Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Br J Haematol ; 195(2): 230-243, 2021 10.
Article in English | MEDLINE | ID: mdl-34341985

ABSTRACT

Lenalidomide and dexamethasone (RD) is a standard treatment in relapsed/refractory immunoglobulin light chain (AL) amyloidosis (RRAL). We retrospectively investigated toxicity, efficacy and prognostic markers in 260 patients with RRAL. Patients received a median of two prior treatment lines (68% had been bortezomib-refractory; 33% had received high-dose melphalan). The median treatment duration was four cycles. The 3-month haematological response rate was 31% [very good haematological response (VGHR) in 18%]. The median follow-up was 56·5 months and the median overall survival (OS) and haematological event-free survival (haemEFS) were 32 and 9 months. The 2-year dialysis rate was 15%. VGHR resulted in better OS (62 vs. 26 months, P < 0·001). Cardiac progression predicted worse survival (22 vs. 40 months, P = 0·027), although N-terminal prohormone of brain natriuretic peptide (NT-proBNP) increase was frequently observed. Multivariable analysis identified these prognostic factors: NT-proBNP for OS [hazard ratio (HR) 1·71; P < 0·001]; gain 1q21 for haemEFS (HR 1·68, P = 0·014), with a trend for OS (HR 1·47, P = 0·084); difference between involved and uninvolved free light chains (dFLC) and light chain isotype for OS (HR 2·22, P < 0·001; HR 1·62, P = 0·016) and haemEFS (HR 1·88, P < 0·001; HR 1·59, P = 0·008). Estimated glomerular filtration rate (HR 0·71, P = 0·004) and 24-h proteinuria (HR 1·10, P = 0·004) were prognostic for renal survival. In conclusion, clonal and organ biomarkers at baseline identify patients with favourable outcome, while VGHR and cardiac progression define prognosis during RD treatment.


Subject(s)
Dexamethasone/therapeutic use , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/diagnosis , Immunoglobulin Light-chain Amyloidosis/drug therapy , Lenalidomide/therapeutic use , Adult , Aged , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Hormonal/toxicity , Biomarkers/metabolism , Cohort Studies , Dexamethasone/administration & dosage , Dexamethasone/toxicity , Drug Therapy, Combination/methods , Female , Follow-Up Studies , Humans , Immunoglobulin Light Chains/immunology , Immunoglobulin Light-chain Amyloidosis/immunology , Immunoglobulin Light-chain Amyloidosis/mortality , Immunologic Factors/administration & dosage , Immunologic Factors/therapeutic use , Immunologic Factors/toxicity , Lenalidomide/administration & dosage , Lenalidomide/toxicity , Male , Middle Aged , Natriuretic Peptide, Brain/metabolism , Peptide Fragments/metabolism , Prognosis , Progression-Free Survival , Recurrence , Retrospective Studies
2.
Xenobiotica ; 51(1): 88-94, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32876521

ABSTRACT

The formation of reactive metabolites (RMs) is a problem in drug development that sometimes results in severe hepatotoxicity. As detecting RMs themselves is difficult, a covalent binding assay using expensive radiolabelled tracers is usually performed for candidate selection. This study aimed to provide a practical approach toward the risk assessment of hepatotoxicity induced by covalent binding before candidate selection. We focused on flutamide because it contains a trifluoromethyl group that shows a strong singlet peak by 19F nuclear magnetic resonance (NMR) spectrometry. The covalent binding of flutamide was evaluated using quantitative NMR and its risk for hepatotoxicity was assessed by estimating the RM burden, an index that reflects the body burden associated with RM exposure by determining the extent of covalent binding, clinical dose and in vivo clearance. The extent of covalent binding and RM burden was 296 pmol/mg/h and 37.9 mg/day, respectively. Flutamide was categorised as high risk with an RM burden >10 mg/day consistent with its clinical hepatotoxicity. These results indicate that a combination of covalent binding assay using 19F-NMR and RM burden is useful for the risk assessment of RMs without using radiolabelled compounds.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Flutamide/toxicity , Antineoplastic Agents, Hormonal/metabolism , Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Flutamide/metabolism , Humans , Magnetic Resonance Spectroscopy , Microsomes, Liver/metabolism
3.
Medicine (Baltimore) ; 99(40): e22620, 2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33019484

ABSTRACT

RATIONALE: Low-dose mitotane has been widely used for many decades in patients with advanced adrenocortical carcinoma (ACC), which exhibited good safety profiles compared with the high-dose regimen. The clinical efficacy and toxicity of mitotane are closely related to its plasma concentration, and therapeutic drug monitoring (TDM) is recommended. Until now, no severe adverse drug reaction (ADR) related to the toxic plasma level after a short-term treatment of low-dose mitotane has been published. PATIENT CONCERNS: A 50-year-old Chinese female presented with severe neurological adverse events related to a toxic plasma levels of 42.8 mg/L after 4 months treatment of low-dose mitotane. DIAGNOSES: During the course of therapy, no other medication could cause neurological adverse events. Therefore, we suspected a high sensitivity to the side effect of mitotane related to a toxic plasma level. INTERVENTIONS: Treatment of mitotane was stopped. OUTCOMES: The trough plasma concentration of mitotane decreased to 18.7 mg/mL after one and a half months, and the neurological symptoms gradually improved after drug discontinuance. LESSONS: The present case provides the first report of severe neurological adverse events induced by the short-term use of low-dose mitotane for adjuvant treatment in a patient with ACC, indicating that potentially severe ADR can also occur when using low-dose regimen in the early stage of treatment. TDM and early recognition could result in a favorable outcome.


Subject(s)
Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/drug therapy , Antineoplastic Agents, Hormonal/toxicity , Mitotane/toxicity , Nervous System Diseases/chemically induced , Antineoplastic Agents, Hormonal/blood , Antineoplastic Agents, Hormonal/therapeutic use , Asian People/ethnology , Drug Monitoring , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Middle Aged , Mitotane/blood , Mitotane/therapeutic use , Neurotoxicity Syndromes , Treatment Outcome , Withholding Treatment
4.
Stem Cells Dev ; 28(3): 186-195, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30511904

ABSTRACT

Hematological patients who accept chemotherapy always develop secondary tumor or even die of severe infections. As an important central lymphoid organ, the thymus is frequently damaged during chemotherapy. Previous studies showed that the mesenchymal stem cells (MSCs) can promote the proliferation and repair of epithelial cells in thymus. The purpose of our study is to investigate the reparative effects of human adipose-derived mesenchymal stem cells (hADMSCs) in chemotherapy-treated damaged thymus. Eighty mice were randomly divided into four groups: normal group, model control group, hADMSCs untreated group, and hADMSCs treated group. The mice were injected intraperitoneally with dexamethasone sodium phosphate (Dex 20 mg/kg), except the normal group. Then, the chemotherapy models were obtained after 1 week; the treated group was infused intraperitoneally with hADMSCs, whereas the model control group was injected with equal volumes of normal saline. The hADMSC's infusion day was regarded as day 0. The mice were sacrificed at different time points (days 3, 7, 10, and 14). The pathological structure and the function of the thymus, the recovery of T-lymphocyte subpopulation, and the proportion of regulatory T (Treg) cells in spleen and peripheral blood were detected. Additionally, we transfected hADMSCs by lentivirus with green fluorescent protein (GFP) to confirm whether they home to thymus and detected the expressions of cytokines that are associated with the development of thymus in hADMSCs and thymus. The results of the study showed that the hADMSCs treated group had a more rapid recovery in terms of thymic pathological structure and function. The hADMSCs could home to the damaged thymus and secrete cytokines that played important roles in repairing damaged thymus. The results indicated that hADMSCs could repair the damaged thymus caused by chemotherapy and improve the immune microenvironment, which may be a potential treatment for hematological patients.


Subject(s)
Lymphatic Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Regeneration , Thymus Gland/physiology , Adipose Tissue/cytology , Animals , Antineoplastic Agents, Hormonal/toxicity , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dexamethasone/toxicity , Humans , Lymphatic Diseases/chemically induced , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Thymus Gland/drug effects , Thymus Gland/metabolism , Thymus Gland/pathology
5.
Toxicol Appl Pharmacol ; 363: 88-97, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30503537

ABSTRACT

The endometrium is a particular sensitive target tissue for estradiol that is able to promptly modify its structure. Tamoxifen (TAM), a selective estrogen receptor modulator, was shown to promote a spectrum of uterine abnormalities, though the morphological and stereological effects of this drug in uterus is not clear. In this way, we have used an established model of ovariectomy followed by estradiol benzoate (EB) or TAM treatment and analyzed their effects in uterine histopathology and proliferation. Administration of EB promotes the unfolding and proliferation of the endometrium stroma, increasing uterine volume. No changes were found in uterine histomorphometric analysis upon TAM administration, except in the thickness of the luminal epithelium and endometrium layer. The latter may result from increased complexity and glandular volume density also observed in TAM treatment. In addition, EB induced PAX2 expression, an oncogene commonly found in epithelial tumors of the female genital tract, an effect that was weakened by previous TAM administration. Although treatments did not affect stroma cells proliferating index, in epithelial cells and, contrary to TAM, EB increased PCNA and not Ki67 expression. Collectively, our data suggest that the acute administration of TAM induces ERα-dependent atrophy of the uterine tissue and decreased the expression of proliferating cellular markers. On the contrary, if administered prior to EB, TAM is able to attenuate the action of the hormone in uterine morphology and biochemistry.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Tamoxifen/toxicity , Uterus/pathology , Animals , Atrophy/chemically induced , Atrophy/pathology , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estrogen Receptor alpha/metabolism , Estrous Cycle/metabolism , Female , Ki-67 Antigen/metabolism , Ovariectomy , PAX2 Transcription Factor/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Wistar , Toxicity Tests, Acute , Uterus/drug effects
6.
J Pharm Pharmacol ; 70(4): 488-497, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29380384

ABSTRACT

OBJECTIVES: Liposomes have attracted the attention of researchers due to their potential to act as drug delivery systems for cancer treatment. The present investigation aimed to develop liposomes loaded with prednisolone base and the evaluation of the antiproliferative effect on human colon carcinoma cell lines. METHODS: Liposomes were elaborated by following a reproducible thin film hydration technique. The physicochemical characterization of liposomes included photon correlation spectroscopy, microscopy analysis, Fourier transform infrared spectroscopy, rheological behaviour and electrophoresis. On the basis of these data and drug loading values, the best formulation was selected. Stability and drug release properties were also tested. KEY FINDINGS: Resulting liposomes exhibited optimal physicochemical and stability properties, an excellent haemocompatibility and direct antiproliferative effect on human colon carcinoma T-84 cell lines. CONCLUSIONS: This study shows direct antitumour effect of prednisolone liposomal formulation, which opens the door for liposomal glucocorticoids as novel antitumour agents.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Colonic Neoplasms , Cytotoxins/toxicity , Nanotechnology/methods , Prednisolone/toxicity , Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cytotoxins/chemistry , Cytotoxins/pharmacokinetics , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Liberation/physiology , Humans , Liposomes , Prednisolone/chemistry , Prednisolone/pharmacokinetics
7.
Cancer Invest ; 35(5): 313-324, 2017 May 28.
Article in English | MEDLINE | ID: mdl-28402678

ABSTRACT

Tamoxifen treatment in breast cancer patients is associated with increased risk of endometrial malignancies. Significantly, higher AGR2 expression was found in endometrial cancers that developed in women previously treated with tamoxifen compared to those who had not been exposed to tamoxifen. An association of elevated AGR2 level with myometrial invasion occurrence and invasion depth was also found. In vitro analyses identified a stimulatory effect of AGR2 on cellular proliferation. Although adverse tamoxifen effects on endometrial cells remain elusive, our work identifies elevated AGR2 as a candidate tamoxifen-dependent mechanism of action responsible for increased incidence of endometrial cancer.


Subject(s)
Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Antineoplastic Agents, Hormonal/toxicity , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Endometrial Neoplasms/chemically induced , Endometrium/drug effects , Proteins/metabolism , Tamoxifen/toxicity , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Humans , MCF-7 Cells , Mucoproteins , Neoplasm Invasiveness , Oncogene Proteins , Proteins/genetics , RNA Interference , Retrospective Studies , Risk Factors , Signal Transduction/drug effects , Transfection , Up-Regulation
8.
Oncotarget ; 8(5): 8406-8419, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28039445

ABSTRACT

Aromatase inhibitors (AIs) cause muscle weakness, bone loss, and joint pain in up to half of cancer patients. Preclinical studies have demonstrated that increased osteoclastic bone resorption can impair muscle contractility and prime the bone microenvironment to accelerate metastatic growth. We hypothesized that AI-induced bone loss could increase breast cancer progression in bone and exacerbate muscle weakness associated with bone metastases. Female athymic nude mice underwent ovariectomy (OVX) or sham surgery and were treated with vehicle or AI (letrozole; Let). An OVX-Let group was then further treated with bisphosphonate (zoledronic acid; Zol). At week three, trabecular bone volume was measured and mice were inoculated with MDA-MB-231 cells into the cardiac ventricle and followed for progression of bone metastases. Five weeks after tumor cell inoculation, tumor-induced osteolytic lesion area was increased in OVX-Let mice and reduced in OVX-Let-Zol mice compared to sham-vehicle. Tumor burden in bone was increased in OVX-Let mice relative to sham-vehicle and OVX-Let-Zol mice. At the termination of the study, muscle-specific force of the extensor digitorum longus muscle was reduced in OVX-Let mice compared to sham-vehicle mice, however, the addition of Zol improved muscle function. In summary, AI treatment induced bone loss and skeletal muscle weakness, recapitulating effects observed in cancer patients. Prevention of AI-induced osteoclastic bone resorption using a bisphosphonate attenuated the development of breast cancer bone metastases and improved muscle function in mice. These findings highlight the bone microenvironment as a modulator of tumor growth locally and muscle function systemically.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Aromatase Inhibitors/toxicity , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Muscle Strength/drug effects , Muscle Weakness/chemically induced , Muscle, Skeletal/drug effects , Nitriles/toxicity , Osteolysis/chemically induced , Receptors, Estrogen/deficiency , Triazoles/toxicity , Animals , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/metabolism , Bone Neoplasms/prevention & control , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Diphosphonates/pharmacology , Disease Progression , Estradiol/blood , Female , Humans , Imidazoles/pharmacology , Letrozole , Mice, Inbred BALB C , Mice, Nude , Muscle Weakness/physiopathology , Muscle, Skeletal/physiopathology , Osteolysis/pathology , Osteolysis/prevention & control , Ovariectomy , Time Factors , Tumor Burden , Tumor Microenvironment , Xenograft Model Antitumor Assays , Zoledronic Acid
9.
Endocr Relat Cancer ; 23(8): 635-50, 2016 08.
Article in English | MEDLINE | ID: mdl-27357940

ABSTRACT

Evidence supports a role of 17&-estradiol (E2) in carcinogenesis and the large majority of breast carcinomas are dependent on estrogen. The anti-estrogen tamoxifen (TAM) is widely used for both treatment and prevention of breast cancer; however, it is also carcinogenic in human uterus and rat liver, highlighting the profound complexity of its actions. The nature of E2- or TAM-induced chromosomal damage has been explored using relatively high concentrations of these agents, and only some numerical aberrations and chromosomal breaks have been analyzed. This study aimed to determine the effects of low doses of E2 and TAM (10(&8 )mol L(&1) and 10(&6 )mol L(&1) respectively) on karyotypes of MCF7, T47D, BT474, and SKBR3 breast cancer cells by comparing the results of conventional karyotyping and multi-FISH painting with cell proliferation. Estrogen receptor (ER)-positive (+) cells showed an increase in cell proliferation after E2 treatment (MCF7, T47D, and BT474) and a decrease after TAM treatment (MCF7 and T47D), whereas in ER& cells (SKBR3), no alterations in cell proliferation were observed, except for a small increase at 96 h. Karyotypes of both ER+ and ER& breast cancer cells increased in complexity after treatments with E2 and TAM leading to specific chromosomal abnormalities, some of which were consistent throughout the treatment duration. This genotoxic effect was higher in HER2+ cells. The ER&/HER2+ SKBR3 cells were found to be sensitive to TAM, exhibiting an increase in chromosomal aberrations. These in vitro results provide insights into the potential role of low doses of E2 and TAM in inducing chromosomal rearrangements in breast cancer cells.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Breast Neoplasms/genetics , Chromosome Aberrations/chemically induced , Estradiol/toxicity , Estrogen Antagonists/toxicity , Tamoxifen/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Karyotype
10.
PLoS One ; 11(5): e0155983, 2016.
Article in English | MEDLINE | ID: mdl-27196739

ABSTRACT

BACKGROUND: The ß-catenin signaling is important in cell growth and differentiation and is frequently dysregulated in various cancers. The most well-known mechanism of endocrine resistance is cross-talk between the estrogen receptor (ER) and other growth factor signaling, such as phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathway. In the present study, we investigated whether ß-catenin could be a potential target to overcome endocrine resistance in breast cancer. METHODS: We established tamoxifen-resistant (TamR) cell line via long-term exposure of MCF-7 breast cancer cells to gradually increasing concentrations of tamoxifen. The levels of protein expression and mRNA transcripts were determined using western blot analysis and real-time quantitative PCR. The transcriptional activity of ß-catenin was measured using luciferase activity assay. RESULTS: TamR cells showed a mesenchymal phenotype, and exhibited a relatively decreased expression of ER and increased expression of human epidermal growth factor receptor 2 and the epidermal growth factor receptor. We confirmed that the expression and transcriptional activity of ß-catenin were increased in TamR cells compared with control cells. The expression and transcriptional activity of ß-catenin were inhibited by ß-catenin small-molecule inhibitor, ICG-001 or ß-catenin siRNA. The viability of TamR cells, which showed no change after treatment with tamoxifen, was reduced by ICG-001 or ß-catenin siRNA. The combination of ICG-001 and mTOR inhibitor, rapamycin, yielded an additive effect on the inhibition of viability in TamR cells. CONCLUSION: These results suggest that ß-catenin plays a role in tamoxifen-resistant breast cancer, and the inhibition of ß-catenin may be a potential target in tamoxifen-resistant breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Drug Resistance, Neoplasm , Estrogen Antagonists/toxicity , Tamoxifen/toxicity , beta Catenin/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Humans , MCF-7 Cells , Pyrimidinones/pharmacology , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
11.
J Appl Toxicol ; 36(6): 853-62, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26584595

ABSTRACT

Tamoxifen is a widely used anticancer drug with both an estrogen agonist and antagonist effect. This study focused on its endocrine disrupting effect, and overall environmental significance. Zebrafish embryos were exposed to different concentrations (0.5, 5, 50 and 500 µg l(-1) ) of tamoxifen for 96 h. The results showed a complex effect of tamoxifen on zebrafish embryo development. For the 500 µg l(-1) exposure group, the heart rate was decreased by 20% and mild defects in caudal fin and skin were observed. Expressions of a series of genes related to endocrine and morphological changes were subsequently tested through quantitative real-time polymerase chain reaction. Bisphenol A as a known estrogen was also tested as an endocrine-related comparison. Among the expression of endocrine-related genes, esr1, ar, cyp19a1b, hsd3b1 and ugt1a1 were all increased by tamoxifen exposure, similar to bisphenol A. The cyp19a1b is a key gene that controls estrogen synthesis. Exposure to 0.5, 5, 50 and 500 µg l(-1) of tamoxifen caused upregulation of cyp19a1b expression to 152%, 568%, 953% and 2024% compared to controls, higher than the effects from the same concentrations of bisphenol A treatment, yet vtg1 was suppressed by 24% from exposure to 500 µg l(-1) tamoxifen. The expression of metabolic-related genes such as cyp1a, cyp1c2, cyp3a65, gpx1a, gstp1, gsr and genes related to observed morphological changes such as krt17 were also found to be upregulated by high concentrations of tamoxifen. These findings indicated the potential environmental effect of tamoxifen on teleost early development. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Endocrine Disruptors/toxicity , Gene Expression Regulation, Developmental/drug effects , Tamoxifen/toxicity , Water Pollutants, Chemical/toxicity , Zebrafish , Animal Fins/abnormalities , Animal Fins/drug effects , Animal Fins/embryology , Animals , Antineoplastic Agents, Hormonal/toxicity , Aromatase/genetics , Aromatase/metabolism , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Estrogens, Non-Steroidal/toxicity , Heart Rate/drug effects , Larva/drug effects , Larva/growth & development , Larva/metabolism , Osmolar Concentration , Random Allocation , Selective Estrogen Receptor Modulators/toxicity , Skin/drug effects , Skin/embryology , Skin Abnormalities/chemically induced , Skin Abnormalities/embryology , Skin Abnormalities/veterinary , Teratogens/toxicity , Vitellogenins/antagonists & inhibitors , Vitellogenins/genetics , Vitellogenins/metabolism , Zebrafish/abnormalities , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/agonists , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zygote/drug effects , Zygote/growth & development , Zygote/metabolism
12.
PLoS One ; 10(12): e0145061, 2015.
Article in English | MEDLINE | ID: mdl-26661278

ABSTRACT

Hereditary, hormonal, and behavioral factors contribute to the development of breast cancer. Alcohol consumption is a modifiable behavior that is linked to increased breast cancer risks and is associated with the development of hormone-dependent breast cancers as well as disease progression and recurrence following endocrine treatment. In this study we examined the molecular mechanisms of action of alcohol by applying molecular, genetic, and genomic approaches in characterizing its effects on estrogen receptor (ER)-positive breast cancer cells. Treatments with alcohol promoted cell proliferation, increased growth factor signaling, and up-regulated the transcription of the ER target gene GREB1 but not the canonical target TFF1/pS2. Microarray analysis following alcohol treatment identified a large number of alcohol-responsive genes, including those which function in apoptotic and cell proliferation pathways. Furthermore, expression profiles of the responsive gene sets in tumors were strongly associated with clinical outcomes in patients who received endocrine therapy. Correspondingly, alcohol treatment attenuated the anti-proliferative effects of the endocrine therapeutic drug tamoxifen in ER-positive breast cancer cells. To determine the contribution and functions of responsive genes, their differential expression in tumors were assessed between outcome groups. The proto-oncogene BRAF was identified as a novel alcohol- and estrogen-induced gene that showed higher expression in patients with poor outcomes. Knock-down of BRAF, moreover, prevented the proliferation of breast cancer cells. These findings not only highlight the mechanistic basis of the effects of alcohol on breast cancer cells and increased risks for disease incidents and recurrence, but may facilitate the discovery and characterization of novel oncogenic pathways and markers in breast cancer research and therapeutics.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Ethanol/pharmacology , Tamoxifen/toxicity , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Estradiol/toxicity , Female , Humans , MCF-7 Cells , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Tamoxifen/therapeutic use , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects
13.
Sci Rep ; 5: 13170, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26255816

ABSTRACT

Breast cancer is a hormone-dependent cancer and usually treated with endocrine therapy using aromatase inhibitors or anti-estrogens such as tamoxifen. A majority of breast cancer, however, will often fail to respond to endocrine therapy. In the present study, we explored miRNAs associated with endocrine therapy resistance in breast cancer. High-throughput miRNA sequencing was performed using RNAs prepared from breast cancer MCF-7 cells and their derivative clones as endocrine therapy resistant cell models, including tamoxifen-resistant (TamR) and long-term estrogen-deprived (LTED) MCF-7 cells. Notably, miR-21 was the most abundantly expressed miRNA in MCF-7 cells and overexpressed in TamR and LTED cells. We found that miR-378a-3p expression was downregulated in TamR and LTED cells as well as in clinical breast cancer tissues. Additionally, lower expression levels of miR-378a-3p were associated with poor prognosis for tamoxifen-treated patients with breast cancer. GOLT1A was selected as one of the miR-378a-3p candidate target genes by in silico analysis. GOLT1A was overexpressed in breast cancer specimens and GOLT1A-specific siRNAs inhibited the growth of TamR cells. Low GOLT1A levels were correlated with better survival in patients with breast cancer. These results suggest that miR-378a-3p-dependent GOLT1A expression contributes to the mechanisms underlying breast cancer endocrine resistance.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/metabolism , Tamoxifen/toxicity , 3' Untranslated Regions , Antineoplastic Agents, Hormonal/therapeutic use , Base Sequence , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Survival/drug effects , Disease-Free Survival , Drug Resistance, Neoplasm/drug effects , Female , High-Throughput Nucleotide Sequencing , Humans , MCF-7 Cells , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , RNA Interference , RNA, Double-Stranded/metabolism , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , Tamoxifen/therapeutic use , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
14.
Cancer Lett ; 367(1): 76-87, 2015 Oct 10.
Article in English | MEDLINE | ID: mdl-26208432

ABSTRACT

Breast cancer cells with stem cell characteristics (CSC) are a distinct cell population with phenotypic similarities to mammary stem cells. CSCs are important drivers of tumorigenesis and the metastatic process. Tamoxifen is the most widely used hormonal therapy for estrogen receptor (ER) positive cancers. In our study, tamoxifen was effective in reducing proliferation of ER + adherent cancer cells, but not their CSC population. We isolated, expanded and incubated CSC from seven breast cancers with or without tamoxifen. By genome-wide transcriptional analysis we identified tamoxifen-induced transcriptional pathways associated with ribosomal biogenesis and mRNA translation, both regulated by the mTOR-pathway. We observed induction of the key mTOR downstream targets S6K1, S6RP and 4E-BP1 in-patient derived CSCs by tamoxifen on protein level. Using the mTOR inhibitors rapamycin, everolimus and PF-04691502 (a dual PI3K/mTOR inhibitor) and in combination with tamoxifen, significant reduction in mammosphere formation was observed. Hence, we suggest that the CSC population play a significant role during endocrine resistance through activity of the mTOR pathway. In addition, tamoxifen further stimulates the mTOR-pathway but can be antagonized using mTOR-inhibitors.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Breast Neoplasms/enzymology , Estrogen Antagonists/toxicity , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tamoxifen/toxicity , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Separation/methods , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Enzyme Activation , Everolimus , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Pyridones/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Spheroids, Cellular , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured
15.
Sci Rep ; 5: 7641, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25560734

ABSTRACT

Most primary breast cancers express estrogen receptor α and can be treated via endocrine therapy using anti-estrogens such as tamoxifen; however, acquired endocrine resistance is a critical issue. To identify tamoxifen response-related microRNAs (miRNAs) in breast cancer, MCF-7 cells infected with a lentiviral miRNA library were treated with 4-hydroxytamoxifen (OHT) or vehicle for 4 weeks, and the amounts of individual miRNA precursors that had integrated into the genome were evaluated by microarray. Compared to the vehicle-treated cells, 5 'dropout' miRNAs, which were downregulated in OHT-treated cells, and 6 'retained' miRNAs, which were upregulated in OHT-treated cells, were identified. Of the dropout miRNAs, we found that miR-574-3p expression was downregulated in clinical breast cancer tissues as compared with their paired adjacent tissues. In addition, anti-miR-574-3p reversed tamoxifen-mediated suppression of MCF-7 cell growth. Clathrin heavy chain (CLTC) was identified as a miR-574-3p target gene by in silico algorithms and luciferase reporter assay using the 3' untranslated region of CLTC mRNA. Interestingly, loss and gain of miR-574-3p function in MCF-7 cells causes CLTC to be upregulated and downregulated, respectively. These results suggest that functional screening mediated by miRNA libraries can provide new insights into the genes essential for tamoxifen response in breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Down-Regulation/drug effects , MicroRNAs/metabolism , Tamoxifen/analogs & derivatives , Up-Regulation/drug effects , 3' Untranslated Regions , Algorithms , Base Sequence , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Clathrin/antagonists & inhibitors , Clathrin/genetics , Clathrin/metabolism , Female , Gene Library , Humans , MCF-7 Cells , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sequence Alignment , Tamoxifen/toxicity
16.
Ecotoxicol Environ Saf ; 111: 228-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25450938

ABSTRACT

Estrogen receptor (ER) antagonistic chemicals in aquatic environments are believed to influence the binding of both endogenous and exogenous estrogens to ERs in aquatic organisms. Although the combined effects of estrogenic compounds have attracted much scientific concern, little work has been done on the influence of such antiestrogens on the biological effects of estrogens. This study focused on how the presence of different amounts of antagonists affects the results of ER agonist activity tests. To achieve this, three questions were stated and answered in sequence. A two-hybrid recombinant yeast assay mediated by ER was adopted, providing a single mode of action and single target of action for this study. Mixtures created by an ER agonist and three antagonists following the fixed-ratio principle were assessed. The concentration of 17ß-estradiol causing maximum induction was set as the fixed dose of estrogen in the antagonist activity test (question 1). When the two classes of chemicals coexisted, antiestrogens, which as a whole behaved according to the concentration addition model (question 2), decreased the response of estrogen and compressed the concentration-response curves along the y-axis in the agonist activity test (question 3). This may cause the estradiol equivalent to be underestimated and potentially mask the action of estrogenic effects in toxicity evaluation of environmental samples.


Subject(s)
Endocrine Disruptors/toxicity , Estrogen Antagonists/toxicity , Estrogens/toxicity , Saccharomyces cerevisiae/drug effects , Two-Hybrid System Techniques , Antineoplastic Agents, Hormonal/toxicity , Dose-Response Relationship, Drug , Estradiol/chemistry , Estrogen Receptor Modulators/toxicity , Humans , In Vitro Techniques , Pesticides/toxicity , beta-Galactosidase/metabolism
17.
Int J Pharm ; 468(1-2): 1-14, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24704438

ABSTRACT

A novel approach of enhancing the Tamoxifen uptake via Intestinal Lymphatic System is executed by developing long chain lipid and oil based nanostructured lipid carrier system (Tmx-NLC). The aim was to achieve improved systemic bioavailability of Tamoxifen, prevent systemic and hepatotoxicity and enhance antitumor efficacy. Following the proof of concept achieved in cell culture experiments and in vivo pharmacokinetic and biodistribution study, the current work focuses on investigation of antitumor efficacy and treatment associated toxicity in murine mammary tumor mice model. The efficacy study demonstrated greater tumor suppression and 100% survival with 1.5 and 3 mg/kg Tmx-NLC compared to 3 mg/kg Tamoxifen suspension and Mamofen(®) (Khandelwal Pharmaceuticals, Mumbai, India). Tmx-NLC treatment for a month demonstrated improved systemic toxicity profile and no evidences of hepatotoxicity. Thus, developed Tmx-NLC could prove to be a promising delivery strategy to confer superior therapeutic efficacy and ability to address the biopharmaceutical and toxicity associated issues of drug.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drug Carriers , Lipids/chemistry , Nanostructures , Tamoxifen/pharmacology , Administration, Oral , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/toxicity , Biological Availability , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Female , Intestinal Absorption , Mice, Inbred BALB C , Nanotechnology , Tamoxifen/administration & dosage , Tamoxifen/chemistry , Tamoxifen/pharmacokinetics , Tamoxifen/toxicity , Technology, Pharmaceutical/methods , Tissue Distribution
18.
Toxicology ; 321: 89-95, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24769059

ABSTRACT

Tamoxifen, as well as most endocrine-disrupting chemicals, affects the reproductive system and sexual development, but little is known about its disruption of the molecular pathways regulating mammalian sex determination. In fetal mice, the expression levels and pattern of key genes involved in controlling sexually dimorphic balance were analyzed both in vivo and in vitro by using whole-mount in situ hybridization and quantitative-PCR. Developmental tamoxifen exposure induced abnormal up-regulation of the testis differentiation marker Pdfgra in Leydig cells and of Sox9 and Fgf9 in Sertoli cells in XX gonad. Immunohistochemistry analysis confirmed the over-expression of SOX9 protein. Accordingly, the ovary development marker Foxl2 was depressed at both the mRNA and protein levels. The increase in testosterone and the reduction in 17ß-estradiol and progesterone were observed by using the in vitro assay with organotypic cultures. Taken together, results indicated that tamoxifen induced the ectopic expression of well-established sex-specific genes during the critical developmental period, thus resulting in abnormal testicular development in the XX gonad of mammals. This study facilitates a better understanding of the molecular mechanisms of antiestrogens and possibly of compounds that interrupt estrogen signaling by other modes of action, and the association with the pathogenesis of human sexual developmental disorders.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Endocrine Disruptors , Ovary/growth & development , Sex Determination Processes/drug effects , Tamoxifen/toxicity , Testis/growth & development , Analysis of Variance , Animals , Anti-Mullerian Hormone/biosynthesis , Anti-Mullerian Hormone/genetics , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gonadal Steroid Hormones/biosynthesis , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred ICR , Organ Culture Techniques , Ovary/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Sex Differentiation/drug effects , Testis/drug effects
19.
Pharm Biol ; 52(8): 1009-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24646302

ABSTRACT

CONTEXT: Tamoxifen (TAM) is widely used for treatment of hormone-dependent breast cancer; however, it may be accompanied with hepatic injury. Allicin is the most abundant thiosulfinate molecule from garlic with the potential to provide beneficial effects on various diseases. OBJECTIVE: To elucidate the effect of commercially available allicin on both antitumor activity and liver injury of TAM. MATERIALS AND METHODS: The cytotoxicity of TAM and/or allicin was evaluated in vitro using cultured Ehrlich ascites carcinoma (EAC) cells and in vivo against murine tumor (solid) model of EAC. TAM induced liver injury in rats by intraperitoneally (i.p.) injection at a dose of 45 mg/kg, for 7 successive days. RESULTS: TAM at a dose of 3 µM (IC50) significantly decreased percent survival of EAC to 52%. TAM combination with allicin (5 or 10 µM) showed a significant cytotoxic effect compared with the TAM-treated group as manifested by a decrease in percent survival of EAC to 35% and 29%, respectively. Allicin (10 mg/kg, orally) enhanced the efficacy of TAM (1 mg/kg, i.p.) in mice as manifested by a significant increase in solid tumor growth inhibition by 82% compared with 70% in the TAM group. In rats, TAM intoxication resulted in a significant decline in SOD, GSH, and total protein with significant elevation in TBARS, ALT and AST, ALP, LDH, total bilirubin, γGT, and TNF-α levels. These changes are abrogated by allicin treatment. DISCUSSION AND CONCLUSION: The results suggest the beneficial role of allicin as an adjuvant to TAM in cancer treatment by alleviating liver injury.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Sulfinic Acids/therapeutic use , Tamoxifen/toxicity , Animals , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Disulfides , Female , Mice , Random Allocation , Rats , Xenograft Model Antitumor Assays/methods
20.
Mater Sci Eng C Mater Biol Appl ; 37: 278-85, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24582250

ABSTRACT

The present work reports a simple one step synthesis of nanoscale graphene oxide magnetic composites (GO-IO) using ferrofluid (GO-IOF). The obtained GO-IO were compared with GO-IO obtained from in situ (GO-IOI) methods. Anastrozole (ANS) was loaded on the GO-IOI and GO-IOF via simple stirring method to form GO-IOA and GO-IOFA respectively. These GO-IO prepared by two techniques were characterized using spectroscopic techniques and vibrating sample magnetometer (VSM) analysis. Particle size and potential were measured using Malvern Zetasizer. Scanning electron microscopy (SEM) was used for studying the surface morphology of GO-IO, and in addition to this elemental analysis was also performed for confirming the presence of iron. The cell viability assay was carried out using the MCF-7 cell line. It revealed that GO-IOFA had reasonably high cytotoxicity (49.7%) compared to GO (13.1%), ANS (16.6), GO-IOI (13%), GO-IOF (13.6) and GO-IOIA (18.34%). Both, GO-IOIA and GO-IOFA showed improved cytotoxicity when compared with pure ANS. GO-IOF were found to exhibit superior magnetic activity due to higher iron content along with smaller particle size and higher loading efficiency compared to GO-IOI. The overall effect suggests that GO-IO can be utilized as efficient carriers for the loading of chemotherapeutic agents.


Subject(s)
Antineoplastic Agents, Hormonal/chemistry , Drug Carriers/chemistry , Graphite/chemistry , Magnetite Nanoparticles/chemistry , Nitriles/chemistry , Triazoles/chemistry , Anastrozole , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/toxicity , Breast Neoplasms/drug therapy , Cell Survival/drug effects , Female , Humans , MCF-7 Cells , Nitriles/administration & dosage , Nitriles/toxicity , Oxides/chemistry , Particle Size , Triazoles/administration & dosage , Triazoles/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...