Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Steroids ; 153: 108521, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31604073

RESUMO

Estrogen receptors (ERs) and the PTEN-Akt-mTor pathway are important growth regulators in human breast cancer cells, which both are known to affect response to tamoxifen therapy. Recently it was reported that ERß activates PTEN expression and tamoxifen sensitivity of human breast cancer cells. In this study we examined whether expression of ERß in turn might be affected by tumor suppressor PTEN, analyzed the effect of this interaction on tamoxifen response and the co-expression of both genes in human breast cancer samples. After siRNA-mediated PTEN knockdown, Western blot analysis revealed a reduction of ERß protein expression by 67.2% in MCF-7 cells and by 73.6% in T-47D cells (both p < 0.01), results which could be verified on the mRNA level. In cells with normal PTEN and ERß status, after 6 days of treatment with 1 µM 4-OH tamoxifen, E2-driven proliferation was decreased by 64.5% in MCF-7 and by 57.7% in T-47D cells (both p < 0.01). After knockdown of PTEN expression, the same concentration of 4-OH TAM reduced E2-triggered growth only by 34.9% (MCF-7) and by 41.8% (T-47D) (both p < 0.01 vs control siRNA). Importantly, treatment with ERß agonist DPN (5 nM) significantly decreased the inhibitory effect of a PTEN knockdown on tamoxifen response of both cell lines (p < 0.05). Additionally, Spearmans rank association analysis of PTEN and ERß 1 mRNA levels in 115 normal and malignant breast tissue samples revealed a strong positive correlation of both genes (rho = 0.6085, p < 0.0001). The data of previous studies reporting an important role of ERß in tamoxifen sensitivity and our findings suggest down-regulation of ERß triggered by PTEN knockdown contributed to the decreased response of breast cancer cells to tamoxifen observed in this study. Our data also suggest expression of ERß might be maintained by tumor suppressor PTEN in human breast cancer cells.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Receptor beta de Estrogênio/antagonistas & inibidores , PTEN Fosfo-Hidrolase/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia , Tamoxifeno/antagonistas & inibidores , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Humanos , Células MCF-7 , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade , Tamoxifeno/farmacologia , Células Tumorais Cultivadas
2.
J Natl Cancer Inst ; 108(3)2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26631176

RESUMO

BACKGROUND: Controversy persists about whether certain antidepressants reduce tamoxifen's effectiveness on lowering breast cancer recurrence. We investigated whether taking tamoxifen and antidepressants (in particular, paroxetine) concomitantly is associated with an increased risk of recurrence or contralateral breast cancer. METHODS: We examined 16 887 breast cancer survivors (TNM stages 0-II) diagnosed between 1996 and 2007 and treated with tamoxifen in two California health plans. Women were followed-up through December 31, 2009, for subsequent breast cancer. The main exposure was the percent of days of overlap when both tamoxifen and an antidepressant (paroxetine, fluoxetine, other selective serotonin reuptake inhibitors, tricyclics, and other classes) were used. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using multivariable Cox regression models with time-varying medication variables. RESULTS: Of the 16 887 women, half (n = 8099) used antidepressants and 2946 women developed subsequent breast cancer during the 14-year study period. We did not find a statistically significant increased risk of subsequent breast cancer in women who concurrently used paroxetine and tamoxifen. For 25%, 50%, and 75% increases in percent overlap days between paroxetine and tamoxifen, hazard ratios were 1.06 (95% CI = 0.98 to 1.14, P = .09), 1.13 (95% CI = 0.98 to 1.30, P = .09), and 1.20 (95% CI = 0.97 to 1.49, P = .09), respectively, in the first year of tamoxifen treatment but were not statistically significant. Hazard ratios decreased to 0.94 (95% CI = 0.81 to 1.10, P = .46), 0.89 (95% CI = 0.66 to 1.20, P = .46), and 0.85 (95% CI = 0.54 to 1.32, P = .46) by the fifth year (all non-statistically significantly). Absolute subsequent breast cancer rates were similar among women who used paroxetine concomitantly with tamoxifen vs tamoxifen-only users. For the other antidepressants, we again found no such associations. CONCLUSIONS: Using the comprehensive electronic health records of insured patients, we did not observe an increased risk of subsequent breast cancer in women who concurrently used tamoxifen and antidepressants, including paroxetine.


Assuntos
Antidepressivos de Segunda Geração/uso terapêutico , Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Depressão/tratamento farmacológico , Recidiva Local de Neoplasia/prevenção & controle , Paroxetina/uso terapêutico , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/uso terapêutico , Adulto , Idoso , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Neoplasias da Mama/psicologia , Estudos de Coortes , Depressão/etiologia , Feminino , Recursos em Saúde/estatística & dados numéricos , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/epidemiologia , Estadiamento de Neoplasias , Razão de Chances , Modelos de Riscos Proporcionais , Sobreviventes , Estados Unidos/epidemiologia
4.
Drug Metab Dispos ; 43(6): 889-97, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25795461

RESUMO

We previously reported upregulation of UGT2B15 by 17ß-estradiol in breast cancer MCF7 cells via binding of the estrogen receptor α (ERα) to an estrogen response unit (ERU) in the proximal UGT2B15 promoter. In the present study, we show that this ERα-mediated upregulation was significantly reduced by two ER antagonists (fulvestrant and raloxifene) but was not affected by a third ER antagonist, 4-hydroxytamoxifen (4-OHTAM), a major active tamoxifen (TAM) metabolite. Furthermore, we found that, similar to 17ß-estradiol, 4-OHTAM and endoxifen (another major active TAM metabolite) elevated UGT2B15 mRNA levels, and that this stimulation was significantly abrogated by fulvestrant. Further experiments using 4-OHTAM revealed a critical role for ERα in this regulation. Specifically; knockdown of ERα expression by anti-ERα small interfering RNA reduced the 4-OHTAM-mediated induction of UGT2B15 expression; 4-OHTAM activated the wild-type but not the ERU-mutated UGT2B15 promoter; and chromatin immunoprecipitation assays showed increased ERα occupancy at the UGT2B15 ERU in MCF7 cells upon exposure to 4-OHTAM. Together, these data indicate that both 17ß-estradiol and the antiestrogen 4-OHTAM upregulate UGT2B15 in MCF7 cells via the same ERα-signaling pathway. This is consistent with previous observations that both 17ß-estradiol and TAM upregulate a common set of genes in MCF7 cells via the ER-signaling pathway. As 4-OHTAM is a UGT2B15 substrate, the upregulation of UGT2B15 by 4-OHTAM in target breast cancer cells is likely to enhance local metabolism and inactivation of 4-OHTAM within the tumor. This represents a potential mechanism that may reduce TAM therapeutic efficacy or even contribute to the development of acquired TAM resistance.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Drogas em Investigação/farmacologia , Indução Enzimática/efeitos dos fármacos , Antagonistas do Receptor de Estrogênio/farmacologia , Glucuronosiltransferase/metabolismo , Tamoxifeno/análogos & derivados , Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/metabolismo , Neoplasias da Mama/metabolismo , Drogas em Investigação/química , Drogas em Investigação/metabolismo , Antagonistas do Receptor de Estrogênio/química , Antagonistas do Receptor de Estrogênio/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Genes Reporter/efeitos dos fármacos , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/genética , Humanos , Células MCF-7 , Mutação , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Interferência de RNA , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia
5.
Carcinogenesis ; 33(4): 895-901, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22266527

RESUMO

The present study examined the effect of dietary genistein, a soy isoflavone, on breast cancer patients who take tamoxifen, an antiestrogen treatment, using a preclinical model. The interaction of various doses of genistein with tamoxifen on the growth of estrogen receptor-positive breast cancer MCF-7 cells was investigated by subcutaneously injecting MCF-7 cells into the flank of ovariectomized athymic mice. Animals were randomized into eight experimental groups with 10-13 mice per group: control (C), estrogen (E) (0.08 mg E implant), tamoxifen (T) (3 mg T implant), estrogen + tamoxifen (E + T), tamoxifen + 500 p.p.m. genistein (T + G500), estrogen + tamoxifen + 250 p.p.m. genistein (E + T + G250), estrogen + tamoxifen + 500 p.p.m. genistein (E + T + G500) and estrogen + tamoxifen + 1000 p.p.m. genistein (E + T + G1000). Treatment of tamoxifen significantly reduced the estrogen-induced MCF-7 tumor prevalence and tumor size. This inhibitory effect of tamoxifen was significantly negated by the low doses of dietary genistein (250 and 500 p.p.m.), whereas the 1000 p.p.m. genistein did not have the same effect. Cells harvested from tamoxifen-treated tumors retained estrogen responsiveness of their progenitor MCF-7 cells, indicating that the abrogating effect of genistein on tamoxifen-treated tumor growth was not caused by a diminished tamoxifen response but directly by genistein. The low doses of dietary genistein abrogated the inhibitory effect of tamoxifen potentially by acting on the tumor cell proliferation/apoptosis ratio and the messenger RNA (mRNA) expression of cyclin D1 in addition to regulating the mRNA expression of progesterone receptor. Therefore, data from the current study suggest that caution is warranted regarding the consumption of dietary genistein by breast cancer patients while on tamoxifen therapy.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Dieta , Genisteína/farmacologia , Tamoxifeno/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Primers do DNA , Relação Dose-Resposta a Droga , Interações Medicamentosas , Estrogênios/sangue , Genisteína/administração & dosagem , Camundongos , Camundongos Nus
6.
Environ Toxicol Pharmacol ; 31(1): 143-52, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21787679

RESUMO

The genotoxic actions of anti-neoplastic drugs can lead to the development of secondary cancers in patients in extended remission. One of the most attractive approaches to disease prevention involves the use of natural antioxidants to protect tissue against toxic injury. We investigated the modulatory effects of exogenously administered taurine, on the genotoxicity of two well known anti-neoplastic drugs methotrexate (MTX) and tamoxifen (TAM) in Swiss albino mice. The animals were randomly divided into six groups consisting of ten mice each. Two groups were received single intraperitoneal injection of MTX (10 mg/kgb.wt.) and TAM (50 mg/kgb.wt.) to induce genotoxicity. Two other groups were treated orally with taurine (100 mg/kgb.wt.) for nine days prior to MTX and TAM administration. A vehicle treated control group and taurine control groups were also included. The protective effects of taurine were monitored by apoptosis assays and level of reduced glutathione (GSH), a key antioxidant, in liver, chromosomal aberrations in somatic and germ cells as well as sperm count, motility and morphology. The results indicated that taurine pre-treatment showed significant increment in the levels of GSH content, reduction in DNA fragmentation and ladder formation in hepatic tissue, suggesting the antioxidant activity of taurine may reduce the toxic effects of MTX and TAM. Treatment with taurine showed also significant reduction in the frequency of chromosomal aberrations in both somatic and germ cells. Moreover, it increases sperm count and motility, and decreases the incidence of sperm abnormalities. In conclusion, it appears that taurine protects against anti-neoplastic drugs-induced genotoxicity in somatic and germ tissues and may be of therapeutic potential in alleviating the risk of secondary tumors in chemotherapy.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Antimutagênicos , Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/toxicidade , Metotrexato/antagonistas & inibidores , Metotrexato/toxicidade , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/toxicidade , Taurina/farmacologia , Androgênios/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Aberrações Cromossômicas/efeitos dos fármacos , Dano ao DNA , Fragmentação do DNA/efeitos dos fármacos , Eletroforese em Gel de Ágar , Células Germinativas/efeitos dos fármacos , Glutationa/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatócitos/efeitos dos fármacos , Compostos de Sulfidrila/metabolismo
7.
Gynecol Endocrinol ; 27(10): 830-6, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21171931

RESUMO

OBJECTIVE: Progesterone (P4) has been implicated as a protective factor for ovarian and endometrial cancers, yet little is known about its mechanism of action. We have shown apoptosis in ovarian and endometrial cancer cells with high doses of P4. Increased generation of reactive oxygen species (ROS) and an altered redox status have long been observed in cancer cells. The goal of this study was to assess the effect of P4 on cell growth, ROS generation, oxidative stress markers, and the expression of antioxidant proteins. METHODS: All experiments were performed in vitro using cancer cell lines. Cell proliferation was determined using MTS proliferation assay. Production of ROS in cells was measured with the ROS indicator dye, aminophenyl fluorescein. Alterations in expression of antioxidant and apoptotic proteins were assessed by Western blotting. RESULTS: The exposure of ovarian and endometrial cancer cell cultures to various doses of P4 caused a dose-dependent decrease in cell viability and the activation of caspase-3. Levels of ROS, markers of oxidative stress, and antioxidant proteins were elevated in cancer cells compared to normal cells and a marked decrease in their expression was seen following P4 treatment. In cancer cells, ROS was elevated while p-53 expression was low. P4 exposure of cells resulted in increased p-53 and BAX and decreased BCL-2 expression. CONCLUSIONS: The data indicates that P4 has antioxidant effects. It alleviates ROS stress and causes apoptosis by upregulating proapoptotic (p-53 and BAX) and decreasing antiapoptotic (BCL-2) gene expression in cancer cells. These findings could have potential therapeutic implications.


Assuntos
Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias do Endométrio/metabolismo , Neoplasias Ovarianas/metabolismo , Progesterona/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos Hormonais/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores/metabolismo , Caspase 3/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Antagonistas de Hormônios/farmacologia , Humanos , Proteínas de Neoplasias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Proteína Supressora de Tumor p53/metabolismo
8.
Food Chem Toxicol ; 47(7): 1689-95, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19394397

RESUMO

Tamoxifen (TAM) is widely used in the treatment and prevention of breast cancer. Adverse effects of TAM include hepatotoxicity. Caffeic acid phenethyl ester (CAPE), an active component of propolis, has been used in folk medicine for diverse ailments. In the current study, the protective effects of CAPE against TAM-induced hepatotoxicity in female rats were evaluated. TAM (45 mg/kg/day, i.p., for 10 consecutive days) resulted in an elevation of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP), depletion of liver reduced glutathione (GSH) and accumulation of oxidized glutathione (GSSG) and lipid peroxidation (LPO). Also, TAM treatment resulted in inhibition of hepatic activity of glutathione reductase (GR), glutathione peroxidase (GPx), superoxide dismutase (SOD) and catalase (CAT). Further, it raised liver tumor necrosis factor-alpha (TNF-alpha) level and induced histopathological changes. Pretreatment with CAPE (2.84 mg/kg/day; i.p., for 20 consecutive days, starting 10 days before TAM injection) significantly prevented the elevation in serum activity of the assessed enzymes. CAPE significantly inhibited TAM-induced hepatic GSH depletion and GSSG and LPO accumulation. Consistently, CAPE normalized the activity of GR, GPx, SOD and CAT, inhibited the rise in TNF-alpha and ameliorated the histopathological changes. In conclusion, CAPE protects against TAM-induced hepatotoxicity.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/toxicidade , Ácidos Cafeicos/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Álcool Feniletílico/análogos & derivados , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/toxicidade , Animais , Antioxidantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Feminino , Glutationa/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Testes de Função Hepática , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Álcool Feniletílico/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Fator de Necrose Tumoral alfa/metabolismo , Xantina Oxidase/metabolismo
10.
Mol Endocrinol ; 20(5): 996-1008, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16455819

RESUMO

The antiestrogen tamoxifen has been widely used for decades as selective estrogen receptor (ER) modulator for ERalpha-positive breast tumors. Tamoxifen significantly reduces tumor recurrence by binding to the activation function-2 (AF-2) domain of the ER. Acquired resistance to tamoxifen in breast cancer patients is a serious therapeutic problem. Antiestrogen-resistant breast cancer often shows increased expression of the epidermal growth factor receptor (EGFR) family members, EGFR and ErbB2. In this report we now show that overexpression of EGFR or activated AKT-2 in MCF-7 cells leads to phosphorylation of Ser167 in the AF-1 domain of ERalpha, enhanced ER-amplified in breast cancer 1 (ER:AIB1) interaction in the presence of tamoxifen, and resistance to tamoxifen. In contrast, transfection of activated MAPK kinase, an immediate upstream activator of MAPK (ERK 1 and 2) into MCF-7 cells leads to phosphorylation of Ser118 in the AF-1 domain of ERalpha, inhibition of ER-amplified in breast cancer 1 (ER:AIB1) interaction in the presence of Tam, and maintenance of sensitivity to tamoxifen. Inhibition of AKT by short inhibitory RNA blocked Ser167 phosphorylation in ER and restored tamoxifen sensitivity. However, maximum sensitivity to tamoxifen was observed when both AKT and MAPK were inhibited. Taken together, these data demonstrate that different phosphorylation sites in the AF-1 domain of ERalpha regulate the agonistic and antagonistic actions of tamoxifen in human breast cancer cells.


Assuntos
Antineoplásicos Hormonais/agonistas , Antineoplásicos Hormonais/antagonistas & inibidores , Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Tamoxifeno/agonistas , Tamoxifeno/antagonistas & inibidores , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/antagonistas & inibidores , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Oncogênicas v-erbB/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Serina/metabolismo , Tamoxifeno/uso terapêutico , Transfecção
11.
Nutr Cancer ; 52(1): 43-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16091003

RESUMO

The prevention of breast cancer through dietary modification is an active area of clinical and epidemiological research. It has been proposed that dietary supplementation of vitamin E may reduce a woman's risk of developing breast cancer. However, the exact mechanism remains unknown. alpha-Tocopherol is the most biologically active form of vitamin E. We investigated the effect of vitamin E (alpha-tocopherol) on breast cancer cell growth. A dose-dependent inhibition of cell proliferation was found in estrogen receptor (ER)-positive cells showing a potent suppression of growth at 100 microM vitamin E in MCF-7 (53%) and T47D (75%) cells. Vitamin E reduced significantly the response of both cell lines to estrogen (10 nM), and cell proliferation was decreased in MCF-7 and T47D cells by 69% and 84%, respectively. No growth inhibition was observed when cells were grown in the absence of estrogen. Vitamin E altered and decreased the growth inhibition induced by tamoxifen (10 microM) in MCF-7 (33%) and T47D (54%) cells. In addition, the immunostaining of ER of MCF-7 cells was reduced by 30% in the presence of vitamin E, suggesting an effect of vitamin E on the expression of ER. This provides evidence that vitamin E may inhibit ER-positive cell growth by altering the cellular response to estrogen.


Assuntos
Neoplasias da Mama/prevenção & controle , Divisão Celular/efeitos dos fármacos , Estrogênios/metabolismo , Receptores de Estrogênio/metabolismo , Vitamina E/administração & dosagem , Vitaminas/administração & dosagem , Antineoplásicos Hormonais/antagonistas & inibidores , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/metabolismo , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Feminino , Humanos , Tamoxifeno/antagonistas & inibidores , Células Tumorais Cultivadas , Vitamina E/farmacologia , Vitaminas/farmacologia , alfa-Tocoferol/administração & dosagem , alfa-Tocoferol/farmacologia
12.
Breast Cancer Res ; 6(3): 119-27, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15084232

RESUMO

Phytoestrogens are polyphenol compounds of plant origin that exhibit a structural similarity to the mammalian steroid hormone 17beta-oestradiol. In Asian nations the staple consumption of phyto-oestrogen-rich foodstuffs correlates with a reduced incidence of breast cancer. Human dietary intervention trials have noted a direct relationship between phyto-oestrogen ingestion and a favourable hormonal profile associated with decreased breast cancer risk. However, these studies failed to ascertain the precise effect of dietary phyto-oestrogens on the proliferation of mammary tissue. Epidemiological and rodent studies crucially suggest that breast cancer chemoprevention by dietary phyto-oestrogen compounds is dependent on ingestion before puberty, when the mammary gland is relatively immature. Phyto-oestrogen supplements are commercially marketed for use by postmenopausal women as natural and safe alternatives to hormone replacement therapy. Of current concern is the effect of phyto-oestrogen compounds on the growth of pre-existing breast tumours. Data are contradictory, with cell culture studies reporting both the oestrogenic stimulation of oestrogen receptor-positive breast cancer cell lines and the antagonism of tamoxifen activity at physiological phyto-oestrogen concentrations. Conversely, phyto-oestrogen ingestion by rodents is associated with the development of less aggressive breast tumours with reduced metastatic potential. Despite the present ambiguity, current data do suggest a potential benefit from use of phyto-oestrogens in breast cancer chemoprevention and therapy. These aspects are discussed.


Assuntos
Anticarcinógenos/uso terapêutico , Neoplasias da Mama/prevenção & controle , Inibidores Enzimáticos/uso terapêutico , Isoflavonas/uso terapêutico , Preparações de Plantas/uso terapêutico , Adolescente , Adulto , Fatores Etários , Idoso , Animais , Anticarcinógenos/química , Anticarcinógenos/farmacologia , Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/uso terapêutico , Ásia/epidemiologia , Ásia/etnologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/epidemiologia , Criança , Emigração e Imigração , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Estrogênios , Comportamento Alimentar , Feminino , Humanos , Lactente , Isoflavonas/efeitos adversos , Isoflavonas/química , Isoflavonas/farmacologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Pessoa de Meia-Idade , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/prevenção & controle , Fitoestrógenos , Preparações de Plantas/efeitos adversos , Preparações de Plantas/química , Preparações de Plantas/farmacologia , Pós-Menopausa , Pré-Menopausa , Prevalência , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Estados Unidos/epidemiologia
14.
Am Surg ; 68(6): 575-7; discussion 577-8, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12079141

RESUMO

Tamoxifen is an antiestrogen used in the treatment of estrogen receptor-positive breast cancer in postmenopausal women. It functions by competitively inhibiting the estrogen receptor and inducing apoptosis and G1 cell cycle arrest. Genistein is a soy phytoestrogen that inhibits breast cancer cell growth in vitro at doses of 10 microM or above. At lower doses genistein may stimulate cell growth and entry into the cell cycle. We hypothesized that treatment with low-dose genistein would reverse the inhibitory effects of tamoxifen in estrogen-receptor-positive breast cancer cells. Cell cycle kinetics and cell proliferation in T47-D human breast cancer cells were examined after exposure to genistein and tamoxifen in a low-estrogen environment designed to mimic a post-menopausal state. Cell proliferation was assessed by a colorimetric assay. Cell cycle kinetics were determined by flow cytometry. Tamoxifen caused G1 arrest and a decrease in proliferation. Genistein reversed the inhibitory effects of tamoxifen on both proliferation and G1 arrest. Thus low-dose genistein was able to inhibit the therapeutic effects of tamoxifen in this postmenopausal model of breast cancer.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Moduladores de Receptor Estrogênico/farmacologia , Genisteína/farmacologia , Inibidores do Crescimento/farmacologia , Tamoxifeno/antagonistas & inibidores , Neoplasias da Mama/metabolismo , Relação Dose-Resposta a Droga , Feminino , Humanos , Pós-Menopausa , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Células Tumorais Cultivadas
15.
Endocrinology ; 143(3): 853-67, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11861507

RESUMO

Short heterodimer partner (SHP) is an orphan nuclear receptor that interacts with ER(alpha) and ERbeta and inhibits E2-induced transcription. We examined how SHP affects tamoxifen's estrogen agonist activity in endometrial cells. We report that SHP interacts with 4-hydroxytamoxifen (4-OHT) or E2-occupied ER(alpha) in a temperature-dependent manner in vitro. In transient transfection assays, SHP inhibited 4-OHT-stimulated reporter gene activity from an estrogen response element (ERE) in ER-positive RL95-2 but not in HEC-1A human endometrial carcinoma cells transfected with ER(alpha) or ERbeta. SHP inhibited E2-induced transcriptional activity in ER(alpha)- or ERbeta-transfected HEC-1A or Chinese hamster ovary-K1 cells. SHP inhibition of E2 activity was greater for ER(alpha) than ERbeta from the nonpalindromic ERE in the pS2 gene promoter in Chinese hamster ovary-K1 but not HEC-1A cells. Thus, ER subtype, cell type, and ERE sequence influence SHP repressor activity. An ER(alpha) mutant lacking activator function-1 showed reduced inhibition by SHP. In glutathione S-transferase pull-down experiments, SHP inhibited ER(alpha) dimerization, providing a possible mechanism to account for the inhibitory effect of SHP on ER activity. These results identify SHP as novel target for blocking 4-OHT agonist activity in endometrial cells.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Neoplasias do Endométrio/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/fisiologia , Tamoxifeno/análogos & derivados , Tamoxifeno/antagonistas & inibidores , Animais , Antineoplásicos Hormonais/farmacologia , Western Blotting , Células CHO , Cricetinae , Estradiol/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Genes Reporter/genética , Glutationa Transferase/metabolismo , Humanos , Plasmídeos/genética , Receptores de Estrogênio/antagonistas & inibidores , Tamoxifeno/farmacologia , Transfecção , Células Tumorais Cultivadas
16.
Anticancer Res ; 22(6C): 4151-6, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12553047

RESUMO

The effects of glucocorticoid (GC) on the proliferation of Dunn Osteosarcoma (OS) cells were examined under in vitro culture conditions. Dexamethasone (Dex) inhibited the proliferation of Dunn OS cells in a dose-dependent manner, while the addition of anti-GC, RU486, to the culture medium in part recovered Dex-induced growth inhibition. The number of maximum binding sites (Bmax) and the dissociation constant (Kd) value of glucocorticoid receptor (GR) in Dunn OS cells were 19,560 sites/cell and 5.2 +/- 0.8 nM, respectively. RU486 competed with labeled Dex against GR at a concentration of 10(-6) M. Western blot analysis of [3H]Dex-mesylate-labeled cell homogenate and immunohistochemical staining against GR further confirmed the presence of GR. Dex treatment of Dunn OS cells resulted in apoptosis with the characteristic internucleosomal DNA cleavage shown by the DNA ladder pattern in agarose gel electrophoresis. These data demonstrate that GC inhibits the proliferation of Dunn OS cells via GR, for which one possible mechanism in vitro is induction of apoptosis.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Dexametasona/farmacologia , Osteossarcoma/tratamento farmacológico , Animais , Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/metabolismo , Apoptose/efeitos dos fármacos , Sítios de Ligação , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Divisão Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Dexametasona/antagonistas & inibidores , Dexametasona/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores do Crescimento/metabolismo , Inibidores do Crescimento/farmacologia , Antagonistas de Hormônios/farmacologia , Cinética , Camundongos , Mifepristona/farmacologia , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Receptores de Glucocorticoides/metabolismo , Trítio , Células Tumorais Cultivadas
17.
Cancer Res ; 61(23): 8452-8, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11731427

RESUMO

In experimental models, human epidermal growth factor receptor-2 (HER-2) amplification leads to estrogen independence and tamoxifen resistance in estrogen receptor (ER)-positive human breast cancer cells. Some but not all reports suggest an association between HER-2 positivity and hormone independence in breast cancer patients. This study aimed to evaluate the antiproliferative effects of endocrine therapy in HER-2-positive/ER-positive primary human breast cancer. The effect on proliferation (Ki67) of hormone therapy was assessed at 2 weeks and/or 12 weeks in biopsies from 115 primary breast cancers with ER-positive tumors. The patients took part in one of 3 neoadjuvant trials of hormonal therapy with a SERM (tamoxifen or idoxifene) or an aromatase inhibitor (anastrozole or vorozole). HER-2 status was assessed by immunocytochemistry and fluorescence in situ hybridization (FISH). Fifteen patients were defined as HER-2 positive by both immunohistochemistry and FISH, with the remaining 100 patients HER-2 negative. Geometric mean Ki67 levels were substantially higher in HER-2-positive than HER-2-negative tumors (27.7% versus 11.5%, respectively; P = 0.003). In HER-2-negative patients, Ki67 was reduced by 62 and 71% at 2 and 12 weeks, respectively (P < 0.0001 for both), but HER-2-positive patients showed no significant fall. The proportional change in Ki67 was significantly different between HER-2-positive and -negative patients (P = 0.014 at 2 weeks; P = 0.047 at 12 weeks). Mean ER levels were lower in the HER-2-positive patients (P = 0.06) but the change in Ki67 was impeded even in those with high ER. Apoptotic index was reduced by 30% at 2 weeks in the HER-2-negative group. However, there were no statistically significant differences in apoptotic index between the groups. It is concluded that ER-positive/HER-2-positive primary breast carcinomas show an impeded antiproliferative response to endocrine therapy that nonetheless may vary between individual treatments. This together with high baseline proliferation is likely to translate to poor clinical response.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Receptor ErbB-2/biossíntese , Receptores de Estrogênio/biossíntese , Tamoxifeno/análogos & derivados , Anastrozol , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/genética , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Feminino , Amplificação de Genes , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Estudos Multicêntricos como Assunto , Nitrilas/antagonistas & inibidores , Nitrilas/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptor ErbB-2/genética , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/farmacologia , Triazóis/antagonistas & inibidores , Triazóis/farmacologia
18.
Res Commun Mol Pathol Pharmacol ; 109(5-6): 275-86, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12889511

RESUMO

Tamoxifen is known to induce hepatocarcinogenesis in experimental animals and reversible chronic liver diseases in humans. Melatonin has been recently introduced as an oncostatic agent, especially for hormone-dependent tumors. This study was designed in order to investigate whether melatonin has an effect onthe tamoxifen-induced hepatotoxicity. Wistar albino rats were injected tamoxifen citrate intraperitoneally in three different doses (10 mg/kg and 20 mg/kg bw for 26 days; and 45 mg/kg bw for three days). Another group of animals were treated with melatonin once a week in addition to daily tamoxifen injections, whereas the third group received melatonin only. The control animals were injected an equal volume of diluent at corresponding intervals. At the end of the experimental period, the animals were sacrificed and the livers were prepared for the flow cytometric DNA analysis. DNA histograms were analyzed using the multicycle program. In experimental groups, all animals had aneuploid cell population. The difference in the diploid/ aneuploid ratio of each experimental group as compared to the control group according to Fischer's exact test was found to be highly significant (p < 0.002 MEL vs control; and p < 0.0001 for both TAM vs control and MEL+TAM vs control). Among the tamoxifen-injected animals, the proportion of multiploidy to aneuploid cell population was 17, similar to those treated solely with melatonin. Although the melatonin plus tamoxifen group had higher multiploidy percentage (38%), the difference was not statistically significant as compared to the tamoxifen (or melatonin) groups. No significant difference was noted between the animals which were treated with three different doses of tamoxifen. S-phase fraction percentage was significantly different in melatonin- and melatonin plus tamoxifen-injected animals with regard to controls, the degree of significancy being < 0.05 for both. According to our data, tamoxifen injections induced DNA aneuploidy, but did not stimulate proliferation in the liver as estimated by S-phase fraction. Melatonin, whether alone or in combination with tamoxifen, stimulated cell proliferation and produced aneuploidy.


Assuntos
Aneuploidia , Antineoplásicos Hormonais/toxicidade , Antioxidantes/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Melatonina/uso terapêutico , Tamoxifeno/toxicidade , Animais , Antineoplásicos Hormonais/antagonistas & inibidores , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Feminino , Citometria de Fluxo , Ploidias , Ratos , Ratos Wistar , Tamoxifeno/antagonistas & inibidores
19.
Eur J Cancer ; 36 Suppl 4: S73, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11056327

RESUMO

Tangeretin, a molecule present in citrus fruits and in certain 'natural' menopausal medications, is an effective tumour growth and invasion inhibitor in vitro of human MCF 7/6 breast cancer cells. However, when added to the drinking water of MCF 7/6 tumour-bearing mice it neutralises the beneficial tumour-suppressing effect of tamoxifen. Tangeretin reduces the number of natural killer cells. This may explain why the beneficial suppressive effect of tangeretin on MCF 7/6 cell proliferation in vitro is completely counteracted in vivo.


Assuntos
Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos/farmacologia , Flavonas , Flavonoides/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Tamoxifeno/antagonistas & inibidores , Animais , Antineoplásicos Hormonais/uso terapêutico , Feminino , Interações Alimento-Droga , Camundongos , Tamoxifeno/uso terapêutico
20.
Cancer Res ; 60(21): 5995-6000, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085519

RESUMO

Tamoxifen (TAM) is widely used in the treatment of breast cancer. The cytostatic effects of TAM have been attributed to the antagonism of estrogen receptor (ER) and inhibition of estrogen-dependent proliferative events. However, the mechanism by which TAM is also effective against certain ER-negative breast tumors remains to be elucidated. Here we report that TAM induced the activity of caspase-3-like proteases in ER-negative breast cancer cell lines MDA-MB-231 and BT-20, as evidenced by the cleavage of fluorogenic tetrapeptide substrate and of poly(ADP-ribose) polymerase. The activation of caspase-3-like proteases preceded TAM-induced chromatin condensation and nuclear fragmentation, the typical apoptotic morphologies. Pretreatment of cells with a specific inhibitor of caspase-3, acetyl-Asp-Glu-Val-Asp-aldehyde, or with a general inhibitor of caspases, benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, prevented TAM-induced apoptosis. TAM also stimulated c-Jun NH2-terminal kinase (JNK) 1 activity, and interfering with the JNK pathway by over-expressing a DN JNK1 mutant attenuated TAM-induced apoptosis. In addition, treatment of cells with a lipid-soluble antioxidant vitamin E blocked TAM-induced caspase-3 and JNK1 activation as well as apoptosis, whereas water-soluble antioxidants N-acetyl L-cysteine and glutathione had little effect. Thus, this study demonstrates that TAM induces apoptosis in ER-negative breast cancer cells through caspase-3 and JNK1 pathways, which are probably initiated at the cell membrane by an oxidative mechanism.


Assuntos
Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/enzimologia , Caspases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Tamoxifeno/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Antineoplásicos Hormonais/antagonistas & inibidores , Antineoplásicos Hormonais/toxicidade , Apoptose/fisiologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Caspase 3 , Inibidores de Caspase , Inibidores de Cisteína Proteinase/farmacologia , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Oligopeptídeos/farmacologia , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/toxicidade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...