Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 28(5): 1005-1014, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28453696

RESUMEN

Background: Recent whole-genome sequencing identified four molecular subtypes of gastric cancer (GC), of which the subgroup of Epstein-Barr virus-associated GC (EBVaGC) showed a significant enrichment of PIK3CA mutations. We here aimed to validate independently the enrichment of PIK3CA mutations in EBVaGC of a Central European GC cohort, to correlate EBV status with clinico-pathological patient characteristics and to test for a major issue of GC, intratumoral heterogeneity. Patients and methods: In a first step, 484 GCs were screened for EBV and PIK3CA hot spot mutations of exon 9/20 using EBER in situ hybridization and pyrosequencing, respectively. Secondly, an extended sequencing of PIK3CA also utilizing next generation sequencing was carried out in all EBVaGCs and 96 corresponding lymph node metastases. Results: Twenty-two GCs were EBER-positive, all being of latency type I. Intratumoral heterogeneity of EBER-positivity was found in 18% of EBVaGCs. Twenty-three GCs held PIK3CA mutations in hot spot regions of exon 9 or 20, being significantly more common in EBVaGCs (P < 0.001). Subsequent extended sequencing of PIK3CA of EBVaGCs showed that 14% harvested three to five different PIK3CA genotypes (including wildtype) in the same primary tumor, albeit in histologically and spatially distinct tumor areas, and that intratumoral heterogeneity of PIK3CA was also present in the corresponding lymph node metastases. Conclusions: Our findings unravel issues of tumor heterogeneity and illustrate that the assessment of the EBV status in tissue biopsies might carry the risk of sampling errors, which may significantly hamper adequate molecular tumor classification in a more clinical setting. Moreover, this is the first report of intratumoral heterogeneity of PIK3CA mutations in GC, and our findings lead to the conclusion that PIK3CA mutant and -wildtype tumor subclones are skilled to metastasize independently to different regional lymph nodes.


Asunto(s)
Adenocarcinoma/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Infecciones por Virus de Epstein-Barr/genética , Neoplasias Gástricas/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adenocarcinoma/virología , Anciano , Infecciones por Virus de Epstein-Barr/mortalidad , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Masculino , Técnicas de Diagnóstico Molecular , Mutación , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología
2.
Eur Biophys J ; 45(7): 749-763, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27652669

RESUMEN

The altered expression and/or activity of ion channels and transporters (transportome) have been associated with malignant behavior of cancer cells and were proposed to be a hallmark of cancer. However, the impact of altered transportome in epithelial cancers, such as pancreatic ductal adenocarcinoma (PDAC), as well as its pathophysiological consequences, still remains unclear. Here, we report the in silico analysis of 840 transportome genes in PDAC patients' tissues. Our study was focused on the transportome changes and their correlation with functional and behavioral responses in PDAC tumor and stromal compartments. The dysregulated gene expression datasets were filtered using a cut-off of fold-change values ≤-2 or ≥2 (adjusted p value ≤0.05). The dysregulated transportome genes were clearly associated with impaired physiological secretory mechanisms and/or pH regulation, control of cell volume, and cell polarity. Additionally, some down-regulated transportome genes were found to be closely linked to epithelial cell differentiation. Furthermore, the observed decrease in genes coding for calcium and chloride transport might be a mechanism for evasion of apoptosis. In conclusion, the current work provides a comprehensive overview of the altered transportome expression and its association with predicted PDAC malignancy with special focus on the epithelial compartment.


Asunto(s)
Adenocarcinoma/metabolismo , Biología Computacional , Simulación por Computador , Perfilación de la Expresión Génica , Canales Iónicos/genética , Canales Iónicos/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Epitelio/metabolismo , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Microambiente Tumoral
3.
Br J Cancer ; 109(3): 694-703, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23839495

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Improvements in the understanding of its molecular mechanism and the characterisation of CRC-specific biomarkers facilitating early detection are considered to increase overall survival. METHODS: A meta-analysis of microarray and Serial Analysis of Gene Expression (SAGE) has been performed to identify differentially regulated genes in CRC. Dipeptidase 1 (DPEP1/MDP/RDP) and Syntenin-2 (SDCBP2/SITAC18) were found to be differentially expressed in tumour tissue compared with normal mucosa. Expression of DPEP1 was assessed in a validation set of 87 normal mucosa samples, 20 hyperplastic polyps, 46 CR adenomas with low- and high-grade intraepithelial neoplasia (IEN) and 217 well-documented CRCs by immunohistochemistry and partially by immunoblotting and real-time PCR. RESULTS: Expression of DPEP1 was specifically increased in human CRC tissue samples compared with normal mucosa (P<0.0001, Mann-Whitney U-test), showing a striking upregulation in high-grade compared with low-grade IEN. Furthermore, high DPEP1 expression was found to strongly correlate with histological stage (P<0.0001, chi-square test) as well as localisation (P<0.0001, chi-square test) and has been recognised as an independent adverse prognostic factor, showing significant prognostic values with an ROC (receiver operating characteristic)-AUC of 0.9230. CONCLUSION: Dipeptidase 1 has been identified as an excellent marker of high-grade IEN and CRC, and may thus be applied for screening of early neoplastic lesions and for prognostic stratification.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma in Situ/enzimología , Carcinoma in Situ/patología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Dipeptidasas/biosíntesis , Biomarcadores de Tumor/genética , Carcinoma in Situ/genética , Neoplasias Colorrectales/genética , Dipeptidasas/genética , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Humanos , Clasificación del Tumor , Pronóstico , ARN Mensajero/biosíntesis , ARN Mensajero/genética
4.
J Cell Biol ; 152(5): 911-22, 2001 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-11238448

RESUMEN

Epimorphin was recently described as a mesenchymal factor modulating morphogenesis of murine mammary ducts, skin, liver, and lung in vitro. In this study epimorphin was analyzed in a human, pancreatic adenocarcinoma cell line (A818-6) which develops single layer epithelial hollow spheres resembling normal pancreatic ductal structures in vitro. Soluble 34- and 31-kD isoforms of epimorphin were found in the culture supernatant of A818-6 cells. In lysates of A818-6 cells we detected the 34-and 31-kD isoforms and the dimers, and in lysates of fibroblasts the 150-kD tetramers of epimorphin additionally. A neutralizing monoclonal antibody against epimorphin (MC-1) efficiently blocked the development of hollow sphere structures from A818-6 cells. Coculture of A818-6 cells with fibroblasts stimulated the development of hollow sphere structures in general and increased differentiation in 5-6-d-old hollow spheres. A818-6 hollow sphere development in the presence of fibroblasts was also blocked by MC-1. In this novel system for human duct-like differentiation of pancreatic epithelial cells, we provide evidence for an autocrine and paracrine function of epimorphin as a major mediator for morphogenesis.


Asunto(s)
Comunicación Autocrina , Glicoproteínas de Membrana/metabolismo , Conductos Pancreáticos/citología , Adenocarcinoma , Anciano , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Comunicación Autocrina/efectos de los fármacos , Biomarcadores/análisis , Adhesión Celular , Agregación Celular/efectos de los fármacos , Diferenciación Celular , División Celular/efectos de los fármacos , Línea Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Dimerización , Femenino , Fibroblastos , Técnica del Anticuerpo Fluorescente , Sustancias de Crecimiento/farmacología , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/inmunología , Morfogénesis/efectos de los fármacos , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/metabolismo , Conductos Pancreáticos/ultraestructura , Isoformas de Proteínas/química , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Solubilidad , Sintaxina 1 , Células Tumorales Cultivadas
5.
J Cell Biol ; 166(6): 839-51, 2004 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-15364960

RESUMEN

Mathematical modeling is required for understanding the complex behavior of large signal transduction networks. Previous attempts to model signal transduction pathways were often limited to small systems or based on qualitative data only. Here, we developed a mathematical modeling framework for understanding the complex signaling behavior of CD95(APO-1/Fas)-mediated apoptosis. Defects in the regulation of apoptosis result in serious diseases such as cancer, autoimmunity, and neurodegeneration. During the last decade many of the molecular mechanisms of apoptosis signaling have been examined and elucidated. A systemic understanding of apoptosis is, however, still missing. To address the complexity of apoptotic signaling we subdivided this system into subsystems of different information qualities. A new approach for sensitivity analysis within the mathematical model was key for the identification of critical system parameters and two essential system properties: modularity and robustness. Our model describes the regulation of apoptosis on a systems level and resolves the important question of a threshold mechanism for the regulation of apoptosis.


Asunto(s)
Apoptosis , Receptor fas/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Simulación por Computador , Bases de Datos Factuales , Regulación hacia Abajo , Activación Enzimática , Humanos , Cinética , Matemática , Modelos Biológicos , Modelos Teóricos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Transducción de Señal , Análisis de Sistemas
6.
Gut ; 57(6): 799-806, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18203806

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinomas (PDACs) are highly resistant to treatment due to changes in various signalling pathways. CK1 isoforms play important regulatory roles in these pathways. AIMS: We analysed the expression levels of CK1 delta and epsilon (CK1delta/in) in pancreatic tumour cells in order to validate the effects of CK1 inhibition by 3-[2,4,6-(trimethoxyphenyl)methylidenyl]-indolin-2-one (IC261) on their proliferation and sensitivity to anti-CD95 and gemcitabine. METHODS: CK1delta/in expression levels were investigated by using western blotting and immunohistochemistry. Cell death was analysed by FACS analysis. Gene expression was assessed by real-time PCR and western blotting. The putative anti-tumoral effects of IC261 were tested in vivo in a subcutaneous mouse xenotransplantation model for pancreatic cancer. RESULTS: We found that CK1delta/in are highly expressed in pancreatic tumour cell lines and in higher graded PDACs. Inhibition of CK1delta/in by IC261 reduced pancreatic tumour cell growth in vitro and in vivo. Moreover, IC261 decreased the expression levels of several anti-apoptotic proteins and sensitised cells to CD95-mediated apoptosis. However, IC261 did not enhance gemcitabine-mediated cell death either in vitro or in vivo. CONCLUSIONS: Targeting CK1 isoforms by IC261 influences both pancreatic tumour cell growth and apoptosis sensitivity in vitro and the growth of induced tumours in vivo, thus providing a promising new strategy for the treatment of pancreatic tumours.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Caseína Cinasa 1 épsilon/antagonistas & inhibidores , Quinasa Idelta de la Caseína/antagonistas & inhibidores , Indoles/farmacología , Neoplasias Pancreáticas/patología , Floroglucinol/análogos & derivados , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/secundario , Caseína Cinasa 1 épsilon/metabolismo , Caseína Cinasa 1 épsilon/fisiología , Quinasa Idelta de la Caseína/metabolismo , Quinasa Idelta de la Caseína/fisiología , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Evaluación Preclínica de Medicamentos , Humanos , Indoles/uso terapéutico , Metástasis Linfática , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Floroglucinol/farmacología , Floroglucinol/uso terapéutico , Trasplante Heterólogo , Células Tumorales Cultivadas , Receptor fas/fisiología , Gemcitabina
7.
Oncogene ; 26(33): 4850-62, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17297450

RESUMEN

In the present study, we have analysed the effects of transforming growth factor-beta (TGF-beta) signaling on the growth behavior of pancreatic carcinoma cells in vitro and on their tumorigenicity in vivo. Ectopic expression of dominant-negative mutants of the TGF-beta type II receptor or type I receptor/activin receptor-like kinase 5 (ALK5) in TGF-beta-sensitive pancreatic ductal adenocarcinoma PANC-1 cells prevented the TGF-beta-induced activation of transfected Smad-responsive reporter genes and growth arrest. The growth-inhibitory effect was mimicked by stable expression of kinase-active ALK5 (ALK5-T204D), and was dependent on ALK5's ability to activate Smad signaling, as a ALK5-derived mutant with an intact kinase domain but deficient in its ability to activate Smads (RImL45) failed to suppress proliferation in the absence of added TGF-beta. Moreover, this mutant often displayed opposite effects to those of ALK5-TD and blocked various ligand-induced responses in vitro, indicating that it acts in a dominant-negative fashion to inhibit endogenous wild-type receptors. ALK5-TD-, but not RImL45-TD-transduced cells underwent epithelial-to-mesenchymal transition, exhibited a higher ratio of thrombospondin-1 to vascular endothelial growth factor-A expression and upregulated various metastasis-associated genes. Upon orthotopic transplantation of PANC-1 clones into immunodeficient mice, ALK5-TD, but not RImL45-TD, greatly reduced tumor size and induced the formation of liver metastases in otherwise non-metastatic PANC-1 cells. These results suggest a causal, dominant role for the endogenous Smad2/3 signaling pathway in the tumor suppressor and prometastatic activities of TGF-beta in pancreatic tumor cells.


Asunto(s)
Receptores de Activinas Tipo I/fisiología , Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Ratones , Ratones SCID , Mutación , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Smad/metabolismo , Transfección , Factor de Crecimiento Transformador beta/farmacología , Carga Tumoral
8.
Oncogene ; 26(14): 2082-94, 2007 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-17043657

RESUMEN

Pancreatic ductal adenocarcinoma is a devastating disease, characterized by a rapid progression and poor treatment response. Using gene expression profiling of pancreatic cancer tissues, we previously identified periostin as a potential diagnostic and therapeutic target. In this study, we report the overexpression of periostin in a larger set of pancreatic cancer tissues and show that although the periostin transcript is exclusively expressed in tumour cells, the protein product is only detected in the extracellular matrix adjacent to cancer cells. Using an enzyme-linked immunosorbent assay (ELISA) assay, we show significantly increased levels of periostin in the sera of pancreatic cancer patients compared to non-cancer controls. We demonstrate that periostin promotes the invasiveness of tumour cells by increasing the motility of cells without inducing expression of proteases, and enhances the survival of tumour cells exposed to hypoxic conditions. At the molecular level, we provide evidence that the alpha(6)beta(4) integrin complex acts as the cell receptor of periostin in pancreatic cancer cells and that interaction promotes phosphorylation of focal adhesion kinase (FAK) and protein kinase B (AKT) though activation of the PI3 kinase pathway, but not the RAS/MEK/ERK pathway. These findings suggest an important role of periostin in pancreatic cancer and provide a rationale to study periostin for diagnostic and therapeutic applications.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Moléculas de Adhesión Celular/fisiología , Integrina beta4/metabolismo , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Apoptosis , Carcinoma Ductal Pancreático/química , Carcinoma Ductal Pancreático/metabolismo , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Hipoxia/metabolismo , Integrina alfa6beta4/metabolismo , Invasividad Neoplásica , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Transcripción Genética
9.
Br J Cancer ; 99(9): 1484-92, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18854834

RESUMEN

Kallikreins play an important role in tumour microenvironment and as cancer biomarkers in different cancer entities. Previous studies suggested an upregulation of KLK10 and KLK6 in pancreatic ductal adenocarcinoma (PDAC). Therefore, we evaluated the clinicopathological role of these kallikreins and their value as biomarkers in PDAC.Differential expression was validated by DNA-microarrays and immunohistochemistry in normal and malignant pancreatic tissues. Sera concentrations of both kallikreins were evaluated using ELISA. In silico analysis of possible protein interactions and gene silencing of KLK10 in vitro using siRNAs gave further insights in the pathomechanisms.Gene expression analysis and immunohistochemistry demonstrated a strong expression for KLK10 and KLK6 in PDAC. Statistical analysis showed that co-expression of these kallikreins correlated with an R1-resection status (P=0.017) and worse outcome for overall survival (P=0.031). Multivariate analysis proofed that co-expression is an independent prognostic factor for survival (P=0.043). Importantly, KLK10 knockdown in AsPC-1 cells significantly reduced cell migration, whereas computational analysis suggested interaction of KLK6 with angiogenetic factors as an important mechanism.Co-expression of KLK10 and KLK6 plays an unfavourable role in PDAC. Our results suggest that this effect is likely mediated by an interaction with the factors of the extracellular matrix and enhancement of cancer cell motility.


Asunto(s)
Adenocarcinoma/química , Carcinoma Ductal Pancreático/química , Calicreínas/análisis , Neoplasias Pancreáticas/química , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Movimiento Celular , Proteínas del Ojo/fisiología , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Inmunohistoquímica , Calicreínas/genética , Calicreínas/fisiología , Factores de Crecimiento Nervioso/fisiología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico , Serpinas/fisiología
10.
Oncogene ; 25(56): 7434-9, 2006 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-16751802

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted considerable attention for its potential use in tumor therapy, as some recombinant variants of this ligand induce apoptosis in tumor cells without harming most normal cells. Here, we show that TRAIL strongly induces the expression of the proinflammatory cytokines interleukin-8 and monocyte chemoattractant protein 1 and enhances the invasion of apoptosis-resistant pancreatic ductal adenocarcinoma cells in vitro by upregulation of the urokinase-type plasminogen activator expression. Most importantly, we also demonstrate for the first time that TRAIL treatment results in strongly increased distant metastasis of pancreatic tumors in vivo. We orthotopically transplanted human pancreatic ductal adenocarcinoma cells to the pancreata of severe combined immunodeficiency mice and observed a dramatic increase in metastatic spread including a sixfold increase in the volume and fourfold increase in the number of liver metastases upon TRAIL treatment. Our results point to the necessity to carefully evaluate in vivo side effects of TRAIL and to select therapy conditions that not only enhance apoptosis induction but in addition prevent proinvasive and proinflammatory non-apoptotic TRAIL signaling.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Humanos , Ratones , Ratones SCID
11.
Cancer Res ; 46(7): 3605-7, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3708591

RESUMEN

The monoclonal antibody CA 19-9 reacts with a carbohydrate epitope (sialylated lacto-N-fucopentaose II), which was shown to be part of a ganglioside extracted from a colon carcinoma cell line as well as of a mucin isolated from gastrointestinal tract tumor patients' sera. Recently, when we compared CA 19-9 levels in pancreatic juices and corresponding serum samples from a large group of patients, we showed the high serum values to be indicative solely for a malignant disease. In contrast, the overall high CA 19-9 content in pancreatic juices from all diagnostic groups raised the question about the antigenic moieties in these samples. By means of thin layer chromatography of glycolipids with subsequent antibody overlay, gel chromatography, and density gradient analysis, we found only the mucin form in all sources investigated. Thus we conclude that the discrimination potential of the CA 19-9 assay in serum is based on an altered secretion or distribution in pancreatic tumors.


Asunto(s)
Antígenos de Neoplasias/análisis , Mucinas/inmunología , Jugo Pancreático/inmunología , Neoplasias Pancreáticas/inmunología , Anticuerpos Monoclonales , Gangliósidos/análisis , Gangliósidos/inmunología , Humanos , Peso Molecular , Pancreatitis/inmunología
12.
Cancer Res ; 55(23 Suppl): 5749s-5755s, 1995 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7493340

RESUMEN

Monoclonal antibodies (mabs) have considerable potential for specific cancer treatment. However, due to the antigen heterogeneity and especially the low uptake in solid tumors, mabs have not been used successfully in most clinical trials to date. This study investigates the effects of a mab-cobra venom factor (CVF) conjugate in vitro and in vivo in an orthotopic pancreatic cancer model using nude rats. CVF, a nontoxic glycoprotein from cobra venom, permanently activates the alternative pathway of complement. Coupled to a mab with tumor-binding properties, the complement activation can be targeted to the tumor tissue. We studied the activity of a mab CA19-9-CVF conjugate with the human pancreatic cancer cells PancTu I. PancTu I cells express the complement resistance factors CD46, CD55, and CD59, as we demonstrated by immunostaining, an observation that may explain the lack of cytotoxicity of the CA19-9-CVF conjugate. However, using ELISA, Western blot, and immunostaining, we showed that CA19-9-CVF activates the complement cascade, including the release of the anaphylatoxin C3a, a mediator of an inflammatory reaction. The in vivo studies of CA19-9-CVF-treated nude rats showed an increased tumor infiltration by natural killer cells and macrophages. The tumor uptake of 99Tc-labeled anti-carcinoembryonic antigen antibody was increased approximately 2-fold in rats pretreated with 70 micrograms of CA19-9-CVF, compared to animals that received an equimolar mixture of noncoupled mab and CVF. This study indicates the value of mab-CVF conjugates in adjuvant immunotherapy. mab-CVF conjugates might be useful in pretargeting approaches by increasing the uptake of a therapeutic mab.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Venenos Elapídicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias Pancreáticas/terapia , Animales , Antígeno Carcinoembrionario/inmunología , Activación de Complemento , Humanos , Células Asesinas Naturales/inmunología , Neoplasias Pancreáticas/inmunología , Radioinmunodetección , Ratas , Ratas Desnudas , Tecnecio , Células Tumorales Cultivadas
13.
Cancer Res ; 58(8): 1741-9, 1998 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9563493

RESUMEN

The Fas system, comprising the Fas receptor (Fas, CD95, APO-1) and its ligand, Fas ligand (FasL), is a central mediator of programmed cell death in various physiological and pathological processes. Recent evidence indicated that tumor cells can exploit this system to their benefit in the dialogue with the host immune system. We have shown that all human pancreatic adenocarcinoma cell lines tested by fluorescence-activated cell sorting analysis (6 of 6) and immunocytochemistry (12 of 12) were positive for Fas expression, as were normal and malignant duct cells in pancreatic tissue sections. However, despite Fas expression, pancreatic tumor cells have become largely resistant toward recombinant FasL- or anti-APO-1 agonistic antibody-induced apoptosis. This resistance correlated with high levels in pancreatic tumor cells of mRNA for FAP-1, a Fas-associated phosphatase that can block the apoptotic function of Fas. Using a variety of methodological approaches, we also present evidence for the production of FasL by pancreatic tumor cells because 6 of 6 pancreatic tumor cell lines were found to contain FasL mRNA as well as the Mr 40,000 and Mr 26,000 forms of the FasL protein. Likewise, pancreatic tissue revealed FasL-specific immunostaining in pancreatic tumor cells but not in the surrounding stroma. In coculture experiments, pancreatic tumor cells displayed a cytotoxic effect toward the Fas-sensitive Jurkat T-cell line, which could be inhibited by a FasL-specific neutralizing antibody. Together, these results support the recently proposed "counterattack model" for local deletion of tumor-reactive T-cells by tumor cell-derived FasL.


Asunto(s)
Adenocarcinoma/metabolismo , Apoptosis , Glicoproteínas de Membrana/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor fas/metabolismo , Anticuerpos/farmacología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , Citoplasma/metabolismo , Proteína Ligando Fas , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Células Jurkat , Glicoproteínas de Membrana/farmacología , Reacción en Cadena de la Polimerasa , Proteína Tirosina Fosfatasa no Receptora Tipo 13 , Proteínas Tirosina Fosfatasas/metabolismo , ARN Mensajero/análisis , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas , Receptor fas/inmunología
14.
Cancer Res ; 56(8): 1823-7, 1996 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8620499

RESUMEN

Five somatostatin receptor subtypes (sst1 to sst5) have been cloned. We demonstrated previously that sst2 and sst5 mediate the antiproliferative effect of the somatostatin analogues octreotide and vapreotide. Using reverse transcription-PCR, we investigated gene expression of the five receptors in 47 human normal and cancerous tissues or cell lines from pancreatic and colorectal origin. mRNAs of somatostatin receptor subtypes were detected in 98% of samples, with more than two mRNA subtypes being expressed in 55% of cases. sst1, sst4, and sst5 were heterogeneously expressed in both normal and cancerous tissues; sst3 was rarely or not expressed. sst2 was present in normal pancreatic tissues but was absent in exocrine pancreatic carcinomas and their metastases. sst2 mRNAs were detected in normal colon, sporadic polyadenomas, and 50% of Dukes' stage B and 20% of Dukes' stage C carcinomas but were undetectable in Dukes' stage D carcinomas, hepatic metastases, and adenomas from familial adenomatous polyposis. The loss of sst2 expression could represent a growth advantage in these tumors and provide an explanation for the lack of therapeutic effect of somatostatin analogues in such adenocarcinomas. A subtyping of somatostatin receptors should be carried out before considering a somatostatin analogue treatment in patients with colorectal or pancreatic cancer.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Expresión Génica , Intestino Grueso/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Somatostatina/biosíntesis , Receptores de Somatostatina/genética , Línea Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Humanos , Estadificación de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Valores de Referencia , Células Tumorales Cultivadas
15.
Cancer Res ; 45(3): 1402-7, 1985 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3882225

RESUMEN

Three pancreatic cancer-associated antigens were characterized by use of monoclonal antibodies in immunobinding studies with various cellular and soluble target antigens, in immunoprecipitation, and in immunoperoxidase staining. C54-0 represents a tumor-associated Mr 122,000 antigen, which appears to be widely distributed on various epithelial tumors and to a lower extent on normal tissue. C1-N3 antigen exhibited a more restricted distribution, reacting with pancreatic and various gastrointestinal tract tumors as well as with chronically inflamed pancreatic tissue. The most specific antigen expression was observed for C1-P83 antigen, found on all exocrine tumors of the pancreas, but not on normal or chronically inflamed pancreatic tissue.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/análisis , Neoplasias Pancreáticas/inmunología , Animales , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos BALB C , Peso Molecular , Páncreas/inmunología , Neoplasias Pancreáticas/patología , Pancreatitis/inmunología
16.
Cancer Res ; 57(15): 3106-10, 1997 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9242433

RESUMEN

We investigated 141 bone marrow and 104 venous blood isolates from gastrointestinal cancer patients with a cytokeratin (CK) 20-specific nested reverse transcription PCR for the detection of disseminated tumor cells at time of primary tumor resection. In colorectal cancer patients, 20 of 65 (31%) bone marrow and 9 of 52 (17%) venous blood isolates yielded a CK 20 mRNA-positive result in a stage-dependent manner. The detection rates for gastric cancer patients were 11 of 49 (22%) and 5 of 30 (17%) for bone marrow and venous blood, respectively. In pancreatic cancer patients, positive signals were found in advanced tumor stage. A duplex PCR system improved the feasibility of the test. After analyzing 70 sets of bone marrow and venous blood isolates from colorectal, gastric, and pancreatic cancer patients, we observed a higher detection rate in bone marrow isolates. Survival of patients with CK 20 mRNA-positive findings was significantly shorter than that of negatively tested patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Médula Ósea/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células Neoplásicas Circulantes , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Humanos , Proteínas de Filamentos Intermediarios/análisis , Proteínas de Filamentos Intermediarios/genética , Queratina-20 , Reacción en Cadena de la Polimerasa/métodos , Pronóstico , ARN Mensajero/análisis , Tasa de Supervivencia
17.
Cancer Res ; 58(24): 5662-6, 1998 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9865718

RESUMEN

Mucins (MUCs) are high molecular weight membrane glycoproteins. The gene expression of MUCs (MUC1-MUC8) may change characteristically during malignant transformation of epithelial tissues. Total RNA was isolated from the four bladder cancer cell lines RT4, 647V, HT1376, and 486P (pathological gradings between G1 and G4) and 17 samples of transitional cell carcinomas, as well as 16 samples of normal human urothelium of the bladder from surgically removed specimens. The RNA samples were studied with MUC1-, MUC2- and MUC7-specific nested reverse transcription-PCRs. Gene expression of MUC1 and MUC2 was found positive in all normal, as well as in malignant, tissue samples and in the tumor cell lines. In contrast, gene expression of MUC7 was only detected in bladder cancer cell lines and samples of invasive transitional cell carcinomas, but neither in superficial, noninvasive bladder tumors nor normal bladder urothelium. Only one of the samples of normal urothelium obtained from 16 different tumor-bearing bladders was positive for MUC7 gene expression. These results suggest a differential MUC7 gene expression with the onset of malignant transformation of the bladder urothelium.


Asunto(s)
Mucina-1/metabolismo , Mucinas/genética , Mucinas/metabolismo , Proteínas y Péptidos Salivales/genética , Neoplasias de la Vejiga Urinaria/genética , Urotelio/metabolismo , Carcinoma de Células Transicionales/metabolismo , Transformación Celular Neoplásica , Expresión Génica , Humanos , Mucina 2 , ARN/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas y Péptidos Salivales/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/metabolismo
18.
Oncogene ; 16(9): 1183-5, 1998 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-9528860

RESUMEN

Soft tissue sarcomas are a heterogeneous group of neoplasms with various histological subtypes. Up to now, no individual causal molecular markers for prognosis and therapeutic success have been identified. A tumorigenic connection between the oncogene product Mdm2 and tumor suppressor p53 is generally accepted, but their possible clinical relevance has not yet been investigated sufficiently in soft tissue sarcoma. In 86 primary soft tissue sarcoma of the extremities (RO-resected, T1/2 N0 M0), Mdm2 and p53 overexpression were investigated by immunohistochemistry. The results were adjusted to clinico-pathological characteristics and evaluated for their prognostic relevance by multivariate analysis. In Cox's multivariate analysis with stratification of Mdm2 to p53 results, we determined four groups which had different prognostic values for relapse-free and overall survival (Mdm2-/p53- < Mdm2-/p53+ < Mdm2+/p53- < Mdm2+/p53+). The most striking finding was a relative risk (rr) for overall survival of 18.77 (P=0.006) for patients with Mdm2/p53 co-overexpression (n=40). It is noticeably higher than the additive risk from both factors. Coincident Mdm2/p53 overexpression is an independent molecular marker with the highest prognostic relevance described for soft tissue sarcoma. Thus, a high risk sarcoma group has been defined which we believe requires alternative therapeutic approaches.


Asunto(s)
Proteínas Nucleares , Proteínas Proto-Oncogénicas/biosíntesis , Sarcoma/genética , Sarcoma/patología , Proteína p53 Supresora de Tumor/biosíntesis , Adulto , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Análisis Multivariante , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/biosíntesis , Estadificación de Neoplasias , Pronóstico , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas c-mdm2 , Riesgo , Sarcoma/mortalidad , Sarcoma/cirugía , Tasa de Supervivencia , Factores de Tiempo , Proteína p53 Supresora de Tumor/análisis
19.
Oncogene ; 6(6): 1015-21, 1991 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1906154

RESUMEN

Human Platelet Derived Growth Factors (PDGF) are potent mitogens for mesenchymal cells and encoded by two related genes, the A- (or 1-) and B- (or 2-) chain. The latter is known as the human homolog (c-sis) of the v-sis oncogene. We investigated the expression and cytokine-mediated regulation of PDGF A- and B-chain mRNA in endoderm-derived cells, i.e. cultured human pancreatic adenocarcinoma cells. Northern blot analysis revealed that out of 14 cells lines 11 were positive for the A-chain and 10 for the B-chain. Tumor Necrosis Factor (TNF) -alpha and -beta, but not Interferon (IFN) -gamma, drastically upregulate the mRNA levels for PDGF B-chain and for the A-chain in a dose-dependent manner in nearly every pancreatic tumor cell line investigated (n = 6). With respect to the signal pathway stimulated by TNF, no evidence emerged for an activation of protein kinase A. The inhibition of protein kinase C by staurosporine (in the absence or presence of TNF) as well as its stimulation by PMA resulted in an increased mRNA level for the B-chain, indicating a functional role of PKC in this system. Furthermore, time course experiments and Cycloheximide treatment showed that the A- and B-chain mRNA are regulated by different mechanisms in transformed epithelial cells. Irrespective of these differences, the sum of their biological functions may contribute to the phenomenon of desmoplasia in pancreatic tumors by epithelial/mesenchymal interactions.


Asunto(s)
Adenocarcinoma/patología , Interferón gamma/farmacología , Neoplasias Pancreáticas/patología , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Adenocarcinoma/metabolismo , Alcaloides/farmacología , Northern Blotting , Línea Celular , Cicloheximida/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Pancreáticas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Proteínas Quinasas/fisiología , Proteínas Proto-Oncogénicas c-sis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estaurosporina , Factores de Tiempo
20.
Oncogene ; 20(7): 859-68, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11314019

RESUMEN

The transcription factor NF-kappaB has anti-apoptotic properties and may confer chemoresistance to cancer cells. Here, we describe human pancreatic carcinoma cell lines that differ in the responsiveness to the topoisomerase-2 inhibitors VP16 (20 microM) and doxorubicin (0.3 microM): Highly sensitive T3M4 [corrected] and PT45-P1 cells, and Capan-1 and A818-4 cells that were almost resistant to both anti cancer drugs. VP16, but not doxorubicin, transiently induced NF-kappaB activity in all cell lines, whereas basal NF-kappaB binding was nearly undetectable in T3M4 [corrected] and PT45-P1 cells, but rather high in Capan-1 and A818-4 cells, as demonstrated by gel-shift and luciferase assays. Treatment with various NF-kappaB inhibitors (Gliotoxin, MG132 and Sulfasalazine), or transfection with the IkappaBalpha super-repressor, strongly enhanced the apoptotic effects of VP16 or doxorubicin on resistant Capan-1 and 818-4 cells. Our results indicate that under certain conditions the resistance of pancreatic carcinoma cells to chemotherapy is due to their constitutive NF-kappaB activity rather than the transient induction of NF-kappaB by some anti-cancer drugs. Blockade of basal NF-kappaB activity by well established drugs efficiently reduces chemoresistance of pancreatic cancer cells and offers the potential for improved therapeutic strategies.


Asunto(s)
Carcinoma/tratamiento farmacológico , Doxorrubicina/farmacología , Etopósido/farmacología , Proteínas I-kappa B , FN-kappa B/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Antineoplásicos/farmacología , Proteínas de Unión al ADN/biosíntesis , Resistencia a Medicamentos , Gliotoxina/farmacología , Humanos , Leupeptinas/farmacología , Inhibidor NF-kappaB alfa , Sulfasalazina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA