Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Mol Pharmacol ; 88(6): 1011-23, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26438213

RESUMEN

Activation of the inositol-requiring enzyme-1 alpha (IRE1α) protein caused by endoplasmic reticulum stress results in the homodimerization of the N-terminal endoplasmic reticulum luminal domains, autophosphorylation of the cytoplasmic kinase domains, and conformational changes to the cytoplasmic endoribonuclease (RNase) domains, which render them functional and can lead to the splicing of X-box binding protein 1 (XBP 1) mRNA. Herein, we report the first crystal structures of the cytoplasmic portion of a human phosphorylated IRE1α dimer in complex with (R)-2-(3,4-dichlorobenzyl)-N-(4-methylbenzyl)-2,7-diazaspiro(4.5)decane-7-carboxamide, a novel, IRE1α-selective kinase inhibitor, and staurosporine, a broad spectrum kinase inhibitor. (R)-2-(3,4-dichlorobenzyl)-N-(4-methylbenzyl)-2,7-diazaspiro(4.5)decane-7-carboxamide inhibits both the kinase and RNase activities of IRE1α. The inhibitor interacts with the catalytic residues Lys599 and Glu612 and displaces the kinase activation loop to the DFG-out conformation. Inactivation of IRE1α RNase activity appears to be caused by a conformational change, whereby the αC helix is displaced, resulting in the rearrangement of the kinase domain-dimer interface and a rotation of the RNase domains away from each other. In contrast, staurosporine binds at the ATP-binding site of IRE1α, resulting in a dimer consistent with RNase active yeast Ire1 dimers. Activation of IRE1α RNase activity appears to be promoted by a network of hydrogen bond interactions between highly conserved residues across the RNase dimer interface that place key catalytic residues poised for reaction. These data implicate that the intermolecular interactions between conserved residues in the RNase domain are required for activity, and that the disruption of these interactions can be achieved pharmacologically by small molecule kinase domain inhibitors.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Línea Celular Tumoral , Cristalización , Endorribonucleasas/química , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Conformación Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
2.
J Biol Chem ; 286(35): 30706-30713, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21737452

RESUMEN

Hepatocellular carcinoma (HCC) is a heterogeneous and highly aggressive malignancy, for which there are no effective cures. Identification of a malignant stemlike subtype of HCC may offer patients with a dismal prognosis a potential targeted therapy using c-MET and Wnt pathway inhibitors. MicroRNAs (miRNAs) show promise as diagnostic and prognostic tools for cancer detection and stratification. Using a TRE-c-Met-driven transgenic HCC mouse model, we identified a cluster of 23 miRNAs that is encoded within the Dlk1-Gtl2 imprinted region on chromosome 12qF1 overexpressed in all of the isolated liver tumors. Interestingly, this region is conserved among mammalian species and maps to the human DLK1-DIO3 region on chromosome 14q32.2. We thus examined the expression of the DLK1-DIO3 miRNA cluster in a cohort of 97 hepatitis B virus-associated HCC patients and identified a subgroup (n = 18) of patients showing strong coordinate overexpression of miRNAs in this cluster but not in other cancer types (breast, lung, kidney, stomach, and colon) that were tested. Expression levels of imprinted gene transcripts from neighboring loci in this 14q32.2 region and from a subset of other imprinted sites were concomitantly elevated in human HCC. Interestingly, overexpression of the DLK1-DIO3 miRNA cluster was positively correlated with HCC stem cell markers (CD133, CD90, EpCAM, Nestin) and associated with a high level of serum α-fetoprotein, a conventional biomarker for liver cancer, and poor survival rate in HCC patients. In conclusion, our findings suggest that coordinate up-regulation of the DLK1-DIO3 miRNA cluster at 14q32.2 may define a novel molecular (stem cell-like) subtype of HCC associated with poor prognosis.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidad , Cromosomas Humanos Par 14/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Yoduro Peroxidasa/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Proteínas de la Membrana/genética , MicroARNs/genética , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio , Estudios de Cohortes , Humanos , Hígado/metabolismo , MicroARNs/metabolismo , Familia de Multigenes , Pronóstico , Distribución Tisular , Resultado del Tratamiento , Regulación hacia Arriba
3.
Bioorg Med Chem Lett ; 22(7): 2613-9, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22365762

RESUMEN

Translation of significant biochemical activity of pyridyl aminothiazole class of Chk1 inhibitors into functional CEA potency required analysis and adjustment of both physical properties and kinase selectivity profile of the series. The steps toward optimization of cellular potency included elimination of CDK7 activity, reduction of molecular weight and polar surface area and increase in lipophilicity of the molecules in the series.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/química , Piridinas/síntesis química , Tiazoles/síntesis química , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/química , Diseño de Fármacos , Halogenación , Humanos , Cinética , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Piridinas/farmacología , Relación Estructura-Actividad , Tiazoles/farmacología , Quinasa Activadora de Quinasas Ciclina-Dependientes
4.
Bioorg Med Chem Lett ; 22(7): 2609-12, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22374217

RESUMEN

Pyridyl aminothiazoles comprise a novel class of ATP-competitive Chk1 inhibitors with excellent inhibitory potential. Modification of the core with ethylenediamine amides provides compounds with low picomolar potency and very high residence times. Investigation of binding parameters of such compounds using X-ray crystallography and molecular dynamics simulations revealed multiple hydrogen bonds to the enzyme backbone as well as stabilization of the conserved water molecules network in the hydrophobic binding region.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/química , Piridinas/síntesis química , Tiazoles/síntesis química , Amidas/química , Antineoplásicos/farmacología , Sitios de Unión , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cristalografía por Rayos X , Diseño de Fármacos , Etilenodiaminas/química , Humanos , Enlace de Hidrógeno , Cinética , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Piridinas/farmacología , Relación Estructura-Actividad , Tiazoles/farmacología , Agua/química
5.
SLAS Discov ; 26(2): 216-229, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33482073

RESUMEN

While c-MYC is well established as a proto-oncogene, its structure and function as a transcription factor have made c-MYC a difficult therapeutic target. To identify small-molecule inhibitors targeting c-MYC for anticancer therapy, we designed a high-throughput screening (HTS) strategy utilizing cellular assays. The novel approach for the HTS was based on the detection of cellular c-MYC protein, with active molecules defined as those that specifically decreased c-MYC protein levels in cancer cells. The assay was based on a dual antibody detection system using Förster/fluorescence resonance energy transfer (FRET) and was utilized to detect endogenous c-MYC protein in the MYC amplified cancer cell lines DMS273 and Colo320 HSR. The assays were miniaturized to 1536-well plate format and utilized to screen the GlaxoSmithKline small-molecule collection of approximately 2 million compounds. In addition to the HTS assay, follow-up assays were developed and used to triage and qualify compounds. Two cellular assays used to eliminate false-positive compounds from the initially selected HTS hits were (1) a cellular toxicity assay and (2) an unstable protein reporter assay. Three positive selection assays were subsequently used to qualify compounds: (1) 384-well cell cycle flow cytometry, (2) 384-well cell growth, and (3) c-MYC gene signature reverse transcription quantitative PCR (RT-qPCR). The HTS and follow-up assays successfully identified three compounds that specifically decreased c-MYC protein levels in cancer cells and phenocopied c-MYC siRNA in terms of cell growth inhibition and gene signatures. The HTS, triage, and three compounds identified are described.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes myc , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia/métodos , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Br J Haematol ; 147(5): 672-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19751238

RESUMEN

This study characterized the preclinical anti-myeloma activity of VE465, a low molecular weight pan-Aurora kinase inhibitor. After 96-h drug exposure, several multiple myeloma (MM) cell lines were more sensitive to VE465 compared to non-malignant cells. The anti-MM activity of VE465 was maintained in the presence of interleukin-6 and, interestingly, enhanced by co-culture with stromal cells. However, primary MM cells were less responsive than cell lines. Combinations with dexamethasone (Dex), doxorubicin (Doxo) and bortezomib showed no antagonism. Our study highlights the potential role of the tumour microenvironment in modulating the activity of this drug class.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple/patología , Piperazinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Aurora Quinasas , Comunicación Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales/métodos , Células Madre Hematopoyéticas/citología , Humanos , Células Tumorales Cultivadas
7.
Arch Biochem Biophys ; 484(1): 1-7, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19467625

RESUMEN

A number of inhibitors of kinesin spindle protein (KSP) have been described, which are known from X-ray crystallography studies to bind to an induced fit pocket defined by the L5 loop. We describe the characterization of eight mutant forms of KSP in which six residues that line this pocket have been altered. Mutants were analyzed by measuring rates of enzyme catalysis, in the presence and absence of six KSP inhibitors of four diverse structural classes and of varied ATP-competition status. Our analysis was in agreement with the model of binding established by the structural studies and suggests that binding energy is well distributed across functional groups in these molecules. The majority of the mutants retained significant enzymatic activity while diminishing inhibitor binding, indicating potential for the development of drug resistance. These data provide detailed information on interactions between inhibitor and binding pocket at the functional group level and enable the development of novel KSP inhibitors.


Asunto(s)
Cinesinas/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Biocatálisis , Cristalografía por Rayos X , Humanos , Cinesinas/química , Cinesinas/genética , Cinesinas/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Homología de Secuencia de Aminoácido
8.
Mol Cell Biol ; 26(10): 3853-63, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648480

RESUMEN

KIF14 is a microtubule motor protein whose elevated expression is associated with poor-prognosis breast cancer. Here we demonstrate KIF14 accumulation in mitotic cells, where it associated with developing spindle poles and spindle microtubules. Cells at later stages of mitosis were characterized by the concentration of KIF14 at the midbody. Time-lapse microscopy revealed that strong RNA interference (RNAi)-mediated silencing of KIF14 induced cytokinesis failure, causing several rounds of endoreduplication and resulting in multinucleated cells. Additionally, less efficacious KIF14-specific short interfering RNAs (siRNAs) induced multiple phenotypes, all of which resulted in acute apoptosis. Our data demonstrate the ability of siRNA-mediated silencing to generate epiallelic hypomorphs associated with KIF14 depletion. Furthermore, the link we observed between siRNA efficacy and phenotypic outcome indicates that distinct stages during cell cycle progression are disrupted by the differential modulation of KIF14 expression.


Asunto(s)
Ciclo Celular/fisiología , Citocinesis/fisiología , Silenciador del Gen , Cinesinas/metabolismo , Proteínas Oncogénicas/metabolismo , Interferencia de ARN , Adenosina Trifosfatasas/análisis , Secuencia de Aminoácidos , Apoptosis , Línea Celular , Clonación Molecular , Secuencia de Consenso , Factor de Crecimiento Epidérmico/metabolismo , Colorantes Fluorescentes , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HeLa , Humanos , Immunoblotting , Indoles , Cinesinas/química , Cinesinas/genética , Microscopía Fluorescente , Microscopía por Video , Datos de Secuencia Molecular , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Penetrancia , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética
9.
Bioorg Med Chem Lett ; 19(4): 1240-4, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19155174

RESUMEN

A high throughput screening campaign was designed to identify allosteric inhibitors of Chk1 kinase by testing compounds at high concentration. Activity was then observed at K(m) for ATP and at near-physiological concentrations of ATP. This strategy led to the discovery of a non-ATP competitive thioquinazolinone series which was optimized for potency and stability. An X-ray crystal structure for the complex of our best inhibitor bound to Chk1 was solved, indicating that it binds to an allosteric site approximately 13A from the ATP binding site. Preliminary data is presented for several of these compounds.


Asunto(s)
Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/efectos de los fármacos , Quinazolinas/síntesis química , Quinazolinas/farmacología , Sitios de Unión , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Técnicas Químicas Combinatorias , Cristalografía por Rayos X , Humanos , Conformación Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Quinazolinas/química
10.
Clin Cancer Res ; 14(19): 6106-15, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18829489

RESUMEN

PURPOSE: We determined the effects of vorinostat (suberoylanalide hydroxamic acid) and/or MK-0457 (VX-680), an Aurora kinase inhibitor on the cultured human (HL-60, OCI-AML3, and K562) and primary acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML), as well as on the murine pro-B BaF3 cells with ectopic expression of the unmutated and mutant forms of Bcr-Abl. EXPERIMENTAL DESIGN: Following exposure to MK-0457 and/or vorinostat, apoptosis, loss of viability, as well as activity and levels of Aurora kinase and Bcr-Abl proteins were determined. RESULTS: Treatment with MK-0457 decreased the phosphorylation of Aurora kinase substrates including serine (S)10 on histone H3 and survivin, and led to aberrant mitosis, DNA endoreduplication as well as apoptosis of the cultured human acute leukemia HL-60, OCI-AML3, and K562 cells. Combined treatment with vorinostat and MK-0457 resulted in greater attenuation of Aurora and Bcr-Abl (in K562) kinase activity and levels as well as synergistically induced apoptosis of OCI-AML3, HL-60, and K562 cells. MK-0457 plus vorinostat also induced synergistic apoptosis of BaF3 cells with ectopic overexpression of wild-type or mutant Bcr-Abl. Finally, cotreatment with MK-0457 and vorinostat induced more loss of viability of primary AML and imatinib-refractory CML than treatment with either agent alone, but exhibited minimal toxicity to normal CD34+ progenitor cells. CONCLUSIONS: Combined in vitro treatment with MK-0457 and vorinostat is highly active against cultured and primary leukemia cells. These findings merit in vivo testing of the combination against human AML and CML cells, especially against imatinib mesylate-resistant Bcr-AblT315I-expressing CML Cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácidos Hidroxámicos/administración & dosificación , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Piperazinas/administración & dosificación , Animales , Línea Celular Tumoral , Sinergismo Farmacológico , Proteínas de Fusión bcr-abl/metabolismo , Células HL-60 , Histonas/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis , Células K562 , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Proteínas de Neoplasias/metabolismo , Survivin , Vorinostat
11.
Arch Biochem Biophys ; 469(2): 220-31, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17999913

RESUMEN

The kinesin spindle protein (KSP, also known as Eg5) is essential for the proper separation of spindle poles during mitosis, and inhibition results in mitotic arrest and the formation of characteristic monoaster spindles. Several distinct classes of KSP inhibitors have been described previously in the public and patent literature. However, most appear to share a common induced-fit allosteric binding site, suggesting a common mechanism of inhibition. In a high-throughput screen for inhibitors of KSP, a novel class of thiazole-containing inhibitors was identified. Unlike the previously described allosteric KSP inhibitors, the thiazoles described here show ATP competitive kinetic behavior, consistent with binding within the nucleotide binding pocket. Although they bind to a pocket that is highly conserved across kinesins, these molecules exhibit significant selectivity for KSP over other kinesins and other ATP-utilizing enzymes. Several of these compounds are active in cells and produce a phenotype similar to that observed with previously published allosteric inhibitors of KSP.


Asunto(s)
Adenosina Trifosfato/metabolismo , Bioquímica/métodos , Cinesinas/antagonistas & inhibidores , Cinesinas/química , Mitosis , Adenosina Trifosfato/química , Sitio Alostérico , Unión Competitiva , Diseño de Fármacos , Humanos , Cinesinas/metabolismo , Modelos Biológicos , Modelos Químicos , Nucleótidos/química , Fenotipo , Unión Proteica , Tiazoles/farmacología
12.
J Mol Biol ; 335(2): 547-54, 2004 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-14672662

RESUMEN

We report here the first inhibitor-bound structure of a mitotic motor protein. The 1.9 A resolution structure of the motor domain of KSP, bound with the small molecule monastrol and Mg2+ x ADP, reveals that monastrol confers inhibition by "induced-fitting" onto the protein some 12 A away from the catalytic center of the enzyme, resulting in the creation of a previously non-existing binding pocket. The structure provides new insights into the biochemical and mechanical mechanisms of the mitotic motor domain. Inhibition of KSP provides a novel mechanism to arrest mitotic spindle formation, a target of several approved and investigative anti-cancer agents. The structural information gleaned from this novel pocket offers a new angle for the design of anti-mitotic agents.


Asunto(s)
Cinesinas/antagonistas & inhibidores , Cinesinas/química , Pirimidinas/farmacología , Tionas/farmacología , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Humanos , Magnesio/metabolismo , Microtúbulos/química , Mitosis , Modelos Moleculares , Proteínas Motoras Moleculares , Unión Proteica/genética , Conformación Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad
13.
Mol Cancer Ther ; 1(9): 747-58, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12479371

RESUMEN

Farnesyl:protein transferase (FPTase) inhibitors were developed as anti-Ras drugs, but they fail to inhibit Ki-Ras activity because Ki-Ras can be modified by geranylgeranyl:protein transferase type-I (GGPTase-I). L-778,123, an inhibitor of FPTase and GGPTase-I, was developed in part because it can completely inhibit Ki-Ras prenylation. To support the clinical development of L-778,123, we developed pharmacodynamic assays using peripheral blood mononuclear cells (PBMCs) to measure the inhibition of prenylation of HDJ2 and Rap1A, proteins that are FPTase- and GGPTase-I substrates, respectively. We validated these assays in animal models and show that inhibition of HDJ2 prenylation in mouse PBMCs correlates with the concentration of FPTase inhibitors in blood. In dogs, continuous infusion of L-778,123 inhibited both HDJ2 and Rap1A prenylation in PBMCs, but we did not detect inhibition of Ki-Ras prenylation. We reported previously results from the first L-778,123 Phase I trial that showed a dose-dependent inhibition of HDJ2 farnesylation in PBMCs. In this report, we present additional analysis of patient samples from this trial and a second Phase I trial of L-778,123, and demonstrate the inhibition of both HDJ2 and Rap1A prenylation in PBMC samples. This study represents the first demonstration of GGPTase-I inhibition in humans. However, no inhibition of Ki-Ras prenylation by L-778,123 was detected in patient samples. These results confirm the pharmacologic profile of L-778,123 in humans as a dual inhibitor of FPTase and GGPTase-I, but indicate that the intended target of the drug, Ki-Ras, was not inhibited.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Animales , Perros , Relación Dosis-Respuesta a Droga , Humanos , Immunoblotting , Leucocitos/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Modelos Químicos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Tiempo , Proteínas de Unión al GTP rap1/metabolismo
14.
J Med Chem ; 46(14): 2973-84, 2003 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12825937

RESUMEN

A series of novel diaryl ether lactams have been identified as very potent dual inhibitors of protein farnesyltransferase (FTase) and protein geranylgeranyltransferase I (GGTase-I), enzymes involved in the prenylation of Ras. The structure of the complex formed between one of these compounds and FTase has been determined by X-ray crystallography. These compounds are the first reported to inhibit the prenylation of the important oncogene Ki-Ras4B in vivo. Unfortunately, doses sufficient to achieve this endpoint were rapidly lethal.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Antineoplásicos/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Proteínas del Choque Térmico HSP40 , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Prenilación de Proteína , Relación Estructura-Actividad , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo
15.
J Med Chem ; 45(12): 2388-409, 2002 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-12036349

RESUMEN

A series of macrocyclic 3-aminopyrrolidinone farnesyltransferase inhibitors (FTIs) has been synthesized. Compared with previously described linear 3-aminopyrrolidinone FTIs such as compound 1, macrocycles such as 49 combined improved pharmacokinetic properties with a reduced potential for side effects. In dogs, oral bioavailability was good to excellent, and increases in plasma half-life were due to attenuated clearance. It was observed that in vivo clearance correlated with the flexibility of the molecules and this concept proved useful in the design of FTIs that exhibited low clearance, such as FTI 78. X-ray crystal structures of compounds 49 and 66 complexed with farnesyltransferase (FTase)-farnesyl diphosphate (FPP) were determined, and they provide details of the key interactions in such ternary complexes. Optimization of this 3-aminopyrrolidinone series of compounds led to significant increases in potency, providing 83 and 85, the most potent inhibitors of FTase in cells described to date.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas , Proteínas de Transporte de Catión , Proteínas de Unión al ADN , Inhibidores Enzimáticos/síntesis química , Naftalenos/síntesis química , Canales de Potasio con Entrada de Voltaje , Pirrolidinas/síntesis química , Transactivadores , Animales , Línea Celular , Cromatografía Liquida , Cristalografía por Rayos X , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Perros , Canal de Potasio ERG1 , Electrocardiografía , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Canales de Potasio Éter-A-Go-Go , Farnesiltransferasa , Humanos , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Modelos Moleculares , Estructura Molecular , Naftalenos/química , Naftalenos/farmacocinética , Oxidorreductasas N-Desmetilantes/antagonistas & inhibidores , Canales de Potasio/metabolismo , Unión Proteica , Pirrolidinas/química , Pirrolidinas/farmacocinética , Estereoisomerismo , Relación Estructura-Actividad , Regulador Transcripcional ERG
16.
Oncotarget ; 5(13): 5076-86, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24970803

RESUMEN

The aim of this study was to assess niraparib (MK-4827), a novel poly(ADP-Ribose) polymerase (PARP) inhibitor, for its ability to radiosensitize human tumor cells. Human tumor cells derived from lung, breast and prostate cancers were tested for radiosensitization by niraparib using clonogenic survival assays. Both p53 wild-type and p53-defective lines were included. The ability of niraparib to alter the repair of radiation-induced DNA double strand breaks (DSBs) was determined using detection of γ-H2AX foci and RAD51 foci. Clonogenic survival analyses indicated that micromolar concentrations of niraparib radiosensitized tumor cell lines derived from lung, breast, and prostate cancers independently of their p53 status but not cell lines derived from normal tissues. Niraparib also sensitized tumor cells to H2O2 and converted H2O2-induced single strand breaks (SSBs) into DSBs during DNA replication. These results indicate that human tumor cells are significantly radiosensitized by the potent and selective PARP-1 inhibitor, niraparib, in the in vitro setting. The mechanism of this effect appears to involve a conversion of sublethal SSBs into lethal DSBs during DNA replication due to the inhibition of base excision repair by the drug. Taken together, our findings strongly support the clinical evaluation of niraparib in combination with radiation.


Asunto(s)
Indazoles/farmacología , Piperidinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Roturas del ADN de Cadena Simple/efectos de los fármacos , Roturas del ADN de Cadena Simple/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Femenino , Histonas/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Microscopía Fluorescente , Oxidantes/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Recombinasa Rad51/metabolismo , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Nat Genet ; 44(7): 765-9, 2012 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-22634754

RESUMEN

To survey hepatitis B virus (HBV) integration in liver cancer genomes, we conducted massively parallel sequencing of 81 HBV-positive and 7 HBV-negative hepatocellular carcinomas (HCCs) and adjacent normal tissues. We found that HBV integration is observed more frequently in the tumors (86.4%) than in adjacent liver tissues (30.7%). Copy-number variations (CNVs) were significantly increased at HBV breakpoint locations where chromosomal instability was likely induced. Approximately 40% of HBV breakpoints within the HBV genome were located within a 1,800-bp region where the viral enhancer, X gene and core gene are located. We also identified recurrent HBV integration events (in ≥ 4 HCCs) that were validated by RNA sequencing (RNA-seq) and Sanger sequencing at the known and putative cancer-related TERT, MLL4 and CCNE1 genes, which showed upregulated gene expression in tumor versus normal tissue. We also report evidence that suggests that the number of HBV integrations is associated with patient survival.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Virus de la Hepatitis B/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Integración Viral/genética , Secuencia de Bases , Inestabilidad Cromosómica/genética , Ciclina E/genética , Variaciones en el Número de Copia de ADN/genética , ADN Viral/genética , Proteínas de Unión al ADN/genética , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Oncogénicas/genética , ARN Viral/genética , Tasa de Supervivencia , Telomerasa/genética
18.
Clin Cancer Res ; 17(17): 5638-48, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21799033

RESUMEN

PURPOSE: Radiotherapy is commonly used to treat a variety of solid tumors. However, improvements in the therapeutic ratio for several disease sites are sorely needed, leading us to assess molecularly targeted therapeutics as radiosensitizers. The aim of this study was to assess the wee1 kinase inhibitor, MK-1775, for its ability to radiosensitize human tumor cells. EXPERIMENTAL DESIGN: Human tumor cells derived from lung, breast, and prostate cancers were tested for radiosensitization by MK-1775 using clonogenic survival assays. Both p53 wild-type and p53-defective lines were included. The ability of MK-1775 to abrogate the radiation-induced G2 block, thereby allowing cells harboring DNA lesions to prematurely progress into mitosis, was determined using flow cytometry and detection of γ-H2AX foci. The in vivo efficacy of the combination of MK-1775 and radiation was assessed by tumor growth delay experiments using a human lung cancer cell line growing as a xenograft tumor in nude mice. RESULTS: Clonogenic survival analyses indicated that nanomolar concentrations of MK-1775 radiosensitized p53-defective human lung, breast, and prostate cancer cells but not similar lines with wild-type p53. Consistent with its ability to radiosensitize, MK-1775 abrogated the radiation-induced G2 block in p53-defective cells but not in p53 wild-type lines. MK-1775 also significantly enhanced the antitumor efficacy of radiation in vivo as shown in tumor growth delay studies, again for p53-defective tumors. CONCLUSIONS: These results indicate that p53-defective human tumor cells are significantly radiosensitized by the potent and selective wee1 kinase inhibitor, MK-1775, in both the in vitro and in vivo settings. Taken together, our findings strongly support the clinical evaluation of MK-1775 in combination with radiation.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Proteína p53 Supresora de Tumor/deficiencia , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Terapia Combinada , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Masculino , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/radioterapia , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinonas , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
PLoS One ; 6(9): e24582, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949730

RESUMEN

Biomarkers derived from gene expression profiling data may have a high false-positive rate and must be rigorously validated using independent clinical data sets, which are not always available. Although animal model systems could provide alternative data sets to formulate hypotheses and limit the number of signatures to be tested in clinical samples, the predictive power of such an approach is not yet proven. The present study aims to analyze the molecular signatures of liver cancer in a c-MET-transgenic mouse model and investigate its prognostic relevance to human hepatocellular carcinoma (HCC). Tissue samples were obtained from tumor (TU), adjacent non-tumor (AN) and distant normal (DN) liver in Tet-operator regulated (TRE) human c-MET transgenic mice (n = 21) as well as from a Chinese cohort of 272 HBV- and 9 HCV-associated HCC patients. Whole genome microarray expression profiling was conducted in Affymetrix gene expression chips, and prognostic significances of gene expression signatures were evaluated across the two species. Our data revealed parallels between mouse and human liver tumors, including down-regulation of metabolic pathways and up-regulation of cell cycle processes. The mouse tumors were most similar to a subset of patient samples characterized by activation of the Wnt pathway, but distinctive in the p53 pathway signals. Of potential clinical utility, we identified a set of genes that were down regulated in both mouse tumors and human HCC having significant predictive power on overall and disease-free survival, which were highly enriched for metabolic functions. In conclusions, this study provides evidence that a disease model can serve as a possible platform for generating hypotheses to be tested in human tissues and highlights an efficient method for generating biomarker signatures before extensive clinical trials have been initiated.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-met/genética , Transcriptoma , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Hígado/citología , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Transgénicos , Pronóstico , Análisis de Supervivencia
20.
Cancer Res ; 70(6): 2476-84, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20197467

RESUMEN

ERBB2/neu and Notch signaling are known to be deregulated in many human cancers. However, pathway cross-talk and dependencies are not well understood. In this study, we use an ERBB2-transgenic mouse model of breast cancer (neuT) to show that Notch signaling plays a critical role in tumor maintenance. Inhibition of the Notch pathway with a gamma-secretase inhibitor (GSI) decreased both the Notch and the mammalian target of rapamycin/AKT pathways. Antitumor activity resulting from GSI treatment was associated with decreased cell proliferation as measured by Ki67 and decreased expression of glucose transporter Glut1. Positron emission tomography (PET) imaging showed that the functional consequences of decreased Glut1 translated to reduced glucose uptake and correlated with antitumor effects as measured by micro-computed tomography imaging. The decrease of Glut1 in neuT tumors was also observed in several human breast cancer cell lines following GSI treatment. We provide evidence that approximately 27% of ERBB2-positive human breast cancer specimens display high expression of HES1, phospho-S6RP, and GLUT1. Together, these results suggest that pathways downstream of Notch signaling are, at least in part, responsible for promoting tumor growth in neuT and also active in both neuT and a subset of human breast cancers. These findings suggest that GSI may provide therapeutic benefit to a subset of ERBB2-positive breast cancers and that [(18)F]FDG-PET imaging may be useful in monitoring clinical response.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Glucosa/farmacocinética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor ErbB-2/metabolismo , Receptores Notch/metabolismo , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Óxidos S-Cíclicos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fluorodesoxiglucosa F18/farmacocinética , Transportador de Glucosa de Tipo 1/biosíntesis , Humanos , Neoplasias Mamarias Experimentales , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Tiadiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA