Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 7(8): 2563-72, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11489840

RESUMEN

We determined the optimal administration schedule of a novel epidermal growth factor receptor (EGFR) protein tyrosine kinase inhibitor (PKI), PKI 166 (4-(R)-phenethylamino-6-(hydroxyl)phenyl-7H-pyrrolo[2.3-d]-pyrimidine), alone or in combination with gemcitabine (administered i.p.) for therapy of L3.6pl human pancreatic carcinoma growing in the pancreas of nude mice. Seven days after orthotopic implantation of L3.6pl cells, the mice received daily oral doses of PKI 166. PKI 166 therapy significantly inhibited phosphorylation of the EGFR without affecting EGFR expression. EGFR phosphorylation was restored 72 h after cessation of therapy. Seven days after orthotopic injection of L3.6pl cells, groups of mice received daily or thrice weekly oral doses of PKI 166 alone or in combination with gemcitabine. Treatment with PKI 166 (daily), PKI 166 (3 times/week), or gemcitabine alone produced a 72%, 69%, or 70% reduction in the volume of pancreatic tumors in mice, respectively. Daily oral PKI 166 or thrice weekly oral PKI 166 in combination with injected gemcitabine produced 97% and 95% decreases in volume of pancreatic cancers and significant inhibition of lymph node and liver metastasis. Daily oral PKI 166 produced a 20% decrease in body weight, whereas treatment 3 times/week did not. Decreased microvessel density, decreased proliferating cell nuclear antigen staining, and increased tumor cell and endothelial cell apoptosis correlated with therapeutic success. Collectively, our results demonstrate that three weekly oral administrations of an EGFR tyrosine kinase inhibitor in combination with gemcitabine are sufficient to significantly inhibit primary and metastatic human pancreatic carcinoma.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Pirimidinas/farmacología , Pirroles/farmacología , Administración Oral , Animales , Antineoplásicos/farmacocinética , División Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Esquema de Medicación , Quimioterapia Combinada , Factores de Crecimiento Endotelial/análisis , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Humanos , Inmunohistoquímica , Interleucina-8/análisis , Linfocinas/análisis , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , Pirimidinas/farmacocinética , Pirroles/farmacocinética , Ribonucleótido Reductasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
3.
Cancer Res ; 60(11): 2926-35, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10850439

RESUMEN

We determined whether down-regulation of the epidermal growth factor-receptor (EGF-R) signaling pathway by oral administration of a novel EGF-R tyrosine kinase inhibitor (PKI166) alone or in combination with gemcitabine (administered i.p.) can inhibit growth and metastasis of human pancreatic carcinoma cells implanted into the pancreas of nude mice. Therapy beginning 7 days after orthotopic injection of L3.6pl human pancreatic cancer cells reduced the volume of pancreatic tumors by 59% in mice treated with gemcitabine only, by 45% in those treated with PKI166 only, and by 85% in those given both drugs. The combination therapy also significantly inhibited lymph node and liver metastasis, which led to a significant increase in overall survival. EGF-R activation was significantly blocked by therapy with PKI166 and was associated with significant reduction in tumor cell production of VEGF and IL-8, which in turn correlated with a significant decrease in microvessel density and an increase in apoptotic endothelial cells. Collectively, our results demonstrate that oral administration of an EGF-R tyrosine kinase inhibitor decreased growth and metastasis of human pancreatic cancer growing orthotopically in nude mice and increased survival. The therapeutic effects were mediated in part by inhibition of tumor-induced angiogenesis attributable to a decrease in production of proangiogenic molecules by tumor cells and increased apoptosis of tumor-associated endothelial cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Administración Oral , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Western Blotting , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Regulación hacia Abajo , Endotelio/patología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Gemcitabina
4.
Clin Cancer Res ; 6(5): 1936-48, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10815919

RESUMEN

Both epidermal growth factor receptor (EGF-R) signaling mechanisms and angiogenesis have been evaluated as independent targets for therapy of human pancreatic carcinoma, but a link between the two processes has been identified only recently. This study evaluated whether EGF-R blockade therapy with anti-EGF-R antibody C225 inhibits pancreatic carcinoma growth and metastasis in an orthotopic nude mouse model via tumor-mediated angiogenesis and whether gemcitabine potentiates this effect. In vitro treatment of human pancreatic carcinoma L3.6pl cells with C225 inhibited EGF-R autophosphorylation, producing a maximum of 20% cytostasis. Treatment with C225 plus gemcitabine resulted in additive cytotoxic effects that increased with increasing gemcitabine concentrations. Dose-dependent decreases in expression of the angiogenic factors vascular endothelial growth factor and interleukin 8 (but not basic fibroblast growth factor) were observed in the C225-treated cells (mRNA and protein levels). In L3.6pl tumors established in the pancreas of nude mice, systemic therapy with C225 alone and C225 in combination with gemcitabine resulted in growth inhibition, tumor regression, and abrogation of metastasis; median tumor volume was reduced from 538 to 0.3 and to 0 mm3, respectively. Gemcitabine treatment alone reduced median tumor volume from 538 to 152 mm3. Liver metastases were present in 50% of the controls, 30% of the gemcitabine-treated animals, and 20% of C225-treated animals. No macroscopically visible liver metastases were observed in the combination treatment group. As early as 11 days after C225 treatment, the median percentage of proliferating cell nuclear antigen-positive cells was substantially reduced compared with gemcitabine treatment alone (26% versus 73%, respectively) versus controls (92%), correlating with in vivo blockade of EGF-R activation. Similarly after 11 days treatment, production of vascular endothelial growth factor and interleukin 8 was significantly lower in C225 and C225 plus gemcitabine-treated tumors versus gemcitabine-treated and control tumors. Significant differences in microvessel density were observed 18 days after C225 or combination treatments (but not gemcitabine alone) in direct correlation with the difference in percentage of apoptotic endothelial cells, as visualized by double immunofluorescence microscopy. These experiments indicate that therapeutic strategies targeting EGF-R have a significant antitumor effect on human L3.6pl pancreatic carcinoma growing in nude mice which is mediated in part by inhibition of tumor-induced angiogenesis, leading to tumor cell apoptosis and regression. Furthermore, this effect is potentiated in combination with gemcitabine.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neovascularización Patológica/prevención & control , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Cetuximab , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Interleucina-8/metabolismo , Linfocinas/metabolismo , Linfocinas/farmacología , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/prevención & control , Trasplante de Neoplasias , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/metabolismo , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Gemcitabina
5.
J Histochem Cytochem ; 46(5): 595-602, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9562568

RESUMEN

Increased epidermal growth factor receptor (EGF-R) gene expression and functional protein levels correlate with the metastatic potential of human colon carcinoma (HCC) cells in nude mice. The purpose of this study was to determine whether the production of liver metastases by HCC cells depends on the EGF-R activation status and whether different organ microenvironments influence this activation. Using two independent monoclonal antibodies specific for the activated (i.e., tyrosine-phosphorylated) EGF-R, increased immunoreactivity was observed in HCC cells growing as metastatic lesions in the livers of athymic nude mice. Staining was observed throughout these lesions, both peripherally and centrally. In contrast, little or no immunoreactivity for activated EGF-R was observed in primary tumors growing orthotopically in the cecum or ectopically in the subcutis of nude mice. Immunohistochemistry for total EGF-R levels (irrelevant of activation status) demonstrated similar levels of immunoreactivity in HCC tumors growing in the cecum, subcutis, or liver of nude mice, indicating that total EGF-R levels are not altered after growth in these different microenvironments. Controls included immunohistochemistry for total and activated EGF-R levels in HCC cells growing in vitro under serum-free or EGF-stimulated conditions and A431-epidermoid carcinoma growing in nude mice. Western blot analyses confirmed the specificity of the antibodies for the activated EGF-R. These results suggest that the production of liver metastasis by HCC cells depends in part on the response of tumor cells to organ-derived growth factors and hence the activation of specific cell surface tyrosine kinase receptors.


Asunto(s)
Neoplasias del Colon/patología , Receptores ErbB/metabolismo , Neoplasias Hepáticas/secundario , Animales , Anticuerpos Monoclonales , Carcinoma de Células Escamosas/patología , Ciego , Neoplasias del Colon/metabolismo , Receptores ErbB/inmunología , Humanos , Técnicas para Inmunoenzimas , Hígado , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Piel , Células Tumorales Cultivadas
6.
Int J Oncol ; 12(2): 329-36, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9458358

RESUMEN

Epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I) receptors are implicated in the development and progression of several malignancies including osteogenic and soft tissue sarcomas (STS). To determine a role for ligand-mediated receptor activation in sarcoma progression, the relative expression and function of EGF-R, IGF-I-R, and several other molecular determinants implicated in the progression of mesenchymal neoplasms were evaluated in human sarcoma cells established from surgical specimens of primary and metastatic tumors. mRNA blot analyses demonstrated the expression of c-Met, p53, and MDM2-specific transcripts. Western blot analyses confirmed the production of high levels of p53 protein; however, minimal levels of MDM2 and c-Met proteins were detected. Analysis of STS cells #23, #26, and #50 originating from an unclassified sarcoma lung metastasis, a malignant fibrous histiocytoma lung metastasis, and a dedifferentiated chondrosarcoma, respectively demonstrated high steady-state levels of EGF-R and IGF-I-R mRNA transcripts and protein correlating with receptor-specific tyrosine kinase activity and autophosphorylation in response to ligand. Treatment of these STS cells with EGF resulted in a >5 fold increase in DNA synthesis and mitogenesis compared with untreated controls. In contrast, treatment with IGF-I showed a variable STS growth response correlating with the origin of the tumor. These data support the involvement of EGF-R and IGF-I-R in the growth and metastasis of human soft tissue sarcoma and may offer new targets for therapeutic intervention in the management of this disease.


Asunto(s)
Receptores ErbB/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas Nucleares , Sarcoma/metabolismo , Western Blotting , ADN de Neoplasias/biosíntesis , Receptores ErbB/fisiología , Expresión Génica , Humanos , Técnicas In Vitro , Factor I del Crecimiento Similar a la Insulina/fisiología , Metástasis de la Neoplasia/patología , Osteogénesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras , Sarcoma/genética , Sarcoma/patología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
7.
Clin Exp Metastasis ; 15(2): 140-50, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9062390

RESUMEN

The purpose of this study was to determine the subpopulation dynamics of human colon carcinoma (HCC) cells growing at orthotopic (cecum, liver) or ectopic (subcutis, kidney, spleen) sites in nude mice and to correlate any outgrowth of distinct clones with the differential expression of metastasis-related genes. Low metastatic KM12C HCC cells were genetically tagged with a retrovirus harboring the neomycin-resistance (Neo(R)) gene. Southern blot analyses demonstrated only minor resolution of the Neo(R) hybridization pattern in DNA isolated from primary tumors growing orthotopically or ectopically, suggesting a polyclonal outgrowth. In contrast, a major resolution of the Neo(R) hybridization pattern was observed in liver-specific metastases, demonstrating the outgrowth of single dominant clones. Expression of epidermal growth factor receptor (EGR-R) increased 20-60% in the liver metastases vs spleen tumors and the KM12C Neo(R) cells. Transforming growth factor alpha (TGF-alpha), amphiregulin (AR), and c-met showed only modest differences in mRNA expression. A 20-80% increase in type IV collagenase mRNA levels was also observed in all tumor specimens. Furthermore, expression of the multi-drug resistance gene PGY-1 and the carcinoembryonic antigen (CEA) gene were elevated in the liver metastases compared with the spleen tumors and cultured cells. Transcript levels of the angiogenic factors interleukin-8 and basic fibroblast growth factor did not correlate with clonal outgrowth. These data demonstrate a correlation between EGF-R, type IV collagenase, CEA, and PGY-1 gene expression and the production of liver metastases. Our results suggest that distinct HCC clones differentially expressing specific mRNA transcripts for metastasis-related genes are the forerunners of the experimental liver metastatic lesions.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Hepáticas/secundario , Animales , Southern Blotting , Antígeno Carcinoembrionario/metabolismo , Neoplasias del Colon/metabolismo , Resistencia a Múltiples Medicamentos/genética , Receptores ErbB/metabolismo , Genes Reporteros , Vectores Genéticos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Desnudos , Neomicina , Proteínas de Neoplasias/metabolismo , Especificidad de Órganos , Proteínas Proto-Oncogénicas c-met , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Retroviridae
8.
Pathobiology ; 65(6): 293-300, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9491849

RESUMEN

Abnormalities in at least 1 of 5 mismatch repair (MMR) genes (hMSH2, hMLH1, hPMS1, hPMS2 and GTBP/hMSH6) are found in hereditary nonpolyposis colon cancer and sporadic colon cancers. We used a single-reaction multiplex reverse transcription (RT)-polymerase chain reaction (PCR), with the beta-actin gene as an internal control, to simultaneously evaluate expression of these 5 known human MMR genes in normal and tumor cell lines with known or uncharacterized mutations in MMR genes. The relative quantitation of the transcripts is demonstrated by controlling the number of PCR cycles and titrating cDNA with a dose-curve. The 13 normal cell lines tested were derived from normal lymphocytes, skin, thymus, breast, lung, colon, liver and kidney. The 26 cancer cell lines were derived from melanoma and cancers of the brain, breast, lung, colon, pancreas and prostate. All 5 MMR genes were ubiquitously expressed in all normal cell lines tested, suggesting their housekeeping roles. Aberrant MMR gene expression was only observed in the colon cancer cell lines. Two previously uncharacterized colon cancer cell lines did not express hMLH1. These data suggest that this nonradioactive multiplex RT-PCR assay for MMR gene expression may be useful for fast screening for genetic alterations that may affect gene expression and so may aid molecular analysis of MMR-related colon cancer.


Asunto(s)
Enzimas Reparadoras del ADN , Reparación del ADN/genética , Reacción en Cadena de la Polimerasa/métodos , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas/genética , Proteínas Portadoras , Línea Celular , Cartilla de ADN/genética , ADN Complementario/análisis , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Expresión Génica/genética , Genes/genética , Humanos , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteína 2 Homóloga a MutS , Mutación/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares , Proteínas Proto-Oncogénicas/genética , ARN/análisis , ARN/genética , Transcripción Genética/genética , Células Tumorales Cultivadas
9.
Oncol Rep ; 4(3): 475-9, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-21590080

RESUMEN

K1735 murine melanoma cells transfected with p53 cDNAs bearing specific point mutations are metastatic in nude mice, whereas the parent and control-transfected cells are nonmetastatic. To determine whether p53 gene mutations regulate genes associated with angiogenesis, growth, and metastasis, we examined expression of vascular endothelial growth factor I, IGF-I receptor, epidermal growth factor insulin-like growth factor I, IGF-I receptor, epidermal growth factor receptor, c-MET, and thrombospondin 1 in K1735 cells transfected with one of four different mutant p53 cDNAs. Northern blot analysis demonstrated differential upregulation of these genes in cells transfected with different mutant p53 cDNAs. Up-regulation of angiogenesis-, growth-, and metastasis-related genes by mutant p53 may contribute to metastasis formation.

10.
Cancer Res ; 55(1): 141-8, 1995 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-7805024

RESUMEN

Metastatic K-1735 murine melanoma cells are amelanotic in culture or in the subcutis of syngeneic mice. When injected into the internal carotid artery, these cells produce melanotic brain metastases. The production of melanin in tumor cells growing in the brain was directly correlated with induction of melanocyte-stimulating hormone receptor (MSH-R) steady-state mRNA transcripts. K-1735 cells isolated from brain lesions and implanted into the subcutis or grown in culture lose MSH-R transcripts and become amelanotic. In contrast to K-1735 cells, B16-BL6 melanoma cells constitutively produce melanin and express high levels of MSH-R mRNA regardless of the site of growth. Somatic cell hybrids between K-1735 and B16 cells produced melanin and expressed high levels of MSH-R mRNA transcripts, regardless of the site of growth, suggesting the dominance of the B16 phenotype. Treatment with alpha-MSH failed to upregulate MSH-R expression in cultured K-1735 cells or to maintain MSH-R expression in K-1735 cells isolated from brain metastases to be grown in culture. Responsiveness to alpha-MSH as determined by cell proliferation, melanin production, and intracellular accumulation of cyclic AMP directly correlated with MSH-R expression. These data demonstrate that a specific organ environment influences the phenotype of metastatic cells by regulation of specific genes that encode for cell surface receptors.


Asunto(s)
Neoplasias Encefálicas/secundario , Regulación Neoplásica de la Expresión Génica , Hormonas Estimuladoras de los Melanocitos/metabolismo , Melanoma/genética , Receptores de la Hormona Hipofisaria/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Femenino , Melaninas/biosíntesis , Ratones , Ratones Endogámicos , Monofenol Monooxigenasa/metabolismo , Trasplante de Neoplasias , ARN Mensajero/análisis , Receptores de la Hormona Hipofisaria/biosíntesis , Transcripción Genética , Células Tumorales Cultivadas , alfa-MSH/farmacología
11.
Int J Cancer ; 58(3): 380-4, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8050821

RESUMEN

We investigated the effects of human recombinant interferons (r-IFNs) on gelatinase production and invasion by human renal-cell carcinoma (HRCC). Incubation of KG-2 HRCC with human r-IFN-beta or -gamma (but not -alpha) suppressed transcription of the 72-kDa gelatinase gene and, hence, production of gelatinase activity. These inhibitory effects of interferons (IFNs) were independent of their antiproliferative effects. Treatment of KG-2 cell with r-IFN-beta or -gamma significantly inhibited cell invasion through reconstituted basement membrane toward chemoattractants produced by kidney fibroblasts. The inhibitory activity of r-IFNs was specific to the KG-2 cells since gelatinase activity by various fibroblasts was unaffected. These findings suggest that r-IFN-beta or IFN-gamma may be used to inhibit the invasive potential of HRCC.


Asunto(s)
Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Gelatinasas/biosíntesis , Interferón beta/farmacología , Interferón gamma/farmacología , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Carcinoma de Células Renales/tratamiento farmacológico , Colágeno/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Gelatinasas/antagonistas & inhibidores , Gelatinasas/genética , Humanos , Interferón Tipo I/farmacología , Interferón beta-1a , Interferon beta-1b , Riñón/citología , Riñón/efectos de los fármacos , Riñón/enzimología , Neoplasias Renales/tratamiento farmacológico , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes , Piel/citología , Piel/efectos de los fármacos , Piel/enzimología , Células Tumorales Cultivadas/efectos de los fármacos
12.
Oncogene ; 9(7): 1877-83, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8208533

RESUMEN

Human SAOS-2 osteogenic sarcoma cells are not metastatic in nude mice and do not express p53. We have selected a variant line (SAOS-LM2) that is tumorigenic and metastatic in nude mice. These cells were transfected with the p53 wild-type (p53wt) or mutated (p53mut 143A) gene, whose expression was verified by reverse transcriptase PCR, cDNA sequencing, and protein immunoprecipitation. Cells were injected i.v. into nude mice, and 4 months later, the mice were necropsied. All cell lines produced a similar number of visible lung metastases, albeit of different sizes. Microscopic examination revealed that most lung metastases in mice injected with p53wt cells (but not p53mut 143A or control cells) consisted of osteoid matrix and apoptotic cells. Expression of either p53wt or p53mut 143A verified the origin of the metastases. These data suggest that transfection of SAOS-LM2 cells with p53wt is associated with in vivo induction of terminal differentiation and apoptosis that inhibit progressive growth of metastases.


Asunto(s)
Apoptosis/genética , Neoplasias Óseas/patología , Genes p53 , Neoplasias Pulmonares/patología , Osteosarcoma/patología , Animales , Secuencia de Bases , Neoplasias Óseas/genética , Diferenciación Celular , Cartilla de ADN , Humanos , Hibridación in Situ , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Osteosarcoma/genética , Osteosarcoma/secundario , Transfección , Células Tumorales Cultivadas
13.
J Natl Cancer Inst ; 86(12): 913-20, 1994 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-7910854

RESUMEN

BACKGROUND: The major cause of death from cancer is metastases that are resistant to conventional therapies. The resistance of metastatic tumor cells to chemotherapy can be caused by their intrinsic properties, such as increased expression of the mdr genes. PURPOSE: The purpose of our present study was to determine some of the mechanisms by which the organ microenvironment influences the response of tumor cells to chemotherapy. METHODS: Murine CT-26 colon cancer cells growing in continuous culture (parental cells) were harvested and injected subcutaneously into the lateral flank (to produce subcutaneous tumors) or the lateral tail vein (to produce experimental lung metastases) of 10 8-week-old syngeneic male BALB/c mice. Seven days after tumor-cell injection, the mice were given intravenous injections of either doxorubicin (10 mg/kg) or 0.9% NaCl (controls). This in vivo injection was repeated 7 days later. Mice with subcutaneous tumors and lung metastases were killed by cervical dislocation on day 21, and tumor samples from control mice were harvested and adapted to culture. The sensitivity of the cultured cells to doxorubicin and fluorouracil (5-FU) was determined at multiple time points. Levels of mdr-1 DNA were measured by slot-blot and Southern-blot analyses. mdr mRNA expression levels were measured by Northern-blot analysis using mdr-1- and mdr-3-specific hybridization probes, and P-glycoprotein level was determined by fluorescence-activated cell sorting using different monoclonal antibodies. RESULTS: Treatment with doxorubicin produced 80% growth inhibition of CT-26 subcutaneous tumors but had little effect on the number (and size) of experimental lung metastases. Collectively, the results suggest that the multidrug-resistant phenotype developed in CT-26 cells growing in the lung environment. Cultures established from lung metastases were initially resistant to doxorubicin (but not to 5-FU) and showed elevated expression of mdr-1 mRNA transcripts and P-glycoprotein. This resistance could be overcome by verapamil and disappeared after 21 days in culture. No mdr gene amplification was detected. The expression level of mdr-specific mRNA (predominance of mdr-1) and P-glycoprotein was directly associated with resistance to doxorubicin. CONCLUSIONS: Results of this study have demonstrated that the in vivo sensitivity of murine CT-26 colon carcinoma cells to doxorubicin depends on the organ environment. The organ environment can influence the P-glycoprotein-mediated multidrug-resistant phenotype in tumor cells, and the increased expression of P-glycoprotein is transient; once removed from the environment (lung), the cell's resistance reverts to that of the sensitive parent cells.


Asunto(s)
Carcinoma/tratamiento farmacológico , Proteínas Portadoras/genética , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Resistencia a Medicamentos , Glicoproteínas de Membrana/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Animales , Carcinoma/genética , Doxorrubicina/administración & dosificación , Expresión Génica , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , ARN Mensajero/genética , ARN Neoplásico/genética , Neoplasias Cutáneas/tratamiento farmacológico , Verapamilo/administración & dosificación
14.
Int J Oncol ; 3(3): 413-22, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21573380

RESUMEN

The purpose of the study was to determine whether the organ environment can influence the response of colon cancer cells to chemotherapy. The highly metastatic human colon cancer cell line KM12L4, previously selected for production of liver metastases in nude mice, was injected into the cecal wall and into the spleen to produce liver metastases, and into the subcutis of nude mice. Doxorubicin (DOX) at 10 mg/kg or saline (control) was injected intravenously on days 7 and 16 after tumor cell injection. The in vivo response of tumors growing in the cecum, liver, and subcutaneous (s.c.) sites as well as the DOX sensitivity of cell lines established from liver and s.c. tumors were compared. Colon cancers growing s.c. were more sensitive to DOX than tumors growing in the cecal wall or liver of nude mice. The difference in response to DOX between s.c. tumors (sensitive) and liver tumors (resistant) was not due to selection of cell populations with different sensitivity to DOX, or differences in DOX distribution. PKC activity was lower in tumors of the liver and the cecum than in s.c. tumors. The expression of P-glycoprotein as determined by flow cytometric analysis of tumor cells harvested from lesions in different organs correlated inversely with their sensitivity to DOX. Increased levels of P-glycoprotein correlated with mdr-1, mdr-3 mRNA expression as determined by Northern analysis. Collectively, the data show that the organ environment influences the response of human colon carcinoma cells to DOX and recommend that animal models of this disease for experimental therapeutic studies employ orthotopic implantation of tumor cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA