Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 390: 21-29, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28043914

RESUMEN

The initiation of mRNA translation has received increasing attention as an attractive target for cancer treatment in the recent years. The oncogenic eukaryotic translation initiation factor 4E (eIF4E) is the major substrate of MAP kinase-interacting kinase 1 (MNK1), and it is located at the junction of the cancer-associated PI3K and MAPK pathways. The fact that MNK1 is linked to cell transformation and tumorigenesis renders the kinase a promising target for cancer therapy. We identified a novel small molecule MNK1 inhibitor, BAY 1143269, by high-throughput screening and lead optimization. In kinase assays, BAY 1143269 showed potent and selective inhibition of MNK1. By targeting MNK1 activity, BAY 1143269 strongly regulated downstream factors involved in cell cycle regulation, apoptosis, immune response and epithelial-mesenchymal transition in vitro or in vivo. In addition, BAY 1143269 demonstrated strong efficacy in monotherapy in cell line and patient-derived non-small cell lung cancer xenograft models as well as delayed tumor regrowth in combination treatment with standard of care chemotherapeutics. In summary, the inhibition of MNK1 activity with a highly potent and selective inhibitor BAY 1143269 may provide an innovative approach for anti-cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Oncogenes/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridazinas/farmacología , Animales , Antineoplásicos/química , Western Blotting , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Imidazoles/química , Concentración 50 Inhibidora , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Piridazinas/química
2.
Int J Cancer ; 140(2): 449-459, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27699769

RESUMEN

The PI3K-AKT-mTOR signaling cascade is activated in the majority of human cancers, and its activation also plays a key role in resistance to chemo and targeted therapeutics. In particular, in both breast and prostate cancer, increased AKT pathway activity is associated with cancer progression, treatment resistance and poor disease outcome. Here, we evaluated the activity of a novel allosteric AKT1/2 inhibitor, BAY 1125976, in biochemical, cellular mechanistic, functional and in vivo efficacy studies in a variety of tumor models. In in vitro kinase activity assays, BAY 1125976 potently and selectively inhibited the activity of full-length AKT1 and AKT2 by binding into an allosteric binding pocket formed by kinase and PH domain. In accordance with this proposed allosteric binding mode, BAY 1125976 bound to inactive AKT1 and inhibited T308 phosphorylation by PDK1, while the activity of truncated AKT proteins lacking the pleckstrin homology domain was not inhibited. In vitro, BAY 1125976 inhibited cell proliferation in a broad panel of human cancer cell lines. Particularly high activity was observed in breast and prostate cancer cell lines expressing estrogen or androgen receptors. Furthermore, BAY 1125976 exhibited strong in vivo efficacy in both cell line and patient-derived xenograft models such as the KPL4 breast cancer model (PIK3CAH1074R mutant), the MCF7 and HBCx-2 breast cancer models and the AKTE17K mutant driven prostate cancer (LAPC-4) and anal cancer (AXF 984) models. These findings indicate that BAY 1125976 is a potent and highly selective allosteric AKT1/2 inhibitor that targets tumors displaying PI3K/AKT/mTOR pathway activation, providing opportunities for the clinical development of new, effective treatments.


Asunto(s)
Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sulfonas/farmacología , Animales , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Células HeLa , Humanos , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
3.
ChemMedChem ; 11(14): 1517-30, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27310202

RESUMEN

The phosphoinositide 3-kinase (PI3K) pathway is aberrantly activated in many disease states, including tumor cells, either by growth factor receptor tyrosine kinases or by the genetic mutation and amplification of key pathway components. A variety of PI3K isoforms play differential roles in cancers. As such, the development of PI3K inhibitors from novel compound classes should lead to differential pharmacological and pharmacokinetic profiles and allow exploration in various indications, combinations, and dosing regimens. A screening effort aimed at the identification of PI3Kγ inhibitors for the treatment of inflammatory diseases led to the discovery of the novel 2,3-dihydroimidazo[1,2-c]quinazoline class of PI3K inhibitors. A subsequent lead optimization program targeting cancer therapy focused on inhibition of PI3Kα and PI3Kß. Herein, initial structure-activity relationship findings for this class and the optimization that led to the identification of copanlisib (BAY 80-6946) as a clinical candidate for the treatment of solid and hematological tumors are described.


Asunto(s)
Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Quinazolinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase Ib/química , Descubrimiento de Drogas , Humanos , Enlace de Hidrógeno , Imidazoles/síntesis química , Imidazoles/química , Simulación del Acoplamiento Molecular , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/síntesis química , Pirimidinas/química , Quinazolinas/síntesis química , Quinazolinas/química , Relación Estructura-Actividad
4.
Mol Cancer Ther ; 15(4): 583-92, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26832791

RESUMEN

Monopolar spindle 1 (Mps1) has been shown to function as the key kinase that activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we report the structure and functional characterization of two novel selective Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nmol/L while showing an excellent selectivity profile. In cellular mechanistic assays, both Mps1 inhibitors abrogated nocodazole-induced SAC activity and induced premature exit from mitosis ("mitotic breakthrough"), resulting in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 nmol/L range). In vivo, BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies. However, in line with its unique mode of action, when combined with paclitaxel, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest by the weakening of SAC activity. As a result, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor monotreatment at the respective MTDs in a broad range of xenograft models, including those showing acquired or intrinsic paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. These preclinical findings validate the innovative concept of SAC abrogation for cancer therapy and justify clinical proof-of-concept studies evaluating the Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs to enhance their efficacy and potentially overcome resistance. Mol Cancer Ther; 15(4); 583-92. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Antineoplásicos/química , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Mitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Ratas , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Med ; 2(5): 611-24, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24403227

RESUMEN

The activation of the transcription factor hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor development, tumor progression, and resistance to chemo- and radiotherapy. In order to identify compounds targeting the HIF pathway, a small molecule library was screened using a luciferase-driven HIF-1 reporter cell line under hypoxia. The high-throughput screening led to the identification of a class of aminoalkyl-substituted compounds that inhibited hypoxia-induced HIF-1 target gene expression in human lung cancer cell lines at low nanomolar concentrations. Lead structure BAY 87-2243 was found to inhibit HIF-1α and HIF-2α protein accumulation under hypoxic conditions in non-small cell lung cancer (NSCLC) cell line H460 but had no effect on HIF-1α protein levels induced by the hypoxia mimetics desferrioxamine or cobalt chloride. BAY 87-2243 had no effect on HIF target gene expression levels in RCC4 cells lacking Von Hippel-Lindau (VHL) activity nor did the compound affect the activity of HIF prolyl hydroxylase-2. Antitumor activity of BAY 87-2243, suppression of HIF-1α protein levels, and reduction of HIF-1 target gene expression in vivo were demonstrated in a H460 xenograft model. BAY 87-2243 did not inhibit cell proliferation under standard conditions. However under glucose depletion, a condition favoring mitochondrial ATP generation as energy source, BAY 87-2243 inhibited cell proliferation in the nanomolar range. Further experiments revealed that BAY 87-2243 inhibits mitochondrial complex I activity but has no effect on complex III activity. Interference with mitochondrial function to reduce hypoxia-induced HIF-1 activity in tumors might be an interesting therapeutic approach to overcome chemo- and radiotherapy-resistance of hypoxic tumors.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Neoplasias Pulmonares/metabolismo , Oxadiazoles/farmacología , Pirazoles/farmacología , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/biosíntesis , Anhidrasas Carbónicas/genética , Hipoxia de la Célula/genética , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Complejo I de Transporte de Electrón/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Relacionados con las Neoplasias , Genes Reporteros , Humanos , Factor 1 Inducible por Hipoxia/biosíntesis , Factor 1 Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Terapia Molecular Dirigida/métodos , Oxadiazoles/administración & dosificación , Oxadiazoles/sangre , Oxadiazoles/uso terapéutico , Pirazoles/administración & dosificación , Pirazoles/sangre , Pirazoles/uso terapéutico , ARN Interferente Pequeño/genética , Bibliotecas de Moléculas Pequeñas , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
J Med Chem ; 50(21): 5202-16, 2007 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17887659

RESUMEN

The peptide hormone ghrelin is the endogenous ligand for the type 1a growth hormone secretagogue receptor (GHS-R1a) and the only currently known circulating appetite stimulant. GHS-R1a antagonism has therefore been proposed as a potential approach for obesity treatment. More recently, ghrelin has been recognized to also play a role in controlling glucose-induced insulin secretion, which suggests another possible benefit for a GHS-R1a antagonist, namely, the role as an insulin secretagogue with potential value for diabetes treatment. In our laboratories, piperidine-substituted quinazolinone derivatives were identified as a new class of small-molecule GHS-R1a antagonists. Starting from an agonist with poor oral bioavailability, optimization led to potent, selective, and orally bioavailable antagonists. In vivo efficacy evaluation of selected compounds revealed suppression of food intake and body weight reduction as well as glucose-lowering effects mediated by glucose-dependent insulin secretion.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Quinazolinonas/síntesis química , Receptores de Ghrelina/antagonistas & inhibidores , Administración Oral , Animales , Unión Competitiva , Glucemia/análisis , Línea Celular , Ingestión de Alimentos/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Quinazolinonas/química , Quinazolinonas/farmacología , Ensayo de Unión Radioligante , Ratas , Ratas Wistar , Estereoisomerismo , Relación Estructura-Actividad , Pérdida de Peso/efectos de los fármacos
7.
J Org Chem ; 70(23): 9463-9, 2005 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-16268621

RESUMEN

[Reaction: see text]. A regioselective and efficient approach toward 6-amino-5-benzoyl-1-substituted 2(1H)-pyridinones by reaction of acyclic ketene aminals with propiolic acid ester was developed. The effect of the solvent and temperature on the regioselectivity of the reaction and the compatibility of the target compounds to functional group manipulations was examined. Substrates with an ortho substituent build atropisomers due to the restricted rotation around the C-N bond. The enantiomers were separated, and the barrier of rotation was determined experimentally. Quantum chemical calculations allowed a ranking of the barrier heights, and a new mechanism of rotation by deformation of the central pyridinone moiety is proposed.


Asunto(s)
Piridonas/síntesis química , Modelos Químicos , Conformación Molecular , Estructura Molecular , Estereoisomerismo
8.
J Dtsch Dermatol Ges ; 3(6): 421-30, 2005 Jun.
Artículo en Alemán | MEDLINE | ID: mdl-15892844

RESUMEN

Malignant melanoma is a primarily cutaneous melanocytic tumour with increasing incidence responsible for 90 % of skin cancer mortality. Genetic predisposition has been identified as the most important risk factor, while UV is second in importance and can be avoided. New diagnostic methods include sentinel lymph node biopsy and the detection of tumour markers in blood. Furthermore, malignant melanoma shows an extraordinary resistance to therapy; at present the only cure lies in early excision of the primary tumour. Thus early recognition is of utmost importance. Experimental approaches, such as dendritic cell vaccination, have shown some effectiveness which must be confirmed in multicenter, randomised trials.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Adulto , Biomarcadores de Tumor , Vacunas contra el Cáncer/uso terapéutico , Niño , Células Dendríticas , Predisposición Genética a la Enfermedad , Humanos , Melanoma/diagnóstico , Melanoma/genética , Melanoma/mortalidad , Melanoma/patología , Melanoma/prevención & control , Melanoma/cirugía , Melanoma/terapia , Pronóstico , Riesgo , Factores de Riesgo , Biopsia del Ganglio Linfático Centinela , Piel/patología , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/cirugía , Neoplasias Cutáneas/terapia , Factores de Tiempo , Rayos Ultravioleta/efectos adversos
9.
Chemistry ; 9(22): 5636-42, 2003 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-14639647

RESUMEN

In the present work the effect of spiroconjugation on the electronic spectrum of the recently synthesized metal complex hexadecaethylspirodicorrolato-dinickel(II) (5) (the corrole units in 5 are isoforms) is investigated. To have a suitable reference compound at our disposal, tetraethylhexamethylisocorrolato nickel(II) (7) has been prepared. On comparing the electronic spectra of this reference compound and the spiro-complex, bathochromic shifts of all absorption bands in the NIR/Vis-region are observed for the latter as well as marked changes in the spectral intensities. A detailed analysis of the spectra supported by semiempirical calculations reveals that at least part of the observed changes can be unambiguously attributed to the spiro effect. This effect is further affirmed by electrochemically measured redox potentials.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA