Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 618(7963): 159-168, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225977

RESUMEN

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Asunto(s)
Regeneración Nerviosa , Humanos , Neoplasias/tratamiento farmacológico , Regeneración Nerviosa/efectos de los fármacos , Isoformas de Proteínas/agonistas , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/química , Fosfatidilinositol 3-Quinasa Clase I/efectos de los fármacos , Cardiotónicos/farmacología , Animales , Biocatálisis/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Neuritas/efectos de los fármacos , Daño por Reperfusión/prevención & control , Compresión Nerviosa , Proliferación Celular/efectos de los fármacos
2.
Antimicrob Agents Chemother ; 68(3): e0106423, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38349161

RESUMEN

Screening a library of 1,200 preselected kinase inhibitors for anti-human rhinovirus 2 (HRV-2) activity in HeLa cells identified a class of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) as effective virus blockers. These were based on the 4-anilinoquinazoline-7-oxypiperidine scaffold, with the most potent representative AZ5385 inhibiting the virus with EC50 of 0.35 µM. Several structurally related analogs confirmed activity in the low µM range, while interestingly, other TKIs targeting EGFR lacked anti-HRV-2 activity. To further probe this lack of association between antiviral activity and EGFR inhibition, we stained infected cells with antibodies specific for activated EGFR (Y1068) and did not observe a dependency on EGFR-TK activity. Instead, consecutive passages of HRV-2 in HeLa cells in the presence of a compound and subsequent nucleotide sequence analysis of resistant viral variants identified the S181T and T210A alterations in the major capsid VP1 protein, with both residues located in the vicinity of a known hydrophobic pocket on the viral capsid. Further characterization of the antiviral effects of AZ5385 showed a modest virus-inactivating (virucidal) activity, while anti-HRV-2 activity was still evident when the inhibitor was added as late as 10 h post infection. The RNA copy/infectivity ratio of HRV-2 propagated in AZ5385 presence was substantially higher than that of control HRV indicating that the compound preferentially targeted HRV progeny virions during their maturation in infected cells. Besides HRV, the compound showed anti-respiratory syncytial virus activity, which warrants its further studies as a candidate compound against viral respiratory infections.


Asunto(s)
Rhinovirus , Humanos , Rhinovirus/química , Rhinovirus/genética , Células HeLa , Proteínas de la Cápside , Antivirales/química , Receptores ErbB
3.
Biochem J ; 477(17): 3329-3347, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32815546

RESUMEN

Despite being catalytically defective, pseudokinases are typically essential players of cellular signalling, acting as allosteric regulators of their active counterparts. Deregulation of a growing number of pseudokinases has been linked to human diseases, making pseudokinases therapeutic targets of interest. Pseudokinases can be dynamic, adopting specific conformations critical for their allosteric function. Interfering with their allosteric role, with small molecules that would lock pseudokinases in a conformation preventing their productive partner interactions, is an attractive therapeutic strategy to explore. As a well-known allosteric activator of epidermal growth factor receptor family members, and playing a major part in cancer progression, the pseudokinase HER3 is a relevant context in which to address the potential of pseudokinases as drug targets for the development of allosteric inhibitors. In this proof-of-concept study, we developed a multiplex, medium-throughput thermal shift assay screening strategy to assess over 100 000 compounds and identify selective small molecule inhibitors that would trap HER3 in a conformation which is unfavourable for the formation of an active HER2-HER3 heterodimer. As a proof-of-concept compound, AC3573 bound with some specificity to HER3 and abrogated HER2-HER3 complex formation and downstream signalling in cells. Our study highlights the opportunity to identify new molecular mechanisms of action interfering with the biological function of pseudokinases.


Asunto(s)
Inhibidores de Proteínas Quinasas , Receptor ErbB-3 , Regulación Alostérica , Animales , Células CHO , Cricetulus , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Prueba de Estudio Conceptual , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Receptor ErbB-3/química , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo
4.
Int J Mol Sci ; 21(24)2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33334026

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) has a large impact on global health. At the onset of disease, NAFLD is characterized by hepatic steatosis defined by the accumulation of triglycerides stored as lipid droplets. Developing therapeutics against NAFLD and progression to non-alcoholic steatohepatitis (NASH) remains a high priority in the medical and scientific community. Drug discovery programs to identify potential therapeutic compounds have supported high throughput/high-content screening of in vitro human-relevant models of NAFLD to accelerate development of efficacious anti-steatotic medicines. Human induced pluripotent stem cell (hiPSC) technology is a powerful platform for disease modeling and therapeutic assessment for cell-based therapy and personalized medicine. In this study, we applied AstraZeneca's chemogenomic library, hiPSC technology and multiplexed high content screening to identify compounds that significantly reduced intracellular neutral lipid content. Among 13,000 compounds screened, we identified hits that protect against hiPSC-derived hepatic endoplasmic reticulum stress-induced steatosis by a mechanism of action including inhibition of the cyclin D3-cyclin-dependent kinase 2-4 (CDK2-4)/CCAAT-enhancer-binding proteins (C/EBPα)/diacylglycerol acyltransferase 2 (DGAT2) pathway, followed by alteration of the expression of downstream genes related to NAFLD. These findings demonstrate that our phenotypic platform provides a reliable approach in drug discovery, to identify novel drugs for treatment of fatty liver disease as well as to elucidate their underlying mechanisms.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Metabolismo de los Lípidos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Biología Computacional/métodos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Diacilglicerol O-Acetiltransferasa/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Gotas Lipídicas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Inhibidores de Proteínas Quinasas/farmacología
5.
Bioorg Med Chem Lett ; 27(13): 3030-3035, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526367

RESUMEN

We report the discovery of a novel aminopyrazine series of PI3Kα inhibitors, designed by hybridizing two known scaffolds of PI3K inhibitors. We describe the progress achieved from the first compounds plagued with poor general kinase selectivity to compounds showing high selectivity for PI3Kα over PI3Kß and excellent general kinase selectivity. This effort culminated with the identification of compound 5 displaying high potency and selectivity, and suitable physiochemical and pharmacokinetic properties for oral administration. In vivo, compound 5 showed good inhibition of tumour growth (86% tumour growth inhibition at 50mg/kg twice daily orally) in the MCF7 xenograft model in mice.


Asunto(s)
Descubrimiento de Drogas , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Fosfatidilinositol 3-Quinasa Clase I , Relación Dosis-Respuesta a Droga , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirazinas/síntesis química , Pirazinas/química , Relación Estructura-Actividad
6.
Bioorg Med Chem Lett ; 27(9): 1949-1954, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28347666

RESUMEN

Attempts to lock the active conformation of compound 4, a PI3Kß/δ inhibitor (PI3Kß cell IC50 0.015µM), led to the discovery of a series of 8-(1-phenylpyrrolidin-2-yl)-6-carboxamide-2-morpholino-4H-chromen-4-ones, which showed high levels of potency and selectivity as PI3Kß/δ inhibitors. Compound 10 proved exquisitely potent and selective: PI3Kß cell IC50 0.0011µM in PTEN null MDA-MB-468 cell and PI3Kδ cell IC50 0.014µM in Jeko-1 B-cell, and exhibited suitable physical properties for oral administration. In vivo, compound 10 showed profound pharmacodynamic modulation of AKT phosphorylation in a mouse PTEN-null PC3 prostate tumour xenograft after a single oral dose and gave excellent tumour growth inhibition in the same model after chronic oral dosing. Based on these results, compound 10 was selected as one of our PI3Kß/δ preclinical candidates.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Benzopiranos/química , Benzopiranos/uso terapéutico , Fosfohidrolasa PTEN/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Benzopiranos/farmacocinética , Benzopiranos/farmacología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Perros , Eliminación de Gen , Humanos , Masculino , Ratones Desnudos , Simulación del Acoplamiento Molecular , Morfolinos/química , Morfolinos/farmacocinética , Morfolinos/farmacología , Morfolinos/uso terapéutico , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
7.
Bioorg Med Chem Lett ; 26(9): 2318-23, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26996374

RESUMEN

We report the discovery and optimisation of a series of 8-(2,3-dihydro-1,4-benzoxazin-4-ylmethyl)-2-morpholino-4-oxo-chromene-6-carboxamides, leading to compound 16 as a potent and selective PI3Kß/δ inhibitor: PI3Kß cell IC50 0.012 µM (in PTEN null MDA-MB-468 cell) and PI3Kδ cell IC50 0.047 µM (in Jeko-1 B-cell), with good pharmacokinetics and physical properties. In vivo, 16 showed profound pharmacodynamic modulation of AKT phosphorylation in a mouse PTEN-deficient PC3 prostate tumour xenograft after a single oral dose and gave excellent tumour growth inhibition in the same model after chronic oral dosing. Compound 16 was selected as a preclinical candidate for the treatment of PTEN-deficient tumours.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Isoenzimas/antagonistas & inhibidores , Morfolinos/química , Morfolinos/farmacología , Fosfohidrolasa PTEN/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Línea Celular Tumoral , Humanos , Ratones , Fosforilación
8.
Bioorg Med Chem Lett ; 25(13): 2679-85, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25980912

RESUMEN

Starting from compound 1, a potent PI3Kα inhibitor having poor general kinase selectivity, we used structural data and modelling to identify key exploitable differences between PI3Kα and the other kinases. This approach led us to design chemical modifications of the central pyrazole, which solved the poor kinase selectivity seen as a strong liability for the initial compound 1. Amongst the modifications explored, a 1,3,4-triazole ring (as in compound 4) as a replacement of the initial pyrazole provided good potency against PI3Kα, with excellent kinase selectivity.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Secuencia de Aminoácidos , Sitios de Unión , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/química , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Humanos , Modelos Moleculares , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/genética , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/genética , Pirazoles/síntesis química , Pirazoles/química , Pirazoles/farmacología , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/química , Triazoles/farmacología
9.
Bioorg Med Chem Lett ; 25(22): 5155-62, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26475521

RESUMEN

Starting from potent inhibitors of PI3Kα having poor general kinase selectivity (e.g., 1 and 2), optimisation of this series led to the identification of 25, a potent inhibitor of PI3Kα (wild type, E545K and H1047R mutations) and PI3Kδ, selective versus PI3Kß and PI3Kγ, with excellent general kinase selectivity. Compound 25 displayed low metabolic turnover and suitable physical properties for oral administration. In vivo, compound 25 showed pharmacodynamic modulation of AKT phosphorylation and near complete inhibition of tumour growth (93% tumour growth inhibition) in a murine H1047R PI3Kα mutated SKOV-3 xenograft tumour model after chronic oral administration at 25mg/kg b.i.d. Compound 25, also known as AZD8835, is currently in phase I clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Oxadiazoles/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Perros , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Simulación del Acoplamiento Molecular , Oxadiazoles/síntesis química , Piperidinas/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Bioorg Med Chem Lett ; 24(16): 3928-35, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24992874

RESUMEN

Starting from TGX-221, we designed a series of 9-(1-anilinoethyl)-2-morpholino-4-oxo-pyrido[1,2-a]pyrimidine-7-carboxamides as potent and selective PI3Kß/δ inhibitors. Structure-activity relationships and structure-property relationships around the aniline and the amide substituents are discussed. We identified compounds 17 and 18, which showed profound pharmacodynamic modulation of phosphorylated Akt in the PC3 prostate tumour xenograft, after a single oral dose. Compound 17 also gave significant inhibition of tumour growth in the PC3 prostate tumour xenograft model after chronic oral dosing.


Asunto(s)
Amidas/farmacología , Antineoplásicos/farmacología , Descubrimiento de Drogas , Neoplasias Experimentales/tratamiento farmacológico , Fosfohidrolasa PTEN/deficiencia , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Administración Oral , Amidas/administración & dosificación , Amidas/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Estructura Molecular , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
11.
Stem Cell Reports ; 18(2): 555-569, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36669494

RESUMEN

Marfan syndrome (MFS) is a rare connective tissue disorder caused by mutations in FBN1. Patients with MFS notably suffer from aortic aneurysm and dissection. Despite considerable effort, animal models have proven to be poorly predictive for therapeutic intervention in human aortic disease. Patient-derived induced pluripotent stem cells can be differentiated into vascular smooth muscle cells (VSMCs) and recapitulate major features of MFS. We have screened 1,022 small molecules in our in vitro model, exploiting the highly proteolytic nature of MFS VSMCs, and identified 36 effective compounds. Further analysis identified GSK3ß as a recurring target in the compound screen. GSK3ß inhibition/knockdown did not ameliorate the proliferation defect in MFS-VSMCs but improved MFS-VSMC proteolysis and apoptosis and partially rescued fibrillin-1 deposition. To conclude, we have identified GSK3ß as a novel target for MFS, forming the foundation for future work in MFS and other aortic diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome de Marfan , Animales , Humanos , Síndrome de Marfan/genética , Músculo Liso Vascular , Aorta , Glucógeno Sintasa Quinasa 3 beta
12.
ACS Med Chem Lett ; 14(12): 1760-1766, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38116421

RESUMEN

A high-throughput fragment-based screen has been employed to discover a series of quinazolinone inositol hexakisphosphate kinase (IP6K) inhibitors. IP6Ks have been studied for their role in glucose homeostasis, metabolic disease, fatty liver disease, chronic kidney disease, blood coagulation, neurological development, and psychiatric disease. IP6Ks phosphorylate inositol hexakisphosphate (IP6) to form pyrophosphate 5-diphospho-1,2,3,4,6-pentakisphosphate (IP7). Molecular docking studies and investigation of structure-activity relationships around the quinazolinone core resulted in compounds with submicromolar potency and interesting selectivity for IP6K1 versus the closely related IP6K2 and IP6K3 isoforms.

13.
Bioorg Med Chem Lett ; 21(16): 4698-701, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21775140

RESUMEN

We disclose a novel series of insulin-like growth factor-1 receptor kinase inhibitors based on the 3-(pyrimidin-4-yl)-imidazo[1,2-a]pyridine scaffold. The influence on the inhibitory activity of substitution on the imidazopyridine and at the C5 position of the pyrimidine is discussed. In the course of this optimization, we discovered a potent and selective inhibitor with suitable pharmacokinetics for oral administration.


Asunto(s)
Descubrimiento de Drogas , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Animales , Perros , Humanos , Ratones , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Piridinas/síntesis química , Piridinas/química , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Distribución Tisular
14.
Cell Death Discov ; 6: 6, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32123582

RESUMEN

Regulated necrosis or necroptosis, mediated by receptor-interacting kinase 1 (RIPK1), RIPK3 and pseudokinase mixed lineage kinase domain-like protein (MLKL), contributes to the pathogenesis of inflammatory, infectious and degenerative diseases. Recently identified necroptosis inhibitors display moderate specificity, suboptimal pharmacokinetics, off-target effects and toxicity, preventing these molecules from reaching the clinic. Here, we developed a cell-based high-throughput screening (HTS) cascade for the identification of small-molecule inhibitors of necroptosis. From the initial library of over 250,000 compounds, the primary screening phase identified 356 compounds that strongly inhibited TNF-α-induced necroptosis, but not apoptosis, in human and murine cell systems, with EC50 < 6.7 µM. From these, 251 compounds were tested for RIPK1 and/or RIPK3 kinase inhibitory activity; some were active and several have novel mechanisms of action. Based on specific chemical descriptors, 110 compounds proceeded into the secondary screening cascade, which then identified seven compounds with maximum ability to reduce MLKL activation, IC50 >100 µM, EC50 2.5-11.5 µM under long-term necroptosis execution in murine fibroblast L929 cells, and full protection from ATP depletion and membrane leakage in human and murine cells. As a proof of concept, compound SN-6109, with binding mode to RIPK1 similar to that of necrostatin-1, confirmed RIPK1 inhibitory activity and appropriate pharmacokinetic properties. SN-6109 was further tested in mice, showing efficacy against TNF-α-induced systemic inflammatory response syndrome. In conclusion, a phenotypic-driven HTS cascade promptly identified robust necroptosis inhibitors with in vivo activity, currently undergoing further medicinal chemistry optimization. Notably, the novel hits highlight the opportunity to identify new molecular mechanisms of action in necroptosis.

15.
Bioorg Med Chem Lett ; 18(16): 4723-6, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18676144

RESUMEN

Tie-2 is a receptor tyrosine kinase which is involved in angiogenesis and thereby growth of human tumours. The discovery and SAR of a novel class of imidazole-vinyl-pyrimidine kinase inhibitors, which inhibit Tie-2 in vitro is reported. Their synthesis was carried out by condensation of imidazole aldehydes with methyl pyrimidines. These compounds are lead-like, with low molecular weight, good physical properties and oral bioavailability.


Asunto(s)
Imidazoles/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/síntesis química , Pirimidinas/farmacología , Receptor TIE-2/antagonistas & inhibidores , Administración Oral , Disponibilidad Biológica , Química Farmacéutica/métodos , Diseño de Fármacos , Humanos , Imidazoles/administración & dosificación , Concentración 50 Inhibidora , Modelos Químicos , Conformación Molecular , Neovascularización Patológica , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/administración & dosificación , Receptor TIE-2/química , Relación Estructura-Actividad
16.
Assay Drug Dev Technol ; 13(2): 88-93, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25710279

RESUMEN

Factorial experimental design (FED) is a powerful approach for efficient optimization of robust in vitro assays-it enables cost and time savings while also improving the quality of assays. Although it is a well-known technique, there can be considerable barriers to overcome to fully exploit it within an industrial or academic organization. The article describes a tactical roll out of FED to a scientist group through: training which demystifies the technical components and concentrates on principles and examples; a user-friendly Excel-based tool for deconvoluting plate data; output which focuses on graphical display of data over complex statistics. The use of FED historically has generally been in conjunction with automated technology; however we have demonstrated a much broader impact of FED on the assay development process. The standardized approaches we have rolled out have helped to integrate FED as a fundamental part of assay development best practice because it can be used independently of the automation and vendor-supplied software. The techniques are applicable to different types of assay, both enzyme and cell, and can be used flexibly in manual and automated processes. This article describes the application of FED for a cellular assay. The challenges of selling FED concepts and rolling out to a wide bioscience community together with recommendations for good working practices and effective implementation are discussed. The accessible nature of these approaches means FED can be used by industrial as well as academic users.


Asunto(s)
Bioensayo/métodos , Interpretación Estadística de Datos , Descubrimiento de Drogas , Análisis Factorial , Modelos Estadísticos , Programas Informáticos , Simulación por Computador , Lenguajes de Programación
17.
J Med Chem ; 58(2): 943-62, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25514658

RESUMEN

Several studies have highlighted the dependency of PTEN deficient tumors to PI3Kß activity and specific inhibition of PI3Kδ has been shown activity against human B-cell cancers. We describe the discovery and optimization of a series of 8-(1-anilino)ethyl)-2-morpholino-4-oxo-4H-chromene-6-carboxamides as PI3Kß/δ inhibitors, which led to the discovery of the clinical candidate 13, also known as AZD8186. On the basis of the lower lipophilicity of the chromen-4-one core compared to the previously utilized pyrido[1,2-a]pyrimid-4-one core, this series of compounds displayed high metabolic stability and suitable physical properties for oral administration. Compound 13 showed profound pharmacodynamic modulation of p-Akt in PTEN-deficient PC3 prostate tumor bearing mice after oral administration and showed complete inhibition of tumor growth in the mouse PTEN-deficient PC3 prostate tumor xenograft model. 13 was selected as a clinical candidate for treatment of PTEN-deficient cancers and has recently entered phase I clinical trials.


Asunto(s)
Compuestos de Anilina/síntesis química , Cromonas/síntesis química , Neoplasias Experimentales/tratamiento farmacológico , Fosfohidrolasa PTEN/deficiencia , Inhibidores de las Quinasa Fosfoinosítidos-3 , Compuestos de Anilina/farmacología , Animales , Cromonas/farmacología , Perros , Descubrimiento de Drogas , Humanos , Masculino , Ratones , Neoplasias Experimentales/química , Relación Estructura-Actividad
18.
Mol Cancer Ther ; 14(1): 48-58, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25398829

RESUMEN

Loss of PTEN protein results in upregulation of the PI3K/AKT pathway, which appears dependent on the PI3Kß isoform. Inhibitors of PI3Kß have potential to reduce growth of tumors in which loss of PTEN drives tumor progression. We have developed a small-molecule inhibitor of PI3Kß and PI3Kδ (AZD8186) and assessed its antitumor activity across a panel of cell lines. We have then explored the antitumor effects as single agent and in combination with docetaxel in triple-negative breast (TNBC) and prostate cancer models. In vitro, AZD8186 inhibited growth of a range of cell lines. Sensitivity was associated with inhibition of the AKT pathway. Cells sensitive to AZD8186 (GI50 < 1 µmol/L) are enriched for, but not exclusively associated with, PTEN deficiency. In vivo, AZD8186 inhibits PI3K pathway biomarkers in prostate and TNBC tumors. Scheduling treatment with AZD8186 shows antitumor activity required only intermittent exposure, and that increased tumor control is achieved when AZD8186 is used in combination with docetaxel. AZD8186 is a potent inhibitor of PI3Kß with activity against PI3Kδ signaling, and has potential to reduce growth of tumors dependent on dysregulated PTEN for growth. Moreover, AZD8186 can be combined with docetaxel, a chemotherapy commonly used to treat advanced TBNC and prostate tumors. The ability to schedule AZD8186 and maintain efficacy offers opportunity to combine AZD8186 more effectively with other drugs.


Asunto(s)
Compuestos de Anilina/administración & dosificación , Antineoplásicos/administración & dosificación , Cromonas/administración & dosificación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias de la Próstata/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Taxoides/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Compuestos de Anilina/farmacología , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromonas/farmacología , Docetaxel , Sinergismo Farmacológico , Femenino , Humanos , Masculino , Ratones , Fosfohidrolasa PTEN/deficiencia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA