Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 3 de 3
Filtre
1.
NPJ Vaccines ; 8(1): 47, 2023 Mar 27.
Article Dans Anglais | MEDLINE | ID: covidwho-2255715

Résumé

SARS-CoV-2 vaccines prevent severe disease but are less efficient in averting infection and transmission of variant strains, making it imperative to explore ways of enhancing protection. Use of inbred mice expressing the human SARS-CoV-2 receptor facilitates such investigations. We employed recombinant MVAs (rMVAs) expressing modified S of several SARS-CoV-2 strains and compared their ability to neutralize variants, bind S proteins and protect K18-hACE2 mice against SARS-CoV-2 challenge when administered intramuscularly or intranasally. The rMVAs expressing Wuhan, Beta and Delta S induced substantial cross neutralizing activities to each other but very low neutralization of Omicron; while rMVA expressing Omicon S induced neutralizing antibody predominanly to Omicron. In mice primed and boosted with rMVA expressing the Wuhan S, neutralizing antibodies to Wuhan increased after one immunization with rMVA expressing Omicron S due to original antigenic sin, but substantial neutralizing antibody to Omicron required a second immunization. Nevertheless, monovalent vaccines with S mismatched to the challenge virus still protected against severe disease and reduced the amounts of virus and subgenomic RNAs in the lungs and nasal turbinates, though not as well as vaccines with matched S. Passive transfer of Wuhan immune serum with Omicron S binding but undetectable neutralizing activity reduced infection of the l-ungs by Omicron suggesting additional effector functions. Notably, there was less infectious virus and viral subgenomic RNAs in the nasal turbinates and lungs when the rMVAs were administered intranasally rather than intramuscularly and this held true for vaccines that were matched or mismatched to the challenge strain of SARS-CoV-2.

2.
Proc Natl Acad Sci U S A ; 119(24): e2202069119, 2022 06 14.
Article Dans Anglais | MEDLINE | ID: covidwho-1890415

Résumé

Current vaccines have greatly diminished the severity of the COVID-19 pandemic, even though they do not entirely prevent infection and transmission, likely due to insufficient immunity in the upper respiratory tract. Here, we compare intramuscular and intranasal administration of a live, replication-deficient modified vaccinia virus Ankara (MVA)-based Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) spike (S) vaccine to raise protective immune responses in the K18-hACE2 mouse model. Using a recombinant MVA expressing firefly luciferase for tracking, live imaging revealed luminescence of the respiratory tract of mice within 6 h and persisting for 3 d following intranasal inoculation, whereas luminescence remained at the site of intramuscular vaccination. Intramuscular vaccination induced S-binding-Immunoglobulin G (IgG) and neutralizing antibodies in the lungs, whereas intranasal vaccination also induced Immunoglobulin A (IgA) and higher levels of antigen-specific CD3+CD8+IFN-γ+ T cells. Similarly, IgG and neutralizing antibodies were present in the blood of mice immunized intranasally and intramuscularly, but IgA was detected only after intranasal inoculation. Intranasal boosting increased IgA after intranasal or intramuscular priming. While intramuscular vaccination prevented morbidity and cleared SARS-CoV-2 from the respiratory tract within several days after challenge, intranasal vaccination was more effective as neither infectious virus nor viral messenger (m)RNAs were detected in the nasal turbinates or lungs as early as 2 d after challenge, indicating prevention or rapid elimination of SARS-CoV-2 infection. Additionally, we determined that neutralizing antibody persisted for more than 6 mo and that serum induced to the Wuhan S protein neutralized pseudoviruses expressing the S proteins of variants, although with less potency, particularly for Beta and Omicron.


Sujets)
Vaccins contre la COVID-19 , COVID-19 , Immunoglobuline A , Appareil respiratoire , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Virus de la vaccine , Administration par voie nasale , Angiotensin-converting enzyme 2/génétique , Animaux , Anticorps neutralisants/sang , Anticorps antiviraux/sang , COVID-19/prévention et contrôle , COVID-19/transmission , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/immunologie , Humains , Immunoglobuline A/sang , Immunoglobuline G/sang , Souris , Souris transgéniques , Appareil respiratoire/immunologie , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Vaccination/méthodes , Virus de la vaccine/génétique , Virus de la vaccine/immunologie
3.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article Dans Anglais | MEDLINE | ID: covidwho-1125261

Résumé

Modified vaccinia virus Ankara (MVA) is a replication-restricted smallpox vaccine, and numerous clinical studies of recombinant MVAs (rMVAs) as vectors for prevention of other infectious diseases, including COVID-19, are in progress. Here, we characterize rMVAs expressing the S protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Modifications of full-length S individually or in combination included two proline substitutions, mutations of the furin recognition site, and deletion of the endoplasmic retrieval signal. Another rMVA in which the receptor binding domain (RBD) is flanked by the signal peptide and transmembrane domains of S was also constructed. Each modified S protein was displayed on the surface of rMVA-infected cells and was recognized by anti-RBD antibody and soluble hACE2 receptor. Intramuscular injection of mice with the rMVAs induced antibodies, which neutralized a pseudovirus in vitro and, upon passive transfer, protected hACE2 transgenic mice from lethal infection with SARS-CoV-2, as well as S-specific CD3+CD8+IFNγ+ T cells. Antibody boosting occurred following a second rMVA or adjuvanted purified RBD protein. Immunity conferred by a single vaccination of hACE2 mice prevented morbidity and weight loss upon intranasal infection with SARS-CoV-2 3 wk or 7 wk later. One or two rMVA vaccinations also prevented detection of infectious SARS-CoV-2 and subgenomic viral mRNAs in the lungs and greatly reduced induction of cytokine and chemokine mRNAs. A low amount of virus was found in the nasal turbinates of only one of eight rMVA-vaccinated mice on day 2 and none later. Detection of low levels of subgenomic mRNAs in turbinates indicated that replication was aborted in immunized animals.


Sujets)
Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Vecteurs génétiques/génétique , SARS-CoV-2/immunologie , Vaccins à ADN/immunologie , Virus de la vaccine/génétique , Angiotensin-converting enzyme 2/génétique , Animaux , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Spécificité des anticorps/immunologie , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/génétique , Modèles animaux de maladie humaine , Expression des gènes , Humains , Immunisation , Immunisation passive , Immunoglobuline G/immunologie , Souris , Souris transgéniques , Glycoprotéine de spicule des coronavirus/immunologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Vaccins à ADN/administration et posologie , Vaccins à ADN/génétique
SÉLECTION CITATIONS
Détails de la recherche