Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Gut ; 62(1): 112-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22466618

RESUMEN

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA. METHODS: Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies. RESULTS: PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility. CONCLUSIONS: The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/fisiología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/fisiología , Ácido Hialurónico/fisiología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/fisiopatología , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/farmacología , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/farmacología , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/fisiopatología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Análisis de Matrices Tisulares , Resultado del Tratamiento , Gemcitabina
2.
Cancer Cell ; 5(4): 375-87, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15093544

RESUMEN

Activating mutations in the ras oncogene are not considered sufficient to induce abnormal cellular proliferation in the absence of cooperating oncogenes. We demonstrate that the conditional expression of an endogenous K-ras(G12D) allele in murine embryonic fibroblasts causes enhanced proliferation and partial transformation in the absence of further genetic abnormalities. Interestingly, K-ras(G12D)-expressing fibroblasts demonstrate attenuation and altered regulation of canonical Ras effector signaling pathways. Widespread expression of endogenous K-ras(G12D) is not tolerated during embryonic development, and directed expression in the lung and GI tract induces preneoplastic epithelial hyperplasias. Our results suggest that endogenous oncogenic ras is sufficient to initiate transformation by stimulating proliferation, while further genetic lesions may be necessary for progression to frank malignancy.


Asunto(s)
Transformación Celular Neoplásica , Anomalías Congénitas/genética , Fibroblastos/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Genes ras/fisiología , Neoplasias/genética , Animales , Ciclo Celular , División Celular , Senescencia Celular , Anomalías Congénitas/patología , Cruzamientos Genéticos , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Embrión de Mamíferos/citología , Femenino , Fibroblastos/metabolismo , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neoplasias/patología , Células Madre/patología , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/metabolismo
3.
Cancer Cell ; 4(6): 437-50, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14706336

RESUMEN

To evaluate the role of oncogenic RAS mutations in pancreatic tumorigenesis, we directed endogenous expression of KRAS(G12D) to progenitor cells of the mouse pancreas. We find that physiological levels of Kras(G12D) induce ductal lesions that recapitulate the full spectrum of human pancreatic intraepithelial neoplasias (PanINs), putative precursors to invasive pancreatic cancer. The PanINs are highly proliferative, show evidence of histological progression, and activate signaling pathways normally quiescent in ductal epithelium, suggesting potential therapeutic and chemopreventive targets for the cognate human condition. At low frequency, these lesions also progress spontaneously to invasive and metastatic adenocarcinomas, establishing PanINs as definitive precursors to the invasive disease. Finally, mice with PanINs have an identifiable serum proteomic signature, suggesting a means of detecting the preinvasive state in patients.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Genes ras/fisiología , Mutación , Neoplasias Pancreáticas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Carcinoma Ductal Pancreático/metabolismo , Ciclooxigenasa 2 , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Isoenzimas/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Proteínas de la Membrana , Ratones , Metástasis de la Neoplasia , Estadificación de Neoplasias , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Factor de Transcripción HES-1
4.
Clin Cancer Res ; 14(15): 4836-42, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676756

RESUMEN

PURPOSE: This study evaluated the safety, maximum tolerated dose, pharmacokinetics, and antitumor activity of sorafenib, a multikinase inhibitor, combined with paclitaxel and carboplatin in patients with solid tumors. PATIENTS AND METHODS: Thirty-nine patients with advanced cancer (24 with melanoma) received oral sorafenib 100, 200, or 400 mg twice daily on days 2 to 19 of a 21-day cycle. All patients received carboplatin corresponding to AUC6 and 225 mg/m(2) paclitaxel on day 1. Pharmacokinetic analyses were done for sorafenib on days 2 and 19 of cycle 1 and for paclitaxel on day 1 of cycles 1 and 2. Pretreatment tumor samples from 17 melanoma patients were analyzed for BRAF mutations. RESULTS: Sorafenib was well tolerated at the doses evaluated. The most frequent severe adverse events were hematologic toxicities (grade 3 or 4 in 33 patients, 85%). Twenty-seven (69%) patients had sorafenib-related adverse events, the most frequent of which were dermatologic events (26 patients, 67%). Exposure to paclitaxel was not altered by intervening treatment with sorafenib. Treatment with sorafenib, paclitaxel, and carboplatin resulted in one complete response and nine partial responses, all among patients with melanoma. There was no correlation between BRAF mutational status and treatment responses in patients with melanoma. CONCLUSIONS: The recommended phase II doses are oral 400 mg twice daily sorafenib, carboplatin at an AUC6 dose, and 225 mg/m(2) paclitaxel. The tumor responses observed with this combined regimen in patients with melanoma warrant further investigation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bencenosulfonatos/administración & dosificación , Carboplatino/administración & dosificación , Paclitaxel/administración & dosificación , Piridinas/administración & dosificación , Administración Oral , Adulto , Anciano , Biopsia , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Sorafenib , Factores de Tiempo , Resultado del Tratamiento
5.
Cancer Res ; 63(17): 5198-202, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500344

RESUMEN

Activating mutations in the BRAF serine/threonine kinase are found in >70% of human melanomas, of which >90% are BRAF(V599E). We sought to investigate the role of the BRAF(V599E) allele in malignant melanoma. We here report that suppression of BRAF(V599E) expression by RNA interference in cultured human melanoma cells inhibits the mitogen-activated protein kinase cascade, causes growth arrest, and promotes apoptosis. Furthermore, knockdown of BRAF(V599E) expression completely abrogates the transformed phenotype as assessed by colony formation in soft agar. Similar targeting of BRAF(V599E) or wild-type BRAF in human fibrosarcoma cells that lack the BRAF(V599E) mutation does not recapitulate these effects. Moreover, these results are specific for BRAF, as targeted interference of CRAF in melanoma cells does not significantly alter their biological properties. Thus, when present, BRAF(V599E) appears to be essential for melanoma cell viability and transformation and, therefore, represents an attractive therapeutic target in the majority of melanomas that harbor the mutation.


Asunto(s)
Transformación Celular Neoplásica/genética , Melanoma/genética , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/genética , Interferencia de ARN , Adhesión Celular/genética , División Celular/genética , Transformación Celular Neoplásica/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patología , Proteínas Proto-Oncogénicas B-raf , Proteínas Proto-Oncogénicas c-raf/biosíntesis , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Transfección , Células Tumorales Cultivadas
6.
Biomed Res Int ; 2014: 817613, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25147816

RESUMEN

Extensive accumulation of the glycosaminoglycan hyaluronan is found in pancreatic cancer. The role of hyaluronan synthases 2 and 3 (HAS2, 3) was investigated in pancreatic cancer growth and the tumor microenvironment. Overexpression of HAS3 increased hyaluronan synthesis in BxPC-3 pancreatic cancer cells. In vivo, overexpression of HAS3 led to faster growing xenograft tumors with abundant extracellular hyaluronan accumulation. Treatment with pegylated human recombinant hyaluronidase (PEGPH20) removed extracellular hyaluronan and dramatically decreased the growth rate of BxPC-3 HAS3 tumors compared to parental tumors. PEGPH20 had a weaker effect on HAS2-overexpressing tumors which grew more slowly and contained both extracellular and intracellular hyaluronan. Accumulation of hyaluronan was associated with loss of plasma membrane E-cadherin and accumulation of cytoplasmic ß-catenin, suggesting disruption of adherens junctions. PEGPH20 decreased the amount of nuclear hypoxia-related proteins and induced translocation of E-cadherin and ß-catenin to the plasma membrane. Translocation of E-cadherin was also seen in tumors from a transgenic mouse model of pancreatic cancer and in a human non-small cell lung cancer sample from a patient treated with PEGPH20. In conclusion, hyaluronan accumulation by HAS3 favors pancreatic cancer growth, at least in part by decreasing epithelial cell adhesion, and PEGPH20 inhibits these changes and suppresses tumor growth.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Ácido Hialurónico/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Microambiente Tumoral/fisiología , Animales , Cadherinas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Humanos , Hialuronano Sintasas , Hialuronoglucosaminidasa/metabolismo , Ratones , beta Catenina/metabolismo
7.
J Exp Med ; 209(3): 437-44, 2012 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-22351932

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is a highly lethal disease that is refractory to medical intervention. Notch pathway antagonism has been shown to prevent pancreatic preneoplasia progression in mouse models, but potential benefits in the setting of an established PDA tumor have not been established. We demonstrate that the gamma secretase inhibitor MRK003 effectively inhibits intratumoral Notch signaling in the KPC mouse model of advanced PDA. Although MRK003 monotherapy fails to extend the lifespan of KPC mice, the combination of MRK003 with the chemotherapeutic gemcitabine prolongs survival. Combination treatment kills tumor endothelial cells and synergistically promotes widespread hypoxic necrosis. These results indicate that the paucivascular nature of PDA can be exploited as a therapeutic vulnerability, and the dual targeting of the tumor endothelium and neoplastic cells by gamma secretase inhibition constitutes a rationale for clinical translation.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Carcinoma Ductal Pancreático/tratamiento farmacológico , Óxidos S-Cíclicos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Tiadiazoles/farmacología , Animales , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Óxidos S-Cíclicos/administración & dosificación , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Sinergismo Farmacológico , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Hipoxia/inducido químicamente , Ratones , Ratones Mutantes , Necrosis , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacología , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Tiadiazoles/administración & dosificación , Investigación Biomédica Traslacional , Gemcitabina
8.
Cancer Chemother Pharmacol ; 68(5): 1243-53, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21431415

RESUMEN

PURPOSE: To develop a sensitive analytical method to quantify gemcitabine (2',2'-difluorodeoxycytidine, dFdC) and its metabolites 2',2'-difluorodeoxyuridine (dFdU) and 2',2'-difluorodeoxycytidine-5'-triphosphate (dFdCTP) simultaneously from tumour tissue. METHODS: Pancreatic ductal adenocarcinoma tumour tissue from genetically engineered mouse models of pancreatic cancer (KP ( FL/FL ) C and KP ( R172H/+) C) was collected after dosing the mice with gemcitabine. (19)F NMR spectroscopy and LC-MS/MS protocols were optimised to detect gemcitabine and its metabolites in homogenates of the tumour tissue. RESULTS: A (19)F NMR protocol was developed, which was capable of distinguishing the three analytes in tumour homogenates. However, it required at least 100 mg of the tissue in question and a long acquisition time per sample, making it impractical for use in large PK/PD studies or clinical trials. The LC-MS/MS protocol was developed using porous graphitic carbon to separate the analytes, enabling simultaneous detection of all three analytes from as little as 10 mg of tissue, with a sensitivity for dFdCTP of 0.2 ng/mg tissue. Multiple pieces of tissue from single tumours were analysed, showing little intra-tumour variation in the concentrations of dFdC or dFdU (both intra- and extra-cellular). Intra-tumoural variation was observed in the concentration of dFdCTP, an intra-cellular metabolite, which may reflect regions of different cellularity within a tumour. CONCLUSION: We have developed a sensitive LC-MS/MS method capable of quantifying gemcitabine, dFdU and dFdCTP in pancreatic tumour tissue. The requirement for only 10 mg of tissue enables this protocol to be used to analyse multiple areas from a single tumour and to spare tissue for additional pharmacodynamic assays.


Asunto(s)
Antineoplásicos/metabolismo , Citidina Trifosfato/análogos & derivados , Desoxicitidina/análogos & derivados , Floxuridina/análogos & derivados , Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas en Tándem/métodos , Animales , Calibración , Citidina Trifosfato/metabolismo , Desoxicitidina/metabolismo , Modelos Animales de Enfermedad , Floxuridina/metabolismo , Flúor , Humanos , Ratones , Estándares de Referencia , Gemcitabina
9.
Science ; 324(5933): 1457-61, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19460966

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is among the most lethal human cancers in part because it is insensitive to many chemotherapeutic drugs. Studying a mouse model of PDA that is refractory to the clinically used drug gemcitabine, we found that the tumors in this model were poorly perfused and poorly vascularized, properties that are shared with human PDA. We tested whether the delivery and efficacy of gemcitabine in the mice could be improved by coadministration of IPI-926, a drug that depletes tumor-associated stromal tissue by inhibition of the Hedgehog cellular signaling pathway. The combination therapy produced a transient increase in intratumoral vascular density and intratumoral concentration of gemcitabine, leading to transient stabilization of disease. Thus, inefficient drug delivery may be an important contributor to chemoresistance in pancreatic cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Proteínas Hedgehog/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Alcaloides de Veratrum/administración & dosificación , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/metabolismo , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Trasplante de Neoplasias , Neovascularización Patológica , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Smoothened , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Alcaloides de Veratrum/farmacocinética , Alcaloides de Veratrum/uso terapéutico , Proteína con Dedos de Zinc GLI1 , Gemcitabina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA