Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.241
Filter
1.
J Med Chem ; 66(16): 11094-11117, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37584263

ABSTRACT

Endocrine resistance remains a significant problem in the clinical treatment of estrogen receptor α-positive (ERα+) breast cancer (BC). In this study, we developed a series of novel dual-functional ERα degraders based on a bridged bicyclic scaffold with selenocyano (SeCN) side chains. These compounds displayed potent ERα degradation and tubulin depolymerization activity. Among them, compounds 35s and 35t exhibited the most promising antiproliferative and ERα degradation activity in multiple ERα+ BC cell lines bearing either wild-type or mutant ERα. Meanwhile, compounds 35s and 35t disrupted the microtubule network by restraining tubulin polymerization, evidenced by 35t inducing cell cycle arrest in the G2/M phase. In MCF-7 and LCC2 xenograft models, compounds 35s and 35t remarkably suppressed tumor growth without noticeable poisonousness. Finally, this study provided guidance for developing new dual-target antitumor drug candidates for the ERα+ BC therapy, especially for the resistant variant.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Receptors, Estrogen , Female , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor alpha/metabolism , MCF-7 Cells , Receptors, Estrogen/antagonists & inhibitors , Tubulin/chemistry , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
2.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35023540

ABSTRACT

Estrogens are well-known to regulate development of sexual dimorphism of the brain; however, their role in embryonic brain development prior to sex-differentiation is unclear. Using estrogen biosensor zebrafish models, we found that estrogen activity in the embryonic brain occurs from early neurogenesis specifically in a type of glia in the olfactory bulb (OB), which we name estrogen-responsive olfactory bulb (EROB) cells. In response to estrogen, EROB cells overlay the outermost layer of the OB and interact tightly with olfactory sensory neurons at the olfactory glomeruli. Inhibiting estrogen activity using an estrogen receptor antagonist, ICI182,780 (ICI), and/or EROB cell ablation impedes olfactory glomerular development, including the topological organisation of olfactory glomeruli and inhibitory synaptogenesis in the OB. Furthermore, activation of estrogen signalling inhibits both intrinsic and olfaction-dependent neuronal activity in the OB, whereas ICI or EROB cell ablation results in the opposite effect on neuronal excitability. Altering the estrogen signalling disrupts olfaction-mediated behaviour in later larval stage. We propose that estrogens act on glia to regulate development of OB circuits, thereby modulating the local excitability in the OB and olfaction-mediated behaviour.


Subject(s)
Estrogens/metabolism , Neurogenesis , Neuroglia/cytology , Olfactory Bulb/embryology , Animals , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/drug effects , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Receptors, Estrogen/antagonists & inhibitors , Synapses/metabolism , Synapses/physiology , Zebrafish
3.
Environ Toxicol Pharmacol ; 89: 103781, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34871798

ABSTRACT

Cell-based bioassays are very sensitive and allow integrative effect screening of the whole environmental sample, which is usually composed of a mixture of agonists and antagonists. Measured toxicity is usually expressed as a bioanalytical equivalent concentration. So far, it is not possible to distinguish which part of this value is caused by the agonists and which by the antagonists. In this article, we present a simple method to analyze the dose-response curve of a mixture and to determine an agonistic bioanalytical equivalent concentration: a concentration of a reference chemical that would elicit the same effect as do only agonists in an unknown mixture. The method has been validated using several artificially prepared mixtures of agonists and competitive antagonists measured in a recombinant yeast assay. No difference was observed between the calculated equivalent concentrations and the used concentrations of the agonist in the mixture.


Subject(s)
Biological Assay/methods , Endocrine Disruptors , Environmental Pollutants/toxicity , Drug Interactions , Estradiol/pharmacology , Estriol/pharmacology , Fulvestrant/pharmacology , Models, Theoretical , Receptors, Estrogen/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects
4.
Bioorg Chem ; 119: 105554, 2022 02.
Article in English | MEDLINE | ID: mdl-34923243

ABSTRACT

Nuclear Estrogen receptors (ER) are cytoplasmic proteins; translocated to the nucleus to induce transcriptional signals after getting bound to the estrogen hormone. ER activation implicated in cancer cell proliferation of female reproductive organs. Thus, the discovery of ER antagonists is a reliable strategy to combat estrogen-dependent breast cancer. Endometrial carcinoma is one of the complications encountered upon long-term therapy by selective estrogen receptor modulators (SERMs) like Tamoxifen (TMX) and methyl piperidinopyrazole (MPP). Thus, the ER-full antagonist is a solution to improve the safety of this class of therapeutics during the treatment of breast cancer. We selected MPP as a lead structure to design conformationally constrained analogs. Structural rigidification is a proven strategy to transform the SERMs into full antagonists. Accordingly, we synthesized 7-methoxy-3-(4-methoxyphenyl)-4,5-dihydro-2H-benzo[g]indazoles (4), (6a-c),(8-12) along with the biphenolic counterparts(13-19)that are the anticipated active metabolites. The 4-nitrophenyl derivative(4)is with the most balanced profile regardingthe in vivoanti-uterotrophic potential (EC50 = 4.160 µM); and the cytotoxicity assay of the corresponding active metabolite(13)against ER+ breast cancer cell lines (MCF-7 IC50 = 7.200 µM, T-47D IC50 = 11.710 µM). The inconsiderable uterotrophic activities of the elaborated ER-antagonists and weak antiproliferative activity of the compound(13)against ovarian cancer (SKOV-3 IC50 = 29.800 µM) highlighted it as a good start point to elaborate potential ER-full antagonists devoid of endometrial carcinoma. Extending the pendant chain that protrudes from the 2-(4-(substituted)-phenyl) ring of the new benzo-indazoles is recommended for enhancing the potency based on the binding mode of compound(13)in the ligand-binding domain (LBD) of ER.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Estrogen Receptor Antagonists/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Female , Humans , Ligands , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rats , Rats, Wistar , Receptors, Estrogen/metabolism , Structure-Activity Relationship
6.
Eur J Pharmacol ; 913: 174644, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34801532

ABSTRACT

The mortality of sepsis-induced cardiac dysfunction (SICD) is very high due to the complex pathophysiological mechanism. Syringaresinol (SYR) is a natural abstract which possesses anti-inflammatory property. The present study aims was to identify the protective impact of SYR on sepsis-induced cardiac dysfunction and investigate the specific mechanisms. We found that SYR improved the cardiac function and alleviated myocardial injury in mice that subjected to cecal ligation and puncture, in addition, SIRT1 expression was significantly elevated after SYR treatment compared to sepsis group both in vivo and in vitro, along with suppression of NLRP3 activation and proinflammatory cytokines release. However, SIRT1 inhibitor EX427 abolished the impact of SYR on LPS-induced pyroptosis in cardiomyocytes. Furthermore, molecular docking analysis predicted that there is high affinity between SYR and estrogen receptor (ER), ER inhibitor ICI182780, the specific ERß inhibitor PHTP and the specific ERαinhibitor AZD9496 were used to examine the role of ER in the protective effect of SYR against SICD, and the results suggested that ER activation was essential for the cardioprotective function of SYR. In conclusion, SYR ameliorates SICD via the ER/SIRT1/NLRP3/GSDMD pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Furans/pharmacology , Heart Diseases/drug therapy , Lignans/pharmacology , Receptors, Estrogen/metabolism , Sepsis/complications , Animals , Cardiotonic Agents/therapeutic use , Cinnamates/pharmacology , Disease Models, Animal , Fulvestrant/pharmacology , Furans/therapeutic use , Heart/drug effects , Heart Diseases/immunology , Heart Diseases/pathology , Humans , Indoles/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Lignans/therapeutic use , Male , Mice , Molecular Docking Simulation , Myocardium/pathology , Pyroptosis/drug effects , Pyroptosis/immunology , Receptors, Estrogen/antagonists & inhibitors , Sepsis/drug therapy , Sepsis/immunology , Sirtuin 1/metabolism
7.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830456

ABSTRACT

Selective estrogen receptor modulators (SERMs) act as estrogen receptor (ERα) agonists or antagonists depending on the target issue. Tamoxifen (TAM) (a non-steroidal triphenylethylene derivative) was the first SERM approved as anti-estrogen for the treatment of metastatic breast cancer. On the hunt for novel SERMs with potential growth inhibitory activity on breast cancer cell lines yet no potential to induce endometrial carcinoma, we designed and synthesized 28 novel TAM analogs. The novel analogs bear a triphenylethylene scaffold. Modifications on rings A, B, and C aim to attenuate estrogenic/anti-estrogenic activities of the novel compounds so they can potentially inhibit breast cancer and provide positive, beneficial estrogenic effects on other tissues with no risk of developing endometrial hyperplasia. Compound 12 (E/Z-1-(2-{4-[1-(4-Chloro-phenyl)-2-(4-methoxy-phenyl)-propenyl]-phenoxy}-ethyl)-piperidine) showed an appreciable relative ERα agonistic activity in a yeast estrogen screen (YES) assay. It successfully inhibited the growth of the MCF-7 cell line with GI50 = 0.6 µM, and it was approximately three times more potent than TAM. It showed no potential estrogenicity on Ishikawa endometrial adenocarcinoma cell line via assaying alkaline phosphatase (AlkP) activity. Compound 12 was tested in vivo to assess its estrogenic properties in an uterotrophic assay in an ovariectomized rat model. Compared to TAM, it induced less increase in wet uterine wet weight and showed no uterotrophic effect. Compound 12 is a promising candidate for further development due to its inhibition activity on MCF-7 proliferation with moderate AlkP activity and no potential uterotrophic effects. The in vitro estrogenic activity encourages further investigations toward potential beneficial properties in cardiovascular, bone, and brain tissues.


Subject(s)
Breast Neoplasms/drug therapy , Endometrial Neoplasms/drug therapy , Estrogen Receptor alpha/genetics , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Estrogen Antagonists/chemical synthesis , Estrogen Antagonists/pharmacology , Female , Humans , MCF-7 Cells , Rats , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Selective Estrogen Receptor Modulators/chemical synthesis , Stilbenes/chemical synthesis , Stilbenes/pharmacology , Tamoxifen/analogs & derivatives
8.
Chem Biol Interact ; 350: 109704, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34655567

ABSTRACT

Pyroptosis is a novel type of pro-inflammatory programmed cell death that has been strongly reported to be related to inflammation, immune, and cancer. Dihydroartemisinin (DHA) has good anti-tumor properties. However, the exact mechanism by which DHA induces pyroptosis to inhibit esophageal squamous cell carcinoma (ESCC) remains unclear. After applying DHA treatment to ESCC, we found that some dying cells exhibited the characteristic morphology of pyroptosis, such as blowing large bubbles from the cell membrane, accompanied by downregulation of pyruvate kinase isoform M2 (PKM2), activation of caspase-8/3, and production of GSDME-NT. Meanwhile, it was accompanied by an increased release of LDH and inflammatory factors (IL-18 and IL-1ß). Both knockdown of GSDME and application of caspase-8/3 specific inhibitors (z-ITED-FMK/Ac-DEVD-CHO) significantly inhibited DHA-induced pyroptosis. However, the former did not affect the activation of caspase-3. In contrast, overexpression of PKM2 inhibited caspase-8/3 activation as well as GSDME-N production. Furthermore, both si-GSDME and OE-PKM2 inhibited DHA-induced pyroptosis in vivo and in vitro. Therefore, the results suggest that DHA can induce pyroptosis of ESCC cells via the PKM2-caspase-8/3-GSDME pathway. Implication: In this study, we identified new mechanism of DHA in inhibiting ESCC development and progression, and provide a potential therapeutic agent for the treatment of ESCC.


Subject(s)
Artemisinins/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Pyroptosis/drug effects , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Pyroptosis/physiology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Up-Regulation , Xenograft Model Antitumor Assays , Thyroid Hormone-Binding Proteins
9.
Exp Mol Med ; 53(9): 1402-1412, 2021 09.
Article in English | MEDLINE | ID: mdl-34580423

ABSTRACT

Our recent studies identified a novel pathway of nicotinamide metabolism that involves 4-pyridone-3-carboxamide-1-ß-D-ribonucleoside (4PYR) and demonstrated its endothelial cytotoxic effect. This study tested the effects of 4PYR and its metabolites in experimental models of breast cancer. Mice were divided into groups: 4T1 (injected with mammary 4T1 cancer cells), 4T1 + 4PYR (4PYR-treated 4T1 mice), and control, maintained for 2 or 21 days. Lung metastasis and endothelial function were analyzed together with blood nucleotides (including 4PYR), plasma amino acids, nicotinamide metabolites, and vascular ectoenzymes of nucleotide catabolism. 4PYR metabolism was also evaluated in cultured 4T1, MDA-MB-231, MCF-7, and T47D cells. An increase in blood 4PYR in 4T1 mice was observed at 2 days. 4PYR and its metabolites were noticed after 21 days in 4T1 only. Higher blood 4PYR was linked with more lung metastases in 4T1 + 4PYR vs. 4T1. Decreased L-arginine, higher asymmetric dimethyl-L-arginine, and higher vascular ecto-adenosine deaminase were observed in 4T1 + 4PYR vs. 4T1 and control. Vascular relaxation caused by flow-dependent endothelial activation in 4PYR-treated mice was significantly lower than in control. The permeability of 4PYR-treated endothelial cells was increased. Decreased nicotinamide but enhanced nicotinamide metabolites were noticed in 4T1 vs. control. Reduced N-methylnicotinamide and a further increase in Met2PY were observed in 4T1 + 4PYR vs. 4T1 and control. In cultured breast cancer cells, estrogen and progesterone receptor antagonists inhibited the production of 4PYR metabolites. 4PYR formation is accelerated in cancer and induces metabolic disturbances that may affect cancer progression and, especially, metastasis, probably through impaired endothelial homeostasis. 4PYR may be considered a new oncometabolite.


Subject(s)
Carcinogens/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Niacinamide/pharmacology , Animals , Cell Line, Tumor , Cell Membrane Permeability , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Extracellular Space/metabolism , Female , Humans , Hydrolysis , Intracellular Space/metabolism , Mice , Niacinamide/analogs & derivatives , Niacinamide/toxicity , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
10.
Int J Mol Sci ; 22(18)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34576301

ABSTRACT

Organochlorine pesticides, such as DDT, methoxychlor, and their metabolites, have been characterized as endocrine disrupting chemicals (EDCs); suggesting that their modes of action involve interaction with or abrogation of endogenous endocrine function. This study examined whether embryonic thymocyte death and alteration of differentiation induced by the primary metabolite of methoxychlor, HPTE, rely upon estrogen receptor binding and concurrent T cell receptor signaling. Estrogen receptor inhibition of ERα or GPER did not rescue embryonic thymocyte death induced by HPTE or the model estrogen diethylstilbestrol (DES). Moreover, adverse effects induced by HPTE or DES were worsened by concurrent TCR and CD2 differentiation signaling, compared with EDC exposure post-signaling. Together, these data suggest that HPTE- and DES-induced adverse effects on embryonic thymocytes do not rely solely on ER alpha or GPER but may require both. These results also provide evidence of a potential collaborative signaling mechanism between TCR and estrogen receptors to mediate adverse effects on embryonic thymocytes, as well as highlight a window of sensitivity that modulates EDC exposure severity.


Subject(s)
Cell Differentiation , Endocrine Disruptors/toxicity , Estrogen Receptor alpha/metabolism , Phenols/toxicity , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Thymocytes/drug effects , Animals , CD2 Antigens/metabolism , Cell Death , Cells, Cultured , Estrogen Receptor alpha/antagonists & inhibitors , Female , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction , Thymocytes/cytology , Thymocytes/metabolism
11.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34480554

ABSTRACT

The selective estrogen receptor (ER) modulator, tamoxifen, is the only endocrine agent with approvals for both the prevention and treatment of premenopausal and postmenopausal estrogen-receptor positive breast cancer as well as for the treatment of male breast cancer. Endoxifen, a secondary metabolite resulting from CYP2D6-dependent biotransformation of the primary tamoxifen metabolite, N-desmethyltamoxifen (NDT), is a more potent antiestrogen than either NDT or the parent drug, tamoxifen. However, endoxifen's antitumor effects may be related to additional molecular mechanisms of action, apart from its effects on ER. In phase 1/2 clinical studies, the efficacy of Z-endoxifen, the active isomer of endoxifen, was evaluated in patients with endocrine-refractory metastatic breast cancer as well as in patients with gynecologic, desmoid, and hormone-receptor positive solid tumors, and demonstrated substantial oral bioavailability and promising antitumor activity. Apart from its potent anticancer effects, Z-endoxifen appears to result in similar or even greater bone agonistic effects while resulting in little or no endometrial proliferative effects compared with tamoxifen. In this review, we summarize the preclinical and clinical studies evaluating endoxifen in the context of breast and other solid tumors, the potential benefits of endoxifen in bone, as well as its emerging role as an antimanic agent in bipolar disorder. In total, the summarized body of literature provides compelling arguments for the ongoing development of Z-endoxifen as a novel drug for multiple indications.


Subject(s)
Receptors, Estrogen/antagonists & inhibitors , Tamoxifen/analogs & derivatives , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Estrogen Antagonists/pharmacology , Estrogen Antagonists/therapeutic use , Female , Humans , Molecular Targeted Therapy/methods , Receptors, Estrogen/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
12.
Mol Cell Endocrinol ; 537: 111426, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34391846

ABSTRACT

Estrogen receptor-positive (ER+) breast carcinomas are the most common subtype, corresponding to 60% of the cases in premenopausal and 75% in postmenopausal women. The third-generation of aromatase inhibitors (AIs), the non-steroidal Anastrozole (Ana) and Letrozole (Let) and the steroidal Exemestane (Exe), are considered a first-line endocrine therapy for postmenopausal women. Despite their clinical success, the development of resistance is the major setback in clinical practice. Nevertheless, the lack of cross-resistance between AIs hints that these drugs may act through distinct mechanisms. Therefore, this work studied the different effects induced by AIs on biological processes, such as cell proliferation, death, autophagy and senescence. Moreover, their effects on the regulation of the hormonal environment were also explored. The non-steroidal AIs induce senescence, through increased YPEL3 expression, on aromatase-overexpressing breast cancer cells (MCF-7aro), whereas Exe promotes a cytoprotective autophagy, thus blocking senescence induction. In addition, in a hormone-enriched environment, the non-steroidal AIs prevent estrogen signaling, despite up-regulating the estrogen receptor alpha (ERα), while Exe down-regulates ERα and maintains its activation. In these conditions, all AIs up-regulate the androgen receptor (AR) which blocks EGR3 transcription in Exe-treated cells. On the other hand, in hormone-depleted conditions, a crosstalk between AR and ERα occurs, enhancing the estrogenic effects of Exe. This indicates that Exe modulates both ERα and AR, while Ana and Let act as pure AIs. Thus, this study highlights the potential clinical benefit of combining AR antagonists with Exe and discourages the sequential use of Exe as second-line therapy in postmenopausal breast cancer.


Subject(s)
Apoptosis , Aromatase Inhibitors/pharmacology , Autophagy , Breast Neoplasms/pathology , Cellular Senescence , Hormones/metabolism , Anastrozole/pharmacology , Androstadienes/pharmacology , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Cyclins/metabolism , Early Growth Response Protein 3/genetics , Early Growth Response Protein 3/metabolism , Estrogens/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Letrozole/pharmacology , Receptors, Androgen/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Breast Cancer Res Treat ; 190(1): 53-67, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34448090

ABSTRACT

PURPOSE: Menopause is associated with an increased risk of estrogen receptor-positive (ER +) breast cancer. To characterize the metabolic shifts associated with reduced estrogen bioavailability on breast tissue, metabolomics was performed from ovary-intact and ovariectomized (OVX) female non-human primates (NHP). The effects of exogenous estrogen administration or estrogen receptor blockade (tamoxifen treatment) on menopause-induced metabolic changes were also investigated. METHODS: Bilateral ovariectomies were performed on female cynomolgus macaques (Macaca fascicularis) to model menopause. OVX NHP were then divided into untreated (n = 13), conjugated equine estrogen (CEE)-treated (n= 13), or tamoxifen-treated (n = 13) subgroups and followed for 3 years. Aged-matched ovary-intact female NHP (n = 12) were used as a premenopausal comparison group. Metabolomics was performed on snap-frozen breast tissue. RESULTS: Changes in several different metabolic biochemicals were noted, particularly in glucose and fatty acid metabolism. Specifically, glycolytic, Krebs cycle, acylcarnitines, and phospholipid metabolites were elevated in breast tissue from ovary-intact NHP and OVX + CEE in relation to the OVX and OVX + tamoxifen group. In contrast, treatment with CEE and tamoxifen decreased several cholesterol metabolites, compared to the ovary-intact and OVX NHP. These changes were accompanied by elevated bile acid metabolites in the ovary-intact group. CONCLUSION: Alterations in estrogen bioavailability are associated with changes in the mammary tissue metabolome, particularly in glucose and fatty acid metabolism. Changes in these pathways may represent a bioenergetic shift in gland metabolism at menopause that may affect breast cancer risk.


Subject(s)
Breast Neoplasms , Estrogens , Hormone Replacement Therapy , Receptors, Estrogen/antagonists & inhibitors , Aged , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Replacement Therapy , Estrogens, Conjugated (USP) , Female , Humans , Macaca fascicularis , Ovariectomy , Receptors, Estrogen/genetics
14.
Breast Cancer Res ; 23(1): 87, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34425869

ABSTRACT

BACKGROUND: In MONARCH 2, abemaciclib plus fulvestrant significantly improved median progression-free survival (PFS, 16.4 vs 9.3 months, hazard ratio [HR] 0.553) and overall survival (OS, 46.7 vs 37.3 months; HR 0.757) compared with placebo plus fulvestrant in hormone receptor-positive (HR-positive), human epidermal growth factor receptor 2-negative (HER2-negative) advanced breast cancer (ABC) patients who were endocrine therapy (ET) resistant, regardless of menopausal status. Here, we report findings in the premenopausal subgroup of the MONARCH 2 trial. METHODS: The premenopausal subgroup included patients with natural menstrual bleeding who received a gonadotropin-releasing hormone agonist at least 4 weeks prior to study treatment start date and for the entire study duration. Of the 669 patients enrolled in the MONARCH 2 trial, 114 were premenopausal (abemaciclib plus fulvestrant, n = 72; placebo plus fulvestrant, n = 42), and were included in this analysis. The primary objective was investigator-assessed PFS and secondary objectives were OS, objective response rate, and safety and tolerability. Exploratory analyses included time to second disease progression (PFS2), time to chemotherapy (TTC), and chemotherapy-free survival (CFS). RESULTS: At the primary objective cutoff (February 14, 2017), median PFS was not reached for the abemaciclib plus fulvestrant arm versus 10.52 months for the placebo plus fulvestrant arm (HR 0.415; 95% CI 0.246-0.698). At the pre-specified OS interim cutoff (20-June-2019), median PFS was 28.6 months in the abemaciclib plus fulvestrant arm compared with 10.26 months in the placebo plus fulvestrant arm (HR 0.477; 95% CI 0.302-0.755). A numerical OS benefit was observed with abemaciclib plus fulvestrant compared to fulvestrant alone (HR 0.689; 95% CI 0.379-1.252, median, not reached vs 47.3 months). Improvements were also observed for the exploratory outcomes of PFS2 (HR 0.599), TTC (HR 0.674), and CFS (HR 0.642) with the addition of abemaciclib to fulvestrant. The safety profile was generally consistent with results disclosed previously. CONCLUSIONS: Results of the premenopausal subgroup in the MONARCH 2 trial were consistent with the improved clinical outcomes observed in the intent-to-treat population. The analysis provides support for the use of abemaciclib plus fulvestrant (with ovarian suppression) as an effective treatment option for premenopausal patients with HR+, HER2- ABC who are ET-resistant. CLINICAL TRIAL REGISTRATION: NCT02107703. Registered April 08, 2014- Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT02107703 .


Subject(s)
Aminopyridines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Breast Neoplasms/drug therapy , Fulvestrant/therapeutic use , Adult , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Female , Humans , Middle Aged , Premenopause , Progression-Free Survival , Receptor, ErbB-2/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Response Evaluation Criteria in Solid Tumors , Survival Rate
15.
Toxicology ; 460: 152871, 2021 08.
Article in English | MEDLINE | ID: mdl-34303733

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a widespread persistent organic pollutant. Both epidemiological survey and our previous in vivo study have revealed the associations between PFOS exposure and spermatogenesis disorder, while the underlying mechanisms are far from clear. In the present study, GC-2 cells, a mouse spermatocyte-derived cell line, was used to investigate the toxic effects of PFOS and its hypothetical mechanism of action. GC-2 cells were treated with PFOS (0, 50, 100 and 150 µM) for 24 h or 48 h. Results demonstrated that PFOS dose-dependently inhibited cell viability, induced G0/G1 cell cycle arrest and triggered apoptosis, which might be partly explained by the decrease in cyclin D1, PCNA and Bcl-2 protein expression; increase in Bax protein expression; and activation of caspase-9, -3. In addition, PFOS did not directly transactivate or repress estrogen receptors (ERs) in gene reporter assays, whereas the protein levels of both ERα and ERß were significantly altered and the downstream ERK1/2 phosphorylation was inhibited by PFOS. Furthermore, pretreatment with specific ERα agonist PPT (1 µM) significantly attenuated the above PFOS-induced effects while specific ERß agonist DPN (1 µM) accelerated them. These results suggest that PFOS may induce growth inhibition and apoptosis via non-genomic estrogen receptor/ERK1/2 signaling pathway in GC-2 cells, which provides a novel insight regarding the potential role of ERs in mediating PFOS-triggered spermatocyte toxicity.


Subject(s)
Alkanesulfonic Acids/toxicity , Apoptosis/drug effects , Fluorocarbons/toxicity , MAP Kinase Signaling System/drug effects , Receptors, Estrogen/antagonists & inhibitors , Spermatocytes/drug effects , Animals , Apoptosis/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Dose-Response Relationship, Drug , MAP Kinase Signaling System/physiology , Male , Mice , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Spermatocytes/metabolism
16.
Environ Toxicol Pharmacol ; 87: 103688, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34119701

ABSTRACT

Multiple substances are considered endocrine disrupting chemicals (EDCs). However, there is a significant gap in the early prioritization of EDC's effects. In this work, in silico and in vitro methods were used to model estrogenicity. Two Quantitative Structure-Activity Relationship (QSAR) models based on Logistic Regression and REPTree algorithms were built using a large and diverse database of estrogen receptor (ESR) agonism. A 10-fold external validation demonstrated their robustness and predictive capacity. Mechanistic interpretations of the molecular descriptors (C-026, nArOH,PW5, B06[Br-Br]) used for modelling suggested that the heteroatomic fragments, aromatic hydroxyls, and bromines, and the relative bond accessibility areas of molecules, are structural determinants in estrogenicity. As validation of the QSARs, ESR transactivity of thirteen persistent organic pollutants (POPs) and suspected EDCs was tested in vitro using the MMV-Luc cell line. A good correspondence between predictions and experimental bioassays demonstrated the value of the QSARs for prioritization of ESR agonist compounds.


Subject(s)
Endocrine Disruptors/toxicity , Estrogens/toxicity , Receptors, Estrogen/metabolism , Algorithms , Cell Line, Tumor , Cell Survival/drug effects , Computer Simulation , Endocrine Disruptors/chemistry , Endocrine Disruptors/classification , Estrogens/chemistry , Estrogens/classification , Humans , Models, Chemical , Quantitative Structure-Activity Relationship , Receptors, Estrogen/antagonists & inhibitors
17.
Breast Cancer Res ; 23(1): 57, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34020697

ABSTRACT

BACKGROUND: Endocrine therapies targeting estrogen signaling have significantly improved breast cancer (BC) patient survival, although 40% of ERα-positive BCs do not respond to those therapies. Aside from genomic signaling, estrogen triggers non-genomic pathways by forming a complex containing methylERα/Src/PI3K, a hallmark of aggressiveness and resistance to tamoxifen. We aimed to confirm the prognostic value of this complex and investigated whether its targeting could improve tumor response in vivo. METHODS: The interaction of ERα/Src and ERα/PI3K was studied by proximity ligation assay (PLA) in a cohort of 440 BC patients. We then treated patient-derived BC xenografts (PDXs) with fulvestrant or the PI3K inhibitor alpelisib (BYL719) alone or in combination. We analyzed their anti-proliferative effects on 6 ERα+ and 3 ERα- PDX models. Genomic and non-genomic estrogen signaling were assessed by measuring ERα/PI3K interaction by PLA and the expression of estrogen target genes by RT-QPCR, respectively. RESULTS: We confirmed that ERα/Src and ERα/PI3K interactions were associated with a trend to poorer survival, the latter displaying the most significant effects. In ERα+ tumors, the combination of BYL719 and fulvestrant was more effective than fulvestrant alone in 3 models, irrespective of PI3K, PTEN status, or ERα/PI3K targeting. Remarkably, resistance to fulvestrant was associated with non-genomic ERα signaling, since genomic degradation of ERα was unaltered in these tumors, whereas the treatment did not diminish the level of ERα/PI3K interaction. Interestingly, in 2 ERα- models, fulvestrant alone impacted tumor growth, and this was associated with a decrease in ERα/PI3K interaction. CONCLUSIONS: Our results demonstrate that ERα/PI3K may constitute a new prognostic marker, as well as a new target in BC. Indeed, resistance to fulvestrant in ERα+ tumors was associated with a lack of impairment of ERα/PI3K interaction in the cytoplasm. In addition, an efficient targeting of ERα/PI3K in ERα- tumors could constitute a promising therapeutic option.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Fulvestrant/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Receptors, Estrogen/metabolism , Thiazoles/therapeutic use , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Female , Genomics , Humans , Mice , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Estrogen/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
18.
Breast Cancer ; 28(5): 1062-1071, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34047951

ABSTRACT

BACKGROUND: Previously, we reported that Wnt5a-positive breast cancer can be classified as estrogen receptor (ER)-positive breast cancer; its prognosis is worse than that of Wnt5a-negative breast cancer. This study aimed to investigate the mechanisms underlying the poor prognosis in Wnt5a-positive breast cancer patients. METHODS: In total, 151 consecutive ER-positive breast cancer patients who underwent resection between January 2011 and February 2014 were enrolled. DNA microarray and pathway analyses were conducted using MCF-7 cells stably expressing Wnt5a [MCF-7/Wnt5a (+)]. Based on the outcomes, cell viability/drug sensitivity assays, and mutation analysis were performed using cell cultures and breast cancer tissues. The relationship between Wnt5a and the PI3K-AKT-mTOR signaling pathway was also examined. RESULTS: The relapse-free survival rate in patients with Wnt5a-positive breast cancer was significantly lower than that in patients with Wnt5a-negative breast cancer (P = 0.047). DNA microarray data suggest that only the cytochrome P450 (CYP) pathway was significantly upregulated in MCF-7/Wnt5a (+) cells (P = 0.0440). Additionally, MCF-7/Wnt5a (+) cells displayed reduced sensitivity to the metabolic substrates of CYP, tamoxifen (P < 0.001), paclitaxel (P < 0.001), and cyclophosphamide (P < 0.001). Of note, PIK3CA mutations were not associated with the expression of Wnt5a in breast cancer tissue and culture cells. CONCLUSIONS: In ER-positive breast cancer, Wnt5a upregulates the CYP metabolic pathway and suppresses tamoxifen, paclitaxel, and cyclophosphamide resistance, all of the three, standard treatment methods for ER-positive breast cancer. Wnt5a is thus potentially involved in the poor prognosis of ER-positive breast cancer independently of the PI3K-AKT-mTOR signaling pathway.


Subject(s)
Breast Neoplasms/genetics , Receptors, Estrogen/antagonists & inhibitors , Wnt-5a Protein/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Longitudinal Studies , MCF-7 Cells , Middle Aged , Progression-Free Survival , Receptor, ErbB-2 , Retrospective Studies , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Up-Regulation
19.
Eur J Med Chem ; 221: 113543, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34022716

ABSTRACT

Endocrine therapy (ET) has benefited patients with estrogen receptor alpha (ERα) positive breast cancer for decades. Selective estrogen receptor modulator (SERM) such as Tamoxifen represents the clinical standard of care (SoC). Despite the therapeutic importance of current SoC agents, 30-50% of prolonged treatment patients inevitably generated resistant tumor cells, usually eventually suffered tumor relapse and developed into metastatic breast cancer (MBC), which was the leading cause of female cancer-related mortality. Among these, most resistant tumors remained dependent on ERα signaling, which reignited the need for the next generation of ERα related agents. We hypothesized that selective estrogen receptor covalent antagonists targeting ERα would provide a therapeutic alternative. In the current work, series of novel benzothiophene hybrids bearing electrophile moieties were synthesized and biologically evaluated. The representative analogue 15c exhibited potent anti-proliferative effect in MCF-7 cell lines in vitro, and further mechanism studies confirmed the necessity of covalent bonding. More importantly, 15c could attenuate the expression of TFF-1, GREB-1 and downregulate the levels of cellular ERα protein.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Design , Estrogen Receptor Antagonists/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Thiophenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Female , Humans , MCF-7 Cells , Molecular Structure , Receptors, Estrogen/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Tumor Cells, Cultured
20.
Int J Mol Sci ; 22(7)2021 Apr 02.
Article in English | MEDLINE | ID: mdl-33918423

ABSTRACT

Estrogen-related receptor α (ERRα), which is overexpressed in a variety of cancers has been considered as an effective target for anticancer therapy. ERRα inverse agonists have been proven to effectively inhibit the migration and invasion of cancer cells. As few crystalline complexes have been reported, molecular dynamics (MD) simulations were carried out in this study to deepen the understanding of the interaction mechanism between inverse agonists and ERRα. The binding free energy was analyzed by the MM-GBSA method. The results show that the total binding free energy was positively correlated with the biological activity of an inverse agonist. The interaction of the inverse agonist with the hydrophobic interlayer composed of Phe328 and Phe495 had an important impact on the biological activity of inverse agonists, which was confirmed by the decomposition of energy on residues. As Glu331 flipped and formed a hydrogen bond with Arg372 in the MD simulation process, the formation of hydrogen bond interaction with Glu331 was not a necessary condition for the compound to act as an inverse agonist. These rules provide guidance for the design of new inverse agonists.


Subject(s)
Drug Inverse Agonism , Receptors, Estrogen/antagonists & inhibitors , Molecular Dynamics Simulation , ERRalpha Estrogen-Related Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...