Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 26(10): 2079-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26209642

RESUMEN

BACKGROUND: The prognostic and predictive role of KRAS mutations in advanced nonsmall-cell lung cancer (NSCLC) is still unclear. TAILOR prospectively assessed the prognostic and predictive value of KRAS mutations in NSCLC patients treated with erlotinib or docetaxel in second line. PATIENTS AND METHODS: NSCLC patients from 52 Italian hospitals were genotyped for KRAS and EGFR mutational status in two independent laboratories. Wild-type EGFR patients (N = 218) received first-line platinum-based chemotherapy and were randomly allocated at progression to erlotinib or docetaxel. Overall survival (OS) according to KRAS mutational status was the primary end point. RESULTS: KRAS mutations were present in 23% of TAILOR randomized cases. The presence of a KRAS mutation did not adversely affect progression-free (PFS) or overall (OS) survival [hazard ratio (HR) PFS = 1.01, 95% confidence interval (CI) 0.71-1.41, P = 0.977; OS = 1.24, 95% CI 0.87-1.77, P = 0.233], nor influenced treatment outcome (test for interaction: OS P = 0.965; PFS P = 0.417). Patients randomized to docetaxel treatment experienced longer survival independently from the KRAS mutational status of their tumors (HR: mutated KRAS 0.81, 95% CI 0.45-1.47; wild-type KRAS 0.79, 95% CI 0.57-1.10). CONCLUSION: In TAILOR, KRAS was neither prognostic nor predictive of benefit for either docetaxel or erlotinib. Docetaxel remains superior independently from KRAS status for second-line treatment in EGFR wild-type advanced NSCLC patients. CLINICAL TRIAL REGISTRATION: NCT00637910.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Docetaxel , Clorhidrato de Erlotinib/administración & dosificación , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Estadificación de Neoplasias , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia , Taxoides/administración & dosificación
2.
Ann Oncol ; 26(5): 838-847, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25411417

RESUMEN

Thymic epithelial tumors (TETs) are rare primary mediastinal tumors arising from thymic epithelium. Their rarity and complexity hinder investigations of their causes and therapy development. Here, we summarize the existing knowledge regarding medical treatment of these tumors, and thoroughly review the known genetic aberrations associated with TETs and the present status of potential biological treatments. Epidermal growth factor receptor (EGFR), stem-cell factor receptor, insulin-like growth factor-1 receptor (IGF1R), and vascular endothelial growth factors (VEGF-A, VEGF-B, and VEGF-2) are overexpressed in TETs. EGFR overexpression in TETs is associated with higher stage, and IGF1R overexpression has poor prognostic value. Data indicate that anti-IGF1R monoclonal antibodies, and inhibitors of angiogenesis, somatostatin receptors, histone deacetylase, mammalian target of rapamycin, and cyclin-dependent kinases may be active against TETs. Continued investigations in this field could lead to advancement of targeted and biological therapies for TETs.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Productos Biológicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Neoplasias del Timo/tratamiento farmacológico , Animales , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Productos Biológicos/efectos adversos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Humanos , Terapia Molecular Dirigida/efectos adversos , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias del Timo/genética , Neoplasias del Timo/metabolismo , Neoplasias del Timo/patología , Resultado del Tratamiento
3.
Pharmacogenomics J ; 14(5): 401-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25001881

RESUMEN

Drug resistance in epithelial ovarian cancer (EOC) limits the efficacy of therapies for this malignancy. This phenomenon might be partially explained by strong inter-individual genomic heterogeneity. Single nucleotide polymorphisms (SNPs) located in specific genes involved in platinum-based drugs inactivation and the metabolism and extrusion of taxanes could be relevant in terms of drugs response prediction. In this paper, we review candidates for genetic markers of treatment outcomes in ovarian cancer. Although an association between SNPs and response to chemotherapy has been detected in several studies, no clear conclusions can be drawn because of conflicting results. Genetic variants in determining response to chemotherapy and clinical outcome need to be clarified in EOC to allow stratification of patients, which would help optimize therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Femenino , Variación Genética/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Resultado del Tratamiento
4.
Ann Oncol ; 24(1): 14-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23110808

RESUMEN

MET is a tyrosine kinase receptor for hepatocyte growth factor (HGF), primarily expressed on epithelial cells; the activation of MET induces several biological responses relevant for the development and growth of many human cancers. Several human malignancies present altered expression of MET and this is usually associated with poor prognosis and aggressive phenotype. The majority of MET inhibitors in clinical development target directly the receptor through the use of monoclonal antibodies (MAbs) or through small molecule inhibitors of MET kinase activity; small molecule inhibitors are very potent but less specific than MAbs. MET inhibitors are of great clinical interest because of the extensive crosstalk of the HGF/MET axis with many other signaling pathways, including growth factor-dependent pathways (like PI3K/AKT/mTOR,RAS/RAF/ERK) and vascular endothelial growth factor (VEGF) axis. In preclinical studies, the treatment with MET inhibitors could prevent or reverse resistance to inhibitors of growth factor-dependent signaling; this hypothesis is currently tested in phase III trials with anti-epidermal growth factor receptor (EGFR) inhibitors in non-small-cell lung cancer (NSCLC). Based on preclinical and preliminary clinical results, a rational strategy for the clinical development of MET antagonists should include a selection of the tumors with MET overexpression, the identification of prognostic/predictive biomarkers, the evaluation of combinations with anti-VEGF compounds.


Asunto(s)
Antineoplásicos/uso terapéutico , Educación Médica Continua , Oncología Médica , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Antineoplásicos/farmacología , Humanos , Recursos Humanos
5.
Pharmacogenomics J ; 13(2): 159-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22158331

RESUMEN

Epithelial ovarian cancer has a poor prognosis owing to late diagnosis and frequent relapse after first-line therapy. Analysis of individual genetic variability could aid in the identification of markers, which could help in stratifying patients with the aim of optimizing individual therapy. In this study we assessed polymorphisms in three genes important in drugs' response in 97 early and 235 late-stage ovarian cancer patients. The Asp1104His polymorphism in xpg, a gene important for removal of platinum adducts, was associated with progression-free survival in early- and late-stage ovarian cancer. Our data indicate that a simple diagnostic analysis such as xpg genotyping can help in predicting response, and extension to other possibly relevant genotypes could be useful in selecting patients with epithelial ovarian cancer for optimal therapy and hence increase the chance of response.


Asunto(s)
Proteínas de Unión al ADN/genética , Endonucleasas/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores Farmacológicos/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN , Supervivencia sin Enfermedad , Femenino , Estudios de Asociación Genética , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/patología , Platino (Metal)/administración & dosificación , Platino (Metal)/efectos adversos , Polimorfismo Genético , Pronóstico , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-mdm2/genética , Resultado del Tratamiento
6.
J Biol Regul Homeost Agents ; 25(2 Suppl): S63-74, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22051172

RESUMEN

Microcarrier culture systems offer an attractive method for cell amplification and as delivery vehicle. At the same time, super paramagnetic iron oxide (SPIO) nanoparticles represent a unique in vivo tracking system, already approved for clinical use. In our study, we tested the combination of clinically approved microcarriers and SPIO nanoparticles for cell-construct delivery and subsequent tracking after implantation. In order to mimic better a clinical setting, biodegradable macroporous microcarriers were employed as an alternative approach to expand human primary chondrocytes in a dynamic culture system for subsequent direct transplantation. In addition, cellseeded microcarriers were labeled with SPIO nanoparticles to evaluate the benefits of cell-constructs tracking with magnetic resonance. In vivo subcutaneous implants were monitored for up to 3 weeks and orthotopic implantation was simulated and monitored in ex vivo osteochondral defects.


Asunto(s)
Condrocitos/citología , Condrocitos/trasplante , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/química , Animales , Células Cultivadas , Células Inmovilizadas/citología , Células Inmovilizadas/trasplante , Femenino , Humanos , Masculino , Ensayo de Materiales/métodos , Ratones , Ratones Desnudos , Trasplante Heterólogo
8.
ESMO Open ; 6(1): 100034, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33422766

RESUMEN

BACKGROUND: Platinum-based therapy, combined or not with immune checkpoint inhibitors, represents a front-line choice for patients with non-small-cell lung cancer (NSCLC). Despite the improved outcomes in the last years for this malignancy, only a sub-group of patients have long-term benefit. Excision repair cross-complementation group 1 (ERCC1) has been considered a potential biomarker to predict the outcome of platinum-based chemotherapy in NSCLC. However, the ERCC1 gene is transcribed in four splice variants where the isoform 202 was described as the only one active and able to complex Xeroderma pigmentosum group F-complementing protein (XPF). Here, we prospectively investigated if the active form of ERCC1, as assessed by the ERCC1/XPF complex (ERCC1/XPF), could predict the sensitivity to platinum compounds. PATIENTS AND METHODS: Prospectively enrolled, patients with advanced NSCLC treated with a first-line regimen containing platinum were centrally evaluated for ERCC1/XPF by a proximity ligation assay. Overall survival (OS), progression-free survival (PFS) and objective response rate (ORR) were analyzed. RESULTS: The absence of the ERCC1/XPF in the tumor suggested a trend of worst outcomes in terms of both OS [hazard ratio (HR) 1.41, 95% confidence interval (CI) 0.67-2.94, P = 0.373] and PFS (HR 1.61, 95% CI 0.88-3.03, P = 0.123). ORR was marginally influenced in ERCC1/XPF-negative and -positive groups [odds ratio (stable disease + progressive disease versus complete response + partial response) 0.87, 95% CI 0.25-3.07, P = 0.832]. CONCLUSION: The lack of ERCC1/XPF complex in NSCLC tumor cells might delineate a group of patients with poor outcomes when treated with platinum compounds. ERCC1/XPF absence might well identify patients for whom a different therapeutic approach could be necessary.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Platino (Metal)/uso terapéutico , Estudios Prospectivos
9.
Br J Cancer ; 102(1): 104-14, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20051961

RESUMEN

BACKGROUND: Owing to its role in cancer, the phosphoinositide 3-kinase (PI3K)/Akt pathway is an attractive target for therapeutic intervention. We previously reported that the inhibition of Akt by inositol 1,3,4,5,6-pentakisphosphate (InsP(5)) results in anti-tumour properties. To further develop this compound we modified its structure to obtain more potent inhibitors of the PI3K/Akt pathway. METHODS: Cell proliferation/survival was determined by cell counting, sulphorhodamine or acridine orange/ethidium bromide assay; Akt activation was determined by western blot analysis. In vivo effect of compounds was tested on PC3 xenografts, whereas in vitro activity on kinases was determined by SelectScreen Kinase Profiling Service. RESULTS: The derivative 2-O-benzyl-myo-inositol 1,3,4,5,6-pentakisphosphate (2-O-Bn-InsP(5)) is active towards cancer types resistant to InsP(5) in vitro and in vivo. 2-O-Bn-InsP(5) possesses higher pro-apoptotic activity than InsP(5) in sensitive cells and enhances the effect of anti-cancer compounds. 2-O-Bn-InsP(5) specifically inhibits 3-phosphoinositide-dependent protein kinase 1 (PDK1) in vitro (IC(50) in the low nanomolar range) and the PDK1-dependent phosphorylation of Akt in cell lines and excised tumours. It is interesting to note that 2-O-Bn-InsP(5) also inhibits the mammalian target of rapamycin (mTOR) in vitro. CONCLUSIONS: InsP(5) and 2-O-Bn-InsP(5) may represent lead compounds to develop novel inhibitors of the PI3K/Akt pathway (including potential dual PDK1/mTOR inhibitors) and novel potential anti-cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Fosfatos de Inositol/química , Fosfatos de Inositol/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/enzimología , Línea Celular Tumoral/trasplante , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Fosfatos de Inositol/síntesis química , Fosfatos de Inositol/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell Death Differ ; 15(5): 849-58, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18259191

RESUMEN

Regulation of the p73 gene is complex due to the presence of two promoters and the very complex mRNA maturation in both the N-terminal and C-terminal parts of the protein. We have found an additional regulation mechanism for the p73-alpha form that occurs through proteolytic cleavage connected to the activity of the serine protease HtrA2. Following apoptotic stimuli, HtrA2 accumulates in the nucleus and cleaves p73alpha in the C-terminal portion, enabling the protein to increase its transactivation activity on the apoptotic gene bax but not on the cell-cycle regulator gene p21. In the presence of HtrA2, p73 is more prone to cause caspase activation and nuclei fragmentation: p73 needs HtrA2 to activate and enhance its apoptotic functions. This new relation between p73 and HtrA2 may help to understand the different behavior of the p73 protein in cell physiology and in the responses of cancer cells to chemotherapy.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Mitocondriales/metabolismo , Proteínas Nucleares/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína X Asociada a bcl-2 , Línea Celular Tumoral , Núcleo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Proteínas Mitocondriales/genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Serina Endopeptidasas/genética , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/genética , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
11.
Ann Oncol ; 19(3): 501-7, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17998283

RESUMEN

BACKGROUND: P63 belongs to the 'p53 family' whose role in cancer progression has been recently revisited in light of the plethora of splicing variants that are generated. We analyzed the expression of the full-length TAp63 gene and its dominant-negative form deltaNp63 in ovarian cancer biopsies to correlate their expression with clinical outcome. MATERIALS AND METHODS: Real-time RT-PCR analysis was used to determine the levels of TAp63 and deltaNp63 in 83 stage I and in 86 stage III ovarian cancer biopsies and in seven human ovarian cancer cell. RESULTS: TAp63 levels were comparable in stage I and stage III, but deltaNp63 levels increased 77-fold in stage III, independently of the p53 status. Patients with high deltaNp63 expression had the worst overall survival (OS); patients with a deltaNp63/TAp63 ratio >2 had a poor OS. Patients with a high deltaNp63/TAp63 ratio were those with a poor response to platinum-based therapy. CONCLUSIONS: Data indicate a role for deltaNp63 as a potential biomarker to predict patient's outcome and tumor progression in ovarian cancer. This would have particularly clinical relevance in ovarian cancer where the high rate of mortality reflects our lack of knowledge of molecular mechanisms underlying cell progression toward malignancy.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Unión al ADN/análisis , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Transactivadores/análisis , Proteínas Supresoras de Tumor/análisis , Biopsia , Progresión de la Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Neoplasias Ováricas/patología , Análisis de Supervivencia , Factores de Transcripción
12.
Reumatismo ; 59(1): 75-84, 2007.
Artículo en Italiano | MEDLINE | ID: mdl-17435846

RESUMEN

Gout was a well known disease in antiquity, even if often confused with other arthritic conditions. In this paper the XVII century physicians' knowledge about gout is discussed. In particular the chapter about gout by Ludovico Settalain his book Animadversionum, & cautionum Medicarum (1652) is reported. A story is also reported, described in many reports of the time, about a man who died under the land-slide which destroyed the town of Piuro in 1618: he was awfully disfigured by the land-slide, but was easily recognized by the severe gout he suffered in his hands..


Asunto(s)
Gota/historia , Libros Ilustrados/historia , Desastres/historia , Historia del Siglo XVII , Humanos , Italia , Masculino , Ilustración Médica/historia
13.
Cell Death Differ ; 12(7): 805-14, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15877106

RESUMEN

The p73 gene has a complex regulation, which leads to the expression of different isoforms, often with opposite biological effects. We have generated in the human colocarcinoma cell line HCT116, expressing a wild-type p53, an inducible DNp73alpha expressing system. Two clones (HCT116/DN3 and HCT116/DN14), upon doxycycline addition, show a strong expression of DNp73alpha. In vitro the two DNp73alpha overexpressing clones grow at similar rate of the control transfected clone (HCT116/8a) and similarly respond to DNA damage. When injected in mice, HCT116/DN3, HCT116/DN14, and HCT116/8a cells grew similarly in the absence or presence of tetracycline. In HCT116/DN3 and HCT116/DN14 tumors, tetracycline induced a strong expression of DNp73alpha both as mRNA and protein. These results indicate that in this system the overexpression of the DNp73alpha does not induce a more aggressive phenotype and does not seem to be associated with a reduced response of the cells to treatment with anticancer agents.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Doxorrubicina/farmacología , Doxiciclina/farmacología , Expresión Génica , Genes Supresores de Tumor , Células HCT116 , Humanos , Ratones , Neoplasias/genética , Fenotipo , Proteína Tumoral p73 , Proteínas Supresoras de Tumor
15.
Nucleic Acids Res ; 28(2): 513-9, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10606650

RESUMEN

The transcriptional activity of the p53 tumor suppressor protein is crucial for the regulation of cell growth, apoptosis and tumor progression. The first identified p53 relative, p73, was reported to be monoallelically expressed in normal tissues. In some tumors, loss of heterozygosity was associated with overexpression of the silent allele. Human p73alpha was transfected into the wild-type p53-expressing human ovarian carcinoma cell line A2780. Unlike human osteosarcoma Saos-2 cells, A2780 cells could tolerate hyperexpression of p73alpha and clones over-expressing p73alpha could be isolated. No p53-p73 protein-protein interaction was found in these clones in co-immunoprecipitation experiments. Endogenous p53 transcriptional activity was markedly decreased both when p73 was integrated into the genome and in transient transfections using a reporter plasmid containing the p53 binding site linked to luciferase. Transient transfection of p73 with a mutation in the DNA-binding domain did not show these effects. The competition for p53 DNA binding by p73alpha was also evident in gel shift experiments. The results suggest that p73 can modulate p53 function by inhibiting its DNA binding and that overexpression of p73 in tumors might be a novel mechanism of inactivation of p53.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Genes Supresores de Tumor , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/patología , Proteína p53 Supresora de Tumor/metabolismo , Sitios de Unión , Cisplatino/farmacología , ADN de Neoplasias/metabolismo , Proteínas de Unión al ADN/genética , Doxorrubicina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Paclitaxel/farmacología , Pruebas de Precipitina , Transcripción Genética/efectos de los fármacos , Transfección , Células Tumorales Cultivadas , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor
16.
J Int Med Res ; 34(4): 348-54, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16989489

RESUMEN

The aim of this study was to evaluate plasma and synovial fluid concentrations of the non-steroidal anti-inflammatory drug nimesulide and its major metabolite (hydroxynimesulide, M1), after a single 100 mg dose of nimesulide and a repeated (14 day) administration, 100 mg twice a day, in patients with osteoarthritis of the knee and joint effusion. Nimesulide was rapidly absorbed in plasma and distributed in synovial fluid. On day 1, effective concentrations were present 30 min after the first dose and on day 14, the synovial fluid concentration of nimesulide was significantly higher than that measured on day 1; no accumulation was observed in plasma. After 14 days of treatment, both the plasma and synovial fluid concentrations of M1 were significantly higher than those measured on day 1. These data may help to explain the rapid onset of the analgesic effect of nimesulide demonstrated in several clinical conditions, including painful osteoarthritis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Osteoartritis de la Rodilla/metabolismo , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética , Líquido Sinovial/metabolismo , Administración Oral , Anciano , Antiinflamatorios no Esteroideos/sangre , Femenino , Humanos , Masculino , Sulfonamidas/análisis , Sulfonamidas/sangre , Líquido Sinovial/química
17.
Cancer Res ; 61(3): 935-8, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11221886

RESUMEN

We examined the consequences of p73alpha overexpression on gene expression and cellular response to anticancer agents in clones from the human ovarian cancer cell line A2780. Using microarray filters, the expression of 588 genes in two clones overexpressing p73alpha (A2780/p73.4 and A2780/ p73.5) in comparison with empty vector-transfected cells was evaluated. There were clear differences in gene expression profiles. Both of the clones showed a marked increase in the expression of genes involved in DNA repair, including genes participating in nucleotide excision repair and mismatch repair. This was confirmed by reverse transcription-PCR and Northern blot analysis and was associated with an increase in the ability of p73alpha-expressing clones to repair two different DDP (cis-dichlorodiammine platinum)-damaged plasmids in a host reactivation assay. p73alpha overexpressing clones were less sensitive than parental cells to alkylating agents treatment or UV radiation but equally sensitive to the topoisomerase I inhibitor topotecan, which indicated that the increase in expression of DNA repair genes has implications for the response to DNA damaging agents.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/biosíntesis , Proteínas Nucleares/biosíntesis , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , División Celular , Cisplatino/farmacología , Células Clonales , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Proteínas Nucleares/genética , Neoplasias Ováricas/tratamiento farmacológico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Topotecan/farmacología , Transfección , Células Tumorales Cultivadas , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor , Rayos Ultravioleta/efectos adversos
18.
Cancer Res ; 57(8): 1442-6, 1997 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9108443

RESUMEN

We tested the ability of synthetic peptides derived from p21(WAF1), fused to the internalization peptide sequence derived from Antennapedia, to inhibit the growth of cancer cells in two human ovarian cancer cell lines expressing wild-type p53 or not. Two fused peptides corresponding to p21(WAF1) regions 17-33 and 63-77 inhibited cell growth in both cell lines while the same peptides without the internalization sequence were inactive. The fused peptides prevented growth at concentrations which inhibited cyclin-dependent kinase 2 and cdc2 activity, thus demonstrating that the peptides act by mimicking the action of p21(WAF1) on kinases. This study illustrates the potential pharmacological use of small peptides fused with the Antennapedia internalization sequence in proliferative disorders. The approach may be extended to other diseases in which cell penetration of a peptide may be of therapeutic benefit. More stable drug-like molecules with better pharmacological properties could be designed based on the results obtained with peptides.


Asunto(s)
Antineoplásicos/farmacología , Quinasas CDC2-CDC28 , Ciclinas/química , Proteínas de Homeodominio/química , Proteínas Nucleares , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Factores de Transcripción , Proteína con Homeodominio Antennapedia , Antineoplásicos/química , Antineoplásicos/metabolismo , División Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Femenino , Citometría de Flujo , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
19.
Cancer Res ; 48(8): 2036-41, 1988 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-3349474

RESUMEN

N10-Propargyl-5,8-dideazafolic acid (CB 3717), a new antifolate which directly inhibits thymidylate synthase and which is now under early clinical investigation, was compared with methotrexate (MTX) for its antiproliferative activity and mode of action on M14 human melanoma cell line and NIH/3T3 murine fibroblasts transfected with human c-Ha-ras oncogene (NIH/3T3R). CB 3717 was as active as MTX on both cell lines in inhibiting colony formation, but 20-100 times less potent. After 24 h of exposure both drugs caused an accumulation of cells in the G1 phase of the cell cycle, probably because of inhibition of DNA synthesis and blockage at the G1-S boundary. In NIH/3T3R treated for 16 h with 2 microM MTX or 200 microM CB 3717, we found DNA single-strand breaks amounting to approximately 130 and 140 rad equivalents, respectively, and a considerable number of DNA double-strand breaks, far more than expected if they had been the result of the proximity of single-strand breaks on the two complementary DNA strands. No DNA-protein cross-links were detected. When cells were incubated in drug-free medium for 8 h, there was a further accumulation of single-strand breaks, possibly due to the effects of the drug retained intracellularly as polyglutamyl derivative. Simultaneous treatment with 1.77 microM cycloheximide prevented DNA damage produced by both drugs. Thymidine (10 microM), renewed in the culture medium every 24 h, also prevented DNA damage and cytotoxicity. Since after 16 h treatment with MTX or CB 3717 cells were completely viable, as assessed by [3H]thymidine release, trypan blue exclusion test, and 51Cr release, DNA damage appears to be an early event preceding cell death and may be a feature of the killing ability of the drugs. The involvement of a protein in the formation of DNA breaks is suggested by the fact that when protein synthesis was inhibited with cycloheximide DNA damage was no longer seen.


Asunto(s)
Daño del ADN , ADN/efectos de los fármacos , Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/análogos & derivados , Metotrexato/farmacología , Quinazolinas/farmacología , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cicloheximida/farmacología , Ácido Fólico/farmacología , Humanos , Proteínas/metabolismo , Timidina/metabolismo , Timidina/farmacología
20.
Cancer Res ; 47(18): 4884-9, 1987 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-3621181

RESUMEN

DNA damage caused by methazolastone [an analogue of 5-(3,3-dimethyl-1-triazeno)imidazole-4-carboxamide which does not require metabolic activation] was investigated in L-1210 leukemia which is sensitive to this drug and in a L-1210 subline [L-1210/N,N-bis(2-chloroethyl)-N-nitrosourea (BCNU)] which is resistant to both chloroethylnitrosoureas and methyltriazenes. Both in vitro and in vivo metazolastone caused formation of DNA alkali-labile sites (assessed by alkaline elution techniques) which were present in similar amounts and repaired at a similar rate in L-1210 and L-1210/BCNU. This suggests that these lesions are not crucial to methyltriazenes activity. DNA alkali-labile sites may be due to the removal of 7-methylguanine by 7-methylguanine-DNA glycosylase which showed the same activity in L-1210 and L-1210/BCNU. Flow cytometry studies revealed that in L-1210 but not in L-1210/BCNU methazolastone induced an arrest of cells in SL-G2-M phases. This blockade was delayed, occurring after at least two cell divisions after drug treatment and therefore appeared temporally unrelated to the presence of DNA alkali-labile sites. There was three times more O6-methylguanine-DNA methyltransferase in L-1210/BCNU than in L-1210 suggesting that methylation of O6-guanine is an important lesion for methyltriazenes activity and resistance to this drug may be linked to its repair.


Asunto(s)
Antineoplásicos/farmacología , Carmustina/farmacología , Daño del ADN , Reparación del ADN , Dacarbazina/análogos & derivados , Leucemia L1210/tratamiento farmacológico , Animales , ADN de Neoplasias/análisis , Dacarbazina/farmacología , Resistencia a Medicamentos , Citometría de Flujo , Metilación , Metiltransferasas/análisis , Ratones , O(6)-Metilguanina-ADN Metiltransferasa , Temozolomida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA