Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 292
Filter
1.
Cancer Res ; 79(20): 5342-5354, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31481501

ABSTRACT

Obese women have higher risk of bearing breast tumors that are highly aggressive and resistant to therapies. Tumor-promoting effects of obesity occur locally via adipose inflammation and related alterations to the extracellular matrix (ECM) as well as systemically via circulating metabolic mediators (e.g., free fatty acids, FFA) associated with excess adiposity and implicated in toll-like receptor-mediated activation of macrophages-key cellular players in obesity-related cancer progression. Although the contribution of macrophages to proneoplastic effects of obesity is well documented, the role of ECM components and their enzymatic degradation is less appreciated. We show that heparanase, the sole mammalian endoglucuronidase that cleaves heparan sulfate in ECM, is preferentially expressed in clinical/experimental obesity-associated breast tumors. Heparanase deficiency abolished obesity-accelerated tumor progression in vivo. Heparanase orchestrated a complex molecular program that occurred concurrently in adipose and tumor tissue and sustained the cancer-promoting action of obesity. Heparanase was required for adipose tissue macrophages to produce inflammatory mediators responsible for local induction of aromatase, a rate-limiting enzyme in estrogen biosynthesis. Estrogen upregulated heparanase in hormone-responsive breast tumors. In subsequent stages, elevated levels of heparanase induced acquisition of procancerous phenotype by tumor-associated macrophages, resulting in activation of tumor-promoting signaling and acceleration of breast tumor growth under obese conditions. As techniques to screen for heparanase expression in tumors become available, these findings provide rational and a mechanistic basis for designing antiheparanase approaches to uncouple obesity and breast cancer in a rapidly growing population of obese patients. SIGNIFICANCE: This study reveals the role of heparanase in promoting obesity-associated breast cancer and provides a mechanistically informed approach to uncouple obesity and breast cancer in a rapidly growing population of obese patients.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma/enzymology , Glucuronidase/physiology , Obesity/complications , Adipose Tissue/metabolism , Animals , Aromatase/biosynthesis , Aromatase/genetics , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Carcinoma/etiology , Carcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Diet, High-Fat/adverse effects , Disease Progression , Estrogens/physiology , Female , Glucuronidase/deficiency , Glucuronidase/genetics , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/etiology , Neoplasms, Hormone-Dependent/pathology , Pancreatic Neoplasms/pathology
2.
Cancer Lett ; 449: 252-262, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30790678

ABSTRACT

Prostate cancer (PCa) is one of the most common cancer in men. Although hormone-sensitive PCa responds to androgen-deprivation, there are no effective therapies for castration-resistant PCa. It has been recently suggested that proton pump inhibitors (PPIs) may increase the risk of certain cancers; however, association with PCa remains elusive. Here, we evaluated the tumorigenic activities of PPIs in vitro, in PCa cell lines and epithelial cells from benign prostatic hyperplasia (BPH) and in vivo, in PCa mice xenografts. PPIs increased survival and proliferation, and inhibited apoptosis in LNCaP cells. These effects were attenuated or absent in androgen-insensitive DU-145 and PC3 cells, respectively. Specifically, omeprazole (OME) promoted cell cycle progression, increased c-Myc expression, ErbB2 activity and PSA secretion. Furthermore, OME induced the phosphorylation of MAPK-ERK1/2, PI3K/Akt and GSK-3ß, and blunted the expression and activity of cellular prostatic acid phosphatase. OME also increased survival, proliferation and PSA levels in BPH cells. In vivo, OME promoted tumor growth in mice bearing LNCaP xenografts. Our results indicate that PPIs display tumorigenic activities in PCa cells, suggesting that their long-term administration in patients should be carefully monitored.


Subject(s)
Acid Phosphatase/antagonists & inhibitors , Cell Proliferation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms, Hormone-Dependent/enzymology , Omeprazole/toxicity , Phosphatidylinositol 3-Kinase/metabolism , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Proton Pump Inhibitors/toxicity , Receptor, ErbB-2/metabolism , Acid Phosphatase/metabolism , Animals , Apoptosis/drug effects , Humans , Male , Mice, Inbred NOD , Mice, SCID , Neoplasms, Hormone-Dependent/pathology , PC-3 Cells , Phosphorylation , Prostatic Neoplasms/pathology , Signal Transduction
3.
Oncogene ; 38(3): 375-389, 2019 01.
Article in English | MEDLINE | ID: mdl-30104711

ABSTRACT

Despite recent improvement in adjuvant therapies, triple-negative, and ER+ subtypes of breast cancer (BC) with metastatic potentials remain the leading cause of BC-related deaths. We investigated the role of phosphatidylinositol-4-phosphate 5-kinase alpha (PIP5Kα), a key upstream factor of PI3K/AKT, and the therapeutic effect of PIP5Kα inhibitor on subtypes of BC. The clinical importance of PIP5K1α and its association with survivals were analyzed using three BC cohorts from Nottingham (n = 913), KM plotter (n = 112) and TCGA (n = 817). Targeted overexpression or knockdown of PIP5K1α were introduced into BC cell lines. The effects of PIP5K1α and its inhibitor on growth and invasion of BC were confirmed by using in vitro assays including proliferation, migration, apoptosis and luciferase reporter assays and in vivo xenograft mouse models. All statistical tests were two-sided. PIP5K1α was associated with poor patient outcome in triple-negative BC (for PIP5K1α protein, p = 0.011 and for mRNA expression, p = 0.028, log-rank test). 29% of triple-negative BC had PIP5K1A gene amplification. Elevated level of PIP5K1α increased expression of pSer-473 AKT (p < 0.001) and invasiveness of triple-negative MDA-MB-231 cells (p < 0.001). Conversely, inhibition of PIP5K1α using its inhibitor ISA-2011B, or via knockdown suppressed growth and invasiveness of MDA-MB-231 xenografts (mean vehicle-treated controls = 2160 mm3, and mean ISA-2011B-treated = 600 mm3, p < 0.001). ISA-2011B-treatment reduced expression of pSer-473 AKT (p < 0.001) and its downstream effectors including cyclin D1, VEGF and its receptors, VEGFR1 and VEGFR2 (p < 0.001) in xenograft tumors. In ER+ cancer cells, PIP5K1α acted on pSer-473 AKT, and was in complexes with VEGFR2, serving as co-factor of ER-alpha to regulate activities of target genes including cyclin D1 and CDK1. Our study suggests that our developed PIP5K1α inhibitor has a great potential on refining targeted therapeutics for treatment of triple-negative and ER+ BC with abnormal PI3K/AKT pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/enzymology , Diketopiperazines/therapeutic use , Estrogens , Indoles/therapeutic use , Isoquinolines/therapeutic use , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/enzymology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Replication/drug effects , Diketopiperazines/pharmacology , Estradiol/pharmacology , Female , Humans , Indoles/pharmacology , Isoquinolines/pharmacology , Kaplan-Meier Estimate , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Phosphatidylinositol 3-Kinases/physiology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/physiology , RNA Interference , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Sci Rep ; 8(1): 12969, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154572

ABSTRACT

Breast cancer is the most common cancer in women worldwide. Hormone receptor breast cancers are the most common ones and, about 2 out of every 3 cases of breast cancer are estrogen receptor (ER) positive. Selective ER modulators, such as tamoxifen, are the first line of endocrine treatment of breast cancer. Despite the expression of hormone receptors some patients develop tamoxifen resistance and 50% present de novo tamoxifen resistance. Recently, we have demonstrated that activated mammalian target of rapamycin (mTOR) is positively associated with overall survival and recurrence free survival in ER positive breast cancer patients who were later treated with tamoxifen. Since altered expression of protein kinase B (PKB)/Akt in breast cancer cells affect N-myristoyltransferase 1 (NMT1) expression and activity, we investigated whether mTOR, a downstream target of PKB/Akt, regulates NMT1 in ER positive breast cancer cells (MCF7 cells). We inhibited mTOR by treating MCF7 cells with rapamycin and observed that the expression of NMT1 increased with rapamycin treatment over the period of time with a concomitant decrease in mTOR phosphorylation. We further employed mathematical modelling to investigate hitherto not known relationship of mTOR with NMT1. We report here for the first time a collection of models and data validating regulation of NMT1 by mTOR.


Subject(s)
Acyltransferases/biosynthesis , Adenocarcinoma/enzymology , Breast Neoplasms/enzymology , Estrogens , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/enzymology , TOR Serine-Threonine Kinases/physiology , Acyltransferases/genetics , Enzyme Induction , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Models, Biological , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/analysis , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
5.
J Inorg Biochem ; 185: 52-62, 2018 08.
Article in English | MEDLINE | ID: mdl-29763768

ABSTRACT

A series of Ni(II), Zn(II) and Cd(II) complexes with the Schiff base derived from the condensation 1:1 from pyridine-2-carboxaldehyde and 5,6-diamino-1,3-dimethyluracil (6-amino-1,3-dimethyl-5-[(pyridin-2-ylmethylidene)-amino]pyrimidine-2,4(1H,3H)-dione, DAAUPic) were synthesized and subsequently characterized by means of elemental analysis, FT-IR, NMR and nine of them by X-ray diffraction. Except the [Zn(µ-O,O'-AcO)(N5,N6,N1F-DAAUPicH-1)]2 and [Cd(O,O'-NO3)(µ-O4,(N5,N6,N1F)-DAAUPicH-1)(H2O)]2·2H2O dimers and the [Cd(µ-S,N-SCN)(N5,N6,N1F-DAAUPicH-1)]n chain-like polymer, all of them display monomeric molecular structures. The anticancer activity of compounds was also explored studying their effects on renin-angiotensin system (RAS)-regulating aminopeptidases on estrogen-dependent and triple negative breast cancer cell lines. At the concentrations used, some of the complexes showed different effects on (RAS) peptidases, which support the idea that their effects on cell growth/proliferation could be related to autocrine/paracrine regulatory functions of their corresponding peptide substrates.


Subject(s)
Aldehydes/chemistry , Aminopeptidases/metabolism , Breast Neoplasms/pathology , Cadmium/chemistry , Estrogens/metabolism , Neoplasms, Hormone-Dependent/pathology , Nickel/chemistry , Pyridines/chemistry , Renin-Angiotensin System , Schiff Bases/chemistry , Triple Negative Breast Neoplasms/pathology , Uracil/analogs & derivatives , Zinc/chemistry , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Crystallography, X-Ray , Humans , Molecular Structure , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/metabolism , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/metabolism , Uracil/chemistry
6.
Apoptosis ; 22(3): 357-368, 2017 03.
Article in English | MEDLINE | ID: mdl-27798717

ABSTRACT

Impaired apoptosis is one of the hallmarks of cancer. Caspase-3 and -8 are key regulators of the apoptotic response and have been shown to interact with the calpain family, a group of cysteine proteases, during tumorigenesis. The current study sought to investigate the prognostic potential of caspase-3 and -8 in breast cancer, as well as the prognostic value of combinatorial caspase and calpain expression. A large cohort (n = 1902) of early stage invasive breast cancer patients was used to explore the expression of caspase-3 and -8. Protein expression was examined using standard immunohistochemistry on tissue microarrays. High caspase-3 expression, but not caspase-8, is significantly associated with adverse breast cancer-specific survival (P = 0.008 and P = 0.056, respectively). Multivariate analysis showed that caspase-3 remained an independent factor when confounding factors were included (hazard ratio (HR) 1.347, 95% confidence interval (CI) 1.086-1.670; P = 0.007). The analyses in individual subgroups demonstrated the significance of caspase-3 expression in clinical outcomes in receptor positive (ER, PR or HER2) subgroups (P = 0.001) and in non-basal like subgroup (P = 0.029). Calpain expression had been previously assessed. Significant association was also found between high caspase-3/high calpain-1 and breast cancer-specific survival in the total patient cohort (P = 0.005) and basal-like subgroup (P = 0.034), as indicated by Kaplan-Meier analysis. Caspase-3 expression is associated with adverse breast cancer-specific survival in breast cancer patients, and provides additional prognostic values in distinct phenotypes. Combinatorial caspase and calpain expression can predict worse prognosis, especially in basal-like phenotypes. The findings warrant further validation studies in independent multi-centre patient cohorts.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma/enzymology , Caspase 3/analysis , Caspase 8/analysis , Estrogens , Genes, erbB-2 , Neoplasm Proteins/analysis , Neoplasms, Hormone-Dependent/enzymology , Progesterone , Adolescent , Adult , Aged , Breast Neoplasms/classification , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Calcium-Binding Proteins/analysis , Calpain/analysis , Carcinoma/classification , Carcinoma/mortality , Carcinoma/pathology , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasms, Hormone-Dependent/mortality , Neoplasms, Hormone-Dependent/pathology , Prognosis , Tissue Array Analysis , Young Adult
7.
Zhonghua Nan Ke Xue ; 23(2): 178-182, 2017 Feb.
Article in Chinese | MEDLINE | ID: mdl-29658259

ABSTRACT

Glycogen synthase kinase3 (GSK3α and GSK3ß) are serine/threonine protein kinases acting on numerous substrates and involved in the regulation of various cellular functions such as their proliferation, survival, glycogen metabolism, and autophagy. Accumulating evidence indicates that the expression of GSK3α is increased mainly in androgendependent while that of GSK3ß in androgenindependent prostate cancer, and that GSK3ß is also involved in the regulation of the transactivation of the androgen receptor (AR) and growth of prostate cancer. Animal experiments have proved that some GSK3 inhibitors, such as lithium, can significantly suppress tumor growth in different animal models of prostate cancer. The GSK3 inhibitor is promising to be an important agent for the clinical management of prostate cancer.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/enzymology , Androgens , Animals , Cell Line, Tumor , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Humans , Male , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism
8.
Eur J Cancer ; 51(18): 2725-31, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26597446

ABSTRACT

BACKGROUND: High-dose oestrogen (HDE) is effective but toxic in postmenopausal women with advanced breast cancer (ABC). Prolonged oestrogen deprivation sensitises BC cell lines to estrogen and we hypothesised that third-generation aromatase inhibitors (AIs) would sensitise BCs to low-dose estradiol (LDE). METHODS: A single-arm phase II study of LDE (2 mg estradiol valerate daily) in postmenopausal women with estrogen receptor-positive (ER+) ABC. The primary end-point was clinical benefit (CB) rate. If LDE was ineffective, HDE was offered. If LDE was effective, retreatment with the pre-LDE AI was offered on progression. RESULTS: Twenty-one patients were recruited before the trial was closed early due to slow accrual; 19 were assessable for efficacy and toxicity. CB was seen in 5 in 19 patients (26%; 95% confidence interval 9.1-51.2%), all with prolonged SD (median duration 16.8 months; range 11.0-29.6). Treatment was discontinued for toxicity in 4 in 19 patients (21%) and 8 in 11 women without hysterectomy experienced vaginal bleeding (VB). After primary LDE failure, three patients received HDE and one achieved a partial response (PR). Following CB on LDE, four patients restarted pre-LDE AI and three achieved CB including one PR. Those with CB to LDE had a significantly longer duration of first-line endocrine therapy for ABC than those without (54.9 versus 16.8 months; p < 0.01) CONCLUSION: LDE is an effective endocrine option in women with evidence of prolonged sensitivity to AI therapy. LDE is reasonably well tolerated although VB is an issue. Re-challenge with the pre-LDE AI following progression confirms re-sensitisation as a true phenomenon.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Aromatase Inhibitors/administration & dosage , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Estradiol/analogs & derivatives , Neoplasms, Hormone-Dependent/drug therapy , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/adverse effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Early Termination of Clinical Trials , England , Estradiol/administration & dosage , Estradiol/adverse effects , Female , Humans , Middle Aged , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Patient Selection , Postmenopause , Quality of Life , Time Factors , Treatment Outcome
9.
Int J Clin Exp Pathol ; 8(5): 5309-17, 2015.
Article in English | MEDLINE | ID: mdl-26191232

ABSTRACT

PURPOSE: The aim was to verify the potential association between CYP19A1 genetic polymorphisms and clinical outcome of hormone therapy in hormone receptor (HR)-positive early breast cancer. METHODS: Genotyping for CYP19A1 rs4646 (C/A) polymorphism was performed on 287 women with HR-positive early breast cancer. Associations were evaluated between CYP19A1 rs4646 genotypes and disease-free survival (DFS). RESULTS: Totally, women with the minor allele (AA or AC) had an improved DFS when compared with those carrying the homozygous common allele (CC) (AA or AC vs. CC: 62.7 months versus 55.6 months; Hazard ratio (HR), 0.745; 95% CI, 0.562-0.988; P=0.04). The difference was further demonstrated by multivariate analyses (HR, 0.681; 95% CI, 0.506-0.917; P=0.011). In premenopausal women, AA genotype was associated with a prolonged DFS (AA versus CC or AC: 98.2 months versus 56.2 months; HR, 0.425; 95% CI, 0.198-0.914; P=0.024). In addition, women with the A allele had an improved DFS when compared with those carrying the homozygous C allele (AA or AC vs. CC: 62.7 months versus 55.6 months; HR, 0.709; 95% CI, 0.516-0.975; P=0.033). These findings were further confirmed by the Cox regression model (HR, 0.336, 0.670; 95% CI, 0.160-0.836, 0.479-0.938; P=0.017, 0.019). In postmenopausal women, rs4646 genotypes were significantly associated with DFS (AA versus AC versus CC: 32.7 months versus not reached versus 56.3 months; P=0.011). Women carrying AA variant had a poorer DFS than those with CC or AC genotypes (32.7 months versus 70.6 months; HR, 3.613; 95% CI, 1.380-9.457; P=0.005). Furthermore, being adjusted by the patients features in multivariate analyses, AA genotype remained an independent prognostic factor for DFS (HR, 3.614; 95% CI, 1.308-9.991; P=0.013). CONCLUSIONS: The homozygous minor allele (AA) of CYP19A1 rs4646 is significantly associated with improved clinical outcome of hormone therapy in premenopausal HR-positive early breast cancer patients, but with a worse impact on postmenopausal women. The findings are novel, if confirmed, genotyping for CYP19A1 rs4646 polymorphism may provide predictive information for better selection of endocrine treatment.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Aromatase/genetics , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Polymorphism, Single Nucleotide , Adult , Aged , Antineoplastic Agents, Hormonal/pharmacokinetics , Aromatase/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Chi-Square Distribution , Disease-Free Survival , Female , Gene Frequency , Heterozygote , Homozygote , Humans , Kaplan-Meier Estimate , Middle Aged , Multivariate Analysis , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/mortality , Patient Selection , Pharmacogenetics , Phenotype , Postmenopause , Precision Medicine , Premenopause , Proportional Hazards Models , Risk Factors , Time Factors , Treatment Outcome , Young Adult
10.
Eur J Cancer ; 51(14): 1890-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26169018

ABSTRACT

PURPOSE: Poor adherence to oral adjuvant hormonal therapy for breast cancer is a common problem, but little is known about the relationship between self-report adherence measures and hormonal suppression. We evaluated the relationship of three self-report measures of medication adherence and oestrogen among patients on aromatase inhibitors (AIs). MATERIALS AND METHODS: We recruited 235 women with breast cancer who were prescribed AI therapy. Participants self-reported AI adherence by completing the following: (1) a single item asking whether they took an AI in the last month, (2) a modified Morisky Medication Adherence Scale-8 (MMAS-8) and (3) the Visual Analog Scale (VAS). Serum estrone and estradiol were analysed using organic solvent extraction and Celite column partition chromatography, followed by radioimmunoassay. RESULTS: Ten percent of participants reported they had not taken an AI in the last month and among this group, median estrone (33.2 pg/ml [interquartile range (IQR)=22.3]) and estradiol levels (7.2 pg/mL [IQR=3.3]) were significantly higher than those in participants who reported AI use (median estrone=11.5 pg/mL [IQR=4.9]; median estradiol=3.4 pg/mL [IQR=2.1]; p<0.001). This relationship held when controlling for race and AI drug type. CONCLUSIONS: A single-item monthly-recall adherence measure for AIs was associated with oestrogen serum levels. This suggests that patient-reported monthly adherence may be a useful measure to identify early non-adherence behaviour and guide interventions to improve patient adherence to hormonal treatment.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Biomarkers, Tumor/blood , Breast Neoplasms/drug therapy , Drug Monitoring , Estradiol/blood , Estrone/blood , Medication Adherence , Neoplasms, Hormone-Dependent/drug therapy , Self Report , Adult , Aged , Aged, 80 and over , Breast Neoplasms/blood , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Drug Monitoring/methods , Female , Humans , Middle Aged , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Pennsylvania , Predictive Value of Tests , Treatment Outcome
11.
Oncotarget ; 6(5): 2865-78, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25605246

ABSTRACT

Herein, we investigated therapeutic potential of a novel histone lysine demethylase 1 (LSD1) inhibitor, NCL1, in prostate cancer. Hormone-sensitive prostate cancer cells, (LNCaP) and castration resistant cancer cells (PC3 and PCai1) were treated with NCL1, and LSD1 expression and cell viability were assessed. Prostate cancer cells showed strong LSD1 expression, and cell viability was decreased by NCL1. ChIP analysis showed that NCL1 induced H3K9me2 accumulation at the promoters of androgen-responsive genes. NCL1 also induced G1 cell cycle arrest and apoptosis. In addition, autophagosomes and autolysosomes were induced by NCL1 treatment in LNCaP. Furthermore, LC3-II expression was significantly increased by NCL1 and chloroquine. In mice injected subcutaneously with PCai1 and intraperitoneally with NCL1, tumor volume was reduced with no adverse effects in NCL1-treated mice. Finally, LSD1 expression in human cancer specimens was significantly higher than that in normal prostate glands. In conclusion, NCL1 effectively suppressed prostate cancer growth without adverse events. We suggest that NCL1 is a potential therapeutic agent for hormone-resistant prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cyclopropanes/pharmacology , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , G1 Phase Cell Cycle Checkpoints/drug effects , Histone Demethylases/metabolism , Humans , Male , Mice, Nude , Molecular Targeted Therapy , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/pathology , RNA Interference , Time Factors , Transfection , Tumor Burden/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
12.
Oncotarget ; 6(5): 3195-210, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25605250

ABSTRACT

Androgen receptor (AR) and MNK activated eIF4E signaling promotes the development and progression of prostate cancer (PCa). In this study, we report that our Novel Retinamides (NRs) target both AR signaling and eIF4E translation in androgen sensitive and castration resistant PCa cells via enhancing AR and MNK degradation through ubiquitin-proteasome pathway. Dual blockade of AR and MNK initiated eIF4E activation by NRs in turn induced cell cycle arrest, apoptosis, and inhibited cell proliferation. NRs also inhibited cell migration and invasion in metastatic cells. Importantly, the inhibitory effects of NRs on AR signaling, eIF4E translation initiation and subsequent oncogenic program were more potent than that observed with clinically relevant retinoids, established MNK inhibitors, and the FDA approved PCa drugs. Our findings provide the first preclinical evidence that simultaneous inhibition of AR and eIF4E activation is a novel and efficacious therapeutic approach for PCa, and that NRs hold significant promise for treatment of advanced prostate cancer.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Prostatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Androgen/drug effects , Tretinoin/analogs & derivatives , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Tretinoin/pharmacology
13.
Cancer Res ; 75(2): 405-14, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25480943

ABSTRACT

Estrogen receptor (ER) α-positive breast cancers initially respond to antiestrogens but eventually become estrogen independent and recur. ER(+) breast cancer cells resistant to long-term estrogen deprivation (LTED) exhibit hormone-independent ER transcriptional activity and growth. A kinome-wide siRNA screen using a library targeting 720 kinases identified Polo-like kinase 1 (PLK1) as one of the top genes whose downregulation resulted in inhibition of estrogen-independent ER transcriptional activity and growth of LTED cells. High PLK1 mRNA and protein correlated with a high Ki-67 score in primary ER(+) breast cancers after treatment with the aromatase inhibitor letrozole. RNAi-mediated knockdown of PLK1 inhibited ER expression, estrogen-independent growth, and ER transcription in MCF7 and HCC1428 LTED cells. Pharmacologic inhibition of PLK1 with volasertib, a small-molecule ATP-competitive PLK1 inhibitor, decreased LTED cell growth, ER transcriptional activity, and ER expression. Volasertib in combination with the ER antagonist, fulvestrant, decreased MCF7 xenograft growth in ovariectomized mice more potently than each drug alone. JUNB, a component of the AP-1 complex, was expressed 16-fold higher in MCF7/LTED compared with parental MCF7 cells. Furthermore, JUNB and BCL2L1 (which encodes antiapoptotic BCL-xL) mRNA levels were markedly reduced upon volasertib treatment in MCF7/LTED cells, while they were increased in parental MCF7 cells. Finally, JUNB knockdown decreased ER expression and transcriptional activity in MCF7/LTED cells, suggesting that PLK1 drives ER expression and estrogen-independent growth via JUNB. These data support a critical role of PLK1 in acquired hormone-independent growth of ER(+) human breast cancer and is therefore a promising target in tumors that have escaped estrogen deprivation therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cell Cycle Proteins/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Drug Synergism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Fulvestrant , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pteridines/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Random Allocation , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic , Xenograft Model Antitumor Assays , bcl-X Protein/biosynthesis , bcl-X Protein/genetics , Polo-Like Kinase 1
14.
Br J Cancer ; 110(10): 2593-603, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24762960

ABSTRACT

BACKGROUND: Accumulating evidence demonstrates high levels of aldehyde dehydrogense (ALDH) activity in human cancer types, in part, because of its association with cancer stem cells. Whereas ALDH1A1 and ALDH7A1 isoforms were reported to associate with prostate tumorigenesis, whether other ALDH isoforms are associated with prostate cancer (PC) remains unclear. METHODS: ALDH3A1 expression was analysed in various PC cell lines. Xenograft tumours and 54 primary and metastatic PC tumours were stained using immunohistochemistry for ALDH3A1 expression. RESULTS: In comparison with the non-stem counterparts, a robust upregulation of ALDH3A1 was observed in DU145-derived PC stem cells (PCSCs). As DU145 PCSCs produced xenograft tumours with more advanced features compared with those derived from DU145 cells, higher levels of ALDH3A1 were detected in the former; a dramatic elevation of ALDH3A1 occurred in DU145 cell-derived lung metastasis compared with local xenograft tumours. Furthermore, while ALDH3A1 was not observed in prostate glands, ALDH3A1 was clearly present in PIN, and further increased in carcinomas. In comparison with the paired local carcinomas, ALDH3A1 was upregulated in lymph node metastatic tumours; the presence of ALDH3A1 in bone metastatic PC was also demonstrated. CONCLUSIONS: We report here the association of ALDH3A1 with PC progression.


Subject(s)
Adenocarcinoma/enzymology , Aldehyde Dehydrogenase/analysis , Neoplasm Proteins/analysis , Prostatic Neoplasms/enzymology , Adenocarcinoma/pathology , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Androgens , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/secondary , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Progression , Enzyme Induction , Epithelial Cells/enzymology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Neoplastic Stem Cells/enzymology , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology
15.
Lancet Oncol ; 15(4): 474-82, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24636210

ABSTRACT

BACKGROUND: Treatment of breast cancer with aromatase inhibitors is associated with damage to bones. NCIC CTG MA.27 was an open-label, phase 3, randomised controlled trial in which women with breast cancer were assigned to one of two adjuvant oral aromatase inhibitors-exemestane or anastrozole. We postulated that exemestane-a mildly androgenic steroid-might have a less detrimental effect on bone than non-steroidal anastrozole. In this companion study to MA.27, we compared changes in bone mineral density (BMD) in the lumbar spine and total hip between patients treated with exemestane and patients treated with anastrozole. METHODS: In MA.27, postmenopausal women with early stage hormone (oestrogen) receptor-positive invasive breast cancer were randomly assigned to exemestane 25 mg versus anastrozole 1 mg, daily. MA.27B recruited two groups of women from MA.27: those with BMD T-scores of -2·0 or more (up to 2 SDs below sex-matched, young adult mean) and those with at least one T-score (hip or spine) less than -2·0. Both groups received vitamin D and calcium; those with baseline T-scores of less than -2·0 also received bisphosphonates. The primary endpoints were percent change of BMD at 2 years in lumbar spine and total hip for both groups. We analysed patients according to which aromatase inhibitor and T-score groups they were allocated to but BMD assessments ceased if patients deviated from protocol. This study is registered with ClinicalTrials.gov, NCT00354302. FINDINGS: Between April 24, 2006, and May 30, 2008, 300 patients with baseline T-scores of -2·0 or more were accrued (147 allocated exemestane, 153 anastrozole); and 197 patients with baseline T-scores of less than -2·0 (101 exemestane, 96 anastrozole). For patients with T-scores greater than -2·0 at baseline, mean change of bone mineral density in the spine at 2 years did not differ significantly between patients taking exemestane and patients taking anastrozole (-0·92%, 95% CI -2·35 to 0·50 vs -2·39%, 95% CI -3·77 to -1·01; p=0·08). Respective mean loss in the hip was -1·93% (95% CI -2·93 to -0·93) versus -2·71% (95% CI -4·32 to -1·11; p=0·10). Likewise for those who started with T-scores of less than -2·0, mean change of spine bone mineral density at 2 years did not differ significantly between the exemestane and anastrozole treatment groups (2·11%, 95% CI -0·84 to 5·06 vs 3·72%, 95% CI 1·54 to 5·89; p=0·26), nor did hip bone mineral density (2·09%, 95% CI -1·45 to 5·63 vs 0·0%, 95% CI -3·67 to 3·66; p=0·28). Patients with baseline T-score of -2·0 or more taking exemestane had two fragility fractures and two other fractures, those taking anastrozole had three fragility fractures and five other fractures. For patients who had baseline T-scores of less than -2·0 taking exemestane, one had a fragility fracture and four had other fractures, whereas those taking anastrozole had five fragility fractures and one other fracture. INTERPRETATION: Our results demonstrate that adjuvant treatment with aromatase inhibitors can be considered for breast cancer patients who have T-scores less than -2·0. FUNDING: Canadian Cancer Society Research Institute, Pfizer, Canadian Institutes of Health Research.


Subject(s)
Androstadienes/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Bone Density/drug effects , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Nitriles/therapeutic use , Triazoles/therapeutic use , Adult , Aged , Aged, 80 and over , Anastrozole , Androstadienes/adverse effects , Antineoplastic Agents, Hormonal/adverse effects , Aromatase Inhibitors/adverse effects , Bone Density Conservation Agents/therapeutic use , Bones of Lower Extremity/diagnostic imaging , Bones of Lower Extremity/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Calcium/therapeutic use , Canada , Chemotherapy, Adjuvant , Dietary Supplements , Diphosphonates/therapeutic use , Female , Fractures, Bone/prevention & control , Humans , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/drug effects , Middle Aged , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Nitriles/adverse effects , Postmenopause , Radiography , Time Factors , Treatment Outcome , Triazoles/adverse effects , United States , Vitamin D/therapeutic use
16.
Eur Urol ; 66(5): 815-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24647231

ABSTRACT

BACKGROUND: Abiraterone acetate (an androgen biosynthesis inhibitor) plus prednisone is approved for treating patients with metastatic castration-resistant prostate cancer (mCRPC). Study COU-AA-302 evaluated abiraterone acetate plus prednisone versus prednisone alone in mildly symptomatic or asymptomatic patients with progressive mCRPC without prior chemotherapy. OBJECTIVE: Report the prespecified third interim analysis (IA) of efficacy and safety outcomes in study COU-AA-302. DESIGN, SETTING, AND PARTICIPANTS: Study COU-AA-302, a double-blind placebo-controlled study, enrolled patients with mCRPC from April 2009 to June 2010. A total of 1088 patients were stratified by Eastern Cooperative Oncology Group performance status (0 vs 1). INTERVENTION: Patients were randomised 1:1 to abiraterone 1000mg plus prednisone 5mg twice daily by mouth versus prednisone. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Co-primary end points were radiographic progression-free survival (rPFS) and overall survival (OS). Median times to event outcomes were estimated using the Kaplan-Meier method. Hazard ratios (HRs) and 95% confidence intervals (CIs) were derived using the Cox model, and treatment comparison used the log-rank test. The O'Brien-Fleming Lan-DeMets α-spending function was used for OS. Adverse events were summarised descriptively. RESULTS AND LIMITATIONS: With a median follow-up duration of 27.1 mo, improvement in rPFS was statistically significant with abiraterone treatment versus prednisone (median: 16.5 vs 8.2 mo; HR: 0.52 [95% CI, 0.45-0.61]; p<0.0001). Abiraterone improved OS (median: 35.3 vs 30.1 mo; HR: 0.79 [95% CI, 0.66-0.95]; p=0.0151) but did not reach the prespecified statistical efficacy boundary (α-level: 0.0035). A post hoc multivariate analysis for OS using known prognostic factors supported the primary results (HR: 0.74 [95% CI, 0.61-0.89]; p=0.0017), and all clinically relevant secondary end points and patient-reported outcomes improved. While the post hoc nature of the long-term safety analysis is a limitation, the safety profile with longer treatment exposure was consistent with prior reports. CONCLUSIONS: The updated IA of study COU-AA-302 in patients with mCRPC without prior chemotherapy confirms that abiraterone delays disease progression, pain, and functional deterioration and has clinical benefit with a favourable safety profile, including in patients treated for ≥24 mo. TRIAL REGISTRATION: Study COU-AA-302, ClinicalTrials.gov number, NCT00887198. PATIENT SUMMARY: The updated results of this ongoing study showed that disease progression was delayed in patients with advanced prostate cancer who were treated with abiraterone acetate and prednisone, and there was a continued trend in prolongation of life compared with patients treated with prednisone alone. Treatment with abiraterone acetate and prednisone was well tolerated by patients who were treated for >2 yr.


Subject(s)
Androstenes/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Abiraterone Acetate , Aged , Androstenes/adverse effects , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytochrome P-450 Enzyme Inhibitors/adverse effects , Disease Progression , Disease-Free Survival , Double-Blind Method , Drug Administration Schedule , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/mortality , Neoplasms, Hormone-Dependent/pathology , Prednisone/administration & dosage , Proportional Hazards Models , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Risk Factors , Steroid 17-alpha-Hydroxylase/metabolism , Time Factors , Treatment Outcome
17.
Mol Biol Cell ; 25(1): 17-29, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24196838

ABSTRACT

Invasion of tumor cells is a key step in metastasis that depends largely on the ability of these cells to degrade the extracellular matrix. Although we have showed that the GTPase ADP-ribosylation factor 1 (ARF1) is overexpressed in highly invasive breast cancer cell lines and that epidermal growth factor stimulation can activate this ARF isoform to regulate migration as well as proliferation, the role of this small GTP-binding protein has not been addressed in the context of invasiveness. Here we report that modulation of ARF1 expression and activity markedly impaired the ability of M.D. Anderson-metastatic breast-231 cells, a prototypical highly invasive breast cancer cell line, to degrade the extracellular matrix by controlling metalloproteinase-9 activity. In addition, we demonstrate that this occurs through inhibition of invadopodia maturation and shedding of membrane-derived microvesicles, the two key structures involved in invasion. To further define the molecular mechanisms by which ARF1 controls invasiveness, we show that ARF1 acts to modulate RhoA and RhoC activity, which in turn affects myosin light-chain (MLC) phosphorylation. Together our findings underscore for the first time a key role for ARF1 in invasion of breast cancer cells and suggest that targeting the ARF/Rho/MLC signaling axis might be a promising strategy to inhibit invasiveness and metastasis.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Breast Neoplasms/enzymology , Myosin Light Chains/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Surface Extensions/metabolism , Cell-Derived Microparticles/metabolism , Epidermal Growth Factor/physiology , ErbB Receptors/metabolism , Female , Humans , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology
18.
Oncol Nurs Forum ; 40(6): 547-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161632

ABSTRACT

The gene CYP2D6 has an extremely important role in drug metabolism. "Cytochrome P450, family 2, subfamily D, polypeptide 6" is the official name of CYP2D6. The gene is located at position 13.1 on the long (q) arm of chromosome 21 and encodes a member of the cytochrome P450 superfamily of enzymes. The cytochrome P450 proteins are monooxygenases that are heavily involved in drug metabolism (Genetics Home Reference, 2013), and many drugs are activated into their biologically active compounds. Because of numerous polymorphisms, the gene also has significant person-to-person variability. To date, more than 80 distinct CYP2D6 alleles and specific types and frequencies have been associated with different ethnic groups. CYP2D6*4 is the most common variant allele in Caucasians and, in that population, has a frequency of about 25%. On the other hand, CYP2D6*10 is common in the Asian population (Stearns & Rae, 2008).


Subject(s)
Antineoplastic Agents, Hormonal/pharmacokinetics , Biotransformation/genetics , Cytochrome P-450 CYP2D6/physiology , Estrogen Receptor Modulators/pharmacokinetics , Polymorphism, Genetic , Prodrugs/pharmacokinetics , Tamoxifen/pharmacokinetics , Alleles , Antidepressive Agents/pharmacokinetics , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carcinoma/drug therapy , Carcinoma/enzymology , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6 Inhibitors , Drug Interactions , Estrogen Receptor Modulators/therapeutic use , Estrogens , Ethnicity/genetics , Female , Genotype , Humans , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/enzymology , Prodrugs/therapeutic use , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tamoxifen/therapeutic use
19.
Korean J Intern Med ; 28(5): 573-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24009453

ABSTRACT

BACKGROUND/AIMS: The higher incidence of gallbladder cancer (GBC) in females has been accredited to the involvement of hormones. The clinical implications of sex hormone receptors in GBC are well established. Cysteine proteases (such as caspase-3-9, etc.) are known to play a central role in the apoptotic pathway. Of these, the downstream enzyme caspase-3 is often activated in the apoptotic pathway. The aim of this work was to examine the status of apoptosis (which directly correlated with the level of active caspase-3) in hormone-responsive GBC. METHODS: We used 10 androgen receptor (AR)-positive, 14 estrogen receptor (ER)-positive, 12 HER/neu-positive, eight triple positive, and 10 triple negative malignant GBC human tissue samples. We isolated the total cellular protein from tumor tissues and carried out Western blotting using antipro-caspase-3 and anti-activated caspase-3 antibodies. RESULTS: ER and HER/neu-positive GBC exhibited high caspase-3 activity and low procaspase-3 activity, whereas AR-positive GBC showed no significant level of apoptosis. We also evaluated the apoptosis status of triple positive GBC and triple negative GBC, and found significant apoptosis in triple positive GBC. CONCLUSIONS: The results indicate that ER and HER/neu-positive GBCs had active apoptosis, whereas AR-positive GBC was highly resistant to apoptosis.


Subject(s)
Apoptosis , Biomarkers, Tumor/analysis , Carcinoma/enzymology , Caspase 3/analysis , Gallbladder Neoplasms/enzymology , Neoplasms, Hormone-Dependent/enzymology , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/drug effects , Blotting, Western , Carcinoma/drug therapy , Carcinoma/pathology , Drug Resistance, Neoplasm , Enzyme Activation , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/pathology , Humans , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Receptor, ErbB-2/analysis , Receptors, Androgen/analysis , Receptors, Estrogen/analysis
20.
Biol Chem ; 394(7): 831-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23509217

ABSTRACT

The identification of the fundamental role of apoptosis in the growth balance and normal homeostasis against cell proliferation led to the recognition of its loss contributing to tumorigenesis. The mechanistic significance of reinstating apoptosis signaling towards selective targeting of malignant cells heavily exploits the caspase family of death-inducing molecules as a powerful therapeutic platform for the development of potent anticancer strategies. Some apoptosis inhibitors induce caspase expression and activity in preclinical models and clinical trials by targeting both the intrinsic and extrinsic apoptotic pathways and restoring the apoptotic capacity in human tumors. Furthermore, up-regulation of caspases emerges as a sensitizing mechanism for tumors exhibiting therapeutic resistance to radiation and adjuvant chemotherapy. This review provides a comprehensive discussion of the functional involvement of caspases in apoptosis control and the current understanding of reactivating caspase-mediated apoptosis signaling towards effective therapeutic modalities in cancer treatment.


Subject(s)
Caspase Inhibitors/pharmacology , Caspases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Apoptosis/drug effects , Caspase Inhibitors/therapeutic use , Cell Proliferation/drug effects , Humans , Male , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...