Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Antiviral Res ; 224: 105842, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38417531

RESUMEN

Enteroviruses are a significant global health concern, causing a spectrum of diseases from the common cold to more severe conditions like hand-foot-and-mouth disease, meningitis, myocarditis, pancreatitis, and poliomyelitis. Current treatment options for these infections are limited, underscoring the urgent need for effective therapeutic strategies. To find better treatment option we analyzed toxicity and efficacy of 12 known broad-spectrum anti-enterovirals both individually and in combinations against different enteroviruses in vitro. We identified several novel, synergistic two-drug and three-drug combinations that demonstrated significant inhibition of enterovirus infections in vitro. Specifically, the triple-drug combination of pleconaril, rupintrivir, and remdesivir exhibited remarkable efficacy against echovirus (EV) 1, EV6, EV11, and coxsackievirus (CV) B5, in human lung epithelial A549 cells. This combination surpassed the effectiveness of single-agent or dual-drug treatments, as evidenced by its ability to protect A549 cells from EV1-induced cytotoxicity across seven passages. Additionally, this triple-drug cocktail showed potent antiviral activity against EV-A71 in human intestinal organoids. Thus, our findings highlight the therapeutic potential of the pleconaril-rupintrivir-remdesivir combination as a broad-spectrum treatment option against a range of enterovirus infections. The study also paves the way towards development of strategic antiviral drug combinations with virus family coverage and high-resistance barriers.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Isoxazoles , Oxadiazoles , Oxazoles , Fenilalanina/análogos & derivados , Pirrolidinonas , Valina/análogos & derivados , Animales , Humanos , Infecciones por Enterovirus/tratamiento farmacológico , Enterovirus Humano B , Antivirales/farmacología , Antivirales/uso terapéutico , Combinación de Medicamentos
2.
Int J Infect Dis ; 137: 75-78, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37852599

RESUMEN

Vaccinated convalescents do not develop severe COVID-19 after infection with new SARS-CoV-2 variants. We questioned how messenger RNA (mRNA) vaccination of convalescents provides protection from emerging virus variants. From the cohort of 71 convalescent plasma donors, we identified a patient who developed immune response to infection with SARS-CoV-2 variant of 20A clade and who subsequently received mRNA vaccine encoding spike (S) protein of strain of 19A clade. We showed that vaccination increased the production of immune cells and anti-S antibodies in the serum. Serum antibodies neutralized not only 19A and 20A, but also 20B, 20H, 21J, and 21K virus variants. One of the serum antibodies (100F8) completely neutralized 20A, 21J, and partially 21K strains. 100F8 was structurally similar to published Ab188 antibody, which recognized non-conserved epitope on the S protein. We proposed that 100F8 and other serum antibodies of the patient which recognized non- and conserved epitopes of the S protein, could have additive or synergistic effects to neutralize various virus variants. Thus, mRNA vaccination could be beneficial for convalescents because it boosts production of neutralizing antibodies with broad-spectrum activity.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/prevención & control , Sueroterapia para COVID-19 , Anticuerpos Neutralizantes , Vacunación , Epítopos , ARN Mensajero/genética , Anticuerpos Antivirales
3.
iScience ; 26(9): 107631, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37664584

RESUMEN

Severe infections with coronaviruses are often accompanied with hyperinflammation, requiring therapeutic strategies to simultaneously tackle the virus and inflammation. By screening a safe-in-human broad-spectrum antiviral agents library, we identified that indomethacin can inhibit pan-coronavirus infection in human cell and airway organoids models. Combining indomethacin with oral antiviral drugs authorized for treating COVID-19 results in synergistic anti-coronavirus activity. Coincidentally, screening a library of FDA-approved drugs identified indomethacin as the most potent potentiator of interferon response through increasing STAT1 phosphorylation. Combining indomethacin with interferon-alpha exerted synergistic antiviral effects against multiple coronaviruses. The anti-coronavirus activity of indomethacin is associated with activating interferon response. In a co-culture system of lung epithelial cells with macrophages, indomethacin inhibited both viral replication and inflammatory response. Collectively, indomethacin is a pan-coronavirus inhibitor that can simultaneously inhibit virus-triggered inflammatory response. The therapeutic potential of indomethacin can be further augmented by combining it with oral antiviral drugs or interferon-alpha.

4.
Cell Mol Life Sci ; 79(12): 605, 2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-36436108

RESUMEN

The viral epidemics and pandemics have stimulated the development of known and the discovery of novel antiviral agents. About a hundred mono- and combination antiviral drugs have been already approved, whereas thousands are in development. Here, we briefly reviewed 7 classes of antiviral agents: neutralizing antibodies, neutralizing recombinant soluble human receptors, antiviral CRISPR/Cas systems, interferons, antiviral peptides, antiviral nucleic acid polymers, and antiviral small molecules. Interferons and some small molecules alone or in combinations possess broad-spectrum antiviral activity, which could be beneficial for treatment of emerging and re-emerging viral infections.


Asunto(s)
Antivirales , Virosis , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/química , Interferones , Virosis/tratamiento farmacológico
5.
Viruses ; 14(10)2022 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-36298676

RESUMEN

Background: Some viruses cause outbreaks, which require immediate attention. Neutralizing antibodies could be developed for viral outbreak management. However, the development of monoclonal antibodies is often long, laborious, and unprofitable. Here, we report the development of chicken polyclonal neutralizing antibodies against SARS-CoV-2 infection. Methods: Layers were immunized twice with 14-day intervals using the purified receptor-binding domain (RBD) of the S protein of SARS-CoV-2/Wuhan or SARS-CoV-2/Omicron. Eggs were harvested 14 days after the second immunization. Polyclonal IgY antibodies were extracted. Binding of anti-RBD IgYs was analyzed by immunoblot and indirect ELISA. Furthermore, the neutralization capacity of anti-RBD IgYs was measured in Vero-E6 cells infected with SARS-CoV-2-mCherry/Wuhan and SARS-CoV-2/Omicron using fluorescence and/or cell viability assays. In addition, the effect of IgYs on the expression of SARS-CoV-2 and host cytokine genes in the lungs of Syrian Golden hamsters was examined using qRT-PCR. Results: Anti-RBD IgYs efficiently bound viral RBDs in situ, neutralized the virus variants in vitro, and lowered viral RNA amplification, with minimal alteration of virus-mediated immune gene expression in vivo. Conclusions: Altogether, our results indicate that chicken polyclonal IgYs can be attractive targets for further pre-clinical and clinical development for the rapid management of outbreaks of emerging and re-emerging viruses.


Asunto(s)
COVID-19 , Animales , COVID-19/prevención & control , Glicoproteína de la Espiga del Coronavirus/genética , Pollos , SARS-CoV-2 , Yema de Huevo , ARN Viral , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Anticuerpos Monoclonales , Antivirales , Citocinas
6.
Viruses ; 14(9)2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36146673

RESUMEN

Background: Enterovirus infections affect people around the world, causing a range of illnesses, from mild fevers to severe, potentially fatal conditions. There are no approved treatments for enterovirus infections. Methods: We have tested our library of broad-spectrum antiviral agents (BSAs) against echovirus 1 (EV1) in human adenocarcinoma alveolar basal epithelial A549 cells. We also tested combinations of the most active compounds against EV1 in A549 and human immortalized retinal pigment epithelium RPE cells. Results: We confirmed anti-enteroviral activities of pleconaril, rupintrivir, cycloheximide, vemurafenib, remdesivir, emetine, and anisomycin and identified novel synergistic rupintrivir-vemurafenib, vemurafenib-pleconaril and rupintrivir-pleconaril combinations against EV1 infection. Conclusions: Because rupintrivir, vemurafenib, and pleconaril require lower concentrations to inhibit enterovirus replication in vitro when combined, their cocktails may have fewer side effects in vivo and, therefore, should be further explored in preclinical and clinical trials against EV1 and other enterovirus infections.


Asunto(s)
Infecciones por Enterovirus , Picornaviridae , Anisomicina/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Cicloheximida/uso terapéutico , Combinación de Medicamentos , Emetina , Humanos , Vemurafenib/uso terapéutico
7.
Nucleic Acids Res ; 50(W1): W272-W275, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35610052

RESUMEN

Viruses can cross species barriers and cause unpredictable outbreaks in man with substantial economic and public health burdens. Broad-spectrum antivirals, (BSAs, compounds inhibiting several human viruses), and BSA-containing drug combinations (BCCs) are deemed as immediate therapeutic options that fill the void between virus identification and vaccine development. Here, we present DrugVirus.info 2.0 (https://drugvirus.info), an integrative interactive portal for exploration and analysis of BSAs and BCCs, that greatly expands the database and functionality of DrugVirus.info 1.0 webserver. Through the data portal that now expands the spectrum of BSAs and provides information on BCCs, we developed two modules for (i) interactive analysis of users' own antiviral drug and combination screening data and their comparison with published datasets, and (ii) exploration of the structure-activity relationship between various BSAs. The updated portal provides an essential toolbox for antiviral drug development and repurposing applications aiming to identify existing and novel treatments of emerging and re-emerging viral threats.


Asunto(s)
Antivirales , Bases de Datos Farmacéuticas , Virus , Humanos , Antivirales/farmacología , Combinación de Medicamentos , Desarrollo de Medicamentos , Virus/efectos de los fármacos , Programas Informáticos , Internet
8.
iScience ; 25(4): 104112, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35402870

RESUMEN

Broadly effective antiviral therapies must be developed to be ready for clinical trials, which should begin soon after the emergence of new life-threatening viruses. Here, we pave the way towards this goal by reviewing conserved druggable virus-host interactions, mechanisms of action, immunomodulatory properties of available broad-spectrum antivirals (BSAs), routes of BSA delivery, and interactions of BSAs with other antivirals. Based on the review, we concluded that the range of indications of BSAs can be expanded, and new pan- and cross-viral mono- and combinational therapies can be developed. We have also developed a new scoring algorithm that can help identify the most promising few of the thousands of potential BSAs and BSA-containing drug cocktails (BCCs) to prioritize their development during the critical period between the identification of a new virus and the development of virus-specific vaccines, drugs, and therapeutic antibodies.

9.
Viruses ; 14(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35215894

RESUMEN

Viral diseases consistently pose a substantial economic and public health burden worldwide [...].


Asunto(s)
Antivirales/farmacología , Virosis/tratamiento farmacológico , Humanos , Virosis/virología , Fenómenos Fisiológicos de los Virus , Virus/clasificación , Virus/efectos de los fármacos , Virus/genética
10.
Sci Adv ; 8(3): eabj5908, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35044825

RESUMEN

Hepatotropic viruses naturally have narrow host and tissue tropisms, challenging the development of robust experimental models. The advent of organoid technology provides a unique opportunity for moving the field forward. Here, we demonstrate that three-dimensional cultured organoids from fetal and adult human liver with cholangiocyte or hepatocyte phenotype support hepatitis E virus (HEV) replication. Inoculation with infectious HEV particles demonstrates that human liver­derived organoids support the full life cycle of HEV infection. By directing organoids toward polarized monolayers in a transwell system, we observed predominantly apical secretion of HEV particles. Genome-wide transcriptomic and tRNAome analyses revealed robust host responses triggered by viral replication. Drug screening in organoids identified brequinar and homoharringtonine as potent HEV inhibitors, which are also effective against the ribavirin resistance variant harboring G1634R mutation. Thus, successful recapitulation of HEV infection in liver-derived organoids shall facilitate the study of virus-host interactions and development of antiviral therapies.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Antivirales/farmacología , Antivirales/uso terapéutico , Hepatitis E/tratamiento farmacológico , Virus de la Hepatitis E/genética , Interacciones Microbiota-Huesped , Humanos , Organoides
11.
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
12.
Viruses ; 13(9)2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34578348

RESUMEN

SARS-CoV-2 and its vaccine/immune-escaping variants continue to pose a serious threat to public health due to a paucity of effective, rapidly deployable, and widely available treatments. Here, we address these challenges by combining Pegasys (IFNα) and nafamostat to effectively suppress SARS-CoV-2 infection in cell culture and hamsters. Our results indicate that Serpin E1 is an important mediator of the antiviral activity of IFNα and that both Serpin E1 and nafamostat can target the same cellular factor TMPRSS2, which plays a critical role in viral replication. The low doses of the drugs in combination may have several clinical advantages, including fewer adverse events and improved patient outcome. Thus, our study may provide a proactive solution for the ongoing pandemic and potential future coronavirus outbreaks, which is still urgently required in many parts of the world.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Benzamidinas/farmacología , COVID-19/metabolismo , COVID-19/virología , Guanidinas/farmacología , Interferón-alfa/farmacología , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Benzamidinas/uso terapéutico , Cricetinae , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Guanidinas/uso terapéutico , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Interferón-alfa/uso terapéutico , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
13.
Arch Virol ; 166(7): 2005-2010, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33987753

RESUMEN

We show that ivermectin, an FDA-approved anti-parasitic drug, effectively inhibits infection with hepatitis E virus (HEV) genotypes 1 and 3 in a range of cell culture models, including hepatic and extrahepatic cells. Long-term treatment showed no clear evidence of the development of drug resistance. Gene silencing of importin-α1, a cellular target of ivermectin and a key member of the host nuclear transport complex, inhibited viral replication and largely abolished the anti-HEV effect of ivermectin.


Asunto(s)
Virus de la Hepatitis E/efectos de los fármacos , Hepatitis E/tratamiento farmacológico , Ivermectina/farmacología , Replicación Viral/efectos de los fármacos , alfa Carioferinas/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virología , Hepatitis E/metabolismo , Hepatitis E/virología , Humanos , Proteínas Nucleares/metabolismo
14.
Viruses ; 13(4)2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33918958

RESUMEN

Therapeutic options for coronaviruses remain limited. To address this unmet medical need, we screened 5406 compounds, including United States Food and Drug Administration (FDA)-approved drugs and bioactives, for activity against a South Korean Middle East respiratory syndrome coronavirus (MERS-CoV) clinical isolate. Among 221 identified hits, 54 had therapeutic indexes (TI) greater than 6, representing effective drugs. The time-of-addition studies with selected drugs demonstrated eight and four FDA-approved drugs which acted on the early and late stages of the viral life cycle, respectively. Confirmed hits included several cardiotonic agents (TI > 100), atovaquone, an anti-malarial (TI > 34), and ciclesonide, an inhalable corticosteroid (TI > 6). Furthermore, utilizing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we tested combinations of remdesivir with selected drugs in Vero-E6 and Calu-3 cells, in lung organoids, and identified ciclesonide, nelfinavir, and camostat to be at least additive in vitro. Our results identify potential therapeutic options for MERS-CoV infections, and provide a basis to treat coronavirus disease 2019 (COVID-19) and other coronavirus-related illnesses.


Asunto(s)
Antivirales/farmacología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Infecciones por Coronavirus/virología , Aprobación de Drogas , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Humanos , Estadios del Ciclo de Vida/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/crecimiento & desarrollo , Bibliotecas de Moléculas Pequeñas/farmacología , Tratamiento Farmacológico de COVID-19
15.
Biomolecules ; 11(1)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477863

RESUMEN

DNA in our cells is constantly modified by internal and external factors [...].


Asunto(s)
Daño del ADN , Animales , Antineoplásicos/farmacología , Reparación del ADN/genética , Modelos Animales de Enfermedad , Epistasis Genética/efectos de los fármacos , Humanos , Ratones
16.
Cell Rep Med ; 2(1): 100189, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33495758

RESUMEN

The SARS-CoV-2 proteome shares regions of conservation with endemic human coronaviruses (CoVs), but it remains unknown to what extent these may be cross-recognized by the antibody response. Here, we study cross-reactivity using a highly multiplexed peptide assay (PepSeq) to generate an epitope-resolved view of IgG reactivity across all human CoVs in both COVID-19 convalescent and negative donors. PepSeq resolves epitopes across the SARS-CoV-2 Spike and Nucleocapsid proteins that are commonly targeted in convalescent donors, including several sites also recognized in some uninfected controls. By comparing patterns of homologous reactivity between CoVs and using targeted antibody-depletion experiments, we demonstrate that SARS-CoV-2 elicits antibodies that cross-recognize pandemic and endemic CoV antigens at two Spike S2 subunit epitopes. We further show that these cross-reactive antibodies preferentially bind endemic homologs. Our findings highlight sites at which the SARS-CoV-2 response appears to be shaped by previous CoV exposures and which have the potential to raise broadly neutralizing responses.

17.
Antiviral Res ; 184: 104967, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33137361

RESUMEN

Exposure to hepatitis E virus (HEV) bears a high risk of developing chronic infection in immunocompromised patients, including organ transplant recipients and cancer patients. We aim to identify effective anti-HEV therapies through screening and repurposing safe-in-human broad-spectrum antiviral agents. In this study, a safe-in-human broad-spectrum antiviral drug library comprising of 94 agents was used. Upon screening, we identified gemcitabine, a widely used anti-cancer drug, as a potent inhibitor of HEV replication. The antiviral effect was confirmed in a range of cell culture models with genotype 1 and 3 HEV strains. As a cytidine analog, exogenous supplementation of pyrimidine nucleosides effectively reversed the antiviral activity of gemcitabine, but the level of pyrimidine nucleosides per se does not affect HEV replication. Surprisingly, similar to interferon-alpha (IFNα) treatment, gemcitabine activates STAT1 phosphorylation. This subsequently triggers activation of interferon-sensitive response element (ISRE) and transcription of interferon-stimulated genes (ISGs). Cytidine or uridine effectively inhibits gemcitabine-induced activation of ISRE and ISGs. As expected, JAK inhibitor 1 blocked IFNα, but not gemcitabine-induced STAT1 phosphorylation, ISRE/ISG activation, and anti-HEV activity. These effects of gemcitabine were completely lost in STAT1 knockout cells. In summary, gemcitabine potently inhibits HEV replication by triggering interferon-like response through STAT1 phosphorylation but independent of Janus kinases. This represents a non-canonical antiviral mechanism, which utilizes the innate defense machinery that is distinct from the classical interferon response. These results support repurposing gemcitabine for treating hepatitis E, especially for HEV-infected cancer patients, leading to dual anti-cancer and antiviral effects.


Asunto(s)
Desoxicitidina/análogos & derivados , Virus de la Hepatitis E/efectos de los fármacos , Interferón-alfa , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Antivirales/farmacología , Línea Celular , Desoxicitidina/farmacología , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Regulación de la Expresión Génica , Hepatitis E/tratamiento farmacológico , Virus de la Hepatitis E/fisiología , Interacciones Microbiota-Huesped , Humanos , Interferón-alfa/farmacología , Quinasas Janus/metabolismo , Ácido Micofenólico/antagonistas & inhibidores , Nucleósidos de Pirimidina/farmacología , Elementos de Respuesta , Ribavirina/antagonistas & inhibidores , Transducción de Señal , Replicación Viral/efectos de los fármacos , Gemcitabina
18.
Viruses ; 12(10)2020 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080984

RESUMEN

Combination therapies have become a standard for the treatment for HIV and hepatitis C virus (HCV) infections. They are advantageous over monotherapies due to better efficacy, reduced toxicity, as well as the ability to prevent the development of resistant viral strains and to treat viral co-infections. Here, we identify new synergistic combinations against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), echovirus 1 (EV1), hepatitis C virus (HCV) and human immunodeficiency virus 1 (HIV-1) in vitro. We observed synergistic activity of nelfinavir with convalescent serum and with purified neutralizing antibody 23G7 against SARS-CoV-2 in human lung epithelial Calu-3 cells. We also demonstrated synergistic activity of nelfinavir with EIDD-2801 or remdesivir in Calu-3 cells. In addition, we showed synergistic activity of vemurafenib with emetine, homoharringtonine, anisomycin, or cycloheximide against EV1 infection in human lung epithelial A549 cells. We also found that combinations of sofosbuvir with brequinar or niclosamide are synergistic against HCV infection in hepatocyte-derived Huh-7.5 cells, and that combinations of monensin with lamivudine or tenofovir are synergistic against HIV-1 infection in human cervical TZM-bl cells. These results indicate that synergy is achieved when a virus-directed antiviral is combined with another virus- or host-directed agent. Finally, we present an online resource that summarizes novel and known antiviral drug combinations and their developmental status.


Asunto(s)
Antivirales/administración & dosificación , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Células A549 , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Antineoplásicos/farmacología , Antivirales/farmacología , COVID-19 , Línea Celular , Infecciones por Coronavirus/virología , Bases de Datos Farmacéuticas , Combinación de Medicamentos , Descubrimiento de Drogas , Sinergismo Farmacológico , Enterovirus Humano B/efectos de los fármacos , VIH-1/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Humanos , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
19.
bioRxiv ; 2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-32743570

RESUMEN

A high-resolution understanding of the antibody response to SARS-CoV-2 is important for the design of effective diagnostics, vaccines and therapeutics. However, SARS-CoV-2 antibody epitopes remain largely uncharacterized, and it is unknown whether and how the response may cross-react with related viruses. Here, we use a multiplexed peptide assay ('PepSeq') to generate an epitope-resolved view of reactivity across all human coronaviruses. PepSeq accurately detects SARS-CoV-2 exposure and resolves epitopes across the Spike and Nucleocapsid proteins. Two of these represent recurrent reactivities to conserved, functionally-important sites in the Spike S2 subunit, regions that we show are also targeted for the endemic coronaviruses in pre-pandemic controls. At one of these sites, we demonstrate that the SARS-CoV-2 response strongly and recurrently cross-reacts with the endemic virus hCoV-OC43. Our analyses reveal new diagnostic and therapeutic targets, including a site at which SARS-CoV-2 may recruit common pre-existing antibodies and with the potential for broadly-neutralizing responses.

20.
Cancers (Basel) ; 12(6)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630560

RESUMEN

BACKGROUND: The evidence that pan-Bcl-2 or Bcl-xL-specific inhibitors prematurely kill virus-infected or RNA/DNA-transfected cells provides rationale for investigating these apoptotic inducers further. We hypothesized that not only invasive RNA or DNA (biological factors) but also DNA/RNA-damaging chemical or physical factors could trigger apoptosis that have been sensitized with pan-Bcl-2 or Bcl-xL-specific agents; Methods: We tested chemical and physical factors plus Bcl-xL-specific inhibitor A-1155463 in cells of various origins and the small roundworms (C. elegans); Results: We show that combination of a A-1155463 along with a DNA-damaging agent, 4-nitroquinoline-1-oxide (4NQO), prematurely kills cells of various origins as well as C. elegans. The synergistic effect is p53-dependent and associated with the release of Bad and Bax from Bcl-xL, which trigger mitochondrial outer membrane permeabilization. Furthermore, we found that combining Bcl-xL-specific inhibitors with various chemical compounds or physical insults also induced cell death; Conclusions: Thus, we were able to identify several biological, chemical and physical triggers of the evolutionarily conserved Bcl-xL-mediated apoptotic pathway, shedding light on strategies and targets for novel drug development.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA