Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 918
Filtrar
1.
N Engl J Med ; 387(21): 1957-1968, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36346079

RESUMEN

BACKGROUND: Bepirovirsen is an antisense oligonucleotide that targets all hepatitis B virus (HBV) messenger RNAs and acts to decrease levels of viral proteins. METHODS: We conducted a phase 2b, randomized, investigator-unblinded trial involving participants with chronic HBV infection who were receiving or not receiving nucleoside or nucleotide analogue (NA) therapy. Participants were randomly assigned (in a 3:3:3:1 ratio) to receive weekly subcutaneous injections of bepirovirsen at a dose of 300 mg for 24 weeks (group 1), bepirovirsen at a dose of 300 mg for 12 weeks then 150 mg for 12 weeks (group 2), bepirovirsen at a dose of 300 mg for 12 weeks then placebo for 12 weeks (group 3), or placebo for 12 weeks then bepirovirsen at a dose of 300 mg for 12 weeks (group 4). Groups 1, 2, and 3 received loading doses of bepirovirsen. The composite primary outcome was a hepatitis B surface antigen (HBsAg) level below the limit of detection and an HBV DNA level below the limit of quantification maintained for 24 weeks after the planned end of bepirovirsen treatment, without newly initiated antiviral medication. RESULTS: The intention-to-treat population comprised 457 participants (227 receiving NA therapy and 230 not receiving NA therapy). Among those receiving NA therapy, a primary-outcome event occurred in 6 participants (9%; 95% credible interval, 0 to 31) in group 1, in 6 (9%; 95% credible interval, 0 to 43) in group 2, in 2 (3%; 95% credible interval, 0 to 16) in group 3, and 0 (0%; post hoc credible interval, 0 to 8) in group 4. Among participants not receiving NA therapy, a primary-outcome event occurred in 7 participants (10%; 95% credible interval, 0 to 38), 4 (6%; 95% credible interval, 0 to 25), 1 (1%; post hoc credible interval, 0 to 6), and 0 (0%; post hoc credible interval, 0 to 8), respectively. During weeks 1 through 12, adverse events, including injection-site reactions, pyrexia, fatigue, and increased alanine aminotransferase levels, were more common with bepirovirsen (groups 1, 2, and 3) than with placebo (group 4). CONCLUSIONS: In this phase 2b trial, bepirovirsen at a dose of 300 mg per week for 24 weeks resulted in sustained HBsAg and HBV DNA loss in 9 to 10% of participants with chronic HBV infection. Larger and longer trials are required to assess the efficacy and safety of bepirovirsen. (Funded by GSK; B-Clear ClinicalTrials.gov number, NCT04449029.).


Asunto(s)
Antivirales , Hepatitis B Crónica , Oligonucleótidos Antisentido , ARN Viral , Humanos , Antivirales/efectos adversos , Antivirales/uso terapéutico , ADN Viral/sangre , Antígenos e de la Hepatitis B/sangre , Antígenos de Superficie de la Hepatitis B/sangre , Virus de la Hepatitis B/genética , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/virología , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/efectos adversos , Oligonucleótidos Antisentido/uso terapéutico , Resultado del Tratamiento , ARN Viral/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Inyecciones Subcutáneas
2.
Nucleic Acids Res ; 50(W1): W266-W271, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35657086

RESUMEN

RNA structures play critical roles in regulating gene expression across all domains of life and viruses. Chemical probing methods coupled with massively parallel sequencing have revolutionized the RNA structure field by enabling the assessment of many structures in their native, physiological context. Previously, we developed Dimethyl-Sulfate-based Mutational Profiling and Sequencing (DMS-MaPseq), which uses DMS to label the Watson-Crick face of open and accessible adenine and cytosine bases in the RNA. We used this approach to determine the genome-wide structures of HIV-1 and SARS-CoV-2 in infected cells, which permitted uncovering new biology and identifying therapeutic targets. Due to the simplicity and ease of the experimental procedure, DMS-MaPseq has been adopted by labs worldwide. However, bioinformatic analysis remains a substantial hurdle for labs that often lack the necessary infrastructure and computational expertise. Here we present a modern web-based interface that automates the analysis of chemical probing profiles from raw sequencing files (http://rnadreem.org). The availability of a simple web-based platform for DMS-MaPseq analysis will dramatically expand studies of RNA structure and aid in the design of structure-based therapeutics.


Asunto(s)
Internet , Conformación de Ácido Nucleico , Pliegue del ARN , ARN , Humanos , ARN/genética , ARN/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Análisis de Secuencia de ARN/métodos , VIH-1/efectos de los fármacos , VIH-1/genética , Adenina , Citosina , Genoma Viral/genética , Diseño de Fármacos , ARN Viral/química , ARN Viral/efectos de los fármacos , ARN Viral/genética
3.
Molecules ; 27(3)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35164069

RESUMEN

The human population is still facing appalling conditions due to several outbreaks of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) virus. The absence of specific drugs, appropriate vaccines for mutants, and knowledge of potential therapeutic agents makes this situation more difficult. Several 1, 2, 4-triazolo [1, 5-a] pyrimidine (TP)-derivative compounds were comprehensively studied for antiviral activities against RNA polymerase of HIV, HCV, and influenza viruses, and showed immense pharmacological interest. Therefore, TP-derivative compounds can be repurposed against the RNA-dependent RNA polymerase (RdRp) protein of SARS-CoV-2. In this study, a meta-analysis was performed to ensure the genomic variability and stability of the SARS-CoV-2 RdRp protein. The molecular docking of natural and synthetic TP compounds to RdRp and molecular dynamic (MD) simulations were performed to analyse the dynamic behaviour of TP compounds at the active site of the RdRp protein. TP compounds were also docked against other non-structural proteins (NSP1, NSP2, NSP3, NSP5, NSP8, NSP13, and NSP15) of SARS-CoV-2. Furthermore, the inhibition potential of TP compounds was compared with Remdesivir and Favipiravir drugs as a positive control. Additionally, TP compounds were analysed for inhibitory activity against SARS-CoV RdRp protein. This study demonstrates that TP analogues (monomethylated triazolopyrimidine and essramycin) represent potential lead molecules for designing an effective inhibitor to control viral replication. Furthermore, in vitro and in vivo studies will strengthen the use of these inhibitors as suitable drug candidates against SARS-CoV-2.


Asunto(s)
ARN Polimerasa Dependiente de ARN de Coronavirus/efectos de los fármacos , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Pirimidinas/farmacología , Triazoles/farmacología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Amidas/farmacología , COVID-19/metabolismo , Dominio Catalítico/efectos de los fármacos , Biología Computacional/métodos , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pirazinas/farmacología , Pirimidinas/química , ARN Viral/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Triazoles/química , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
4.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054766

RESUMEN

After a long limbo, RNA has gained its credibility as a druggable target, fully earning its deserved role in the next generation of pharmaceutical R&D. We have recently probed the trans-activation response (TAR) element, an RNA stem-bulge-loop domain of the HIV-1 genome with bis-3-chloropiperidines (B-CePs), and revealed the compounds unique behavior in stabilizing TAR structure, thus impairing in vitro the chaperone activity of the HIV-1 nucleocapsid (NC) protein. Seeking to elucidate the determinants of B-CePs inhibition, we have further characterized here their effects on the target TAR and its NC recognition, while developing quantitative analytical approaches for the study of multicomponent RNA-based interactions.


Asunto(s)
VIH-1/efectos de los fármacos , Proteínas de la Nucleocápside/metabolismo , Piperidinas/farmacología , ARN Viral/efectos de los fármacos , VIH-1/metabolismo , Conformación de Ácido Nucleico , Piperidinas/química , ARN Viral/química , ARN Viral/metabolismo
5.
Sci Rep ; 12(1): 310, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013354

RESUMEN

Influenza virus is a persistent threat to human health; indeed, the deadliest modern pandemic was in 1918 when an H1N1 virus killed an estimated 50 million people globally. The intent of this work is to better understand influenza from an RNA-centric perspective to provide local, structural motifs with likely significance to the influenza infectious cycle for therapeutic targeting. To accomplish this, we analyzed over four hundred thousand RNA sequences spanning three major clades: influenza A, B and C. We scanned influenza segments for local secondary structure, identified/modeled motifs of likely functionality, and coupled the results to an analysis of evolutionary conservation. We discovered 185 significant regions of predicted ordered stability, yet evidence of sequence covariation was limited to 7 motifs, where 3-found in influenza C-had higher than expected amounts of sequence covariation.


Asunto(s)
Betainfluenzavirus/genética , Gammainfluenzavirus/genética , Virus de la Influenza A/genética , Estabilidad del ARN , ARN Viral/ultraestructura , Antivirales/farmacología , Virus de la Influenza A/efectos de los fármacos , Betainfluenzavirus/efectos de los fármacos , Gammainfluenzavirus/efectos de los fármacos , Modelos Moleculares , Conformación de Ácido Nucleico , Motivos de Nucleótidos , ARN Viral/efectos de los fármacos , ARN Viral/genética , Análisis de Secuencia de ARN , Relación Estructura-Actividad
6.
PLoS Pathog ; 18(1): e1010245, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35041707

RESUMEN

Activation of the NF-κB signaling pathway by Protein Kinase C (PKC) agonists is a potent mechanism for human immunodeficiency virus (HIV) latency disruption in vitro. However, significant toxicity risks and the lack of evidence supporting their activity in vivo have limited further evaluation of PKC agonists as HIV latency-reversing agents (LRA) in cure strategies. Here we evaluated whether GSK445A, a stabilized ingenol-B derivative, can induce HIV/simian immunodeficiency virus (SIV) transcription and virus production in vitro and demonstrate pharmacological activity in nonhuman primates (NHP). CD4+ T cells from people living with HIV and from SIV+ rhesus macaques (RM) on antiretroviral therapy (ART) exposed in vitro to 25 nM of GSK445A produced cell-associated viral transcripts as well as viral particles at levels similar to those induced by PMA/Ionomycin, indicating that GSK445A can potently reverse HIV/SIV latency. Importantly, these concentrations of GSK445A did not impair the proliferation or survival of HIV-specific CD8+ T cells, but instead, increased their numbers and enhanced IFN-γ production in response to HIV peptides. In vivo, GSK445A tolerability was established in SIV-naïve RM at 15 µg/kg although tolerability was reduced in SIV-infected RM on ART. Increases in plasma viremia following GSK445A administration were suggestive of increased SIV transcription in vivo. Collectively, these results indicate that GSK445A is a potent HIV/SIV LRA in vitro and has a tolerable safety profile amenable for further evaluation in vivo in NHP models of HIV cure/remission.


Asunto(s)
Diterpenos/farmacología , VIH , Proteína Quinasa C/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Animales , Humanos , Macaca mulatta , Proteína Quinasa C/metabolismo , ARN Viral/efectos de los fármacos , Transcripción Genética
7.
Org Biomol Chem ; 20(3): 606-612, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34927652

RESUMEN

As the importance of RNA as a therapeutic target has become increasingly recognized, the need for new chemotypes able to bind RNA has grown in significance. We hypothesized that diketopiperazines (DKPs), common substructures in natural products and protein-targeting therapeutic agents, could serve as effective scaffolds for targeting RNA. To confirm this hypothesis, we designed and synthesized two analogs, one incorporating a DKP and one not, of compounds previously demonstrated to bind an RNA critical to the life cycle of HIV-1 with high affinity and specificity. Prior to compound synthesis, calculations employing density functional methods and molecular mechanics conformational searches were used to confirm that the DKP could present functionality in a similar (albeit not identical) orientation to the non DKP-containing compound. We found that both the DKP-containing and parent compound had similar affinities to the target RNA as measured by surface plasmon resonance (SPR). Both compounds were found to have modest but equal anti-HIV activity. These results establish the feasibility of using DKPs to target RNA.


Asunto(s)
Fármacos Anti-VIH/farmacología , Productos Biológicos/farmacología , Dicetopiperazinas/farmacología , VIH/efectos de los fármacos , ARN Viral/efectos de los fármacos , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Teoría Funcional de la Densidad , Dicetopiperazinas/síntesis química , Dicetopiperazinas/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular
8.
Antiviral Res ; 197: 105232, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34968527

RESUMEN

We report the in vitro antiviral activity of DZNep (3-Deazaneplanocin A; an inhibitor of S-adenosylmethionine-dependent methyltransferase) against SARS-CoV-2, besides demonstrating its protective efficacy against lethal infection of infectious bronchitis virus (IBV, a member of the Coronaviridae family). DZNep treatment resulted in reduced synthesis of SARS-CoV-2 RNA and proteins without affecting other steps of viral life cycle. We demonstrated that deposition of N6-methyl adenosine (m6A) in SARS-CoV-2 RNA in the infected cells recruits heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), an RNA binding protein which serves as a m6A reader. DZNep inhibited the recruitment of hnRNPA1 at m6A-modified SARS-CoV-2 RNA which eventually suppressed the synthesis of the viral genome. In addition, m6A-marked RNA and hnRNPA1 interaction was also shown to regulate early translation to replication switch of SARS-CoV-2 genome. Furthermore, abrogation of methylation by DZNep also resulted in defective synthesis of the 5' cap of viral RNA, thereby resulting in its failure to interact with eIF4E (a cap-binding protein), eventually leading to a decreased synthesis of viral proteins. Most importantly, DZNep-resistant mutants could not be observed upon long-term sequential passage of SARS-CoV-2 in cell culture. In summary, we report the novel role of methylation in the life cycle of SARS-CoV-2 and propose that targeting the methylome using DZNep could be of significant therapeutic value against SARS-CoV-2 infection.


Asunto(s)
Adenosina/análogos & derivados , Genoma Viral/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , SARS-CoV-2/efectos de los fármacos , Adenosina/farmacología , Animales , Embrión de Pollo , Chlorocebus aethiops , Secuenciación de Inmunoprecipitación de Cromatina , Metilación de ADN/efectos de los fármacos , Metilación de ADN/fisiología , Farmacorresistencia Viral/efectos de los fármacos , Genoma Viral/genética , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Humanos , Dosificación Letal Mediana , Ratones , Biosíntesis de Proteínas/efectos de los fármacos , ARN Viral/efectos de los fármacos , ARN Viral/metabolismo , Conejos , SARS-CoV-2/genética , Organismos Libres de Patógenos Específicos , Transcripción Genética/efectos de los fármacos , Células Vero
9.
Viruses ; 13(12)2021 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-34960758

RESUMEN

BACKGROUND: There is an urgent need for new antivirals with powerful therapeutic potential and tolerable side effects. METHODS: Here, we tested the antiviral properties of interferons (IFNs), alone and with other drugs in vitro. RESULTS: While IFNs alone were insufficient to completely abolish replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), IFNα, in combination with remdesivir, EIDD-2801, camostat, cycloheximide, or convalescent serum, proved to be more effective. Transcriptome and metabolomic analyses revealed that the IFNα-remdesivir combination suppressed SARS-CoV-2-mediated changes in Calu-3 cells and lung organoids, although it altered the homeostasis of uninfected cells and organoids. We also demonstrated that IFNα combinations with sofosbuvir, telaprevir, NITD008, ribavirin, pimodivir, or lamivudine were effective against HCV, HEV, FLuAV, or HIV at lower concentrations, compared to monotherapies. CONCLUSIONS: Altogether, our results indicated that IFNα can be combined with drugs that affect viral RNA transcription, protein synthesis, and processing to make synergistic combinations that can be attractive targets for further pre-clinical and clinical development against emerging and re-emerging viral infections.


Asunto(s)
Antivirales/farmacología , Interferón-alfa/farmacología , SARS-CoV-2/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/virología , Metaboloma/efectos de los fármacos , Organoides , ARN Viral/biosíntesis , ARN Viral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus/clasificación , Virus/efectos de los fármacos
10.
Viruses ; 13(12)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34960780

RESUMEN

Broad-spectrum antiviral therapies hold promise as a first-line defense against emerging viruses by blunting illness severity and spread until vaccines and virus-specific antivirals are developed. The nucleobase favipiravir, often discussed as a broad-spectrum inhibitor, was not effective in recent clinical trials involving patients infected with Ebola virus or SARS-CoV-2. A drawback of favipiravir use is its rapid clearance before conversion to its active nucleoside-5'-triphosphate form. In this work, we report a synergistic reduction of flavivirus (dengue, Zika), orthomyxovirus (influenza A), and coronavirus (HCoV-OC43 and SARS-CoV-2) replication when the nucleobases favipiravir or T-1105 were combined with the antimetabolite 6-methylmercaptopurine riboside (6MMPr). The 6MMPr/T-1105 combination increased the C-U and G-A mutation frequency compared to treatment with T-1105 or 6MMPr alone. A further analysis revealed that the 6MMPr/T-1105 co-treatment reduced cellular purine nucleotide triphosphate synthesis and increased conversion of the antiviral nucleobase to its nucleoside-5'-monophosphate, -diphosphate, and -triphosphate forms. The 6MMPr co-treatment specifically increased production of the active antiviral form of the nucleobases (but not corresponding nucleosides) while also reducing levels of competing cellular NTPs to produce the synergistic effect. This in-depth work establishes a foundation for development of small molecules as possible co-treatments with nucleobases like favipiravir in response to emerging RNA virus infections.


Asunto(s)
Antimetabolitos/farmacología , Antivirales/farmacología , Virus ARN/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Amidas/farmacología , Animales , Línea Celular , Sinergismo Farmacológico , Guanosina Trifosfato/metabolismo , Humanos , Metiltioinosina/farmacología , Mutación/efectos de los fármacos , Fosforribosil Pirofosfato/metabolismo , Pirazinas/farmacología , Virus ARN/clasificación , Virus ARN/genética , ARN Viral/efectos de los fármacos , ARN Viral/genética , Replicación Viral/efectos de los fármacos
11.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769072

RESUMEN

Inhaled nebulized interferon (IFN)-α and IFN-ß have been shown to be effective in the management of coronavirus disease 2019 (COVID-19). We aimed to construct a virus-free rapid detection system for high-throughput screening of IFN-like compounds that induce viral RNA degradation and suppress the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We prepared a SARS-CoV-2 subreplicon RNA expression vector which contained the SARS-CoV-2 5'-UTR, the partial sequence of ORF1a, luciferase, nucleocapsid, ORF10, and 3'-UTR under the control of the cytomegalovirus promoter. The expression vector was transfected into Calu-3 cells and treated with IFN-α and the IFNAR2 agonist CDM-3008 (RO8191) for 3 days. SARS-CoV-2 subreplicon RNA degradation was subsequently evaluated based on luciferase levels. IFN-α and CDM-3008 suppressed SARS-CoV-2 subreplicon RNA in a dose-dependent manner, with IC50 values of 193 IU/mL and 2.54 µM, respectively. HeLa cells stably expressing SARS-CoV-2 subreplicon RNA were prepared and treated with the IFN-α and pan-JAK inhibitor Pyridone 6 or siRNA-targeting ISG20. IFN-α activity was canceled with Pyridone 6. The knockdown of ISG20 partially canceled IFN-α activity. Collectively, we constructed a virus-free rapid detection system to measure SARS-CoV-2 RNA suppression. Our data suggest that the SARS-CoV-2 subreplicon RNA was degraded by IFN-α-induced ISG20 exonuclease activity.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Interferón-alfa/farmacología , ARN Viral/metabolismo , SARS-CoV-2/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Exorribonucleasas/genética , Vectores Genéticos , Células HeLa , Humanos , Interferón-alfa/administración & dosificación , Luciferasas/genética , Luciferasas/metabolismo , Naftiridinas/administración & dosificación , Naftiridinas/farmacología , Oxadiazoles/administración & dosificación , Oxadiazoles/farmacología , ARN Viral/efectos de los fármacos , Replicón
12.
Cell Rep ; 37(4): 109882, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34653416

RESUMEN

Remdesivir (RDV), a nucleotide analog with broad-spectrum features, has exhibited effectiveness in COVID-19 treatment. However, the precise working mechanism of RDV when targeting the viral RNA-dependent RNA polymerase (RdRP) has not been fully elucidated. Here, we solve a 3.0-Å structure of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RdRP elongation complex (EC) and assess RDV intervention in polymerase elongation phase. Although RDV could induce an "i+3" delayed termination in meta-stable complexes, only pausing and subsequent elongation are observed in the EC. A comparative investigation using an enterovirus RdRP further confirms similar delayed intervention and demonstrates that steric hindrance of the RDV-characteristic 1'-cyano at the -4 position is responsible for the "i+3" intervention, although two representative Flaviviridae RdRPs do not exhibit similar behavior. A comparison of representative viral RdRP catalytic complex structures indicates that the product RNA backbone encounters highly conserved structural elements, highlighting the broad-spectrum intervention potential of 1'-modified nucleotide analogs in anti-RNA virus drug development.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , ARN Polimerasa Dependiente del ARN/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Proteínas Virales/efectos de los fármacos , Adenosina Monofosfato/farmacología , Alanina/farmacología , Microscopía por Crioelectrón , Humanos , ARN Viral/química , ARN Viral/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/química , SARS-CoV-2/química , Proteínas Virales/química , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
13.
Antiviral Res ; 195: 105188, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34648875

RESUMEN

Zika virus (ZIKV) has been the cause of some epidemics since 2007. The correlations of microcephaly and Guillain-Barré syndrome with ZIKV have been noticed. Unfortunately, researchers have yet to develop an effective vaccine or drug approved for ZIKV infection. Anidulafungin is a member of echinocandins that is used to treat candida infections. This study assessed the antiviral capability of anidulafungin against ZIKV. Anidulafungin was shown to significantly decrease viral RNA levels, protein expression levels, viral yields, and the rate of infection. In time of addition assays, anidulafungin exhibited inhibitory activities in the early stages of ZIKV infection. In binding and entry assays, administering anidulafungin did not lead to a corresponding decrease in quantity of viral RNA, but a significant decrease in ZIKV infectivity was observed in virucidal assays. This indicated that anidulafungin interferes directly with virions. T-1105 is a viral polymerase inhibitor, which functions in the late stage of ZIKV infection. When anidulafungin was administered in combination with T-1105, an obvious synergistic effect was observed, resulting in a combination index (CI) value of 0.85 ± 0.13. Finally, we evaluated the effects of echinocandins in terms of half-maximal inhibitory concentration (IC50), calculation of cytotoxicity concentration 50% (CC50), selectivity index (SI), and Patchdock score. Among the tests, anidulafungin bears the lowest IC50 and highest Patchdock score. Although anidulafungin is classified as a pregnancy category C agent; however, combination therapy of anidulafungin with a viral RNA replication inhibitor could expand treatment options for ZIKV infection.


Asunto(s)
Anidulafungina/farmacología , Antivirales/síntesis química , Pirazinas/farmacología , ARN Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos , Animales , Antivirales/farmacología , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Concentración 50 Inhibidora , Células Vero , Virión/efectos de los fármacos , Acoplamiento Viral/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología
14.
Viruses ; 13(9)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34578365

RESUMEN

Rhinoviruses (RVs) are the main cause of recurrent infections with rather mild symptoms characteristic of the common cold. Nevertheless, RVs give rise to enormous numbers of absences from work and school and may become life-threatening in particular settings. Vaccination is jeopardised by the large number of serotypes eliciting only poorly cross-neutralising antibodies. Conversely, antivirals developed over the years failed FDA approval because of a low efficacy and/or side effects. RV species A, B, and C are now included in the fifteen species of the genus Enteroviruses based upon the high similarity of their genome sequences. As a result of their comparably low pathogenicity, RVs have become a handy model for other, more dangerous members of this genus, e.g., poliovirus and enterovirus 71. We provide a short overview of viral proteins that are considered potential drug targets and their corresponding drug candidates. We briefly mention more recently identified cellular enzymes whose inhibition impacts on RVs and comment novel approaches to interfere with infection via aggregation, virus trapping, or preventing viral access to the cell receptor. Finally, we devote a large part of this article to adding the viral RNA genome to the list of potential drug targets by dwelling on its structure, folding, and the still debated way of its exit from the capsid. Finally, we discuss the recent finding that G-quadruplex stabilising compounds impact on RNA egress possibly via obfuscating the unravelling of stable secondary structural elements.


Asunto(s)
Antivirales/farmacología , ARN Viral/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Aminoquinolinas/farmacología , Animales , Cápside/metabolismo , Proteínas de la Cápside/genética , Enterovirus/genética , Infecciones por Enterovirus/virología , Genoma Viral/efectos de los fármacos , Humanos , Ácidos Picolínicos/farmacología , Poliovirus/genética , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas Virales/genética
15.
Hepatol Commun ; 5(11): 1873-1887, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34558823

RESUMEN

Therapy with nucleic acid polymers (NAPs), tenofovir disoproxil fumarate (TDF), and pegylated interferon (pegIFN) achieve high rates of HBsAg loss/seroconversion and functional cure in chronic hepatitis B virus (HBV) infection. The role of hepatitis B surface antigen (HBsAg) seroconversion and inactivation of covalently closed circular DNA (cccDNA) in establishing functional cure were examined. Archived serum from the REP 401 study was analyzed using the Abbott ARCHITECT HBsAg NEXT assay (Chicago, IL), Abbott research use-only assays for HBsAg immune complexes (HBsAg ICs), circulating HBV RNA, and the Fujirebio assay for hepatitis B core-related antigen (HBcrAg; Malvern, PA). HBsAg became < 0.005 IU/mL in 23 participants during NAP exposure, which persisted in all participants with functional cure. HBsAg IC declined during lead-in TDF monotherapy and correlated with minor declines in HBsAg. Following the addition of NAPs and pegIFN, minor HBsAg IC increases (n = 13) or flares (n = 2) during therapy were not correlated with HBsAg decline, hepatitis B surface antibody (anti-HBs) titers, or alanine aminotransferase. HBsAg IC universally declined during follow-up in participants with virologic control or functional cure. Universal declines in HBV RNA and HBcrAg during TDF monotherapy continued with NAP + pegIFN regardless of therapeutic outcome. At the end of therapy, HBV RNA was undetectable in only 5 of 14 participants with functional cure but became undetectable after removal of therapy in all participants with functional cure. Undetectable HBV RNA at the end of therapy in 5 participants was followed by relapse to virologic control or viral rebound. Conclusion: Anti-HBs-independent mechanisms contribute to HBsAg clearance during NAP therapy. Inactivation of cccDNA does not predict functional cure following NAP-based therapy; however, functional cure is accompanied by persistent inactivation of cccDNA. Persistent HBsAg loss with functional cure may also reflect reduction/clearance of integrated HBV DNA. Clinicaltrials.org number NCT02565719.


Asunto(s)
Antivirales/uso terapéutico , ADN Circular/sangre , Antígenos de Superficie de la Hepatitis B/sangre , Virus de la Hepatitis B/genética , Hepatitis B Crónica/inmunología , Seroconversión/efectos de los fármacos , Adulto , Alanina Transaminasa/sangre , Alanina Transaminasa/efectos de los fármacos , Estudios Cruzados , ADN Circular/efectos de los fármacos , Quimioterapia Combinada , Femenino , Antígenos del Núcleo de la Hepatitis B/sangre , Antígenos del Núcleo de la Hepatitis B/efectos de los fármacos , Antígenos del Núcleo de la Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/inmunología , Hepatitis B Crónica/sangre , Hepatitis B Crónica/tratamiento farmacológico , Humanos , Interferones/uso terapéutico , Masculino , Ácidos Nucleicos/uso terapéutico , Polímeros/uso terapéutico , ARN Viral/sangre , ARN Viral/efectos de los fármacos , ARN Viral/inmunología , Tenofovir/uso terapéutico , Resultado del Tratamiento , Inactivación de Virus/efectos de los fármacos
17.
Viruses ; 13(5)2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065382

RESUMEN

SARS-CoV-2 is the causative agent of COVID-19, which is a global pandemic. SARS-CoV-2 is transmitted rapidly via contaminated surfaces and aerosols, emphasizing the importance of environmental disinfection to block the spread of virus. Ultraviolet C radiation and chemical compounds are effective for SARS-CoV-2 disinfection, but can only be applied in the absence of humans due to their toxicities. Therefore, development of disinfectants that can be applied in working spaces without evacuating people is needed. Here we showed that TiO2-mediated photocatalytic reaction inactivates SARS-CoV-2 in a time-dependent manner and decreases its infectivity by 99.9% after 20 min and 120 min of treatment in aerosol and liquid, respectively. The mechanistic effects of TiO2 photocatalyst on SARS-CoV-2 virion included decreased total observed virion count, increased virion size, and reduced particle surface spike structure, as determined by transmission electron microscopy. Damage to viral proteins and genome was further confirmed by western blotting and RT-qPCR, respectively. The multi-antiviral effects of TiO2-mediated photocatalytic reaction implies universal disinfection potential for different infectious agents. Notably, TiO2 has no adverse effects on human health, and therefore, TiO2-induced photocatalytic reaction is suitable for disinfection of SARS-CoV-2 and other emerging infectious disease-causing agents in human habitation.


Asunto(s)
Desinfección/métodos , SARS-CoV-2/efectos de los fármacos , Titanio/farmacología , Animales , COVID-19/metabolismo , Línea Celular , Chlorocebus aethiops , Humanos , Pandemias , ARN Viral/efectos de los fármacos , SARS-CoV-2/patogenicidad , Titanio/química , Células Vero
18.
Medicine (Baltimore) ; 100(24): e26312, 2021 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-34128871

RESUMEN

ABSTRACT: All-oral direct-acting antiviral therapies are becoming the choice for hepatitis C (HCV) treatment. In this study, we aimed to evaluate the efficacy and safety of ritonavir-boosted danoprevir (DNVr) plus sofosbuvir±ribavirin on HCV genotype 1, 2, 3, or 6 in the real world in China.In this observational, prospective, multicenter cohort, we enrolled a total of 58 patients with HCV genotype 1, 2, 3, or 6 patients from July 2018 to December 2019. All patients were treated with DNVr plus sofosbuvir ± ribavirin for 12 weeks and then followed up for 12 weeks. The primary endpoint was the rate of sustained virologic response at week 12 after the end of treatment (SVR12). The secondary endpoint was virologic response rate at end-of-treatment and adverse event outcome.Of the 58 patients who were enrolled, 5.2% (n = 3) had genotype 1a; 43.1% (n = 25) had HCV genotype 1b; 17.2% (n = 10) had genotype 2a; 5.2% (n = 3) had genotype 3a; 8.6% (n = 5) had genotype 3b; and 20.7% (n = 12) had genotype 6a. The virologic response rate at end-of-treatment was 100% (58/58). The HCV-RNA results of 5 patients were absent at week 12 after treatment. Among the 53 patients, SVR12 rate achieved 100% (53/53) with DNVr plus sofosbuvir ± ribavirin treatment in patients with HCV genotype 1b, 2a, 3, and 6a. For compensated cirrhosis and noncirrhosis patients, SVR12 was 100% with DNVr plus sofosbuvir ± ribavirin treatment. No serious event was observed during the treatment and follow-up. Only 5 patients had mild adverse events.DNVr plus sofosbuvir ± ribavirin for 12 weeks provided 100% SVR12 in a broad patient population and were well tolerated, which may be a promising regimen for CHC treatment.


Asunto(s)
Antivirales/administración & dosificación , Ciclopropanos/administración & dosificación , Hepacivirus/genética , Hepatitis C Crónica/tratamiento farmacológico , Isoindoles/administración & dosificación , Lactamas Macrocíclicas/administración & dosificación , Prolina/análogos & derivados , Ribavirina/administración & dosificación , Sofosbuvir/administración & dosificación , Sulfonamidas/administración & dosificación , Adulto , China , Quimioterapia Combinada , Femenino , Genotipo , Hepatitis C Crónica/virología , Humanos , Cirrosis Hepática/virología , Masculino , Persona de Mediana Edad , Prolina/administración & dosificación , Estudios Prospectivos , ARN Viral/efectos de los fármacos , Respuesta Virológica Sostenida
19.
Curr Opin Virol ; 49: 81-85, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34052732

RESUMEN

The nucleotide analogue prodrug remdesivir remains the only FDA-approved antiviral small molecule for the treatment of infection with SARS-CoV-2. Biochemical studies revealed that the active form of the drug targets the viral RNA-dependent RNA polymerase and causes delayed chain-termination. Delayed chain-termination is incomplete, but the continuation of RNA synthesis enables a partial escape from viral proofreading. Remdesivir becomes embedded in the copy of the RNA genome that later serves as a template. Incorporation of an incoming nucleotide triphosphate is now inhibited by the modified template. Knowledge on the mechanism of action matters. Enzymatic inhibition links to antiviral effects in cell cultures, animal models and viral load reduction in patients, which provides the logical chain that is expected for a direct acting antiviral. Hence, remdesivir also serves as a benchmark in current drug development efforts that will hopefully lead to orally available treatments to the benefit of a broader population.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/farmacología , Alanina/uso terapéutico , ARN Polimerasa Dependiente de ARN de Coronavirus/antagonistas & inhibidores , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Humanos , Profármacos/farmacología , Profármacos/uso terapéutico , ARN Viral/biosíntesis , ARN Viral/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , SARS-CoV-2/fisiología , Especificidad por Sustrato , Replicación Viral/efectos de los fármacos
20.
Int Immunopharmacol ; 97: 107702, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33930706

RESUMEN

BACKGROUND: The clinical characteristics and treatment of patients who tested positive for COVID-19 after recovery remained elusive. Effective antiviral therapy is important for tackling these patients. We assessed the efficacy and safety of favipiravir for treating these patients. METHODS: This is a multicenter, open-label, randomized controlled trial in SARS-CoV-2 RNA re-positive patients. Patients were randomly assigned in a 2:1 ratio to receive either favipiravir, in addition to standard care, or standard care alone. The primary outcome was time to achieve a consecutive twice (at intervals of more than 24 h) negative RT-PCR result for SARS-CoV-2 RNA in nasopharyngeal swab and sputum sample. RESULTS: Between March 27 and May 9, 2020, 55 patients underwent randomization; 36 were assigned to the favipiravir group and 19 were assigned to the control group. Favipiravir group had a significantly shorter time from start of study treatment to negative nasopharyngeal swab and sputum than control group (median 17 vs. 26 days); hazard ratio 2.1 (95% CI [1.1-4.0], p = 0.038). The proportion of virus shedding in favipiravir group was higher than control group (80.6% [29/36] vs. 52.6% [10/19], p = 0.030, respectively). C-reactive protein decreased significantly after treatment in the favipiravir group (p = 0.016). The adverse events were generally mild and self-limiting. CONCLUSION: Favipiravir was safe and superior to control in shortening the duration of viral shedding in SARS-CoV-2 RNA recurrent positive after discharge. However, a larger scale and randomized, double-blind, placebo-controlled trial is required to confirm our conclusion.


Asunto(s)
Amidas/administración & dosificación , Antivirales/administración & dosificación , Tratamiento Farmacológico de COVID-19 , Pirazinas/administración & dosificación , Reinfección/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Amidas/efectos adversos , Antivirales/efectos adversos , COVID-19/sangre , Femenino , Humanos , Subgrupos Linfocitarios/efectos de los fármacos , Masculino , Persona de Mediana Edad , Alta del Paciente , Pirazinas/efectos adversos , ARN Viral/análisis , ARN Viral/efectos de los fármacos , Reinfección/sangre , SARS-CoV-2/efectos de los fármacos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA