Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Lancet ; 393(10189): 2404-2415, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31079938

RESUMEN

BACKGROUND: A phase 2 trial showed improved progression-free survival for atezolizumab plus bevacizumab versus sunitinib in patients with metastatic renal cell carcinoma who express programmed death-ligand 1 (PD-L1). Here, we report results of IMmotion151, a phase 3 trial comparing atezolizumab plus bevacizumab versus sunitinib in first-line metastatic renal cell carcinoma. METHODS: In this multicentre, open-label, phase 3, randomised controlled trial, patients with a component of clear cell or sarcomatoid histology and who were previously untreated, were recruited from 152 academic medical centres and community oncology practices in 21 countries, mainly in Europe, North America, and the Asia-Pacific region, and were randomly assigned 1:1 to either atezolizumab 1200 mg plus bevacizumab 15 mg/kg intravenously once every 3 weeks or sunitinib 50 mg orally once daily for 4 weeks on, 2 weeks off. A permuted-block randomisation (block size of 4) was applied to obtain a balanced assignment to each treatment group with respect to the stratification factors. Study investigators and participants were not masked to treatment allocation. Patients, investigators, independent radiology committee members, and the sponsor were masked to PD-L1 expression status. Co-primary endpoints were investigator-assessed progression-free survival in the PD-L1 positive population and overall survival in the intention-to-treat (ITT) population. This trial is registered with ClinicalTrials.gov, number NCT02420821. FINDINGS: Of 915 patients enrolled between May 20, 2015, and Oct 12, 2016, 454 were randomly assigned to the atezolizumab plus bevacizumab group and 461 to the sunitinib group. 362 (40%) of 915 patients had PD-L1 positive disease. Median follow-up was 15 months at the primary progression-free survival analysis and 24 months at the overall survival interim analysis. In the PD-L1 positive population, the median progression-free survival was 11·2 months in the atezolizumab plus bevacizumab group versus 7·7 months in the sunitinib group (hazard ratio [HR] 0·74 [95% CI 0·57-0·96]; p=0·0217). In the ITT population, median overall survival had an HR of 0·93 (0·76-1·14) and the results did not cross the significance boundary at the interim analysis. 182 (40%) of 451 patients in the atezolizumab plus bevacizumab group and 240 (54%) of 446 patients in the sunitinib group had treatment-related grade 3-4 adverse events: 24 (5%) in the atezolizumab plus bevacizumab group and 37 (8%) in the sunitinib group had treatment-related all-grade adverse events, which led to treatment-regimen discontinuation. INTERPRETATION: Atezolizumab plus bevacizumab prolonged progression-free survival versus sunitinib in patients with metastatic renal cell carcinoma and showed a favourable safety profile. Longer-term follow-up is necessary to establish whether a survival benefit will emerge. These study results support atezolizumab plus bevacizumab as a first-line treatment option for selected patients with advanced renal cell carcinoma. FUNDING: F Hoffmann-La Roche Ltd and Genentech Inc.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Bevacizumab/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Sunitinib/uso terapéutico , Anciano , Anticuerpos Monoclonales Humanizados , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/secundario , Supervivencia sin Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Tasa de Supervivencia , Resultado del Tratamiento
2.
J Biol Chem ; 284(31): 21057-65, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19494112

RESUMEN

Krüppel-like factor 6 (Klf6) belongs to a family of zinc finger transcription factors known to play a role in development and tumor suppression. Although Klf6 is highly mutated in prostate cancer, its function in prostate development is unknown. We have generated a prostate-specific Klf6-deficient mouse model and report here a novel role for Klf6 in the regulation of prostate branching morphogenesis. Importantly, our study reveals a novel relationship between Klf6 and the Shh pathway. Klf6-deficiency leads to elevated levels of hedgehog pathway components (Shh, Ptc, and Gli) and loss of their localized expression, which in turn causes impaired lateral branching.


Asunto(s)
Silenciador del Gen , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Morfogénesis , Próstata/crecimiento & desarrollo , Próstata/metabolismo , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Animales , Proteína Morfogenética Ósea 4/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Factor 6 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Músculo Liso/crecimiento & desarrollo , Especificidad de Órganos , Próstata/anomalías , Próstata/patología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Regulación hacia Arriba/genética , beta-Galactosidasa/metabolismo
3.
Nature ; 429(6987): 86-92, 2004 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15103385

RESUMEN

COP1 (constitutively photomorphogenic 1) is a RING-finger-containing protein that functions to repress plant photomorphogenesis, the light-mediated programme of plant development. Mutants of COP1 are constitutively photomorphogenic, and this has been attributed to their inability to negatively regulate the proteins LAF1 (ref. 1) and HY5 (ref. 2). The role of COP1 in mammalian cells is less well characterized. Here we identify the tumour-suppressor protein p53 as a COP1-interacting protein. COP1 increases p53 turnover by targeting it for degradation by the proteasome in a ubiquitin-dependent fashion, independently of MDM2 or Pirh2, which are known to interact with and negatively regulate p53. Moreover, COP1 serves as an E3 ubiquitin ligase for p53 in vitro and in vivo, and inhibits p53-dependent transcription and apoptosis. Depletion of COP1 by short interfering RNA (siRNA) stabilizes p53 and arrests cells in the G1 phase of the cell cycle. Furthermore, we identify COP1 as a p53-inducible gene, and show that the depletion of COP1 and MDM2 by siRNA cooperatively sensitizes U2-OS cells to ionizing-radiation-induced cell death. Overall, these results indicate that COP1 is a critical negative regulator of p53 and represents a new pathway for maintaining p53 at low levels in unstressed cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Apoptosis , Secuencia de Bases , Proteínas Portadoras/genética , Línea Celular , Línea Celular Tumoral , Cisteína Endopeptidasas/metabolismo , Fase G1 , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Complejos Multienzimáticos/metabolismo , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/química , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Nature ; 428(6984): 754-8, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15085134

RESUMEN

Vascular development is a complex but orderly process that is tightly regulated. A number of secreted factors produced by surrounding cells regulate endothelial cell (EC) differentiation, proliferation, migration and coalescence into cord-like structures. Vascular cords then undergo tubulogenesis to form vessels with a central lumen. But little is known about how tubulogenesis is regulated in vivo. Here we report the identification and characterization of a new EC-derived secreted factor, EGF-like domain 7 (Egfl7). Egfl7 is expressed at high levels in the vasculature associated with tissue proliferation, and is downregulated in most of the mature vessels in normal adult tissues. Loss of Egfl7 function in zebrafish embryos specifically blocks vascular tubulogenesis. We uncover a dynamic process during which gradual separation and proper spatial arrangement of the angioblasts allow subsequent assembly of vascular tubes. This process fails to take place in Egfl7 knockdown embryos, leading to the failure of vascular tube formation. Our study defines a regulator that controls a specific and important step in vasculogenesis.


Asunto(s)
Vasos Sanguíneos/embriología , Embrión de Mamíferos/irrigación sanguínea , Células Endoteliales/metabolismo , Proteínas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Vasos Sanguíneos/citología , Proteínas de Unión al Calcio , Adhesión Celular , Recuento de Células , Familia de Proteínas EGF , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/citología , Embrión no Mamífero/anomalías , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/citología , Células Endoteliales/citología , Hibridación in Situ , Ratones , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Pez Cebra/anomalías , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Mol Cell Biol ; 25(16): 7054-68, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16055717

RESUMEN

The Drosophila Fused (Fu) kinase is an integral component of the Hedgehog (Hh) pathway that helps promote Hh-dependent gene transcription. Vertebrate homologues of Fu function in the Hh pathway in vitro, suggesting that Fu is evolutionarily conserved. We have generated fused (stk36) knockout mice to address the in vivo function of the mouse Fu (mFu) homologue. fused knockouts develop normally, being born in Mendelian ratios, but fail to thrive within 2 weeks, displaying profound growth retardation with communicating hydrocephalus and early mortality. The fused gene is expressed highly in ependymal cells and the choroid plexus, tissues involved in the production and circulation of cerebral spinal fluid (CSF), suggesting that loss of mFu disrupts CSF homeostasis. Similarly, fused is highly expressed in the nasal epithelium, where fused knockouts display bilateral suppurative rhinitis. No obvious defects were observed in the development of organs where Hh signaling is required (limbs, face, bones, etc.). Specification of neuronal cell fates by Hh in the neural tube was normal in fused knockouts, and induction of Hh target genes in numerous tissues is not affected by the loss of mFu. Furthermore, stimulation of fused knockout cerebellar granule cells to proliferate with Sonic Hh revealed no defect in Hh signal transmission. These results show that the mFu homologue is not required for Hh signaling during embryonic development but is required for proper postnatal development, possibly by regulating the CSF homeostasis or ciliary function.


Asunto(s)
Líquido Cefalorraquídeo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hidrocefalia/etiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Animales , Proteína Axina , Linaje de la Célula , Proliferación Celular , Relación Dosis-Respuesta a Droga , Genes Reporteros , Genotipo , Heterocigoto , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hibridación in Situ , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Modelos Genéticos , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rinitis/genética , Transducción de Señal , Factores de Tiempo , Distribución Tisular , Transcripción Genética , beta-Galactosidasa/metabolismo
6.
Mol Cell Biol ; 24(4): 1608-13, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14749376

RESUMEN

EDA-A1 and EDA-A2 are members of the tumor necrosis factor family of ligands. The products of alternative splicing of the ectodysplasin (EDA) gene, EDA-A1 and EDA-A2 differ by an insertion of two amino acids and bind to distinct receptors. The longer isoform, EDA-A1, binds to EDAR and plays an important role in sweat gland, hair, and tooth development; mutations in EDA, EDAR, or the downstream adaptor EDARADD cause hypohidrotic ectodermal dysplasia. EDA-A2 engages the receptor XEDAR, but its role in the whole organism is less clear. We have generated XEDAR-deficient mice by gene targeting and transgenic mice expressing secreted forms of EDA-A1 or EDA-A2 downstream of the skeletal muscle-specific myosin light-chain 2 or skin-specific keratin 5 promoter. Mice lacking XEDAR were indistinguishable from their wild-type littermates, but EDA-A2 transgenic mice exhibited multifocal myodegeneration. This phenotype was not observed in the absence of XEDAR. Skeletal muscle in EDA-A1 transgenic mice was unaffected, but their sebaceous glands were hypertrophied and hyperplastic, consistent with a role for EDA-A1 in the development of these structures. These data indicate that XEDAR-transduced signals are dispensable for development of ectoderm-derived organs but might play a role in skeletal muscle homeostasis.


Asunto(s)
Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Receptores de Superficie Celular/deficiencia , Animales , Células Cultivadas , Ectodisplasinas , Receptor Edar , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de la Ectodisplasina , Receptores del Factor de Necrosis Tumoral , Glándulas Sebáceas/metabolismo , Glándulas Sebáceas/patología , Transducción de Señal , Receptor Xedar
7.
Clin Cancer Res ; 12(9): 2676-88, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16675559

RESUMEN

Activated fibroblasts are thought to play important roles in the progression of many solid tumors, but little is known about the mechanisms responsible for the recruitment of fibroblasts in tumors. Using several methods, we identified platelet-derived growth factor A (PDGFA) as the major fibroblast chemoattractant and mitogen from conditioned medium generated by the Calu-6 lung carcinoma cell line. In addition, we showed that Calu-6 tumors express significant levels of PDGFC, and that the levels of expression of these two PDGFRalpha ligands correlate strongly with the degree of stromal fibroblast infiltration into the tumor mass. The most intense expression of PDGFRalpha was observed in fibroblasts in the tumor outer rim. We subsequently showed that disrupting PDGFRalpha-mediated signaling results in significant inhibition of tumor growth in vivo. Furthermore, analysis of a compendium of microarray data revealed significant expression of PDGFA, PDGFC, and PDGFRalpha in human lung tumors. We propose that therapies targeting this stromal cell type may be effective in treating certain types of solid tumors.


Asunto(s)
Neoplasias Pulmonares/fisiopatología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Células del Estroma/patología , Células 3T3 , Animales , Anticuerpos , División Celular , Línea Celular Tumoral , Medios de Cultivo Condicionados , Cartilla de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Pulmón/citología , Pulmón/fisiología , Neoplasias Pulmonares/patología , Linfocinas/genética , Ratones , Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/aislamiento & purificación , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
8.
Cancer Res ; 65(21): 9751-61, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16266996

RESUMEN

To identify genes that could serve as targets for novel cancer therapeutics, we used a bioinformatic analysis of microarray data comparing gene expression between normal and tumor-derived primary human tissues. From this approach, we have found that maternal embryonic leucine zipper kinase (Melk), a member of the AMP serine/threonine kinase family, exhibits multiple features consistent with the potential utility of this gene as an anticancer target. An oligonucleotide microarray analysis of multiple human tumor samples and cell lines suggests that Melk expression is frequently elevated in cancer relative to normal tissues, a pattern confirmed by quantitative reverse transcription-PCR and Western blotting of selected primary tumor samples. In situ hybridization localized Melk expression to malignant epithelial cells in 96%, 23%, and 13% of colorectal, lung, and ovarian tissue tumor samples, respectively. Expression of this gene is also elevated in spontaneous tumors derived from the ApcMin and Apc1638N murine models of intestinal tumorigenesis. To begin addressing whether Melk is relevant for tumorigenesis, RNA interference-mediated silencing within human and murine tumor cell lines was done. We show that Melk knockdown decreases proliferation and anchorage-independent growth in vitro as well as tumor growth in a xenograft model. Together, these results suggest that Melk may provide a growth advantage for neoplastic cells and, therefore, inactivation may be therapeutically beneficial.


Asunto(s)
Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/biosíntesis , Células 3T3 , Secuencia de Aminoácidos , Animales , Biología Computacional , Células HeLa , Humanos , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/terapia , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Methods Mol Biol ; 326: 255-64, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16780208

RESUMEN

Tissue microarrays enable the rapid histological localization of gene expression in hundreds of archival samples by in situ hybridization. However, the scoring of tissue microarray data may be influenced by intra- and inter-observer variations, and categorizing continuous variables risks discarding potentially meaningful information. Quantitation imposes a greater degree of objectivity, is more reproducible than subjective discriminations, and facilitates the communication and clarity of definitions. Phosphorimaging has been successfully used to quantitate the hybridization signal intensity from arrayed tissues. The process is rapid and has a wide dynamic range, surpassing the densitometric analysis of autoradiograms. This paper presents a detailed method for quantitative isotopic in situ hybridization on formalin-fixed paraffin-embedded tissue microarrays. In addition, the method includes a protocol for the development of synthetic agarose cores to control for the specificity and sensitivity of hybridization.


Asunto(s)
Hibridación in Situ/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Fijadores , Formaldehído , Técnicas Histológicas , Humanos , Radioisótopos de Fósforo , Sensibilidad y Especificidad , Adhesión del Tejido , Fijación del Tejido
10.
Clin Cancer Res ; 11(14): 5181-7, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16033834

RESUMEN

A receptor tyrosine kinase for ephrin ligands, EphB2 is expressed in colorectal cancer and has been proposed as a target for immunoconjugate therapy. The aim of this study was to perform a detailed histologic analysis of EphB2 expression in normal and neoplastic colorectal tissues. In addition, we sought to evaluate EphB2 expression as a prognostic factor in colorectal cancer. Expression of EphB2 was examined in normal colon (n = 28), colorectal cell lines (n = 20), colorectal adenomas (n = 148), primary cancers (n = 28), and metastases (n = 39) using immunohistochemistry. In addition, a series of primary cancers and matched normal (n = 342) with outcome data were profiled in tissue microarrays. The intensity of EphB2 expression was assessed in the entire series by immunohistochemistry, and in a subset by in situ hybridization. Overall survival and recurrence-free survival were correlated with EphB2 protein expression in retrospective subset analyses. Epithelial EphB2 expression was shown at all stages of colorectal tumorigenesis, including the base of all normal crypts, 77% of adenomas, 82% of primary cancers, and 64% of metastases. Although homogeneous expression was observed in adenomas, the pattern of staining was focal (mean 25%) in most malignant lesions. Patients whose tumor stained 2+ for EphB2 expression (versus 0/1+) exhibited significantly prolonged overall survival: mean duration of survival, 2,514 versus 1,044 days; hazard ratio, 0.45; 95% confidence interval, 0.18 to 0.95 (P = 0.035). In summary, EphB2 is expressed in normal crypts, colorectal adenomas, primary cancers, and metastases. High levels of EphB2 expression are associated with a longer mean duration of survival in colorectal cancer.


Asunto(s)
Adenoma/genética , Adenoma/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptor EphB2/biosíntesis , Receptor EphB2/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Células Tumorales Cultivadas
11.
Cancer Res ; 64(20): 7226-30, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15492238

RESUMEN

The tumor suppressor protein p53 plays a central role in protecting normal cells from undergoing transformation. Thus, it is fitting that cancer cells selectively dampen the p53 response to gain a selective growth advantage. In fact, the p53 gene is the most commonly mutated tumor suppressor gene in human cancers, and if the gene is not mutated, then other components of the p53 pathways are skewed to dampen the p53 response to stress. We recently identified COP1 as a novel and critical negative regulator of p53. COP1 is a RING finger-containing protein that targets p53 for degradation to the proteasome and is necessary for p53 turnover in normal and cancer cells. However, the association between COP1 and cancer remains to be determined. We performed expression analysis of COP1 in ovarian and breast cancer tissue microarrays. COP1 is significantly overexpressed in 81% (25 of 32) of breast and 44% (76 of 171) of ovarian adenocarcinoma as assessed by in situ hybridization and immunohistochemistry. Overexpression of COP1 correlated with a striking decrease in steady state p53 protein levels and attenuation of the downstream target gene, p21, in cancers that retain a wild-type p53 gene status. Overall, these results suggest that overexpression of COP1 contributes to the accelerated degradation of p53 protein in cancers and attenuates the tumor suppressor function of p53.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias Ováricas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
12.
Cancer Res ; 62(9): 2546-53, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11980648

RESUMEN

We conducted an expression analysis of prostate stem cell antigen (PSCA)in normal urogenital tissues, benign prostatic hyperplasia (n = 21), prostatic intraepithelial neoplasia (n = 33), and primary (n = 137) and metastatic (n = 42) prostate adenocarcinoma, using isotopic in situ hybridization on tissue microarrays. In normal prostate, we observe PSCA expression in the terminally differentiated, secretory epithelium; strong expression was also seen in normal urothelium. Forty-eight percent of primary and 64% of metastatic prostatic adenocarcinomas expressed PSCA RNA. Our studies did not confirm a positive correlation between level of PSCA RNA expression and high Gleason grade. We characterized monoclonal anti-PSCA antibodies that recognize PSCA expressed on the surface of live cells, are efficiently internalized after antigen recognition, and kill tumor cells in vitro in an antigen-specific fashion upon conjugation with maytansinoid. Unconjugated anti-PSCA antibodies demonstrated efficacy against PSCA-positive tumors by delaying progressive tumor growth in vivo. Maytansinoid-conjugated antibodies caused complete regression of established tumors in a large proportion of animals. Our results strongly suggest that maytansinoid-conjugated anti-PSCA monoclonal antibodies should be evaluated as a therapeutic modality for patients with advanced prostate cancer.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/terapia , Anticuerpos Monoclonales/farmacología , Glicoproteínas de Membrana/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Adenocarcinoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Antígenos de Neoplasias , Femenino , Proteínas Ligadas a GPI , Humanos , Inmunización Pasiva/métodos , Inmunotoxinas/farmacocinética , Inmunotoxinas/farmacología , Hibridación in Situ , Masculino , Maitansina/farmacocinética , Maitansina/farmacología , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/metabolismo
13.
Endocrinology ; 144(6): 2606-16, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12746324

RESUMEN

We recently described human endocrine gland-derived vascular endothelial growth factor (EG-VEGF) as an endothelial cell mitogen with a novel selective activity and an expression pattern essentially limited to steroidogenic glands. Herein we present the identification and characterization of the mouse ortholog. The mouse cDNA and predicted amino acid sequences are, respectively, 86% and 88% identical with the human. Surprisingly, the mouse EG-VEGF transcript is predominantly expressed in liver and kidney. A comparison of human and mouse EG-VEGF promoter sequences revealed a potential binding site for NR5A1, which is known to be a pivotal element for steroidogenic-specific transcription, in the human but not mouse promoter. In situ hybridization studies localized expression of mouse EG-VEGF mRNA to hepatocytes and renal tubule cells. Interestingly, capillary endothelial cells in these sites share several common structural features with those found in steroidogenic glands. Within liver and kidney, EG-VEGF receptor expression was largely restricted to endothelial cells. Mouse EG-VEGF promoted proliferation and survival of endothelial cells. We propose that mouse EG-VEGF, like human EG-VEGF, plays a role in regulating the phenotype and growth properties of endothelial cells within distinct capillary beds.


Asunto(s)
Inductores de la Angiogénesis/genética , Endotelio Vascular/fisiología , Hormonas Gastrointestinales/genética , Neovascularización Fisiológica/fisiología , Neuropéptidos , Factor A de Crecimiento Endotelial Vascular , Animales , Supervivencia Celular/fisiología , Mapeo Cromosómico , ADN Complementario , Glándulas Endocrinas/fisiología , Endotelio Vascular/química , Endotelio Vascular/citología , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Riñón/irrigación sanguínea , Hígado/irrigación sanguínea , Ratones , Mitógenos/genética , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , Receptores de Factores de Crecimiento Endotelial Vascular/análisis , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina
14.
J Clin Endocrinol Metab ; 89(8): 4078-88, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15292351

RESUMEN

Angiogenesis is essential for tumor growth and metastasis. A new human angiogenic mitogen, endocrine gland-derived vascular endothelial growth factor (EG-VEGF), has been recently identified; its expression pattern is restricted to endocrine glands, with the highest expression in testis. We used in situ hybridization and newly generated monoclonal antibodies to investigate the expression of EG-VEGF in normal human prenatal and adult testis and in 48 human testicular tumors of different subtypes. We found that EG-VEGF was expressed from 14 wk until birth in human fetal testis. In the adult testis, EG-VEGF was strongly expressed only in Leydig cells. In testicular tumors, EG-VEGF was expressed specifically in Leydig cell tumors, whereas germ cell-derived neoplasms, including carcinoma in situ, seminoma, and nonseminomatous germ cell tumors, were negative for this antigen. In contrast, VEGF, another powerful angiogenic factor, was expressed in seminoma, but very weakly in Leydig cell tumors. Interestingly, we found that Leydig cell tumors presented vessel surface density 3.2-fold higher than seminoma. These findings argue that human EG-VEGF may play a role in angiogenesis both during the early endocrine development of testis and in the adult testis as well as in Leydig cell tumor growth.


Asunto(s)
Tumor de Células de Leydig/metabolismo , Neoplasias Testiculares/metabolismo , Testículo/embriología , Testículo/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Adulto , Feto/metabolismo , Humanos , Masculino , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Testículo/irrigación sanguínea
15.
Cancer Res ; 69(6): 2358-64, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19258515

RESUMEN

Antibody-drug conjugates (ADC), potent cytotoxic drugs covalently linked to antibodies via chemical linkers, provide a means to increase the effectiveness of chemotherapy by targeting the drug to neoplastic cells while reducing side effects. Here, we systematically examine the potential targets and linker-drug combinations that could provide an optimal ADC for the treatment for non-Hodgkin's lymphoma. We identified seven antigens (CD19, CD20, CD21, CD22, CD72, CD79b, and CD180) for potential treatment of non-Hodgkin's lymphoma with ADCs. ADCs with cleavable linkers mediated in vivo efficacy via all these targets; ADCs with uncleavable linkers were only effective when targeted to CD22 and CD79b. In target-independent safety studies in rats, the uncleavable linker ADCs showed reduced toxicity, presumably due to the reduced release of free drug or other toxic metabolites into the circulation. Thus, our data suggest that ADCs with cleavable linkers work on a broad range of targets, and for specific targets, ADCs with uncleavable linkers provide a promising opportunity to improve the therapeutic window for ADCs in humans.


Asunto(s)
Antineoplásicos/administración & dosificación , Inmunotoxinas/farmacología , Linfoma no Hodgkin/tratamiento farmacológico , Maitansina/análogos & derivados , Oligopéptidos/administración & dosificación , Compuestos de Sulfhidrilo/administración & dosificación , Animales , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Antineoplásicos/farmacocinética , Linfocitos B/inmunología , Reactivos de Enlaces Cruzados/administración & dosificación , Reactivos de Enlaces Cruzados/farmacocinética , Femenino , Inmunotoxinas/inmunología , Inmunotoxinas/farmacocinética , Linfoma no Hodgkin/inmunología , Maitansina/administración & dosificación , Maitansina/farmacocinética , Ratones , Ratones Endogámicos ICR , Ratones SCID , Oligopéptidos/farmacocinética , Ratas , Compuestos de Sulfhidrilo/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 2(6): e575, 2007 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-17593974

RESUMEN

BACKGROUND: Heparan sulfate proteoglycans (HSPGs) use highly sulfated polysaccharide side-chains to interact with several key growth factors and morphogens, thereby regulating their accessibility and biological activity. Various sulfotransferases and sulfatases with differing specificities control the pattern of HSPG sulfation, which is functionally critical. Among these enzymes in the mouse are two secreted 6-O-endosulfatases, Sulf1 and Sulf2, which modify HSPGs in the extracellular matrix and on the cell surface. The roles of Sulf1 and Sulf2 during normal development are not well understood. METHODS/RESULTS: To investigate the importance of Sulf1 and Sulf2 for embryonic development, we generated mice genetically deficient in these genes and assessed the phenotypes of the resulting secreted sulfatase-deficient mice. Surprisingly, despite the established crucial role of HSPG interactions during development, neither Sulf1- nor Sulf2-deficient mice showed significant developmental flaws. In contrast, mice deficient in both Sulf1and Sulf2 exhibited highly penetrant neonatal lethality. Loss of viability was associated with multiple, although subtle, developmental defects, including skeletal and renal abnormalities. CONCLUSIONS: These results show that Sulf1 and Sulf2 play overlapping yet critical roles in mouse development and are redundant and essential for neonatal survival.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Genes Letales/fisiología , Proteoglicanos de Heparán Sulfato/metabolismo , Sulfatasas/fisiología , Sulfotransferasas/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Hibridación in Situ , Riñón/anomalías , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/anomalías , Embarazo , Transducción de Señal , Tasa de Supervivencia
17.
J Clin Oncol ; 24(2): 217-27, 2006 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-16365183

RESUMEN

PURPOSE: Bevacizumab is a monoclonal antibody to vascular endothelial growth factor-A (VEGF). In the pivotal trial in metastatic colorectal cancer (mCRC), addition of bevacizumab to first-line irinotecan, fluorouracil, and leucovorin (IFL) significantly prolonged median survival. The aim of these retrospective subset analyses was to evaluate VEGF, thrombospondin-2 (THBS-2), and microvessel density (MVD) as prognostic factors and/or predictors of benefit from bevacizumab. PATIENTS AND METHODS: In the pivotal trial, 813 patients with untreated mCRC were randomly assigned to receive IFL plus bevacizumab or placebo. Of 312 tissue samples collected (285 primaries, 27 metastases), outcome data were available for 278 (153 bevacizumab, 125 placebo). Epithelial and stromal VEGF expression were assessed by in situ hybridization (ISH) and immunohistochemistry on tissue microarrays and whole sections. Stromal THBS-2 expression was examined by ISH on tissue microarrays. MVD was quantified by Chalkley count. Overall survival was associated with these variables in retrospective subset analyses. RESULTS: In all subgroups, estimated hazard ratios (HRs) for risk of death were < 1 for bevacizumab-treated patients regardless of the level of VEGF or THBS-2 expression or MVD. Patients with a high THBS-2 score showed a nonsignificant improvement in survival following bevacizumab treatment (HR = 0.11; 95% CI, 0.02 to 0.51) compared to patients with a low score (HR = 0.65; 95% CI, 0.41 to 1.02); interaction analysis P = .22. VEGF or THBS-2 expression and MVD were not significant prognostic factors. CONCLUSION: These exploratory analyses suggest that in patients with mCRC addition of bevacizumab to IFL improves survival regardless of the level of VEGF or THBS-2 expression, or MVD.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Trombospondinas/análisis , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bevacizumab , Neoplasias Colorrectales/mortalidad , Femenino , Humanos , Inmunohistoquímica , Masculino , Microcirculación/efectos de los fármacos , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , ARN Mensajero/análisis , Trombospondinas/genética , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/genética
18.
J Immunol ; 176(4): 2069-73, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16455961

RESUMEN

Chemokines play an important role in the immune system by regulating cell trafficking in homeostasis and inflammation. In this study, we report the identification and characterization of a novel cytokine-like protein, DMC (dendritic cell and monocyte chemokine-like protein), which attracts dendritic cells and monocytes. The key to the identification of this putative new chemokine was the application of threading techniques to its uncharacterized sequence. Based on our studies, DMC is predicted to have an IL-8-like chemokine fold and to be structurally and functionally related to CXCL8 and CXCL14. Consistent with our predictions, DMC induces migration of monocytes and immature dendritic cells. Expression studies show that DMC is constitutively expressed in lung, suggesting a potential role for DMC in recruiting monocytes and dendritic cells from blood into lung parenchyma.


Asunto(s)
Movimiento Celular , Quimiocinas/química , Quimiocinas/metabolismo , Células Dendríticas/citología , Monocitos/citología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Quimiocinas/inmunología , Quimiocinas CXC , Células Dendríticas/química , Células Dendríticas/inmunología , Regulación de la Expresión Génica , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Monocitos/química , Monocitos/inmunología , Especificidad de Órganos , Pliegue de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
19.
J Pathol ; 206(4): 466-75, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15971170

RESUMEN

Vascular endothelial growth factor-A (VEGF) is an important regulator of vascular permeability. In preclinical studies, VEGF induces endothelial fenestrations in pre-existing and neo-vasculature, while inhibition of VEGF leads to a reduction in endothelial fenestrations. Recently, vascular regression in response to VEGF inhibition has been shown to correlate with the presence of endothelial fenestrations. Plasmalemmal vesicle-associated protein (PLVAP) is believed to be a component of diaphragmed endothelial fenestrations, but a direct relationship with VEGF signalling has not been established. The aim of this study was to characterize the expression pattern of PLVAP and investigate whether PLVAP is a transcriptional target of VEGF signal transduction. The expression pattern of PLVAP was characterized in normal and neoplastic human tissues by in situ hybridization and/or immunohistochemistry. The role of VEGF signal transduction in the regulation of PLVAP expression was investigated in vitro using receptor-selective engineered forms of VEGF, a neutralizing monoclonal antibody against VEGF, and inhibitors of downstream signalling pathways. PLVAP mRNA and protein were widely expressed in the endothelium of normal and neoplastic tissues. In cultured endothelial cells, VEGF signalling through receptor 2 stimulated expression of PLVAP total RNA and protein. This induction could be blocked with an anti-VEGF monoclonal antibody and by inhibitors of phosphatidylinositol 3-kinase (LY294002) or p38 mitogen-activated protein kinase (SB203580), but not by PD98059, a mitogen-activated protein/extracellular signal-regulated kinase 1 inhibitor. These data show that PLVAP is more widely expressed in the vasculature of normal tissues than previously thought and that it is expressed in the vasculature of most human tumours. We suggest that PLVAP is a downstream target of VEGF signalling. This work solidifies the association between VEGF and the appearance and maintenance of fenestrations by providing a potential mechanistic link.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Transducción de Señal/genética , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/genética , Línea Celular Tumoral , Células Cultivadas , Cromonas/farmacología , Células Endoteliales/fisiología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Imidazoles/farmacología , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Morfolinas/farmacología , Piridinas/farmacología , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transducción de Señal/efectos de los fármacos , Transcripción Genética/genética
20.
Proc Natl Acad Sci U S A ; 100(5): 2685-90, 2003 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-12604792

RESUMEN

We recently identified an angiogenic mitogen, endocrine-gland-derived vascular endothelial growth factor (EG-VEGF), with selective activity for endothelial cells of endocrine tissues. Here we describe the characterization of a highly related molecule, Bv8, also known as prokineticin-2. Human Bv8 shares 60% identity and 75% similarity with EG-VEGF. The human and mouse Bv8 genes share a common structure. Like EG-VEGF, Bv8 is able to induce proliferation, survival and migration of adrenal cortical capillary endothelial cells. Bv8 gene expression is induced by hypoxic stress. Bv8 expression occurs predominantly in the testis and is largely restricted to primary spermatocytes. Adenoviral delivery of Bv8 or EG-VEGF to the mouse testis resulted in a potent angiogenic response. We have localized the expression of the Bv8EG-VEGF receptors within the testis to vascular endothelial cells. The testis exhibits relatively high turnover of endothelial cells. Therefore, Bv8 and EG-VEGF, along with other factors such as VEGF-A, may maintain the integrity and also regulate proliferation of the blood vessels in the testis.


Asunto(s)
Hormonas Gastrointestinales/metabolismo , Hormonas Gastrointestinales/fisiología , Neovascularización Patológica , Neuropéptidos , Receptores de Péptidos/biosíntesis , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bovinos , División Celular , Movimiento Celular , Células Cultivadas , Quimiotaxis , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Endotelial/metabolismo , Endotelio/metabolismo , Endotelio Vascular/citología , Humanos , Hipoxia , Immunoblotting , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Linfocinas/metabolismo , Masculino , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Isoformas de Proteínas , ARN/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Espermatozoides/citología , Testículo/irrigación sanguínea , Testículo/metabolismo , Testículo/patología , Distribución Tisular , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA