Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-32152074

RESUMEN

In the past few decades, enterovirus A71 (EVA71) has caused devastating outbreaks in the Asia-Pacific region, resulting in serious sequelae in infected young children. No preventive or therapeutic interventions are currently available for curing EVA71 infection, highlighting a great unmet medical need for this disease. Here, we showed that one novel single-domain antibody (sdAb), F1, isolated from an immunized llama, could alleviate EVA71 infection both in vitro and in vivo We also confirmed that the sdAb clone F1 recognizes EVA71 through a novel conformational epitope comprising the highly conserved region of VP3 capsid protein by using competitive-binding and overlapping-peptide enzyme-linked immunosorbent assays (ELISAs). Because of the virion's icosahedral structure, we reasoned that adjacent epitopes must be clustered within molecular ranges that may be simultaneously bound by an engineered antibody with multiple valency. Therefore, two single-domain binding modules (F1) were fused to generate an sdAb-in-tandem design so that the capture of viral antigens could be further increased by valency effects. We showed that the tetravalent construct F1×F1-hFc, containing two sdAb-in-tandem on a fragment crystallizable (Fc) scaffold, exhibits more potent neutralization activity against EVA71 than does the bivalent sdAb F1-hFc by at least 5.8-fold. We also demonstrated that, using a human scavenger receptor class B member 2 (hSCARB2) transgenic mouse model, a half dose of the F1×F1-hFc provided better protection against EVA71 infection than did the F1-hFc. Thus, our study furnishes important insights into multivalent sdAb engineering against viral infection and provides a novel strategic deployment approach for preparedness of emerging infectious diseases such as EVA71.


Asunto(s)
Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/inmunología , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/terapia , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , Antígenos Virales/inmunología , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/prevención & control , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Ratones , Ratones Transgénicos , Anticuerpos de Dominio Único/farmacología
2.
Artículo en Inglés | MEDLINE | ID: mdl-32669265

RESUMEN

The coronavirus (CoV) disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome CoV-2 (SARS-CoV-2) is a health threat worldwide. Viral main protease (Mpro, also called 3C-like protease [3CLpro]) is a therapeutic target for drug discovery. Herein, we report that GC376, a broad-spectrum inhibitor targeting Mpro in the picornavirus-like supercluster, is a potent inhibitor for the Mpro encoded by SARS-CoV-2, with a half-maximum inhibitory concentration (IC50) of 26.4 ± 1.1 nM. In this study, we also show that GC376 inhibits SARS-CoV-2 replication with a half-maximum effective concentration (EC50) of 0.91 ± 0.03 µM. Only a small portion of SARS-CoV-2 Mpro was covalently modified in the excess of GC376 as evaluated by mass spectrometry analysis, indicating that improved inhibitors are needed. Subsequently, molecular docking analysis revealed that the recognition and binding groups of GC376 within the active site of SARS-CoV-2 Mpro provide important new information for the optimization of GC376. Given that sufficient safety and efficacy data are available for GC376 as an investigational veterinary drug, expedited development of GC376, or its optimized analogues, for treatment of SARS-CoV-2 infection in human is recommended.


Asunto(s)
Antivirales/química , Betacoronavirus/efectos de los fármacos , Cisteína Endopeptidasas/química , Inhibidores de Proteasas/química , Pirrolidinas/química , Proteínas no Estructurales Virales/química , Secuencias de Aminoácidos , Animales , Antivirales/farmacología , Betacoronavirus/patogenicidad , Dominio Catalítico , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Expresión Génica , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/farmacología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Pirrolidinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , SARS-CoV-2 , Ácidos Sulfónicos , Termodinámica , Células Vero , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
3.
Nano Lett ; 14(1): 134-8, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24351061

RESUMEN

Noble metal nanoparticles (e.g., gold and platinum) supported on TiO2 surfaces are utilized in many technological applications such as heterogeneous catalysts. To fully understand their enhanced catalytic activity, it is essential to unravel the interfacial interaction between the metal atoms and TiO2 surfaces at the level of atomic dimensions. However, it has been extremely difficult to directly characterize the atomic-scale structures that result when individual metal atoms are adsorbed on the TiO2 surfaces. Here, we show direct atomic-resolution images of individual Pt atoms adsorbed on TiO2 (110) surfaces using aberration-corrected scanning transmission electron microscopy. Subangstrom spatial resolution enables us to identify five different Pt atom adsorption sites on the TiO2 (110) surface. Combining this with systematic density functional theory calculations reveals that the most favorable Pt adsorption sites are on vacancy sites of basal oxygen atoms that are located in subsurface positions relative to the top surface bridging oxygen atoms.

4.
BMC Biotechnol ; 12: 83, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23134743

RESUMEN

BACKGROUND: There are few studies that have examined the potential of RNA inference (RNAi) to increase protein production in the baculovirus expression vector system (BEVS). Spodoptera frugiperda (fall armyworm) (Sf)-caspase-1-repressed stable cells exhibit resistance to apoptosis and enhancement of recombinant protein production. However, the mechanism of recombinant protein augmentation in baculovirus-infected Caspase-repressed insect cells has not been elucidated. RESULTS: In the current study, we utilized RNAi-mediated Sf-caspase-1-repressed stable cells to clarify how the resistance to apoptosis can enhance both intracellular (firefly luciferase) and extracellular (secreted alkaline phosphatase [SEAP]) recombinant protein production in BEVS. Since the expression of molecular chaperones is strongly associated with the maximal production of exogenous proteins in BEVS, the differential expression of molecular chaperones in baculovirus-infected stable cells was also analyzed in this study. CONCLUSION: The data indicated that the retention of expression of molecular chaperones in baculovirus-infected Sf-caspase-1-repressed stable cells give the higher recombinant protein accumulation.


Asunto(s)
Caspasas/metabolismo , Chaperonas Moleculares/metabolismo , Animales , Baculoviridae/genética , Caspasas/química , Caspasas/genética , Chaperonas Moleculares/genética , Plásmidos/genética , Plásmidos/metabolismo , Interferencia de ARN , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Células Sf9 , Spodoptera , Transfección
5.
J Nat Prod ; 74(4): 580-4, 2011 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-21391660

RESUMEN

Diosgenin (3ß-hydroxy-5-spirostene, 1), a plant-derived sapogenin, is used as a dietary supplement. However, the biological effects of 1 related to viral replication remain unexplored. In this study, the effects of 1 on hepatitis C virus (HCV) replication were evaluated. Based on a reporter-based HCV subgenomic replicon system, 1 was found to inhibit HCV replication at low micromolar concentrations. The EC(50) (concentration at which 50% of HCV replication is inhibited) of 1 was 3.8 µM. No cellular toxicity was observed at this concentration. Diosgenin (1) also significantly reduced the levels of viral RNA and viral proteins as evaluated by quantitative real-time reverse transcriptase PCR and Western blot analysis, respectively. In addition, in an alternative HCV antiviral system more closely aligned to all steps involved in the HCV infection and life cycle, 1 totally abolished HCV replication at 20 µM. Moreover, 1 reduced the phosphorylation of signal transducer and activator of transcription 3. A combination of 1 and interferon-α exerted an additive effect on the resultant anti-HCV activity.


Asunto(s)
Antivirales/farmacología , Diosgenina/farmacología , Hepacivirus/efectos de los fármacos , Sapogeninas/farmacología , Antivirales/química , Antivirales/aislamiento & purificación , Suplementos Dietéticos/análisis , Diosgenina/química , Diosgenina/aislamiento & purificación , Hepacivirus/genética , Humanos , Estructura Molecular , ARN Viral/análisis , ARN Viral/efectos de los fármacos , Sapogeninas/química , Sapogeninas/aislamiento & purificación , Proteínas no Estructurales Virales/efectos de los fármacos
6.
Sci Rep ; 10(1): 8929, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488021

RESUMEN

Zika virus (ZIKV) of the flaviviridae family, is the cause of emerging infections characterized by fever, Guillain-Barré syndrome (GBS) in adults and microcephaly in newborns. There exists an urgent unmet clinical need for anti-ZIKV drugs for the treatment of infected individuals. In the current work, we aimed at the promising virus drug target, ZIKV NS3 protease and constructed a Pharmacophore Anchor (PA) model for the active site. The PA model reveals a total of 12 anchors (E, H, V) mapped across the active site subpockets. We further identified five of these anchors to be critical core anchors (CEH1, CH3, CH7, CV1, CV3) conserved across flaviviral proteases. The ZIKV protease PA model was then applied in anchor-enhanced virtual screening yielding 14 potential antiviral candidates, which were tested by in vitro assays. We discovered FDA drugs Asunaprevir and Simeprevir to have potent anti-ZIKV activities with EC50 values 4.7 µM and 0.4 µM, inhibiting the viral protease with IC50 values 6.0 µM and 2.6 µM respectively. Additionally, the PA model anchors aided in the exploration of inhibitor binding mechanisms. In conclusion, our PA model serves as a promising guide map for ZIKV protease targeted drug discovery and the identified 'previr' FDA drugs are promising for anti-ZIKV treatments.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas/métodos , Serina Endopeptidasas/efectos de los fármacos , Proteínas Virales/efectos de los fármacos , Virus Zika/efectos de los fármacos , Dominio Catalítico/efectos de los fármacos , Modelos Químicos , Simulación del Acoplamiento Molecular , Alineación de Secuencia , Virus Zika/enzimología , Virus Zika/genética
7.
Anal Biochem ; 377(1): 89-94, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18358823

RESUMEN

Epidermal growth factor receptor (EGFR) is a valid drug target for development of target-based therapeutics against non-small-cell lung cancer. In this study, we established a high-throughput cell-based assay to screen for compounds that may inhibit EGFR activation and/or EGFR-mediated downstream signaling pathway. This drug screening platform is based on the characterization of an EGFR-transfected 32D cell line (32D-EGFR). The expression of EGFR in 32D cells allowed cell proliferation in the presence of either epidermal growth factor (EGF) or interleukin 3 (IL-3) and provided a system for both screening and counterscreening of EGFR pathway-inhibitory compounds. After the completion of primary and secondary screenings in which 32D-EGFR cells were grown under the stimulation of either EGF or IL-3, 9 of 20,000 compounds were found to selectively inhibit the EGF-dependent proliferation, but not the IL-3-dependent proliferation, of 32D-EGFR cells. Subsequent analysis showed that 3 compounds of the 9 initial hits directly inhibited the kinase activity of recombinant EGFR in vitro and the phosphorylation of EGFR in H1299 cells transfected with EGFR. Thus, this 32D-EGFR assay system provides a promising approach for identifying novel EGFR and EGFR signaling pathway inhibitors with potential antitumor activity.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Fosforilación/efectos de los fármacos , Reproducibilidad de los Resultados
8.
Bioorg Med Chem Lett ; 18(5): 1623-7, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18243698

RESUMEN

Tricyclic 6,7-dihydro-4H-pyrazolo[1,5-a]pyrrolo[3,4-d]pyrimidine-5,8-dione was identified as a novel scaffold for Aurora kinase A inhibition through virtual screening. SAR exploration coupled with molecular modeling of 8a reveals the minimum pharmacophore requirements for Aurora kinase A inhibition.


Asunto(s)
Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/química , Pirimidinonas/química , Pirroles/química , Aurora Quinasas , Sitios de Unión , Modelos Moleculares , Estructura Molecular , Proteínas Serina-Treonina Quinasas/química , Relación Estructura-Actividad
9.
J Med Chem ; 49(16): 4971-80, 2006 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-16884309

RESUMEN

A potent SARS coronavirus (CoV) 3CL protease inhibitor (TG-0205221, Ki = 53 nM) has been developed. TG-0205221 showed remarkable activity against SARS CoV and human coronavirus (HCoV) 229E replications by reducing the viral titer by 4.7 log (at 5 microM) for SARS CoV and 5.2 log (at 1.25 microM) for HCoV 229E. The crystal structure of TG-0205221 (resolution = 1.93 A) has revealed a unique binding mode comprising a covalent bond, hydrogen bonds, and numerous hydrophobic interactions. Structural comparisons between TG-0205221 and a natural peptide substrate were also discussed. This information may be applied toward the design of other 3CL protease inhibitors.


Asunto(s)
Antivirales/síntesis química , Carbamatos/síntesis química , Cisteína Endopeptidasas/química , Dipéptidos/síntesis química , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Animales , Antivirales/química , Antivirales/farmacología , Carbamatos/química , Carbamatos/farmacología , Línea Celular , Chlorocebus aethiops , Coronavirus Humano 229E/efectos de los fármacos , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Dipéptidos/química , Dipéptidos/farmacología , Estabilidad de Medicamentos , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Modelos Moleculares , Estructura Molecular , Ratas , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
10.
J Phys Condens Matter ; 28(17): 175002, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27033403

RESUMEN

Atomic adsorption of Au and Pt on the rutile (1 1 0) surface was investigated by atomic-resolution aberration-corrected scanning transmission electron microscopy (STEM) measurements combined with density functional theory calculations. Au single atoms were deposited on the surface in a vacuum condition, and the observed results were compared with Pt single atoms on the same surface prepared by the same experimental manner. It was found that Au single atoms are stably adsorbed only at the bridging oxygen vacancy sites, which is quite different from Pt single atoms exhibiting the most frequently observed adsorption at the basal oxygen vacancy sites. Such a difference in oxygen-vacancy effect between Au and Pt can be explained by electronic structures of the surface vacancies as well as characters of outermost atomic orbitals of Au and Pt.

11.
Theranostics ; 6(1): 118-30, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26722378

RESUMEN

The overexpression of HER2/neu and EGFR receptors plays important roles in tumorigenesis and tumor progression. Targeting these two receptors simultaneously can have a more widespread application in early diagnosis of cancers. In this study, a new multifunctional nanoparticles (MnMEIO-CyTE777-(Bis)-mPEG NPs) comprising a manganese-doped iron oxide nanoparticle core (MnMEIO), a silane-amino functionalized poly(ethylene glycol) copolymer shell, a near infrared fluorescence dye (CyTE777), and a covalently conjugated anti-HER2/neu and anti-EGFR receptors bispecific antibody (Bis) were successfully developed. In vitro T2-weighted MR imaging studies in SKBR-3 and A431 tumor cells incubated with MnMEIO-CyTE777-(Bis)-mPEG NPs showed - 94.8 ± 3.8 and - 84.1 ± 2.8% negative contrast enhancement, respectively. Pharmacokinetics study showed that MnMEIO-CyTE777-(Bis)-mPEG NPs were eliminated from serum with the half-life of 21.3 mins. In vivo MR imaging showed that MnMEIO-CyTE777-(Bis)-mPEG NPs could specifically and effectively target to HER2/neu- and EGFR-expressing tumors in mice; the relative contrast enhancements were 11.8 (at 2 hrs post-injection) and 61.5 (at 24 hrs post-injection) fold higher in SKBR-3 tumors as compared to Colo-205 tumors. T2-weighted MR and optical imaging studies revealed that the new contrast agent (MnMEIO-CyTE777-(Bis)-mPEG NPs) could specifically and effectively target to HER2/neu- and/or EGFR-expressing tumors. Our results demonstrate that MnMEIO-CyTE777-(Bis)-mPEG NPs are able to recognize the tumors expressing both HER2/neu and/or EGFR, and may provide a novel molecular imaging tool for early diagnosis of cancers expressing HER2/neu and/or EGFR.


Asunto(s)
Anticuerpos Biespecíficos/farmacocinética , Receptores ErbB/análisis , Compuestos Férricos/farmacocinética , Imagen por Resonancia Magnética/métodos , Manganeso/farmacocinética , Neoplasias/diagnóstico , Receptor ErbB-2/análisis , Animales , Magnetismo , Ratones , Neoplasias/patología , Coloración y Etiquetado/métodos
12.
PLoS One ; 9(11): e111331, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25412347

RESUMEN

Enterovirus 71 (EV-A71) is a neurotropic virus that can cause severe complications involving the central nervous system. No effective antiviral therapeutics are available for treating EV-A71 infection and drug discovery efforts are rarely focused to target this disease. Thus, the main goal of this study was to discover existing drugs with novel indications that may effectively inhibit EV-A71 replication and the inflammatory cytokines elevation. In this study, we showed that LiCl, a GSK3ß inhibitor, effectively suppressed EV-A71 replication, apoptosis and inflammatory cytokines production (Interleukin 6, Interleukin-1ß) in infected cells. Furthermore, LiCl and an immunomodular agent were shown to strongly synergize with each other in suppressing EV-A71 replication. The results highlighted potential new treatment regimens in suppressing sequelae caused by EV-A71 replication.


Asunto(s)
Compuestos de Anilina/administración & dosificación , Antivirales/administración & dosificación , Enterovirus Humano A/efectos de los fármacos , Infecciones por Enterovirus/tratamiento farmacológico , Hidroxibutiratos/administración & dosificación , Cloruro de Litio/administración & dosificación , Compuestos de Anilina/farmacología , Animales , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Crotonatos , Citocinas/genética , Citocinas/metabolismo , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/inmunología , Humanos , Hidroxibutiratos/farmacología , Cloruro de Litio/farmacología , Nitrilos , Toluidinas , Células Vero , Replicación Viral/efectos de los fármacos
13.
Mol Oncol ; 6(3): 299-310, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22366308

RESUMEN

An acquired mutation (T790M) in the epidermal growth factor receptor (EGFR) accounts for half of all relapses in non-small cell lung cancer (NSCLC) patients who initially respond to EGFR kinase inhibitors. In this study, we demonstrated for the first time that EGFR-T790M interacts with the cytoskeletal components, myosin heavy chain 9 (MYH9) and ß-actin, in the nucleus of H1975 cells carrying the T790M-mutant EGFR. The interactions of EGFR with MYH9 and ß-actin were reduced in the presence of blebbistatin, a specific inhibitor for the MYH9-ß-actin interaction, suggesting that the EGFR interaction with MYH9 and ß-actin is affected by the integrity of the cytoskeleton. These physical interactions among MYH9, ß-actin, and EGFR were also impaired by CL-387,785, a kinase inhibitor for EGFR-T790M. Furthermore, CL-387,785 and blebbistatin interacted in a synergistic fashion to suppress cell proliferation and induce apoptosis in H1975 cells. The combination of CL-387,785 and blebbistatin enhanced the down-regulation of cyclooxygenase-2 (COX-2), a transcriptional target of nuclear EGFR. Overall, our findings demonstrate that disrupting EGFR interactions with the cytoskeletal components enhanced the anti-cancer effects of CL-387,785 against H1975 cells, suggesting a novel therapeutic approach for NSCLC cells that express the drug-resistant EGFR-T790M.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas Motoras Moleculares/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Actinas/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Citometría de Flujo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Inmunoprecipitación , Miosina Tipo II/antagonistas & inhibidores , Unión Proteica , Quinazolinas/farmacología , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Anticancer Res ; 32(1): 147-51, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22213300

RESUMEN

A high-throughput 32D(L858R/T790M) cell-based assay to identify inhibitors of the L858R/T790M mutant epidermal growth factor receptor (EGFR) pathway was established. After screening, ten hits from among 60,000 compounds in our in-house compound library were initially identified. In the secondary assays, one hit, 1-[2-(decyloxy)-2-oxoethyl]-3-methyl-2-[(4-methylphenoxy) methyl]-1H-benzimidazol-3-ium, was confirmed to directly inhibit the kinase activity of recombinant L858R/T790M EGFR and the phosphorylation of EGFR-L858R/T790M in gefitinib-resistant H1975 cells. Thus, this high-throughput assay system may be useful for identifying novel inhibitors which suppress mutant EGFR-T790M signalling and for overcoming T790M-mediated acquired resistance for future anticancer drug discovery.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Neoplasias Pulmonares/tratamiento farmacológico , Mutación/efectos de los fármacos , Quinazolinas/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Gefitinib , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/genética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos
15.
ChemMedChem ; 5(2): 255-67, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20039358

RESUMEN

Herein we reveal a simple method for the identification of novel Aurora kinase A inhibitors through substructure searching of an in-house compound library to select compounds for testing. A hydrazone fragment conferring Aurora kinase activity and heterocyclic rings most frequently reported in kinase inhibitors were used as substructure queries to filter the in-house compound library collection prior to testing. Five new series of Aurora kinase inhibitors were identified through this strategy, with IC(50) values ranging from approximately 300 nM to approximately 15 microM, by testing only 133 compounds from a database of approximately 125,000 compounds. Structure-activity relationship studies and X-ray co-crystallographic analysis of the most potent compound, a furanopyrimidine derivative with an IC(50) value of 309 nM toward Aurora kinase A, were carried out. The knowledge gained through these studies could help in the future design of potent Aurora kinase inhibitors.


Asunto(s)
Furanos/química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/química , Aurora Quinasas , Sitios de Unión , Cristalografía por Rayos X , Ensayos Analíticos de Alto Rendimiento , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/síntesis química , Pirimidinas/farmacología , Relación Estructura-Actividad
16.
J Med Chem ; 53(20): 7316-26, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20961149

RESUMEN

HTS hit 7 was modified through hybrid design strategy to introduce a chiral side chain followed by introduction of Michael acceptor group to obtain potent EGFR kinase inhibitors 11 and 19. Both 11 and 19 showed over 3 orders of magnitude enhanced HCC827 antiproliferative activity compared to HTS hit 7 and also inhibited gefitinib-resistant double mutant (DM, T790M/L858R) EGFR kinase at nanomolar concentration. Moreover, treatment with 19 shrinked tumor in nude mice xenograft model.


Asunto(s)
Antineoplásicos/síntesis química , Receptores ErbB/antagonistas & inhibidores , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Pirimidinas/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas , Línea Celular Tumoral , Diseño de Fármacos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/genética , Gefitinib , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Masculino , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Pirimidinas/química , Pirimidinas/farmacología , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo
17.
Langmuir ; 25(1): 179-84, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19063618

RESUMEN

3-Mercapto-1-propanesulfonic acid (MPS) and bis(3-sulfopropyl) disulfide (SPS) adsorbed on a Au(111) electrode were studied by using in situ scanning tunneling microscopy (STM). Although the adsorptions of MPS and SPS are known to be oxidative and reductive, respectively, on an Au(111) electrode, these two admolecules behave similarly in terms of phase evolution, surface coverage, potential for stripping, and characteristics of cyclic voltammetry. However, different adsorption mechanisms of these molecules result in different structures. Raising electrode potential causes more MPS and SPS molecules to adsorb, yielding ordered adlattices between 0.67 and 0.8 V (vs reversible hydrogen electrode). The ordered adlattices of MPS and SPS appear as striped and netlike structures with molecules adsorbed parallel to the Au(111) surface. Switching potential to 0.9 V or more positive still does not result in upright molecular orientation, possibly inhibited by electrostatic interaction between the end group of -SO(3)(-) and the Au(111) electrode. Lowering the potential to 0.4 V disrupted the ordered adlayer. Stripping voltammetric experiments show that MPS and SPS admolecules are desorbed from Au(111) at the same potential, suggesting that these molecules are both adsorbed via their sulfur headgroups. The S-S bond in SPS is likely broken upon its adsorption on Au(111).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA