60 results on '"Touret F"'
Search Results
2. Favipiravir strikes the SARS-CoV-2 at its Achilles heel, the RNA polymerase
- Author
-
Shannon, A., primary, Selisko, B., additional, Le, NTT, additional, Huchting, J., additional, Touret, F., additional, Piorkowski, G., additional, Fattorini, V., additional, Ferron, F., additional, Decroly, E., additional, Meier, C, additional, Coutard, B., additional, Peersen, O., additional, and Canard, B., additional
- Published
- 2020
- Full Text
- View/download PDF
3. Dose Rationale for Favipiravir Use in Patients Infected With SARS-CoV-2
- Author
-
Eloy, P., Solas, C., Touret, F., Mentre, F., Malvy, Denis, De Lamballerie, X., GUEDJ, J., Bordeaux population health (BPH), and Université de Bordeaux (UB)-Institut de Santé Publique, d'Épidémiologie et de Développement (ISPED)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
IDLIC ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie - Published
- 2020
- Full Text
- View/download PDF
4. Non-Polio Enterovirus Inhibitors: Scaffolds, Targets, and Potency─What's New?
- Author
-
Roux HFG, Touret F, Rathelot P, Sciò P, Coluccia A, Vanelle P, and Roche M
- Subjects
- Humans, Structure-Activity Relationship, Enterovirus Infections virology, Enterovirus Infections drug therapy, Hand, Foot and Mouth Disease virology, Hand, Foot and Mouth Disease drug therapy, Hand, Foot and Mouth Disease prevention & control, Animals, Antiviral Agents pharmacology, Antiviral Agents chemistry, Enterovirus drug effects
- Abstract
Enterovirus (EV) is a genus that includes a large diversity of viruses spread around the world. They are the main cause of numerous diseases with seasonal clusters, like hand-foot-mouth disease (HFMD). A vaccine is marketed in China for the prevention of HFMD caused by EV-A71. Despite the need, no antiviral is marketed to date. Therefore, several compounds have been currently evaluated to inhibit non-polio Enterovirus (NPEV), namely direct antiviral agents and host target inhibitor. We propose to make a review of the latest molecules evaluated as NPEV inhibitors and to summarize structure-activity relationships between these inhibitors and their target. We provide access to all recent information on Enterovirus inhibitors, regardless of the species, to facilitate the design of future broad-spectrum drugs.
- Published
- 2025
- Full Text
- View/download PDF
5. Epidemic Zika virus strains from the Asian lineage induce an attenuated fetal brain pathogenicity.
- Author
-
Darmuzey M, Touret F, Slowikowski E, Gladwyn-Ng I, Ahuja K, Sanchez-Felipe L, de Lamballerie X, Verfaillie C, Marques PE, Neyts J, and Kaptein SJF
- Subjects
- Animals, Female, Mice, Humans, Pregnancy, Fetus virology, Disease Models, Animal, Virus Replication, Epidemics, Asia epidemiology, Macrophages virology, Macrophages immunology, Microglia virology, Microglia pathology, Virulence, Mice, Inbred C57BL, Zika Virus pathogenicity, Zika Virus genetics, Zika Virus Infection virology, Brain virology, Brain pathology
- Abstract
The 2015-2016 Zika virus (ZIKV) outbreak in the Americas revealed the ability of ZIKV from the Asian lineage to cause birth defects, generically called congenital Zika syndrome (CZS). Notwithstanding the long circulation history of Asian ZIKV, no ZIKV-associated CZS cases were reported prior to the outbreaks in French Polynesia (2013) and Brazil (2015). Whether the sudden emergence of CZS resulted from an evolutionary event of Asian ZIKV has remained unclear. We performed a comparative analysis of the pathogenicity of pre-epidemic and epidemic Asian ZIKV strains in mouse embryonic brains using a female immunocompetent intraplacental infection mouse model. All studied Asian ZIKV strains are neurovirulent, but pre-epidemic strains are consistently more pathogenic in the embryos than their epidemic equivalents. Pathogenicity is not directly linked to viral replication. By contrast, an influx of macrophages/microglial cells is noted in infected fetal brains for both pre-epidemic and epidemic ZIKV strains. Moreover, all tested ZIKV strains trigger an immunological response, whereby the intensity of the response differs between strains, and with epidemic ZIKV strains generally mounting a more attenuated immunostimulatory response. Our study reveals that Asian ZIKV strains evolved towards pathogenic attenuation, potentially resulting in CZS emergence in neonates rather than premature death in utero., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. The effects of Remdesivir's functional groups on its antiviral potency and resistance against the SARS-CoV-2 polymerase.
- Author
-
Sama B, Selisko B, Falcou C, Fattorini V, Piorkowski G, Touret F, Donckers K, Neyts J, Jochmans D, Shannon A, Coutard B, and Canard B
- Subjects
- Humans, RNA, Viral genetics, RNA, Viral biosynthesis, Coronavirus RNA-Dependent RNA Polymerase antagonists & inhibitors, Chlorocebus aethiops, Animals, Vero Cells, Alanine analogs & derivatives, Alanine pharmacology, Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacology, Antiviral Agents pharmacology, SARS-CoV-2 drug effects, Virus Replication drug effects, COVID-19 Drug Treatment, Drug Resistance, Viral
- Abstract
Remdesivir (RDV, Veklury®) is the first FDA-approved antiviral treatment for COVID-19. It is a nucleotide analogue (NA) carrying a 1'-cyano (1'-CN) group on the ribose and a pseudo-adenine nucleobase whose contributions to the mode of action (MoA) are not clear. Here, we dissect these independent contributions by employing RDV-TP analogues. We show that while the 1'-CN group is directly responsible for transient stalling of the SARS-CoV-2 replication/transcription complex (RTC), the nucleobase plays a role in the strength of this stalling. Conversely, RNA extension assays show that the 1'-CN group plays a role in fidelity and that RDV-TP can be incorporated as a GTP analogue, albeit with lower efficiency. However, a mutagenic effect by the viral polymerase is not ascertained by deep sequencing of viral RNA from cells treated with RDV. We observe that once added to the 3' end of RNA, RDV-MP is sensitive to excision and its 1'-CN group does not impact its nsp14-mediated removal. A >14-fold RDV-resistant SARS-CoV-2 isolate can be selected carrying two mutations in the nsp12 sequence, S759A and A777S. They confer both RDV-TP discrimination over ATP by nsp12 and stalling during RNA synthesis, leaving more time for excision-repair and potentially dampening RDV efficiency. We conclude that RDV presents a multi-faced MoA. It slows down or stalls overall RNA synthesis but is efficiently repaired from the primer strand, whereas once in the template, read-through inhibition adds to this effect. Its efficient incorporation may corrupt proviral RNA, likely disturbing downstream functions in the virus life cycle., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Neutralizing and enhancing monoclonal antibodies in SARS-CoV-2 convalescent patients: lessons from early variant infection and impact on shaping emerging variants.
- Author
-
Coutant F, Touret F, Pin JJ, Alonzo M, Baronti C, Munier S, Attia M, de Lamballerie X, Ferry T, and Miossec P
- Subjects
- Humans, Antibodies, Blocking, Antibodies, Neutralizing, Antibodies, Monoclonal, Antibodies, Viral, Spike Glycoprotein, Coronavirus, SARS-CoV-2, COVID-19
- Abstract
Serological studies of COVID-19 convalescent patients have identified polyclonal lineage-specific and cross-reactive antibodies (Abs), with varying effector functions against virus variants. Individual specificities of anti-SARS-CoV-2 Abs and their impact on infectivity by other variants have been little investigated to date. Here, we dissected at a monoclonal level neutralizing and enhancing Abs elicited by early variants and how they affect infectivity of emerging variants. B cells from 13 convalescent patients originally infected by D614G or Alpha variants were immortalized to isolate 445 naturally-produced anti-SARS-CoV-2 Abs. Monoclonal antibodies (mAbs) were tested for their abilities to impact the cytopathic effect of D614G, Delta, and Omicron (BA.1) variants. Ninety-eight exhibited robust neutralization against at least one of the three variant types, while 309 showed minimal or no impact on infectivity. Thirty-eight mAbs enhanced infectivity of SARS-CoV-2. Infection with D614G/Alpha variants generated variant-specific (65 neutralizing Abs, 35 enhancing Abs) and cross-reactive (18 neutralizing Abs, 3 enhancing Abs) mAbs. Interestingly, among the neutralizing mAbs with cross-reactivity restricted to two of the three variants tested, none demonstrated specific neutralization of the Delta and Omicron variants. In contrast, cross-reactive mAbs enhancing infectivity ( n = 3) were found exclusively specific to Delta and Omicron variants. Notably, two mAbs that amplified in vitro the cytopathic effect of the Delta variant also exhibited neutralization against Omicron. These findings shed light on functional diversity of cross-reactive Abs generated during SARS-CoV-2 infection and illustrate how the balance between neutralizing and enhancing Abs facilitate variant emergence.
- Published
- 2024
- Full Text
- View/download PDF
8. Genomic surveillance reveals a dengue 2 virus epidemic lineage with a marked decrease in sensitivity to Mosnodenvir.
- Author
-
Bouzidi HS, Sen S, Piorkowski G, Pezzi L, Ayhan N, Fontaine A, Canivez T, Geulen M, Amaral R, Grard G, Durand GA, de Lamballerie X, Touret F, and Klitting R
- Subjects
- Humans, Genomics methods, Epidemics, Viral Nonstructural Proteins genetics, Animals, Dengue Virus genetics, Dengue Virus drug effects, Dengue virology, Dengue epidemiology, Phylogeny, Genome, Viral genetics, Drug Resistance, Viral genetics, Antiviral Agents pharmacology, Mutation
- Abstract
Dengue fever is the most important arbovirosis for public health, with more than 5 million cases worldwide in 2023. Mosnodenvir is the first anti-dengue compound with very high preclinical pan-serotype activity, currently undergoing phase 2 clinical evaluation. Here, by analyzing dengue virus (DENV) genomes from the 2023-2024 epidemic in the French Caribbean Islands, we show that they all exhibit mutation NS4B:V91A, previously associated with a marked decrease in sensitivity to mosnodenvir in vitro. Using antiviral activity tests on four clinical and reverse-genetic strains, we confirm a marked decrease in mosnodenvir sensitivity for DENV-2 ( > 1000 fold). Finally, combining phylogenetic analysis and experimental testing for resistance, we find that virus lineages with low sensitivity to mosnodenvir due to the V91A mutation likely emerged multiple times over the last 30 years in DENV-2 and DENV-3. These results call for increased genomic surveillance, in particular to track lineages with resistance mutations. These efforts should allow to better assess the activity profile of DENV treatments in development against circulating strains., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. New potent EV-A71 antivirals targeting capsid.
- Author
-
Roux H, Touret F, Coluccia A, Khoumeri O, Di Giorgio C, Majdi C, Sciò P, Silvestri R, Vanelle P, and Roche M
- Subjects
- Humans, Structure-Activity Relationship, Capsid Proteins antagonists & inhibitors, Capsid Proteins metabolism, Capsid Proteins chemistry, Molecular Structure, Microbial Sensitivity Tests, Dose-Response Relationship, Drug, Antiviral Agents pharmacology, Antiviral Agents chemistry, Antiviral Agents chemical synthesis, Enterovirus A, Human drug effects, Capsid drug effects, Capsid metabolism
- Abstract
The enterovirus is a genus of single-stranded, highly diverse positive-sense RNA viruses, including Human Enterovirus A-D and Human Rhinovirus A-C species. They are responsible for numerous diseases and some infections can progress to life-threatening complications, particularly in children or immunocompromised patients. To date, there is no treatment against enteroviruses on the market, except for polioviruses (vaccine) and EV-A71 (vaccine in China). Following a decrease in enterovirus infections during and shortly after the (SARS-Cov2) lockdown, enterovirus outbreaks were once again detected, notably in young children. This reemergence highlights on the need to develop broad-spectrum treatment against enteroviruses. Over the last year, our research team has identified a new class of small-molecule inhibitors showing anti-EV activity. Targeting the well-known hydrophobic pocket in the viral capsid, these compounds show micromolar activity against EV-A71 and a high selectivity index (SI) (5h: EC
50, MRC-5 = 0.57 μM, CC50, MRC-5 >20 μM, SI > 35; EC50, RD = 4.38 μM, CC50, RD > 40 μM, SI > 9; 6c: EC50, MRC-5 = 0.29 μM, CC50, MRC-5 >20 μM, SI > 69; EC50, RD = 1.66 μM, CC50, RD > 40 μM, SI > 24; Reference: Vapendavir EC50, MRC-5 = 0.36 μM, CC50, MRC-5 > 20 μM, EC50, RD = 0.53 μM, CC50, RD > 40 μM, SI > 63). The binding mode of these compounds in complex with enterovirus capsids was analyzed and showed a series of conserved interactions. Consequently, 6c and its derivatives are promising candidates for the treatment of enterovirus infections., Competing Interests: Declaration of competing interest The authors declare no competing financial interest., (Copyright © 2024. Published by Elsevier Masson SAS.)- Published
- 2024
- Full Text
- View/download PDF
10. From the "One-Molecule, One-Target, One-Disease" Concept towards Looking for Multi-Target Therapeutics for Treating Non-Polio Enterovirus (NPEV) Infections.
- Author
-
Roux H, Touret F, Rathelot P, Vanelle P, and Roche M
- Abstract
Non-polio enteroviruses (NPEVs), namely coxsackieviruses (CV), echoviruses (E), enteroviruses (EV), and rhinoviruses (RV), are responsible for a wide variety of illnesses. Some infections can progress to life-threatening conditions in children or immunocompromised patients. To date, no treatments have been approved. Several molecules have been evaluated through clinical trials without success. To overcome these failures, the multi-target directed ligand (MTDL) strategy could be applied to tackle enterovirus infections. This work analyzes registered clinical trials involving antiviral drugs to highlight the best candidates and develops filters to apply to a selection for MTDL synthesis. We explicitly stated the methods used to answer the question: which solution can fight NPEVs effectively? We note the originality and relevance of this proposal in relation to the state of the art in the enterovirus-inhibitors field. Several combinations are possible to broaden the antiviral spectrum and potency. We discuss data related to the virus and data related to each LEAD compound identified so far. Overall, this study proposes a perspective on different strategies to overcome issues identified in clinical trials and evaluate the "MTDL" potential to improve the efficacy of drugs, broaden the antiviral targets, possibly reduce the adverse effects, drug design costs and limit the selection of drug-resistant virus variants.
- Published
- 2024
- Full Text
- View/download PDF
11. Preclinical in vivo assessment of the activity of AZD7442 anti-SARS-CoV-2 monoclonal antibodies against Omicron sublineages.
- Author
-
Driouich JS, Cochin M, Lingas G, Luciani L, Baronti C, Bernadin O, Gilles M, Villarroel PMS, Moureau G, Petit PR, Dupont A, Izopet J, Kamar N, Autran B, Paintaud G, Caillard S, le Bourgeois A, Richez C, Couzi L, Xhaard A, Marjanovic Z, Avouac J, Jacquet C, Anglicheau D, Cheminant M, Nguyen S, Terrier B, Gottenberg JE, Besson C, Letrou S, Tine J, Basilua JM, Angoulvant D, Tardivon C, Blancho G, Martin-Blondel G, Yazdanpanah Y, Mentré F, Lévy V, Touret F, Guedj J, de Lamballerie X, and Nougairède A
- Abstract
Therapeutic monoclonal antibodies have been successful in protecting vulnerable populations against SARS-CoV-2. However, their effectiveness has been hampered by the emergence of new variants. To adapt the therapeutic landscape, health authorities have based their recommendations mostly on in vitro neutralization tests. However, these do not provide a reliable understanding of the changes in the dose-effect relationship and how they may translate into clinical efficacy. Taking the example of EvusheldTM (AZD7442), we aimed to investigate how in vivo data can provide critical quantitative results and project clinical effectiveness. We used the Golden Syrian hamster model to estimate 90 % effective concentrations (EC90) of AZD7442 in vivo against SARS-CoV-2 Omicron BA.1, BA.2 and BA.5 variants. While our in vivo results confirmed the partial loss of AZD7442 activity for BA.1 and BA.2, they showed a much greater loss of efficacy against BA.5 than that obtained in vitro. We analyzed in vivo EC90s in perspective with antibody levels measured in a cohort of immunocompromised patients who received 300 mg of AZD7442. We found that a substantial proportion of patients had serum levels of anti-SARS-CoV-2 spike protein IgG above the estimated in vivo EC90 for BA.1 and BA.2 (21 % and 92 % after 1 month, respectively), but not for BA.5. These findings suggest that AZD7442 is likely to retain clinical efficacy against BA.2 and BA.1, but not against BA.5. Overall, the present study illustrates the importance of complementing in vitro investigations by preclinical studies in animal models to help predict the efficacy of monoclonal antibodies in humans., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Current status of pathogen handling in European laboratories: focus on viral inactivation process.
- Author
-
Pastorino B, Touret F, Gilles M, De Lamballerie X, and Charrel RN
- Abstract
For handling safely infectious agents, European laboratories must comply with specific EC Directives, national regulations and recommendations from the World Health Organization (WHO). To prevent laboratory acquired infections (LAIs) and pathogens dissemination, a key biosafety rule requires that any infectious material (clinical specimens or research samples) manipulated outside a biosafety cabinet (BSC) must be inactivated unless the lack of infectivity is proven. This inactivation process is a crucial step for biosafety and must be guided by a rigorous experimental qualification and validation procedure. However, for diagnostic or research laboratories, this process is not harmonized with common standard operation procedures (SOPs) but based on individual risk assessment and general international guidelines which can pose problems in emergency situations such as major outbreaks or pandemics. This review focuses on viral inactivation method, outlining the current regulatory framework, its limitations and a number of ways in which biosafety can be improved., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Pastorino, Touret, Gilles, De Lamballerie and Charrel.)
- Published
- 2024
- Full Text
- View/download PDF
13. Broad-spectrum dengue virus detection using the commercial RealStar dengue RT-PCR kit 3.0 (Altona) and an in-house combined real-time RT-PCR assay.
- Author
-
Luciani L, Combe P, Touret F, Gazin C, Klitting R, Pezzi L, Thirion L, Charrel R, Grard G, de Lamballerie X, and Nougairède A
- Abstract
In endemic areas, the genetic diversity among co-circulating dengue virus (DENV) strains is considerable and new, highly divergent strains are identified on a regular basis. It is thus critical to ensure that molecular diagnostic tools effectively detect virus genomes even in case of important genetic variation. Here, we tested both the pan-DENV detection capacity and the limit of detection of two real-time RT-PCR assays: (i) the commercial RealStar Altona 3.0 system and (ii) a laboratory developed test (LDT) combining two RT-PCR systems in a single reaction tube (DenAllDUO). We used a panel of DENV strains representative of the genetic diversity within DENV species, combined with three in vitro transcribed RNAs as surrogates for unavailable strains corresponding to recently discovered strains with substantial genetic divergence: DENV serotype 1 (DENV-1) Brun2014, DENV-2 QML22 and DENV-4 DKE121. Both systems (i) targeted the genome 3' untranslated region, (ii) displayed a broad detection spectrum, encompassing most of DENV species diversity, and (iii) detected the three aforementioned divergent strains. DenAllDUO detected all the strains tested, whereas the RealStar system failed to detect strains from DENV-4 genotype III. Altogether, our findings support the value of these two RT-PCR systems as part of the Dengue diagnostic arsenal., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 The Authors. Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
14. Further preclinical characterization of molnupiravir against SARS-CoV-2: Antiviral activity determinants and viral genome alteration patterns.
- Author
-
Petit PR, Touret F, Driouich JS, Cochin M, Luciani L, Bernadin O, Laprie C, Piorkowski G, Fraisse L, Sjö P, Mowbray CE, Escudié F, Scandale I, Chatelain E, de Lamballerie X, Solas C, and Nougairède A
- Abstract
The SARS-CoV-2 pandemic has highlighted the need for broad-spectrum antiviral drugs to respond promptly to viral emergence. We conducted a preclinical study of molnupiravir (MOV) against SARS-CoV-2 to fully characterise its antiviral properties and mode of action. The antiviral activity of different concentrations of MOV was evaluated ex vivo on human airway epithelium (HAE) and in vivo in a hamster model at three escalating doses (150, 300 and 400 mg/kg/day) according to three different regimens (preventive, pre-emptive and curative). We assessed viral loads and infectious titres at the apical pole of HAE and in hamster lungs, and MOV trough concentration in plasma and lungs. To explore the mode of action of the MOV, the entire genomes of the collected viruses were deep-sequenced. MOV effectively reduced viral titres in HAE and in the lungs of treated animals. Early treatment after infection was a key factor in efficacy, probably associated with high lung concentrations of MOV, suggesting good accumulation in the lung. MOV induced genomic alteration in viral genomes with an increase in the number of minority variants, and predominant G to A transitions. The observed reduction in viral replication and its mechanism of action leading to lethal mutagenesis, supported by clinical trials showing antiviral action in humans, provide a convincing basis for further research as an additional means in the fight against COVID-19 and other RNA viruses., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
15. Generation and evaluation of protease inhibitor-resistant SARS-CoV-2 strains.
- Author
-
Bouzidi HS, Driouich JS, Klitting R, Bernadin O, Piorkowski G, Amaral R, Fraisse L, Mowbray CE, Scandale I, Escudié F, Chatelain E, de Lamballerie X, Nougairède A, and Touret F
- Subjects
- Animals, Cricetinae, Protease Inhibitors pharmacology, SARS-CoV-2 genetics, Enzyme Inhibitors, Antiviral Agents pharmacology, Mesocricetus, COVID-19, Anti-Infective Agents
- Abstract
Since the start of the SARS-CoV-2 pandemic, the search for antiviral therapies has been at the forefront of medical research. To date, the 3CLpro inhibitor nirmatrelvir (Paxlovid®) has shown the best results in clinical trials and the greatest robustness against variants. A second SARS-CoV-2 protease inhibitor, ensitrelvir (Xocova®), has been developed. Ensitrelvir, currently in Phase 3, was approved in Japan under the emergency regulatory approval procedure in November 2022, and is available since March 31, 2023. One of the limitations for the use of antiviral monotherapies is the emergence of resistance mutations. Here, we experimentally generated mutants resistant to nirmatrelvir and ensitrelvir in vitro following repeating passages of SARS-CoV-2 in the presence of both antivirals. For both molecules, we demonstrated a loss of sensitivity for resistance mutants in vitro. Using a Syrian golden hamster infection model, we showed that the ensitrelvir M49L mutation, in the multi-passage strain, confers a high level of in vivo resistance. Finally, we identified a recent increase in the prevalence of M49L-carrying sequences, which appears to be associated with multiple repeated emergence events in Japan and may be related to the use of Xocova® in the country since November 2022. These results highlight the strategic importance of genetic monitoring of circulating SARS-CoV-2 strains to ensure that treatments administered retain their full effectiveness., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Hybrid molecules based on an emodin scaffold. Synthesis and activity against SARS-CoV-2 and Plasmodium .
- Author
-
Li Y, Touret F, de Lamballerie X, Nguyen M, Laurent M, Benoit-Vical F, Robert A, Liu Y, and Meunier B
- Subjects
- Humans, SARS-CoV-2, Emodin pharmacology, COVID-19, Plasmodium, Antimalarials pharmacology
- Abstract
Since the Covid-19 epidemic, it has been clear that the availability of small and affordable drugs that are able to efficiently control viral infections in humans is still a challenge in medicinal chemistry. The synthesis and biological activities of a series of hybrid molecules that combine an emodin moiety and other structural moieties expected to act as possible synergistic pharmacophores in a single molecule were studied. Emodin has been reported to block the entry of the SARS-CoV-2 virus into human cells and might also inhibit cytokine production, resulting in the reduction of pulmonary injury induced by SARS-CoV-2. The pharmacophore associated with emodin was either a polyamine residue (emodin-PA series), a choice driven by the fact that a natural alkyl PA like spermine and spermidine play regulatory roles in immune cell functions, or a diphenylmethylpiperazine derivative of the norchlorcyclizine series (emoxyzine series). In fact, diphenylmethylpiperazine antagonists of the H1 histamine receptor display activity against several viruses by multiple interrelated mechanisms. In the emoxyzine series, the most potent drug against SARS-CoV-2 was ( R )-emoxyzine-2, with an EC
50 value = 1.9 μM, which is in the same range as that of the reference drug remdesivir. However, the selectivity index was rather low, indicating that the dissociation of antiviral potency and cytotoxicity remains a challenge. In addition, since emodin was also reported to be a relatively high-affinity inhibitor of the virulence regulator FIKK kinase from the malaria parasite Plasmodium vivax , the antimalarial activity of the synthesized hybrid compounds has been evaluated. However, these molecules cannot efficiently compete with the currently used antimalarial drugs.- Published
- 2023
- Full Text
- View/download PDF
17. Structure-guided optimization of adenosine mimetics as selective and potent inhibitors of coronavirus nsp14 N7-methyltransferases.
- Author
-
Hausdorff M, Delpal A, Barelier S, Nicollet L, Canard B, Touret F, Colmant A, Coutard B, Vasseur JJ, Decroly E, and Debart F
- Subjects
- Humans, Adenosine pharmacology, Pandemics, SARS-CoV-2 genetics, Viral Nonstructural Proteins metabolism, Antiviral Agents pharmacology, S-Adenosylmethionine, RNA, Viral genetics, Adenine, Methyltransferases metabolism, COVID-19
- Abstract
The COVID-19 pandemic reveals the urgent need to develop new therapeutics targeting the SARS-CoV-2 replication machinery. The first antiviral drugs were nucleoside analogues targeting RdRp and protease inhibitors active on nsp5 Mpro. In addition to these common antiviral targets, SARS-CoV-2 codes for the highly conserved protein nsp14 harbouring N7-methyltransferase (MTase) activity. Nsp14 is involved in cap N7-methylation of viral RNA and its inhibition impairs viral RNA translation and immune evasion, making it an attractive new antiviral target. In this work, we followed a structure-guided drug design approach to design bisubstrates mimicking the S-adenosylmethionine methyl donor and RNA cap. We developed adenosine mimetics with an N-arylsulfonamide moiety in the 5'-position, recently described as a guanine mimicking the cap structure in a potent adenosine-derived nsp14 inhibitor. Here, the adenine moiety was replaced by hypoxanthine, N
6 -methyladenine, or C7-substituted 7-deaza-adenine. 26 novel adenosine mimetics were synthesized, one of which selectively inhibits nsp14 N7-MTase activity with a subnanomolar IC50 (and seven with a single-digit nanomolar IC50 ). In the most potent inhibitors, adenine was replaced by two different 7-deaza-adenines bearing either a phenyl or a 3-quinoline group at the C7-position via an ethynyl linker. These more complex compounds are barely active on the cognate human N7-MTase and docking experiments reveal that their selectivity of inhibition might result from the positioning of their C7 substitution in a SAM entry tunnel present in the nsp14 structure and absent in the hN7-MTase. These compounds show moderate antiviral activity against SARS-CoV-2 replication in cell culture, suggesting delivery or stability issue., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
18. Activity of Sotrovimab against BQ.1.1 and XBB.1 Omicron sublineages in a hamster model.
- Author
-
Driouich JS, Bernadin O, Touret F, de Lamballerie X, and Nougairède A
- Subjects
- Animals, Cricetinae, Humans, SARS-CoV-2, Antibodies, Monoclonal, Humanized, Antibodies, Neutralizing, Antibodies, Viral, COVID-19
- Abstract
The successive emergence of SARS-CoV-2 Omicron variants has completely changed the modalities of use of therapeutic monoclonal antibodies. Recent in vitro studies indicated that only Sotrovimab has maintained partial activity against BQ.1.1 and XBB.1. In the present study, we used the hamster model to determine whether Sotrovimab retains antiviral activity against these Omicron variants in vivo. Our results show that at exposures consistent with those observed in humans, Sotrovimab remains active against BQ.1.1 and XBB.1, although for BQ.1.1 the efficacy is lower than that observed against the first globally dominant Omicron sublineages BA.1 and BA.2., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. Sotrovimab retains activity against SARS-CoV-2 omicron variant BQ.1.1 in a non-human primate model.
- Author
-
Hérate C, Marlin R, Touret F, Dereuddre-Bosquet N, Donati F, Relouzat F, Junges L, Galhaut M, Dehan O, Sconosciuti Q, Nougairède A, de Lamballerie X, van der Werf S, and Le Grand R
- Abstract
The SARS-CoV2 Omicron variants have acquired new Spike mutations leading to escape from the most of the currently available monoclonal antibody treatments reducing the options for patients suffering from severe Covid-19. Recently, both in vitro and in vivo data have suggested that Sotrovimab could retain partial activity against recent omicron sub-lineage such as BA.5 variants, including BQ.1.1. Here we report full efficacy of Sotrovimab against BQ.1.1 viral replication as measure by RT-qPCR in a non-human primate challengemodel., Competing Interests: The authors have declared that no conflict of interest exists., (© 2023 Université Paris-Saclay.)
- Published
- 2023
- Full Text
- View/download PDF
20. Enhanced neutralization escape to therapeutic monoclonal antibodies by SARS-CoV-2 omicron sub-lineages.
- Author
-
Touret F, Giraud E, Bourret J, Donati F, Tran-Rajau J, Chiaravalli J, Lemoine F, Agou F, Simon-Lorière E, van der Werf S, and de Lamballerie X
- Abstract
The landscape of SARS-CoV-2 variants dramatically diversified with the simultaneous appearance of multiple subvariants originating from BA.2, BA.4, and BA.5 Omicron sub-lineages. They harbor a specific set of mutations in the spike that can make them more evasive to therapeutic monoclonal antibodies. In this study, we compared the neutralizing potential of monoclonal antibodies against the Omicron BA.2.75.2, BQ.1, BQ.1.1, and XBB variants, with a pre-Omicron Delta variant as a reference. Sotrovimab retains some activity against BA.2.75.2, BQ.1, and XBB as it did against BA.2/BA.5, but is less active against BQ.1.1. Within the Evusheld/AZD7442 cocktail, Cilgavimab lost all activity against all subvariants studied, resulting in loss of Evusheld activity. Finally, Bebtelovimab, while still active against BA.2.75, also lost all neutralizing activity against BQ.1, BQ.1.1, and XBB variants., Competing Interests: The authors declare that there is no conflict of interest., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
21. Blocking NS3-NS4B interaction inhibits dengue virus in non-human primates.
- Author
-
Goethals O, Kaptein SJF, Kesteleyn B, Bonfanti JF, Van Wesenbeeck L, Bardiot D, Verschoor EJ, Verstrepen BE, Fagrouch Z, Putnak JR, Kiemel D, Ackaert O, Straetemans R, Lachau-Durand S, Geluykens P, Crabbe M, Thys K, Stoops B, Lenz O, Tambuyzer L, De Meyer S, Dallmeier K, McCracken MK, Gromowski GD, Rutvisuttinunt W, Jarman RG, Karasavvas N, Touret F, Querat G, de Lamballerie X, Chatel-Chaix L, Milligan GN, Beasley DWC, Bourne N, Barrett ADT, Marchand A, Jonckers THM, Raboisson P, Simmen K, Chaltin P, Bartenschlager R, Bogers WM, Neyts J, and Van Loock M
- Subjects
- Animals, Humans, Mice, Clinical Trials, Phase I as Topic, Dose-Response Relationship, Drug, Drug Resistance, Viral, In Vitro Techniques, Molecular Targeted Therapy, Protein Binding drug effects, Virus Replication, Antiviral Agents adverse effects, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Dengue drug therapy, Dengue prevention & control, Dengue virology, Dengue Virus classification, Dengue Virus drug effects, Primates virology, Viral Nonstructural Proteins antagonists & inhibitors, Viral Nonstructural Proteins metabolism
- Abstract
Dengue is a major health threat and the number of symptomatic infections caused by the four dengue serotypes is estimated to be 96 million
1 with annually around 10,000 deaths2 . However, no antiviral drugs are available for the treatment or prophylaxis of dengue. We recently described the interaction between non-structural proteins NS3 and NS4B as a promising target for the development of pan-serotype dengue virus (DENV) inhibitors3 . Here we present JNJ-1802-a highly potent DENV inhibitor that blocks the NS3-NS4B interaction within the viral replication complex. JNJ-1802 exerts picomolar to low nanomolar in vitro antiviral activity, a high barrier to resistance and potent in vivo efficacy in mice against infection with any of the four DENV serotypes. Finally, we demonstrate that the small-molecule inhibitor JNJ-1802 is highly effective against viral infection with DENV-1 or DENV-2 in non-human primates. JNJ-1802 has successfully completed a phase I first-in-human clinical study in healthy volunteers and was found to be safe and well tolerated4 . These findings support the further clinical development of JNJ-1802, a first-in-class antiviral agent against dengue, which is now progressing in clinical studies for the prevention and treatment of dengue., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
22. Biosynthetic proteins targeting the SARS-CoV-2 spike as anti-virals.
- Author
-
Thébault S, Lejal N, Dogliani A, Donchet A, Urvoas A, Valerio-Lepiniec M, Lavie M, Baronti C, Touret F, Da Costa B, Bourgon C, Fraysse A, Saint-Albin-Deliot A, Morel J, Klonjkowski B, de Lamballerie X, Dubuisson J, Roussel A, Minard P, Le Poder S, Meunier N, and Delmas B
- Subjects
- Antiviral Agents chemistry, Antiviral Agents pharmacology, Humans, Peptidyl-Dipeptidase A metabolism, Protein Binding, Recombinant Proteins pharmacology, Recombinant Proteins therapeutic use, SARS-CoV-2 genetics, Angiotensin-Converting Enzyme 2 chemistry, Angiotensin-Converting Enzyme 2 metabolism, Recombinant Fusion Proteins pharmacology, Recombinant Fusion Proteins therapeutic use, Spike Glycoprotein, Coronavirus metabolism, COVID-19 Drug Treatment
- Abstract
The binding of the SARS-CoV-2 spike to angiotensin-converting enzyme 2 (ACE2) promotes virus entry into the cell. Targeting this interaction represents a promising strategy to generate antivirals. By screening a phage-display library of biosynthetic protein sequences build on a rigid alpha-helicoidal HEAT-like scaffold (named αReps), we selected candidates recognizing the spike receptor binding domain (RBD). Two of them (F9 and C2) bind the RBD with affinities in the nM range, displaying neutralisation activity in vitro and recognizing distinct sites, F9 overlapping the ACE2 binding motif. The F9-C2 fusion protein and a trivalent αRep form (C2-foldon) display 0.1 nM affinities and EC50 of 8-18 nM for neutralization of SARS-CoV-2. In hamsters, F9-C2 instillation in the nasal cavity before or during infections effectively reduced the replication of a SARS-CoV-2 strain harbouring the D614G mutation in the nasal epithelium. Furthermore, F9-C2 and/or C2-foldon effectively neutralized SARS-CoV-2 variants (including delta and omicron variants) with EC50 values ranging from 13 to 32 nM. With their high stability and their high potency against SARS-CoV-2 variants, αReps provide a promising tool for SARS-CoV-2 therapeutics to target the nasal cavity and mitigate virus dissemination in the proximal environment., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
23. Need for a Standardized Translational Drug Development Platform: Lessons Learned from the Repurposing of Drugs for COVID-19.
- Author
-
Assmus F, Driouich JS, Abdelnabi R, Vangeel L, Touret F, Adehin A, Chotsiri P, Cochin M, Foo CS, Jochmans D, Kim S, Luciani L, Moureau G, Park S, Pétit PR, Shum D, Wattanakul T, Weynand B, Fraisse L, Ioset JR, Mowbray CE, Owen A, Hoglund RM, Tarning J, Lamballerie X, Nougairède A, Neyts J, Sjö P, Escudié F, Scandale I, and Chatelain E
- Abstract
In the absence of drugs to treat or prevent COVID-19, drug repurposing can be a valuable strategy. Despite a substantial number of clinical trials, drug repurposing did not deliver on its promise. While success was observed with some repurposed drugs (e.g., remdesivir, dexamethasone, tocilizumab, baricitinib), others failed to show clinical efficacy. One reason is the lack of clear translational processes based on adequate preclinical profiling before clinical evaluation. Combined with limitations of existing in vitro and in vivo models, there is a need for a systematic approach to urgent antiviral drug development in the context of a global pandemic. We implemented a methodology to test repurposed and experimental drugs to generate robust preclinical evidence for further clinical development. This translational drug development platform comprises in vitro, ex vivo, and in vivo models of SARS-CoV-2, along with pharmacokinetic modeling and simulation approaches to evaluate exposure levels in plasma and target organs. Here, we provide examples of identified repurposed antiviral drugs tested within our multidisciplinary collaboration to highlight lessons learned in urgent antiviral drug development during the COVID-19 pandemic. Our data confirm the importance of assessing in vitro and in vivo potency in multiple assays to boost the translatability of pre-clinical data. The value of pharmacokinetic modeling and simulations for compound prioritization is also discussed. We advocate the need for a standardized translational drug development platform for mild-to-moderate COVID-19 to generate preclinical evidence in support of clinical trials. We propose clear prerequisites for progression of drug candidates for repurposing into clinical trials. Further research is needed to gain a deeper understanding of the scope and limitations of the presented translational drug development platform.
- Published
- 2022
- Full Text
- View/download PDF
24. Pre-clinical evaluation of antiviral activity of nitazoxanide against SARS-CoV-2.
- Author
-
Driouich JS, Cochin M, Touret F, Petit PR, Gilles M, Moureau G, Barthélémy K, Laprie C, Wattanakul T, Chotsiri P, Hoglund RM, Tarning J, Fraisse L, Sjö P, Mowbray CE, Escudié F, Scandale I, Chatelain E, de Lamballerie X, Solas C, and Nougairède A
- Subjects
- Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Cricetinae, Humans, Nitro Compounds, Thiazoles, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
Background: To address the emergence of SARS-CoV-2, multiple clinical trials in humans were rapidly started, including those involving an oral treatment by nitazoxanide, despite no or limited pre-clinical evidence of antiviral efficacy., Methods: In this work, we present a complete pre-clinical evaluation of the antiviral activity of nitazoxanide against SARS-CoV-2., Findings: First, we confirmed the in vitro efficacy of nitazoxanide and tizoxanide (its active metabolite) against SARS-CoV-2. Then, we demonstrated nitazoxanide activity in a reconstructed bronchial human airway epithelium model. In a SARS-CoV-2 virus challenge model in hamsters, oral and intranasal treatment with nitazoxanide failed to impair viral replication in commonly affected organs. We hypothesized that this could be due to insufficient diffusion of the drug into organs of interest. Indeed, our pharmacokinetic study confirmed that concentrations of tizoxanide in organs of interest were always below the in vitro EC
50 ., Interpretation: These preclinical results suggest, if directly applicable to humans, that the standard formulation and dosage of nitazoxanide is not effective in providing antiviral therapy for Covid-19., Funding: This work was supported by the Fondation de France "call FLASH COVID-19", project TAMAC, by "Institut national de la santé et de la recherche médicale" through the REACTing (REsearch and ACTion targeting emerging infectious diseases), by REACTING/ANRS MIE under the agreement No. 21180 ('Activité des molécules antivirales dans le modèle hamster'), by European Virus Archive Global (EVA 213 GLOBAL) funded by the European Union's Horizon 2020 research and innovation program under grant agreement No. 871029 and DNDi under support by the Wellcome Trust Grant ref: 222489/Z/21/Z through the COVID-19 Therapeutics Accelerator"., Competing Interests: Declaration of interests None to declare., (Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
25. Identification of potent inhibitors of arenavirus and SARS-CoV-2 exoribonucleases by fluorescence polarization assay.
- Author
-
Hernández S, Feracci M, De Jesus CT, El Kazzi P, Kaci R, Garlatti L, Mondielli C, Bailly F, Cotelle P, Touret F, de Lamballerie X, Coutard B, Decroly E, Canard B, Ferron F, and Alvarez K
- Subjects
- Animals, Chlorocebus aethiops, Fluorescence Polarization, Vero Cells, Viral Nonstructural Proteins antagonists & inhibitors, Antiviral Agents pharmacology, Arenavirus drug effects, Exoribonucleases antagonists & inhibitors, SARS-CoV-2 drug effects
- Abstract
Viral exoribonucleases are uncommon in the world of RNA viruses. To date, they have only been identified in the Arenaviridae and the Coronaviridae families. The exoribonucleases of these viruses play a crucial role in the pathogenicity and interplay with host innate immune response. Moreover, coronaviruses exoribonuclease is also involved in a proofreading mechanism ensuring the genetic stability of the viral genome. Because of their key roles in virus life cycle, they constitute attractive target for drug design. Here we developed a sensitive, robust and reliable fluorescence polarization assay to measure the exoribonuclease activity and its inhibition in vitro. The effectiveness of the method was validated on three different viral exoribonucleases, including SARS-CoV-2, Lymphocytic Choriomeningitis and Machupo viruses. We performed a screening of a focused library consisting of 113 metal chelators. Hit compounds were recovered with an IC
50 at micromolar level. We confirmed 3 hits in SARS-CoV-2 infected Vero-E6 cells., (Copyright © 2022. Published by Elsevier B.V.)- Published
- 2022
- Full Text
- View/download PDF
26. In vitro activity of therapeutic antibodies against SARS-CoV-2 Omicron BA.1, BA.2 and BA.5.
- Author
-
Touret F, Baronti C, Pastorino B, Villarroel PMS, Ninove L, Nougairède A, and de Lamballerie X
- Subjects
- Antibodies, Monoclonal, Antibodies, Monoclonal, Humanized, Antibodies, Neutralizing, Antibodies, Viral, Drug Combinations, Humans, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, COVID-19 Drug Treatment
- Abstract
The replacement of the Omicron BA.1 variant of SARS-CoV-2 by the BA.2 and the rapid growth of the BA.5 sub lineage, which have both different sets of mutations in the spike glycoprotein, alters the spectrum of activity of therapeutic antibodies currently licensed in the European Union. Using clinical strains of the Omicron BA.2 and BA.5 variants, we compared the neutralising power of monoclonal antibodies against the Omicron BA.1, BA.2 and BA.5 variants, using an ancestral strain (lineage B.1, D614G) and a Delta variant strain as reference. Sotrovimab/Vir-7831 is less active against BA.2 than against BA.1 (fold change reduction ~ 1,4) and even less active against BA.5 (fold change reduction ~ 2.7). Within the Evusheld /AZD7442 cocktail, Cilgavimab/AZD1061 is more active against BA.2 and BA.5 than against BA.1 (fold change increase ~ 32), whilst the very low activity of Tixagevimab/AZD8895 against BA.1 is not enhanced against BA.2 nor BA.5. In total, compared to BA.1, the activity of the Evusheld/AZD7442 is significantly improved against BA.2 while BA.5 is intermediate but closer to BA.2., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. A simple reverse genetics method to generate recombinant coronaviruses.
- Author
-
Mélade J, Piorkowski G, Touret F, Fourié T, Driouich JS, Cochin M, Bouzidi HS, Coutard B, Nougairède A, and de Lamballerie X
- Subjects
- Animals, Antiviral Agents, Cats, Reverse Genetics, COVID-19 genetics, SARS-CoV-2 genetics
- Abstract
Engineering recombinant viruses is a pre-eminent tool for deciphering the biology of emerging viral pathogens such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the large size of coronavirus genomes renders the current reverse genetics methods challenging. Here, we describe a simple method based on "infectious subgenomic amplicons" (ISA) technology to generate recombinant infectious coronaviruses with no need for reconstruction of the complete genomic cDNA and apply this method to SARS-CoV-2 and also to the feline enteric coronavirus. In both cases we rescue wild-type viruses with biological characteristics similar to original strains. Specific mutations and fluorescent red reporter genes can be readily incorporated into the SARS-CoV-2 genome enabling the generation of a genomic variants and fluorescent reporter strains for in vivo experiments, serological diagnosis, and antiviral assays. The swiftness and simplicity of the ISA method has the potential to facilitate the advance of coronavirus reverse genetics studies, to explore the molecular biological properties of the SARS-CoV-2 variants, and to accelerate the development of effective therapeutic reagents., (© 2022 Unité des Virus Emergents. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
28. Potent Inhibition of SARS-CoV-2 nsp14 N 7-Methyltransferase by Sulfonamide-Based Bisubstrate Analogues.
- Author
-
Ahmed-Belkacem R, Hausdorff M, Delpal A, Sutto-Ortiz P, Colmant AMG, Touret F, Ogando NS, Snijder EJ, Canard B, Coutard B, Vasseur JJ, Decroly E, and Debart F
- Subjects
- Exoribonucleases antagonists & inhibitors, Exoribonucleases chemistry, Humans, Methyltransferases, Molecular Docking Simulation, RNA, Viral genetics, S-Adenosylmethionine, Sulfonamides pharmacology, Viral Nonstructural Proteins antagonists & inhibitors, Viral Nonstructural Proteins chemistry, COVID-19 virology, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, COVID-19 Drug Treatment
- Abstract
Enzymes involved in RNA capping of SARS-CoV-2 are essential for the stability of viral RNA, translation of mRNAs, and virus evasion from innate immunity, making them attractive targets for antiviral agents. In this work, we focused on the design and synthesis of nucleoside-derived inhibitors against the SARS-CoV-2 nsp14 ( N 7-guanine)-methyltransferase ( N 7-MTase) that catalyzes the transfer of the methyl group from the S -adenosyl-l-methionine (SAM) cofactor to the N 7-guanosine cap. Seven compounds out of 39 SAM analogues showed remarkable double-digit nanomolar inhibitory activity against the N 7-MTase nsp14. Molecular docking supported the structure-activity relationships of these inhibitors and a bisubstrate-based mechanism of action. The three most potent inhibitors significantly stabilized nsp14 (Δ T
m ≈ 11 °C), and the best inhibitor demonstrated high selectivity for nsp14 over human RNA N 7-MTase.- Published
- 2022
- Full Text
- View/download PDF
29. In vitro evaluation of therapeutic antibodies against a SARS-CoV-2 Omicron B.1.1.529 isolate.
- Author
-
Touret F, Baronti C, Bouzidi HS, and de Lamballerie X
- Subjects
- Antibodies, Monoclonal, Antibodies, Monoclonal, Humanized, Antibodies, Neutralizing, Antibodies, Viral therapeutic use, Humans, Membrane Glycoproteins, Neutralization Tests, SARS-CoV-2, Spike Glycoprotein, Coronavirus, Viral Envelope Proteins, COVID-19 Drug Treatment
- Abstract
The emergence and rapid spread of the Omicron variant of SARS-CoV-2, which has more than 30 substitutions in the spike glycoprotein, compromises the efficacy of currently available vaccines and therapeutic antibodies. Using a clinical strain of the Omicron variant, we analyzed the neutralizing power of eight currently used monoclonal antibodies compared to the ancestral B.1 BavPat1 D614G strain. We observed that six of these antibodies have lost their ability to neutralize the Omicron variant. Of the antibodies still having neutralizing activity, Sotrovimab/Vir-7831 shows the smallest reduction in activity, with a factor change of 3.1. Cilgavimab/AZD1061 alone shows a reduction in efficacy of 15.8, resulting in a significant loss of activity for the Evusheld cocktail (42.6-fold reduction) in which the other antibody, Tixagevimab, does not retain significant activity against Omicron. Our results suggest that the clinical efficacy of the initially proposed doses should be rapidly evaluated and the possible need to modify doses or propose combination therapies should be considered., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
30. The SARS-CoV-2 Alpha variant exhibits comparable fitness to the D614G strain in a Syrian hamster model.
- Author
-
Cochin M, Luciani L, Touret F, Driouich JS, Petit PR, Moureau G, Baronti C, Laprie C, Thirion L, Maes P, Boudewijns R, Neyts J, de Lamballerie X, and Nougairède A
- Subjects
- Animals, Antibodies, Neutralizing blood, Cytokines genetics, Female, Gastrointestinal Tract virology, Genome, Viral, Lung virology, Nasal Lavage Fluid virology, SARS-CoV-2 genetics, Virus Replication, COVID-19 blood, COVID-19 immunology, COVID-19 virology, Disease Models, Animal, Mesocricetus, SARS-CoV-2 physiology
- Abstract
Late 2020, SARS-CoV-2 Alpha variant emerged in United Kingdom and gradually replaced G614 strains initially involved in the global spread of the pandemic. In this study, we use a Syrian hamster model to compare a clinical strain of Alpha variant with an ancestral G614 strain. The Alpha variant succeed to infect animals and to induce a pathology that mimics COVID-19. However, both strains replicate to almost the same level and induced a comparable disease and immune response. A slight fitness advantage is noted for the G614 strain during competition and transmission experiments. These data do not corroborate the epidemiological situation observed during the first half of 2021 in humans nor reports that showed a more rapid replication of Alpha variant in human reconstituted bronchial epithelium. This study highlights the need to combine data from different laboratories using various animal models to decipher the biological properties of newly emerging SARS-CoV-2 variants., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
31. Hydroxychloroquine and azithromycin used alone or combined are not effective against SARS-CoV-2 ex vivo and in a hamster model.
- Author
-
Cochin M, Touret F, Driouich JS, Moureau G, Petit PR, Laprie C, Solas C, de Lamballerie X, and Nougairède A
- Subjects
- Animals, Anti-Infective Agents administration & dosage, Anti-Infective Agents therapeutic use, Azithromycin administration & dosage, Azithromycin pharmacokinetics, Azithromycin therapeutic use, Bronchi cytology, Bronchi virology, Chlorocebus aethiops, Cricetinae, Disease Models, Animal, Drug Therapy, Combination, Female, Humans, Hydroxychloroquine administration & dosage, Hydroxychloroquine therapeutic use, Lung pathology, Mesocricetus, Middle Aged, Plasma virology, Real-Time Polymerase Chain Reaction, Vero Cells, Anti-Infective Agents pharmacology, Azithromycin pharmacology, Hydroxychloroquine pharmacology, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
Drug repositioning has been used extensively since the beginning of the COVID-19 pandemic in an attempt to identify antiviral molecules for use in human therapeutics. Hydroxychloroquine and azithromycin have shown inhibitory activity against SARS-CoV-2 replication in different cell lines. Based on such in vitro data and despite the weakness of preclinical assessment, many clinical trials were set up using these molecules. In the present study, we show that hydroxychloroquine and azithromycin alone or combined does not block SARS-CoV-2 replication in human bronchial airway epithelia. When tested in a Syrian hamster model, hydroxychloroquine and azithromycin administrated alone or combined displayed no significant effect on viral replication, clinical course of the disease and lung impairments, even at high doses. Hydroxychloroquine quantification in lung tissues confirmed strong exposure to the drug, above in vitro inhibitory concentrations. Overall, this study does not support the use of hydroxychloroquine and azithromycin as antiviral drugs for the treatment of SARS-CoV-2 infections., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
32. Niclosamide shows strong antiviral activity in a human airway model of SARS-CoV-2 infection and a conserved potency against the Alpha (B.1.1.7), Beta (B.1.351) and Delta variant (B.1.617.2).
- Author
-
Weiss A, Touret F, Baronti C, Gilles M, Hoen B, Nougairède A, de Lamballerie X, and Sommer MOA
- Subjects
- Animals, Caco-2 Cells, Chlorocebus aethiops, Humans, Inhibitory Concentration 50, Respiratory Mucosa virology, Vero Cells, Antiviral Agents therapeutic use, Niclosamide therapeutic use, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
SARS-CoV-2 variants are emerging with potential increased transmissibility highlighting the great unmet medical need for new therapies. Niclosamide is a potent anti-SARS-CoV-2 agent that has advanced in clinical development. We validate the potent antiviral efficacy of niclosamide in a SARS-CoV-2 human airway model. Furthermore, niclosamide remains its potency against the D614G, Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) variants. Our data further support the potent anti-SARS-CoV-2 properties of niclosamide and highlights its great potential as a therapeutic agent for COVID-19., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: MOAS and AW are shareholders or benefit from an employee incentive scheme in UNION therapeutics. This does not alter our adherence to PLOS ONE policies on sharing data and materials. Further, UNION had no role in study design, data collection, data analysis, data interpretation, or writing of the report. All other authors have nothing to disclose.
- Published
- 2021
- Full Text
- View/download PDF
33. Publisher Correction: A pan-serotype dengue virus inhibitor targeting the NS3-NS4B interaction.
- Author
-
Kaptein SJF, Goethals O, Kiemel D, Marchand A, Kesteleyn B, Bonfanti JF, Bardiot D, Stoops B, Jonckers THM, Dallmeier K, Geluykens P, Thys K, Crabbe M, Chatel-Chaix L, Münster M, Querat G, Touret F, de Lamballerie X, Raboisson P, Simmen K, Chaltin P, Bartenschlager R, Van Loock M, and Neyts J
- Published
- 2021
- Full Text
- View/download PDF
34. A pan-serotype dengue virus inhibitor targeting the NS3-NS4B interaction.
- Author
-
Kaptein SJF, Goethals O, Kiemel D, Marchand A, Kesteleyn B, Bonfanti JF, Bardiot D, Stoops B, Jonckers THM, Dallmeier K, Geluykens P, Thys K, Crabbe M, Chatel-Chaix L, Münster M, Querat G, Touret F, de Lamballerie X, Raboisson P, Simmen K, Chaltin P, Bartenschlager R, Van Loock M, and Neyts J
- Subjects
- Animals, Antiviral Agents pharmacokinetics, Antiviral Agents therapeutic use, Dengue drug therapy, Dengue Virus genetics, Dengue Virus metabolism, Disease Models, Animal, Female, Male, Membrane Proteins antagonists & inhibitors, Mice, RNA Helicases antagonists & inhibitors, RNA Helicases metabolism, Serine Endopeptidases metabolism, Viral Load drug effects, Viral Nonstructural Proteins antagonists & inhibitors, Viremia drug therapy, Viremia virology, Virus Replication drug effects, Antiviral Agents pharmacology, Dengue virology, Dengue Virus classification, Dengue Virus drug effects, Membrane Proteins metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Dengue virus causes approximately 96 million symptomatic infections annually, manifesting as dengue fever or occasionally as severe dengue
1,2 . There are no antiviral agents available to prevent or treat dengue. Here, we describe a highly potent dengue virus inhibitor (JNJ-A07) that exerts nanomolar to picomolar activity against a panel of 21 clinical isolates that represent the natural genetic diversity of known genotypes and serotypes. The molecule has a high barrier to resistance and prevents the formation of the viral replication complex by blocking the interaction between two viral proteins (NS3 and NS4B), thus revealing a previously undescribed mechanism of antiviral action. JNJ-A07 has a favourable pharmacokinetic profile that results in outstanding efficacy against dengue virus infection in mouse infection models. Delaying start of treatment until peak viraemia results in a rapid and significant reduction in viral load. An analogue is currently in further development., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
35. A Bioluminescent 3CL Pro Activity Assay to Monitor SARS-CoV-2 Replication and Identify Inhibitors.
- Author
-
Mathieu C, Touret F, Jacquemin C, Janin YL, Nougairède A, Brailly M, Mazelier M, Décimo D, Vasseur V, Hans A, Valle-Casuso JC, de Lamballerie X, Horvat B, André P, Si-Tahar M, Lotteau V, and Vidalain PO
- Subjects
- Animals, Antiviral Agents pharmacology, Cell Line, Chlorocebus aethiops, Drug Discovery, Drug Evaluation, Preclinical, Enzyme Activation, HEK293 Cells, Humans, Luciferases, Firefly metabolism, Nasal Mucosa virology, Pyrazolones pharmacology, Pyridones pharmacology, SARS-CoV-2 metabolism, Vero Cells, Virus Internalization drug effects, Antiviral Agents isolation & purification, Biosensing Techniques methods, Coronavirus 3C Proteases metabolism, SARS-CoV-2 physiology, Virus Replication drug effects
- Abstract
Our therapeutic arsenal against viruses is very limited and the current pandemic of SARS-CoV-2 highlights the critical need for effective antivirals against emerging coronaviruses. Cellular assays allowing a precise quantification of viral replication in high-throughput experimental settings are essential to the screening of chemical libraries and the selection of best antiviral chemical structures. To develop a reporting system for SARS-CoV-2 infection, we generated cell lines expressing a firefly luciferase maintained in an inactive form by a consensus cleavage site for the viral protease 3CL
Pro of coronaviruses, so that the luminescent biosensor is turned on upon 3CLPro expression or SARS-CoV-2 infection. This cellular assay was used to screen a metabolism-oriented library of 492 compounds to identify metabolic vulnerabilities of coronaviruses for developing innovative therapeutic strategies. In agreement with recent reports, inhibitors of pyrimidine biosynthesis were found to prevent SARS-CoV-2 replication. Among the top hits, we also identified the NADPH oxidase (NOX) inhibitor Setanaxib. The anti-SARS-CoV-2 activity of Setanaxib was further confirmed using ACE2-expressing human pulmonary cells Beas2B as well as human primary nasal epithelial cells. Altogether, these results validate our cell-based functional assay and the interest of screening libraries of different origins to identify inhibitors of SARS-CoV-2 for drug repurposing or development.- Published
- 2021
- Full Text
- View/download PDF
36. Preclinical evaluation of Imatinib does not support its use as an antiviral drug against SARS-CoV-2.
- Author
-
Touret F, Driouich JS, Cochin M, Petit PR, Gilles M, Barthélémy K, Moureau G, Mahon FX, Malvy D, Solas C, de Lamballerie X, and Nougairède A
- Subjects
- Animals, COVID-19 epidemiology, COVID-19 virology, Cell Line, Chlorocebus aethiops, Drug Evaluation, Preclinical, Drug Repositioning, Enzyme Inhibitors pharmacology, Epithelium, Female, Humans, Lung pathology, Male, Mesocricetus, Vero Cells, Virus Replication drug effects, Antiviral Agents pharmacology, Imatinib Mesylate pharmacology, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
Following the emergence of SARS-CoV-2, the search for an effective and rapidly available treatment was initiated worldwide based on repurposing of available drugs. Previous reports described the antiviral activity of certain tyrosine kinase inhibitors (TKIs) targeting the Abelson kinase 2 against pathogenic coronaviruses. Imatinib, one of them, has more than twenty years of safe utilization for the treatment of hematological malignancies. In this context, Imatinib was rapidly evaluated in clinical trials against Covid-19. Here, we present the pre-clinical evaluation of imatinib in multiple models. Our results indicated that imatinib and another TKI, the masitinib, exhibit an antiviral activity in VeroE6 cells. However, imatinib was inactive in a reconstructed bronchial human airway epithelium model. In vivo, imatinib therapy failed to impair SARS-CoV-2 replication in a golden Syrian hamster model despite high concentrations in plasma and in the lung. Overall, these results do not support the use of imatinib and similar TKIs as antivirals in the treatment of Covid-19., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. Replicative Fitness of a SARS-CoV-2 20I/501Y.V1 Variant from Lineage B.1.1.7 in Human Reconstituted Bronchial Epithelium.
- Author
-
Touret F, Luciani L, Baronti C, Cochin M, Driouich JS, Gilles M, Thirion L, Nougairède A, and de Lamballerie X
- Subjects
- Animals, COVID-19 pathology, Caco-2 Cells, Cell Line, Chlorocebus aethiops, Humans, Respiratory Mucosa virology, Vero Cells, Viral Load, COVID-19 transmission, Genetic Fitness genetics, SARS-CoV-2 genetics, SARS-CoV-2 growth & development, Virus Replication genetics
- Abstract
Since its emergence in 2019, circulating populations of the new coronavirus (CoV) continuously acquired genetic diversity. At the end of 2020, a variant named 20I/501Y.V1 (lineage B.1.1.7) emerged and replaced other circulating strains in several regions. This phenomenon has been poorly associated with biological evidence that this variant and the original strain exhibit different phenotypic characteristics. Here, we analyze the replication ability of this new variant in different cellular models using for comparison an ancestral D614G European strain (lineage B1). Results from comparative replication kinetics experiments in vitro and in a human reconstituted bronchial epithelium showed no difference. However, when both viruses were put in competition in human reconstituted bronchial epithelium, the 20I/501Y.V1 variant outcompeted the ancestral strain. All together, these findings demonstrate that this new variant replicates more efficiently and may contribute to a better understanding of the progressive replacement of circulating strains by the severe acute respiratory CoV-2 (SARS-CoV-2) 20I/501Y.V1 variant. IMPORTANCE The emergence of several SARS-CoV-2 variants raised numerous questions concerning the future course of the pandemic. We are currently observing a replacement of the circulating viruses by the variant from the United Kingdom known as 20I/501Y.V1, from the B.1.1.7 lineage, but there is little biological evidence that this new variant exhibits a different phenotype. In the present study, we used different cellular models to assess the replication ability of the 20I/501Y.V1 variant. Our results showed that this variant replicates more efficiently in human reconstituted bronchial epithelium, which may explain why it spreads so rapidly in human populations.
- Published
- 2021
- Full Text
- View/download PDF
38. Favipiravir antiviral efficacy against SARS-CoV-2 in a hamster model.
- Author
-
Driouich JS, Cochin M, Lingas G, Moureau G, Touret F, Petit PR, Piorkowski G, Barthélémy K, Laprie C, Coutard B, Guedj J, de Lamballerie X, Solas C, and Nougairède A
- Subjects
- Animals, COVID-19 virology, Chlorocebus aethiops, Cricetinae, Disease Models, Animal, Female, Genome, Viral, Lung virology, Mesocricetus, SARS-CoV-2 genetics, Vero Cells, Viral Load drug effects, Amides pharmacology, Antiviral Agents pharmacology, Pyrazines pharmacology, SARS-CoV-2 drug effects, COVID-19 Drug Treatment
- Abstract
Despite no or limited pre-clinical evidence, repurposed drugs are massively evaluated in clinical trials to palliate the lack of antiviral molecules against SARS-CoV-2. Here we use a Syrian hamster model to assess the antiviral efficacy of favipiravir, understand its mechanism of action and determine its pharmacokinetics. When treatment is initiated before or simultaneously to infection, favipiravir has a strong dose effect, leading to reduction of infectious titers in lungs and clinical alleviation of the disease. Antiviral effect of favipiravir correlates with incorporation of a large number of mutations into viral genomes and decrease of viral infectivity. Antiviral efficacy is achieved with plasma drug exposure comparable with those previously found during human clinical trials. Notably, the highest dose of favipiravir tested is associated with signs of toxicity in animals. Thereby, pharmacokinetic and tolerance studies are required to determine whether similar effects can be safely achieved in humans.
- Published
- 2021
- Full Text
- View/download PDF
39. SARS-CoV-2 viral dynamics in non-human primates.
- Author
-
Gonçalves A, Maisonnasse P, Donati F, Albert M, Behillil S, Contreras V, Naninck T, Marlin R, Solas C, Pizzorno A, Lemaitre J, Kahlaoui N, Terrier O, Ho Tsong Fang R, Enouf V, Dereuddre-Bosquet N, Brisebarre A, Touret F, Chapon C, Hoen B, Lina B, Rosa Calatrava M, de Lamballerie X, Mentré F, Le Grand R, van der Werf S, and Guedj J
- Subjects
- Animals, Antiviral Agents pharmacology, Basic Reproduction Number, COVID-19 blood, COVID-19 prevention & control, Cytokines blood, Disease Models, Animal, Nasopharynx virology, SARS-CoV-2 drug effects, Trachea virology, Viral Load, Virus Replication drug effects, COVID-19 virology, Macaca fascicularis virology, SARS-CoV-2 physiology
- Abstract
Non-human primates infected with SARS-CoV-2 exhibit mild clinical signs. Here we used a mathematical model to characterize in detail the viral dynamics in 31 cynomolgus macaques for which nasopharyngeal and tracheal viral load were frequently assessed. We identified that infected cells had a large burst size (>104 virus) and a within-host reproductive basic number of approximately 6 and 4 in nasopharyngeal and tracheal compartment, respectively. After peak viral load, infected cells were rapidly lost with a half-life of 9 hours, with no significant association between cytokine elevation and clearance, leading to a median time to viral clearance of 10 days, consistent with observations in mild human infections. Given these parameter estimates, we predict that a prophylactic treatment blocking 90% of viral production or viral infection could prevent viral growth. In conclusion, our results provide estimates of SARS-CoV-2 viral kinetic parameters in an experimental model of mild infection and they provide means to assess the efficacy of future antiviral treatments., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: A.G. PhD grant has been provided by ROCHE Company. J.G. has worked as consultant for ROCHE Company.
- Published
- 2021
- Full Text
- View/download PDF
40. Synthesis and Antiviral Evaluation of (1,4-Disubstituted-1,2,3-Triazol)-( E )-2-Methyl-but-2-Enyl Nucleoside Phosphonate Prodrugs.
- Author
-
Abuduaini T, Roy V, Marlet J, Gaudy-Graffin C, Brand D, Baronti C, Touret F, Coutard B, McBrayer TR, Schinazi RF, and Agrofoglio LA
- Subjects
- Alkenes chemistry, Animals, Cell Line, Tumor, Chlorocebus aethiops, HIV-1 drug effects, Hepatitis B virus drug effects, Humans, Magnetic Resonance Spectroscopy, Methylation, SARS-CoV-2 drug effects, Structure-Activity Relationship, Triazoles chemistry, Vero Cells, Antiviral Agents chemical synthesis, Antiviral Agents pharmacology, Azo Compounds chemistry, Nucleosides chemistry, Organophosphonates chemistry, Prodrugs chemical synthesis, Prodrugs pharmacology
- Abstract
A series of hitherto unknown (1,4-disubstituted-1,2,3-triazol)-( E )-2-methyl-but-2-enyl nucleosides phosphonate prodrugs bearing 4-substituted-1,2,3-triazoles were prepared in a straight approach through an olefin acyclic cross metathesis as the key synthetic step. All novel compounds were evaluated for their antiviral activities against HBV, HIV and SARS-CoV-2. Among these molecules, only compound 15j , a hexadecyloxypropyl (HDP)/( isopropyloxycarbonyl -oxymethyl)-ester (POC) prodrug, showed activity against HBV in Huh7 cell cultures with 62% inhibition at 10 μM, without significant cytotoxicity (IC
50 = 66.4 μM in HepG2 cells, IC50 = 43.1 μM in HepG2 cells) at 10 μM.- Published
- 2021
- Full Text
- View/download PDF
41. Drugs against SARS-CoV-2: What do we know about their mode of action?
- Author
-
Valle C, Martin B, Touret F, Shannon A, Canard B, Guillemot JC, Coutard B, and Decroly E
- Subjects
- Combined Modality Therapy, Disease Management, Disease Susceptibility, Drug Development, Drug Repositioning, Host-Pathogen Interactions, Humans, Treatment Outcome, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, COVID-19 virology, SARS-CoV-2 drug effects, SARS-CoV-2 physiology, COVID-19 Drug Treatment
- Abstract
The health emergency caused by the recent Covid-19 pandemic highlights the need to identify effective treatments against the virus causing this disease (SARS-CoV-2). The first clinical trials have been testing repurposed drugs that show promising anti-SARS-CoV-2 effects in cultured cells. Although more than 2400 clinical trials are already under way, the actual number of tested compounds is still limited to approximately 20, alone or in combination. In addition, knowledge on their mode of action (MoA) is currently insufficient. Their first results reveal some inconsistencies and contradictory results and suggest that cohort size and quality of the control arm are two key issues for obtaining rigorous and conclusive results. Moreover, the observed discrepancies might also result from differences in the clinical inclusion criteria, including the possibility of early treatment that may be essential for therapy efficacy in patients with Covid-19. Importantly, efforts should also be made to test new compounds with a documented MoA against SARS-CoV-2 in clinical trials. Successful treatment will probably be based on multitherapies with antiviral compounds that target different steps of the virus life cycle. Moreover, a multidisciplinary approach that combines artificial intelligence, compound docking, and robust in vitro and in vivo assays will accelerate the development of new antiviral molecules. Finally, large retrospective studies on hospitalized patients are needed to evaluate the different treatments with robust statistical tools and to identify the best treatment for each Covid-19 stage. This review describes different candidate antiviral strategies for Covid-19, by focusing on their mechanism of action., (© 2020 John Wiley & Sons, Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
42. Rapid incorporation of Favipiravir by the fast and permissive viral RNA polymerase complex results in SARS-CoV-2 lethal mutagenesis.
- Author
-
Shannon A, Selisko B, Le NT, Huchting J, Touret F, Piorkowski G, Fattorini V, Ferron F, Decroly E, Meier C, Coutard B, Peersen O, and Canard B
- Subjects
- Amides pharmacokinetics, Animals, Antiviral Agents pharmacokinetics, COVID-19, Chlorocebus aethiops, Coronavirus Infections virology, Coronavirus RNA-Dependent RNA Polymerase, Models, Molecular, Mutagenesis drug effects, Pandemics, Pneumonia, Viral virology, Pyrazines pharmacokinetics, RNA, Viral genetics, RNA, Viral metabolism, RNA-Dependent RNA Polymerase chemistry, RNA-Dependent RNA Polymerase metabolism, SARS-CoV-2, Sequence Analysis, Vero Cells, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins metabolism, Virus Replication drug effects, COVID-19 Drug Treatment, Amides pharmacology, Antiviral Agents pharmacology, Betacoronavirus drug effects, Betacoronavirus genetics, Coronavirus Infections drug therapy, Pneumonia, Viral drug therapy, Pyrazines pharmacology
- Abstract
The ongoing Corona Virus Disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has emphasized the urgent need for antiviral therapeutics. The viral RNA-dependent-RNA-polymerase (RdRp) is a promising target with polymerase inhibitors successfully used for the treatment of several viral diseases. We demonstrate here that Favipiravir predominantly exerts an antiviral effect through lethal mutagenesis. The SARS-CoV RdRp complex is at least 10-fold more active than any other viral RdRp known. It possesses both unusually high nucleotide incorporation rates and high-error rates allowing facile insertion of Favipiravir into viral RNA, provoking C-to-U and G-to-A transitions in the already low cytosine content SARS-CoV-2 genome. The coronavirus RdRp complex represents an Achilles heel for SARS-CoV, supporting nucleoside analogues as promising candidates for the treatment of COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
43. Hydroxychloroquine use against SARS-CoV-2 infection in non-human primates.
- Author
-
Maisonnasse P, Guedj J, Contreras V, Behillil S, Solas C, Marlin R, Naninck T, Pizzorno A, Lemaitre J, Gonçalves A, Kahlaoui N, Terrier O, Fang RHT, Enouf V, Dereuddre-Bosquet N, Brisebarre A, Touret F, Chapon C, Hoen B, Lina B, Calatrava MR, van der Werf S, de Lamballerie X, and Le Grand R
- Subjects
- Animals, Azithromycin pharmacology, Azithromycin therapeutic use, COVID-19, Chlorocebus aethiops, Coronavirus Infections pathology, Coronavirus Infections physiopathology, Cytokines blood, Disease Models, Animal, Female, Humans, Hydroxychloroquine pharmacokinetics, Hydroxychloroquine pharmacology, In Vitro Techniques, Kinetics, Macaca fascicularis, Male, Pandemics, Pneumonia, Viral pathology, Pneumonia, Viral physiopathology, Pre-Exposure Prophylaxis, Respiratory Mucosa cytology, Respiratory Mucosa drug effects, Respiratory Mucosa virology, SARS-CoV-2, Time Factors, Treatment Failure, Vero Cells, Viral Load drug effects, COVID-19 Drug Treatment, Betacoronavirus drug effects, Coronavirus Infections drug therapy, Coronavirus Infections virology, Hydroxychloroquine therapeutic use, Pneumonia, Viral drug therapy, Pneumonia, Viral virology
- Abstract
Coronavirus disease 2019 (COVID-19) has rapidly become a global pandemic and no antiviral drug or vaccine is yet available for the treatment of this disease
1-3 . Several clinical studies are ongoing to evaluate the efficacy of repurposed drugs that have demonstrated antiviral efficacy in vitro. Among these candidates, hydroxychloroquine (HCQ) has been given to thousands of individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-the virus that causes COVID-19-worldwide but there is no definitive evidence that HCQ is effective for treating COVID-194-7 . Here we evaluated the antiviral activity of HCQ both in vitro and in SARS-CoV-2-infected macaques. HCQ showed antiviral activity in African green monkey kidney cells (Vero E6) but not in a model of reconstituted human airway epithelium. In macaques, we tested different treatment strategies in comparison to a placebo treatment, before and after peak viral load, alone or in combination with azithromycin (AZTH). Neither HCQ nor the combination of HCQ and AZTH showed a significant effect on viral load in any of the analysed tissues. When the drug was used as a pre-exposure prophylaxis treatment, HCQ did not confer protection against infection with SARS-CoV-2. Our findings do not support the use of HCQ, either alone or in combination with AZTH, as an antiviral drug for the treatment of COVID-19 in humans.- Published
- 2020
- Full Text
- View/download PDF
44. Prolonged Infectivity of SARS-CoV-2 in Fomites.
- Author
-
Pastorino B, Touret F, Gilles M, de Lamballerie X, and Charrel RN
- Subjects
- Aluminum analysis, COVID-19, Coronavirus Infections virology, Glass analysis, Humans, Pandemics, Plastics analysis, Pneumonia, Viral virology, SARS-CoV-2, Time Factors, Betacoronavirus isolation & purification, Coronavirus Infections transmission, Disease Transmission, Infectious, Fomites virology, Pneumonia, Viral transmission
- Abstract
We spotted severe acute respiratory syndrome coronavirus 2 on polystyrene plastic, aluminum, and glass for 96 hours with and without bovine serum albumin (3 g/L). We observed a steady infectivity (<1 log
10 drop) on plastic, a 3.5 log10 decrease on glass, and a 6 log10 drop on aluminum. The presence of proteins noticeably prolonged infectivity.- Published
- 2020
- Full Text
- View/download PDF
45. In vitro screening of a FDA approved chemical library reveals potential inhibitors of SARS-CoV-2 replication.
- Author
-
Touret F, Gilles M, Barral K, Nougairède A, van Helden J, Decroly E, de Lamballerie X, and Coutard B
- Subjects
- Animals, Antiviral Agents chemistry, Antiviral Agents metabolism, Antiviral Agents pharmacology, Betacoronavirus isolation & purification, COVID-19, Caco-2 Cells, Cell Survival drug effects, Chlorocebus aethiops, Coronavirus Infections pathology, Coronavirus Infections virology, Drug Approval, Drug Evaluation, Preclinical, Drug Repositioning, Humans, Pandemics, Pneumonia, Viral pathology, Pneumonia, Viral virology, SARS-CoV-2, Small Molecule Libraries metabolism, Small Molecule Libraries pharmacology, Vero Cells, Virus Replication drug effects, Betacoronavirus physiology, Small Molecule Libraries chemistry
- Abstract
A novel coronavirus, named SARS-CoV-2, emerged in 2019 in China and rapidly spread worldwide. As no approved therapeutics exists to treat COVID-19, the disease associated to SARS-Cov-2, there is an urgent need to propose molecules that could quickly enter into clinics. Repurposing of approved drugs is a strategy that can bypass the time-consuming stages of drug development. In this study, we screened the PRESTWICK CHEMICAL LIBRARY composed of 1,520 approved drugs in an infected cell-based assay. The robustness of the screen was assessed by the identification of drugs that already demonstrated in vitro antiviral effect against SARS-CoV-2. Thereby, 90 compounds were identified as positive hits from the screen and were grouped according to their chemical composition and their known therapeutic effect. Then EC50 and CC50 were determined for a subset of 15 compounds from a panel of 23 selected drugs covering the different groups. Eleven compounds such as macrolides antibiotics, proton pump inhibitors, antiarrhythmic agents or CNS drugs emerged showing antiviral potency with 2 < EC50 ≤ 20 µM. By providing new information on molecules inhibiting SARS-CoV-2 replication in vitro, this study provides information for the selection of drugs to be further validated in vivo. Disclaimer: This study corresponds to the early stages of antiviral development and the results do not support by themselves the use of the selected drugs to treat SARS-CoV-2 infection.
- Published
- 2020
- Full Text
- View/download PDF
46. Dose Rationale for Favipiravir Use in Patients Infected With SARS-CoV-2.
- Author
-
Eloy P, Solas C, Touret F, Mentré F, Malvy D, de Lamballerie X, and Guedj J
- Subjects
- Amides, Betacoronavirus, COVID-19, Humans, Pyrazines, SARS-CoV-2, Coronavirus Infections, Pandemics, Pneumonia, Viral, Severe acute respiratory syndrome-related coronavirus
- Published
- 2020
- Full Text
- View/download PDF
47. Heat Inactivation of Different Types of SARS-CoV-2 Samples: What Protocols for Biosafety, Molecular Detection and Serological Diagnostics?
- Author
-
Pastorino B, Touret F, Gilles M, de Lamballerie X, and Charrel RN
- Subjects
- Antibodies, Neutralizing immunology, COVID-19, Containment of Biohazards standards, Coronavirus Infections diagnosis, Coronavirus Infections prevention & control, Enzyme-Linked Immunosorbent Assay, Hot Temperature, Humans, Neutralization Tests, Pandemics prevention & control, Pneumonia, Viral diagnosis, Pneumonia, Viral prevention & control, SARS-CoV-2, Serologic Tests standards, Betacoronavirus immunology, Containment of Biohazards methods, Coronavirus Infections virology, Pneumonia, Viral virology, Serologic Tests methods, Virus Inactivation
- Abstract
Standard precautions to minimize the risk of SARS-CoV-2 transmission implies that infected cell cultures and clinical specimens may undergo some sort of inactivation to reduce or abolish infectivity. We evaluated three heat inactivation protocols (56 °C-30 min, 60 °C-60 min and 92 °C-15 min) on SARS-CoV-2 using (i) infected cell culture supernatant, (ii) virus-spiked human sera (iii) and nasopharyngeal samples according to the recommendations of the European norm NF EN 14476-A2. Regardless of the protocol and the type of samples, a 4 Log
10 TCID50 reduction was observed. However, samples containing viral loads > 6 Log10 TCID50 were still infectious after 56 °C-30 min and 60 °C-60 min, although infectivity was < 10 TCID50 . The protocols 56 °C-30 min and 60 °C-60 min had little influence on the RNA copies detection, whereas 92 °C-15 min drastically reduced the limit of detection, which suggests that this protocol should be avoided for inactivation ahead of molecular diagnostics. Lastly, 56 °C-30 min treatment of serum specimens had a negligible influence on the results of IgG detection using a commercial ELISA test, whereas a drastic decrease in neutralizing titers was observed.- Published
- 2020
- Full Text
- View/download PDF
48. Evaluation of Chemical Protocols for Inactivating SARS-CoV-2 Infectious Samples.
- Author
-
Pastorino B, Touret F, Gilles M, Luciani L, de Lamballerie X, and Charrel RN
- Subjects
- Animals, Betacoronavirus genetics, Betacoronavirus isolation & purification, Betacoronavirus physiology, COVID-19, Cell Culture Techniques methods, Chlorocebus aethiops, Containment of Biohazards methods, Containment of Biohazards standards, Humans, Nasopharynx virology, Pandemics, RNA, Viral isolation & purification, SARS-CoV-2, Specimen Handling methods, Vero Cells, Viral Load methods, Betacoronavirus drug effects, Coronavirus Infections virology, Pneumonia, Viral virology, Virus Inactivation drug effects
- Abstract
Clinical samples collected in coronavirus disease 19 (COVID-19), patients are commonly manipulated in biosafety level 2 laboratories for molecular diagnostic purposes. Here, we tested French norm NF-EN-14476+A2 derived from European standard EN-14885 to assess the risk of manipulating infectious viruses prior to RNA extraction. SARS-CoV-2 cell-culture supernatant and nasopharyngeal samples (virus-spiked samples and clinical samples collected in COVID-19 patients) were used to measure the reduction of infectivity after 10 minute contact with lysis buffer containing various detergents and chaotropic agents. A total of thirteen protocols were evaluated. Two commercially available formulations showed the ability to reduce infectivity by at least 6 log 10, whereas others proved less effective.
- Published
- 2020
- Full Text
- View/download PDF
49. Favipiravir strikes the SARS-CoV-2 at its Achilles heel, the RNA polymerase.
- Author
-
Shannon A, Selisko B, Le N, Huchting J, Touret F, Piorkowski G, Fattorini V, Ferron F, Decroly E, Meier C, Coutard B, Peersen O, and Canard B
- Abstract
The ongoing Corona Virus Disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has emphasized the urgent need for antiviral therapeutics. The viral RNA-dependent-RNA-polymerase (RdRp) is a promising target with polymerase inhibitors successfully used for the treatment of several viral diseases. Here we show that Favipiravir exerts an antiviral effect as a nucleotide analogue through a combination of chain termination, slowed RNA synthesis and lethal mutagenesis. The SARS-CoV RdRp complex is at least 10-fold more active than any other viral RdRp known. It possesses both unusually high nucleotide incorporation rates and high-error rates allowing facile insertion of Favipiravir into viral RNA, provoking C-to-U and G-to-A transitions in the already low cytosine content SARS-CoV-2 genome. The coronavirus RdRp complex represents an Achilles heel for SARS-CoV, supporting nucleoside analogues as promising candidates for the treatment of COVID-19., Competing Interests: Competing interests: Authors declare no competing interests.
- Published
- 2020
- Full Text
- View/download PDF
50. Of chloroquine and COVID-19.
- Author
-
Touret F and de Lamballerie X
- Subjects
- Antiviral Agents adverse effects, Antiviral Agents pharmacology, Antiviral Agents standards, Betacoronavirus drug effects, COVID-19, China, Chloroquine adverse effects, Chloroquine pharmacology, Chloroquine standards, Clinical Trials as Topic standards, Humans, Hydroxychloroquine adverse effects, Hydroxychloroquine pharmacology, Hydroxychloroquine standards, Hydroxychloroquine therapeutic use, Pandemics, SARS-CoV-2, Antiviral Agents therapeutic use, Chloroquine therapeutic use, Coronavirus Infections drug therapy, Pneumonia, Viral drug therapy
- Abstract
Recent publications have brought attention to the possible benefit of chloroquine, a broadly used antimalarial drug, in the treatment of patients infected by the novel emerged coronavirus (SARS-CoV-2). The scientific community should consider this information in light of previous experiments with chloroquine in the field of antiviral research., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.