Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 365
Filter
3.
Br J Pharmacol ; 171(17): 4062-72, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24819221

ABSTRACT

BACKGROUND AND PURPOSE: Triphenylethylene (TPE)-like compounds were the first agents to be used in the treatment of metastatic breast cancer in postmenopausal women. Although structurally related to the anti-oestrogen, 4-hydroxytamoxifen, TPEs possess oestrogenic properties in fully oestrogenized breast cancer cells but do not induce apoptosis with short-term treatment in long-term oestrogen-deprived breast cancer cells. This study determined the differential effects of bisphenol, a TPE, on growth and apoptosis based on the modulation of the shape of the ligand-oestrogen receptor complex. EXPERIMENTAL APPROACH: Apoptotic flow cytometric studies were used to evaluate apoptosis over time. Proliferation of the breast cancer cells was assessed using DNA quantification and cell cycle analysis. Real-time PCR was performed to quantify mRNA levels of apoptotic genes. Regulation of cell cycle and apoptotic genes was determined using PCR-based arrays. KEY RESULTS: Bisphenol induced an up-regulation of cell cycle genes similar to those induced by 17ß oestradiol (E2 ). Unlike the changes induced by E2 that occur after 24 h, the apoptosis evoked by bisphenol occurred after 4 days, with quantifiable apoptotic changes noted at 6 days. A prolonged up-regulation of endoplasmic reticulum stress and inflammatory stress response genes was observed with subsequent activation of apoptosis-related genes in the second week of treatment with bisphenol. CONCLUSIONS AND IMPLICATIONS: The bisphenol: ERα complex induces delayed biological effects on the growth and apoptosis of breast cancer cells. Both the shape of the complex and the duration of treatment control the initiation of apoptosis.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Estradiol/pharmacology , Estrogens/pharmacology , Stilbenes/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Estradiol/chemistry , Female , Humans , MCF-7 Cells , Stilbenes/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
4.
Br J Cancer ; 110(6): 1488-96, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24548860

ABSTRACT

BACKGROUND: Oestrogen (E2) induces apoptosis in long-term E2-deprived MCF7 cells (MCF7:5C). Taxanes have been used extensively in the treatment of early and advanced breast cancer. We have interrogated the sequence of events that involve the apoptotic signalling pathway induced by E2 in comparison with paclitaxel. METHODS: DNA quantification and cell cycle analysis were used to assess proliferation of cancer cells. Apoptosis was evaluated using annexin V and DNA staining methods. Regulation of apoptotic genes was determined by performing PCR-based arrays and RT-PCR. RESULTS: E2-induced apoptosis is a delayed process, whereas paclitaxel immediately inhibits the growth and induces death of MCF7:5C cells. The cellular commitment for E2-triggered apoptosis occur after 24 h. Activation of the intrinsic pathway was observed by 36 h of E2 treatment with subsequent induction of the extrinsic apoptotic pathway by 48 h. Paclitaxel exclusively activated extramitochondrial apoptotic genes and caused rapid G2/M blockade by 12 h of treatment. By contrast, E2 causes an initial proliferation with elevated S phase of cell cycles followed by apoptosis of the MCF7:5C cells. Most importantly, we are the first to document that E2-induced apoptosis can be reversed after 24 h treatment. CONCLUSIONS: These data indicate that E2-induced apoptosis involves a novel, multidynamic process that is distinctly different from that of a classic cytotoxic chemotherapeutic drug used in breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Estrogens/pharmacology , Paclitaxel/pharmacology , Breast Neoplasms/pathology , Cell Division/drug effects , Female , Humans , MCF-7 Cells
5.
Clin Pharmacol Ther ; 95(2): 216-27, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24060820

ABSTRACT

The International Tamoxifen Pharmacogenomics Consortium was established to address the controversy regarding cytochrome P450 2D6 (CYP2D6) status and clinical outcomes in tamoxifen therapy. We performed a meta-analysis on data from 4,973 tamoxifen-treated patients (12 globally distributed sites). Using strict eligibility requirements (postmenopausal women with estrogen receptor-positive breast cancer, receiving 20 mg/day tamoxifen for 5 years, criterion 1); CYP2D6 poor metabolizer status was associated with poorer invasive disease-free survival (IDFS: hazard ratio = 1.25; 95% confidence interval = 1.06, 1.47; P = 0.009). However, CYP2D6 status was not statistically significant when tamoxifen duration, menopausal status, and annual follow-up were not specified (criterion 2, n = 2,443; P = 0.25) or when no exclusions were applied (criterion 3, n = 4,935; P = 0.38). Although CYP2D6 is a strong predictor of IDFS using strict inclusion criteria, because the results are not robust to inclusion criteria (these were not defined a priori), prospective studies are necessary to fully establish the value of CYP2D6 genotyping in tamoxifen therapy.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Cytochrome P-450 CYP2D6/genetics , Tamoxifen/therapeutic use , Aged , Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/genetics , Female , Genetic Variation/genetics , Genotype , Humans , Menopause , Middle Aged , Pharmacogenetics/methods , Survival Analysis , Tamoxifen/pharmacokinetics , Treatment Outcome
6.
Br J Pharmacol ; 169(1): 167-78, 2013 May.
Article in English | MEDLINE | ID: mdl-23373633

ABSTRACT

BACKGROUND AND PURPOSE: Oestrogen receptor alpha (ERα) binds to different ligand which can function as complete/partial oestrogen-agonist or antagonist. This depends on the chemical structure of the ligands which modulates the transcriptional activity of the oestrogen-responsive genes by altering the conformation of the liganded-ERα complex. This study determined the molecular mechanism of oestrogen-agonistic/antagonistic action of structurally similar ligands, bisphenol (BP) and bisphenol A (BPA) on cell proliferation and apoptosis of ERα + ve breast cancer cells. EXPERIMENTAL APPROACH: DNA was measured to assess the proliferation and apoptosis of breast cancer cells. RT-PCR and ChIP assays were performed to quantify the transcripts of TFF1 gene and recruitment of ERα and SRC3 at the promoter of TFF1 gene respectively. Molecular docking was used to delineate the binding modes of BP and BPA with the ERα. PCR-based arrays were used to study the regulation of the apoptotic genes. KEY RESULTS: BP and BPA induced the proliferation of breast cancer cells; however, unlike BPA, BP failed to induce apoptosis. BPA consistently acted as an agonist in our studies but BP exhibited mixed agonistic/antagonistic properties. Molecular docking revealed agonistic and antagonistic mode of binding for BPA and BP respectively. BPA treatment resembled E2 treatment in terms of PCR-based regulation of apoptotic genes whereas BP was similar to 4OHT treatment. CONCLUSIONS AND IMPLICATIONS: The chemical structure of ERα ligand determines the agonistic or antagonistic biological responses by the virtue of their binding mode, conformation of the liganded-ERα complex and the context of the cellular function.


Subject(s)
Apoptosis/drug effects , Benzhydryl Compounds/pharmacology , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Phenols/pharmacology , Apoptosis/genetics , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/chemistry , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Female , Humans , MCF-7 Cells , Molecular Docking Simulation , Phenols/administration & dosage , Phenols/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
7.
Oncogene ; 32(27): 3274-85, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-22907432

ABSTRACT

Tamoxifen (TAM) has both cytostatic and cytotoxic properties for breast cancer. TAM engaged mitochondrial estrogen receptor beta (ERß) as an antagonist in MCF7-BK cells, increasing reactive oxygen species (ROS) concentrations from the mitochondria that were required for cytotoxicity. In part, this derived from TAM downregulating manganese superoxide dismutase (MnSOD) activity by causing the nitrosylation of tyrosine 34, thereby increasing ROS. ROS-activated protein kinase C delta and c-jun N-terminal kinases, resulting in the mitochondrial translocation of Bax and cytochrome C release. Interestingly, TAM failed to cause high ROS levels or induce cell death in MCF7-BK-TR cells due to stimulation of MnSOD activity through agonistic effects at mitochondrial ERß. In several mouse xenograft models, lentiviral shRNA-induced knockdown of MnSOD caused tumors that grew in the presence of TAM to undergo substantial apoptosis. Tumor MnSOD and mitochondrial ERß are therefore targets for therapeutic intervention to reverse TAM resistance and enhance a cell death response.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Estrogen Receptor beta/metabolism , Mitochondria/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Xenograft Model Antitumor Assays
8.
Br J Pharmacol ; 150(1): 3-4, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17115068

ABSTRACT

Selective oestrogen receptor modulators (SERMs) can build bone in the postmenopausal woman and lower circulating cholesterol. These oestrogen-like properties contrast with the anti-oestrogenic properties observed in the breast where SERMs inhibit the oestrogen-mediated development and growth of ER positive breast cancers. The two clinically useful SERMs, tamoxifen and its chemical cousin raloxifene, are currently used successfully either for the treatment and prevention of breast cancer (tamoxifen) or the treatment and prevention of osteoporosis (raloxifene). However, raloxifene has the beneficial side-effect of breast cancer prevention. These multifunction medicines provide proof of concept that novel molecules can be selectively targeted to diseases mediated by the endocrine system.


Subject(s)
Breast Neoplasms/prevention & control , Osteoporosis/prevention & control , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Humans
9.
J Steroid Biochem Mol Biol ; 94(1-3): 131-41, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15862958

ABSTRACT

Estrogen suppression through the use of an aromatase inhibitor is an effective endocrine treatment option for postmenopausal breast cancer patients with estrogen receptor (ER)-positive disease, however, there are concerns that long-term estrogen deprivation will inevitably lead to resistance. To address the issue of acquired resistance to long-term estrogen deprivation our laboratory has developed an ER+/PR- hormone-independent breast cancer cell line, MCF-7:5C which is a variant clone of wild-type MCF-7 cells. Originally, these cells were cultured in estrogen-free MEM containing 5% charcoal-stripped calf serum and were found to be resistant to both estradiol (E(2)) and antiestrogens. Interestingly, a completely different phenomenon was observed when MCF-7:5C cells were cultured in phenol red-free RPMI 1640 medium containing 10% charcoal-stripped fetal bovine serum (SFS). Using DNA quantitation assays, we examined the effect of E(2) on the growth of MCF-7:5C cells under different media conditions. Our results showed that 10(-9)M E(2) caused a dramatic 90% reduction in the growth of MCF-7:5C cells cultured in RPMI medium containing 10% SFS but did not have any significant inhibitory effects on cells cultured in MEM media. Additional experiments were performed to determine whether the medium or the serum facilitated the inhibitory effects of E(2) and the results indicated that it was the serum. Annexin V and DAPI staining confirmed that the E(2)-induced growth inhibition of MCF-7:5C cells was due to apoptosis. We also examined the tumorigenic potential of MCF-7:5C cells by injecting 1x10(7)cells/site into ovariectomized athymic mice and found that these cells, previously cultured in RPMI media, spontaneously grew into tumors in the absence of E(2). Overall, these results show that low concentrations (>10(-11)M) of E(2) are capable of inducing apoptosis in an aromatase resistant breast cancer cell model and that this effect is highly influenced by the medium in which the cells are grown.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Estrogens/pharmacology , Annexin A5/analysis , Cell Division/drug effects , Cell Line, Tumor , Culture Media , Estradiol/pharmacology , Female , Humans , Kinetics , Substance Withdrawal Syndrome
10.
Br J Cancer ; 90(5): 944-9, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-14997187

ABSTRACT

The link between sex steroids and the development and growth of breast cancer has proved to be an invaluable clue for advances in the prevention and treatment of breast cancer. The identification of the oestrogen receptor (ER) not only allowed advances in the molecular endocrinology of oestrogen action, but also provided a target for antioestrogenic therapeutic agents. However, the application of long-term or indefinite treatment regimens has consequences for the breast cancer. New forms of resistance, based upon enhanced cellular survival networks independent of ER and the suppression of apoptotic mechanisms, develop and then evolve. Remarkably, low concentrations of oestrogen collapse survival pathways and induce apoptosis in completely antihormonally refractory breast cancer. However, recurrent oestrogen-stimulated disease is again sensitive to antihormonal therapy. The novel reapplication of the ER as a therapeutic target for apoptosis is emerging as a new strategy for the long-term targeted maintenance treatment of breast cancer, and in formulating a targeted strategy for endocrine independent cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Estrogens/therapeutic use , Neoplasms, Hormone-Dependent/metabolism , Neoplasms/metabolism , Receptors, Estrogen/drug effects , Animals , Cell Division/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans
11.
Breast ; 12(6): 432-41, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14659118

ABSTRACT

The oestrogen receptor (ER) has proven to be an extraordinarily successful target for breast cancer treatment and prevention. The clinical use of tamoxifen, a nonsteroidal antioestrogen, demonstrated (1) that the strategic use of adjuvant tamoxifen in ER-positive patients could save lives and (2) that a selective ER modulator (SERM) could reduce the incidence of breast cancer in high-risk women. The ER is now the target for new and safer therapies such as the aromatase inhibitors and the pure antioestrogens that either block oestrogen synthesis or destroy the ER. However, the use of raloxifene, a SERM to prevent osteoporosis with the potential to prevent breast cancer has introduced a new dimension into preventive oncology. The widespread use of endocrine modulators (SERMs, aromatase inhibitors, and pure antioestrogens) raised the question of drug resistance. It is now clear that endocrine resistance can evolve through stages. Once a breast tumour becomes resistant to SERMs, the growth is stimulated by either the SERM or oestrogen. This is why an aromatase inhibitor is effective following SERM resistance and withdrawal. However, the extended use of repeated endocrine therapies now supersensitized the cells to oestrogen that causes apoptosis through the ER. We suggest that future clinical treatment strategies incorporate an 'oestrogen purge' to both enhance the actions of chemotherapy or completely reverse endocrine resistance and restore endocrine sensitivity. These new data build on the idea that breast cancer can be controlled as a chronic disease and will permit patients to live long and productive lives during targeted maintenance treatment.


Subject(s)
Breast Neoplasms/physiopathology , Receptors, Estrogen/physiology , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Drug Resistance , Female , Humans , Phosphorylation , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
13.
Eur J Cancer ; 39(7): 891-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12706357

ABSTRACT

The aim of this study was to determine the effects of low doses of tamoxifen (5 and 10mg/day) for 50 days compared with the standard dose (20 mg/day) on breast biomarkers measured in normal breast tissue from premenopausal patients. A randomised double-blind study was performed using tissue from 56 premenopausal women with a diagnosis of fibroadenoma of the breast. Excisional biopsy was performed on the 50th day of therapy. Normal breast tissue samples were collected during surgery. The patients were divided in groups: A (placebo, n=11); group B (5 mg, n=16), group C (10 mg, n=14) and group D (20 mg, n=15). In this cross-sectional study, differences in the expression of Oestrogen Receptor alpha (ERalpha), Progesterone Receptor (PR), Ki-67, apoptotic bodies and mitotic index between the different groups after treatment can be seen on the normal breast tissue. We believe that a lower dose of tamoxifen could reduce the side-effects associated with treatment without affecting its chemopreventive activity in the breast.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Biomarkers, Tumor/analysis , Breast/drug effects , Tamoxifen/administration & dosage , Adolescent , Adult , Apoptosis , Breast/chemistry , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fibroadenoma/drug therapy , Fibroadenoma/pathology , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Mitosis , Premenopause , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
14.
Br J Cancer ; 87(4): 449-56, 2002 Aug 12.
Article in English | MEDLINE | ID: mdl-12177783

ABSTRACT

The purpose of this study was to classify selective oestrogen receptor modulators based on gene expression profiles produced in breast cancer cells expressing either wtERalpha or mutant(351)ERalpha. In total, 54 microarray experiments were carried out by using a commercially available Atlas cDNA Expression Arrays (Clontech), containing 588 cancer-related genes. Nine sets of data were generated for each cell line following 24 h of treatment: expression data were obtained for cells treated with vehicle EtOH (Control); with 10(-9) or 10(-8) M oestradiol; with 10(-6) M 4-hydroxytamoxifen; with 10(-6) M raloxifene; with 10(-6) M idoxifene, with 10(-6) M EM 652, with 10(-6) M GW 7604; with 5 x 10(-5) M resveratrol and with 10(-6) M ICI 182,780. We developed a new algorithm 'Expression Signatures' to classify compounds on the basis of differential gene expression profiles. We created dendrograms for each cell line, in which branches represent relationships between compounds. Additionally, clustering analysis was performed using different subsets of genes to assess the robustness of the analysis. In general, only small differences between gene expression profiles treated with compounds were observed with correlation coefficients ranged from 0.83 to 0.98. This observation may be explained by the use of the same cell context for treatments with compounds that essentially belong to the same class of drugs with oestrogen receptors related mechanisms. The most surprising observation was that ICI 182,780 clustered together with oestrodiol and raloxifene for cells expressing wtERalpha and clustered together with EM 652 for cells expressing mutant(351)ERalpha. These data provide a rationale for a more precise and elaborate study in which custom made oligonucleotide arrays can be used with comprehensive sets of genes known to have consensus and putative oestrogen response elements in their promoter regions.


Subject(s)
Breast Neoplasms/genetics , Receptors, Estrogen/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Receptors, Estrogen/metabolism , Tumor Cells, Cultured
15.
Gynecol Oncol ; 85(3): 498-506, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12051881

ABSTRACT

OBJECTIVE: Tamoxifen has mixed agonist/antagonist activities, leading to tissue-specific estrogen-like actions and endometrial cancer. The purpose of this study was to evaluate the effects of antiestrogens on the growth of estrogen receptor (ER)-positive ECC-1 endometrial cancer cells in vitro and in vivo. METHODS: We performed growth studies and luciferase assays using ERE-tK and AP-1 reporters. ERalpha protein expression was measured by Western blot after antiestrogen treatments. We investigated the actions of antiestrogens on the transcription of the pS2 gene in situ measured by Northern blot and the actions of antiestrogens on the VEGF protein secreted by ELISA. ERalpha, ERbeta, EGFR, and HER2/neu mRNAs were determined by RT-PCR. Last, ECC-1 tumors were developed by inoculation of cells into ovariectomized athymic mice and treated with estradiol (E2), tamoxifen, raloxifene, and a combination. RESULTS: E2 induced cell proliferation while antiestrogens did not. E2 and raloxifene down regulated ERalpha protein; in contrast, 4OHT did not. ICI182,780 completely degraded the receptor. ECC-1 cells express ERbeta at insignificant levels. Luciferase assays did not show any induction in ERE- nor AP-1-mediated transcription by antiestrogens. E2 caused a concentration-dependent increase in pS2 mRNA but antiestrogens did not. E2 increased VEGF expression in a dose-dependent manner and antiestrogens blocked E2 action. E2 down regulated HER2/neu while 4OHT and raloxifene did not change HER2/neu levels compared to control. In addition, EGFR mRNA was down regulated by E2 but raloxifene did not change it. Tamoxifen and raloxifene did not promote tumor growth in vivo. However, raloxifene (1.5 mg daily) only partially blocked E2-stimulated growth. CONCLUSION: Tamoxifen and raloxifene are antiproliferative agents and antiestrogens in ECC-1 endometrial cells in vitro and in vivo. The observation that selective estrogen-receptor modulators do not down regulate EGFR and HER2/neu mRNA may provide a potential role for these oncogenes in the development of raloxifene- or tamoxifen-stimulated endometrial cancer. The ECC-1 cell line could provide important new clues about the evolution of drug resistance to tamoxifen and raloxifene.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Selective Estrogen Receptor Modulators/pharmacology , Adenocarcinoma/metabolism , Animals , Blotting, Northern , Cell Division/drug effects , Endometrial Neoplasms/metabolism , Endothelial Growth Factors/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Luciferases/biosynthesis , Luciferases/genetics , Luciferases/metabolism , Lymphokines/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raloxifene Hydrochloride/pharmacology , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology , Transcription, Genetic/drug effects , Trefoil Factor-1 , Tumor Suppressor Proteins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Clin Cancer Res ; 7(12): 4149-55, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751515

ABSTRACT

PURPOSE: Arzoxifene (Arzox) is a novel benzothiophene analogue with selective estrogen receptor modulator activity similar to raloxifene. Arzox is being developed as a treatment for breast cancer and has a predominantly antiestrogenic effect on the rodent uterus. Our objectives were to verify whether the novel selective estrogen receptor modulator, Arzox, can be a good first-line agent and also be effective at controlling the growth of endometrial cancer after exposure to tamoxifen (Tam). EXPERIMENTAL DESIGN: We compared the effects of Tam and Arzox on the growth of estrogen responsive ECC-1 endometrial cancer cells in vitro, and we determined their antitumor effects on ECC-1 and EnCa101 endometrial carcinoma growth in athymic mice. RESULTS: We observed that estrogen receptor protein expression is down-regulated by Arzox to the same extent as raloxifene, whereas 4-hydroxytamoxifen, the active metabolite of Tam, does not affect estrogen receptor protein levels. Tam and Arzox inhibit the growth of Tam-naïve ECC-1 tumors in athymic mice. However when Tam-stimulated or estrogen-stimulated (which had been treated with Tam previously) EnCa101 endometrial tumors were treated with Tam or Arzox, we observed a stimulatory effect of both compounds in these models. CONCLUSIONS: The results indicate that Arzox may be a good first-line agent, but it may be ineffective at controlling the growth of endometrial cancer after exposure to Tam. Our data suggest that Arzox stimulates endometrial tumor growth to at least the same extent as Tam, thereby suggesting a limited role as a second-line agent for the patient on Tam who develops occult endometrial cancer.


Subject(s)
Endometrial Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Piperidines/therapeutic use , Tamoxifen/analogs & derivatives , Thiophenes/therapeutic use , Animals , Cell Division/drug effects , Down-Regulation/drug effects , Endometrial Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Receptors, Estrogen/genetics , Tamoxifen/therapeutic use , Transplantation, Heterologous , Tumor Cells, Cultured
19.
Clin Cancer Res ; 7(10): 3156-65, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595710

ABSTRACT

PURPOSE: Resistance to tamoxifen (TAM) represents a significant challenge to the management of breast cancer. We previously reported that the estrogen receptor (ER)-negative hormone-independent T47D:C42 cell line has both elevated protein kinase Calpha (PKCalpha) protein expression and basal activator protein-1 activity compared with the parental ER+ (hormone-dependent) T47D:A18 cell line. Stable transfection of PKCalpha to the T47D:A18 breast cancer cell line results in increased basal activator protein-1 activity, reduced ER function, increased proliferation rate, and hormone-independent growth (Tonetti et al., Br. J. Cancer, 83: 782-791, 2000). In this report, we further characterize the role of PKCalpha overexpression in vivo to elucidate a possible molecular mechanism of tamoxifen resistance. EXPERIMENTAL DESIGN: To determine whether the T47D:A18/PKCalpha cell line would produce hormone-independent tumors in athymic mice, we injected T47D:A18, T47D:A18/neo, or the T47D:A18/PKCalpha20 cell clones bilaterally into the mammary fat pads of athymic mice. Tumor growth was evaluated following treatment with estradiol (E2), TAM, and the pure antiestrogen, ICI 182,780. RESULTS: Mice receiving either T47D:A18 or T47D:A18/neo cells produced tumors that grew in response to E2 treatment, whereas the untreated control and TAM-treated groups showed no tumor growth. Interestingly, mice receiving the T47D:A18/PKCalpha20 clone produced tumors in both the control and TAM groups, whereas tumor growth was inhibited in mice treated with E2. PKCalpha was also overexpressed in an MCF-7 tumor model that also exhibited TAM-stimulated and E2-induced regression. CONCLUSIONS: These results suggest that overexpression of PKCalpha in breast tumors results in hormone-independent tumor growth that cannot be inhibited by TAM treatment. Furthermore, the finding that E2 has an antitumor effect on breast tumors overexpressing PKCalpha is a novel observation that may have important therapeutic implications.


Subject(s)
Antineoplastic Agents/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Isoenzymes/metabolism , Mammary Neoplasms, Experimental/prevention & control , Protein Kinase C/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Division/drug effects , Estradiol/therapeutic use , Estrogen Antagonists/pharmacology , Estrogen Antagonists/therapeutic use , Estrogen Receptor alpha , Female , Fulvestrant , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/prevention & control , Protein Kinase C-alpha , Receptors, Estrogen/metabolism , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Xenograft Model Antitumor Assays
20.
J Natl Cancer Inst ; 93(19): 1449-57, 2001 Oct 03.
Article in English | MEDLINE | ID: mdl-11584060

ABSTRACT

The recognition of selective estrogen receptor modulation in the laboratory has resulted in the development of two selective estrogen receptor modulators (SERMs), tamoxifen and raloxifene, for clinical application in healthy women. SERMs are antiestrogenic in the breast but estrogen-like in the bones and reduce circulating cholesterol levels. SERMs also have different degrees of estrogenicity in the uterus. Tamoxifen is used specifically to reduce the incidence of breast cancer in premenopausal and postmenopausal women at risk for the disease. In contrast, raloxifene is used specifically to reduce the risk of osteoporosis in postmenopausal women at high risk for osteoporosis. The study of tamoxifen and raloxifene (STAR) trial is currently comparing the ability of these SERMs to reduce breast cancer incidence in high-risk postmenopausal women. There is intense interest in understanding the molecular mechanism(s) of action of SERMs at target sites in a woman's body. An understanding of the targeted actions of this novel drug group will potentially result in the introduction of new multifunctional medicines with applications as preventive agents or treatments of breast cancer and endometrial cancer, coronary heart disease, and osteoporosis.


Subject(s)
Breast Neoplasms/prevention & control , Coronary Disease/prevention & control , Osteoporosis/prevention & control , Selective Estrogen Receptor Modulators/therapeutic use , Adult , Aged , Bone and Bones/drug effects , Breast/drug effects , Breast Neoplasms/chemistry , Breast Neoplasms/epidemiology , Cardiovascular System/drug effects , Cinnamates/pharmacology , Clinical Trials as Topic , Coronary Disease/epidemiology , Endometrial Neoplasms/chemically induced , Estrogen Replacement Therapy , Female , Heart/drug effects , Hot Flashes/chemically induced , Humans , Middle Aged , Models, Biological , Organ Specificity , Osteoporosis/epidemiology , Postmenopause , Premenopause , Prospective Studies , Protein Structure, Tertiary/drug effects , Raloxifene Hydrochloride/adverse effects , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Randomized Controlled Trials as Topic , Receptors, Estrogen/analysis , Receptors, Estrogen/drug effects , Risk , Risk Assessment , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/pharmacology , Stilbenes/pharmacology , Tamoxifen/adverse effects , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Thrombophilia/chemically induced , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...