Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Nature ; 615(7953): 678-686, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922586

RESUMEN

Dengue is a major health threat and the number of symptomatic infections caused by the four dengue serotypes is estimated to be 96 million1 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.


Asunto(s)
Antivirales , Virus del Dengue , Dengue , Primates , Proteínas no Estructurales Virales , Animales , Humanos , Ratones , Antivirales/efectos adversos , Antivirales/farmacología , Antivirales/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Dengue/tratamiento farmacológico , Dengue/prevención & control , Dengue/virología , Virus del Dengue/clasificación , Virus del Dengue/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral , Técnicas In Vitro , Terapia Molecular Dirigida , Primates/virología , Unión Proteica/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
2.
Nature ; 598(7881): 504-509, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616043

RESUMEN

Dengue virus causes approximately 96 million symptomatic infections annually, manifesting as dengue fever or occasionally as severe dengue1,2. 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.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/clasificación , Virus del Dengue/efectos de los fármacos , Dengue/virología , Proteínas de la Membrana/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/farmacocinética , Antivirales/uso terapéutico , Dengue/tratamiento farmacológico , Virus del Dengue/genética , Virus del Dengue/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/metabolismo , Serina Endopeptidasas/metabolismo , Carga Viral/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Viremia/tratamiento farmacológico , Viremia/virología , Replicación Viral/efectos de los fármacos
4.
Artículo en Inglés | MEDLINE | ID: mdl-32340991

RESUMEN

Despite the worldwide reemergence of the chikungunya virus (CHIKV) and the high morbidity associated with CHIKV infections, there is no approved vaccine or antiviral treatment available. Here, we aimed to identify the target of a novel class of CHIKV inhibitors, i.e., the CHVB series. CHVB compounds inhibit the in vitro replication of CHIKV isolates with 50% effective concentrations in the low-micromolar range. A CHVB-resistant variant (CHVBres) was selected that carried two mutations in the gene encoding nsP1 (responsible for viral RNA capping), one mutation in nsP2, and one mutation in nsP3. Reverse genetics studies demonstrated that both nsP1 mutations were necessary and sufficient to achieve ∼18-fold resistance, suggesting that CHVB targets viral mRNA capping. Interestingly, CHVBres was cross-resistant to the previously described CHIKV capping inhibitors from the MADTP series, suggesting they share a similar mechanism of action. In enzymatic assays, CHVB inhibited the methyltransferase and guanylyltransferase activities of alphavirus nsP1 proteins. To conclude, we identified a class of CHIKV inhibitors that targets the viral capping machinery. The potent anti-CHIKV activity makes this chemical scaffold a potential candidate for CHIKV drug development.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Animales , Antivirales/farmacología , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/genética , Chlorocebus aethiops , Células Vero , Proteínas no Estructurales Virales , Replicación Viral
5.
J Gen Virol ; 98(11): 2676-2688, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29022865

RESUMEN

Toscana virus (TOSV) is an arthropod-borne phlebovirus within the family Phenuiviridae in the order Bunyavirales. It seems to be an important agent of human meningoencephalitis in the warm season in the Mediterranean area. Because the polymerase of Bunyavirales lacks a capping activity, it cleaves short-capped RNA leaders derived from the host cell, and uses them to initiate viral mRNA synthesis. To determine the size and nucleotide composition of the host-derived RNA leaders, and to elucidate the first steps of TOSV transcription initiation, we performed a high-throughput sequencing of the 5' end of TOSV mRNAs in infected cells at different times post-infection. Our results indicated that the viral polymerase cleaved the host-capped RNA leaders within a window of 11-16 nucleotides. A single population of cellular mRNAs could be cleaved at different sites to prime the synthesis of several viral mRNA species. The majority of the mRNA resulted from direct priming, but we observed mRNAs resulting from several rounds of prime-and-realign events. Our data suggest that the different rounds of the prime-and-realign mechanism result from the blocking of the template strand in a static position in the active site, leading to the slippage of the nascent strand by two nucleotides when the growing duplex is sorted out from the active site. To minimize this rate-limiting step, TOSV polymerase cleaves preferentially capped RNA leaders after GC, so as to greatly reduce the number of cycles of priming and realignment, and facilitate the separation of the growing duplex.


Asunto(s)
ARN Mensajero/biosíntesis , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/metabolismo , Virus de Nápoles de la Fiebre de la Mosca de los Arenales/enzimología , Virus de Nápoles de la Fiebre de la Mosca de los Arenales/genética , Transcripción Genética , Células Cultivadas , Células Epiteliales/virología , Humanos , ARN Mensajero/química , ARN Mensajero/genética , ARN Viral/química , ARN Viral/genética
6.
Crit Rev Microbiol ; 43(6): 753-778, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28418734

RESUMEN

Bunyaviridae family is the largest and most diverse family of RNA viruses. It has more than 350 members divided into five genera: Orthobunyavirus, Phlebovirus, Nairovirus, Hantavirus, and Tospovirus. They are present in the five continents, causing recurrent epidemics, epizootics, and considerable agricultural loss. The genome of bunyaviruses is divided into three segments of negative single-stranded RNA according to their relative size: L (Large), M (Medium) and S (Small) segment. Bunyaviridae RNA-dependent RNA polymerase (RdRp) is encoded by the L segment, and is in charge of the replication and transcription of the viral RNA in the cytoplasm of the infected cell. Viral RdRps share a characteristic right hand-like structure with three subdomains: finger, palm, and thumb subdomains that define the formation of the catalytic cavity. In addition to the N-terminal endonuclease domain, eight conserved motifs (A-H) have been identified in the RdRp of Bunyaviridae. In this review, we have summarized the recent insights from the structural and functional studies of RdRp to understand the roles of different motifs shared by RdRps, the mechanism of viral RNA replication, genome segment packaging by the nucleoprotein, cap-snatching, mRNA transcription, and other RNA mechanisms of bunyaviruses.


Asunto(s)
Bunyaviridae/genética , Bunyaviridae/metabolismo , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/genética , Ensamble de Virus/genética , Replicación Viral/genética , Secuencia de Aminoácidos/genética , Infecciones por Bunyaviridae/virología , Genoma Viral/genética , ARN Viral/genética
7.
J Gen Virol ; 95(Pt 11): 2462-2467, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25053561

RESUMEN

Reverse genetics is a key methodology for producing genetically modified RNA viruses and deciphering cellular and viral biological properties, but methods based on the preparation of plasmid-based complete viral genomes are laborious and unpredictable. Here, both wild-type and genetically modified infectious RNA viruses were generated in days using the newly described ISA (infectious-subgenomic-amplicons) method. This new versatile and simple procedure may enhance our capacity to obtain infectious RNA viruses from PCR-amplified genetic material.


Asunto(s)
Virus ARN/genética , Virus ARN/fisiología , Genética Inversa/métodos , Animales , Línea Celular , Cricetinae , ADN Complementario/genética , ADN Viral/genética , Flavivirus/genética , Flavivirus/fisiología , Genoma Viral , Humanos , Datos de Secuencia Molecular , ARN Viral/genética , Replicación Viral/genética , Replicación Viral/fisiología
8.
J Antimicrob Chemother ; 69(10): 2770-84, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24951535

RESUMEN

OBJECTIVES: T-705, also known as favipiravir, is a small-molecule inhibitor that is currently in clinical development for the treatment of influenza virus infections. This molecule also inhibits the replication of a broad spectrum of other RNA viruses. The objective of this study was to investigate the antiviral effect of favipiravir on chikungunya virus (CHIKV) replication and to contribute to unravelling the molecular mechanism of action against this virus. METHODS: The anti-CHIKV effect of favipiravir was examined in cell culture and in a mouse model of lethal infection. A five-step protocol was used to select for CHIKV variants with reduced susceptibility to favipiravir. The resistant phenotype was confirmed in cell culture and the whole genome was sequenced. The identified mutations were reverse-engineered into an infectious clone to confirm their impact on the antiviral efficacy of favipiravir. RESULTS: Favipiravir inhibits the replication of laboratory strains and clinical isolates of CHIKV, as well as of a panel of other alphaviruses. Several favipiravir-resistant CHIKV variants were independently selected and all of them in particular acquired the unique K291R mutation in the RNA-dependent RNA polymerase (RdRp). Reverse-engineering of this K291R mutation into an infectious clone of CHIKV confirmed the link between the mutant genotype and the resistant phenotype. Interestingly, this particular lysine is also highly conserved in the RdRp of positive-stranded RNA viruses in general. CONCLUSIONS: This study provides an important insight into the precise molecular mechanism by which favipiravir exerts its antiviral activity against (alpha)viruses, which may be of help in designing other potent broad-spectrum antivirals.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/genética , Farmacorresistencia Viral/genética , Mutación , Pirazinas/farmacología , Proteínas no Estructurales Virales/genética , Amidas/química , Animales , Antivirales/química , Línea Celular , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Efecto Citopatogénico Viral/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ratones , Pruebas de Sensibilidad Microbiana , Fenotipo , Pirazinas/química , Reproducibilidad de los Resultados , Replicación Viral/efectos de los fármacos
9.
Antiviral Res ; 182: 104883, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32750467

RESUMEN

Alphaviruses are arthropod-borne viruses of public health concern. To date no efficient vaccine nor antivirals are available for safe human use. During viral replication the nonstructural protein 1 (nsP1) catalyzes capping of genomic and subgenomic RNAs. The capping reaction is unique to the Alphavirus genus. The whole three-step process follows a particular order: (i) transfer of a methyl group from S-adenosyl methionine (SAM) onto a GTP forming m7GTP; (ii) guanylylation of the enzyme to form a m7GMP-nsP1adduct; (iii) transfer of m7GMP onto 5'-diphosphate RNA to yield capped RNA. Specificities of these reactions designate nsP1 as a promising target for antiviral drug development. In the current study we performed a mutational analysis on two nsP1 positions associated with Sindbis virus (SINV) ribavirin resistance in the Venezuelan equine encephalitis virus (VEEV) context through reverse genetics correlated to enzyme assays using purified recombinant VEEV nsP1 proteins. The results demonstrate that the targeted positions are strongly associated to the regulation of the capping reaction by increasing the affinity between GTP and nsP1. Data also show that in VEEV the S21A substitution, naturally occurring in Chikungunya virus (CHIKV), is a hallmark of ribavirin susceptibility. These findings uncover the specific mechanistic contributions of these residues to nsp1-mediated methyl-transfer and guanylylation reactions.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Mutación , Caperuzas de ARN/metabolismo , Ribavirina/farmacología , Proteínas no Estructurales Virales/genética , Animales , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/genética , Chlorocebus aethiops , Farmacorresistencia Viral , Virus de la Encefalitis Equina Venezolana/genética , Células Vero , Replicación Viral/efectos de los fármacos
10.
Antiviral Res ; 168: 109-113, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31085207

RESUMEN

Dengue fever is the most widespread of the human arbovirus diseases, with approximately one third of the world's population at risk of infection. Dengue viruses are members of the genus Flavivirus (family Flaviviridae) and, antigenically, they separate as four closely related serotypes (1-4) that share 60-75% amino acid homology. This genetic diversity complicates the process of antiviral drug discovery. Thus, currently no approved dengue-specific therapeutic treatments are available. With the aim of providing an efficient tool for dengue virus drug discovery, a collection of nineteen dengue viruses, representing the genotypic diversity within the four serotypes, was developed. After phylogenetic analysis of the full-length genomes, we selected relevant strains from the EVAg collection at Aix-Marseille University and completed the virus collection, using a reverse genetic system based on the infectious sub-genomic amplicons technique. Finally, we evaluated this dengue virus collection against three published dengue inhibitory compounds. NITD008, which targets the highly conserved active site of the viral NS5 polymerase enzyme, exhibited similar antiviral potencies against each of the different dengue genotypes in the panel. Compounds targeting less conserved protein subdomains, such as the capsid inhibitor ST-148, or SDM25N, a ∂ opioid receptor antagonist which indirectly targets NS4B, exhibited larger differences in potency against the various genotypes of dengue viruses. These results illustrate the importance of a phylogenetically based dengue virus reference panel for dengue antiviral research. The collection developed in this study, which includes such representative dengue viruses, has been made available to the scientific community through the European Virus Archive to evaluate novel DENV antiviral candidates.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , Virus del Dengue/clasificación , Evaluación Preclínica de Medicamentos , Variación Genética , Genoma Viral/genética , Genotipo , Humanos , Filogenia , Serogrupo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética
11.
Eur J Med Chem ; 161: 323-333, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30368131

RESUMEN

No antiviral drugs to treat or prevent life-threatening flavivirus infections such as those caused by mosquito-borne Dengue (DENV) and more recently Zika (ZIKV) viruses are yet available. We aim to develop, through a structure-based drug design approach, novel inhibitors targeting the NS5 AdoMet-dependent mRNA methyltransferase (MTase), a viral protein involved in the RNA capping process essential for flaviviruses replication. Herein, we describe the optimization of a hit (5) identified using fragment-based and structure-guided linking techniques, which binds to a proximal site of the AdoMet binding pocket. X-ray crystallographic structures and computational docking were used to guide our optimization process and lead to compounds 30 and 33 (DENV IC50 = 26 µM and 23 µM; ZIKV IC50 = 28 µM and 19  µM, respectively), two representatives of novel non-nucleoside inhibitors of flavivirus MTases.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Metiltransferasas/antagonistas & inhibidores , Virus Zika/efectos de los fármacos , Antivirales/síntesis química , Antivirales/química , Cristalografía por Rayos X , Virus del Dengue/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Metiltransferasas/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Virus Zika/enzimología
12.
Antiviral Res ; 167: 6-12, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30849420

RESUMEN

We report the design, synthesis, and biological evaluation of a class of 1H-pyrido[2,1-b][1,3]benzothiazol-1-ones originated from compound 1, previously identified as anti-flavivirus agent. Some of the new compounds showed activity in low µM range with reasonable selectivity against Dengue 2, Yellow fever (Bolivia strain), and West Nile viruses. One of the most interesting molecules, compound 16, showed broad antiviral activity against additional flaviviruses such as Dengue 1, 3 and 4, Zika, Japanese encephalitis, several strains of Yellow fever, and tick-borne encephalitis viruses. Compound 16 did not exert any effect on alphaviruses and phleboviruses and its activity was maintained in YFV infected cells from different species. The activity of 16 appears specific for flavivirus with respect to other virus families, suggesting, but not proving, that it might be targeting a viral factor. We demonstrated that the antiviral effect of 16 is not related to reduced viral RNA synthesis or virion release. On the contrary, viral particles grown in the presence of 16 showed reduced infectivity, being unable to perform a second round of infection. The chemical class herein presented thus emerges as suitable to provide pan-flavivirus inhibitors.


Asunto(s)
Antivirales , Flaviviridae/efectos de los fármacos , Oxazocinas , Piridinas , Animales , Antivirales/síntesis química , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus de la Encefalitis Transmitidos por Garrapatas/efectos de los fármacos , Humanos , Oxazocinas/síntesis química , Oxazocinas/farmacología , Piridinas/síntesis química , Piridinas/farmacología , ARN Viral/efectos de los fármacos , Virión/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus Zika/efectos de los fármacos
13.
Antiviral Res ; 163: 59-69, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30639438

RESUMEN

Alphaviruses such as the Venezuelan equine encephalitis virus (VEEV) are important human emerging pathogens transmitted by mosquitoes. They possess a unique viral mRNA capping mechanism catalyzed by the viral non-structural protein nsP1, which is essential for virus replication. The alphaviruses capping starts by the methylation of a GTP molecule by the N7-guanine methyltransferase (MTase) activity; nsP1 then forms a covalent link with m7GMP releasing pyrophosphate (GT reaction) and the m7GMP is next transferred onto the 5'-diphosphate end of the viral mRNA to form a cap-0 structure. The cap-0 structure decreases the detection of foreign viral RNAs, prevents RNA degradation by cellular exonucleases, and promotes viral RNA translation into proteins. Additionally, reverse-genetic studies have demonstrated that viruses mutated in nsP1 catalytic residues are both impaired towards replication and attenuated. The nsP1 protein is thus considered an attractive antiviral target for drug discovery. We have previously demonstrated that the guanylylation of VEEV nsP1 can be monitored by Western blot analysis using an antibody recognizing the cap structure. In this study, we developed a high throughput ELISA screening assay to monitor the GT reaction through m7GMP-nsP1 adduct quantitation. This assay was validated using known nsP1 inhibitors before screening 1220 approved compounds. 18 compounds inhibiting the nsP1 guanylylation were identified, and their IC50 determined. Compounds from two series were further characterized and shown to inhibit the nsP1 MTase activity. Conversely, these compounds barely inhibited a cellular MTase demonstrating their specificity towards nsP1. Analogues search and SAR were also initiated to identify the active pharmacophore features. Altogether the results show that this HT enzyme-based assay is a convenient way to select potent and specific hit compounds targeting the viral mRNA capping of Alphaviruses.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/enzimología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Chlorocebus aethiops , Aprobación de Drogas , Ensayo de Inmunoadsorción Enzimática , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Caperuzas de ARN , Células Vero , Replicación Viral/efectos de los fármacos
14.
Retrovirology ; 5: 45, 2008 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-18533995

RESUMEN

Uracil is a natural base of RNA but may appear in DNA through two different pathways including cytosine deamination or misincorporation of deoxyuridine 5'-triphosphate nucleotide (dUTP) during DNA replication and constitutes one of the most frequent DNA lesions. In cellular organisms, such lesions are faithfully cleared out through several universal DNA repair mechanisms, thus preventing genome injury. However, several recent studies have brought some pieces of evidence that introduction of uracil bases in viral genomic DNA intermediates during genome replication might be a way of innate immune defence against some viruses. As part of countermeasures, numerous viruses have developed powerful strategies to prevent emergence of uracilated viral genomes and/or to eliminate uracils already incorporated into DNA. This review will present the current knowledge about the cellular and viral countermeasures against uracils in DNA and the implications of these uracils as weapons against viruses.


Asunto(s)
ADN Viral/biosíntesis , Inmunidad Innata , Uracilo/metabolismo , Virus/genética , Virus/inmunología , Animales , ADN Viral/química , Humanos
15.
Eur J Med Chem ; 143: 1667-1676, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29137867

RESUMEN

Over recent years, many RNA viruses have been "re-discovered", including life-threatening flaviviruses, such as Dengue, Zika, and several encephalitis viruses. Since no specific inhibitors are currently available to treat these infections, there is a pressing need for new therapeutics. Among the flaviviral proteins, NS5 RNA-dependent RNA polymerase (RdRp) represents a validated target being essential for viral replication and it has no human analog. To date, few NS5 RdRp inhibitor chemotypes have been reported and no inhibitors are currently in clinical development. In this context, after an in vitro screening against Dengue 3 NS5 RdRp of our in-house HCV NS5B inhibitors focused library, we found that 2,1-benzothiazine 2,2-dioxides are promising non-nucleoside inhibitors of flaviviral RdRp with compounds 8 and 10 showing IC50 of 0.6 and 0.9 µM, respectively. Preliminary structure-activity relationships indicated a key role for the C-4 benzoyl group and the importance of a properly functionalized C-6 phenoxy moiety to modulate potency. Compound 8 acts as non-competitive inhibitor and its proposed pose in the so-called N pocket of the RdRp thumb domain allowed to explain the key contribution of the benzoyl and the phenoxy moieties for the ligand binding.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Animales , Antivirales/síntesis química , Antivirales/química , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Virus del Dengue/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , ARN Polimerasa Dependiente del ARN/metabolismo , Relación Estructura-Actividad , Células Vero
16.
ACS Infect Dis ; 4(4): 605-619, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29406692

RESUMEN

The re-emergence of chikungunya virus (CHIKV) is a serious global health threat. CHIKV is an alphavirus that is transmitted to humans by Aedes mosquitoes; therefore, their wide distribution significantly contributes to the globalization of the disease. Unfortunately, no effective antiviral drugs are available. We have identified a series of 3-aryl-[1,2,3]triazolo[4,5- d]pyrimidin-7(6 H)-ones as selective inhibitors of CHIKV replication. New series of compounds have now been synthesized with the aim to improve their physicochemical properties and to potentiate the inhibitory activity against different CHIKV strains. Among these newly synthesized compounds modified at position 3 of the aryl ring, tetrahydropyranyl and N- t-butylpiperidine carboxamide derivatives have shown to elicit potent antiviral activity against different clinically relevant CHIKV isolates with 50% effective concentration (EC50) values ranging from 0.30 to 4.5 µM in Vero cells, as well as anti-CHIKV activity in human skin fibroblasts (EC50 = 0.1 µM), a clinically relevant cell system for CHIKV infection.


Asunto(s)
Antivirales/aislamiento & purificación , Virus Chikungunya/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/química , Línea Celular , Fenómenos Químicos , Virus Chikungunya/fisiología , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular
17.
Antiviral Res ; 140: 95-105, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28132865

RESUMEN

Ebola virus (EBOV) haemorrhagic fever remains a threat to global public health with an urgent need for an effective treatment. In order to achieve these goals, access to non-human primate (NHP) laboratory models is an essential requirement. Here, we present the first NHP-EBOV laboratory model readily available to the European scientific community, based on infection of Mauritian cynomolgus macaques using a Central-African EBOV strain and increasing virus challenge dose (10, 100, or 1000 focus forming units per animal). The outcome of these experiments was assessed using clinical, hematological, and biochemical criteria. All challenge doses resulted in fatal infections within 8-11 days. Symptoms appeared from day 5 after infection onwards and disease progression was slower than in previous reports based on Asian cynomolgus macaques. Thus, our model resembled human disease more closely than previous models (onset of symptoms estimated 2-21 days after infection) extending the period of time available for therapeutic intervention. To establish the dynamics of virus genome variation, the study included the first detailed analysis of major and minor genomic EBOV variants during the course of the disease. Major variants were scarce and the population of minor variants was shaped by selective pressure similar to genomic mutations observed in Nature. This primate model provides a robust baseline for future genomic studies in the context of therapeutic methods for treating Ebola virus-infected patients.


Asunto(s)
Ebolavirus/genética , Fiebre Hemorrágica Ebola/virología , Macaca fascicularis , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ebolavirus/aislamiento & purificación , Ebolavirus/patogenicidad , Genoma Viral , Fiebre Hemorrágica Ebola/terapia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
18.
Eur J Med Chem ; 125: 865-880, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27750202

RESUMEN

With the aim to help drug discovery against dengue virus (DENV), a fragment-based drug design approach was applied to identify ligands targeting a main component of DENV replication complex: the NS5 AdoMet-dependent mRNA methyltransferase (MTase) domain, playing an essential role in the RNA capping process. Herein, we describe the identification of new inhibitors developed using fragment-based, structure-guided linking and optimization techniques. Thermal-shift assay followed by a fragment-based X-ray crystallographic screening lead to the identification of three fragment hits binding DENV MTase. We considered linking two of them, which bind to proximal sites of the AdoMet binding pocket, in order to improve their potency. X-ray crystallographic structures and computational docking were used to guide the fragment linking, ultimately leading to novel series of non-nucleoside inhibitors of flavivirus MTase, respectively N-phenyl-[(phenylcarbamoyl)amino]benzene-1-sulfonamide and phenyl [(phenylcarbamoyl)amino]benzene-1-sulfonate derivatives, that show a 10-100-fold stronger inhibition of 2'-O-MTase activity compared to the initial fragments.


Asunto(s)
Antivirales/química , Virus del Dengue/enzimología , Inhibidores Enzimáticos/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/farmacología , Sitios de Unión , Cristalografía por Rayos X , Descubrimiento de Drogas , Humanos , Ligandos , Metiltransferasas/antagonistas & inhibidores , Sulfatos/farmacología , Sulfonamidas/farmacología
19.
Antiviral Res ; 144: 330-339, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28676301

RESUMEN

Two highly pathogenic human coronaviruses associated with severe respiratory syndromes emerged since the beginning of the century. The severe acute respiratory syndrome SARS-coronavirus (CoV) spread first in southern China in 2003 with about 8000 infected cases in few months. Then in 2012, the Middle East respiratory syndrome (MERS-CoV) emerged from the Arabian Peninsula giving a still on-going epidemic associated to a high fatality rate. CoVs are thus considered a major health threat. This is especially true as no vaccine nor specific therapeutic are available against either SARS- or MERS-CoV. Therefore, new drugs need to be identified in order to develop antiviral treatments limiting CoV replication. In this study, we focus on the nsp14 protein, which plays a key role in virus replication as it methylates the RNA cap structure at the N7 position of the guanine. We developed a high-throughput N7-MTase assay based on Homogenous Time Resolved Fluorescence (HTRF®) and screened chemical libraries (2000 compounds) on the SARS-CoV nsp14. 20 compounds inhibiting the SARS-CoV nsp14 were further evaluated by IC50 determination and their specificity was assessed toward flavivirus- and human cap N7-MTases. Our results reveal three classes of compounds: 1) molecules inhibiting several MTases as well as the dengue virus polymerase activity unspecifically, 2) pan MTases inhibitors targeting both viral and cellular MTases, and 3) inhibitors targeting one viral MTase more specifically showing however activity against the human cap N7-MTase. These compounds provide a first basis towards the development of more specific inhibitors of viral methyltransferases.


Asunto(s)
Antivirales/aislamiento & purificación , Evaluación Preclínica de Medicamentos/métodos , Exorribonucleasas/antagonistas & inhibidores , Metiltransferasas/antagonistas & inhibidores , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Fluorometría , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana
20.
Antiviral Res ; 144: 216-222, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28619679

RESUMEN

Chikungunya virus (CHIKV) is a re-emerging alphavirus transmitted to humans by Aedes mosquitoes. Since 2005, CHIKV has been spreading worldwide resulting in epidemics in Africa, the Indian Ocean islands, Asia and more recently in the Americas. CHIKV is thus considered as a global health concern. There is no specific vaccine or drug available for the treatment of this incapacitating viral infection. We previously identified 3-aryl-[1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones as selective inhibitors of CHIKV replication and proposed the viral capping enzyme nsP1 as a target. This work describes the synthesis of novel series of related compounds carrying at the aryl moiety a methylketone and related oximes combined with an ethyl or an ethyl-mimic at 5-position of the triazolopyrimidinone. These compounds have shown antiviral activity against different CHIKV isolates in the very low µM range based on both virus yield reduction and virus-induced cell-killing inhibition assays. Moreover, these antivirals inhibit the in vitro guanylylation of alphavirus nsP1, as determined by Western blot using an anti-cap antibody. Thus, the data obtained seem to indicate that the anti-CHIKV activity might be related to the inhibition of this crucial step in the viral RNA capping machinery.


Asunto(s)
Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Chlorocebus aethiops , Pruebas de Sensibilidad Microbiana , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA