Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.04.18.440302

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV-2) is responsible for the COVID-19 pandemic, causing health and economic problems. Currently, as dangerous mutations emerge there is an increased demand for specific treatments for SARS-CoV-2 infected patients. The spike glycoprotein on the virus membrane binds to the angiotensin converting enzyme 2 (ACE2) receptor on host cells through its receptor binding domain (RBD) to mediate virus entry. Thus, blocking this interaction may inhibit viral entry and consequently stop infection. Here, we generated fusion proteins composed of the extracellular portions of ACE2 and RBD fused to the Fc portion of human IgG1 (ACE2-Ig and RBD-Ig, respectively). We demonstrate that ACE2-Ig is enzymatically active and that it can be recognized by the SARS-CoV-2 RBD, independently of its enzymatic activity. We further show that RBD-Ig efficiently inhibits in vitro and in vivo SARS-CoV-2 infection, better than ACE2-Ig. Mechanistically we show that anti-spike antibodies generation, ACE2 enzymatic activity and ACE2 surface expression were not affected by RBD-Ig. Finally, we show that RBD-Ig is more efficient than ACE2-Ig at neutralizing high virus concentration infection. We thus propose that RBD-Ig physically blocks virus infection by binding to ACE2 and that RBD-Ig should be used for the treatment of SARS-CoV-2-infected patients.


Subject(s)
Severe Acute Respiratory Syndrome , Tumor Virus Infections , COVID-19
2.
researchsquare; 2020.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-136702.v1

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause for the ongoing COVID-19 pandemic1. The continued spread of SARS-CoV-2 along with the imminent flu season increase the probability of influenza-SARS-CoV-2 dual infection which might result in a severe disease. In this study, we examined the disease outcome of influenza A virus (IAV) and SARS-CoV-2 co-infection in K18-hACE2 mice. Our data indicates that IAV-infected mice are more susceptible to develop severe disease upon co-infection with SARS-CoV-2 two days post influenza infection. This co-infection results in severe morbidity and nearly uniform fatality as compared to the non-fatal influenza disease, or the partial fatality of SARS-CoV-2 alone. Co-infection was associated with elevated influenza viral load in respiratory organs. Remarkably, prior immunity to influenza, but not to SARS-CoV-2, prevented the severe disease and mortality. These data provide an experimental support that flu intervention by prior vaccination may be valuable in reducing the risk of sever Flu - SARS-CoV-2 comorbidity, and highlight the importance of vaccination.


Subject(s)
Coinfection , Severe Acute Respiratory Syndrome , COVID-19 , Influenza, Human
3.
- The COVID Moonshot Consortium; Hagit Achdout; Anthony Aimon; Elad Bar-David; Haim Barr; Amir Ben-Shmuel; James Bennett; Melissa L Bobby; Juliane Brun; Sarma BVNBS; Mark Calmiano; Anna Carbery; Emma Cattermole; John D. Chodera; Austin Clyde; Joseph E. Coffland; Galit Cohen; Jason Cole; Alessandro Contini; Lisa Cox; Milan Cvitkovic; Alex Dias; Alice Douangamath; Shirly Duberstein; Tim Dudgeon; Louise Dunnett; Peter K. Eastman; Noam Erez; Michael Fairhead; Daren Fearon; Oleg Fedorov; Matteo Ferla; Holly Foster; Richard Foster; Ronen Gabizon; Paul Gehrtz; Carina Gileadi; Charline Giroud; William G. Glass; Robert Glen; Itai Glinert; Marian Gorichko; Tyler Gorrie-Stone; Edward J Griffen; Jag Heer; Michelle Hill; Sam Horrell; Matthew F.D. Hurley; Tomer Israely; Andrew Jajack; Eric Jnoff; Tobias John; Anastassia L. Kantsadi; Peter W. Kenny; John L. Kiappes; Lizbe Koekemoer; Boris Kovar; Tobias Krojer; Alpha Albert Lee; Bruce A. Lefker; Haim Levy; Nir London; Petra Lukacik; Hannah Bruce Macdonald; Beth MacLean; Tika R. Malla; Tatiana Matviiuk; Willam McCorkindale; Sharon Melamed; Oleg Michurin; Halina Mikolajek; Aaron Morris; Garrett M. Morris; Melody Jane Morwitzer; Demetri Moustakas; Jose Brandao Neto; Vladas Oleinikovas; 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; Matthew C. Robinson; Ralph P. Robinson; Dominic Rufa; Christopher Schofield; Aarif Shaikh; Jiye Shi; Khriesto Shurrush; Assa Sittner; Rachael Skyner; Adam Smalley; Mihaela D. Smilova; John Spencer; Claire Strain-Damerell; Vishwanath Swamy; Hadas Tamir; Rachael Tennant; Andrew Thompson; Warren Thompson; Susana Tomasio; Anthony Tumber; Ioannis Vakonakis; Ronald P. van Rij; Finny S. Varghese; Mariana Vaschetto; Einat B. Vitner; Vincent Voelz; Annette von Delft; Frank von Delft; Martin Walsh; Walter Ward; Charlie Weatherall; Shay Weiss; Conor Francis Wild; Matthew Wittmann; Nathan Wright; Yfat Yahalom-Ronen; Daniel Zaidmann; Hadeer Zidane; Nicole Zitzmann.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.29.339317

ABSTRACT

Herein we provide a living summary of the data generated during the COVID Moonshot project focused on the development of SARS-CoV-2 main protease (Mpro) inhibitors. Our approach uniquely combines crowdsourced medicinal chemistry insights with high throughput crystallography, exascale computational chemistry infrastructure for simulations, and machine learning in triaging designs and predicting synthetic routes. This manuscript describes our methodologies leading to both covalent and non-covalent inhibitors displaying protease IC50 values under 150 nM and viral inhibition under 5 uM in multiple different viral replication assays. Furthermore, we provide over 200 crystal structures of fragment-like and lead-like molecules in complex with the main protease. Over 1000 synthesized and ordered compounds are also reported with the corresponding activity in Mpro enzymatic assays using two different experimental setups. The data referenced in this document will be continually updated to reflect the current experimental progress of the COVID Moonshot project, and serves as a citable reference for ensuing publications. All of the generated data is open to other researchers who may find it of use.

4.
Xun Chen; Matteo Gentili; Nir Hacohen; Aviv Regev; Haim Barr; Amir Ben-Shmuel; James Bennett; Melissa L Bobby; Juliane Brun; Sarma BVNBS; Mark Calmiano; Anna Carbery; Emma Cattermole; John D. Chodera; Austin Clyde; Joseph E. Coffland; Galit Cohen; Jason Cole; Alessandro Contini; Lisa Cox; Milan Cvitkovic; Alex Dias; Alice Douangamath; Shirly Duberstein; Tim Dudgeon; Louise Dunnett; Peter K. Eastman; Noam Erez; Michael Fairhead; Daren Fearon; Oleg Fedorov; Matteo Ferla; Holly Foster; Richard Foster; Ronen Gabizon; Paul Gehrtz; Carina Gileadi; Charline Giroud; William G. Glass; Robert Glen; Itai Glinert; Marian Gorichko; Tyler Gorrie-Stone; Edward J Griffen; Jag Heer; Michelle Hill; Sam Horrell; Matthew F.D. Hurley; Tomer Israely; Andrew Jajack; Eric Jnoff; Tobias John; Anastassia L. Kantsadi; Peter W. Kenny; John L. Kiappes; Lizbe Koekemoer; Boris Kovar; Tobias Krojer; Alpha Albert Lee; Bruce A. Lefker; Haim Levy; Nir London; Petra Lukacik; Hannah Bruce Macdonald; Beth MacLean; Tika R. Malla; Tatiana Matviiuk; Willam McCorkindale; Sharon Melamed; Oleg Michurin; Halina Mikolajek; Aaron Morris; Garrett M. Morris; Melody Jane Morwitzer; Demetri Moustakas; Jose Brandao Neto; Vladas Oleinikovas; 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; Matthew C. Robinson; Ralph P. Robinson; Dominic Rufa; Christopher Schofield; Aarif Shaikh; Jiye Shi; Khriesto Shurrush; Assa Sittner; Rachael Skyner; Adam Smalley; Mihaela D. Smilova; John Spencer; Claire Strain-Damerell; Vishwanath Swamy; Hadas Tamir; Rachael Tennant; Andrew Thompson; Warren Thompson; Susana Tomasio; Anthony Tumber; Ioannis Vakonakis; Ronald P. van Rij; Finny S. Varghese; Mariana Vaschetto; Einat B. Vitner; Vincent Voelz; Annette von Delft; Frank von Delft; Martin Walsh; Walter Ward; Charlie Weatherall; Shay Weiss; Conor Francis Wild; Matthew Wittmann; Nathan Wright; Yfat Yahalom-Ronen; Daniel Zaidmann; Hadeer Zidane; Nicole Zitzmann.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.29.361287

ABSTRACT

Antibody engineering technologies face increasing demands for speed, reliability and scale. We developed CeVICA, a cell-free antibody engineering platform that integrates a novel generation method and design for camelid heavy-chain antibody VHH domain-based synthetic libraries, optimized in vitro selection based on ribosome display and a computational pipeline for binder prediction based on CDR-directed clustering. We applied CeVICA to engineer antibodies against the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike proteins and identified >800 predicted binder families. Among 14 experimentally-tested binders, 6 showed inhibition of pseudotyped virus infection. Antibody affinity maturation further increased binding affinity and potency of inhibition. Additionally, the unique capability of CeVICA for efficient and comprehensive binder prediction allowed retrospective validation of the fitness of our synthetic VHH library design and revealed direction for future refinement. CeVICA offers an integrated solution to rapid generation of divergent synthetic antibodies with tunable affinities in vitro and may serve as the basis for automated and highly parallel antibody generation.


Subject(s)
Severe Acute Respiratory Syndrome , Tumor Virus Infections
5.
Saumyabrata Mazumder; Ruchir Rastogi; Avinash Undale; Kajal Arora; Nupur Mehrotra Arora; Biswa Pratim Das Purkayastha; Dilip Kumar; Abyson Joseph; Bhupesh Mali; Vidya Bhushan Arya; Sriganesh Kalyanaraman; Abhishek Mukherjee; Aditi Gupta; Swaroop Potdar; Sourav Singha Roy; Deepak Parashar; Jeny Paliwal; Sudhir Kumar Singh; Aelia Naqvi; Apoorva Srivastava; Manglesh Kumar Singh; Devanand Kumar; Sarthi Bansal; Satabdi Rautray; Indrajeet Singh; Pankaj Fengade; Bivekanand Kumar; Manish Saini; Kshipra Jain; Reeshu Gupta; Prabuddha K Kundu; Matteo Ferla; Holly Foster; Richard Foster; Ronen Gabizon; Paul Gehrtz; Carina Gileadi; Charline Giroud; William G. Glass; Robert Glen; Itai Glinert; Marian Gorichko; Tyler Gorrie-Stone; Edward J Griffen; Jag Heer; Michelle Hill; Sam Horrell; Matthew F.D. Hurley; Tomer Israely; Andrew Jajack; Eric Jnoff; Tobias John; Anastassia L. Kantsadi; Peter W. Kenny; John L. Kiappes; Lizbe Koekemoer; Boris Kovar; Tobias Krojer; Alpha Albert Lee; Bruce A. Lefker; Haim Levy; Nir London; Petra Lukacik; Hannah Bruce Macdonald; Beth MacLean; Tika R. Malla; Tatiana Matviiuk; Willam McCorkindale; Sharon Melamed; Oleg Michurin; Halina Mikolajek; Aaron Morris; Garrett M. Morris; Melody Jane Morwitzer; Demetri Moustakas; Jose Brandao Neto; Vladas Oleinikovas; 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; Matthew C. Robinson; Ralph P. Robinson; Dominic Rufa; Christopher Schofield; Aarif Shaikh; Jiye Shi; Khriesto Shurrush; Assa Sittner; Rachael Skyner; Adam Smalley; Mihaela D. Smilova; John Spencer; Claire Strain-Damerell; Vishwanath Swamy; Hadas Tamir; Rachael Tennant; Andrew Thompson; Warren Thompson; Susana Tomasio; Anthony Tumber; Ioannis Vakonakis; Ronald P. van Rij; Finny S. Varghese; Mariana Vaschetto; Einat B. Vitner; Vincent Voelz; Annette von Delft; Frank von Delft; Martin Walsh; Walter Ward; Charlie Weatherall; Shay Weiss; Conor Francis Wild; Matthew Wittmann; Nathan Wright; Yfat Yahalom-Ronen; Daniel Zaidmann; Hadeer Zidane; Nicole Zitzmann.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.30.360115

ABSTRACT

The rapid development of safe and effective vaccines against SARS CoV-2 is the need of the hour for the coronavirus outbreak. Here, we have developed PRAK-03202, the world's first triple antigen VLP vaccine candidate in a highly characterized S. cerevisiae-based D-Crypt platform, which induced SARS CoV-2 specific neutralizing antibodies in BALB/c mice. Immunizations using three different doses of PRAK-03202 induces antigen specific (Spike, envelope and membrane proteins) humoral response and neutralizing potential. PBMCs from convalescent patients, when exposed to PRAK-03202, showed lymphocyte proliferation and elevated IFN-{gamma} levels suggestive of conservation of epitopes and induction of T helper 1 (Th1)-biased cellular immune responses. These data support the clinical development and testing of PRAK-03202 for use in humans.

6.
Kathryn Kistler; Trevor Bedford; Avinash Undale; Kajal Arora; Nupur Mehrotra Arora; Biswa Pratim Das Purkayastha; Dilip Kumar; Abyson Joseph; Bhupesh Mali; Vidya Bhushan Arya; Sriganesh Kalyanaraman; Abhishek Mukherjee; Aditi Gupta; Swaroop Potdar; Sourav Singha Roy; Deepak Parashar; Jeny Paliwal; Sudhir Kumar Singh; Aelia Naqvi; Apoorva Srivastava; Manglesh Kumar Singh; Devanand Kumar; Sarthi Bansal; Satabdi Rautray; Indrajeet Singh; Pankaj Fengade; Bivekanand Kumar; Manish Saini; Kshipra Jain; Reeshu Gupta; Prabuddha K Kundu; Matteo Ferla; Holly Foster; Richard Foster; Ronen Gabizon; Paul Gehrtz; Carina Gileadi; Charline Giroud; William G. Glass; Robert Glen; Itai Glinert; Marian Gorichko; Tyler Gorrie-Stone; Edward J Griffen; Jag Heer; Michelle Hill; Sam Horrell; Matthew F.D. Hurley; Tomer Israely; Andrew Jajack; Eric Jnoff; Tobias John; Anastassia L. Kantsadi; Peter W. Kenny; John L. Kiappes; Lizbe Koekemoer; Boris Kovar; Tobias Krojer; Alpha Albert Lee; Bruce A. Lefker; Haim Levy; Nir London; Petra Lukacik; Hannah Bruce Macdonald; Beth MacLean; Tika R. Malla; Tatiana Matviiuk; Willam McCorkindale; Sharon Melamed; Oleg Michurin; Halina Mikolajek; Aaron Morris; Garrett M. Morris; Melody Jane Morwitzer; Demetri Moustakas; Jose Brandao Neto; Vladas Oleinikovas; 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; Matthew C. Robinson; Ralph P. Robinson; Dominic Rufa; Christopher Schofield; Aarif Shaikh; Jiye Shi; Khriesto Shurrush; Assa Sittner; Rachael Skyner; Adam Smalley; Mihaela D. Smilova; John Spencer; Claire Strain-Damerell; Vishwanath Swamy; Hadas Tamir; Rachael Tennant; Andrew Thompson; Warren Thompson; Susana Tomasio; Anthony Tumber; Ioannis Vakonakis; Ronald P. van Rij; Finny S. Varghese; Mariana Vaschetto; Einat B. Vitner; Vincent Voelz; Annette von Delft; Frank von Delft; Martin Walsh; Walter Ward; Charlie Weatherall; Shay Weiss; Conor Francis Wild; Matthew Wittmann; Nathan Wright; Yfat Yahalom-Ronen; Daniel Zaidmann; Hadeer Zidane; Nicole Zitzmann.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.30.352914

ABSTRACT

Seasonal coronaviruses (OC43, 229E, NL63 and HKU1) are endemic to the human population, regularly infecting and reinfecting humans while typically causing asymptomatic to mild respiratory infections. It is not known to what extent reinfection by these viruses is due to waning immune memory or antigenic drift of the viruses. Here, we address the influence of antigenic drift on immune evasion of seasonal coronaviruses. We provide evidence that at least two of these viruses, OC43 and 229E, are undergoing adaptive evolution in regions of the viral spike protein that are exposed to human humoral immunity. This suggests that reinfection may be due, in part, to positively-selected genetic changes in these viruses that enable them to escape recognition by the immune system. It is possible that, as with seasonal influenza, these adaptive changes in antigenic regions of the virus would necessitate continual reformulation of a vaccine made against them.


Subject(s)
Respiratory Tract Infections
7.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.26.354811

ABSTRACT

Coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), exhibits high levels of mortality and morbidity and has dramatic consequences on human life, sociality and global economy. Neutralizing antibodies constitute a highly promising approach for treating and preventing infection by this novel pathogen. In the present study, we characterized and further evaluated the recently identified human monoclonal MD65 antibody for its ability to provide protection against a lethal SARS-CoV-2 infection of K18-hACE2 transgenic mice. Eighty percent of the untreated mice succumbed 6-9 days post-infection while administration of the MD65 antibody as late as 3 days after exposure, rescued all infected animals. In addition, the efficiency of the treatment is supported by prevention of morbidity and ablation of the load of infective virions in the lungs of treated animals. The data unprecedentedly demonstrate, the therapeutic value of human monoclonal antibodies as a life-saving treatment of severe COVID-19 infection.


Subject(s)
COVID-19
8.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.06.18.160655

ABSTRACT

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.


Subject(s)
COVID-19
9.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.05.18.103283

ABSTRACT

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.


Subject(s)
COVID-19 , Infections , Gaucher Disease
SELECTION OF CITATIONS
SEARCH DETAIL