Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
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
2.
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
4.
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
5.
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
6.
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
7.
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
8.
Curr Med Chem ; 19(8): 1164-76, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22257058

RESUMEN

The RAS/RAF/MEK signaling pathway plays a central role in mediating both proliferation and survival of cancer cells. These proteins are a group of serine/threonine kinases activated in response to a variety of extracellular stimuli and mediate signal transduction from the cell surface towards both nuclear and cytosolic targets. In combination with several other signaling pathways, they can differentially alter phosphorylation status of the transcription factors. A controlled regulation of these cascades is involved in cell proliferation and differentiation, whereas an unregulated activation of these kinases can result in oncogenesis. Dysregulation of the RAS/RAF/MEK pathway has been detected in more than 30% of human tumors, however mutations in the MEK1 and MEK2 genes are seldom, so that hyperactivation of MEK1/2 usually results from gain-of-function mutations in RAS and/or B-RAF. In addition, alteration of the pathways is often associated with drug resistance in the clinic, such as the case of K-RAS mutant expressing tumors. Since RAS protein is a difficult target, alternative ways altering post-translational modifications using farnesyl transferase inhibitors have been adopted. Drug discovery programs have therefore largely focused on B-RAF and MEK. In this review we will discuss the most promising strategies developed to target these kinases and the most recent inhibitors facing the preclinical and clinical setting, also considering their structure-activity relationship (SAR).


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Quinasas raf/antagonistas & inhibidores , Proteínas ras/antagonistas & inhibidores , Animales , Antineoplásicos/química , Inhibidores Enzimáticos/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Relación Estructura-Actividad , Quinasas raf/metabolismo , Proteínas ras/metabolismo
9.
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
10.
Eur J Cancer ; 47(7): 1086-94, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21216588

RESUMEN

The purpose of this study was to investigate retrospectively the mRNA expression of genes involved in different DNA repair pathways implicated in processing platinum-induced damage in 171 chemotherapy-naïve ovarian tumours and correlate the expression of the different genes with clinical parameters. The expression of genes involved in DNA repair pathways (PARP1, ERCC1, XPA, XPF, XPG, BRCA1, FANCA, FANCC, FANCD2, FANCF and PolEta), and in DNA damage transduction (Chk1 and Claspin) was measured by RT-PCR in 13 stage I borderline and 77 stage I and 88 III ovarian carcinomas. ERCC1, XPA, XPF and XPG genes were significantly less expressed in stage III than in stage I carcinoma; BRCA1, FANCA, FANCC, FANCD2 gene expressions were low in borderline tumours, higher in stage I carcinomas and lower in stage III samples. High levels of ERCC1, XPA, FANCC, XPG and PolEta correlated with an increase in Overall Survival (OS) and Progression Free Survival (PFS), whilst high BRCA1 levels were associated with PFS on univariate analysis. With multivariate analyses no genes retained an association when adjusted by stage, grade and residual tumour. A tendency towards a better PFS was observed in patients with the highest level of ERCC1 and BRCA1 after platinum-based therapy than those given both platinum and taxol. The expression of DNA repair genes differed in borderline stage I, stage I and stage III ovarian carcinomas. The role of DNA repair genes in predicting the response in ovarian cancer patients seems far from being established.


Asunto(s)
Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Proteínas de Unión al ADN/genética , Supervivencia sin Enfermedad , Endonucleasas/genética , Femenino , Genes BRCA1 , Humanos , Persona de Mediana Edad , Análisis Multivariante , Paclitaxel/farmacología , ARN Mensajero/metabolismo , Resultado del Tratamiento
12.
Curr Med Chem ; 17(36): 4433-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21062259

RESUMEN

Phisiological activation of PI3K pathway is necessary for cells to regulate many different physiological processes such as transcription, protein synthesis, metabolic responses and membrane trafficking. Abnormal activation of the PI3K pathway leads to an increased activity resulting in tumor onset, maintenance, progression and invasion. Both genetic and epigenetic alterations could affect the normal pathway's activation. Ovarian cancer is the leading cause of death from gynaecological malignancies in the western world. PI3K pathway has been recorded as one of the most deregulated signalling pathway in many tumors, including ovarian ones. So it could be considered an attractive target to be investigated with the various classes of chemical compounds already present or in development. In this rewiew we'll try to discuss the published data of the inhibitors targeting members of the PI3K/ akt/ mTOR pathway in the ovarian cancer setting from a preclinical and clinical point of view, with particular emphasis on drugs combination and strategies of administration. Relevant issues and limitations to the use of particular compounds will be also addressed.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Antineoplásicos/química , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
13.
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
14.
Eur J Cancer ; 45(3): 443-53, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19008096

RESUMEN

The p73 locus gene has a complex structure encoding a plethora of isoforms. The different DeltaN truncated isoforms of p73 may exert different activities depending on the cellular context. The beta isoform of DeltaNp73 seems to have a particular pattern of action even if its role in cell cycle and mitosis is still under investigation. To gain further knowledge of DeltaNp73beta's function, we investigated the effects of its over-expression in tumour cellular models, using the tetracycline-inducible expression system. In the human lung carcinoma cell line H1299, DeltaNp73beta over-expression resulted in suppression of cell growth and in cell death. Surprisingly stable over-expression of DeltaNp73beta impaired the genomic stability of tumour cells, leading to the formation of tetraploid cells. The cells become enlarged and multinucleate, with incorrect mitotic figures, and died by apoptotic-independent pathways. Our data suggest that DeltaNp73beta-induced aberrant mitosis evades the control of the mitotic spindle assay checkpoint, leading to tetraploidy and cell death through mitotic catastrophe rather than apoptosis. The various C-terminal regions of DeltaNp73 may influence the final cellular phenotype and we assume that the beta one in particular could be important in both cell growth control and regulation of mitosis.


Asunto(s)
Apoptosis/genética , Proteínas de Unión al ADN/genética , Mitosis/genética , Proteínas Nucleares/genética , Poliploidía , Carcinoma Pulmonar de Células Pequeñas/genética , Proteínas Supresoras de Tumor/genética , Apoptosis/fisiología , Ciclo Celular/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Mitosis/fisiología , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
16.
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
17.
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
18.
Oncogene ; 27(12): 1805-11, 2008 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17891178

RESUMEN

RASSF2 is a recently identified member of a class of novel tumour suppressor genes, all containing a ras-association domain. RASSF2 resides at 20p13, a region frequently lost in human cancers. In this report we investigated methylation status of the RASSF2 promoter CpG island in a series of breast, ovarian and non-small cell lung cancers (NSCLC). RASSF2 was frequently methylated in breast tumour cell lines (65%, 13/20) and in primary breast tumours (38%, 15/40). RASSF2 expression could be switched back on in methylated breast tumour cell lines after treatment with 5'-aza-2'deoxycytidine. RASSF2 was also frequently methylated in NSCLC tumours (44%, (22/50). The small number of corresponding normal breast and lung tissue DNA samples analysed were unmethylated. We also did not detect RASSF2 methylation in ovarian tumours (0/17). Furthermore no mutations were found in the coding region of RASSF2 in these ovarian tumours. We identified a highly conserved putative bipartite nuclear localization signal (NLS) and demonstrated that endogenous RASSF2 localized to the nucleus. Mutation of the putative NLS abolished the nuclear localization. RASSF2 suppressed breast tumour cell growth in vitro and in vivo, while the ability of NLS-mutant RASSF2 to suppress growth was much diminished. Hence we demonstrate that RASSF2 has a functional NLS that is important for its tumour suppressor gene function. Our data from this and a previous report indicate that RASSF2 is frequently methylated in colorectal, breast and NSCLC tumours. We have identified RASSF2 as a novel methylation marker for multiple malignancies and it has the potential to be developed into a valuable marker for screening several cancers in parallel using promoter hypermethylation profiles.


Asunto(s)
Neoplasias de la Mama/genética , Epigénesis Genética/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Genes Supresores de Tumor/fisiología , Neoplasias Pulmonares/genética , Proteínas/genética , Secuencia de Aminoácidos , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular , Línea Celular Tumoral , Metilación de ADN , Perros , Femenino , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Neoplasias Ováricas/genética , Proteínas/metabolismo , Ratas , Proteínas Supresoras de Tumor
19.
Int J Clin Pract ; 61(8): 1270-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17590218

RESUMEN

OBJECTIVE: This study was designed to investigate the analgesic effects of nimesulide and celecoxib in patients with knee osteoarthritis (OA). In patients with joint effusion, the effects of these non-steroidal anti-inflammatory drugs (NSAIDs) on synovial fluid concentrations of substance P (SP), interleukin (IL)-6 and IL-8 also were evaluated. METHODS: Patients were randomly assigned either nimesulide (100 mg twice a day) or celecoxib (200 mg once a day) for 2 weeks. The intensity of joint pain was assessed with a 100-mm visual analogue scale (VAS). Furthermore, patients completed questions about analgesic efficacy and overall tolerability of the treatments on a five-point categorical scale. Synovial fluid samples were drawn at baseline, 30 min after the first drug intake (day 1), and 30 min after the last drug intake (day 14). RESULTS: We enrolled 44 patients, 20 of whom had a joint effusion. In this group, the effects of nimesulide were more marked than for celecoxib, with evidence of a faster onset of the analgesic action. Both after a single or repeated administration, nimesulide significantly reduced the synovial fluid concentrations of SP and IL-6. Celecoxib, on the other hand, did not change the concentrations of SP and significantly reduced the levels of IL-6 only on day 14. None of the drugs affected IL-8. Both drugs were generally well tolerated. CONCLUSIONS: These results provide evidence that nimesulide is an effective agent for the symptomatic treatment of OA. The effect on inflammatory pain mediators is consistent with the fast analgesic action of this NSAID.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Artralgia/tratamiento farmacológico , Osteoartritis de la Rodilla/tratamiento farmacológico , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Anciano , Celecoxib , Método Doble Ciego , Femenino , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Dimensión del Dolor , Sustancia P/metabolismo , Líquido Sinovial/química , Resultado del Tratamiento
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA