Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Intervalo de año de publicación
1.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21268336

RESUMEN

BackgroundAge/frailty are strong predictors of COVID-19 mortality. After the second BNT162b2 dose, immunity wanes faster in older ([≥]65 years) versus younger adults. The durability of response after the third vaccine is unclear. MethodsThis prospective cohort study included healthcare workers/family members[≥]60 years who received a third BNT162b2 dose. Blood samples were drawn immediately before (T0), 10{square}19 (T1), and 74{square}103 (T2) days after the third dose. Anti-spike IgG titers were determined using a commercial assay, seropositivity was defined as[≥]50 AU/mL. Neutralising antibody titres were determined at T2. Adverse events, COVID-19 infections, and clinical frailty scale (CFS) levels were documented. FindingsThe analysis included 97 participants (median age, 70 years [IQR, 66{square}74], 58% CFS level 2). IgG titres, which increased significantly from T0 to T1 (medians, 440 AU/mL [IQR, 294{square}923] and 25,429 [14203{square}36114] AU/mL, respectively; p<0{middle dot}001), decreased significantly by T2, but all remained seropositive (median, 8306 AU/mL [IQR, 4595{square}14701], p<0{middle dot}001 vs T1). In a multivariable analysis, only time from the first vaccine was significantly associated with lower IgG levels at T2 (p=0{middle dot}004). At T2, 60 patients were evaluated for neutralising antibodies; all were seropositive (median, 1294 antibody titre [IQR, 848{square}2072]). Neutralising antibody and anti-spike IgG levels were correlated (R=0{middle dot}6, p<0{middle dot}001). No major adverse events or COVID-19 infections were reported. InterpretationAnti-spike IgG and neutralising antibody levels remain adequate 3 months after the third BNT162b2 vaccine in healthy adults[≥]60 years, although the decline in IgG is concerning. A third vaccine dose in this population should be top priority. FundingNo external funding. Research in contextO_ST_ABSEvidence before this studyC_ST_ABSWe searched PubMed on Aug 1, 2021, for published research articles with no date restrictions, using the search terms of "SARS-Cov-2", "COVID-19", "vaccine", "dose", "antibody response", and "adults" with English as a filter. Several studies were identified that investigated waning of immunity in healthy adults. It is well established through epidemiology and serology studies that in healthy adults, the protection conferred by the BNT162b2 messenger RNA (mRNA) vaccine (Pfizer/BioNtech) wanes significantly after several months. Studies have also shown that the immune response to the vaccine varies with age, and that after the second dose of the BNT162b2 vaccine, the older adult population (65-85 years of age) typically has a lower immune response (as reflected in an analysis of anti-spike IgG antibodies and neutralising antibody titres), than younger adults (18-55 years of age), and that the immunity wanes in all age groups within several months. Added value of this studyThis is, to our knowledge, the first study that examined anti-spike IgG and neutralising antibody titres three months after the third BNT162b2 vaccine dose. The study has demonstrated that three months after that dose, participants, who were healthy adults aged 60 years and older, remained anti-spike IgG seropositive, although a significant decrease in anti-spike IgG titres was observed (compared to two weeks after the third dose). In addition, a statistically significant correlation was observed between the neutralising antibody titres and the anti-spike IgG titres, and all participants were seropositive for neutralising antibodies three months after the third dose. Also, no major adverse events or COVID-19 infections were reported. Implications of all the available evidenceAs our data suggest that a third dose of the BNT162b2 vaccine is effective in maintaining adequate immune response against COVID-19 for at least several months in healthy adults aged 60 years and older, and as it is well established that older adults are at higher risk of severe COVID-19 disease and COVID-19 mortality, providing a third dose to this population should be a top priority. Our data also highlight that understanding the waning of the immune response in other age groups is key for making evidence-based policies regarding booster vaccinations for the population at large.

2.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21266673

RESUMEN

The emergence of rapidly spreading variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a major challenge to the ability of vaccines and therapeutic antibodies to provide immunity. These variants contain mutations at specific amino acids that might impede vaccine efficacy. BriLife(R) (rVSV-{Delta}G-spike) is a newly developed SARS-CoV-2 vaccine candidate currently in Phase II clinical trials. It is based on a replication competent vesicular stomatitis virus (VSV) platform. rVSV-{Delta}G-spike contains several spontaneously-acquired spike mutations that correspond to SARS-CoV-2 variants mutations. We show that human sera from BriLife(R) vaccinees preserve comparable neutralization titers towards alpha, gamma and delta variants, and show less than 3-fold reduction in neutralization capacity of beta and omicron compared to the original virus. Taken together, we show that human sera from BriLife(R) vaccinees overall maintain neutralizing antibody response against all tested variants. We suggest that BriLife(R) acquired mutations may prove advantageous against future SARS-CoV-2 VOCs.

3.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-451119

RESUMEN

rVSV-{Delta}G-SARS-CoV-2-S is a clinical stage (Phase 2) replication competent recombinant vaccine against SARS-CoV-2. Nonclinical safety, immunogenicity and efficacy studies were conducted in 4 animal species, using multiple dose levels (up to 108 PFU/animal) and various dosing regimens. There were no treatment related mortalities in any study, or any noticeable clinical signs. Compared to unvaccinated controls, hematology and biochemistry parameters were unremarkable and no adverse histopathological findings gave cause for safety concern in any of the studies. There was no viral shedding in urine, nor viral RNA detected in whole blood or serum samples 7 days post vaccination. The rVSV-{Delta}G-SARS-CoV-2-S vaccine immune response gave rise to neutralizing antibodies, cellular immune response, and increased lymphocytic cellularity in the spleen germinal centers and regional lymph node. No evidence for neurovirulence was found in C57BL/6 immune competent mice or in highly sensitive IFNAR KO mice. Vaccine virus replication and distribution in K18 hACE2 transgenic mice showed a gradual clearance from the vaccination site with no vaccine virus recovered from the lungs. The rVSV-{Delta}G-SARS-CoV-2-S vaccine was well tolerated locally and systemically and elicited an effective immunogenic response up to the highest dose tested, supporting further clinical development.

4.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-447687

RESUMEN

COVID-19 pandemic initiated a worldwide race toward the development of treatments and vaccines. Small animal models were the Syrian golden hamster and the K18-hACE2 mice infected with SARS-CoV-2 to display a disease state with some aspects of the human COVID-19. Group activity of animals in their home cage continuously monitored by the HCMS100 was used as a sensitive marker of disease, successfully detecting morbidity symptoms of SARS-CoV-2 infection in hamsters and in K18-hACE2 mice. COVID-19 convalescent hamsters re-challenged with SARS-CoV-2, exhibited minor reduction in group activity compared to naive hamsters. To evaluate rVSV-{Delta}G-spike vaccination efficacy against SARS-CoV-2, we used the HCMS100 to monitor group activity of hamsters in their home cage. Single-dose rVSV-{Delta}G-spike vaccination of immunized group showed a faster recovery compared to the non-immunized infected hamsters, substantiating the efficacy of rVSV-{Delta}G-spike vaccine. HCMS100 offers non-intrusive, hands-free monitoring of a number of home cages of hamsters or mice modeling COVID-19.

5.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-424878

RESUMEN

Replication-restricted modified vaccinia virus Ankara (MVA) is a licensed smallpox vaccine and numerous clinical studies investigating recombinant MVAs (rMVAs) as vectors for prevention of other infectious diseases have been completed or are in progress. Two rMVA COVID-19 vaccine trials are at an initial stage, though no animal protection studies have been reported. Here, we characterize rMVAs expressing the S protein of 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) 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 human cells and was recognized by anti-RBD antibody and by soluble hACE2 receptor. Intramuscular injection of mice with the rMVAs induced S-binding and pseudovirus-neutralizing antibodies. Boosting occurred following a second homologous rMVA but was higher with adjuvanted purified RBD protein. Weight loss and lethality following intranasal infection of transgenic hACE2 mice with CoV-2 was prevented by one or two immunizations with rMVAs or by passive transfer of serum from vaccinated mice. One or two rMVA vaccinations also prevented recovery of infectious CoV-2 from the lungs. A low amount of virus was detected in the nasal turbinates of only one of eight rMVA-vaccinated mice on day 2 and none later. Detection of subgenomic mRNA in turbinates on day 2 only indicated that replication was abortive in immunized animals. SignificanceVaccines are required to control COVID-19 during the pandemic and possibly afterwards. Recombinant nucleic acids, proteins and virus vectors that stimulate immune responses to the CoV-2 S protein have provided protection in experimental animal or human clinical trials, though questions remain regarding their ability to prevent spread and the duration of immunity. The present study focuses on replication-restricted modified vaccinia virus Ankara (MVA), which has been shown to be a safe, immunogenic and stable smallpox vaccine and a promising vaccine vector for other infectious diseases and cancer. In a transgenic mouse model, one or two injections of recombinant MVAs that express modified forms of S inhibited CoV-2 replication in the upper and lower respiratory tracts and prevented severe disease.

6.
- The COVID Moonshot Initiative; Hagit Achdout; Anthony Aimon; Elad Bar-David; Haim Barr; Amir Ben-Shmuel; James Bennett; Vitaliy A. Bilenko; Vitaliy A. Bilenko; Melissa L. Boby; Bruce Borden; Gregory R. Bowman; Juliane Brun; Sarma BVNBS; Mark Calmiano; Anna Carbery; Daniel Carney; Emma Cattermole; Edcon Chang; Eugene Chernyshenko; John D. Chodera; Austin Clyde; Joseph E. Coffland; Galit Cohen; Jason Cole; Alessandro Contini; Lisa Cox; Milan Cvitkovic; Alex Dias; Kim Donckers; David L. Dotson; Alice Douangamath; Shirly Duberstein; Tim Dudgeon; Louise Dunnett; Peter K. Eastman; Noam Erez; Charles J. Eyermann; Mike Fairhead; Gwen Fate; Daren Fearon; Oleg Fedorov; Matteo Ferla; Rafaela S. Fernandes; Lori Ferrins; Richard Foster; Holly Foster; Ronen Gabizon; Adolfo Garcia-Sastre; Victor O. Gawriljuk; Paul Gehrtz; Carina Gileadi; Charline Giroud; William G. Glass; Robert Glen; Itai Glinert; Andre S. Godoy; Marian Gorichko; Tyler Gorrie-Stone; Ed J. Griffen; Storm Hassell Hart; Jag Heer; Michael Henry; Michelle Hill; Sam Horrell; Victor D. Huliak; Matthew F.D. Hurley; Tomer Israely; Andrew Jajack; Jitske Jansen; Eric Jnoff; Dirk Jochmans; Tobias John; Steven De Jonghe; Anastassia L. Kantsadi; Peter W. Kenny; J. L. Kiappes; Serhii O. Kinakh; Lizbe Koekemoer; Boris Kovar; Tobias Krojer; Alpha Lee; Bruce A. Lefker; Haim Levy; Ivan G. Logvinenko; Nir London; Petra Lukacik; Hannah Bruce Macdonald; Beth MacLean; Tika R. Malla; Tatiana Matviiuk; Willam McCorkindale; Briana L. McGovern; Sharon Melamed; Kostiantyn P. Melnykov; Oleg Michurin; Halina Mikolajek; Bruce F. Milne; Aaron Morris; Garrett M. Morris; Melody Jane Morwitzer; Demetri Moustakas; Aline M. Nakamura; Jose Brandao Neto; Johan Neyts; Luong Nguyen; Gabriela D. Noske; Vladas Oleinikovas; Glaucius Oliva; Gijs J. Overheul; David Owen; Ruby Pai; Jin Pan; Nir Paran; Benjamin Perry; Maneesh Pingle; Jakir Pinjari; Boaz Politi; Ailsa Powell; Vladimir Psenak; Reut Puni; Victor L. Rangel; Rambabu N. Reddi; St Patrick Reid; Efrat Resnick; Emily Grace Ripka; Matthew C. Robinson; Ralph P. Robinson; Jaime Rodriguez-Guerra; Romel Rosales; Dominic Rufa; Kadi Saar; Kumar Singh Saikatendu; Chris Schofield; Mikhail Shafeev; Aarif Shaikh; Jiye Shi; Khriesto Shurrush; Sukrit Singh; Assa Sittner; Rachael Skyner; Adam Smalley; Bart Smeets; Mihaela D. Smilova; Leonardo J. Solmesky; John Spencer; Claire Strain-Damerell; Vishwanath Swamy; Hadas Tamir; Rachael Tennant; Warren Thompson; Andrew Thompson; Susana Tomasio; Igor S. Tsurupa; Anthony Tumber; Ioannis Vakonakis; Ronald P. van Rij; Laura Vangeel; Finny S. Varghese; Mariana Vaschetto; Einat B. Vitner; Vincent Voelz; Andrea Volkamer; Frank von Delft; Annette von Delft; Martin Walsh; Walter Ward; Charlie Weatherall; Shay Weiss; Kris M. White; Conor Francis Wild; Matthew Wittmann; Nathan Wright; Yfat Yahalom-Ronen; Daniel Zaidmann; Hadeer Zidane; Nicole Zitzmann.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-339317

RESUMEN

The COVID-19 pandemic is a stark reminder that a barren global antiviral pipeline has grave humanitarian consequences. Future pandemics could be prevented by accessible, easily deployable broad-spectrum oral antivirals and open knowledge bases that derisk and accelerate novel antiviral discovery and development. Here, we report the results of the COVID Moonshot, a fully open-science structure-enabled drug discovery campaign targeting the SARS-CoV-2 main protease. We discovered a novel chemical scaffold that is differentiated from current clinical candidates in terms of toxicity, resistance, and pharmacokinetics liabilities, and developed it into noncovalent orally-bioavailable nanomolar inhibitors with clinical potential. Our approach leveraged crowdsourcing, high-throughput structural biology, machine learning, and exascale molecular simulations. In the process, we generated a detailed map of the structural plasticity of the main protease, extensive structure-activity relationships for multiple chemotypes, and a wealth of biochemical activity data. In a first for a structure-based drug discovery campaign, all compound designs (>18,000 designs), crystallographic data (>500 ligand-bound X-ray structures), assay data (>10,000 measurements), and synthesized molecules (>2,400 compounds) for this campaign were shared rapidly and openly, creating a rich open and IP-free knowledgebase for future anti-coronavirus drug discovery.

7.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-160655

RESUMEN

The COVID-19 pandemic caused by SARS-CoV-2 that emerged in December 2019 in China resulted in over 7.8 million infections and over 430,000 deaths worldwide, imposing an urgent need for rapid development of an efficient and cost-effective vaccine, suitable for mass immunization. Here, we generated a replication competent recombinant VSV-{Delta}G-spike vaccine, in which the glycoprotein of VSV was replaced by the spike protein of the SARS-CoV-2. In vitro characterization of the recombinant VSV-{Delta}G-spike indicated expression and presentation of the spike protein on the viral membrane with antigenic similarity to SARS-CoV-2. A golden Syrian hamster in vivo model for COVID-19 was implemented. We show that vaccination of hamsters with recombinant VSV-{Delta}G-spike results in rapid and potent induction of neutralizing antibodies against SARS-CoV-2. Importantly, single-dose vaccination was able to protect hamsters against SARS-CoV-2 challenge, as demonstrated by the abrogation of body weight loss of the immunized hamsters compared to unvaccinated hamsters. Furthermore, whereas lungs of infected hamsters displayed extensive tissue damage and high viral titers, immunized hamsters lungs showed only minor lung pathology, and no viral load. Taken together, we suggest recombinant VSV-{Delta}G-spike as a safe, efficacious and protective vaccine against SARS-CoV-2 infection.

8.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-103283

RESUMEN

The need for antiviral drugs is real and relevant. Broad spectrum antiviral drugs have a particular advantage when dealing with rapid disease outbreaks, such as the current COVID-19 pandemic. Since viruses are completely dependent on internal cell mechanisms, they must cross cell membranes during their lifecycle, creating a dependence on processes involving membrane dynamics. Thus, in this study we examined whether the synthesis of glycosphingolipids, biologically active components of cell membranes, can serve as an antiviral therapeutic target. We examined the antiviral effect of two specific inhibitors of GlucosylCeramide synthase (GCS); (i) Genz-123346, an analogue of the FDA-approved drug Cerdelga(R), (ii) GENZ-667161, an analogue of venglustat which is currently under phase III clinical trials. We found that both GCS inhibitors inhibit the replication of four different enveloped RNA viruses of different genus, organ-target and transmission route: (i) Neuroinvasive Sindbis virus (SVNI), (ii) West Nile virus (WNV), (iii) Influenza A virus, and (iv) SARS-CoV-2. Moreover, GCS inhibitors significantly increase the survival rate of SVNI-infected mice. Our data suggest that GCS inhibitors can potentially serve as a broad-spectrum antiviral therapy and should be further examined in preclinical and clinical trial. Analogues of the specific compounds tested have already been studied clinically, implying they can be fast-tracked for public use. With the current COVID-19 pandemic, this may be particularly relevant to SARS-CoV-2 infection. One Sentence SummaryAn analogue of Cerdelga(R), an FDA-approved drug, is effective against a broad range of RNA-viruses including the newly emerging SARS-CoV-2.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...