10β-Hydroxyestra-1,4-diene-3,17-dione as potential antiproliferative agent: in vitro biological evaluation and in silico studies (original) (raw)
Related papers
10β-Hydroxyestra-1,4-diene-3,17-dione Does Not Bind to the Nuclear Estrogen Receptor α
bioRxiv (Cold Spring Harbor Laboratory), 2022
The lack of nuclear estrogen receptor (ERα and ERβ) bindings of 10β-hydroxyestra-1,4-diene-3,17-dione (HEDD) and structurally related steroidal para-quinols have been shown by an extensive series of multidisciplinary investigational evidence including specific receptor binding studies. In support of the latter, the absence of estrogen-derived para-quinols' in vivo uterotrophic effects has also been well documented. Via in silico docking, a recent publication by Canário et al. (2022) reported a robust binding of HEDD (Figure 1B) to ERα. The authors claimed a strong binding of HEDD as strong as that of its natural ligand, 17β-estradiol (E2), the main human estrogen. However, an examination of the virtual binding pocket revealed that at least one residue near the critical ligand-binding site of their reported HEDD-ERα complex was labelled as "unknown" indicating thereby alteration of the receptor's published structure (Tannenbaum et al, 1998; Bafna et al., 2020) to fit the ligand. Based on these arguments, the contradictory result by Canário et al. (2022) on HEDD's binding to ERα should be dismissed.
Apoptosis induction in HeLa cervical cancer cell line by steroidal 16,17-seco-16,17a-dinitriles
JSCS, 2022
Steroids are good candidates for drug development, thanks to their low general toxicity and possibility for structure modifications connected with change of their activity. Several 16,17-secoandrost-4-ene-16,17a-dinitrile compounds were synthesized and screened for anticancer effect previously, including 6-oxo and 6-hydroxyimino compounds. This research is continued with the attempts for different synthetic strategy and evaluation of anticancer effect mechanism. Synthesis of 3-hydroxyimino compounds was successful, but inseparable mix of isomers was excluded from biological tests. Tested secodinitriles expressed cytotoxic effect on HeLa cervix cancer cells as a model system, with submicromolar to molar IC 50 values, where 6-substituted derivatives were more effective. After 72 h treatment with equitoxic concentrations equal IC 50 values of test compounds the mechanism of this effect was studied using flow cytometry and specific fluorescent dyes. Modest change in both G0/G1 and G2/M resting phases and change in mitochondrial membrane potential were noticed, while the most pronounced effect was apoptosis induction. Total apoptosis was in range 50.72-58.31 % in all cell samples treated with secodinitriles, compared to 7.44 % in control samples. Total percent of dead cells, including both apoptotic and necrotic, ranged from 55.24 to 65.34 %, compared to 10.68 % in control. Selectivity towards cancer cells is very important feature of these compounds indicating their potential use as lead compounds in the drug development for the treatment of cancers of steroid hormone-dependent tissues.
ACS chemical neuroscience, 2018
Uterotrophic effect of 17α-estradiol, the C17 epimer of the main human estrogen 17β-estradiol, was shown to manifest in animal models at doses lower than those necessary for central outcome raising concerns about its potential to treat maladies of the central nervous system. We introduce here 10β,17α-dihydroxyestra-1,4-dien-3-one (α-DHED) that acts as a bioprecursor prodrug producing 17α-estradiol with remarkable selectivity to the brain and, therefore, without appreciable exposure of the periphery to the parent steroid. This distinguishing feature of α-DHED is shown by using an estrogen-responsive mouse model with complementary LC-MS/MS measurement of drug contents in target tissues. Our data warrant further research to fully establish the potential of α-DHED for a safe and efficacious 17α-estradiol-based neurotherapy.
Cancer research, 2002
2-Methoxyestradiol (2ME(2)) is an endogenous metabolite of 17beta-estradiol (E(2)) that arises from the hydroxylation and subsequent methylation at the 2-position. In vitro 2ME(2) inhibits a large variety of tumor and nontumor cell lines from diverse origins, as well as several stages of the angiogenic cascade. In vivo it has been shown to be a very effective inhibitor of tumor growth and angiogenesis in numerous models. Although various molecular targets have been proposed for this compound, the mechanism of action is still uncertain. As this molecule emerges as a drug candidate it is important to assess the estrogen receptors (ERs) as molecular targets for 2ME(2). The purpose of this study was to investigate whether 2ME(2) is able to engage ERs as an agonist and whether its antiproliferative activities are mediated through ERs. We confirm that 2ME(2) has a lower binding affinity for ERalpha as compared with E(2) and other E(2) metabolites and antagonists, and we demonstrate that t...
2-Methoxyestradiol, a Promising Anticancer Agent
Pharmacotherapy, 2003
Estrogens occurring naturally in the body are metabolized to catecholestrogens (2-and 4-hydroxyestradiol) by the cytochrome P450 enzymes. 2-Hydroxy catecholestrogens are further metabolized by catechol-O-methyltransferase to 2-methoxyestradiol, which is known to be protective against tumor formation. 2-Methoxyestradiol exhibits potent apoptotic activity against rapidly growing tumor cells. It also possesses antiangiogenic activity through a direct apoptotic effect on endothelial cells. Other molecular mechanisms, including microtubule stabilization by inhibition of the colchicine-binding site, have been reported. The exact mechanism of action of 2-methoxyestradiol is still unclear, but it has been shown to be effective in preventing tumor growth in a variety of cell lines. 2-Methoxyestradiol also possesses cardioprotective activity by inhibiting vascular smooth muscle cell growth in arteries. It has a lower binding affinity for estrogen receptor ␣ compared with that of estradiol, and its affinity for estrogen receptor  is even lower than that of estrogen receptor ␣, thus it has minimal estrogenic activity. 2-Methoxyestradiol is distinct because of its inability to engage estrogen receptors as an agonist, and its unique antiproliferative and apoptotic activities are mediated independently of estrogen receptors ␣ and . A phase I clinical trial of 2-methoxyestradiol 200, 400, 600, 800, and 1000 mg/day in 15 patients with breast cancer showed significant reduction in bone pain and analgesic intake in some patients, with no significant adverse effects. Another phase I study of 2-methoxyestradiol 200-1000 mg/day in combination with docetaxel 35 mg/m 2 /week for 4-6 weeks performed in 15 patients with advanced refractory metastatic breast cancer showed no serious drug-related adverse effects. A phase II randomized, double-blind trial of 2-methoxyestradiol 400 and 1200 mg/day in 33 patients with hormone-refractory prostate cancer showed that it was well tolerated and showed prostate specific antigen stabilizations and declines. We have started a phase I clinical trial to explore dosages greater than 1000 mg/day.
Molecules, 2014
Objective: 2-Methoxyestradiol, one of the natural 17β-estradiol derivatives, is a novel, potent anticancer agent currently being evaluated in advanced phases of clinical trials. The main goal of the study was to investigate the anticancer activity of 2-methoxy-estradiol towards osteosarcoma cells and its possible neurodegenerative effects. We used an experimental model of neurotoxicity and anticancer activity of the physiological agent, 2-methoxyestradiol. Thus, we used highly metastatic osteosarcoma 143B and mouse immortalized hippocampal HT22 cell lines. The cells were treated with pharmacological (1 μM, 10 μM) concentrations of 2-methoxyestradiol. Experimental: Neuronal nitric oxide synthase and 3-nitrotyrosine protein levels were determined by western blotting. Cell viability and induction of cell death were measured by MTT and PI/Annexin V staining and a DNA fragmentation ELISA kit, respectively. Intracellular levels of nitric oxide were determined by flow cytometry. Results: Here we demonstrated that the signaling pathways of neurodegenerative diseases and cancer may overlap. We presented evidence that 2-methoxyestradiol, in contrast to 17β-estradiol, specifically affects neuronal nitric oxide synthase and augments 3-nitrotyrosine level leading to osteosarcoma and immortalized hippocampal cell death. Conclusions: We report the dual facets of 2-methoxyestradiol, that causes cancer cell death, but on the other hand may play a key role as a neurotoxin.
Cancer research, 2002
2-Methoxyestradiol (2ME 2) is an endogenous metabolite of 17-estradiol (E 2) that arises from the hydroxylation and subsequent methylation at the 2-position. In vitro 2ME 2 inhibits a large variety of tumor and nontumor cell lines from diverse origins, as well as several stages of the angiogenic cascade. In vivo it has been shown to be a very effective inhibitor of tumor growth and angiogenesis in numerous models. Although various molecular targets have been proposed for this compound, the mechanism of action is still uncertain. As this molecule emerges as a drug candidate it is important to assess the estrogen receptors (ERs) as molecular targets for 2ME 2. The purpose of this study was to investigate whether 2ME 2 is able to engage ERs as an agonist and whether its antiproliferative activities are mediated through ERs. We confirm that 2ME 2 has a lower binding affinity for ER␣ as compared with E 2 and other E 2 metabolites and antagonists, and we demonstrate that the affinity of 2ME 2 for ER is even lower. When assessed in the presence of galangin, a cytochrome P450 enzyme inhibitor, at concentrations at which 2ME 2 interacts with ER␣ in an in vitro binding assay, it does not stimulate the proliferation of an estrogen-dependent breast carcinoma cell line. Similar IC 50 values for inhibition of proliferation and induction of apoptosis are obtained in estrogen-dependent and estrogen-independent human breast cancer cell lines, irrespective of the expression of ER␣ and ER. Moreover, the estrogen antagonist ICI 182,780 does not inhibit the antiproliferative activity of 2ME 2. In E 2-responsive cells such as MCF-7 and human umbilical vascular endothelial cells, high levels of E 2 inhibit the antiproliferative activity of ICI 182,780 but not of 2ME 2. Collectively, these results suggest that 2ME 2 is distinct among estradiol metabolites because of its inability to engage ERs as an agonist, and its unique antiproliferative and apoptotic activities are mediated independently of ER␣ and ER.
Journal of Experimental & Clinical Cancer Research, 2013
Background Tumors are diseases characterized by uncontrolled cell growth and, in spite of the progress of medicine over the years, continue to represent a major threat to the health, requiring new therapies. Several synthetic compounds, such as those derived from natural sources, have been identified as anticancer drugs; among these compounds quinone represent the second largest class of anticancer agents in use. Several studies have shown that these act on tumor cells through several mechanisms. An important objective of this work is to develop quinoidscompounds showing antitumor activity, but with fewer side effects. The parachinone cannabinol HU-331, is a small molecule that with its core 4-hydroxy-1,4-benzoquinone, exhibits a potent and selective cytotoxic activity on different tumor cell lines. A series of derivatives 3-hydroxy-1,4-benzochinoni were thus developed through HU-331 chemical modifications. The purpose of the work is to test the ability of the compounds to induce pr...
RSC Advances, 2014
Syntheses of 2-methoxyestradiol (1) and eugenol template based conformationally flexible and rigid diarylpropenes, 14(a-l) and 20(a-e), as nonsteroidal anticancer agents have been performed. The synthesized compounds were evaluated for their anticancer activity in in vitro using a panel of human cancer cell lines viz. MCF-7, A549, DU 145, KB and MDA-MB-231by SRB assay. Compounds 14i, 14k and 15a showed significant anticancer activity at IC 50 between 10.27 mM to 27.91 mM in different cancer cell lines. The most active molecule, 14k, inhibited proliferation of cells by inducing apoptosis and arresting the cell cycle at the G2/M phase. In vitro toxicity of these compounds (14i, 14k and 15a) in healthy hepatic monocyte (THP-1) cells showed high selectivity of compounds towards cancerous vs. healthy cells. Scheme 1 Reagent and conditions (a) KOH in ethanol, 35-40 C; (b) 10% Pd/C, ethylacetate-methanol (1 : 1), 35-40 C; (c) sodium borohydride, dry ethanol, 35-40 C; (d) HCl, methanol, reflux. a In case of hydroxy derivatives of 10, R ¼ THP which was deprotected during workup and gave hydroxy derivative of 11. Scheme 2 Reagent and conditions (a) (i) KOH in ethanol, 35-40 C (ii) HCl; (b) 10% Pd/C, ethylacetate-methaol (1 : 1), 35-40 C; (c) sodium borohydride, dry ethanol, 35-40 C; (d) HCl, methanol, reflux. a In case of hydroxy derivatives of 10, R ¼ THP which was deprotected during workup and gave hydroxy derivative of 17. 35172 | RSC Adv., 2014, 4, 35171-35185 This journal is