Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Ann Oncol ; 28(9): 2278-2285, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911087

RESUMEN

BACKGROUND: The transforming growth factor (TGF)-ß pathway is a well-described inducer of immunosuppression and can act as an oncogenic factor in advanced tumors. Several preclinical and clinical studies show that the TGF-ß pathway can be considered a promising molecular target for cancer therapy. The human genome has three TGF-ß isoforms and not much is known about the oncogenic response to each of the isoforms. Here, we studied the antitumor response to ISTH0047, a recently developed locked nucleic acid-modified antisense oligonucleotide targeting TGF-ß2. MATERIALS AND METHODS: We have studied the anticancer response to ISTH0047 using gymnotic delivery in tumor cell cultures and in in vivo preclinical orthotopic mouse models for primary tumors (breast and kidney tumors) and lung metastasis. RESULTS: We observed that ISTH0047 is able to significantly reduce TGF-ß2 mRNA and protein levels without altering the levels of TGF-ß1 and TGF-ß3. ISTH0047 prevented lung metastasis in syngeneic orthotopic renal cell carcinoma (RENCA) and breast cancer (4T1) tumor models. In addition, using an orthotopic xenograft model of a lung cancer cell line (CRL5807) that mainly expresses TGF-ß2, we observed that ISTH0047 had an important effect on the lung microenvironment inhibiting the growth of lung lesions. ISTH0047 treatment re-educated macrophages in the lung parenchyma to express the tumor-suppressive factor, CD86. CONCLUSION: Overall, our data point to TGF-ß2 as a therapeutic target and ISTH0047 as a novel anticancer drug to prevent lung metastasis by impacting on the tumor niche, in part, through the induction of CD86 in tumor-associated macrophages.


Asunto(s)
Antígeno B7-2/inmunología , Neoplasias de la Mama/patología , Neoplasias Renales/patología , Neoplasias Pulmonares/secundario , Macrófagos/inmunología , Oligonucleótidos Antisentido/genética , Factor de Crecimiento Transformador beta2/genética , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Natl Cancer Inst ; 89(20): 1505-15, 1997 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9337347

RESUMEN

BACKGROUND: Growth factor receptor-signaling pathways are potentially important targets for anticancer therapy. The interaction of anticancer agents with specific molecular targets can be identified by correlating target expression patterns with cytotoxicity patterns. We sought to identify new agents that target and inhibit the activity of the epidermal growth factor (EGF) receptor and of c-erbB2 (also called HER2 or neu), by correlating EGF receptor, transforming growth factor (TGF)-alpha (a ligand for EGF receptor), and c-erbB2 messenger RNA (mRNA) expression levels with the results of cytotoxicity assays of the 49000 compounds in the National Cancer Institute (NCI) drug screen database. METHODS: The levels of mRNAs were measured and used to generate a molecular target database for the 60 cell lines of the NCI anticancer drug screen. The computer analysis program, COMPARE, was used to search for cytotoxicity patterns in the NCI drug screen database that were highly correlated with EGF receptor, TGF-alpha, or c-erbB2 mRNA expression patterns. The putative EGF receptor-inhibiting compounds were tested for effects on basal tyrosine phosphorylation, in vitro EGF receptor tyrosine kinase activity, and EGF-dependent growth. Putative ErbB2-inhibiting compounds were tested for effects on antibody-induced ErbB2 tyrosine kinase activity. RESULTS: EGF receptor mRNA and TGF-alpha mRNA levels were highest in cell lines derived from renal cancers, and c-erbB2 mRNA levels were highest in cells derived from breast, ovarian, and colon cancers. Twenty-five compounds with high correlation coefficients (for cytotoxicity and levels of the measured mRNAs) were tested as inhibitors of the EGF receptor or c-erbB2 signaling pathways; 14 compounds were identified as inhibitors of these pathways. The most potent compound, B4, inhibited autophosphorylation (which occurs following activation) of ErbB2 by 50% in whole cells at 7.7 microM. CONCLUSIONS: Novel EGF receptor or c-erbB2 pathway inhibitors can be identified in the NCI drug screen by correlation of cytotoxicity patterns with EGF receptor or c-erbB2 mRNA expression levels.


Asunto(s)
Antineoplásicos/toxicidad , Receptores ErbB/biosíntesis , Receptor ErbB-2/biosíntesis , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador alfa/biosíntesis , Neoplasias de la Mama , División Celular/efectos de los fármacos , Línea Celular , Análisis por Conglomerados , Neoplasias del Colon , Ensayos de Selección de Medicamentos Antitumorales , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Renales , Neoplasias Ováricas , ARN Mensajero/biosíntesis , Relación Estructura-Actividad , Células Tumorales Cultivadas
3.
Cancer Res ; 56(16): 3737-42, 1996 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8706017

RESUMEN

Brain-derived neurotrophic factor (BDNF) and its receptors are necessary for the survival and development of many neuronal cells. Because BDNF and TrkB are expressed in many poor-prognosis neuroblastoma (NB) tumors, we evaluated the role of BDNF in affecting sensitivity to chemotherapeutic agents. We investigated the effects of activation of the BDNF-TrkB signal transduction pathway in two NB cell lines, 15N and SY5Y. 15N cells lack the high-affinity receptor p145TrkB and express BDNF; 15N cells were used along with 15N-TrkB cells, a subline transfected with a TrkB expression vector. In cytotoxicity assays, 15N-TrkB cells were consistently 1.4-2 fold more resistant to vinblastine than 15N cells. Drug accumulation assays showed a 50% reduction in[3H]vinblastine accumulation in 15N-TrkB cells compared with control 15N cells. Addition of 30 ng/ml BDNF resulted in a reduction to 46% of control in 15N cells and a reduction to 28% of control in 15N-TrkB cells. SY5Y cells were chosen as a second model because they lack both endogenous BDNF and TrkB expression. p145TrkB expression is induced by 1 nM retinoic acid. Vinblastine accumulation was not significantly affected by 1 nM retinoic acid in SY5Y cells. Addition of 30 ng/ml BDNF decreased [3H]vinblastine accumulation to 58% of control in SY5Y cells and decreased [3H]vinblastine accumulation to 62% of control in TrkB-expressing SY5Y cells. Although an increase in BDNF expression in seen in multidrug-resistant sublines of SY5Y and BE(2)-C NB cells, the protective effect of BDNF in vinblastine toxicity may be unrelated to mdr-1, because the activity of other agents transported by P-glycoprotein was not affected. There was no increase in mdr-1 expression in 1 nM RA SY5Y cells and 15N-TrkB cells, as assessed by Northern blot analysis. In addition to the effects of BDNF on vinblastine cytotoxicity and accumulation, there was an inhibition in the ability of vinblastine to depolymerize tubulin in BDNF-treated cells. Thus, BDNF and TrkB may partially rescue NB cells from vinblastine toxicity and thereby may contribute to a more chemoresistant phenotype.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteínas del Tejido Nervioso/farmacología , Neuroblastoma/tratamiento farmacológico , Vinblastina/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Factor Neurotrófico Derivado del Encéfalo , Resistencia a Medicamentos , Humanos , Proteínas del Tejido Nervioso/genética , ARN Mensajero/análisis , Tretinoina/farmacología , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas , Vinblastina/farmacocinética
4.
Biochim Biophys Acta ; 1497(2): 215-26, 2000 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-10903426

RESUMEN

We examined transforming growth factor (TGF) alpha, epidermal growth factor (EGF) and EGF receptor (EGFR) expression and signaling in three drug resistant MCF-7 human breast cancer sublines and asked whether these pathways contribute to the drug resistance phenotype. In the resistant sublines, upregulation of both TGFalpha and EGFR mRNA was observed. In an apparent contrast with upregulated growth factor and receptor gene expression, the drug resistant sublines displayed a reduced growth rate. Defects in the EGFR signaling pathway cascade were found in all examined drug resistant sublines, including altered EGF-induced Shc, Raf-1, or mitogen-activated protein kinase phosphorylation. Induction of c-fos mRNA expression by EGF was impaired in the sublines compared to parental MCF-7 cells. In contrast, the induction of the stress-activated protein kinase activity was similar in both parental and drug resistant cells. Evaluating the link between the reduced growth rate and drug resistance, serum starvation experiments were performed. These studies demonstrated that a reduced proliferative activity resulted in a marked reduction in sensitivity to cytotoxic agents in the parental MCF-7 cells. We propose that the altered EGFR levels frequently observed in drug resistant breast cancer cells are associated with perturbations in the signaling pathway that mediate a reduced proliferative rate and thereby contribute to drug resistance.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Anisomicina , Antineoplásicos/farmacología , Neoplasias de la Mama , División Celular , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/aislamiento & purificación , Doxorrubicina/farmacología , Resistencia a Medicamentos , Receptores ErbB/efectos de los fármacos , Humanos , Immunoblotting , Proteína Quinasa 9 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Paclitaxel/farmacología , Fenotipo , Pruebas de Precipitina , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/análisis , Transducción de Señal , Células Tumorales Cultivadas
5.
Clin Cancer Res ; 3(12 Pt 1): 2405-14, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9815641

RESUMEN

It is increasingly recognized that drug-resistant cells undergo transitions not directly linked to "classical" drug resistance. We examined the expression of growth factors, growth factor receptors, and the estrogen receptor in 17 drug-resistant and 2 revertant human breast cancer sublines to provide an understanding of the phenotypic changes that occur and how these changes could affect the biology of the cell. These sublines were derived from five parental human breast cancer cell lines (MCF-7, ZR75B, T47D, MDA-MB-231, and MDA-MB-453). The expression of estrogen receptor was absent or decreased in 6 of the 15 resistant MCF-7, ZR75B, and T47D sublines. Increases of as much as 49-fold compared to parental levels were observed in transforming growth factor alpha, epidermal growth factor receptor, c-erbB2, and/or c-erbB3 mRNA expression in 14 of the 17 resistant sublines. Altered amphiregulin and insulin-like growth factor-I receptor expression was observed in nine and four drug-resistant sublines, respectively. No major alterations were observed in epidermal growth factor and c-erbB4 expression. Few alterations were observed in two sublines derived from estrogen receptor-negative cells. Higher levels of phosphotyrosine residues were detected in a subset of the resistant sublines, indicating an increased tyrosine kinase activity in these cells. Interestingly, decreased growth rates were observed in all of the sublines, despite up-regulated growth factor-related gene expression. Taken together, these data suggest that loss of estrogen receptor, increased expression of growth factor pathway genes, and decreased growth rate regularly occur in drug-resistant breast cancer cells. Although we do not know whether the altered expression of growth factor pathway genes is linked as a cause or a consequence of the reduced growth rate, it is well established that decreased growth rate confers drug resistance. These phenotypic changes in drug-resistant human breast cancer cells could serve to initiate, support, or extend the drug resistance.


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias de la Mama/genética , Resistencia a Múltiples Medicamentos/genética , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Genes erbB-2 , Proteínas Proto-Oncogénicas/genética , División Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/biosíntesis , ADN-Topoisomerasas de Tipo II/genética , Femenino , Humanos , Proto-Oncogenes , ARN Mensajero/genética , Receptor ErbB-2/genética , Receptor ErbB-3 , Receptores de Estrógenos/análisis , Receptores de Estrógenos/genética , Transcripción Genética , Células Tumorales Cultivadas
6.
Mol Cell Endocrinol ; 110(1-2): 205-11, 1995 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-7672450

RESUMEN

Human breast cancer cells selected for multidrug resistance frequently overexpress ligands and receptors in the epidermal growth factor (EGF) receptor family. To determine whether this overexpression contributes to the drug resistant phenotype, EGF receptor transfected ZR75B human breast cancer cells were examined. Two EGF receptor overexpressing clones were evaluated: clone 11 with > 1 x 10(6) sites, and clone 13 with 310,000 receptor sites/cell. These were compared with clone 2-neo, which was transfected with the neomycin gene only and contained 43,000 receptor sites/cell. The EGF receptor overexpressing clones and the neo transfected control clone displayed comparable growth rates. Cytotoxicity analyses were performed with doxorubicin, vinblastine, cisplatin and 5-fluorouracil to determine the sensitivity of the clones to antineoplastic drugs. The EGF receptor overexpressing clones were found to be 1.5-5.6 times more resistant to the four drugs tested. This increase in the IC50 conferred a selective advantage when grown in the presence of 2, 3 and 6 ng/ml doxorubicin. Clone 13 cells overtook a mixed population which began with clone 2-neo comprising 95% of the cells. Clone 2-neo remained the dominant clone in the absence of drug. Finally, after long-term selection of the clones with 6 ng/ml doxorubicin, clone 2-neo became fourfold more resistant than the unselected clone 2-neo, a level which was comparable to that found in the EGF receptor overexpressing clones 11 and 13. No additional increase in resistance was observed for these clones, suggesting that clone 2-neo had developed additional resistance mechanisms.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Múltiples Medicamentos , Receptores ErbB/genética , Expresión Génica , Transfección , Cisplatino/farmacología , Doxorrubicina/farmacología , Receptores ErbB/fisiología , Fluorouracilo/farmacología , Humanos , Neomicina , Fenotipo , ARN Mensajero/metabolismo , Células Tumorales Cultivadas , Vinblastina/farmacología
7.
Am J Clin Oncol ; 14 Suppl 2: S5-14, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1962598

RESUMEN

Pharmacologic investigations with droloxifene in vitro and in vivo revealed that droloxifene is a more efficient antiestrogen than tamoxifen. Droloxifene differs from tamoxifen in the following ways: it has a more than 10-fold higher binding affinity to the estrogen receptor; it shows lower estrogenic and higher antiestrogenic effects on rat uterus, indicating a higher therapeutic index; it more potently inhibits growth of various human ER-positive mammary carcinoma cell lines; short-term exposures with clinically relevant concentrations of droloxifene produce long-term growth inhibition of human ER-positive cancer cells and are more effective than continuous treatment with tamoxifen; it more effectively reduces S-phases and arrests ER-positive cells in G1-phase of the cell cycle; it antagonizes estrogen independent, growth factor stimulated proliferation of MCF-7 cells with higher efficiency; it blocks estrogen activated c-myc expression better than tamoxifen; it more effectively inhibits growth of various experimental tumors of animal (R 3230, DMBA) and human (T61) origin. Therefore, in all experimental systems, it was found that droloxifene is a more potent antiestrogen than tamoxifen.


Asunto(s)
Antineoplásicos/farmacología , Antagonistas de Estrógenos/farmacología , Tamoxifeno/análogos & derivados , Animales , Antineoplásicos/uso terapéutico , División Celular/efectos de los fármacos , Antagonistas de Estrógenos/uso terapéutico , Femenino , Genes myc/efectos de los fármacos , Humanos , Neoplasias Experimentales/tratamiento farmacológico , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Células Tumorales Cultivadas/efectos de los fármacos
8.
Biochem Biophys Res Commun ; 188(3): 1067-76, 1992 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-1445344

RESUMEN

Insulin-like growth factor I (IGF-I) was 3 times more potent in pagating MCF-7 cell proliferation than epidermal growth factor (EGF). IGF-I stimulated c-fos mRNA expression about 5 times less than EGF. Both growth factors were equipotent in inducing c-jun and c-myc mRNA expressions. The protein level of c-Myc correlated with the mRNA level. IGF-I and EGF stimulated the transcriptional activity dependent on the phorbol 12-myristate 13-acetate-responsive element (TRE) to the same extent, when measured by the chloramphenicol acetyl transferase activity of a transiently transfected multiple TRE construct. These results strongly indicate that the expression levels of the measured proto-oncogenes do not correlate with the increase of growth stimulation by IGF-I and EGF and are not growth rate limiting for the human MCF-7 breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes fos , Genes jun , Genes myc , División Celular , Factor de Crecimiento Epidérmico/farmacología , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , ARN Mensajero/análisis , Transcripción Genética , Células Tumorales Cultivadas
9.
Eur J Biochem ; 221(2): 631-7, 1994 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8174543

RESUMEN

In breast cancer, hormone dependency is inversely correlated with the number of surface epidermal-growth-factor (EGF) receptors on the tumor cells. In vitro, EGF stimulated only hormone-dependent immortalized human breast cancer cells to grow with an increased rate whereas hormone-independent cells were not affected by EGF. The number of EGF surface receptors is about 5-10-times smaller on hormone-dependent cells than on hormone-independent cells. Two cell lines representing the two cell types were used to demonstrate the signal-transduction capabilities of the EGF receptors. The two cell lines were the hormone-dependent MCF-7 cells and the hormone-independent MDA-MB-231 cells. Incubation at 37 degrees C for 15 min with 10(-8) M EGF increased the surface EGF-receptor density substantially on MCF-7 cells (50%) and reduced the number of these receptors on MDA-MB-231 cells to about 65% of the control. Both cell lines internalized a fluorescein-isothiocyanate-labeled EGF with similar kinetics. EGF triggered tyrosine phosphorylation of several targets in isolated MCF-7 cell membranes. One of these targets was shown by immunoprecipitation to be the EGF receptor. In MDA-MB-231 cell membranes, the EGF receptor was demonstrated to be the main target for tyrosine phosphorylation. The mRNA expression of the immediate early proto-oncogene c-fos was stimulated by EGF only in MCF-7 cells. In contrast, the mRNA of the EGF receptors was stimulated by EGF in both cell lines. These results demonstrate that, although EGF-binding sites are present on both cell lines, their signal-transduction capacity and activities are substantially different and resulted in a divergent response of the two cell types to EGF.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Neoplasias Hormono-Dependientes/metabolismo , Transducción de Señal , Northern Blotting , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Citometría de Flujo , Fluoresceína-5-Isotiocianato , Genes fos , Humanos , Neoplasias Hormono-Dependientes/patología , Fosforilación , Proto-Oncogenes Mas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas , Tirosina/metabolismo
10.
Int J Cancer ; 53(2): 290-7, 1993 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-8425767

RESUMEN

Recently, it was reported that the anti-estrogen tamoxifen not only inhibits estradiol-stimulated growth of MCF-7 cells but also significantly reduces the proliferation rate of cells stimulated by growth factors. We have confirmed this finding and also shown that the new anti-estrogen droloxifene inhibits the proliferation of epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I)-stimulated MCF-7 cells. The growth-factor-induced proliferation was inhibited in a dose-dependent manner by the anti-estrogens in the complete absence of estrogen and FCS. Of the anti-estrogens, droloxifene was considerably more potent than tamoxifen. Because the expression of the proto-oncogenes c-fos and c-myc has been considered a key event in development of the mitogenic response, we examined the effects of anti-estrogens on c-myc and c-fos gene expression. We included in these investigations the steroidal anti-estrogen ICI 164,384 because this compound has no or very little estrogenic activity. The studies revealed that all 3 anti-estrogens transiently induced c-myc mRNA expression. However, the anti-estrogens inhibited estradiol-induced c-myc mRNA expression, although with different potencies. Pre-incubation of MCF-7 cells with droloxifene and tamoxifen resulted in elevated levels of growth-factor-induced c-myc mRNA expression. In contrast, the anti-estrogens did not induce c-fos mRNA or affect the expression of c-fos mRNA induced by growth factors. In conclusion, non-steroidal anti-estrogens inhibit growth-factor-stimulated proliferation of MCF-7 cells without inhibiting growth-factor-induced c-myc or c-fos mRNA expression.


Asunto(s)
Neoplasias de la Mama/patología , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Antagonistas de Estrógenos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Neoplasias de la Mama/genética , División Celular/efectos de los fármacos , Estradiol/farmacología , Genes fos/efectos de los fármacos , Genes myc/efectos de los fármacos , Humanos , ARN Mensajero/efectos de los fármacos , ARN Neoplásico/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA