall for Dectin-1 Receptor-Mediated Enhancement of Mucosal Immunity. Mol Pharm 2025; 22:1241-1252. [PMID: 39960883 DOI: 10.1021/acs.molpharmaceut.4c00943] [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: 03/04/2025]
Abstract
Mucosal vaccines can generate localized mucosal immunity, effectively preventing initial pathogen infection and providing more effective protection. Oral vaccines are an attractive option for inducing mucosal immunity. The yeast cell wall, primarily composed of natural β-1,3-d glucan, can be recognized by the apical membrane receptor, dectin-1, which has a high expression on macrophages and intestinal M cells. In this study, by using vortexing methods to break yeast cell walls into nanometer-sized fragments, which retain the negatively charged β-glucan components on their surface and employing electrostatic adsorption/coextrusion techniques, these fragments were attached onto the surface of PS-DNA NPs, as verified by a scanning electron microscope (SEM), a transmission electron microscope (TEM), and dynamic light scattering (DLS) data. YCW-coated NPs (YNPs) showed greater drug stability compared to NPs in a simulated gastrointestinal environment. In vitro cell evaluation further demonstrated that YNPs were rapidly and efficiently taken up by antigen-presenting cells via receptor dectin-1-mediated endocytosis. In vivo experiments revealed that the oral vaccine elicited high levels of RBD-specific antibodies and triggered extensive cellular immunity in the intestinal mucosa. This study provides new insights into mucosal vaccine research.
Collapse
Affiliation(s)
- Yingqi Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fan Meng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wanting Feng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zehong Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Haonan Xing
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aiping Zheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
14
|
Venturi G, Gallinaro A, Fortuna C, Pirillo MF, Scoglio A, Di Carlo B, Marsili G, Michelini Z, Amendola A, Carocci A, Dispinseri S, Borghi M, Canitano A, Falce C, Zappitelli A, Scarlatti G, Lixi ML, Aste A, Masala L, Baroncelli S, Cara A, Negri D. Viral and immune profiles during the first wave of SARS-CoV-2 infection in hospitalized patients in Sardinia, Italy. Sci Rep 2025; 15:6660. [PMID: 39994243 PMCID: PMC11850715 DOI: 10.1038/s41598-025-90324-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
We performed a retrospective immunological analysis of the antibody response in serum and in nasopharyngeal swabs (NPS) obtained from 46 individuals infected with ancestral SARS-CoV-2 Wuhan-Hu-1 strain during the first COVID-19 wave in Cagliari (Sardinia, Italy), with a 4-month follow-up after the hospital admission. We implemented a comprehensive antibody response in serum and in mucosal samples using assays established in our laboratories. In NPS we evaluated the viral load by real time PCR, presence and kinetics of anti-Spike IgG and IgA by ELISA as well as their anti-Wuhan neutralization activity, showing induction and persistence of anti-viral immunity at the mucosal level. Neutralizing antibodies were measured in serum and NPS using a safe pseudovirus-based assay validated after comparison with a standard neutralization test using live SARS-CoV-2. We evaluated cross-neutralizing antibodies against all the major early variants of concerns (VoC) in sera. Of note, we detected a remarkable reduction of neutralizing activity against BA.1 compared to BA.2 and BA.5 Omicron subvariants, which was confirmed in sera from an analogous cohort of patients at the San Raffaele hospital in Milan, a geographically distant region of Italy, infected with the ancestral virus during the same period of time.
Collapse
Affiliation(s)
- Giulietta Venturi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Claudia Fortuna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Arianna Scoglio
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, Rome, Italy
| | - Beatrice Di Carlo
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, Rome, Italy
| | - Giulia Marsili
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Antonello Amendola
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Alberto Carocci
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Dispinseri
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Falce
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alice Zappitelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maria Luisa Lixi
- Laboratory Medicine, Santissima Trinità Hospital, Cagliari, Italy
| | - Alessandra Aste
- Laboratory Medicine, Santissima Trinità Hospital, Cagliari, Italy
| | - Laura Masala
- Laboratory Medicine, Santissima Trinità Hospital, Cagliari, Italy
| | - Silvia Baroncelli
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
15
|
De Voss CJ, Korompis M, Li S, Ateere A, McShane H, Stylianou E. Novel mRNA vaccines induce potent immunogenicity and afford protection against tuberculosis. Front Immunol 2025; 16:1540359. [PMID: 40018046 PMCID: PMC11865049 DOI: 10.3389/fimmu.2025.1540359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a disease with a severe global burden. The intractability of Mtb has prevented the identification of clear correlates of protection against TB and hindered the development of novel TB vaccines that are urgently required. Lipid nanoparticle (LNP)-formulated mRNA is a highly promising vaccine platform that has yet to be thoroughly applied to TB. Methods We selected five Mtb antigens (PPE15, ESAT6, EspC, EsxI, MetE) and evaluated their potential as LNP-formulated mRNA vaccines, both when each antigen was delivered individually, and when all five antigens were combined in a mix regimen (m-Mix). Results Each mRNA construct demonstrated unique cellular and humoral immunogenicity, and both m-Mix, as well as the single antigen EsxI, conferred significant protection in a murine Mtb challenge model. Whilst the potent immune responses of each mRNA were maintained when applied as a boost to BCG, there was no additional increase to the efficacy of BCG. Combination of m-Mix with a recombinant, replication-deficient chimpanzee adenovirus (ChAdOx1), in a heterologous prime-boost delivery (C-m-Mix), appeared to result in increased protection upon murine Mtb infection, than either regimen alone. Discussion This work warrants further investigation of LNP-formulated mRNA vaccines for TB, whilst indicating the potential of m-Mix and C-m-Mix to progress to further stages of vaccine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Elena Stylianou
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
| |
Collapse
|
16
|
Pilapitiya D, Lee WS, Vu MN, Kelly A, Webster RH, Koutsakos M, Kent SJ, Juno JA, Tan HX, Wheatley AK. Mucosal vaccination against SARS-CoV-2 using recombinant influenza viruses delivering self-assembling nanoparticles. Vaccine 2025; 46:126668. [PMID: 39740385 DOI: 10.1016/j.vaccine.2024.126668] [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: 08/30/2024] [Revised: 11/07/2024] [Accepted: 12/21/2024] [Indexed: 01/02/2025]
Abstract
Recombinant influenza viruses are promising vectors that can bolster antibody and resident lymphocyte responses within mucosal sites. This study evaluates recombinant influenza viruses with SARS-CoV-2 RBD genes in eliciting mucosal and systemic responses. Using reverse genetics, we generated replication-competent recombinant influenza viruses carrying heterologous RBD genes in monomeric, trimeric, or ferritin-based nanoparticle forms. Following intranasal immunisation, mice developed potent serological anti-RBD responses, with ferritin nanoparticles superseding monomeric or trimeric RBD responses. While parenteral and mucosal immunisation elicited robust anti-RBD IgG in serum, mucosal immunisation seeded respiratory IgA, RBD-specific lung-resident memory and germinal centre (GC) B cells. In animals with prior intramuscular vaccination, intranasal boosting with recombinant influenza vectors augmented mucosal IgG, IgA, GC and memory B cells, and SARS-CoV-2 lung neutralising titres. Recall of RBD-specific memory B cells via antigen re-exposure in the lung increased antibody-secreting cells in the lung-draining lymph nodes, with maintenance of lung GC B cells. Recombinant influenza-based vaccines effectively deliver highly immunogenic self-assembling nanoparticles, generating antibodies and B cells in the respiratory mucosa. This strategy provides a tractable pathway to augment lung-localised responses against recurrent respiratory viral infections.
Collapse
MESH Headings
- Animals
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Mice
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Female
- Administration, Intranasal
- Immunity, Mucosal
- Immunoglobulin G/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Mice, Inbred BALB C
- Antibodies, Neutralizing/immunology
- Immunoglobulin A/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Vaccination/methods
- Lung/immunology
- B-Lymphocytes/immunology
- Humans
- Orthomyxoviridae/genetics
- Orthomyxoviridae/immunology
Collapse
Affiliation(s)
- Devaki Pilapitiya
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Mai N Vu
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Andrew Kelly
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Rosela H Webster
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
17
|
Erbs G, Jakobsen JT, Schmidt ST, Christensen D, Bailey M, Jungersen G. Retinoic acid-adjuvanted vaccine induces antigen-specific secretory IgA in the gut of newborn piglets. Vaccine 2025; 46:126672. [PMID: 39733479 DOI: 10.1016/j.vaccine.2024.126672] [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: 06/26/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
Mucosal secretory IgA (SIgA) produced by subepithelial plasma cells in the lamina propria is the major antigen-specific defense mechanism against mucosal infections. We investigated if a retinoic acid (RA)-containing adjuvant in parenteral immunization, can induce vaccine-specific SIgA in the jejunal lumen in a dose-dependent manner in neonatal pigs immunized with a Chlamydia hybrid antigen. To accurately quantify SIgA responses in mucosal secretions, an antigen-specific ELISA method with secondary detection of porcine secretory component rather than IgA was developed. RA facilitated a stronger (or faster) IgG, IgA, IgM and SIgA response in serum after primary immunization, and a more than 10-fold significantly increased level of vaccine-specific SIgA in jejunum at termination 2 weeks after the secondary boost, whereas IgA or SIgA responses in bronchoalveolar lavage (BAL) were not significantly increased after immunization with RA. Analyses of different isotype responses to vaccination and different sampling sites, revealed significant correlations between IgG and IgA responses in serum, and between IgG in serum and jejunum, while IgA in jejunum was neither correlated with IgA in serum nor with IgG in jejunum. This is indicative of IgG in jejunum being primarily a transudate from serum, while IgA is not. Jejunum SIgA correlated significantly with jejunum IgA and with both serum SIgA and IgA. Our results thus support the use of SC-specific reagents for mucosal SIgA responses, although IgA reagents to a lesser extent also reflects local antibodies. Although the IgA and SIgA levels in BAL were not significantly different with or without RA, we observed a significant correlation of vaccine-specific SIgA in jejunum and BAL, indicating a level of commonality in the regulation of mucosal antibodies in gut and respiratory system. In conclusion, an adjuvant with high concentration of RA was shown to increase the local intestinal mucosal antibody response after parenteral immunization in pigs.
Collapse
Affiliation(s)
- Gitte Erbs
- Infectious Disease Immunology, Center for Vaccine Research, SSI, Copenhagen, Denmark
| | - Jeanne Toft Jakobsen
- Infectious Disease Immunology, Center for Vaccine Research, SSI, Copenhagen, Denmark
| | - Signe Tandrup Schmidt
- Infectious Disease Immunology, Center for Vaccine Research, SSI, Copenhagen, Denmark
| | - Dennis Christensen
- Infectious Disease Immunology, Center for Vaccine Research, SSI, Copenhagen, Denmark
| | - Mick Bailey
- Bristol Veterinary School, Langford House, University of Bristol, UK
| | - Gregers Jungersen
- Infectious Disease Immunology, Center for Vaccine Research, SSI, Copenhagen, Denmark.
| |
Collapse
|
18
|
Zhou M, Xiao H, Yang X, Cheng T, Yuan L, Xia N. Novel vaccine strategies to induce respiratory mucosal immunity: advances and implications. MedComm (Beijing) 2025; 6:e70056. [PMID: 39830020 PMCID: PMC11739453 DOI: 10.1002/mco2.70056] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/31/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Rapid advances in vaccine technology are becoming increasingly important in tackling global health crises caused by respiratory virus infections. While traditional vaccines, primarily administered by intramuscular injection, have proven effective, they often fail to provide the broad upper respiratory tract mucosal immunity, which is urgently needed for first-line control of respiratory viral infections. Furthermore, traditional intramuscular vaccines may not adequately address the immune escape of emerging virus variants. In contrast, respiratory mucosal vaccines developed using the body's mucosal immune response mechanism can simultaneously establish both systemic and mucosal immunity. This dual action effectively allows the respiratory mucosal immune system to function as the first line of defense, preventing infections at the entry points. This review highlights the efficacy of respiratory mucosal vaccines, including innovative delivery methods such as nasal and oral formulations, in enhancing local and systemic immune barriers. Notably, respiratory mucosal vaccines offer potential advantages in protecting against emerging virus variants and maintaining long-term and multidimensional immune memory in the upper respiratory tract. In addition, a combination of intramuscular and respiratory mucosal delivery of vaccines largely improves their coverage and effectiveness, providing valuable insights for future vaccine development and public inoculation strategies.
Collapse
Affiliation(s)
- Ming Zhou
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Haiqin Xiao
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Xinyi Yang
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| |
Collapse
|
19
|
Slamanig S, Lemus N, Lai TY, Singh G, Mishra M, Abdeljawad A, Boza M, Dolange V, Singh G, Lee B, González-Domínguez I, Schotsaert M, Krammer F, Palese P, Sun W. A single immunization with intranasal Newcastle disease virus (NDV)-based XBB.1.5 variant vaccine reduces disease and transmission in animals against matched-variant challenge. Vaccine 2025; 45:126586. [PMID: 39667115 DOI: 10.1016/j.vaccine.2024.126586] [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: 08/06/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
The rapid development of coronavirus disease 2019 (COVID-19) vaccines has helped mitigate the initial impact of the pandemic. However, in order to reduce transmission rates and protect more vulnerable and immunocompromised individuals unable to mount an effective immune response, development of a next-generation of mucosal vaccines is necessary. Here, we developed an intranasal Newcastle disease virus (NDV)-based vaccine expressing the spike of the XBB.1.5 variant stabilized in its pre-fusion conformation (NDV-HXP-S). We demonstrated that one or two intranasal immunizations with live NDV-HXP-S expressing the XBB.1.5 spike induces systemic and mucosal antibody responses in mice and protects them from a challenge with the XBB.1.5 variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Furthermore, one or two intranasal vaccinations with NDV-HXP-S XBB.1.5 protected hamsters from variant matched infection and reduced virus emission, thereby providing complete protection to naïve animals in a direct contact transmission study. The data shown in this study supports the notion that intranasal vaccination with variant-adapted NDV-HXP-S induces protective mucosal immunity and reduces transmission rates, highlighting the robust protective efficacy of a single mucosal vaccination in mice and hamsters.
Collapse
Affiliation(s)
- Stefan Slamanig
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Nicholas Lemus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tsoi Ying Lai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mitali Mishra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam Abdeljawad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta Boza
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria Dolange
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health 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 Institute for Precision Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), 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; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Peter Palese
- Department of Microbiology, 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.
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
20
|
Lin Y, Liao X, Cao X, Zhang Z, Wang X, He X, Liao H, Ju B, Qi F, Xu H, Ren Z, Wang Y, Hu Z, Yang J, Fu YX, Zhao J, Zhang Z, Peng H. Sequential intranasal booster triggers class switching from intramuscularly primed IgG to mucosal IgA against SARS-CoV-2. J Clin Invest 2025; 135:e175233. [PMID: 39808503 PMCID: PMC11870729 DOI: 10.1172/jci175233] [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: 08/29/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
The persistent emergence of COVID-19 variants and recurrent waves of infection worldwide underscores the urgent need for vaccines that effectively reduce viral transmission and prevent infections. Current intramuscular (IM) COVID-19 vaccines inadequately protect the upper respiratory mucosa. In response, we have developed a nonadjuvanted, IFN-armed SARS-CoV-2 fusion protein vaccine with IM priming and intranasal (IN) boost sequential immunization. Our study showed that this sequential vaccination strategy of the IM+IN significantly enhanced both upper respiratory and systemic antiviral immunity in a mouse model, characterized by the rapid increase in systemic and mucosal T and B cell responses, particularly the mucosal IgA antibody response. The IN boost triggered a swift secondary immune response, rapidly inducing antigen-specific IgA+ B cells. Further B cell receptor-seq (BCR-seq) analysis indicated that these IgA+ B cells primarily arose through direct class switching from preexisting IgG+ B cells in draining lymph nodes. Notably, our clinical studies revealed that the IN boost after IM vaccination elicited a robust systemic IgA antibody response in humans, as measured in serum. Thus, we believe that our cytokine-armed protein vaccine presents a promising strategy for inducing rapid and potent mucosal protection against respiratory viral infections.
Collapse
Affiliation(s)
- Yifan Lin
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xuezhi Cao
- Guangzhou Laboratory, Guangzhou International Bio-Island, 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, Guangdong, China
| | - Xiuye Wang
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Xiaomeng He
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | | | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hairong Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | - Yanqun Wang
- 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, Guangdong, China
| | | | | | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Jincun Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- 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, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hua Peng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- 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, Guangdong, China
| |
Collapse
|
21
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
22
|
Aljehani ND, Tamming L, Khan MY, Abdulal RH, Alfaleh MA, Ghazwani A, Helal A, Alsulaiman RM, Sanki MA, Alluhaybi K, Sukareh FA, Alharbi RH, Alyami FH, ElAssouli MZ, Shebbo S, Abdulaal WH, Algaissi A, Mahmoud AB, Basabrain M, Duque D, Bavananthasivam J, Chen W, Wang L, Sauve S, Abujamel TS, Altorki T, Alhabbab R, Tran A, Li X, Hashem AM. Mucosal SARS-CoV-2 S1 adenovirus-based vaccine elicits robust systemic and mucosal immunity and protects against disease in animals. mBio 2025; 16:e0217024. [PMID: 39629990 PMCID: PMC11708039 DOI: 10.1128/mbio.02170-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/04/2024] [Indexed: 01/11/2025] Open
Abstract
The COVID-19 pandemic has emphasized the importance and need for accessible safe, effective, and versatile vaccine platforms. While approved SARS-CoV-2 vaccines have been instrumental in saving lives and reducing healthcare and economic burdens, the induction of mucosal immunity remains an unmet need. Here, we engineered and evaluated a non-replicating adenovirus 5 (rAd5)-based vaccine expressing the SARS-CoV-2 S1 subunit (rAd5-SARS2-S1). We assessed the immunogenicity, durability, and protective efficacy of intramuscular (IM) and intranasal (IN) administration of rAd5-SARS2-S1 in mice and Syrian hamsters. Two IM or IN doses of rAd5-SARS2-S1 elicited robust and sustained Th1-skewed S1-specific serum IgG, neutralizing antibodies (nAbs) against several SARS-CoV-2 variants and systemic antigen-specific memory T cell responses in mice. Additionally, IN vaccination induced potent and long-lasting mucosal S1-specific IgG, IgA, and nAbs and pulmonary memory T cells. Importantly, while IM vaccine significantly ameliorated disease severity in hamsters by reducing viral burden, lung pathology, and, to some extent, weight loss, IN immunization significantly reduced viral replication and provided superior protection against disease and weight loss. Together, our study demonstrates that the rAd5-SARS2-S1 vaccine is immunogenic in both mice and hamsters when administered intramuscularly or intranasally, with IN administration providing better protection. These findings suggest that IN delivery of rAd5-SARS2-S1 could be a promising approach for inducing mucosal and systemic immunity, offering enhanced protection against SARS-CoV-2 and emerging variants. IMPORTANCE This publication presents an assessment of the immune response and effectiveness of a vaccine containing genetically modified non-replicating recombinant that expresses the S1 subunit protein of SARS-CoV-2. We conducted a comparative analysis of the immune response potency, durability, and protective effectiveness of this vaccine using intramuscular (IM) and intranasal (IN) inoculation in mice and Syrian hamsters. Our findings indicate that both vaccinations were effective in stimulating strong and long-lasting immune responses, both locally and across the body, when administered through either IM or IN methods. Crucially, our study demonstrated that the IN vaccination outperformed the IM vaccine by effectively and significantly suppressing the multiplication of the virus in the lungs and nasal turbinates. Additionally, the IN vaccine provided protection against disease-related weight loss and lung damage in the animals. This work showcases the potential of intranasal administration as a viable method to stimulate both mucosal and systemic immunity. This technique provides improved defense against SARS-CoV-2 and maybe additional variations.
Collapse
Affiliation(s)
- Najwa D. Aljehani
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Levi Tamming
- Centre for Oncology, Radiopharmaceuticals and Research Biologics and Radiopharmaceutical Drug Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Muhammad Yasir Khan
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rwaa H. Abdulal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A. Alfaleh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aishah Ghazwani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Asalah Helal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Reem M. Alsulaiman
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad A. Sanki
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalid Alluhaybi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farah Ayman Sukareh
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rahaf H. Alharbi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Faris H. Alyami
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - M-Zaki ElAssouli
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salima Shebbo
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- College of Dental Medicine, QU Health, Qatar University, Doha, Qatar
| | - Wesam H. Abdulaal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Mohammad Basabrain
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Diana Duque
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Ontario, Canada
| | - Jegarubee Bavananthasivam
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Ontario, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Ontario, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon Sauve
- Centre for Oncology, Radiopharmaceuticals and Research Biologics and Radiopharmaceutical Drug Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Ontario, Canada
| | - Turki S. Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tarfa Altorki
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rowa Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anh Tran
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Ontario, Canada
| | - Xuguang Li
- Centre for Oncology, Radiopharmaceuticals and Research Biologics and Radiopharmaceutical Drug Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
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: 2] [Impact Index Per Article: 2.0] [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
|
24
|
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
|
25
|
Stephenson S, Eid W, Wong CH, Mercier E, D'Aoust PM, Kabir MP, Baral S, Gilbride KA, Oswald C, Straus SE, Mackenzie A, Delatolla R, Graber TE. Urban wastewater contains a functional human antibody repertoire of mucosal origin. WATER RESEARCH 2024; 267:122532. [PMID: 39369505 DOI: 10.1016/j.watres.2024.122532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024]
Abstract
Wastewater-based surveillance of human disease offers timely insights to public health, helping to mitigate infectious disease outbreaks and decrease downstream morbidity and mortality. These systems rely on nucleic acid amplification tests for monitoring disease trends, while antibody-based seroprevalence surveys gauge community immunity. However, serological surveys are resource-intensive and subject to potentially long lead times and sampling bias. We identified and characterized a human antibody repertoire, predominantly secretory IgA, isolated from a central wastewater treatment plant and building-scale wastewater collection points. These antibodies partition to the solids fraction and retain immunoaffinity for SARS-CoV-2 and Influenza A virus antigens. This stable pool could enable real-time tracking for correlates of vaccination, infection, and immunity, aiding in establishing population-level thresholds for immune protection and assessing the efficacy of future vaccine campaigns.
Collapse
Affiliation(s)
- Sean Stephenson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Walaa Eid
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Chandler Hayyin Wong
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Elisabeth Mercier
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Patrick M D'Aoust
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Md Pervez Kabir
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Stefan Baral
- Knowledge Translation Program, Unity Health Toronto, Toronto, Ontario, Canada
| | - Kimberly A Gilbride
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Claire Oswald
- Department of Geography and Environmental Studies, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Sharon E Straus
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alex Mackenzie
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Robert Delatolla
- Department of Civil Engineering, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada.
| |
Collapse
|
26
|
Xu JW, Wang BS, Gao P, Huang HT, Wang FY, Qiu W, Zhang YY, Xu Y, Gou JB, Yu LL, Liu X, Wang RJ, Zhu T, Hou LH, Wang Q. Safety and immunogenicity of heterologous boosting with orally administered aerosolized bivalent adenovirus type-5 vectored COVID-19 vaccine and B.1.1.529 variant adenovirus type-5 vectored COVID-19 vaccine in adults 18 years and older: a randomized, double blinded, parallel controlled trial. Emerg Microbes Infect 2024; 13:2281355. [PMID: 37933089 PMCID: PMC11025474 DOI: 10.1080/22221751.2023.2281355] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/04/2023] [Indexed: 11/08/2023]
Abstract
Vaccination strategies that can induce a broad spectrum immune response are important to enhance protection against SARS-CoV-2 variants. We conducted a randomized, double-blind and parallel controlled trial to evaluate the safety and immunogenicity of the bivalent (5×1010viral particles) and B.1.1.529 variant (5×1010viral particles) adenovirus type-5 (Ad5) vectored COVID-19 vaccines administrated via inhalation. 451 eligible subjects aged 18 years and older who had been vaccinated with three doses inactivated COVID-19 vaccines were randomly assigned to inhale one dose of either B.1.1.529 variant Ad5 vectored COVID-19 vaccine (Ad5-nCoVO-IH group, N=150), bivalent Ad5 vectored COVID-19 vaccine (Ad5-nCoV/O-IH group, N=151), or Ad5 vectored COVID-19 vaccine (5×1010viral particles; Ad5-nCoV-IH group, N=150). Adverse reactions reported by 37 (24.67%) participants in the Ad5-nCoVO-IH group, 28 (18.54%) in the Ad5-nCoV/O-IH group, and 26 (17.33%) in the Ad5-nCoV-IH group with mainly mild to moderate dry mouth, oropharyngeal pain, headache, myalgia, cough, fever and fatigue. No serious adverse events related to the vaccine were reported. Investigational vaccines were immunogenic, with significant difference in the GMTs of neutralizing antibodies against Omicron BA.1 between Ad5-nCoV/O-IH (43.70) and Ad5-nCoV-IH (29.25) at 28 days after vaccination (P=0.0238). The seroconversion rates of neutralizing antibodies against BA.1 in Ad5-nCoVO-IH, Ad5-nCoV/O-IH, and Ad5-nCoV-IH groups were 56.00%, 59.60% and 48.67% with no significant difference among the groups. Overall, the investigational vaccines were demonstrated to be safe and well tolerated in adults, and was highly effective in inducing mucosal immunities in addition to humoral and cellular immune responses defending against SARS-CoV-2 variants.Trial registration: Chictr.org identifier: ChiCTR2200063996.
Collapse
Affiliation(s)
- Jia-Wei Xu
- Expanded Program on Immunization, Chongqing Center for Disease Control and Prevention, Chongqing, People’s Republic of China
| | - Bu-Sen Wang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Ping Gao
- Logistics University of Chinese People’s Armed Police Force, Tianjin, People’s Republic of China
| | - Hai-Tao Huang
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Fei-Yu Wang
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Wei Qiu
- Expanded Program on Immunization, Chongqing Center for Disease Control and Prevention, Chongqing, People’s Republic of China
| | - Yuan-Yuan Zhang
- Expanded Program on Immunization, Chongqing Center for Disease Control and Prevention, Chongqing, People’s Republic of China
| | - Yu Xu
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Jin-Bo Gou
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Lin-Ling Yu
- Expanded Program on Immunization, Yubei District Center for Disease Control and Prevention, Chongqing, People’s Republic of China
| | - Xuan Liu
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Rui-Jie Wang
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Tao Zhu
- CanSino Biologics Inc., Tianjin, People’s Republic of China
| | - Li-Hua Hou
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Qing- Wang
- Expanded Program on Immunization, Chongqing Center for Disease Control and Prevention, Chongqing, People’s Republic of China
| |
Collapse
|
27
|
Gao P, Morita N, Shinkura R. Role of mucosal IgA antibodies as novel therapies to enhance mucosal barriers. Semin Immunopathol 2024; 47:1. [PMID: 39567378 PMCID: PMC11579142 DOI: 10.1007/s00281-024-01027-4] [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: 10/12/2023] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
To prevent infection, the experience of the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) pandemic has led to recognition of the importance of not only vaccines but also the strengthening of mucosal barriers by secretory immunoglobulin A (IgA). Strong mucosal barrier provided by IgA is also possible to prevent allergies and chronic inflammatory conditions in the intestinal tract, since it can protect foreign enemies or antigens at the first line of defense before their invasion. Therefore, it is important to understand the role of IgA antibodies secreted by the mucosa of the body. In this section, we discuss the role of mucosal IgA antibodies in relation to three disease states: control of intestinal microbiota, protection against infection, and allergy. In addition, we provide the evidence in which the quality as well as the quantity of IgA is critical for disease prevention. Therefore, we discuss about novel strategies to enhance mucosal barriers by induction of high-quality IgA.
Collapse
Affiliation(s)
- Peng Gao
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-0032, Japan
| | - Naoki Morita
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-0032, Japan
| | - Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-0032, Japan.
| |
Collapse
|
28
|
Augello M, Wagenhäuser I, Krone M, Dauby N, Ferrara P, Sabbatucci M, Ruta S, Rezahosseini O, Velikov P, Gkrania-Klotsas E, Montes J, Franco-Paredes C, Goodman AL, Küçükkaya S, Tuells J, Harboe ZB, Epaulard O. Should SARS-CoV-2 serological testing be used in the decision to deliver a COVID-19 vaccine booster? A pro-con assessment. Vaccine 2024; 42:126184. [PMID: 39097440 DOI: 10.1016/j.vaccine.2024.126184] [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: 02/15/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Anti-SARS-CoV-2 vaccination has saved millions of lives in the past few years. To maintain a high level of protection, particularly in at-risk populations, booster doses are recommended to counter the waning of circulating antibody levels over time and the continuous emergence of immune escape variants of concern (VOCs). As anti-spike serology is now widely available, it may be considered a useful tool to identify individuals needing an additional vaccine dose, i.e., to screen certain populations to identify those whose plasma antibody levels are too low to provide protection. However, no recommendations are currently available on this topic. We reviewed the relevant supporting and opposing arguments, including areas of uncertainty, and concluded that in most populations, spike serology should not be used to decide about the administration of a booster dose. The main counterarguments are as follows: correlates of protection are imperfectly characterised, essentially owing to the emergence of VOCs; spike serology has an intrinsic inability to comprehensively reflect the whole immune memory; and booster vaccines are now VOC-adapted, while the commonly available commercial serological assays explore antibodies against the original virus.
Collapse
Affiliation(s)
- Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Isabell Wagenhäuser
- University Hospital Würzburg, Infection Control and Antimicrobial Stewardship Unit, Würzburg, Germany
| | - Manuel Krone
- University Hospital Würzburg, Infection Control and Antimicrobial Stewardship Unit, Würzburg, Germany
| | - Nicolas Dauby
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Environmental health and occupational health, School of Public Health, Université Libre de Bruxelles (ULB), Brussel, Belgium
| | - Pietro Ferrara
- Center for Public Health Research, University of Milan - Bicocca, Monza, Italy; IRCCS Istituto Auxologico Italiano, Laboratory of Public Health, Milan, Italy
| | | | - Simona Ruta
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Omid Rezahosseini
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, Hillerød, Denmark
| | - Petar Velikov
- Clinic for Pediatric Infectious Diseases, Infectious Disease Hospital "Prof. Ivan Kirov", Sofia, Bulgaria; Department of Global Public Health, University of Tsukuba, Tsukuba, Japan
| | | | - Jose Montes
- Investigación en Resistencia Antibiótica (INVERA), Buenos Aires, Argentina; Fundación del Centro de Estudios Infectológicos (FUNCEI), Buenos Aires, Argentina
| | - Carlos Franco-Paredes
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, USA; Hospital Infantil de Mexico, Mexico City, Mexico
| | - Anna L Goodman
- Centre for Infection Diagnostics research, Department of Infection at at King's College London and Guys' and St Thomas NHS Foundation trust, London, UK
| | - Sertaç Küçükkaya
- Department of Medical Microbiology, Istanbul Faculty of Medicine, İstanbul University, Istanbul, Turkey
| | - Jose Tuells
- Departamento de Enfermería Comunitaria, Medicina Preventiva y Salud Pública e historia de la ciencia, Universidad de Alicante, Alicante, Spain
| | | | - Olivier Epaulard
- Université Grenoble Alpes, Infectiologie, CHU Grenoble Alpes, Grenoble, France.
| |
Collapse
|
29
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), 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.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
30
|
Picó Munyoz R, Tárrega A, Laguna L. Exploring the impact of bubble type on sensory stimulation in drinks. Physiol Behav 2024; 286:114656. [PMID: 39111644 DOI: 10.1016/j.physbeh.2024.114656] [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: 04/23/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/30/2024]
Abstract
This study explores the impact of various types of carbonation on sensory stimulation in the mouth, salivary secretion and the neurotransmitter substance P (SP), as well as body responses such as heart rate (HR) and Galvanic Skin Response (GSR). Three types of carbonation (one made using a soda machine, another carbonated with a gasifier, and the last commercial sparkling water) were used to produce different bubbles resulting in distinct sensory characteristics assessed by a trained panel. The impact of carbonation was measured by recording changes in salivary flow rate, SP levels, salivary secretory immunoglobulin A (SIgA), HR, and GSR in fifteen healthy participants. The results showed that the bubble type only affected the sensory perception of carbonation. Regardless of bubble type, carbonation increased salivary flow rate and SP values, SigA and HR. These characteristics are being sought to improve treatments for dysphagia or dry mouth. Therefore, these findings highlight the potential therapeutic application of carbonation in these situations.
Collapse
Affiliation(s)
- Ruth Picó Munyoz
- Consumer Perception and Behavior and Adapted Nutrition Laboratory, Institute of Agrochemistry and Food Technology (IATA, CSIC), Paterna, Spain
| | - Amparo Tárrega
- Consumer Perception and Behavior and Adapted Nutrition Laboratory, Institute of Agrochemistry and Food Technology (IATA, CSIC), Paterna, Spain
| | - Laura Laguna
- Consumer Perception and Behavior and Adapted Nutrition Laboratory, Institute of Agrochemistry and Food Technology (IATA, CSIC), Paterna, Spain.
| |
Collapse
|
31
|
Fuchs J, Hübner J, Schmidt A, Irrgang P, Maier C, Vieira Antão A, Oltmanns F, Thirion C, Lapuente D, Tenbusch M. Evaluation of adenoviral vector Ad19a encoding RSV-F as novel vaccine against respiratory syncytial virus. NPJ Vaccines 2024; 9:205. [PMID: 39472590 PMCID: PMC11522487 DOI: 10.1038/s41541-024-01001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in infants and toddlers. Since natural infections do not induce persistent immunity, there is the need of vaccines providing long-term protection. Here, we evaluated a new adenoviral vector (rAd) vaccine based on the rare serotype rAd19a and compared the immunogenicity and efficacy to the highly immunogenic rAd5. Given as an intranasal boost in DNA primed mice, both vectors encoding the F protein provided efficient protection against a subsequent RSV infection. However, intramuscular immunization with rAd19a vectors provoked vaccine-enhanced disease after RSV infection compared to non-vaccinated animals. While mucosal IgA antibodies and tissue-resident memory T-cells in intranasally vaccinated mice rapidly control RSV replication, a strong anamnestic systemic T-cell response in absence of local immunity might be the reason for immune-mediated enhanced disease. Our study highlighted the potential benefits of developing effective mucosal against respiratory pathogens.
Collapse
Affiliation(s)
- Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Julian Hübner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Anna Schmidt
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Ana Vieira Antão
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany.
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054, Erlangen, Germany.
| |
Collapse
|
32
|
Declercq J, Gerlo S, Van Nevel S, De Ruyck N, Holtappels G, Delesie L, Tobback E, Lammens I, Gerebtsov N, Sedeyn K, Saelens X, Lambrecht BN, Gevaert P, Vandekerckhove L, Vanhee S. Repeated COVID-19 mRNA-based vaccination contributes to SARS-CoV-2 neutralizing antibody responses in the mucosa. Sci Transl Med 2024; 16:eadn2364. [PMID: 39441904 DOI: 10.1126/scitranslmed.adn2364] [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: 12/19/2023] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
To prevent infection by respiratory viruses and consequently limit virus circulation, vaccines need to promote mucosal immunity. The extent to which the currently used messenger RNA (mRNA)-based COVID-19 vaccines induce mucosal immunity remains poorly characterized. We evaluated mucosal neutralizing antibody responses in a cohort of 183 individuals. Participants were sampled at several time points after primary adenovirus vector-based or mRNA-based COVID-19 vaccination and after mRNA-based booster vaccinations. Our findings revealed that repeated vaccination with mRNA boosters promoted severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies in nasal secretions. Nasal and serum neutralizing antibody titers of both IgG and IgA isotypes correlated to one another. We investigated the source of these mucosal antibodies in a mouse model wherein mice received repeated mRNA vaccines for SARS-CoV-2. These experiments indicated that neutralizing antibody-producing cells reside in the spleen and bone marrow, whereas no proof of tissue homing to the respiratory mucosa was observed, despite the detection of mucosal antibodies. Serum transfer experiments confirmed that circulating antibodies were able to migrate to the respiratory mucosa. Collectively, these results demonstrate that, especially upon repeated vaccination, the currently used COVID-19 mRNA vaccines can elicit mucosal neutralizing antibodies and that vaccination might also stimulate mucosal immunity induced by previous SARS-CoV-2 infection. Moreover, migration of circulating antibodies to the respiratory mucosa might be a main mechanism. These findings advance our understanding of mRNA vaccine-induced immunity and have implications for the design of vaccine strategies to combat respiratory infections.
Collapse
Affiliation(s)
- Jozefien Declercq
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sarah Gerlo
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Sharon Van Nevel
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Natalie De Ruyck
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Gabriele Holtappels
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Liesbeth Delesie
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Els Tobback
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Inés Lammens
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Nikita Gerebtsov
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Koen Sedeyn
- VIB Center for Medical Biotechnology, 9000 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Xavier Saelens
- VIB Center for Medical Biotechnology, 9000 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus Medical Center, 3015 GD Rotterdam, Netherlands
| | - Philippe Gevaert
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Stijn Vanhee
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
33
|
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
|
34
|
Holder KA, Ings DP, Fifield KE, Barnes DA, Barnable KA, Harnum DOA, Russell RS, Grant MD. Sequence Matters: Primary COVID-19 Vaccination after Infection Elicits Similar Anti-spike Antibody Levels, but Stronger Antibody Dependent Cell-mediated Cytotoxicity than Breakthrough Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1105-1114. [PMID: 39248629 PMCID: PMC11457723 DOI: 10.4049/jimmunol.2400250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Infection before primary vaccination (herein termed "hybrid immunity") engenders robust humoral immunity and broad Ab-dependent cell-mediated cytotoxicity (ADCC) across SARS-CoV-2 variants. We measured and compared plasma IgG and IgA against Wuhan-Hu-1 and Omicron (B.1.1.529) full-length spike (FLS) and receptor binding domain after three mRNA vaccines encoding Wuhan-Hu-1 spike (S) and after Omicron breakthrough infection. We also measured IgG binding to Wuhan-Hu-1 and Omicron S1, Wuhan-Hu-1 S2 and Wuhan-Hu-1 and Omicron cell-based S. We compared ADCC using human embryonic lung fibroblast (MRC-5) cells expressing Wuhan-Hu-1 or Omicron S. The effect of Omicron breakthrough infection on IgG anti-Wuhan-Hu-1 and Omicron FLS avidity was also considered. Despite Omicron breakthrough infection increasing IgG and IgA against FLS and receptor binding domain to levels similar to those seen with hybrid immunity, there was no boost to ADCC. Preferential recognition of Wuhan-Hu-1 persisted following Omicron breakthrough infection, which increased IgG avidity against Wuhan-Hu-1 FLS. Despite similar total anti-FLS IgG levels following breakthrough infection, 4-fold higher plasma concentrations were required to elicit ADCC comparable to that elicited by hybrid immunity. The greater capacity for hybrid immunity to elicit ADCC was associated with a differential IgG reactivity pattern against S1, S2, and linear determinants throughout FLS. Immunity against SARS-CoV-2 following Omicron breakthrough infection manifests significantly less ADCC capacity than hybrid immunity. Thus, the sequence of antigenic exposure by infection versus vaccination and other factors such as severity of infection affect antiviral functions of humoral immunity in the absence of overt quantitative differences in the humoral response.
Collapse
Affiliation(s)
- Kayla A. Holder
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Danielle P. Ings
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Kathleen E. Fifield
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - David A. Barnes
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Keeley A. Barnable
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | | | - Rodney S. Russell
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Michael D. Grant
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| |
Collapse
|
35
|
Vieira J, de Oliveira TVV, Queiroz LRR, Camargo CTS, Nardy A, Monteiro FR, do Amaral JB, Paixão V, Vaisberg M, Amirato GR, Dos Santos CAF, Durigon EL, Oliveira DBL, Aguiar AS, Alvares-Saraiva AM, Heller D, Mantoanelli PGV, Siqueira MF, da Silva Nali LH, Bachi ALL. Salivary assessment of the immune/inflammatory responses and oxidative stress in older adults vaccinated with CoronaVac or ChadOx-1. BMC Geriatr 2024; 24:807. [PMID: 39363197 PMCID: PMC11448442 DOI: 10.1186/s12877-024-05357-8] [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: 10/02/2023] [Accepted: 09/04/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Although important information concerning COVID-19 vaccination is available, the effects of the CoronaVac and ChadOx-1 vaccines on immunity and the redox balance in the upper airway mucosa of the aged population are not fully understood. Therefore, the aim of this study was to investigate the impacts of two doses of the CoronaVac or ChadOx-1 vaccine on immune/inflammatory responses and oxidative stress in the airway mucosa of older adults. METHODS Seventy-six older adults of both sexes, with a mean age of 75.1 ± 6.4 years, were separated according to vaccination status into the CoronaVac (n = 52) and ChadOx-1 (n = 24) groups. Saliva samples were collected before (pre) and 30 days after (post) the administration of the second dose of the CoronaVac or ChadOx-1 vaccine to assess the levels of antibodies (sIgA and IgG), antimicrobial peptides, cytokines, and oxidant/antioxidant agents. RESULTS The immunogenicity in the ChadOx-1 group was 37.5% for sIgA and 25% for IgG, while that in the CoronaVac group was 18.9% for sIgA and 13.2% for IgG. Intergroup analysis revealed that (1) lower levels of IFN-α, IFN-γ, and IL-10 and a greater IFN-γ/IL-10 ratio, in addition to a greater IL-6/IL-10 ratio, were found in both the pre- and postvaccination periods, and (2) lower levels of total sIgA, IL-12p70, IL-17A, TNF-α, and the IL-12p70/IL-10 ratio, in addition to higher levels of specific sIgA for SARS-CoV-2 antigens and lysozyme, were observed only in the postvaccination period in the ChadOx-1 group than in the CoronaVac group. Intragroup analysis revealed (1) a significant increase in the salivary levels of total peroxides in the postvaccination period compared to those in the prevaccination period in both volunteer groups; (2) a decrease in the levels of lysozyme and the ratio between total antioxidant capacity (TAC) and total peroxides in the postvaccination period in the CoronaVac group compared with those in the prevaccination period; and (3) decreases in the TNF-α, IL-6, and IL-12p70 levels, and the IL-12p70/IL-10 ratio in the ChadoX-1 group, as well as a higher lactoferrin concentration in the postvaccination period than in the prevaccination period. Several positive and negative correlations between the parameters assessed here were found. CONCLUSIONS In general, the ChadOx-1 group exhibited improvements in both immune/inflammatory responses and redox balance and greater immunogenicity than did the CoronaVac group.
Collapse
Affiliation(s)
- Jeniffer Vieira
- Faculty of Dentistry, Campus 1, Santo Amaro University (UNISA), São Paulo, Brazil
| | | | | | | | - Ariane Nardy
- Postgraduate Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, Brazil
| | | | - Jônatas Bussador do Amaral
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Vitória Paixão
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Mauro Vaisberg
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Gislene Rocha Amirato
- Mane Garrincha Sport Education Center, Sports Department of the Municipality of São Paulo (SEME), São Paulo, Brazil
| | - Carlos André Freitas Dos Santos
- Discipline of Geriatrics and Gerontology, Department of Medicine, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- Postgraduate Program in Translational Medicine, Department of Medicine, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Edison Luiz Durigon
- Institute of Biomedical Science of University of São Paulo (USP), São Paulo, Brazil
- Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Danielle Bruna Leal Oliveira
- Institute of Biomedical Science of University of São Paulo (USP), São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Andressa Simões Aguiar
- Institute of Biomedical Science of University of São Paulo (USP), São Paulo, Brazil
- Infection Control Service, São Luiz Gonzaga Hospital of Santa Casa de Misericordia of São Paulo, São Paulo, Brazil
| | | | - Débora Heller
- Postgraduate Program in Dentistry, Cruzeiro do Sul University, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
36
|
Murphy QM, Lewis GK, Sajadi MM, Forde JE, Ciupe SM. Understanding antibody magnitude and durability following vaccination against SARS-CoV-2. Math Biosci 2024; 376:109274. [PMID: 39218212 DOI: 10.1016/j.mbs.2024.109274] [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: 05/11/2024] [Revised: 07/14/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) results in transient antibody response against the spike protein. The individual immune status at the time of vaccination influences the response. Using mathematical models of antibody decay, we determined the dynamics of serum immunoglobulin G (IgG) and serum immunoglobulin A (IgA) over time. Data fitting to longitudinal IgG and IgA titers was used to quantify differences in antibody magnitude and antibody duration among infection-naïve and infection-positive vaccinees. We found that prior infections result in more durable serum IgG and serum IgA responses, with prior symptomatic infections resulting in the most durable serum IgG response and prior asymptomatic infections resulting in the most durable serum IgA response. These findings can guide vaccine boosting schedules.
Collapse
Affiliation(s)
- Quiyana M Murphy
- Department of Mathematics, Virginia Polytechnic Institute and State University, 225 Stanger Street, Blacksburg, 24060, VA, USA; Virginia Tech Center for the Mathematics of Biosystems, Virginia Tech, Blacksburg, VA, USA
| | - George K Lewis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mohammad M Sajadi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jonathan E Forde
- Department of Mathematics and Computer Sciences, Hobart and William Smith Colleges, Geneva, NY, USA
| | - Stanca M Ciupe
- Department of Mathematics, Virginia Polytechnic Institute and State University, 225 Stanger Street, Blacksburg, 24060, VA, USA; Virginia Tech Center for the Mathematics of Biosystems, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
37
|
Acúrcio RC, Kleiner R, Vaskovich‐Koubi D, Carreira B, Liubomirski Y, Palma C, Yeheskel A, Yeini E, Viana AS, Ferreira V, Araújo C, Mor M, Freund NT, Bacharach E, Gonçalves J, Toister‐Achituv M, Fabregue M, Matthieu S, Guerry C, Zarubica A, Aviel‐Ronen S, Florindo HF, Satchi‐Fainaro R. Intranasal Multiepitope PD-L1-siRNA-Based Nanovaccine: The Next-Gen COVID-19 Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404159. [PMID: 39116324 PMCID: PMC11515909 DOI: 10.1002/advs.202404159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/28/2024] [Indexed: 08/10/2024]
Abstract
The first approved vaccines for human use against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are nanotechnology-based. Although they are modular, rapidly produced, and can reduce disease severity, the currently available vaccines are restricted in preventing infection, stressing the global demand for novel preventive vaccine technologies. Bearing this in mind, we set out to develop a flexible nanovaccine platform for nasal administration to induce mucosal immunity, which is fundamental for optimal protection against respiratory virus infection. The next-generation multiepitope nanovaccines co-deliver immunogenic peptides, selected by an immunoinformatic workflow, along with adjuvants and regulators of the PD-L1 expression. As a case study, we focused on SARS-CoV-2 peptides as relevant antigens to validate the approach. This platform can evoke both local and systemic cellular- and humoral-specific responses against SARS-CoV-2. This led to the secretion of immunoglobulin A (IgA), capable of neutralizing SARS-CoV-2, including variants of concern, following a heterologous immunization strategy. Considering the limitations of the required cold chain distribution for current nanotechnology-based vaccines, it is shown that the lyophilized nanovaccine is stable for long-term at room temperature and retains its in vivo efficacy upon reconstitution. This makes it particularly relevant for developing countries and offers a modular system adaptable to future viral threats.
Collapse
Affiliation(s)
- Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Ron Kleiner
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Bárbara Carreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Yulia Liubomirski
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Carolina Palma
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Adva Yeheskel
- The Blavatnik Center for Drug DiscoveryTel Aviv UniversityTel Aviv6997801Israel
| | - Eilam Yeini
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Ana S. Viana
- Center of Chemistry and BiochemistryFaculty of SciencesUniversity of LisbonLisbon1749‐016Portugal
| | - Vera Ferreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Carlos Araújo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Michael Mor
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Natalia T. Freund
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
| | - Eran Bacharach
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv6997801Israel
| | - João Gonçalves
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | | | - Manon Fabregue
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Solene Matthieu
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Capucine Guerry
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | - Ana Zarubica
- Centre d'ImmunophénomiqueAix Marseille UniversitéInserm, CNRS, PHENOMINMarseille13284France
| | | | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineTel Aviv UniversityTel Aviv6997801Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv6997801Israel
| |
Collapse
|
38
|
O’Reilly S, Byrne J, Feeney ER, Mallon PWG, Gautier V. Navigating the Landscape of B Cell Mediated Immunity and Antibody Monitoring in SARS-CoV-2 Vaccine Efficacy: Tools, Strategies and Clinical Trial Insights. Vaccines (Basel) 2024; 12:1089. [PMID: 39460256 PMCID: PMC11511438 DOI: 10.3390/vaccines12101089] [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: 07/31/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
Correlates of Protection (CoP) are biomarkers above a defined threshold that can replace clinical outcomes as primary endpoints, predicting vaccine effectiveness to support the approval of new vaccines or follow up studies. In the context of COVID-19 vaccination, CoPs can help address challenges such as demonstrating vaccine effectiveness in special populations, against emerging SARS-CoV-2 variants or determining the durability of vaccine-elicited immunity. While anti-spike IgG titres and viral neutralising capacity have been characterised as CoPs for COVID-19 vaccination, the contribution of other components of the humoral immune response to immediate and long-term protective immunity is less well characterised. This review examines the evidence supporting the use of CoPs in COVID-19 clinical vaccine trials, and how they can be used to define a protective threshold of immunity. It also highlights alternative humoral immune biomarkers, including Fc effector function, mucosal immunity, and the generation of long-lived plasma and memory B cells and discuss how these can be applied to clinical studies and the tools available to study them.
Collapse
Affiliation(s)
- Sophie O’Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Joanne Byrne
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
39
|
Clark RD, Rabito F, Munyonho FT, Remcho TP, Kolls JK. Evaluation of anti-vector immune responses to adenovirus-mediated lung gene therapy and modulation by αCD20. Mol Ther Methods Clin Dev 2024; 32:101286. [PMID: 39070292 PMCID: PMC11283059 DOI: 10.1016/j.omtm.2024.101286] [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: 08/16/2023] [Accepted: 06/21/2024] [Indexed: 07/30/2024]
Abstract
Although the last decade has seen tremendous progress in drugs that treat cystic fibrosis (CF) due to mutations that lead to protein misfolding, there are approximately 8%-10% of subjects with mutations that result in no significant CFTR protein expression demonstrating the need for gene editing or gene replacement with inhaled mRNA or vector-based approaches. A limitation for vector-based approaches is the formation of neutralizing humoral responses. Given that αCD20 has been used to manage post-transplant lymphoproliferative disease in CF subjects with lung transplants, we studied the ability of αCD20 to module both T and B cell responses in the lung to one of the most immunogenic vectors, E1-deleted adenovirus serotype 5. We found that αCD20 significantly blocked luminal antibody responses and efficiently permitted re-dosing. αCD20 had more limited impact on the T cell compartment, but reduced tissue resident memory T cell responses in bronchoalveolar lavage fluid. Taken together, these pre-clinical studies suggest that αCD20 could be re-purposed for lung gene therapy protocols to permit re-dosing.
Collapse
Affiliation(s)
- Robert D.E. Clark
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Felix Rabito
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ferris T. Munyonho
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - T. Parks Remcho
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay K. Kolls
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
40
|
Lee YR, Liou CW, Liu IH, Chang JM. A nonadjuvanted HLA-restricted peptide vaccine induced both T and B cell immunity against SARS-CoV-2 spike protein. Sci Rep 2024; 14:20579. [PMID: 39242614 PMCID: PMC11379847 DOI: 10.1038/s41598-024-71663-1] [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: 05/13/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
During COVID-19 pandemic, cases of postvaccination infections and restored SARS-CoV-2 virus have increased after full vaccination, which might be contributed to by immune surveillance escape or virus rebound. Here, artificial linear 9-mer human leucocyte antigen (HLA)-restricted UC peptides were designed based on the well-conserved S2 region of the SARS-CoV-2 spike protein regardless of rapid mutation and glycosylation hindrance. The UC peptides were characterized for its effect on immune molecules and cells by HLA-tetramer refolding assay for HLA-binding ability, by HLA-tetramer specific T cell assay for engaged cytotoxic T lymphocytes (CTLs) involvement, by HLA-dextramer T cell assay for B cell activation, by intracellular cytokine release assay for polarization of immune response, Th1 or Th2. The specific lysis activity assay of T cells was performed for direct activation of cytotoxic T lymphocytes by UC peptides. Mice were immunized for immunogenicity of UC peptides in vivo and immunized sera was assay for complement cytotoxicity assay. Results appeared that through the engagement of UC peptides and immune molecules, HLA-I and II, that CTLs elicited cytotoxic activity by recognizing SARS-CoV-2 spike-bearing cells and preferably secreting Th1 cytokines. The UC peptides also showed immunogenicity and generated a specific antibody in mice by both intramuscular injection and oral delivery without adjuvant formulation. In conclusion, a T-cell vaccine could provide long-lasting protection against SARS-CoV-2 either during reinfection or during SARS-CoV-2 rebound. Due to its ability to eradicate SARS-CoV-2 virus-infected cells, a COVID-19 T-cell vaccine might provide a solution to lower COVID-19 severity and long COVID-19.
Collapse
Affiliation(s)
- Yi-Ru Lee
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC
| | - Chiung-Wen Liou
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC
| | - I-Hua Liu
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC
| | - Jia-Ming Chang
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC.
| |
Collapse
|
41
|
Nguyen K, Relja B, Epperson M, Park SH, Thornburg NJ, Costantini VP, Vinjé J. Salivary immune responses after COVID-19 vaccination. PLoS One 2024; 19:e0307936. [PMID: 39226256 PMCID: PMC11371244 DOI: 10.1371/journal.pone.0307936] [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: 12/08/2023] [Accepted: 07/16/2024] [Indexed: 09/05/2024] Open
Abstract
mRNA-based COVID-19 vaccines have played a critical role in reducing severe outcomes of COVID-19. Humoral immune responses against SARS-CoV-2 after vaccination have been extensively studied in blood; however, limited information is available on the presence and duration of SARS-CoV-2 specific antibodies in saliva and other mucosal fluids. Saliva offers a non-invasive sampling method that may also provide a better understanding of mucosal immunity at sites where the virus enters the body. Our objective was to evaluate the salivary immune response after vaccination with the COVID-19 Moderna mRNA-1273 vaccine. Two hundred three staff members of the U.S. Centers for Disease Control and Prevention were enrolled prior to receiving their first dose of the mRNA-1273 vaccine. Participants were asked to self-collect 6 saliva specimens at days 0 (prior to first dose), 14, 28 (prior to second dose), 42, and 56 using a SalivaBio saliva collection device. Saliva specimens were tested for anti-spike protein SARS-CoV-2 specific IgA and IgG enzyme immunoassays. Overall, SARS-CoV-2-specific salivary IgA titers peaked 2 weeks after each vaccine dose, followed by a sharp decrease during the following weeks. In contrast to IgA titers, IgG antibody titers increased substantially 2 weeks after the first vaccine dose, peaked 2 weeks after the second dose and persisted at an elevated level until at least 8 weeks after the first vaccine dose. Additionally, no significant differences in IgA/IgG titers were observed based on age, sex, or race/ethnicity. All participants mounted salivary IgA and IgG immune responses against SARS-CoV-2 after receiving the mRNA-1273 COVID-19 vaccine. Because of the limited follow-up time for this study, more data are needed to assess the antibody levels beyond 2 months after the first dose. Our results confirm the potential utility of saliva in assessing immune responses elicited by immunization and possibly by infection.
Collapse
Affiliation(s)
- Kenny Nguyen
- National Foundation for the Centers for Disease Control and Prevention Inc., Atlanta, GA, United States of America
| | - Boris Relja
- National Foundation for the Centers for Disease Control and Prevention Inc., Atlanta, GA, United States of America
- Cherokee Nation Assurance, Arlington, VA, United States of America
| | - Monica Epperson
- Laboratory Branch, Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - So Hee Park
- Eagle Global Scientific, LLC, Atlanta, GA, United States of America
| | - Natalie J. Thornburg
- Laboratory Branch, Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Veronica P. Costantini
- Division of Viral Diseases, Viral Gastroenteritis Branch, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Jan Vinjé
- Division of Viral Diseases, Viral Gastroenteritis Branch, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| |
Collapse
|
42
|
Goh YS, Fong SW, Hor PX, Loh CY, Tay MZ, Wang B, Salleh SNM, Ngoh EZX, Lee RTC, Poh XY, Lee IR, Rao S, Chia PY, Maurer-Stroh S, Wang CI, Leo YS, Lye DC, Young BE, Ng LFP, Renia L. Imprinting of IgA responses in previously infected individuals receiving bivalent mRNA vaccines (WT and BA.4/BA.5 or WT and BA.1). Int J Infect Dis 2024; 146:107147. [PMID: 38945433 DOI: 10.1016/j.ijid.2024.107147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
OBJECTIVES The emergence of new SARS-CoV-2 variants has led to the development of Omicron-targeting bivalent mRNA vaccines. It is crucial to understand how bivalent vaccines may improve antibody responses against new variants. METHODS A total of 107 participants, who had three COVID-19 WT mRNA vaccine doses, were recruited, and given either a monovalent (WT) or a bivalent mRNA vaccination (Pfizer/BioNTech Bivalent (WT and BA.4/BA.5) or Moderna Bivalent (WT and BA.1). Blood samples were taken before booster and at 28 days post-booster. RESULTS We found significantly lower fold change in serum binding IgA responses against BA.1, BA.5 and EG.5.1 spike in the bivalent booster group, compared with the monovalent (WT) booster group, following vaccination. However, this was only observed in individuals with prior infection. The relative fold change in serum binding IgA response was more skewed towards WT over variant (BA.1, BA.5 or EG.5.1) spike in previously infected bivalent-booster-vaccinees, as compared with previously infected monovalent-(WT)-booster-vaccinees. CONCLUSION The findings suggest imprinting of antibody responses that is shaped by the first vaccination (WT spike). Previous infection also affects the boosting effect of follow-up vaccination. Studies are needed to understand how to induce a robust and long-lasting IgA immunity for protection against COVID-19 infection.
Collapse
Affiliation(s)
- Yun Shan Goh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siew-Wai Fong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Xiang Hor
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Matthew Zirui Tay
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bei Wang
- Singapore Immunology Network, A*STAR, Singapore, Singapore
| | | | | | - Raphael Tze Chuen Lee
- Bioinformatics Institute, A*STAR, Singapore, Singapore; GISAID Global Data Science Initiative (GISAID), Munich, Germany
| | - Xuan Ying Poh
- National Centre for Infectious Diseases, Singapore, Singapore
| | - I Russel Lee
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Suma Rao
- National Centre for Infectious Diseases, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sebastian Maurer-Stroh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Bioinformatics Institute, A*STAR, Singapore, Singapore; GISAID Global Data Science Initiative (GISAID), Munich, Germany; National Public Health Laboratory, Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, A*STAR, Singapore, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
43
|
Panickar A, Manoharan A, Anbarasu A, Ramaiah S. Respiratory tract infections: an update on the complexity of bacterial diversity, therapeutic interventions and breakthroughs. Arch Microbiol 2024; 206:382. [PMID: 39153075 DOI: 10.1007/s00203-024-04107-z] [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: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Respiratory tract infections (RTIs) have a significant impact on global health, especially among children and the elderly. The key bacterial pathogens Streptococcus pneumoniae, Haemophilus influenzae, Klebsiella pneumoniae, Staphylococcus aureus and non-fermenting Gram Negative bacteria such as Acinetobacter baumannii and Pseudomonas aeruginosa are most commonly associated with RTIs. These bacterial pathogens have evolved a diverse array of resistance mechanisms through horizontal gene transfer, often mediated by mobile genetic elements and environmental acquisition. Treatment failures are primarily due to antimicrobial resistance and inadequate bacterial engagement, which necessitates the development of alternative treatment strategies. To overcome this, our review mainly focuses on different virulence mechanisms and their resulting pathogenicity, highlighting different therapeutic interventions to combat resistance. To prevent the antimicrobial resistance crisis, we also focused on leveraging the application of artificial intelligence and machine learning to manage RTIs. Integrative approaches combining mechanistic insights are crucial for addressing the global challenge of antimicrobial resistance in respiratory infections.
Collapse
Affiliation(s)
- Avani Panickar
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Anand Manoharan
- Infectious Diseases Medical and Scientific Affairs, GlaxoSmithKline (GSK), Worli, Maharashtra, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
44
|
Kim DH, Lee J, Lee DY, Lee SH, Jeong JH, Kim JY, Kim J, Choi YK, Lee JB, Park SY, Choi IS, Lee SW, Youk S, Song CS. Intranasal Administration of Recombinant Newcastle Disease Virus Expressing SARS-CoV-2 Spike Protein Protects hACE2 TG Mice against Lethal SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:921. [PMID: 39204044 PMCID: PMC11359043 DOI: 10.3390/vaccines12080921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged as a global outbreak in 2019, profoundly affecting both human health and the global economy. Various vaccine modalities were developed and commercialized to overcome this challenge, including inactivated vaccines, mRNA vaccines, adenovirus vector-based vaccines, and subunit vaccines. While intramuscular vaccines induce high IgG levels, they often fail to stimulate significant mucosal immunity in the respiratory system. We employed the Newcastle disease virus (NDV) vector expressing the spike protein of the SARS-CoV-2 Beta variant (rK148/beta-S), and evaluated the efficacy of intranasal vaccination with rK148/beta-S in K18-hACE2 transgenic mice. Intranasal vaccination with a low dose (106.0 EID50) resulted in an 86% survival rate after challenge with the SARS-CoV-2 Beta variant. Administration at a high dose (107.0 EID50) led to a reduction in lung viral load and 100% survival against the SARS-CoV-2 Beta and Delta variants. A high level of the SARS-CoV-2 spike-specific IgA was also induced in vaccinated mice lungs following the SARS-CoV-2 challenge. Our findings suggest that rK148/beta-S holds promise as an intranasal vaccine candidate that effectively induces mucosal immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Deok-Hwan Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA
| | - Da-Ye Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seung-Hun Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jei-Hyun Jeong
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ji-Yun Kim
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiwon Kim
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
| | - Joong-Bok Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Seung-Young Park
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - In-Soo Choi
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Sang-Won Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Sungsu Youk
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 28160, Republic of Korea
- Biomedical Research Institute, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
45
|
Goh YS, Fong SW, Hor PX, Loh CY, Wang B, Salleh SNM, Ngoh EZX, Lee RTC, Poh XY, Rao S, Chia PY, Ong SWX, Lee TH, Lim C, Teo J, Pada S, Sun LJ, Ong DLS, Somani J, Lee ES, Maurer-Stroh S, Wang CI, Leo YS, Lye DC, Young BE, Ng LFP, Renia L. Variant-Specific IgA Protects Against Omicron Infection. J Infect Dis 2024; 230:e287-e291. [PMID: 37996071 PMCID: PMC11326848 DOI: 10.1093/infdis/jiad525] [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: 09/01/2023] [Revised: 10/25/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The emergence of rapidly evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, coupled with waning vaccine-induced immunity, has contributed to the rise of vaccine breakthrough infections. It is crucial to understand how vaccine-induced protection is mediated. METHODS We examined 2 prospective cohorts of mRNA vaccinated and boosted individuals during the Omicron wave of infection in Singapore. RESULTS We found that individuals who remain uninfected over the follow-up period had a higher variant-specific IgA, but not IgG, antibody response at 1 month after booster vaccination, compared with individuals who became infected. CONCLUSIONS We conclude that IgA may have a potential contributory role in protection against Omicron infection. Clinical Trials Registration . NCT05142319.
Collapse
Affiliation(s)
- Yun Shan Goh
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Siew-Wai Fong
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Pei Xiang Hor
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Eve Zi Xian Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Raphael Tze Chuen Lee
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
- GISAID Global Data Science Initiative, Munich, Germany
| | - Xuan Ying Poh
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Suma Rao
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sean W X Ong
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tau Hong Lee
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Clarissa Lim
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Jefanie Teo
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Surinder Pada
- Division of Infectious Diseases, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Louisa Jin Sun
- Infectious Diseases, Alexandra Hospital, Singapore, Singapore
| | | | - Jyoti Somani
- Division of Infectious Diseases, Department of Medicine, National University Hospital, National University Health System, Singapore, Singapore
| | - Eng Sing Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Healthcare Group Polyclinics, Singapore, Singapore
| | - Sebastian Maurer-Stroh
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
- GISAID Global Data Science Initiative, Munich, Germany
- National Public Health Laboratory, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- Health Protection Research Unit in Emerging and Zoonotic Infections, National Institute of Health Research, University of Liverpool, Liverpool, United Kingdom
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Laurent Renia
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
46
|
Tsoi HW, Ng MKW, Cai JP, Poon RWS, Chan BPC, Chan KH, Tam AR, Chu WM, Hung IFN, To KKW. The impact of vaccine type and booster dose on the magnitude and breadth of SARS-CoV-2-specific systemic and mucosal antibodies among COVID-19 vaccine recipients. Heliyon 2024; 10:e35334. [PMID: 39166006 PMCID: PMC11334685 DOI: 10.1016/j.heliyon.2024.e35334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024] Open
Abstract
The COVID-19 pandemic has had a major impact on global health and economy, which was significantly mitigated by the availability of COVID-19 vaccines. The levels of systemic and mucosal antibodies against SARS-CoV-2 correlated with protection. However, there is limited data on how vaccine type and booster doses affect mucosal antibody response, and how the breadth of mucosal and systemic antibodies compares. In this cross-sectional study, we compared the magnitude and breadth of mucosal and systemic antibodies in 108 individuals who received either the BNT162b2 (Pfizer) or CoronaVac (SinoVac) vaccine. We found that BNT162b2 (vs CoronaVac) or booster doses (vs two doses) were significantly associated with higher serum IgG levels, but were not significantly associated with salivary IgA levels, regardless of prior infection status. Among non-infected individuals, serum IgG, serum IgA and salivary IgG levels were significantly higher against the ancestral strain than the Omicron BA.2 sublineage, but salivary IgA levels did not differ between the strains. Salivary IgA had the weakest correlation with serum IgG (r = 0.34) compared with salivary IgG (r = 0.63) and serum IgA (r = 0.60). Our findings suggest that intramuscular COVID-19 vaccines elicit a distinct mucosal IgA response that differs from the systemic IgG response. As mucosal IgA independently correlates with protection, vaccine trials should include mucosal IgA as an outcome measure.
Collapse
Affiliation(s)
- Hoi-Wah Tsoi
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Miko Ka-Wai Ng
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Rosana Wing-Shan Poon
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Brian Pui-Chun Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anthony Raymond Tam
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wing-Ming Chu
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| |
Collapse
|
47
|
Chupp DP, Rivera CE, Zhou Y, Xu Y, Ramsey PS, Xu Z, Zan H, Casali P. A humanized mouse that mounts mature class-switched, hypermutated and neutralizing antibody responses. Nat Immunol 2024; 25:1489-1506. [PMID: 38918608 PMCID: PMC11291283 DOI: 10.1038/s41590-024-01880-3] [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: 11/20/2023] [Accepted: 05/18/2024] [Indexed: 06/27/2024]
Abstract
Humanized mice are limited in terms of modeling human immunity, particularly with regards to antibody responses. Here we constructed a humanized (THX) mouse by grafting non-γ-irradiated, genetically myeloablated KitW-41J mutant immunodeficient pups with human cord blood CD34+ cells, followed by 17β-estradiol conditioning to promote immune cell differentiation. THX mice reconstitute a human lymphoid and myeloid immune system, including marginal zone B cells, germinal center B cells, follicular helper T cells and neutrophils, and develop well-formed lymph nodes and intestinal lymphoid tissue, including Peyer's patches, and human thymic epithelial cells. These mice have diverse human B cell and T cell antigen receptor repertoires and can mount mature T cell-dependent and T cell-independent antibody responses, entailing somatic hypermutation, class-switch recombination, and plasma cell and memory B cell differentiation. Upon flagellin or a Pfizer-BioNTech coronavirus disease 2019 (COVID-19) mRNA vaccination, THX mice mount neutralizing antibody responses to Salmonella or severe acute respiratory syndrome coronavirus 2 Spike S1 receptor-binding domain, with blood incretion of human cytokines, including APRIL, BAFF, TGF-β, IL-4 and IFN-γ, all at physiological levels. These mice can also develop lupus autoimmunity after pristane injection. By leveraging estrogen activity to support human immune cell differentiation and maturation of antibody responses, THX mice provide a platform to study the human immune system and to develop human vaccines and therapeutics.
Collapse
Affiliation(s)
- Daniel P Chupp
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
- Invivyd, Waltham, MA, USA
| | - Carlos E Rivera
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Yulai Zhou
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Yijiang Xu
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Patrick S Ramsey
- Department of Obstetrics & Gynecology, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Zhenming Xu
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Hong Zan
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
- Prellis Biologics, Berkeley, CA, USA
| | - Paolo Casali
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA.
- Department of Medicine, The University of Texas Long School of Medicine, San Antonio, TX, USA.
| |
Collapse
|
48
|
Woelfel S, Dütschler J, Junker D, König M, Leinenkugel G, Graf N, Krieger C, Truniger S, Franke A, Koller S, Metzger-Peter K, Oberholzer M, Frei N, Geissler N, Schaub P, STAR SIGN Investigators, Albrich WC, Friedrich M, Niess JH, Schneiderhan-Marra N, Dulovic A, Korte W, Bürgi JJ, Brand S. Systemic and Mucosal Immunogenicity of Monovalent XBB.1.5-Adapted COVID-19 mRNA Vaccines in Patients with Inflammatory Bowel Disease. Vaccines (Basel) 2024; 12:774. [PMID: 39066413 PMCID: PMC11281571 DOI: 10.3390/vaccines12070774] [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: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Recently updated COVID-19 mRNA vaccines encode the spike protein of the omicron subvariant XBB.1.5 and are recommended for patients with inflammatory bowel disease (IBD) on immunosuppressive treatment. Nonetheless, their immunogenicity in patients with IBD against rapidly expanding virus variants remains unknown. This prospective multicenter cohort study is the first study to investigate the immunogenicity of XBB.1.5-adapted vaccines in patients with IBD. Systemic and mucosal antibodies targeting the receptor-binding domains (RBDs) of the omicron subvariants XBB.1.5, EG.5.1, and BA.2.86, as well as their neutralization were quantified before and two to four weeks after vaccination with monovalent XBB.1.5-adapted mRNA vaccines. Vaccination increased levels of serum anti-RBD IgG targeting XBB.1.5, EG.5.1, and BA.2.86 (1.9-fold, 1.8-fold, and 2.6-fold, respectively) and enhanced corresponding neutralization responses (2.3-fold, 3.1-fold, and 3.5-fold, respectively). Following vaccination, anti-TNF-treated patients had reduced virus neutralization compared to patients on treatments with other cellular targets. 11.1% and 16.7% of patients lacked EG.5.1 and BA.2.86 neutralization, respectively; all these patients received anti-TNF treatment. At mucosal sites, vaccination induced variant-specific anti-RBD IgG but failed to induce RBD-targeting IgA. Our findings provide a basis for future vaccine recommendations while highlighting the importance of frequent booster vaccine adaptation and the need for mucosal vaccination strategies in patients with IBD.
Collapse
Affiliation(s)
- Simon Woelfel
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, Ludwig Maximilian University (LMU), 80333 Munich, Germany
| | - Joel Dütschler
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
- Outpatient Clinic, Ambulatory Services Rorschach, 9400 Rorschach, Switzerland
| | - Daniel Junker
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Marius König
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Georg Leinenkugel
- Department of Gastroenterology and Hepatology, University Digestive Healthcare Center, Clarunis, 4002 Basel, Switzerland
| | - Nicole Graf
- Clinical Trials Unit, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Claudia Krieger
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Samuel Truniger
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
- Outpatient Clinic, Ambulatory Services Rorschach, 9400 Rorschach, Switzerland
| | - Annett Franke
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
- Outpatient Clinic, Ambulatory Services Rorschach, 9400 Rorschach, Switzerland
| | - Seraina Koller
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Katline Metzger-Peter
- Department of Gastroenterology and Hepatology, University Digestive Healthcare Center, Clarunis, 4002 Basel, Switzerland
| | | | - Nicola Frei
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Nora Geissler
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Peter Schaub
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | | | - Werner C. Albrich
- Division of Infectious Diseases, Infection Prevention, & Travel Medicine, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Matthias Friedrich
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Jan Hendrik Niess
- Department of Gastroenterology and Hepatology, University Digestive Healthcare Center, Clarunis, 4002 Basel, Switzerland
- Gastroenterology Group, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | | | - Alex Dulovic
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Wolfgang Korte
- Center for Laboratory Medicine, 9001 St. Gallen, Switzerland
| | - Justus J. Bürgi
- Center for Laboratory Medicine, 9001 St. Gallen, Switzerland
| | - Stephan Brand
- Department of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| |
Collapse
|
49
|
Esih H, Mezgec K, Billmeier M, Malenšek Š, Benčina M, Grilc B, Vidmar S, Gašperlin M, Bele M, Zidarn M, Zupanc TL, Morgan T, Jordan I, Sandig V, Schrödel S, Thirion C, Protzer U, Wagner R, Lainšček D, Jerala R. Mucoadhesive film for oral delivery of vaccines for protection of the respiratory tract. J Control Release 2024; 371:179-192. [PMID: 38795814 DOI: 10.1016/j.jconrel.2024.05.041] [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: 02/05/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
The delivery of vaccines plays a pivotal role in influencing the strength and longevity of the immune response and controlling reactogenicity. Mucosal immunization, as compared to parenteral vaccination, could offer greater protection against respiratory infections while being less invasive. While oral vaccination has been presumed less effective and believed to target mainly the gastrointestinal tract, trans-buccal delivery using mucoadhesive films (MAF) may allow targeted delivery to the mucosa. Here we present an effective strategy for mucosal delivery of several vaccine platforms incorporated in MAF, including DNA plasmids, viral vectors, and lipid nanoparticles incorporating mRNA (mRNA/LNP). The mRNA/LNP vaccine formulation targeting SARS-CoV-2 as a proof of concept remained stable within MAF consisting of slowly releasing water-soluble polymers and an impermeable backing layer, facilitating enhanced penetration into the oral mucosa. This formulation elicited antibody and cellular responses comparable to the intramuscular injection, but also induced the production of mucosal IgAs, highlighting its efficacy, particularly for use as a booster vaccine and the potential advantage for protection against respiratory infections. The MAF vaccine preparation demonstrates significant advantages, such as efficient delivery, stability, and simple noninvasive administration with the potential to alleviate vaccine hesitancy.
Collapse
Affiliation(s)
- Hana Esih
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Klemen Mezgec
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Martina Billmeier
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia
| | - Blaž Grilc
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ljubljana 1000, Slovenia
| | - Sara Vidmar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mirjana Gašperlin
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ljubljana 1000, Slovenia
| | - Marjan Bele
- Department of Materials Chemistry, National Institute of Chemistry, Ljubljana 1000, Slovenia
| | - Mihaela Zidarn
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia
| | | | - Tina Morgan
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia
| | - Ingo Jordan
- Applied Science & Technologies, ProBioGen AG, Berlin, Germany
| | - Volker Sandig
- Applied Science & Technologies, ProBioGen AG, Berlin, Germany
| | - Silke Schrödel
- SIRION Biotech GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
| | | | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, Helmholtz Zentrum München, Munich, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology & Hygiene, University Hospital, Regensburg, Germany
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia.
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia.
| |
Collapse
|
50
|
Medrano-Arranz C, Rincón S, Zurita L, Ponz F, Truchado DA. Antigen-functionalized turnip mosaic virus nanoparticles increase antibody sensing in saliva. A case study with SARS-CoV-2 RBD. Diagn Microbiol Infect Dis 2024; 109:116298. [PMID: 38604075 DOI: 10.1016/j.diagmicrobio.2024.116298] [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: 01/23/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Nanoparticles derived from plant viruses play an important role in nanomedicine due to their biocompatibility, self-assembly and easily-modifiable surface. In this study, we developed a novel platform for increasing antibody sensing using viral nanoparticles derived from turnip mosaic virus (TuMV) functionalized with SARS-CoV-2 receptor binding domain (RBD) through three different methods: chemical conjugation, gene fusion and the SpyTag/SpyCatcher technology. Even though gene fusion turned out to be unsuccessful, the other two constructs were proven to significantly increase antibody sensing when tested with saliva of patients with different infection and vaccination status to SARS-CoV-2. Our findings show the high potential of TuMV nanoparticles in the fields of diagnostics and immunodetection, being especially attractive for the development of novel antibody sensing devices.
Collapse
Affiliation(s)
- Carlos Medrano-Arranz
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Sara Rincón
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Lucía Zurita
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Ponz
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, 28223 Pozuelo de Alarcón, Madrid, Spain.
| | - Daniel A Truchado
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, 28223 Pozuelo de Alarcón, Madrid, Spain
| |
Collapse
|