Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 65(10): 7371-7379, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35544687

RESUMEN

Precise identification of the tumor margins during breast-conserving surgery (BCS) remains a challenge given the lack of visual discrepancy between malignant and surrounding normal tissues. Therefore, we developed a fluorescent imaging agent, ICG-p28, for intraoperative imaging guidance to better aid surgeons in achieving negative margins in BCS. Here, we determined the pharmacokinetics (PK), biodistribution, and preclinical toxicity of ICG-p28. The PK and biodistribution of ICG-p28 indicated rapid tissue uptake and localization at tumor lesions. There were no dose-related effect and no significant toxicity in any of the breast cancer and normal cell lines tested. Furthermore, ICG-p28 was evaluated in clinically relevant settings with transgenic mice that spontaneously developed invasive mammary tumors. Intraoperative imaging with ICG-p28 showed a significant reduction in the tumor recurrence rate. This simple, nontoxic, and cost-effective method can offer a new approach that enables surgeons to intraoperatively identify tumor margins and potentially improves overall outcomes by reducing recurrence rates.


Asunto(s)
Neoplasias de la Mama , Mastectomía Segmentaria , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Diagnóstico por Imagen , Femenino , Humanos , Márgenes de Escisión , Mastectomía Segmentaria/métodos , Ratones , Imagen Óptica/métodos , Distribución Tisular
2.
EBioMedicine ; 76: 103850, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35108666

RESUMEN

BACKGROUND: Given the lack of visual discrepancy between malignant and surrounding normal tissue, current breast conserving surgery (BCS) is associated with a high re-excision rate. Due to the increasing cases of BCS, a novel method of complete tumour removal at the initial surgical resection is critically needed in the operating room to help optimize the surgical procedure and to confirm tumour-free edges. METHODS: We developed a unique near-infrared (NIR) fluorescence imaging probe, ICG-p28, composed of the clinically nontoxic tumour-targeting peptide p28 and the FDA-approved NIR dye indocyanine green (ICG). ICG-p28 was characterized in vitro and evaluated in multiple breast cancer animal models with appropriate control probes. Our experimental approach with multiple-validations and -blinded procedures was designed to determine whether ICG-p28 can accurately identify tumour margins in mimicked intraoperative settings. FINDINGS: The in vivo kinetics were analysed to optimize settings for potential clinical use. Xenograft tumours stably expressing iRFP as a tumour marker showed significant colocalization with ICG-p28, but not ICG alone. Image-guided surgery with ICG-p28 showed an over 6.6 × 103-fold reduction in residual normalized tumour DNA at the margin site relative to control approaches (i.e., surgery with ICG or palpation/visible inspection alone), resulting in an improved tumour recurrence rate (92% specificity) in multiple breast cancer animal models independent of the receptor expression status. ICG-p28 allowed accurate identification of tumour cells in the margin to increase the complete resection rate. INTERPRETATION: Our simple and cost-effective approach has translational potential and offers a new surgical procedure that enables surgeons to intraoperatively identify tumour margins in a real-time, 3D fashion and that notably improves overall outcomes by reducing re-excision rates. FUNDING: This work was supported by NIH/ National Institute of Biomedical Imaging and Bioengineering, R01EB023924.


Asunto(s)
Recurrencia Local de Neoplasia , Cirugía Asistida por Computador , Animales , Humanos , Verde de Indocianina , Márgenes de Escisión , Imagen Óptica/métodos , Cirugía Asistida por Computador/métodos
3.
Int J Oncol ; 47(1): 35-42, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25997921

RESUMEN

Most studies on cellular senescence (CS) have been performed in vitro by employing cytotoxic agents, irradiation, chromatin and telomerase modulators or by activating certain oncogenes. All these approaches usually lead to DNA damage, gene instability and/or chromatin alterations that primarily affect p53-p21 signaling. Little is known on whether retinoids and rexinoids, which are cell differentiation agents, can also induce CS in vitro and in vivo, and which molecular mechanisms are involved in promoting the senescent phenotype. We reviewed the recent publications on CS induced by retinoids and rexinoids in ER+ and ER- breast cancer cell lines and in corresponding animal models of mammary carcinogenesis which simulate those of human breast cancer. The role of retinoic acid receptors ß2 and 5 (RARß2 and RARß5) and of receptor independent genes involved in mediating the senescence program of retinoids and rexinoids in ER+ and ER- breast cancer cells is discussed. Potential strategists for clinical implication of CS as biomarker of prognosis and of response to treatment with retinoids, rexinoids and with other cell differentiation and antitumor agents are outlined.


Asunto(s)
Neoplasias de la Mama/metabolismo , Senescencia Celular/efectos de los fármacos , Retinoides/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Receptores de Estrógenos/metabolismo , Receptores de Ácido Retinoico/metabolismo
4.
Cancer Prev Res (Phila) ; 6(4): 299-308, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23430755

RESUMEN

Previous studies have shown that retinoids and rexinoids can prevent breast cancer in animal models and in women with increased risk of developing the disease. The cellular effects of these vitamin A analogues have been primarily associated with induction of differentiation and inhibition of proliferation. In this study, we tested the hypothesis that bexarotene (LGD1069, Targretin), a rexinoid, can not only inhibit cell proliferation but also induce cellular senescence in mammary epithelial cells, premalignant lesions, and tumors of the MMTV-Neu model of mammary carcinogenesis, which develops estrogen receptor-negative tumors. Mice with palpable mammary tumors were treated for 4 weeks with bexarotene at 80 or 40 mg/kg body weight, and senescent cells were determined by SA-ß-Gal assay. Bexarotene decreased in a dose-dependent manner the multiplicity of premalignant lesions and tumors, and this was associated with inhibition of cell proliferation and induction of cellular senescence and apoptosis. By double labeling of senescent cells, first by SA-ß-Gal and then by antibodies against genes related to cellular senescence, we found that p21, p16, and RARß, but not p53, were upregulated by bexarotene in mammary tumors and in breast cancer cell lines, suggesting involvement of multiple signaling pathways in mediating the senescence program of rexinoids. These findings indicate that, in addition to cell proliferation and apoptosis, cellular senescence could be used as a potential biomarker of response in breast cancer prevention and therapy studies with rexinoids and possibly with other antitumor agents.


Asunto(s)
Anticarcinógenos/administración & dosificación , Transformación Celular Neoplásica/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Tetrahidronaftalenos/administración & dosificación , Animales , Bexaroteno , Transformación Celular Neoplásica/genética , Femenino , Genes erbB-2 , Virus del Tumor Mamario del Ratón , Ratones , Ratones Transgénicos , Regulación hacia Arriba/efectos de los fármacos
5.
Int J Oncol ; 39(3): 577-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21667021

RESUMEN

The effect of retinoids on breast cancer has been predominantly studied in vitro, on established cell lines, which in biology differ significantly from primary tumor cells. Little is known on whether early in vitro passages of breast cancer cells (EPBCCs) are differentially sensitive to retinoids and differentially express retinoid acid receptors (RARs) and retinoid X receptors (RXRs). We have previously identified a novel RARß isoform (RARß5) and hypothesized that it may serve as a potential target of retinoids in EPBCCs. Breast cancer cells isolated from primary tumors were cultured in vitro for 6-12 passages (EPBCCs) and their epithelial origin was confirmed by a cocktail of antibodies against cytokeratins. EPBCCs were treated for 4 days with 1.0 µM of all-trans retinoic acid (atRA), 9-cis retinoic acid (9cRA) or 4-hydroxy-phenylretinamide (4-HPR) and their viability determined by MTT assay. Among nine EPBCCs consistently grown in vitro, three were resistant to the above retinoids, five were susceptible to atRA, four to 4-HPR and two to 9cRA, suggesting that patients with breast carcinomas may differentially respond to various retinoids. All EPBBCs differentially expressed RARα, RARγ, RXRα, RXRß proteins and RARß5 and RARß2 mRNAs. However, only one EPBCC (BCA-2) expressed RARß5 at mRNA and protein level and it was resistant to retinoids, both in vitro and in a xenograft tumor assay. RARß5 suppression by siRNA in BCA-2 cells increased their susceptibility to atRA. No correlation was found between sensitivity of EPBCCs to the above retinoids and RARß5 and RARß2 mRNA expression. atRA reduced RARß expression in most EPBCCs suggesting that this retinoid receptor is most probably the prime target of retinoids in breast cancer. These data may have clinical implication in selecting patients with breast cancer that would benefit the most from clinical trials with retinoids.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Receptores de Ácido Retinoico/biosíntesis , Retinoides/farmacología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones Desnudos , Isoformas de Proteínas , ARN Interferente Pequeño/genética , Receptores de Ácido Retinoico/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Angiogenesis ; 14(3): 355-69, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21667138

RESUMEN

Amino acids 50-77 (p28) of azurin, a 128 aa cupredoxin isolated from Pseudomonas aeruginosa, is essentially responsible for azurin's preferential penetration of cancer cells. We now report that p28 also preferentially penetrates human umbilical vein endothelial cells (HUVEC), co-localized with caveolin-1 and VEGFR-2, and inhibits VEGF- and bFGF-induced migration, capillary tube formation and neoangiogenesis in multiple xenograft models. The antiangiogenic effect of p28 in HUVEC is associated with a dose-related non-competitive inhibition of VEGFR-2 kinase activity. However, unlike other antiangiogenic agents that inhibit the VEGFR-2 kinase, p28 decreased the downstream phosphorylation of FAK and Akt that normally precedes cellular repositioning of the cytoskeletal (F-actin), focal adhesion (FAK and paxillin), and cell to cell junction protein PECAM-1, inhibiting HUVEC motility and migration. The decrease in pFAK and pAkt levels suggests that p28 induces a pFAK-mediated loss of HUVEC motility and migration and a parallel Akt-associated reduction in cell matrix attachment and survival. This novel, direct antiangiogenic effect of p28 on endothelial cells may enhance the cell cycle inhibitory and apoptotic properties of this prototype peptide on tumor cell proliferation as it enters a Phase II clinical trial.


Asunto(s)
Antineoplásicos/farmacocinética , Azurina/farmacología , Péptidos de Penetración Celular/farmacología , Células Endoteliales/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Actinas/metabolismo , Animales , Antineoplásicos/química , Azurina/química , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular , Péptidos de Penetración Celular/química , Ensayos Clínicos Fase II como Asunto , Células Endoteliales/patología , Adhesiones Focales/metabolismo , Humanos , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fragmentos de Péptidos/química , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Pseudomonas aeruginosa/química , Venas Umbilicales/metabolismo , Venas Umbilicales/patología
7.
BMC Cancer ; 10: 541, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20932324

RESUMEN

BACKGROUND: p27 is a cell cycle suppressor gene, whose protein is a negative regulator of cyclin/cdk complexes. p27 is also a potential target of retinoids in cancer prevention studies. In benign prostate hyperplasia (BPH), and in most carcinomas, p27(Kip1) is down-regulated, suggesting its potential resistance to retinoids. To test this hypothesis, we examined the efficacy of 9-cis retinoic acid (9cRA) to suppress prostate cell proliferation (PECP) and carcinogenesis in p27(Kip1) deficient mice. METHODS: p27(Kip1) deficient (-/-), heterozygous (+/-) and homozygous (+/+) mice were treated for 7 days with testosterone, 9cRA, or with both, and cell proliferation in dorsolateral prostate (DLP) was determined by BrdU labeling. Prostate carcinogenesis was induced by N-Methyl-N-Nitrosourea (MNU) and hormone stimulation. RESULTS: PECP in DLP of two-month-old mice of all genotypes was similar but significantly increased in old p27-/- mice only. Testosterone treatment increased PECP in all three p27 genotypes with the highest values in p27-/- mice. p27(Kip1) deficiency did not affect the response of PEC to 9cRA and to 9cRA+testosterone. The decrease of p27(Kip1) in p27+/- and p27-/- mice progressively increased the incidence and frequency of PIN and tumors. 9cRA suppressed PIN in all three p27 genotypes and this was associated with decreased PECP and increased cellular senescence. CONCLUSIONS: This data indicates that p27(Kip1) deficiency promotes prostate cell proliferation and carcinogenesis but does not affect 9cRA's potential to suppress prostate carcinogenesis, suggesting that patients with PIN and carcinomas lacking or having a low level of p27(Kip1) expression may also benefit from clinical trials with retinoids.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Retinoides/metabolismo , Alitretinoína , Animales , Antineoplásicos/farmacología , Ciclo Celular , Proliferación Celular , Modelos Animales de Enfermedad , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tretinoina/farmacología
8.
Mol Cancer Ther ; 8(10): 2947-58, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19808975

RESUMEN

We report that amino acids 50 to 77 of azurin (p28) preferentially enter the human breast cancer cell lines MCF-7, ZR-75-1, and T47D through a caveolin-mediated pathway. Although p28 enters p53 wild-type MCF-7 and the isogenic p53 dominant-negative MDD2 breast cancer cell lines, p28 only induces a G(2)-M-phase cell cycle arrest and apoptosis in MCF-7 cells. p28 exerts its antiproliferative activity by reducing proteasomal degradation of p53 through formation of a p28:p53 complex within a hydrophobic DNA-binding domain (amino acids 80-276), increasing p53 levels and DNA-binding activity. Subsequent elevation of the cyclin-dependent kinase inhibitors p21 and p27 reduces cyclin-dependent kinase 2 and cyclin A levels in a time-dependent manner in MCF-7 cells but not in MDD2 cells. These results suggest that p28 and similar peptides that significantly reduce proteasomal degradation of p53 by a MDM2-independent pathway(s) may provide a unique series of cytostatic and cytotoxic (apoptotic) chemotherapeutic agents.


Asunto(s)
Azurina/química , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclinas/metabolismo , Femenino , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res ; 69(2): 537-46, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19147567

RESUMEN

Azurin, a member of the cupredoxin family of copper containing redox proteins, preferentially penetrates human cancer cells and exerts cytostatic and cytotoxic (apoptotic) effects with no apparent activity on normal cells. Amino acids 50 to 77 (p28) of azurin seem responsible for cellular penetration and at least part of the antiproliferative, proapoptotic activity of azurin against a number of solid tumor cell lines. We show by confocal microscopy and fluorescence-activated cell sorting that amino acids 50 to 67 (p18) are a minimal motif (protein transduction domain) responsible for the preferential entry of azurin into human cancer cells. A combination of inhibitors that interfere with discrete steps of the endocytotic process and antibodies for caveolae and Golgi-mediated transport revealed that these amphipathic, alpha-helical peptides are unique. Unlike the cationic cell-penetrating peptides, alpha-helical antennapedia-like, or VP22 type peptides, p18 and p28 are not bound by cell membrane glycosaminoglycans and preferentially penetrate cancer cells via endocytotic, caveosome-directed, and caveosome-independent pathways. Once internalized, p28, but not p18, inhibits cancer cell proliferation initially through a cytostatic mechanism. These observations suggest the azurin fragments, p18 and p28, account for the preferential entry of azurin into human cancer cells and a significant amount of the antiproliferative activity of azurin on human cancer cells, respectively.


Asunto(s)
Azurina/farmacocinética , Neoplasias/metabolismo , Fragmentos de Péptidos/farmacocinética , Secuencia de Aminoácidos , Azurina/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Células HCT116 , Humanos , Datos de Secuencia Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fragmentos de Péptidos/farmacología , Estructura Terciaria de Proteína
10.
Clin Cancer Res ; 13(18 Pt 1): 5488-96, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875779

RESUMEN

PURPOSE: The methylnitrosourea (MNU)-induced mammary cancer model in rats is similar to estrogen receptor-positive breast cancer in women. In prevention studies using this model, tumor incidence and multiplicity were typically primary end points. The ability of various agents administered for a short period to modulate cell proliferation [proliferation index (PI)] and apoptosis [apoptotic index (AI)] in mammary cancers was compared with their efficacy in long-term prevention and therapy studies. EXPERIMENTAL DESIGN: Rats were injected with MNU to induce mammary cancers. For the prevention studies, agents were administered by gavage or in the diet beginning 5 days after MNU. For proliferation (PI) and apoptosis (AI) experiments, animals with a palpable mammary cancer were treated with the agents for only 4 to 7 days. PI was determined following 5-bromodeoxyuridine labeling whereas AI was determined using the terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling assay. Therapeutic efficacy was evaluated by measuring cancer size over a 6-week period. RESULTS: Treatments with differing chemopreventive efficacy and mechanism(s) of action were examined: (a) hormonal treatments [tamoxifen, vorozole (an aromatase inhibitor), and ovariectomy]; (b) retinoid X receptor agonists (targretin, 9-cis retinoic acid, and UAB30); (c) inducers of drug-metabolizing enzymes (indole-3-carbinol, 5,6 benzoflavone, and diindoylmethane); (d) agents that alter signal transduction (R115777, a farnesyltransferase inhibitor); Iressa (an epidermal growth factor receptor inhibitor); sulindac and celecoxib (cyclooxygenase 1/2 and cyclooxygenase 2 inhibitors); and (e) diverse agents including meclizine, vitamin C, and sodium phenylbutyrate. Correlations between inhibition of PI, increase of AI, and chemopreventive efficacy were observed. Although most agents with moderate or low preventive efficacy suppressed PI, they minimally affected AI. CONCLUSIONS: The data confirmed that the short-term effects of various agents on cell proliferation and apoptosis in small mammary cancers can predict their preventive/therapeutic efficacy. Thus, these biomarkers can be used to help determine the efficacy of compounds in phase II clinical prevention trials.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis , Biomarcadores/análisis , Proliferación Celular/efectos de los fármacos , Femenino , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Metilnitrosourea/toxicidad , Ratas , Ratas Sprague-Dawley
11.
Cancer Lett ; 222(2): 153-63, 2005 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-15863264

RESUMEN

Recently we found that retinoic acid receptors (alpha, beta, gamma) and retinoid X receptors (alpha, beta, gamma) are variably expressed in MCF10A model of breast cancer development and progression. Here we employed this model to assess both in vitro and in vivo sensitivity of cells to retinoids and the role of RARbeta2 in mediating the antitumor potential of retinoids. In vitro, we found that transformation of the benign MCF10A cells into premalignant MCF10AT and malignant MCF10CA1a cells increased their sensitivity to 4-(Hydroxyphenyl)retinamide (4-HPR) and all-trans-retinoic acid (atRA) but not to 9-cis-retinoic acid (9cRA) and LGD1069 and this was associated with loss of induction of RARbeta2 by retinoids. RARbeta2 expression in premalignant MCF10AT cells decreased their proliferating activity and increased their sensitivity to atRA. In vivo, when transplanted into the mammary fat pads of nude mice, MCF10A cells did not grow, MCF10AT cells formed slow-growing tumor nodules, and MCF10CA1a cells were highly malignant, grew quickly and infiltrated the surrounding tissues. Of the retinoids used, only 4-HPR suppressed the growth of slow-growing hyperplastic and premalignant MCF10AT but not of the malignant MCF10CA1a tumor nodules. These data may have clinical implication in selecting women with hyperplastic and premalignant breast lesions that may benefit the most from clinical trials with retinoids.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/efectos de los fármacos , Lesiones Precancerosas , Receptores de Ácido Retinoico/biosíntesis , Receptores de Ácido Retinoico/fisiología , Retinoides/farmacología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/patología , Femenino , Humanos , Células Tumorales Cultivadas
12.
Breast Cancer Res ; 7(6): R1132-40, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16457693

RESUMEN

INTRODUCTION: Dehydroepiandrosterone (DHEA), an adrenal 17-ketosteroid, is a precursor of testosterone and 17beta-estradiol. Studies have shown that DHEA inhibits carcinogenesis in mammary gland and prostate as well as other organs, a process that is not hormone dependent. Little is known about the molecular mechanisms of DHEA-mediated inhibition of the neoplastic process. Here we examine whether DHEA and its analog DHEA 8354 can suppress the progression of hyperplastic and premalignant (carcinoma in situ) lesions in mammary gland toward malignant tumors and the cellular mechanisms involved. METHODS: Rats were treated with N-nitroso-N-methylurea and allowed to develop mammary hyperplastic and premalignant lesions with a maximum frequency 6 weeks after carcinogen administration. The animals were then given DHEA or DHEA 8354 in the diet at 125 or 1,000 mg/kg diet for 6 weeks. The effect of these agents on induction of apoptosis, senescence, cell proliferation, tumor burden and various effectors of cellular signaling were determined. RESULTS: Both agents induced a dose-dependent decrease in tumor multiplicity and in tumor burden. In addition they induced a senescent phenotype in tumor cells, inhibited cell proliferation and increased the number of apoptotic cells. The DHEA-induced cellular effects were associated with increased expression of p16 and p21, but not p53 expression, implicating a p53-independent mechanism in their action. CONCLUSION: We provide evidence that DHEA and DHEA 8354 can suppress mammary carcinogenesis by altering various cellular functions, inducing cellular senescence, in tumor cells with the potential involvement of p16 and p21 in mediating these effects.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Neoplasias de la Mama/prevención & control , Transformación Celular Neoplásica , Senescencia Celular/efectos de los fármacos , Deshidroepiandrosterona/farmacología , Neoplasias Mamarias Experimentales/prevención & control , Alquilantes/administración & dosificación , Animales , Carcinoma in Situ/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Deshidroepiandrosterona/análogos & derivados , Femenino , Humanos , Metilnitrosourea/administración & dosificación , Ratas , Ratas Sprague-Dawley , Proteína p53 Supresora de Tumor/fisiología
13.
Breast Cancer Res Treat ; 77(3): 253-64, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12602925

RESUMEN

Tamoxifen has been widely used for treatment, and more recently, for the prevention of breast cancer. Since breast carcinomas are composed of heterogeneous populations of estrogen receptor-positive (ER+) cells, we hypothesized that tamoxifen may suppress tumor growth by differentially affecting cell proliferation and apoptosis. ER+ mammary tumors were induced in Sprague-Dawley rats by N-methyl-N-nitrosourea (MNU) and when they became palpable, the animals were treated for 5, 10, or 20 days with tamoxifen, 1.0 mg/kg body weight. Tamoxifen induced a time-dependent decrease in proliferating (BrdU-labeled) cells, arrested the cells in G1/0 phase, and differentially decreased the cyclin E and cyclin D1 expression at mRNA and protein levels. In the same tumors, apoptotic cells increased during the first 10 days of treatment, but their number remained unchanged with extension of the treatment to 20 days. Thus, we provide data that tamoxifen may differentially affect cell proliferation and apoptosis in mammary tumors and that the expression levels of cyclin D1 and cyclin E might also be considered potential intermediate biomarkers of response of mammary tumors to tamoxifen and possibly to other selective estrogen receptor modulators (SERMs).


Asunto(s)
Biomarcadores de Tumor/metabolismo , Ciclina D1/metabolismo , Ciclina E/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Tamoxifeno/uso terapéutico , Animales , Western Blotting , División Celular/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Ciclina E/efectos de los fármacos , Femenino , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Metilnitrosourea , Neoplasias Hormono-Dependientes/inducido químicamente , Neoplasias Hormono-Dependientes/patología , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Células Tumorales Cultivadas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA