Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Cell Biochem ; 113(11): 3363-70, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22644871

RESUMEN

Several different cytokines and growth factors secreted by mesenchymal stem cells (MSCs) have been hypothesized to play a role in breast cancer progression. By using a small panel of breast cancer cell lines (MCF-7, T47D, and SK-Br-3 cells), we analyzed the role of interleukin-6 (IL-6) and vascular endothelial growth factor A (VEGF) in the cross-talk between MSCs and breast cancer cells. We performed migration assays in which breast cancer cells were allowed to migrate in response to conditioned medium from MSCs (MSCs-CM), in absence or in presence of the anti-VEGF antibody bevacizumab or an anti-IL-6 antibody, alone or in combination. We found that anti-VEGF and anti-IL-6 antibodies inhibited the migration of breast cancer cells and that the combination had an higher inhibitory effect. We next evaluated the effects of recombinant VEGF and IL-6 proteins on breast cancer cell growth and migration. IL-6 and VEGF had not significant effects on the proliferation of breast carcinoma cells. In contrast, both VEGF and IL-6 significantly increased the ability to migrate of MCF-7, T47D and SK-Br-3 cells, with the combination showing a greater effect as compared with treatment with a single protein. The combination of VEGF and IL-6 produced in breast cancer cells a more significant and more persistent activation of MAPK, AKT, and p38MAPK intracellular signaling pathways. These results suggest that MSC-secreted IL-6 and VEGF may act as paracrine factors to sustain breast cancer cell migration.


Asunto(s)
Neoplasias de la Mama/genética , Interleucina-6/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Bevacizumab , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Comunicación Celular , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/farmacología , Células Madre Mesenquimatosas/patología , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Expert Opin Drug Metab Toxicol ; 7(7): 911-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21563999

RESUMEN

INTRODUCTION: Increased bone resorption is associated with several diseases, including osteoporosis, bone metastases and Paget's disease of bone. Zoledronic acid (ZA) is the most potent of the clinically available bisphosphonates. In addition to its antiresorptive activity, there has been increasing evidence to suggest that it also has anticancer properties. AREAS COVERED: This article is complied through PubMed and Medline databases searches on ZA. In this review, the authors summarize the current knowledge (up to December 2010) on the pharmacodynamic and pharmacokinetic properties of ZA. EXPERT OPINION: ZA is a well-tolerated and effective drug in the management of metabolic as well as cancer-related bone disease. Clinical benefits in cancer patients include improvement in bone pain, reduction in skeletal events and delay of time to first skeletal event. However, novel indications for this drug are emerging from clinical studies in early breast cancer. Recent findings suggest that the addition of ZA to endocrine therapy can significantly prevent bone loss in premenopausal patients. Increasing evidence also indicates a potential anticancer activity of ZA, although this property needs to be further explored.


Asunto(s)
Antineoplásicos/farmacocinética , Conservadores de la Densidad Ósea/farmacocinética , Enfermedades Óseas/tratamiento farmacológico , Neoplasias Óseas/tratamiento farmacológico , Quimioterapia Adyuvante/métodos , Difosfonatos/farmacocinética , Imidazoles/farmacocinética , Adyuvantes Farmacéuticos/farmacocinética , Enfermedades Óseas/inducido químicamente , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Ensayos Clínicos como Asunto , Evaluación de Medicamentos , Femenino , Humanos , Osteoporosis/tratamiento farmacológico , Ácido Zoledrónico
4.
J Cell Physiol ; 226(8): 2131-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21520065

RESUMEN

Increasing evidence suggests that bone marrow-derived mesenchymal stem cells (MSCs) are recruited into the stroma of developing tumors where they contribute to cancer progression. MSCs produce different growth factors that sustain tumor-associated neo-angiogenesis. Since the majority of carcinomas secrete ligands of the epidermal growth factor receptor (EGFR), we assessed the role of EGFR signaling in regulating the release of angiogenic factors in MSCs. Treatment of human primary MSCs and of the human osteoblastic cell line hFOB with transforming growth factor α (TGF-α), one of the main ligands of the EGFR, significantly induced activation of this receptor and of different intracellular signaling proteins, including the PI3K/AKT and the MEK/MAPK pathways. TGF-α induced a significant increase in the levels of secretion of vascular endothelial growth factor in both MSCs and hFOB. Conditioned medium from TGF-α treated MSCs showed an higher in vivo angiogenic effect as compared with medium from untreated cells. Treatment of MSCs with TGF-α also produced a significant increase in the secretion of other angiogenic growth factors such as angiopoietin-2, granulocyte-colony stimulating factor, hepatocyte growth factor, interleukin (IL)-6, IL-8, and platelet-derived growth factor-BB. Using selective MEK and PI3K inhibitors, we found that both MEK/MAPK and the PI3K/AKT signaling pathways mediate the ability of TGF-α to induce secretion of angiogenic factors in MSCs. Finally, stimulation with TGF-α increased the ability of MSCs to induce migration of MCF-7 breast cancer cells. These data suggest that EGFR signaling regulates the ability of MSCs to sustain cancer progression through the release of growth factors that promote neo-angiogenesis and tumor cell migration.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Receptores ErbB/metabolismo , Células Madre Mesenquimatosas/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Ligandos , Células Madre Mesenquimatosas/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Factor de Crecimiento Transformador alfa/farmacología
5.
Expert Rev Anticancer Ther ; 11(2): 299-307, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21342047

RESUMEN

Bisphosphonates (BPs) are cornerstones in the treatment of patients with compromised skeletal integrity (either cancer related or not). However, a major indication for BPs use remains the treatment of patients with advanced cancer metastatic to the bone. Recently, several observations derived from clinical trials, primarily aimed at establishing the impact of BPs on the bone health of cancer patients, suggested a potential role for these agents as direct anti-tumor drugs. Consequently, a series of preclinical works were produced with the aim of clarifying the mechanism underlying this observed effect. However, the impact of such data is still under debate owing to the intrinsic weakness of observations from trials not adequately powered to support them. In conclusion, the entire matter remains one of the most intriguing in oncology, and data from ongoing and planned future studies will surely provide us with more information on the great potential of BPs in the adjuvant setting.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/prevención & control , Difosfonatos/farmacología , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Quimioterapia Adyuvante , Ensayos Clínicos como Asunto , Difosfonatos/efectos adversos , Difosfonatos/química , Difosfonatos/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Estructura Molecular
6.
Breast Cancer Res Treat ; 123(2): 387-96, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19946741

RESUMEN

Treatment of breast cancer cells with a combination of the EGFR-tyrosine kinase inhibitor (EGFR-TKI) gefitinib and the anti-ErbB-2 monoclonal antibody trastuzumab results in a synergistic antitumor effect. In this study, we addressed the mechanisms involved in this phenomenon. The activation of signaling pathways and the expression of cell cycle regulatory proteins were studied in SK-Br-3 and BT-474 breast cancer cells, following treatment with EGFR and/or ErbB-2 inhibitors. Treatment with the gefitinib/trastuzumab combination produced, as compared with a single agent, a more prolonged blockade of AKT and MAPK activation, a more pronounced accumulation of cells in the G0/G1 phase of the cell cycle, a more significant increase in the levels of p27(kip1) and of hypophosphorylated pRb2, and a decrease in the levels of Cyclin D1 and survivin. Similar findings were observed with the EGFR/ErbB-2 inhibitor lapatinib. Gefitinib, trastuzumab, and their combination increased the stability of p27(kip1), with the combination showing the highest effects. Blockade of both receptors with gefitinib/trastuzumab or lapatinib induced a significant increase in the levels of p27(kip1) mRNA and in the nuclear levels of the p27(kip1) transcription factor FKHRL-1. Inhibition of PI3K signaling also produced a significant raise in p27(kip1) mRNA. Finally, down-modulation of FKHRL-1 with siRNAs prevented the lapatinib-induced increase of p27(kip1) mRNA. The synergism deriving from EGFR and ErbB-2 blockade is mediated by several different alterations in the activation of signaling proteins and in the expression of cell cycle regulatory proteins, including transcriptional and posttranscriptional regulation of p27(kip1) expression.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/enzimología , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Gefitinib , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lapatinib , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/farmacología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor ErbB-2/metabolismo , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Factores de Tiempo , Trastuzumab
7.
Oncology ; 77 Suppl 1: 2-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20130425

RESUMEN

Analysis by DNA microarrays has led to the identification of molecular subtypes of breast carcinomas that show a distinct expression profile. Several studies have demonstrated that this 'intrinsic subtype' classification has a strong prognostic value. In addition, gene expression profiling techniques have been used to identify gene signatures that could be associated with the outcome of breast cancer patients. Several different genomic tests have been shown to better define the prognosis of early-stage breast cancer patients as compared with conventional clinical and pathological characteristics of the tumors, and some assays are already commercially available. However, it must be emphasized that the prognostic power of these genetic classifiers has not been confirmed yet in prospective trials. Genetic signatures that might predict the activity of specific chemotherapy agents have also been developed by using gene expression profiling techniques. The same approach has been used to identify gene signatures associated with the activation of oncogenic pathways that might represent targets for molecular therapy of breast cancer. By using these approaches, gene expression techniques might significantly improve our ability to predict the risk of recurrence and to tailor the treatment for each individual breast cancer patient.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Femenino , Humanos , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA