Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 16(4)2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38675980

RESUMEN

Clofazimine and Arbidol have both been reported to be effective in vitro SARS-CoV-2 fusion inhibitors. Both are promising drugs that have been repurposed for the treatment of COVID-19 and have been used in several previous and ongoing clinical trials. Small-molecule bindings to expressed constructs of the trimeric S2 segment of Spike and the full-length SARS-CoV-2 Spike protein were measured using a Surface Plasmon Resonance (SPR) binding assay. We demonstrate that Clofazimine, Toremifene, Arbidol and its derivatives bind to the S2 segment of the Spike protein. Clofazimine provided the most reliable and highest-quality SPR data for binding with S2 over the conditions explored. A molecular docking approach was used to identify the most favorable binding sites on the S2 segment in the prefusion conformation, highlighting two possible small-molecule binding sites for fusion inhibitors. Results related to molecular docking and modeling of the structure-activity relationship (SAR) of a newly reported series of Clofazimine derivatives support the proposed Clofazimine binding site on the S2 segment. When the proposed Clofazimine binding site is superimposed with other experimentally determined coronavirus structures in structure-sequence alignments, the changes in sequence and structure may rationalize the broad-spectrum antiviral activity of Clofazimine in closely related coronaviruses such as SARS-CoV, MERS, hCoV-229E, and hCoV-OC43.


Asunto(s)
Clofazimina , Indoles , Simulación del Acoplamiento Molecular , Unión Proteica , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Sulfuros , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Clofazimina/farmacología , Clofazimina/química , Clofazimina/metabolismo , SARS-CoV-2/efectos de los fármacos , Humanos , Sitios de Unión , Antivirales/farmacología , Antivirales/química , Tratamiento Farmacológico de COVID-19 , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Inhibidores de Proteínas Virales de Fusión/farmacología , Inhibidores de Proteínas Virales de Fusión/química
2.
Antiviral Res ; 206: 105399, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36007601

RESUMEN

Filoviruses enter cells through macropinocytosis and trafficking into the endosomes in which they bind to the receptor Niemann-Pick C1 protein (NPC1) for membrane fusion and entry into the cytoplasm. The endosomal receptor-binding is critical step for filovirus entry. Designing inhibitors to block receptor binding will prevent viral entry. Using available binding structural information from the co-crystal structures of the viral GP with the receptor NPC1 or with monoclonal antibodies, we have conducted structure-based design of peptide inhibitors to target the receptor binding site (RBS). The designed peptides were tested for their inhibition activity against pseudo-typed or replication-competent viruses in a cell-based assay. The results indicate that these peptides exhibited strong activities against both Ebola and Marburg virus infection. It is expected that these peptides can be further developed for therapeutic use to treat filovirus infection and combat the outbreaks.


Asunto(s)
Filoviridae , Receptores Virales , Inhibidores de Proteínas Virales de Fusión , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Ebolavirus/fisiología , Endosomas/metabolismo , Filoviridae/química , Filoviridae/efectos de los fármacos , Fiebre Hemorrágica Ebola , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ligandos , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/farmacología , Internalización del Virus/efectos de los fármacos
3.
Viruses ; 14(4)2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35458385

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by infection of SARS-CoV-2 and its variants has posed serious threats to global public health, thus calling for the development of potent and broad-spectrum antivirals. We previously designed and developed a peptide-based pan-coronavirus (CoV) fusion inhibitor, EK1, which is effective against all human CoVs (HCoV) tested by targeting the HCoV S protein HR1 domain. However, its relatively short half-life may limit its clinical use. Therefore, we designed, constructed, and expressed a recombinant protein, FL-EK1, which consists of a modified fibronectin type III domain (FN3) with albumin-binding capacity, a flexible linker, and EK1. As with EK1, we found that FL-EK1 could also effectively inhibit infection of SARS-CoV-2 and its variants, as well as HCoV-OC43. Furthermore, it protected mice from infection by the SARS-CoV-2 Delta variant and HCoV-OC43. Importantly, the half-life of FL-EK1 (30 h) is about 15.7-fold longer than that of EK1 (1.8 h). These results suggest that FL-EK1 is a promising candidate for the development of a pan-CoV fusion inhibitor-based long-acting antiviral drug for preventing and treating infection by current and future SARS-CoV-2 variants, as well as other HCoVs.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Inhibidores de Proteínas Virales de Fusión , Animales , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Dominio de Fibronectina del Tipo III , Semivida , Ratones , Proteínas Recombinantes de Fusión , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/farmacología
4.
FEBS Open Bio ; 11(12): 3253-3261, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34710289

RESUMEN

A key step in infections by enveloped viruses, such as influenza, is the fusion between the viral envelope and the host cell membrane, which allows the virus to insert its genetic material into the host cell and replicate. The influenza virus fusion process is promoted by hemagglutinin (HA), a glycoprotein that contains three identical monomers composed of two polypeptide chains (HA1 and HA2). Early studies on this protein revealed that HA-mediated fusion involves the insertion of the HA2 N-terminal segment into the host membrane and that this segment, known as the fusion peptide, is a key player in the fusion process. This mini-review highlights the main findings that have been obtained by experimental and computational studies on the HA fusion peptide, which give us a glimpse of its mode of action.


Asunto(s)
Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Proteínas Virales de Fusión/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Membranas , Modelos Moleculares , Péptidos , Conformación Proteica , Inhibidores de Proteínas Virales de Fusión/farmacología , Proteínas Virales de Fusión/genética
5.
Viruses ; 13(9)2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34578344

RESUMEN

Lassa virus (LASV)-a member of the family Arenaviridae-causes Lassa fever in humans and is endemic in West Africa. Currently, no approved drugs are available. We screened 2480 small compounds for their potential antiviral activity using pseudotyped vesicular stomatitis virus harboring the LASV glycoprotein (VSV-LASVGP) and a related prototypic arenavirus, lymphocytic choriomeningitis virus (LCMV). Follow-up studies confirmed that CP100356 hydrochloride (CP100356), a specific P-glycoprotein (P-gp) inhibitor, suppressed VSV-LASVGP, LCMV, and LASV infection with half maximal inhibitory concentrations of 0.52, 0.54, and 0.062 µM, respectively, without significant cytotoxicity. Although CP100356 did not block receptor binding at the cell surface, it inhibited low-pH-dependent membrane fusion mediated by arenavirus glycoproteins. P-gp downregulation did not cause a significant reduction in either VSV-LASVGP or LCMV infection, suggesting that P-gp itself is unlikely to be involved in arenavirus entry. Finally, our data also indicate that CP100356 inhibits the infection by other mammarenaviruses. Thus, our findings suggest that CP100356 can be considered as an effective virus entry inhibitor for LASV and other highly pathogenic mammarenaviruses.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Arenaviridae/metabolismo , Isoquinolinas/farmacología , Virus Lassa/efectos de los fármacos , Quinazolinas/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/farmacología , Chlorocebus aethiops , Humanos , Fiebre de Lassa/tratamiento farmacológico , Fiebre de Lassa/virología , Virus de la Coriomeningitis Linfocítica , Receptores Virales , Células Vero , Estomatitis Vesicular/virología , Inhibidores de Proteínas Virales de Fusión/farmacología
6.
Molecules ; 26(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946996

RESUMEN

Respiratory syncytial virus (RSV) is a major pathogen that causes severe lower respiratory tract infection in infants, the elderly and the immunocompromised worldwide. At present no approved specific drugs or vaccines are available to treat this pathogen. Recently, several promising candidates targeting RSV entry and multiplication steps are under investigation. However, it is possible to lead to drug resistance under the long-term treatment. Therapeutic combinations constitute an alternative to prevent resistance and reduce antiviral doses. Therefore, we tested in vitro two-drug combinations of fusion inhibitors (GS5806, Ziresovir and BMS433771) and RNA-dependent RNA polymerase complex (RdRp) inhibitors (ALS8176, RSV604, and Cyclopamine). The statistical program MacSynergy II was employed to determine synergism, additivity or antagonism between drugs. From the result, we found that combinations of ALS8176 and Ziresovir or GS5806 exhibit additive effects against RSV in vitro, with interaction volume of 50 µM2% and 31 µM2% at 95% confidence interval, respectively. On the other hand, all combinations between fusion inhibitors showed antagonistic effects against RSV in vitro, with volume of antagonism ranging from -50 µM2 % to -176 µM2 % at 95% confidence interval. Over all, our results suggest the potentially therapeutic combinations in combating RSV in vitro could be considered for further animal and clinical evaluations.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Antivirales/química , Antivirales/uso terapéutico , Descubrimiento de Drogas/métodos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Quinazolinas/química , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/virología , Bibliotecas de Moléculas Pequeñas , Sulfonas , Tiazepinas/química , Tiazepinas/farmacología , Tiazepinas/uso terapéutico , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/farmacología , Inhibidores de Proteínas Virales de Fusión/uso terapéutico
7.
Science ; 371(6536): 1379-1382, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33597220

RESUMEN

Containment of the COVID-19 pandemic requires reducing viral transmission. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is initiated by membrane fusion between the viral and host cell membranes, which is mediated by the viral spike protein. We have designed lipopeptide fusion inhibitors that block this critical first step of infection and, on the basis of in vitro efficacy and in vivo biodistribution, selected a dimeric form for evaluation in an animal model. Daily intranasal administration to ferrets completely prevented SARS-CoV-2 direct-contact transmission during 24-hour cohousing with infected animals, under stringent conditions that resulted in infection of 100% of untreated animals. These lipopeptides are highly stable and thus may readily translate into safe and effective intranasal prophylaxis to reduce transmission of SARS-CoV-2.


Asunto(s)
COVID-19/transmisión , Lipopéptidos/administración & dosificación , Fusión de Membrana/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Inhibidores de Proteínas Virales de Fusión/administración & dosificación , Internalización del Virus/efectos de los fármacos , Administración Intranasal , Animales , COVID-19/prevención & control , COVID-19/virología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Diseño de Fármacos , Hurones , Lipopéptidos/química , Lipopéptidos/farmacocinética , Lipopéptidos/farmacología , Ratones , Profilaxis Pre-Exposición , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Distribución Tisular , Células Vero , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/farmacocinética , Inhibidores de Proteínas Virales de Fusión/farmacología
9.
J Gen Virol ; 102(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054904

RESUMEN

Although enveloped viruses canonically mediate particle entry through virus-cell fusion, certain viruses can spread by cell-cell fusion, brought about by receptor engagement and triggering of membrane-bound, viral-encoded fusion proteins on the surface of cells. The formation of pathogenic syncytia or multinucleated cells is seen in vivo, but their contribution to viral pathogenesis is poorly understood. For the negative-strand paramyxoviruses respiratory syncytial virus (RSV) and Nipah virus (NiV), cell-cell spread is highly efficient because their oligomeric fusion protein complexes are active at neutral pH. The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has also been reported to induce syncytia formation in infected cells, with the spike protein initiating cell-cell fusion. Whilst it is well established that fusion protein-specific antibodies can block particle attachment and/or entry into the cell (canonical virus neutralization), their capacity to inhibit cell-cell fusion and the consequences of this neutralization for the control of infection are not well characterized, in part because of the lack of specific tools to assay and quantify this activity. Using an adapted bimolecular fluorescence complementation assay, based on a split GFP-Renilla luciferase reporter, we have established a micro-fusion inhibition test (mFIT) that allows the identification and quantification of these neutralizing antibodies. This assay has been optimized for high-throughput use and its applicability has been demonstrated by screening monoclonal antibody (mAb)-mediated inhibition of RSV and NiV fusion and, separately, the development of fusion-inhibitory antibodies following NiV vaccine immunization in pigs. In light of the recent emergence of coronavirus disease 2019 (COVID-19), a similar assay was developed for SARS-CoV-2 and used to screen mAbs and convalescent patient plasma for fusion-inhibitory antibodies. Using mFITs to assess antibody responses following natural infection or vaccination is favourable, as this assay can be performed entirely at low biocontainment, without the need for live virus. In addition, the repertoire of antibodies that inhibit cell-cell fusion may be different to those that inhibit particle entry, shedding light on the mechanisms underpinning antibody-mediated neutralization of viral spread.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , COVID-19/diagnóstico , Infecciones por Henipavirus/diagnóstico , Ensayos Analíticos de Alto Rendimiento , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/aislamiento & purificación , Anticuerpos Antivirales/metabolismo , COVID-19/inmunología , COVID-19/virología , Fusión Celular , Convalecencia , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Humanos , Sueros Inmunes/química , Luciferasas/genética , Luciferasas/metabolismo , Modelos Moleculares , Virus Nipah/inmunología , Virus Nipah/patogenicidad , Conformación Proteica , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/patogenicidad , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Porcinos , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/metabolismo , Inhibidores de Proteínas Virales de Fusión/farmacología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
10.
J Med Chem ; 63(15): 8043-8045, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32786236

RESUMEN

Inhibitors of the respiratory syncytial virus (RSV) fusion protein block entry of the virus into the cell and have shown varying efficacy in a human challenge model of RSV disease. Trials in patient populations are yet to show significant benefits. Jonckers et al. ( J. Med. Chem. 2020, DOI: 10.1021/acs.jmedchem.0c00226) describe the discovery of JNJ-53718678 which can now claim the leading position in clinical evaluation. For RSV inhibitors, the current status of the clinical development of the compound is discussed.


Asunto(s)
Antivirales/química , Imidazolidinas/química , Indoles/química , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Inhibidores de Proteínas Virales de Fusión/química , Antivirales/farmacología , Antivirales/uso terapéutico , Cristalografía por Rayos X/métodos , Humanos , Imidazolidinas/farmacología , Imidazolidinas/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/fisiología , Resultado del Tratamiento , Inhibidores de Proteínas Virales de Fusión/farmacología
11.
Int J Mol Sci ; 21(11)2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32481690

RESUMEN

In the past 17 years, three novel coronaviruses have caused severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and the coronavirus disease 2019 (COVID-19). As emerging infectious diseases, they were characterized by their novel pathogens and transmissibility without available clinical drugs or vaccines. This is especially true for the newly identified COVID-19 caused by SARS coronavirus 2 (SARS-CoV-2) for which, to date, no specific antiviral drugs or vaccines have been approved. Similar to SARS and MERS, the lag time in the development of therapeutics is likely to take months to years. These facts call for the development of broad-spectrum anti-coronavirus drugs targeting a conserved target site. This review will systematically describe potential broad-spectrum coronavirus fusion inhibitors, including antibodies, protease inhibitors, and peptide fusion inhibitors, along with a discussion of their advantages and disadvantages.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Inhibidores de Proteínas Virales de Fusión/uso terapéutico , Animales , Betacoronavirus/fisiología , Humanos , Inhibidores de Proteasas/efectos adversos , Inhibidores de Proteasas/farmacología , SARS-CoV-2 , Inhibidores de Proteínas Virales de Fusión/efectos adversos , Inhibidores de Proteínas Virales de Fusión/farmacología , Internalización del Virus/efectos de los fármacos
13.
J Am Chem Soc ; 141(32): 12648-12656, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31268705

RESUMEN

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) cause lower respiratory infection in infants and young children. There are no vaccines for these pathogens, and existing treatments have limited or questionable efficacy. Infection by HPIV3 or RSV requires fusion of the viral and cell membranes, a process mediated by a trimeric fusion glycoprotein (F) displayed on the viral envelope. Once triggered, the pre-fusion form of F undergoes a series of conformational changes that first extend the molecule to allow for insertion of the hydrophobic fusion peptide into the target cell membrane and then refold the trimeric assembly into an energetically stable post-fusion state, a process that drives the merger of the viral and host cell membranes. Peptides derived from defined regions of HPIV3 F inhibit infection by HPIV3 by interfering with the structural transitions of the trimeric F assembly. Here we describe lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F that potently inhibit infection by both HPIV3 and RSV. The lead peptide inhibits RSV infection as effectively as does a peptide corresponding to the RSV HRC domain itself. We show that the inhibitors bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F with high affinity. Co-crystal structures of inhibitors bound to the HRN domains of HPIV3 or RSV F reveal remarkably different modes of binding in the N-terminal segment of the inhibitor.


Asunto(s)
Lipopéptidos/farmacología , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Inhibidores de Proteínas Virales de Fusión/farmacología , Proteínas Virales de Fusión/farmacología , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Lipopéptidos/metabolismo , Pruebas de Sensibilidad Microbiana , Virus de la Parainfluenza 3 Humana/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Mucosa Respiratoria/virología , Virus Sincitiales Respiratorios/química , Inhibidores de Proteínas Virales de Fusión/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
14.
Sci Adv ; 5(4): eaav4580, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30989115

RESUMEN

Continuously emerging highly pathogenic human coronaviruses (HCoVs) remain a major threat to human health, as illustrated in past SARS-CoV and MERS-CoV outbreaks. The development of a drug with broad-spectrum HCoV inhibitory activity would address this urgent unmet medical need. Although previous studies have suggested that the HR1 of HCoV spike (S) protein is an important target site for inhibition against specific HCoVs, whether this conserved region could serve as a target for the development of broad-spectrum pan-CoV inhibitor remains controversial. Here, we found that peptide OC43-HR2P, derived from the HR2 domain of HCoV-OC43, exhibited broad fusion inhibitory activity against multiple HCoVs. EK1, the optimized form of OC43-HR2P, showed substantially improved pan-CoV fusion inhibitory activity and pharmaceutical properties. Crystal structures indicated that EK1 can form a stable six-helix bundle structure with both short α-HCoV and long ß-HCoV HR1s, further supporting the role of HR1 region as a viable pan-CoV target site.


Asunto(s)
Infecciones por Coronavirus/virología , Coronavirus/efectos de los fármacos , Coronavirus/metabolismo , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/química , Inhibidores de Proteínas Virales de Fusión/química , Inhibidores de Proteínas Virales de Fusión/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular , Coronavirus/clasificación , Coronavirus/genética , Infecciones por Coronavirus/tratamiento farmacológico , Modelos Animales de Enfermedad , Humanos , Fusión de Membrana/efectos de los fármacos , Ratones , Modelos Moleculares , Péptidos/química , Péptidos/farmacología , Filogenia , Conformación Proteica , Dominios Proteicos , Solubilidad
15.
Science ; 363(6431)2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30846569

RESUMEN

Recent characterization of broadly neutralizing antibodies (bnAbs) against influenza virus identified the conserved hemagglutinin (HA) stem as a target for development of universal vaccines and therapeutics. Although several stem bnAbs are being evaluated in clinical trials, antibodies are generally unsuited for oral delivery. Guided by structural knowledge of the interactions and mechanism of anti-stem bnAb CR6261, we selected and optimized small molecules that mimic the bnAb functionality. Our lead compound neutralizes influenza A group 1 viruses by inhibiting HA-mediated fusion in vitro, protects mice against lethal and sublethal influenza challenge after oral administration, and effectively neutralizes virus infection in reconstituted three-dimensional cell culture of fully differentiated human bronchial epithelial cells. Cocrystal structures with H1 and H5 HAs reveal that the lead compound recapitulates the bnAb hotspot interactions.


Asunto(s)
Anticuerpos Neutralizantes/química , Materiales Biomiméticos/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/prevención & control , Piperazinas/farmacología , Piridinas/farmacología , Tetrazoles/farmacología , Inhibidores de Proteínas Virales de Fusión/farmacología , Internalización del Virus/efectos de los fármacos , Administración Oral , Animales , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/farmacocinética , Bronquios/virología , Células Cultivadas , Perros , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Células de Riñón Canino Madin Darby , Ratones , Piperazinas/administración & dosificación , Piperazinas/farmacocinética , Piridinas/administración & dosificación , Piridinas/farmacocinética , Mucosa Respiratoria/virología , Tetrazoles/administración & dosificación , Tetrazoles/farmacocinética , Inhibidores de Proteínas Virales de Fusión/administración & dosificación , Inhibidores de Proteínas Virales de Fusión/farmacocinética
17.
J Theor Biol ; 456: 62-73, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30048719

RESUMEN

Respiratory syncytial virus (RSV) is a respiratory infection that can cause serious illness, particularly in infants. In this study, we test four different model implementations for the effect of a fusion inhibitor, including one model that combines different drug effects, by fitting the models to data from a study of TMC353121 in African green monkeys. We use mathematical modeling to estimate the drug efficacy parameters, εmax, the maximum efficacy of the drug, and EC50, the drug concentration needed to achieve half the maximum effect. We find that if TMC353121 is having multiple effects on viral kinetics, more detailed data, using different treatment delays, is needed to detect this effect.


Asunto(s)
Bencimidazoles/uso terapéutico , Modelos Biológicos , Piridinas/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/efectos de los fármacos , Inhibidores de Proteínas Virales de Fusión/uso terapéutico , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Piridinas/administración & dosificación , Piridinas/farmacología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/aislamiento & purificación , Virus Sincitiales Respiratorios/fisiología , Inhibidores de Proteínas Virales de Fusión/administración & dosificación , Inhibidores de Proteínas Virales de Fusión/farmacología , Carga Viral , Replicación Viral/efectos de los fármacos
18.
Int J Mol Sci ; 19(2)2018 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-29415501

RESUMEN

Human coronavirus 229E (HCoV-229E) infection in infants, elderly people, and immunocompromised patients can cause severe disease, thus calling for the development of effective and safe therapeutics to treat it. Here we reported the design, synthesis and characterization of two peptide-based membrane fusion inhibitors targeting HCoV-229E spike protein heptad repeat 1 (HR1) and heptad repeat 2 (HR2) domains, 229E-HR1P and 229E-HR2P, respectively. We found that 229E-HR1P and 229E-HR2P could interact to form a stable six-helix bundle and inhibit HCoV-229E spike protein-mediated cell-cell fusion with IC50 of 5.7 and 0.3 µM, respectively. 229E-HR2P effectively inhibited pseudotyped and live HCoV-229E infection with IC50 of 0.5 and 1.7 µM, respectively. In a mouse model, 229E-HR2P administered intranasally could widely distribute in the upper and lower respiratory tracts and maintain its fusion-inhibitory activity. Therefore, 229E-HR2P is a promising candidate for further development as an antiviral agent for the treatment and prevention of HCoV-229E infection.


Asunto(s)
Coronavirus Humano 229E/efectos de los fármacos , Péptidos/farmacología , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/química , Inhibidores de Proteínas Virales de Fusión/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Células Gigantes/virología , Humanos , Fusión de Membrana , Ratones , Péptidos/química , Unión Proteica , Inhibidores de Proteínas Virales de Fusión/química
19.
Sci Rep ; 8(1): 2769, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29426822

RESUMEN

Current anti-hepatitis B virus (HBV) agents including interferons and nucleos(t)ide analogs efficiently suppress HBV infection. However, as it is difficult to eliminate HBV from chronically infected liver, alternative anti-HBV agents targeting a new molecule are urgently needed. In this study, we applied a chemical array to high throughput screening of small molecules that interacted with sodium taurocholate cotransporting polypeptide (NTCP), an entry receptor for HBV. From approximately 30,000 compounds, we identified 74 candidates for NTCP interactants, and five out of these were shown to inhibit HBV infection in cell culture. One of such compound, NPD8716, a coumarin derivative, interacted with NTCP and inhibited HBV infection without causing cytotoxicity. Consistent with its NTCP interaction capacity, this compound was shown to block viral attachment to host hepatocytes. NPD8716 also prevented the infection with hepatitis D virus, but not hepatitis C virus, in agreement with NPD8716 specifically inhibiting NTCP-mediated infection. Analysis of derivative compounds showed that the anti-HBV activity of compounds was apparently correlated with the affinity to NTCP and the capacity to impair NTCP-mediated bile acid uptake. These results are the first to show that the chemical array technology represents a powerful platform to identify novel viral entry inhibitors.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Transportadores de Anión Orgánico Sodio-Dependiente/agonistas , Simportadores/agonistas , Inhibidores de Proteínas Virales de Fusión/aislamiento & purificación , Inhibidores de Proteínas Virales de Fusión/farmacología , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Ácidos y Sales Biliares/metabolismo , Cumarinas/química , Cumarinas/aislamiento & purificación , Cumarinas/farmacología , Células Hep G2 , Hepacivirus/efectos de los fármacos , Virus de la Hepatitis Delta/efectos de los fármacos , Humanos , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Inhibidores de Proteínas Virales de Fusión/química
20.
Nat Commun ; 8(1): 167, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28761099

RESUMEN

Respiratory syncytial virus is a major cause of acute lower respiratory tract infection in young children, immunocompromised adults, and the elderly. Intervention with small-molecule antivirals specific for respiratory syncytial virus presents an important therapeutic opportunity, but no such compounds are approved today. Here we report the structure of JNJ-53718678 bound to respiratory syncytial virus fusion (F) protein in its prefusion conformation, and we show that the potent nanomolar activity of JNJ-53718678, as well as the preliminary structure-activity relationship and the pharmaceutical optimization strategy of the series, are consistent with the binding mode of JNJ-53718678 and other respiratory syncytial virus fusion inhibitors. Oral treatment of neonatal lambs with JNJ-53718678, or with an equally active close analog, efficiently inhibits established acute lower respiratory tract infection in the animals, even when treatment is delayed until external signs of respiratory syncytial virus illness have become visible. Together, these data suggest that JNJ-53718678 is a promising candidate for further development as a potential therapeutic in patients at risk to develop respiratory syncytial virus acute lower respiratory tract infection.Respiratory syncytial virus causes lung infections in children, immunocompromised adults, and in the elderly. Here the authors show that a chemical inhibitor to a viral fusion protein is effective in reducing viral titre and ameliorating infection in rodents and neonatal lambs.


Asunto(s)
Imidazolidinas/metabolismo , Indoles/metabolismo , Virus Sincitial Respiratorio Humano/metabolismo , Inhibidores de Proteínas Virales de Fusión/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Chlorocebus aethiops , Células Epiteliales , Humanos , Imidazolidinas/farmacología , Imidazolidinas/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Estructura Molecular , Neumonía Viral/tratamiento farmacológico , Ratas , Mucosa Respiratoria/citología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/metabolismo , Ovinos , Relación Estructura-Actividad , Células Vero , Inhibidores de Proteínas Virales de Fusión/farmacología , Inhibidores de Proteínas Virales de Fusión/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...