Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtre
1.
Front Immunol ; 14: 1166589, 2023.
Article Dans Anglais | MEDLINE | ID: covidwho-2321884

Résumé

Since early 2022, various Omicron variants have dominated the SARS-CoV-2 pandemic in most countries. All Omicron variants are B-cell immune escape variants, and antibodies induced by first-generation COVID-19 vaccines or by infection with earlier SARS-CoV-2 variants largely fail to protect individuals from Omicron infection. In the present study, we investigated the effect of Omicron infections in triple-vaccinated and in antigen-naive individuals. We show that Omicron breakthrough infections occurring 2-3.5 months after the third vaccination restore B-cell and T-cell immune responses to levels similar to or higher than those measured 14 days after the third vaccination, including the induction of Omicron-neutralizing antibodies. Antibody responses in breakthrough infection derived mostly from cross-reacting B cells, initially induced by vaccination, whereas Omicron infections in antigen-naive individuals primarily generated B cells binding to the Omicron but not the Wuhan spike protein. Although antigen-naive individuals mounted considerable T-cell responses after infection, B-cell responses were low, and neutralizing antibodies were frequently below the limit of detection. In summary, the detection of Omicron-associated B-cell responses in primed and in antigen-naive individuals supports the application of Omicron-adapted COVID-19 vaccines, but calls into question their suitability if they also contain/encode antigens of the original Wuhan virus.


Sujets)
COVID-19 , Humains , Vaccins contre la COVID-19 , SARS-CoV-2 , Anticorps neutralisants ,
2.
Cell Genom ; : 100232, 2022 Dec 02.
Article Dans Anglais | MEDLINE | ID: covidwho-2237461

Résumé

SARS-CoV-2 infection causes severe COVID-19 in some patients and mild in others. Dysfunctional innate immune responses have been identified to contribute to COVID-19 severity, but the key regulators are still unknown. Here, we present an integrative single-cell multi-omics analysis of peripheral blood mononuclear cells from hospitalized and convalescent COVID-19 patients. In classical monocytes, we identified genes that were potentially regulated by differential chromatin accessibility. Then, sub-clustering and motif-enrichment analyses reveals disease condition-specific regulation by transcription factors and their targets, including an interaction between C/EBPs and a long-noncoding RNA LUCAT1, which we validated through loss-of-function experiments. Finally, we investigated genetic risk variants that exhibit allele-specific open chromatin (AsoC) in COVID-19 patients and identified a SNP rs6800484-C, which is associated with lower expression of CCR2 and may contribute to higher viral loads and higher risk of COVID-19 hospitalization. Altogether, our study highlights the diverse genetic and epigenetic regulators that contribute to COVID-19.

4.
Nat Commun ; 13(1): 4872, 2022 08 18.
Article Dans Anglais | MEDLINE | ID: covidwho-1991596

Résumé

Heterologous prime/boost vaccination with a vector-based approach (ChAdOx-1nCov-19, ChAd) followed by an mRNA vaccine (e.g. BNT162b2, BNT) has been reported to be superior in inducing protective immunity compared to repeated application of the same vaccine. However, data comparing immunity decline after homologous and heterologous vaccination as well as effects of a third vaccine application after heterologous ChAd/BNT vaccination are lacking. Here we show longitudinal monitoring of ChAd/ChAd (n = 41) and ChAd/BNT (n = 88) vaccinated individuals and the impact of a third vaccination with BNT. The third vaccination greatly augments waning anti-spike IgG but results in only moderate increase in spike-specific CD4 + and CD8 + T cell numbers in both groups, compared to cell frequencies already present after the second vaccination in the ChAd/BNT group. More importantly, the third vaccination efficiently restores neutralizing antibody responses against the Alpha, Beta, Gamma, and Delta variants of the virus, but neutralizing activity against the B.1.1.529 (Omicron) variant remains severely impaired. In summary, inferior SARS-CoV-2 specific immune responses following homologous ChAd/ChAd vaccination can be compensated by heterologous BNT vaccination, which might influence the choice of vaccine type for subsequent vaccination boosts.


Sujets)
COVID-19 , Anticorps neutralisants , Anticorps antiviraux , Production d'anticorps , Vaccin BNT162 , COVID-19/prévention et contrôle , Humains , SARS-CoV-2 , Vaccination , Vaccins synthétiques , Vaccins à ARNm
5.
Front Immunol ; 13: 863039, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-1775685

Résumé

Evaluating long-term protection against SARS-CoV-2 variants of concern in convalescing individuals is of high clinical relevance. In this prospective study of a cohort of 46 SARS-CoV-2 patients infected with the Wuhan strain of SARS-CoV-2 we longitudinally analyzed changes in humoral and cellular immunity upon early and late convalescence. Antibody neutralization capacity was measured by surrogate virus neutralization test and cellular responses were investigated with 31-colour spectral flow cytometry. Spike-specific, isotype-switched B cells developed already during the disease phase, showed a memory phenotype and did not decrease in numbers even during late convalescence. Otherwise, no long-lasting perturbations of the immune compartment following COVID-19 clearance were observed. During convalescence anti-Spike (S1) IgG antibodies strongly decreased in all patients. We detected neutralizing antibodies against the Wuhan strain as well as the Alpha and Delta but not against the Beta, Gamma or Omicron variants for up to 7 months post COVID-19. Furthermore, correlation analysis revealed a strong association between sera anti-S1 IgG titers and their neutralization capacity against the Wuhan strain as well as Alpha and Delta. Overall, our data suggest that even 7 month after the clearance of COVID-19 many patients possess a protective layer of immunity, indicated by the persistence of Spike-specific memory B cells and by the presence of neutralizing antibodies against the Alpha and Delta variants. However, lack of neutralizing antibodies against the Beta, Gamma and Omicron variants even during the peak response is of major concern as this indicates viral evasion of the humoral immune response.


Sujets)
COVID-19 , SARS-CoV-2 , Anticorps neutralisants , Convalescence , Humains , Immunité humorale , Immunoglobuline G , Études prospectives , Glycoprotéine de spicule des coronavirus/génétique
7.
Eur J Immunol ; 52(2): 356-359, 2022 02.
Article Dans Anglais | MEDLINE | ID: covidwho-1555488

Résumé

Sera of vaccines were assessed by surrogate virus neutralization tests for their capacity to neutralize the SARS-CoV-2 Delta variant. Homologous prime-boost immunization with Moderna's Spikevax as well as heterologous immunization with AstraZeneca's Vaxzevria followed by Moderna's Spikevax were identified as highly potent vaccination regimens for the induction of Delta-neutralizing antibodies.


Sujets)
Anticorps neutralisants/sang , Anticorps antiviraux/sang , Vaccins contre la COVID-19/administration et posologie , COVID-19/sang , SARS-CoV-2/métabolisme , Vaccination , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , Femelle , Humains , Mâle , SARS-CoV-2/immunologie
8.
Front Immunol ; 12: 772240, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1551510

Résumé

Antigen-specific tissue-resident memory T cells (Trms) and neutralizing IgA antibodies provide the most effective protection of the lungs from viral infections. To induce those essential components of lung immunity against SARS-CoV-2, we tested various immunization protocols involving intranasal delivery of a novel Modified Vaccinia virus Ankara (MVA)-SARS-2-spike vaccine candidate. We show that a single intranasal MVA-SARS-CoV-2-S application in mice strongly induced pulmonary spike-specific CD8+ T cells, albeit restricted production of neutralizing antibodies. In prime-boost protocols, intranasal booster vaccine delivery proved to be crucial for a massive expansion of systemic and lung tissue-resident spike-specific CD8+ T cells and the development of Th1 - but not Th2 - CD4+ T cells. Likewise, very high titers of IgG and IgA anti-spike antibodies were present in serum and broncho-alveolar lavages that possessed high virus neutralization capacities to all current SARS-CoV-2 variants of concern. Importantly, the MVA-SARS-2-spike vaccine applied in intramuscular priming and intranasal boosting treatment regimen completely protected hamsters from developing SARS-CoV-2 lung infection and pathology. Together, these results identify intramuscular priming followed by respiratory tract boosting with MVA-SARS-2-S as a promising approach for the induction of local, respiratory as well as systemic immune responses suited to protect from SARS-CoV-2 infections.


Sujets)
Anticorps antiviraux/sang , Lymphocytes T CD8+/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Administration par voie nasale , Animaux , Anticorps neutralisants/sang , Lignée cellulaire , Chlorocebus aethiops , Cricetinae , Vecteurs génétiques , Rappel de vaccin , Immunoglobuline A/sang , Immunoglobuline G/sang , Poumon/immunologie , Mâle , Souris , Souris de lignée C57BL , Lymphocytes auxiliaires Th1/immunologie , Vaccination , Vaccins sous-unitaires/immunologie , Virus de la vaccine/immunologie , Cellules Vero , Charge virale/immunologie
9.
Front Immunol ; 12: 721738, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1378191

Résumé

Here, we described the case of a B cell-deficient patient after CD19 CAR-T cell therapy for refractory B cell Non-Hodgkin Lymphoma with protracted coronavirus disease 2019 (COVID-19). For weeks, this patient only inefficiently contained the virus while convalescent plasma transfusion correlated with virus clearance. Interestingly, following convalescent plasma therapy natural killer cells matured and virus-specific T cells expanded, presumably allowing virus clearance and recovery from the disease. Our findings, thus, suggest that convalescent plasma therapy can activate cellular immune responses to clear SARS-CoV-2 infections. If confirmed in larger clinical studies, these data could be of general importance for the treatment of COVID-19 patients.


Sujets)
Lymphocytes B , COVID-19/immunologie , COVID-19/thérapie , Déficits immunitaires/immunologie , Immunothérapie adoptive , Cellules tueuses naturelles/immunologie , Lymphocytes T/immunologie , Lymphocytes B/immunologie , COVID-19/complications , Femelle , Humains , Immunisation passive , Immunoglobulines par voie veineuse , Déficits immunitaires/complications , Activation des lymphocytes , Lymphopoïèse , SARS-CoV-2 , Charge virale ,
11.
Nat Med ; 27(9): 1525-1529, 2021 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1310811

Résumé

Currently approved viral vector-based and mRNA-based vaccine approaches against coronavirus disease 2019 (COVID-19) consider only homologous prime-boost vaccination. After reports of thromboembolic events, several European governments recommended using AstraZeneca's ChAdOx1-nCov-19 (ChAd) only in individuals older than 60 years, leaving millions of already ChAd-primed individuals with the decision to receive either a second shot of ChAd or a heterologous boost with mRNA-based vaccines. However, such combinations have not been tested so far. We used Hannover Medical School's COVID-19 Contact Study cohort of healthcare professionals to monitor ChAd-primed immune responses before and 3 weeks after booster with ChAd (n = 32) or BioNTech/Pfizer's BNT162b2 (n = 55). Although both vaccines boosted prime-induced immunity, BNT162b2 induced significantly higher frequencies of spike-specific CD4+ and CD8+ T cells and, in particular, high titers of neutralizing antibodies against the B.1.1.7, B.1.351 and P.1 variants of concern of severe acute respiratory syndrome coronavirus 2.


Sujets)
Anticorps neutralisants/sang , Anticorps antiviraux/sang , Vaccins contre la COVID-19/effets indésirables , Vaccins contre la COVID-19/immunologie , SARS-CoV-2/immunologie , Vaccin BNT162 , Numération des lymphocytes CD4 , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , COVID-19/immunologie , Vaccin ChAdOx1 nCoV-19 , Humains , Rappel de vaccin/méthodes , Immunogénicité des vaccins/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Vaccination
12.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article Dans Anglais | MEDLINE | ID: covidwho-1284760

Résumé

Severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) has emerged as the infectious agent causing the pandemic coronavirus disease 2019 (COVID-19) with dramatic consequences for global human health and economics. Previously, we reached clinical evaluation with our vector vaccine based on modified vaccinia virus Ankara (MVA) against the Middle East respiratory syndrome coronavirus (MERS-CoV), which causes an infection in humans similar to SARS and COVID-19. Here, we describe the construction and preclinical characterization of a recombinant MVA expressing full-length SARS-CoV-2 spike (S) protein (MVA-SARS-2-S). Genetic stability and growth characteristics of MVA-SARS-2-S, plus its robust expression of S protein as antigen, make it a suitable candidate vaccine for industrial-scale production. Vaccinated mice produced S-specific CD8+ T cells and serum antibodies binding to S protein that neutralized SARS-CoV-2. Prime-boost vaccination with MVA-SARS-2-S protected mice sensitized with a human ACE2-expressing adenovirus from SARS-CoV-2 infection. MVA-SARS-2-S is currently being investigated in a phase I clinical trial as aspirant for developing a safe and efficacious vaccine against COVID-19.


Sujets)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Glycoprotéine de spicule des coronavirus/immunologie , Animaux , Vaccins contre la COVID-19/normes , Relation dose-réponse (immunologie) , Humains , Souris , Souris de lignée BALB C , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/génétique , Lymphocytes T , Vaccination , Virus de la vaccine
13.
Cell Mol Immunol ; 18(4): 936-944, 2021 04.
Article Dans Anglais | MEDLINE | ID: covidwho-899921

Résumé

Neutralizing antibodies targeting the receptor-binding domain (RBD) of the SARS-CoV-2 spike (S) block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry into cells via surface-expressed angiotensin-converting enzyme 2 (ACE2). We used a surrogate virus neutralization test (sVNT) and SARS-CoV-2 S protein-pseudotyped vesicular stomatitis virus (VSV) vector-based neutralization assay (pVNT) to assess the degree to which serum antibodies from coronavirus disease 2019 (COVID-19) convalescent patients interfere with the binding of SARS-CoV-2 S to ACE2. Both tests revealed neutralizing anti-SARS-CoV-2 S antibodies in the sera of ~90% of mildly and 100% of severely affected COVID-19 convalescent patients. Importantly, sVNT and pVNT results correlated strongly with each other and to the levels of anti-SARS-CoV-2 S1 IgG and IgA antibodies. Moreover, levels of neutralizing antibodies correlated with the duration and severity of clinical symptoms but not with patient age. Compared to pVNT, sVNT is less sophisticated and does not require any biosafety labs. Since this assay is also much faster and cheaper, sVNT will not only be important for evaluating the prevalence of neutralizing antibodies in a population but also for identifying promising plasma donors for successful passive antibody therapy.


Sujets)
Anticorps neutralisants/sang , Anticorps antiviraux/sang , COVID-19/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Adulte , Sujet âgé , Angiotensin-converting enzyme 2/métabolisme , COVID-19/sang , Lignée cellulaire , Convalescence , Femelle , Humains , Immunoglobuline A/sang , Immunoglobuline G/sang , Mâle , Adulte d'âge moyen , Tests de neutralisation/méthodes
14.
Infect Dis Ther ; 9(4): 837-849, 2020 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-743789

Résumé

BACKGROUND: Serology testing is explored for epidemiological research and to inform individuals after suspected infection. During the coronavirus disease 2019 (COVID-19) pandemic, frontline healthcare professionals (HCP) may be at particular risk for infection. No longitudinal data on functional seroconversion in HCP in regions with low COVID-19 prevalence and low pre-test probability exist. METHODS: In a large German university hospital, we performed weekly questionnaire assessments and anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin G (IgG) measurements with various commercial tests, a novel surrogate virus neutralisation test, and a neutralisation assay using live SARS-CoV-2. RESULTS: From baseline to week 6, 1080 screening measurements for anti-SARS CoV-2 (S1) IgG from 217 frontline HCP (65% female) were performed. Overall, 75.6% of HCP reported at least one symptom of respiratory infection. Self-perceived infection probability declined over time (from mean 20.1% at baseline to 12.4% in week 6, p < 0.001). In sera of convalescent patients with PCR-confirmed COVID-19, we measured high anti-SARS-CoV-2 IgG levels, obtained highly concordant results from enzyme-linked immunosorbent assays (ELISA) using e.g. the spike 1 (S1) protein domain and the nucleocapsid protein (NCP) as targets, and confirmed antiviral neutralisation. However, in HCP the cumulative incidence for anti-SARS-CoV-2 (S1) IgG was 1.86% for positive and 0.93% for equivocal positive results over the study period of 6 weeks. Except for one HCP, none of the eight initial positive results were confirmed by alternative serology tests or showed in vitro neutralisation against live SARS-CoV-2. The only true seroconversion occurred without symptoms and mounted strong functional humoral immunity. Thus, the confirmed cumulative incidence for neutralizing anti-SARS-CoV-2 IgG was 0.47%. CONCLUSION: When assessing anti-SARS-CoV-2 immune status in individuals with low pre-test probability, we suggest confirming positive results from single measurements by alternative serology tests or functional assays. Our data highlight the need for a methodical serology screening approach in regions with low SARS-CoV-2 infection rates. TRIAL REGISTRATION: The study is registered at DRKS00021152.

15.
Front Immunol ; 11: 1959, 2020.
Article Dans Anglais | MEDLINE | ID: covidwho-732901

Résumé

The lung is the vital target organ of coronavirus disease 2019 (COVID-19) caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In the majority of patients the most active virus replication seems to be found in the upper respiratory tract, severe cases however suffer from SARS-like disease associated with virus replication in lung tissues. Due to the current lack of suitable anti-viral drugs the induction of protective immunity such as neutralizing antibodies in the lung is the key aim of the only alternative approach-the development and application of SARS-CoV-2 vaccines. However, past experience from experimental animals, livestock, and humans showed that induction of immunity in the lung is limited following application of vaccines at peripheral sides such as skin or muscles. Based on several considerations we therefore propose here to consider the application of a Modified Vaccinia virus Ankara (MVA)-based vaccine to mucosal surfaces of the respiratory tract as a favorable approach to combat COVID-19.


Sujets)
Betacoronavirus/composition chimique , Infections à coronavirus/prévention et contrôle , Pandémies/prévention et contrôle , Pneumopathie virale/prévention et contrôle , Glycoprotéine de spicule des coronavirus/immunologie , Virus de la vaccine/immunologie , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/immunologie , Administration par voie muqueuse , Animaux , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Bronches/immunologie , COVID-19 , Infections à coronavirus/virologie , Humains , Immunoglobuline A/métabolisme , Tissu lymphoïde/immunologie , Plasmocytes/immunologie , Pneumopathie virale/virologie , Muqueuse respiratoire/effets des médicaments et des substances chimiques , Muqueuse respiratoire/immunologie , SARS-CoV-2 , Lymphocytes T/immunologie , Vaccination , Vaccins atténués/immunologie
16.
EBioMedicine ; 57: 102885, 2020 Jul.
Article Dans Anglais | MEDLINE | ID: covidwho-633885

Résumé

BACKGROUND: Elucidating the role of T cell responses in COVID-19 is of utmost importance to understand the clearance of SARS-CoV-2 infection. METHODS: 30 hospitalized COVID-19 patients and 60 age- and gender-matched healthy controls (HC) participated in this study. We used two comprehensive 11-colour flow cytometric panels conforming to Good Laboratory Practice and approved for clinical diagnostics. FINDINGS: Absolute numbers of lymphocyte subsets were differentially decreased in COVID-19 patients according to clinical severity. In severe disease (SD) patients, all lymphocyte subsets were reduced, whilst in mild disease (MD) NK, NKT and γδ T cells were at the level of HC. Additionally, we provide evidence of T cell activation in MD but not SD, when compared to HC. Follow up samples revealed a marked increase in effector T cells and memory subsets in convalescing but not in non-convalescing patients. INTERPRETATION: Our data suggest that activation and expansion of innate and adaptive lymphocytes play a major role in COVID-19. Additionally, recovery is associated with formation of T cell memory as suggested by the missing formation of effector and central memory T cells in SD but not in MD. Understanding T cell-responses in the context of clinical severity might serve as foundation to overcome the lack of effective anti-viral immune response in severely affected COVID-19 patients and can offer prognostic value as biomarker for disease outcome and control. FUNDING: Funded by State of Lower Saxony grant 14-76,103-184CORONA-11/20 and German Research Foundation, Excellence Strategy - EXC2155"RESIST"-Project ID39087428, and DFG-SFB900/3-Project ID158989968, grants SFB900-B3, SFB900-B8.


Sujets)
Betacoronavirus/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Infections à coronavirus/immunologie , Activation des lymphocytes/immunologie , Pneumopathie virale/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques , Lymphocytes T CD4+/cytologie , Lymphocytes T CD8+/cytologie , COVID-19 , Femelle , Humains , Mémoire immunologique/immunologie , Numération des lymphocytes , Mâle , Adulte d'âge moyen , Pandémies , Pronostic , SARS-CoV-2 , Indice de gravité de la maladie , Jeune adulte
SÉLECTION CITATIONS
Détails de la recherche