Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
Plus de filtres










Base de données
Gamme d'année
2.
Sci Rep ; 13(1): 13912, 2023 08 25.
Article de Anglais | MEDLINE | ID: mdl-37626085

RÉSUMÉ

The development of new therapies against SARS-CoV-2 is required to extend the toolkit of intervention strategies to combat the global pandemic. In this study, hyperimmune plasma from sheep immunised with whole spike SARS-CoV-2 recombinant protein has been used to generate candidate products. In addition to purified IgG, we have refined candidate therapies by removing non-specific IgG via affinity binding along with fragmentation to eliminate the Fc region to create F(ab')2 fragments. These preparations were evaluated for in vitro activity and demonstrated to be strongly neutralising against a range of SARS-CoV-2 strains, including Omicron B2.2. In addition, their protection against disease manifestations and viral loads were assessed using a hamster SARS-CoV-2 infection model. Results demonstrated protective effects of both IgG and F(ab')2, with the latter requiring sequential dosing to maintain in vivo activity due to rapid clearance from the circulation.


Sujet(s)
COVID-19 , SARS-CoV-2 , Cricetinae , Animaux , Ovis , Immunisation passive , Cinétique , Immunoglobuline G
3.
J Gen Virol ; 104(7)2023 07.
Article de Anglais | MEDLINE | ID: mdl-37432877

RÉSUMÉ

The 2',5'- oligoadenylate synthetase (OAS) - ribonuclease L (RNAseL) - phosphodiesterase 12 (PDE12) pathway is an essential interferon-induced effector mechanism against RNA virus infection. Inhibition of PDE12 leads to selective amplification of RNAseL activity in infected cells. We aimed to investigate PDE12 as a potential pan-RNA virus antiviral drug target and develop PDE12 inhibitors that elicit antiviral activity against a range of viruses. A library of 18 000 small molecules was screened for PDE12 inhibitor activity using a fluorescent probe specific for PDE12. The lead compounds (CO-17 or CO-63) were tested in cell-based antiviral assays using encephalomyocarditis virus (EMCV), hepatitis C virus (HCV), dengue virus (DENV), West Nile virus (WNV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in vitro. Cross reactivity of PDE12 inhibitors with other PDEs and in vivo toxicity were measured. In EMCV assays, CO-17 potentiated the effect of IFNα by 3 log10. The compounds were selective for PDE12 when tested against a panel of other PDEs and non-toxic at up to 42 mg kg-1 in rats in vivo. Thus, we have identified PDE12 inhibitors (CO-17 and CO-63), and established the principle that inhibitors of PDE12 have antiviral properties. Early studies suggest these PDE12 inhibitors are well tolerated at the therapeutic range, and reduce viral load in studies of DENV, HCV, WNV and SARS-CoV-2 in human cells and WNV in a mouse model.


Sujet(s)
COVID-19 , Virus à ARN , Humains , Souris , Animaux , Rats , Antiviraux/pharmacologie , SARS-CoV-2 , Interféron alpha , Virus de l'encéphalomyocardite , Phosphodiesterases
5.
Genome Biol ; 24(1): 47, 2023 03 13.
Article de Anglais | MEDLINE | ID: mdl-36915185

RÉSUMÉ

BACKGROUND: The mutational landscape of SARS-CoV-2 varies at the dominant viral genome sequence and minor genomic variant population. During the COVID-19 pandemic, an early substitution in the genome was the D614G change in the spike protein, associated with an increase in transmissibility. Genomes with D614G are accompanied by a P323L substitution in the viral polymerase (NSP12). However, P323L is not thought to be under strong selective pressure. RESULTS: Investigation of P323L/D614G substitutions in the population shows rapid emergence during the containment phase and early surge phase during the first wave. These substitutions emerge from minor genomic variants which become dominant viral genome sequence. This is investigated in vivo and in vitro using SARS-CoV-2 with P323 and D614 in the dominant genome sequence and L323 and G614 in the minor variant population. During infection, there is rapid selection of L323 into the dominant viral genome sequence but not G614. Reverse genetics is used to create two viruses (either P323 or L323) with the same genetic background. L323 shows greater abundance of viral RNA and proteins and a smaller plaque morphology than P323. CONCLUSIONS: These data suggest that P323L is an important contribution in the emergence of variants with transmission advantages. Sequence analysis of viral populations suggests it may be possible to predict the emergence of a new variant based on tracking the frequency of minor variant genomes. The ability to predict an emerging variant of SARS-CoV-2 in the global landscape may aid in the evaluation of medical countermeasures and non-pharmaceutical interventions.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/génétique , Pandémies , Contexte génétique , Génome viral , Mutation
6.
ACS Cent Sci ; 8(5): 527-545, 2022 May 25.
Article de Anglais | MEDLINE | ID: mdl-35647275

RÉSUMÉ

Heparan sulfate (HS) is a cell surface polysaccharide recently identified as a coreceptor with the ACE2 protein for the S1 spike protein on SARS-CoV-2 virus, providing a tractable new therapeutic target. Clinically used heparins demonstrate an inhibitory activity but have an anticoagulant activity and are supply-limited, necessitating alternative solutions. Here, we show that synthetic HS mimetic pixatimod (PG545), a cancer drug candidate, binds and destabilizes the SARS-CoV-2 spike protein receptor binding domain and directly inhibits its binding to ACE2, consistent with molecular modeling identification of multiple molecular contacts and overlapping pixatimod and ACE2 binding sites. Assays with multiple clinical isolates of SARS-CoV-2 virus show that pixatimod potently inhibits the infection of monkey Vero E6 cells and physiologically relevant human bronchial epithelial cells at safe therapeutic concentrations. Pixatimod also retained broad potency against variants of concern (VOC) including B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta), and B.1.1.529 (Omicron). Furthermore, in a K18-hACE2 mouse model, pixatimod significantly reduced SARS-CoV-2 viral titers in the upper respiratory tract and virus-induced weight loss. This demonstration of potent anti-SARS-CoV-2 activity tolerant to emerging mutations establishes proof-of-concept for targeting the HS-Spike protein-ACE2 axis with synthetic HS mimetics and provides a strong rationale for clinical investigation of pixatimod as a potential multimodal therapeutic for COVID-19.

7.
Theranostics ; 12(1): 1-17, 2022.
Article de Anglais | MEDLINE | ID: mdl-34987630

RÉSUMÉ

Background: Administration of potent anti-receptor-binding domain (RBD) monoclonal antibodies has been shown to curtail viral shedding and reduce hospitalization in patients with SARS-CoV-2 infection. However, the structure-function analysis of potent human anti-RBD monoclonal antibodies and its links to the formulation of antibody cocktails remains largely elusive. Methods: Previously, we isolated a panel of neutralizing anti-RBD monoclonal antibodies from convalescent patients and showed their neutralization efficacy in vitro. Here, we elucidate the mechanism of action of antibodies and dissect antibodies at the epitope level, which leads to a formation of a potent antibody cocktail. Results: We found that representative antibodies which target non-overlapping epitopes are effective against wild type virus and recently emerging variants of concern, whilst being encoded by antibody genes with few somatic mutations. Neutralization is associated with the inhibition of binding of viral RBD to ACE2 and possibly of the subsequent fusion process. Structural analysis of representative antibodies, by cryo-electron microscopy and crystallography, reveals that they have some unique aspects that are of potential value while sharing some features in common with previously reported neutralizing monoclonal antibodies. For instance, one has a common VH 3-53 public variable region yet is unusually resilient to mutation at residue 501 of the RBD. We evaluate the in vivo efficacy of an antibody cocktail consisting of two potent non-competing anti-RBD antibodies in a Syrian hamster model. We demonstrate that the cocktail prevents weight loss, reduces lung viral load and attenuates pulmonary inflammation in hamsters in both prophylactic and therapeutic settings. Although neutralization of one of these antibodies is abrogated by the mutations of variant B.1.351, it is also possible to produce a bi-valent cocktail of antibodies both of which are resilient to variants B.1.1.7, B.1.351 and B.1.617.2. Conclusions: These findings support the up-to-date and rational design of an anti-RBD antibody cocktail as a therapeutic candidate against COVID-19.


Sujet(s)
Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/pharmacologie , Traitements médicamenteux de la COVID-19 , SARS-CoV-2/métabolisme , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , Animaux , Anticorps monoclonaux/métabolisme , Anticorps neutralisants/composition chimique , Anticorps neutralisants/immunologie , Anticorps neutralisants/pharmacologie , Sites de fixation , Fixation compétitive , COVID-19/virologie , Cricetinae , Cryomicroscopie électronique , Cristallographie aux rayons X , Chiens , Épitopes , Femelle , Humains , Cellules rénales canines Madin-Darby , Tests de neutralisation , Domaines protéiques , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/pathogénicité , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/métabolisme
8.
Nat Commun ; 12(1): 5469, 2021 09 22.
Article de Anglais | MEDLINE | ID: mdl-34552091

RÉSUMÉ

SARS-CoV-2 remains a global threat to human health particularly as escape mutants emerge. There is an unmet need for effective treatments against COVID-19 for which neutralizing single domain antibodies (nanobodies) have significant potential. Their small size and stability mean that nanobodies are compatible with respiratory administration. We report four nanobodies (C5, H3, C1, F2) engineered as homotrimers with pmolar affinity for the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. Crystal structures show C5 and H3 overlap the ACE2 epitope, whilst C1 and F2 bind to a different epitope. Cryo Electron Microscopy shows C5 binding results in an all down arrangement of the Spike protein. C1, H3 and C5 all neutralize the Victoria strain, and the highly transmissible Alpha (B.1.1.7 first identified in Kent, UK) strain and C1 also neutralizes the Beta (B.1.35, first identified in South Africa). Administration of C5-trimer via the respiratory route showed potent therapeutic efficacy in the Syrian hamster model of COVID-19 and separately, effective prophylaxis. The molecule was similarly potent by intraperitoneal injection.


Sujet(s)
Anticorps neutralisants/pharmacologie , Traitements médicamenteux de la COVID-19 , Anticorps à domaine unique/pharmacologie , Glycoprotéine de spicule des coronavirus/métabolisme , Administration par voie nasale , Animaux , Anticorps neutralisants/administration et posologie , Anticorps neutralisants/génétique , Anticorps neutralisants/immunologie , Cryomicroscopie électronique , Cristallographie aux rayons X , Modèles animaux de maladie humaine , Relation dose-réponse (immunologie) , Épitopes/composition chimique , Épitopes/métabolisme , Femelle , Mâle , Mesocricetus , Tests de neutralisation , SARS-CoV-2/effets des médicaments et des substances chimiques , Anticorps à domaine unique/administration et posologie , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/métabolisme , Glycoprotéine de spicule des coronavirus/composition chimique
10.
Nat Commun ; 12(1): 1260, 2021 02 24.
Article de Anglais | MEDLINE | ID: mdl-33627662

RÉSUMÉ

A novel coronavirus, SARS-CoV-2, has been identified as the causative agent of the current COVID-19 pandemic. Animal models, and in particular non-human primates, are essential to understand the pathogenesis of emerging diseases and to assess the safety and efficacy of novel vaccines and therapeutics. Here, we show that SARS-CoV-2 replicates in the upper and lower respiratory tract and causes pulmonary lesions in both rhesus and cynomolgus macaques. Immune responses against SARS-CoV-2 are also similar in both species and equivalent to those reported in milder infections and convalescent human patients. This finding is reiterated by our transcriptional analysis of respiratory samples revealing the global response to infection. We describe a new method for lung histopathology scoring that will provide a metric to enable clearer decision making for this key endpoint. In contrast to prior publications, in which rhesus are accepted to be the preferred study species, we provide convincing evidence that both macaque species authentically represent mild to moderate forms of COVID-19 observed in the majority of the human population and both species should be used to evaluate the safety and efficacy of interventions against SARS-CoV-2. Importantly, accessing cynomolgus macaques will greatly alleviate the pressures on current rhesus stocks.


Sujet(s)
COVID-19/immunologie , COVID-19/virologie , Poumon/anatomopathologie , Poumon/virologie , Animaux , Modèles animaux de maladie humaine , Femelle , Immunité cellulaire/physiologie , Interféron gamma/métabolisme , Macaca fascicularis , Macaca mulatta , Mâle , Pandémies , SARS-CoV-2/immunologie , SARS-CoV-2/pathogénicité
11.
Nat Commun ; 12(1): 542, 2021 01 22.
Article de Anglais | MEDLINE | ID: mdl-33483491

RÉSUMÉ

There is need for effective and affordable vaccines against SARS-CoV-2 to tackle the ongoing pandemic. In this study, we describe a protein nanoparticle vaccine against SARS-CoV-2. The vaccine is based on the display of coronavirus spike glycoprotein receptor-binding domain (RBD) on a synthetic virus-like particle (VLP) platform, SpyCatcher003-mi3, using SpyTag/SpyCatcher technology. Low doses of RBD-SpyVLP in a prime-boost regimen induce a strong neutralising antibody response in mice and pigs that is superior to convalescent human sera. We evaluate antibody quality using ACE2 blocking and neutralisation of cell infection by pseudovirus or wild-type SARS-CoV-2. Using competition assays with a monoclonal antibody panel, we show that RBD-SpyVLP induces a polyclonal antibody response that recognises key epitopes on the RBD, reducing the likelihood of selecting neutralisation-escape mutants. Moreover, RBD-SpyVLP is thermostable and can be lyophilised without losing immunogenicity, to facilitate global distribution and reduce cold-chain dependence. The data suggests that RBD-SpyVLP provides strong potential to address clinical and logistic challenges of the COVID-19 pandemic.


Sujet(s)
Anticorps antiviraux/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Peptides/immunologie , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Angiotensin-converting enzyme 2/immunologie , Animaux , Anticorps bloquants/immunologie , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , COVID-19/immunologie , Lignée cellulaire , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Motifs et domaines d'intéraction protéique , Multimérisation de protéines , Suidae
12.
Nat Commun ; 12(1): 81, 2021 01 04.
Article de Anglais | MEDLINE | ID: mdl-33398055

RÉSUMÉ

There is a vital need for authentic COVID-19 animal models to enable the pre-clinical evaluation of candidate vaccines and therapeutics. Here we report a dose titration study of SARS-CoV-2 in the ferret model. After a high (5 × 106 pfu) and medium (5 × 104 pfu) dose of virus is delivered, intranasally, viral RNA shedding in the upper respiratory tract (URT) is observed in 6/6 animals, however, only 1/6 ferrets show similar signs after low dose (5 × 102 pfu) challenge. Following sequential culls pathological signs of mild multifocal bronchopneumonia in approximately 5-15% of the lung is seen on day 3, in high and medium dosed groups. Ferrets re-challenged, after virus shedding ceased, are fully protected from acute lung pathology. The endpoints of URT viral RNA replication & distinct lung pathology are observed most consistently in the high dose group. This ferret model of SARS-CoV-2 infection presents a mild clinical disease.


Sujet(s)
COVID-19/immunologie , Modèles animaux de maladie humaine , Furets/immunologie , SARS-CoV-2/immunologie , Animaux , Anticorps antiviraux/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/pharmacologie , Relation dose-effet des médicaments , Femelle , Poumon/immunologie , Poumon/anatomopathologie , ARN viral/isolement et purification , SARS-CoV-2/génétique , Réplication virale/effets des médicaments et des substances chimiques , Réplication virale/immunologie , Excrétion virale/effets des médicaments et des substances chimiques , Excrétion virale/immunologie
13.
Br J Pharmacol ; 178(3): 626-635, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33125711

RÉSUMÉ

BACKGROUND AND PURPOSE: Currently, there are no licensed vaccines and limited antivirals for the treatment of COVID-19. Heparin (delivered systemically) is currently used to treat anticoagulant anomalies in COVID-19 patients. Additionally, in the United Kingdom, Brazil and Australia, nebulised unfractionated heparin (UFH) is being trialled in COVID-19 patients as a potential treatment. A systematic comparison of the potential antiviral effect of various heparin preparations on live wild type SARS-CoV-2, in vitro, is needed. EXPERIMENTAL APPROACH: Seven different heparin preparations including UFH and low MW heparins (LMWH) of porcine or bovine origin were screened for antiviral activity against live SARS-CoV-2 (Australia/VIC01/2020) using a plaque inhibition assay with Vero E6 cells. Interaction of heparin with spike protein RBD was studied using differential scanning fluorimetry and the inhibition of RBD binding to human ACE2 protein using elisa assays was examined. KEY RESULTS: All the UFH preparations had potent antiviral effects, with IC50 values ranging between 25 and 41 µg·ml-1 , whereas LMWHs were less inhibitory by ~150-fold (IC50 range 3.4-7.8 mg·ml-1 ). Mechanistically, we observed that heparin binds and destabilizes the RBD protein and furthermore, we show heparin directly inhibits the binding of RBD to the human ACE2 protein receptor. CONCLUSION AND IMPLICATIONS: This comparison of clinically relevant heparins shows that UFH has significantly stronger SARS-CoV-2 antiviral activity compared to LMWHs. UFH acts to directly inhibit binding of spike protein to the human ACE2 protein receptor. Overall, the data strongly support further clinical investigation of UFH as a potential treatment for patients with COVID-19.


Sujet(s)
Héparine/pharmacologie , SARS-CoV-2/croissance et développement , Angiotensin-converting enzyme 2/métabolisme , Animaux , Antiviraux/pharmacologie , Chlorocebus aethiops , Héparine/métabolisme , Héparine/usage thérapeutique , Héparine bas poids moléculaire/pharmacologie , Liaison aux protéines/effets des médicaments et des substances chimiques , Glycoprotéine de spicule des coronavirus/métabolisme , Méthode des plages virales , Traitements médicamenteux de la COVID-19
16.
Nat Struct Mol Biol ; 27(10): 950-958, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32737466

RÉSUMÉ

The COVID-19 pandemic has had an unprecedented health and economic impact and there are currently no approved therapies. We have isolated an antibody, EY6A, from an individual convalescing from COVID-19 and have shown that it neutralizes SARS-CoV-2 and cross-reacts with SARS-CoV-1. EY6A Fab binds the receptor binding domain (RBD) of the viral spike glycoprotein tightly (KD of 2 nM), and a 2.6-Å-resolution crystal structure of an RBD-EY6A Fab complex identifies the highly conserved epitope, away from the ACE2 receptor binding site. Residues within this footprint are key to stabilizing the pre-fusion spike. Cryo-EM analyses of the pre-fusion spike incubated with EY6A Fab reveal a complex of the intact spike trimer with three Fabs bound and two further multimeric forms comprising the destabilized spike attached to Fab. EY6A binds what is probably a major neutralizing epitope, making it a candidate therapeutic for COVID-19.


Sujet(s)
Anticorps antiviraux/composition chimique , Betacoronavirus/composition chimique , Infections à coronavirus/immunologie , Pneumopathie virale/immunologie , Glycoprotéine de spicule des coronavirus/composition chimique , Adulte , Angiotensin-converting enzyme 2 , Animaux , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/métabolisme , Betacoronavirus/immunologie , Betacoronavirus/métabolisme , Sites de fixation , COVID-19 , Chlorocebus aethiops , Réactions croisées , Cryomicroscopie électronique , Cristallographie aux rayons X , Épitopes , Humains , Fragments Fab d'immunoglobuline/composition chimique , Fragments Fab d'immunoglobuline/métabolisme , Mâle , Pandémies , Peptidyl-Dipeptidase A/métabolisme , Conformation des protéines , Domaines protéiques , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/métabolisme , Cellules Vero
17.
Nat Commun ; 11(1): 4198, 2020 08 21.
Article de Anglais | MEDLINE | ID: mdl-32826914

RÉSUMÉ

COVID-19 caused by SARS-CoV-2 has become a global pandemic requiring the development of interventions for the prevention or treatment to curtail mortality and morbidity. No vaccine to boost mucosal immunity, or as a therapeutic, has yet been developed to SARS-CoV-2. In this study, we discover and characterize a cross-reactive human IgA monoclonal antibody, MAb362. MAb362 binds to both SARS-CoV and SARS-CoV-2 spike proteins and competitively blocks ACE2 receptor binding, by overlapping the ACE2 structural binding epitope. Furthermore, MAb362 IgA neutralizes both pseudotyped SARS-CoV and SARS-CoV-2 in 293 cells expressing ACE2. When converted to secretory IgA, MAb326 also neutralizes authentic SARS-CoV-2 virus while the IgG isotype shows no neutralization. Our results suggest that SARS-CoV-2 specific IgA antibodies, such as MAb362, may provide effective immunity against SARS-CoV-2 by inducing mucosal immunity within the respiratory system, a potentially critical feature of an effective vaccine.


Sujet(s)
Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Betacoronavirus/immunologie , Immunoglobuline A/immunologie , Peptidyl-Dipeptidase A/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Angiotensin-converting enzyme 2 , Animaux , Anticorps monoclonaux/métabolisme , Anticorps neutralisants/métabolisme , Chlorocebus aethiops , Réactions croisées , Épitopes , Cellules HEK293 , Humains , Immunoglobuline A/métabolisme , Immunoglobuline A sécrétoire/immunologie , Immunoglobuline A sécrétoire/métabolisme , Immunoglobuline G/immunologie , Immunoglobuline G/métabolisme , Modèles moléculaires , Mutation , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Virus du SRAS/immunologie , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Cellules Vero
18.
Nat Struct Mol Biol ; 27(9): 846-854, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32661423

RÉSUMÉ

The SARS-CoV-2 virus is more transmissible than previous coronaviruses and causes a more serious illness than influenza. The SARS-CoV-2 receptor binding domain (RBD) of the spike protein binds to the human angiotensin-converting enzyme 2 (ACE2) receptor as a prelude to viral entry into the cell. Using a naive llama single-domain antibody library and PCR-based maturation, we have produced two closely related nanobodies, H11-D4 and H11-H4, that bind RBD (KD of 39 and 12 nM, respectively) and block its interaction with ACE2. Single-particle cryo-EM revealed that both nanobodies bind to all three RBDs in the spike trimer. Crystal structures of each nanobody-RBD complex revealed how both nanobodies recognize the same epitope, which partly overlaps with the ACE2 binding surface, explaining the blocking of the RBD-ACE2 interaction. Nanobody-Fc fusions showed neutralizing activity against SARS-CoV-2 (4-6 nM for H11-H4, 18 nM for H11-D4) and additive neutralization with the SARS-CoV-1/2 antibody CR3022.


Sujet(s)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Betacoronavirus/immunologie , Infections à coronavirus , Pandémies , Peptidyl-Dipeptidase A/métabolisme , Pneumopathie virale , Récepteurs viraux/métabolisme , Anticorps à domaine unique/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Séquence d'acides aminés , Angiotensin-converting enzyme 2 , Anticorps neutralisants/métabolisme , Anticorps neutralisants/ultrastructure , Anticorps antiviraux/métabolisme , Anticorps antiviraux/ultrastructure , Affinité des anticorps , Réaction antigène-anticorps/immunologie , Betacoronavirus/métabolisme , Fixation compétitive , COVID-19 , Cryomicroscopie électronique , Cristallographie aux rayons X , Épitopes/immunologie , Humains , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/immunologie , Modèles moléculaires , Banque de peptides , Peptidyl-Dipeptidase A/ultrastructure , Liaison aux protéines , Conformation des protéines , Récepteurs viraux/ultrastructure , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/métabolisme , SARS-CoV-2 , Similitude de séquences d'acides aminés , Anticorps à domaine unique/métabolisme , Anticorps à domaine unique/ultrastructure , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/ultrastructure
19.
Cell Host Microbe ; 28(3): 445-454.e6, 2020 09 09.
Article de Anglais | MEDLINE | ID: mdl-32585135

RÉSUMÉ

There are as yet no licensed therapeutics for the COVID-19 pandemic. The causal coronavirus (SARS-CoV-2) binds host cells via a trimeric spike whose receptor binding domain (RBD) recognizes angiotensin-converting enzyme 2, initiating conformational changes that drive membrane fusion. We find that the monoclonal antibody CR3022 binds the RBD tightly, neutralizing SARS-CoV-2, and report the crystal structure at 2.4 Å of the Fab/RBD complex. Some crystals are suitable for screening for entry-blocking inhibitors. The highly conserved, structure-stabilizing CR3022 epitope is inaccessible in the prefusion spike, suggesting that CR3022 binding facilitates conversion to the fusion-incompetent post-fusion state. Cryogenic electron microscopy (cryo-EM) analysis confirms that incubation of spike with CR3022 Fab leads to destruction of the prefusion trimer. Presentation of this cryptic epitope in an RBD-based vaccine might advantageously focus immune responses. Binders at this epitope could be useful therapeutically, possibly in synergy with an antibody that blocks receptor attachment.


Sujet(s)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Betacoronavirus/composition chimique , Betacoronavirus/immunologie , Infections à coronavirus/thérapie , Pneumopathie virale/thérapie , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/immunologie , Site allostérique , Séquence d'acides aminés , Angiotensin-converting enzyme 2 , Anticorps monoclonaux/immunologie , Anticorps neutralisants/usage thérapeutique , Anticorps antiviraux/usage thérapeutique , Complexe antigène-anticorps/composition chimique , Betacoronavirus/génétique , COVID-19 , Vaccins contre la COVID-19 , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/immunologie , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/virologie , Cryomicroscopie électronique , Cristallographie aux rayons X , Interactions hôte-microbes/immunologie , Humains , Modèles moléculaires , Tests de neutralisation , Pandémies , Peptidyl-Dipeptidase A/composition chimique , Pneumopathie virale/immunologie , Pneumopathie virale/virologie , Récepteurs viraux/composition chimique , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/usage thérapeutique , Pénétration virale , Traitements médicamenteux de la COVID-19
20.
Hum Vaccin Immunother ; 12(7): 1795-801, 2016 07 02.
Article de Anglais | MEDLINE | ID: mdl-26836234

RÉSUMÉ

Concern over the release of variola virus as an agent of bioterrorism remains high and a rapid vaccination regimen is desirable for use in the event of a confirmed release of virus. A single, high-dose (5×10(8) TCID50) of Bavarian Nordic's IMVAMUNE was tested in a Phase-II clinical trial, in humans, as a substitute for the standard (1×10(8) TCID50), using a 2-dose, 28-days apart regimen. Prior to this clinical trial taking place a Good Laboratory Practice, repeated high-dose, toxicology study was performed using IMVAMUNE, in New Zealand white rabbits and the results are reported here. Male and female rabbits were dosed twice, subcutaneously, with 5×10(8) TCID50 of IMVAMUNE (test) or saline (control), 7-days apart. The clinical condition, body-weight, food consumption, haematology, blood chemistry, immunogenicity, organ-weight, and macroscopic and microscopic pathology were investigated. Haematological investigations indicated changes within the white blood cell profile that were attributed to treatment with IMVAMUNE; these comprised slight increases in neutrophil and monocyte numbers, on study days 1-3 and a marginal increase in lymphocyte numbers on day 10. Macroscopic pathology revealed reddening at the sites of administration and thickened skin in IMVAMUNE, treated animals. After the second dose of IMVAMUNE 9/10 rabbits seroconverted, as detected by antibody ELISA on day 10, by day 21, 10/10 rabbits seroconverted. Treatment-related changes were not detected in other parameters. In conclusion, the subcutaneous injection of 2 high-doses of IMVAMUNE, to rabbits, was well tolerated producing only minor changes at the site of administration. Vaccinia-specific antibodies were raised in IMVAMUNE-vaccinated rabbits only.


Sujet(s)
Effets secondaires indésirables des médicaments/épidémiologie , Effets secondaires indésirables des médicaments/anatomopathologie , Vaccin antivariolique/administration et posologie , Vaccin antivariolique/effets indésirables , Animaux , Anticorps antiviraux/sang , Évaluation préclinique de médicament , Test ELISA , Femelle , Injections sous-cutanées , Mâle , Lapins , Vaccins atténués/administration et posologie , Vaccins atténués/effets indésirables
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...