Cargando…

Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations

INTRODUCTION: Humoral immunity after SARS-CoV-2 vaccination has been extensively investigated in blood. Aim of this study was to develop an ELISA method in order to determine the prevalence of IgG and IgA SARS-CoV-2 domain 1 spike-protein (S) specific antibodies (Abs) in buccal and nasal mucosal sur...

Descripción completa

Detalles Bibliográficos
Autores principales: Tsamadou, Chrysanthi, Ludwig, Carolin, Scholz, Judith, Proffen, Matthias, Hägele, Janina, Rode, Immanuel, Körper, Sixten, Fabricius, Dorit, Jahrsdörfer, Bernd, Neuchel, Christine, Amann, Elisa, Schrezenmeier, Hubert, Fürst, Daniel
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9712976/
https://www.ncbi.nlm.nih.gov/pubmed/36466833
http://dx.doi.org/10.3389/fimmu.2022.999693
_version_ 1784841905618550784
author Tsamadou, Chrysanthi
Ludwig, Carolin
Scholz, Judith
Proffen, Matthias
Hägele, Janina
Rode, Immanuel
Körper, Sixten
Fabricius, Dorit
Jahrsdörfer, Bernd
Neuchel, Christine
Amann, Elisa
Schrezenmeier, Hubert
Fürst, Daniel
author_facet Tsamadou, Chrysanthi
Ludwig, Carolin
Scholz, Judith
Proffen, Matthias
Hägele, Janina
Rode, Immanuel
Körper, Sixten
Fabricius, Dorit
Jahrsdörfer, Bernd
Neuchel, Christine
Amann, Elisa
Schrezenmeier, Hubert
Fürst, Daniel
author_sort Tsamadou, Chrysanthi
collection PubMed
description INTRODUCTION: Humoral immunity after SARS-CoV-2 vaccination has been extensively investigated in blood. Aim of this study was to develop an ELISA method in order to determine the prevalence of IgG and IgA SARS-CoV-2 domain 1 spike-protein (S) specific antibodies (Abs) in buccal and nasal mucosal surfaces of vaccinees. METHODS: To this end, we analyzed 69 individuals who received their first vaccine dose between February and July 2021. Vaccines administered were BNT162b2, mRNA-1273 or ChAdOx1-nCoV-19. Detection of IgG and IgA Abs was performed using commercial ELISA kits for both blood and swab samples after protocol modification for the latter. RESULTS: Anti-spike IgG and IgA Abs in the buccal and/or nasal swabs were detectable in >81% of the study subjects after the second dose. The IgG measurements in buccal swabs appeared to correlate in a more consistent way with the respective measurements in blood with a correlation coefficient of r=0.74. It is of note that IgA Abs appeared to be significantly more prevalent in the nasal compared to the buccal mucosa. Optimal selection of the assay cut-off for the IgG antibody detection in buccal swabs conferred a sensitivity of 91.8% and a specificity of 100%. Last, individuals vaccinated with mRNA-based vaccines exhibited higher antibody levels in both blood and mucosal surfaces compared to those receiving ChAdOx1-nCoV-19 confirming previously reported results. CONCLUSION: In conclusion, our findings show a differential prevalence of anti-S Abs on mucosal surfaces after vaccination for SARS-CoV-2, while they also set the basis for potential future use of IgG antibody detection in buccal swabs for extended immunity screening in large populations.
format Online
Article
Text
id pubmed-9712976
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-97129762022-12-02 Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations Tsamadou, Chrysanthi Ludwig, Carolin Scholz, Judith Proffen, Matthias Hägele, Janina Rode, Immanuel Körper, Sixten Fabricius, Dorit Jahrsdörfer, Bernd Neuchel, Christine Amann, Elisa Schrezenmeier, Hubert Fürst, Daniel Front Immunol Immunology INTRODUCTION: Humoral immunity after SARS-CoV-2 vaccination has been extensively investigated in blood. Aim of this study was to develop an ELISA method in order to determine the prevalence of IgG and IgA SARS-CoV-2 domain 1 spike-protein (S) specific antibodies (Abs) in buccal and nasal mucosal surfaces of vaccinees. METHODS: To this end, we analyzed 69 individuals who received their first vaccine dose between February and July 2021. Vaccines administered were BNT162b2, mRNA-1273 or ChAdOx1-nCoV-19. Detection of IgG and IgA Abs was performed using commercial ELISA kits for both blood and swab samples after protocol modification for the latter. RESULTS: Anti-spike IgG and IgA Abs in the buccal and/or nasal swabs were detectable in >81% of the study subjects after the second dose. The IgG measurements in buccal swabs appeared to correlate in a more consistent way with the respective measurements in blood with a correlation coefficient of r=0.74. It is of note that IgA Abs appeared to be significantly more prevalent in the nasal compared to the buccal mucosa. Optimal selection of the assay cut-off for the IgG antibody detection in buccal swabs conferred a sensitivity of 91.8% and a specificity of 100%. Last, individuals vaccinated with mRNA-based vaccines exhibited higher antibody levels in both blood and mucosal surfaces compared to those receiving ChAdOx1-nCoV-19 confirming previously reported results. CONCLUSION: In conclusion, our findings show a differential prevalence of anti-S Abs on mucosal surfaces after vaccination for SARS-CoV-2, while they also set the basis for potential future use of IgG antibody detection in buccal swabs for extended immunity screening in large populations. Frontiers Media S.A. 2022-11-17 /pmc/articles/PMC9712976/ /pubmed/36466833 http://dx.doi.org/10.3389/fimmu.2022.999693 Text en Copyright © 2022 Tsamadou, Ludwig, Scholz, Proffen, Hägele, Rode, Körper, Fabricius, Jahrsdörfer, Neuchel, Amann, Schrezenmeier and Fürst https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Tsamadou, Chrysanthi
Ludwig, Carolin
Scholz, Judith
Proffen, Matthias
Hägele, Janina
Rode, Immanuel
Körper, Sixten
Fabricius, Dorit
Jahrsdörfer, Bernd
Neuchel, Christine
Amann, Elisa
Schrezenmeier, Hubert
Fürst, Daniel
Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations
title Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations
title_full Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations
title_fullStr Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations
title_full_unstemmed Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations
title_short Differentially induced immunity in buccal and nasal mucosae after vaccination for SARS–CoV–2: Prospects for mass scale immunity-screening in large populations
title_sort differentially induced immunity in buccal and nasal mucosae after vaccination for sars–cov–2: prospects for mass scale immunity-screening in large populations
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9712976/
https://www.ncbi.nlm.nih.gov/pubmed/36466833
http://dx.doi.org/10.3389/fimmu.2022.999693
work_keys_str_mv AT tsamadouchrysanthi differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT ludwigcarolin differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT scholzjudith differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT proffenmatthias differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT hagelejanina differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT rodeimmanuel differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT korpersixten differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT fabriciusdorit differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT jahrsdorferbernd differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT neuchelchristine differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT amannelisa differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT schrezenmeierhubert differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations
AT furstdaniel differentiallyinducedimmunityinbuccalandnasalmucosaeaftervaccinationforsarscov2prospectsformassscaleimmunityscreeninginlargepopulations