Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-22270043

RESUMO

BackgroundDespite the availability of effective vaccines against coronavirus disease 2019 (Covid-19), the emergence of variant strains and breakthrough infections pose a challenging new reality. Booster vaccinations are needed to maintain vaccine-induced protection. MethodsENSEMBLE2 is an ongoing, randomized, double-blind, placebo-controlled, phase 3 pivotal trial including crossover vaccination after emergency authorization of Covid-19 vaccines. Adults aged [≥]18 years were randomized to receive Ad26.COV2.S or placebo as a primary dose plus a booster dose at two months. The primary endpoint was vaccine efficacy against the first occurrence of molecularly-confirmed moderate to severe-critical Covid-19 with onset [≥]14 days after booster vaccination in the per-protocol population. Key efficacy, safety, and immunogenicity endpoints were also assessed. ResultsThe double-blind phase enrolled 31,300 participants, 14,492 of whom received 2 doses and were evaluable for efficacy (per-protocol set, Ad26.COV2.S n=7484; placebo n=7008). Baseline demographics and characteristics were balanced. Vaccine efficacy was 75.2% (adjusted 95% CI, 54.6-87.3) against moderate to severe-critical Covid-19 and was similar against symptomatic infection (75.6% [55.5-99.9]). Efficacy was consistent across participants with and without comorbidities, and reached 93.7% (58.5-99.9) in the US. Vaccine efficacy against severe-critical Covid-19 was 100% (32.6-100.0; 0 vs 8 cases). The booster vaccine induced robust humoral responses and exhibited an acceptable safety profile. ConclusionsA homologous Ad26.COV2.S booster administered 2 months after primary single-dose vaccination in adults led to high vaccine efficacy, including against any symptomatic infection and SARS-CoV-2 variants prevalent during the study. (Funding: Janssen Research and Development and others; ENSEMBLE2 ClinicalTrials.gov number, NCT04614948.)

2.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21268268

RESUMO

BackgroundCOVID-19 has so far affected more than 250 million individuals worldwide, causing more than 5 million deaths. Several risk factors for severe disease have been identified, most of which coincide with advanced age. In younger individuals, severe COVID-19 often occurs in the absence of obvious comorbidities. Guided by the finding of cytomegalovirus (CMV)-specific T cells with some cross-reactivity to SARS-CoV-2 in a COVID-19 intensive care unit (ICU) patient, we decided to investigate whether CMV seropositivity is associated with severe or critical COVID-19. MethodsNational German COVID-19 bio-sample and data banks were used to retrospectively analyze the CMV serostatus of patients who experienced mild (n=101), moderate (n=130) or severe to critical (n=80) disease by CMV IgG serology. We then investigated the relationship between disease severity and CMV serostatus via statistical models. ResultsNon-geriatric patients (< 70 years) with severe COVID-19 were found to have a very high prevalence of CMV-seropositivity, while CMV status distribution in individuals with mild disease was similar to the prevalence in the German population; interestingly, this was not detectable in older patients. Prediction models support the hypothesis that the CMV serostatus might be a strong biomarker in identifying younger individuals with a higher risk of developing severe COVID-19. ConclusionsWe identified CMV-seropositivity as a potential novel risk factor for severe COVID-19 in non-geriatric individuals in the studied cohorts. More mechanistic analyses as well as confirmation of similar findings in cohorts representing the currently most relevant SARS-CoV-2 variants should be performed shortly.

3.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21262037

RESUMO

BackgroundSARS-CoV2 has evolved to enter the host via the ACE2 receptor which is part of the Kinin-kallirein pathway. This complex pathway is only poorly understood in context of immune regulation but critical to control infection. This study examines SARS-CoV2 infection and epithelial mechanisms of the kinin-kallikrein system at the kinin B2 receptor level in SARS-CoV-2 infection that is of direct translational relevance. MethodsFrom acute SARS-CoV-2-positive patients and -negative controls, transcriptomes of nasal brushings were analyzed. Primary airway epithelial cells (NHBEs) were infected with SARS-CoV-2 and treated with the approved B2R antagonist icatibant. SARS-CoV-2 RNA RT-qPCR, cytotoxicity assays, plaque assays and transcriptome analyses were performed. The treatment effect was further studied in a murine airway inflammation model in vivo. ResultsHere, we report a broad and strong upregulation of kallikreins and the kinin B2 receptor (B2R) in the nasal mucosa of acutely symptomatic SARS-CoV-2-positive patients. A B2R antagonist impeded SARS-CoV-2 replication and spread in NHBEs, as determined in plaque assays on Vero E6 cells. B2R antagonism reduced the expression of SARS-CoV-2 entry receptor ACE2 in vitro and in a murine airway inflammation model in vivo. In addition, it suppressed gene expression broadly, particularly genes involved in G-protein-coupled-receptor signaling and ion transport. ConclusionsIn summary, this study provides evidence that treatment with B2R antagonists protects airway epithelial cells from SARS-CoV-2 by inhibiting its replication and spread, through the reduction of ACE2 levels and the interference with several cellular signaling processes. Future clinical studies need to shed light on the airway protection potential of approved B2R antagonists, like icatibant, in the treatment of early-stage COVID-19.

4.
Frauke Degenhardt; David Ellinghaus; Simonas Juzenas; Jon Lerga-Jaso; Mareike Wendorff; Douglas Maya-Miles; Florian Uellendahl-Werth; Hesham ElAbd; Malte Christoph Ruehlemann; Jatin Arora; Onur Oezer; Ole Bernt Lenning; Ronny Myhre; May Sissel Vadla; Eike Matthias Wacker; Lars Wienbrandt; Aaron Blandino Ortiz; Adolfo de Salazar; Adolfo Garrido Chercoles; Adriana Palom; Agustin Ruiz; Alba-Estela Garcia-Fernandez; Albert Blanco-Grau; Alberto Mantovani; Alberto Zanella; Aleksander Rygh Holten; Alena Mayer; Alessandra Bandera; Alessandro Cherubini; Alessandro Protti; Alessio Aghemo; Alessio Gerussi; Alfredo Ramirez; Alice Braun; Almut Nebel; Ana Barreira; Ana Lleo; Ana Teles; Anders Kildal; Andrea Biondi; Andrea Caballero-Garralda; Andrea Ganna; Andrea Gori; Andreas Glueck; Andreas Lind; Anja Tanck; Anke Hinney; Anna Carreras Carreras Nolla; Anna Ludovica Fracanzani; Anna Peschuck; Annalisa Cavallero; Anne Ma Dyrhol-Riise; Antonella Ruello; Antonio Julia; Antonio Muscatello; Antonio Pesenti; Antonio Voza; Ariadna Rando-Segura; Aurora Solier; Axel Schmidt; Beatriz Cortes; Beatriz Mateos; Beatriz Nafria-Jimenez; Benedikt Schaefer; Bjoern Jensen; Carla Bellinghausen; Carlo Maj; Carlos Ferrando; Carmen de la Horra; Carmen Quereda; Carsten Skurk; Charlotte Thibeault; Chiara Scollo; Christian Herr; Christoph D Spinner; Christoph Gassner; Christoph Lange; Cinzia Hu; Cinzia Paccapelo; Clara Lehmann; Claudio Angelini; Claudio Cappadona; Clinton Azuure; Cristiana Bianco; Cristina Cea; Cristina Sancho; Dag Arne Lihaug Hoff; Daniela Galimberti; Daniele Prati; David Haschka; David Jimenez; David Pestana; David Toapanta; Eduardo Muniz-Diaz; Elena Azzolini; Elena Sandoval; Eleonora Binatti; Elio Scarpini; Elisa T Helbig; Elisabetta Casalone; Eloisa Urrechaga; Elvezia Maria Paraboschi; Emanuele Pontali; Enric Reverter; Enrique J Calderon; Enrique Navas; Erik Solligard; Ernesto Contro; Eunate Arana-Arri; Fatima Aziz; Federico Garcia; Felix Garcia Sanchez; Ferruccio Ceriotti; Filippo Martinelli-Boneschi; Flora Peyvandi; Florian Kurth; Francesco Blasi; Francesco Malvestiti; Francisco J Medrano; Francisco Mesonero; Francisco Rodriguez-Frias; Frank Hanses; Fredrik Mueller; Georg Hemmrich-Stanisak; Giacomo Bellani; Giacomo Grasselli; Gianni Pezzoli; Giorgio Costantino; Giovanni Albano; Giulia Cardamone; Giuseppe Bellelli; Giuseppe Citerio; Giuseppe Foti; Giuseppe Lamorte; Giuseppe Matullo; Guido Baselli; Hayato Kurihara; Holger Neb; Ilaria My; Ingo Kurth; Isabel Hernandez; Isabell Pink; Itziar de Rojas; Ivan Galvan-Femenia; Jan Cato Holter; Jan Egil Afset; Jan Heyckendorf; Jan Kaessens; Jan Kristian Damas; Jan Rybniker; Janine Altmueller; Javier Ampuero; Javier Martin; Jeanette Erdmann; Jesus M Banales; Joan Ramon Badia; Joaquin Dopazo; Jochen Schneider; Jonas Bergan; Jordi Barretina; Joern Walter; Jose Hernandez Quero; Josune Goikoetxea; Juan Delgado; Juan M Guerrero; Julia Fazaal; Julia Kraft; Julia Schroeder; Kari Risnes; Karina Banasik; Karl Erik Mueller; Karoline I Gaede; Koldo Garcia-Etxebarria; Kristian Tonby; Lars Heggelund; Laura Izquierdo-Sanchez; Laura Rachele Bettini; Lauro Sumoy; Leif Erik Sander; Lena J Lippert; Leonardo Terranova; Lindokuhle Nkambule; Lisa Knopp; Lise Tuset Gustad; Lucia Garbarino; Luigi Santoro; Luis Tellez; Luisa Roade; Mahnoosh Ostadreza; Maider Intxausti; Manolis Kogevinas; Mar Riveiro-Barciela; Marco Schaefer; Mari EK Niemi; Maria A Gutierrez-Stampa; Maria Carrabba; Maria E Figuera Basso; Maria Grazia Valsecchi; Maria Hernandez-Tejero; Maria JGT Vehreschild; Maria Manunta; Marialbert Acosta-Herrera; Mariella D'Angio; Marina Baldini; Marina Cazzaniga; Marit M Grimsrud; Markus Cornberg; Markus M Noethen; Marta Marquie; Massimo Castoldi; Mattia Cordioli; Maurizio Cecconi; Mauro D'Amato; Max Augustin; Melissa Tomasi; Merce Boada; Michael Dreher; Michael J Seilmaier; Michael Joannidis; Michael Wittig; Michela Mazzocco; Michele Ciccarelli; Miguel Rodriguez-Gandia; Monica Bocciolone; Monica Miozzo; Natale Imaz-Ayo; Natalia Blay; Natalia Chueca; Nicola Montano; Nicole Braun; Nicole Ludwig; Nikolaus Marx; Nilda Martinez; Oliver A Cornely; Oliver Witzke; Orazio Palmieri; Paola Faverio; Paoletta Preatoni; Paolo Bonfanti; Paolo Omodei; Paolo Tentorio; Pedro Castro; Pedro M Rodrigues; Pedro Pablo Espana; Per Hoffmann; Philip Rosenstiel; Philipp Schommers; Phillip Suwalski; Raul de Pablo; Ricard Ferrer; Robert Bals; Roberta Gualtierotti; Rocio Gallego-Duran; Rosa Nieto; Rossana Carpani; Ruben Morilla; Salvatore Badalamenti; Sammra Haider; Sandra Ciesek; Sandra May; Sara Bombace; Sara Marsal; Sara Pigazzini; Sebastian Klein; Serena Pelusi; Sibylle Wilfling; Silvano Bosari; Sonja Volland; Soren Brunak; Soumya Raychaudhuri; Stefan Schreiber; Stefanie Heilmann-Heimbach; Stefano Aliberti; Stephan Ripke; Susanne Dudman; Tanja Wesse; Tenghao Zheng; Thomas Bahmer; Thomas Eggermann; Thomas Illig; Thorsten Brenner; Tomas Pumarola; Torsten Feldt; Trine Folseraas; Trinidad Gonzalez Cejudo; Ulf Landmesser; Ulrike Protzer; Ute Hehr; Valeria Rimoldi; Valter Monzani; Vegard Skogen; Verena Keitel; Verena Kopfnagel; Vicente Friaza; Victor Andrade; Victor Moreno; Wolfgang Albrecht; Wolfgang Peter; Wolfgang Poller; Xavier Farre; Xiaoli Yi; Xiaomin Wang; Yascha Khodamoradi; Zehra Karadeniz; Anna Latiano; Siegfried Goerg; Petra Bacher; Philipp Koehler; Florian Tran; Heinz Zoller; Eva C Schulte; Bettina Heidecker; Kerstin U Ludwig; Javier Fernandez; Manuel Romero-Gomez; Agustin Albillos; Pietro Invernizzi; Maria Buti; Stefano Duga; Luis Bujanda; Johannes R Hov; Tobias L Lenz; Rosanna Asselta; Rafael de Cid; Luca Valenti; Tom Hemming Karlsen; Mario Caceres; Andre Franke; - COVICAT study group; - Covid-19 Aachen Study (COVAS); - Pa COVID-19 Study Group; - The Humanitas COVID-19 Task Force; - The Humanitas Gavazzeni COVID-19 Task Force; - Norwegian SARS-CoV-2 Study group.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21260624

RESUMO

Given the highly variable clinical phenotype of Coronavirus disease 2019 (COVID-19), a deeper analysis of the host genetic contribution to severe COVID-19 is important to improve our understanding of underlying disease mechanisms. Here, we describe an extended GWAS meta-analysis of a well-characterized cohort of 3,260 COVID-19 patients with respiratory failure and 12,483 population controls from Italy, Spain, Norway and Germany/Austria, including stratified analyses based on age, sex and disease severity, as well as targeted analyses of chromosome Y haplotypes, the human leukocyte antigen (HLA) region and the SARS-CoV-2 peptidome. By inversion imputation, we traced a reported association at 17q21.31 to a highly pleiotropic [~]0.9-Mb inversion polymorphism and characterized the potential effects of the inversion in detail. Our data, together with the 5th release of summary statistics from the COVID-19 Host Genetics Initiative, also identified a new locus at 19q13.33, including NAPSA, a gene which is expressed primarily in alveolar cells responsible for gas exchange in the lung.

5.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21257324

RESUMO

SARS-CoV-2 infection induces a coagulopathy characterized by platelet activation and a hypercoagulable state with an increased incidence of cardiovascular events. The viral spike protein S has been reported to enhance thrombosis formation, stimulate platelets to release pro-coagulant factors and promote the formation of platelet-leukocyte aggregates even in absence of the virus. Although SARS-CoV-2 vaccines induce spike protein overexpression to trigger SARS-CoV-2-specific immune protection, thrombocyte activity has not been investigated in this context. Here, we provide the first phenotypic platelet characterization of healthy human subjects undergoing BNT162b2 vaccination. Using mass cytometry, we analyzed the expression of constitutive transmembrane receptors, adhesion proteins and platelet activation markers in 12 healthy donors before and at five different timepoints within four weeks after the first BNT162b2 administration. We measured platelet reactivity by stimulating thrombocyte activation with thrombin receptor-activating peptide (TRAP). Activation marker expression (P-Selectin, LAMP-3, LAMP-1, CD40L and PAC-1) did not change after vaccination. All investigated constitutive transmembrane proteins showed similar expressions over time. Platelet reactivity was not altered after BNT162b2 administration. Activation marker expression was significantly lower compared to an independent cohort of mild symptomatic COVID-19 patients analyzed with the same platform. This study reveals that BNT162b2 administration does not alter platelet protein expression and reactivity.

6.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21252875

RESUMO

BackgroundThere is considerable variability in COVID-19 outcomes amongst younger adults--and some of this variation may be due to genetic predisposition. We characterized the clinical implications of the major genetic risk factor for COVID-19 severity, and its age-dependent effect, using individual-level data in a large international multi-centre consortium. MethodThe major common COVID-19 genetic risk factor is a chromosome 3 locus, tagged by the marker rs10490770. We combined individual level data for 13,424 COVID-19 positive patients (N=6,689 hospitalized) from 17 cohorts in nine countries to assess the association of this genetic marker with mortality, COVID-19-related complications and laboratory values. We next examined if the magnitude of these associations varied by age and were independent from known clinical COVID-19 risk factors. FindingsWe found that rs10490770 risk allele carriers experienced an increased risk of all-cause mortality (hazard ratio [HR] 1{middle dot}4, 95% confidence interval [CI] 1{middle dot}2-1{middle dot}6) and COVID-19 related mortality (HR 1{middle dot}5, 95%CI 1{middle dot}3-1{middle dot}8). Risk allele carriers had increased odds of several COVID-19 complications: severe respiratory failure (odds ratio [OR] 2{middle dot}0, 95%CI 1{middle dot}6-2{middle dot}6), venous thromboembolism (OR 1{middle dot}7, 95%CI 1{middle dot}2-2{middle dot}4), and hepatic injury (OR 1{middle dot}6, 95%CI 1{middle dot}2-2{middle dot}0). Risk allele carriers [≤] 60 years had higher odds of death or severe respiratory failure (OR 2{middle dot}6, 95%CI 1{middle dot}8-3{middle dot}9) compared to those > 60 years OR 1{middle dot}5 (95%CI 1{middle dot}3-1{middle dot}9, interaction p-value=0{middle dot}04). Amongst individuals [≤] 60 years who died or experienced severe respiratory COVID-19 outcome, we found that 31{middle dot}8% (95%CI 27{middle dot}6-36{middle dot}2) were risk variant carriers, compared to 13{middle dot}9% (95%CI 12{middle dot}6-15{middle dot}2%) of those not experiencing these outcomes. Prediction of death or severe respiratory failure among those [≤] 60 years improved when including the risk allele (AUC 0{middle dot}82 vs 0{middle dot}84, p=0{middle dot}016) and the prediction ability of rs10490770 risk allele was similar to, or better than, most established clinical risk factors. InterpretationThe major common COVID-19 risk locus on chromosome 3 is associated with increased risks of morbidity and mortality--and these are more pronounced amongst individuals [≤] 60 years. The effect on COVID-19 severity was similar to, or larger than most established risk factors, suggesting potential implications for clinical risk management. FundingFunding was obtained by each of the participating cohorts individually.

7.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-20248509

RESUMO

Non-pharmaceutical interventions (NPIs) are important to mitigate the spread of infectious diseases as long as no vaccination or outstanding medical treatments are available. We assess the effectiveness of the sets of non-pharmaceutical interventions that were in place during the course of the Coron-avirus disease 2019 (Covid-19) pandemic in Germany. Our results are based on hybrid models, combining SIR-type models on local scales with spatial resolution. In order to account for the age-dependence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we include realistic pre-pandemic and recently recorded contact patterns between age groups. The implementation of non-pharmaceutical interventions will occur on changed contact patterns, improved isolation, or reduced infectiousness when, e.g., wearing masks. In order to account for spatial heterogeneity, we use a graph approach and we include high-quality information on commuting activities combined with traveling information from social networks. The remaining uncertainty will be accounted for by a large number of randomized simulation runs. Based on the derived factors for the effectiveness of different non-pharmaceutical interventions over the past months, we provide different forecast scenarios for the upcoming time. Mathematics Subject Classification (2010) 00A72 {middle dot} 65L05 {middle dot} 68U20

8.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-20206136

RESUMO

BackgroundHospital staff are at high risk of infection during the coronavirus disease (COVID-19) pandemic. We analysed the exposure characteristics, efficacy of protective measures, and transmission dynamics in this hospital-wide prospective seroprevalence study. Methods and FindingsOverall, 4554 individuals were tested for anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG antibodies using a chemiluminescent immunoassay. Individual risk factors, use of personal protective equipment (PPE), occupational exposure, previous SARS-CoV-2 infection, and symptoms were assessed using a questionnaire and correlated to anti-SARS-CoV-2 IgG antibody titres and PCR testing results. Odds ratios with corresponding exact 95% confidence intervals were used to evaluate associations between individual factors and seropositivity. Spatio-temporal trajectories of SARS-CoV-2-infected patients and staff mobility within the hospital were visualised to identify local hotspots of virus transmission. The overall seroprevalence of anti-SARS-CoV-2-IgG antibody was 2.4% [95% CI 1.9-2.9]. Patient-facing staff, including those working in COVID-19 areas, had a similar probability of being seropositive as non-patient-facing staff. Prior interaction with SARS-CoV-2-infected co-workers or private contacts and unprotected exposure to COVID-19 patients increased the probability of seropositivity. Loss of smell and taste had the highest positive predictive value for seropositivity. The rate of asymptomatic SARS-CoV-2 infections was 25.9%, and higher anti-SARS-CoV-2 IgG antibody titres were observed in symptomatic individuals. Spatio-temporal hotspots of SARS-CoV-2-positive staff and patients only showed partial overlap. ConclusionsPatient-facing work in a healthcare facility during the SARS-CoV-2 pandemic may be safe if adequate PPE and hygiene measures are applied. The high numbers of asymptomatic SARS-CoV-2 infections that escaped detection by symptomatic testing underline the value of cross-sectional seroprevalence studies. Unprotected contact is a major risk factor for infection and argues for the rigorous implementation of hygiene measures.

9.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-20076349

RESUMO

The evolving dynamics of coronavirus disease 2019 (COVID-19) and the increasing infection numbers require diagnostic tools to identify patients at high risk for a severe disease course. Here we evaluate clinical and imaging parameters for estimating the need of intensive care unit (ICU) treatment. We collected clinical, laboratory and imaging data from 65 patients with confirmed COVID-19 infection based on PCR testing. Two radiologists evaluated the severity of findings in computed tomography (CT) images on a scale from 1 (no characteristic signs of COVID-19) to 5 (confluent ground glass opacities in over 50% of the lung parenchyma). The volume of affected lung was quantified using commercially available software. Machine learning modelling was performed to estimate the risk for ICU treatment. Patients with a severe course of COVID-19 had significantly increased IL-6, CRP and leukocyte counts and significantly decreased lymphocyte counts. The radiological severity grading was significantly increased in ICU patients. Multivariate random forest modelling showed a mean {+/-} standard deviation sensitivity, specificity and accuracy of 0.72 {+/-} 0.1, 0.86 {+/-} 0.16 and 0.80 {+/-} 0.1 and a ROC-AUC of 0.79 {+/-} 0.1. The need for ICU treatment is independently associated with affected lung volume, radiological severity score, CRP and IL-6.

10.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-20073767

RESUMO

Data on people living with human immunodeficiency virus (PLWH) in the current SARS-CoV-2 pandemic is still scarce. This case series of 33 PLWH patients with COVID-19 reveals symptoms and outcome in this special population. Three out of 32 patients with documented outcome died (9%). However, 91% of the patients recovered and 76% have been classified as mild cases, indicating that there is no excess morbidity and mortality among PLWH with symptomatic COVID-19. All patients were on antiretroviral treatment, of them 22 on tenofovir-containing regimen, and 4 on the protease inhibitor darunavir.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...