Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-39059983

ABSTRACT

Glioblastoma (GBM), a malignant brain tumor originating in glial cells, is one of the most common primary brain malignancies, affecting one in 100,000 people, typically in the frontal lobe. Estrogens, like estradiol-17 (E2), significantly influence GBM progression, metastasis, and angiogenesis. Estrogen receptors (ERs) are crucial in signal transduction and physiology, making them potential therapeutic targets. However, their roles in GBM pathogenesis remain unclear. This review explores ERs in GBM, focusing on their involvement in tumor immune evasion, modulation of the tumor microenvironment, and the mechanisms underlying GBM progression. Additionally, therapeutic opportunities targeting ERs for GBM treatment are discussed. Estrogen, synthesized primarily in ovaries and in smaller amounts by adrenal glands and fat tissues, regulates reproductive systems, bone density, skin health, and cardiovascular function. The invasive nature and heterogeneity of GBM complicate therapy development. Preclinical findings suggest that endocrine therapy with hormone receptor agonists or antagonists can extend patient survival and improve post-treatment quality of life. The ERß pathway, in particular, shows tumor-suppressive potential, limiting glioma progression with fewer side effects. ERß agonists could become a novel drug class for GBM treatment. Identifying biomarkers and specific therapeutic targets is crucial for early detection and improved prognosis. Estrogen and its receptors are advantageous for GBM treatment due to their regulation of numerous biological processes, ability to penetrate the blood-brain barrier, and genomic and non-genomic control of transcription, making them promising targets for GBM therapy.


Subject(s)
Brain Neoplasms , Glioblastoma , Receptors, Estrogen , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Receptors, Estrogen/metabolism , Receptors, Estrogen/antagonists & inhibitors , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Antagonists/chemistry , Tumor Microenvironment/drug effects
2.
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
3.
Eur J Med Chem ; 227: 113869, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34710747

ABSTRACT

New twelve in silico designed coumarin-based ERα antagonists, namely 3DQ-1a to 3DQ-1е, were synthesized and confirmed as selective ERα antagonists, showing potencies ranging from single-digit nanomolar to picomolar. The hits were confirmed as selective estrogen receptor modulators and validated as antiproliferative agents using MCF-7 breast cancer cell lines exerting from picomolar to low nanomolar potency, at the same time showing no agonistic activity within endometrial cell lines. Their mechanism of action was inspected and revealed to be through the inhibition of the Raf-1/MAPK/ERK signal transduction pathway, preventing hormone-mediated gene expression on either genomic direct or genomic indirect level, and stopping the MCF-7 cells proliferation at G0/G1 phase. In vivo experiments, by means of the per os administration to female Wistar rats with pre-induced breast cancer, distinguished six derivatives, 3DQ-4a, 3DQ-2a, 3DQ-1a, 3DQ-1b, 3DQ-2b, and 3DQ-3b, showing remarkable potency as tumor suppressors endowed with optimal pharmacokinetic profiles and no significant histopathological profiles. The presented data indicate the new compounds as potential candidates to be submitted in clinical trials for breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coumarins/pharmacology , Drug Design , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Molecular Structure , Rats , Rats, Wistar , Structure-Activity Relationship
4.
J Med Chem ; 64(16): 11841-11856, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34251202

ABSTRACT

Breast cancer remains a leading cause of cancer death in women, representing a significant unmet medical need. Here, we disclose our discovery efforts culminating in a clinical candidate, 35 (GDC-9545 or giredestrant). 35 is an efficient and potent selective estrogen receptor degrader (SERD) and a full antagonist, which translates into better antiproliferation activity than known SERDs (1, 6, 7, and 9) across multiple cell lines. Fine-tuning the physiochemical properties enabled once daily oral dosing of 35 in preclinical species and humans. 35 exhibits low drug-drug interaction liability and demonstrates excellent in vitro and in vivo safety profiles. At low doses, 35 induces tumor regressions either as a single agent or in combination with a CDK4/6 inhibitor in an ESR1Y537S mutant PDX or a wild-type ERα tumor model. Currently, 35 is being evaluated in Phase III clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Carbolines/therapeutic use , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carbolines/chemistry , Carbolines/pharmacokinetics , Dogs , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacokinetics , Female , Humans , MCF-7 Cells , Macaca fascicularis , Mice , Molecular Structure , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
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
6.
Eur J Med Chem ; 192: 112191, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32169784

ABSTRACT

Up to 80% of mammary carcinoma initially exhibit estrogen-dependent growth, which can be treated by aromatase inhibitors or SERMs/SERDs. To increase the options after failure of the hormonal therapy with these drugs, the search for alternatives with a different mode of action to prevent estrogen action is of high relevance. Therefore, this study focused on the inhibition of coactivator recruitment at the estrogen receptor (ER) by targeted attachment of bivalent compounds at the coactivator binding site besides the primary binding at the ligand binding domain. Eight homodimeric 4-[1-(4-hydroxyphenyl)-2-phenyl-1-butenyl]cinnamic acid (GW7604)- or cyclofenilacrylic acid-based ER ligands with diaminoalkane linkers (C2-C5) were synthesized and their effects on the ER subtypes were assessed in vitro. All compounds possessed full antagonistic potency at ERα/ß as determined in a transactivation assay. Furthermore, they exerted medium downregulatory effects dependent on the spacer length and did not stimulate the ER expression as observed for 4-hydroxytamoxifen. The cyclofenil-derived dimer with C4 spacer (15b) showed the highest binding affinity to ERα (RBA = 79.2%) and downregulated the ER content in MCF-7 cells with an efficiency of 38% at 1 µM.


Subject(s)
Alkenes/pharmacology , Cyclofenil/pharmacology , Down-Regulation/drug effects , Drug Development , Estrogen Receptor Antagonists/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Alkenes/chemical synthesis , Alkenes/chemistry , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cyclofenil/chemical synthesis , Cyclofenil/chemistry , Dose-Response Relationship, Drug , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Humans , Molecular Structure , Receptors, Estrogen/metabolism , Structure-Activity Relationship
7.
Bioorg Med Chem ; 28(3): 115274, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31879182

ABSTRACT

17ß-Estradiol (E2) is a natural steroid ligand for the structurally and physiologically independent estrogen receptors (ERs) ERα and ERß. We recently observed that CF3-containing bisphenol AF (BPAF) works as an agonist for ERα but as an antagonist for ERß. Similar results were also observed for the CCl3-containing bisphenol designated as HPTE. Both BPAF and HPTE are comprised of a tri-halogenated methyl group in the central alkyl moiety of their bisphenol structures, which strongly suggests that halogens contribute directly to the agonist/antagonist dual biological functions. We conducted this study to investigate the structure-activity relationships by assessing together newly synthesized CF3- and CBr3-containing bisphenol E analogs (BPE-X). We first tested bisphenols for their receptor binding ability and then for their transcriptional activities. Halogen-containing bisphenols were found to be fully active for ERα, but almost completely inactive for ERß. When we examined these bisphenols for their inhibitory activities for E2 in ERß, we observed that they worked as distinct antagonists. The ascending order of agonist/antagonist dual biological functions was BPE-F < BPE-Cl (HPTE) ≤ BPAF < BPE-Br, demonstrating that the electrostatic halogen bonding effect is a major driving force of the bifunctional ERα agonist and ERß antagonist activities of BPAF.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogens/pharmacology , Phenols/pharmacology , Benzhydryl Compounds/chemical synthesis , Benzhydryl Compounds/chemistry , Dose-Response Relationship, Drug , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/chemical synthesis , Estrogens/chemistry , HeLa Cells , Humans , Molecular Structure , Phenols/chemical synthesis , Phenols/chemistry , Structure-Activity Relationship
8.
Yakugaku Zasshi ; 139(8): 1101-1105, 2019.
Article in Japanese | MEDLINE | ID: mdl-31366846

ABSTRACT

In order to implement academic detailing, it is extremely important to understand the pharmacological activity of drugs based on their chemical structures. To construct a database for academic detailing, a viewpoint for clinically utilizing the fundamental pharmacological features of a drug is required. These fundamental pharmacological features include chemical characteristics such as chemical structures, physical characteristics such as pharmaceutical formulations for efficient drug delivery to target organs, and a pharmacological viewpoint, which is a mechanism by which a drug is determined to be effective. In addition, in vivo kinetics are included in a drug's pharmacological features, i.e., a drug's excretion through urine and feces via the kidneys, its side effects due to differences in enzyme type when the drug is metabolized in the liver, as well as the capacity of the patient's current enzyme profile. This review describes academic detailing based on the chemical structure of drugs for breast cancer hormone therapy, as an example.


Subject(s)
Drug Information Services , Molecular Structure , Pharmaceutical Preparations/chemistry , Pharmacology , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Chemical Phenomena , Drug Compounding , Drug Delivery Systems , Drug-Related Side Effects and Adverse Reactions , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/therapeutic use , Female , Humans , Liver/metabolism , Molecular Conformation , Pharmacokinetics
9.
Curr Top Med Chem ; 19(15): 1318-1337, 2019.
Article in English | MEDLINE | ID: mdl-31215379

ABSTRACT

Breast cancer is the most common cancer suffered by female, and the second highest cause of cancer-related death among women worldwide. At present, hormone therapy is still the main treatment route and can be divided into three main categories: selective estrogen receptor modulators (SERMs), selective estrogen receptor downregulators (SERDs), and aromatase inhibitors (AIs). However, breast cancer is difficult to cure even after several rounds of anti-estrogen therapy and most drugs have serious side-effects. Here, we review the literature published over the past five years regarding the isolation and synthesis of analogs and their derivatives.


Subject(s)
Antineoplastic Agents/pharmacology , Aromatase Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/isolation & purification , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/isolation & purification , Estrogen Receptor Modulators/chemistry , Estrogen Receptor Modulators/isolation & purification , Estrogen Receptor alpha/metabolism , Female , Humans , Molecular Structure
10.
J Med Chem ; 61(18): 8155-8173, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30053783

ABSTRACT

In this work, we developed a small library of novel OBHS-RES hybrid compounds with dual inhibition activities targeting both the estrogen receptor α (ERα) and NF-κB by incorporating resveratrol (RES), a known inhibitor of NF-κB, into a privileged indirect antagonism structural motif (OBHS, oxabicycloheptene sulfonate) of estrogen receptor (ER). The OBHS-RES conjugates could bind well to ER and showed remarkable ERα antagonistic activity, and they also exhibited excellent NO inhibition in macrophage RAW 264.7 cells. Compared with 4-hydroxytamoxifen, some of them showed better antiproliferative efficacy in MCF-7 cell lines with IC50 up to 3.7 µM. In vivo experiments in a MCF-7 breast cancer model in Balb/c nude mice indicated that compound 26a was more potent than tamoxifen. Exploration of the compliancy of the structure against ER specificity utilizing these types of isomeric three-dimensional ligands indicated that one enantiomer had much better biological activity than the other.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Estrogen Receptor alpha/antagonists & inhibitors , Inflammation/drug therapy , NF-kappa B/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cells, Cultured , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Ligands , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Structure , Protein Conformation , Resveratrol/chemistry , Signal Transduction , Structure-Activity Relationship , Sulfones/chemistry
11.
Cancer Discov ; 8(9): 1176-1193, 2018 09.
Article in English | MEDLINE | ID: mdl-29991605

ABSTRACT

Mutations in estrogen receptor alpha (ERα) that confer resistance to existing classes of endocrine therapies are detected in up to 30% of patients who have relapsed during endocrine treatments. Because a significant proportion of therapy-resistant breast cancer metastases continue to be dependent on ERα signaling, there remains a critical need to develop the next generation of ERα antagonists that can overcome aberrant ERα activity. Through our drug-discovery efforts, we identified H3B-5942, which covalently inactivates both wild-type and mutant ERα by targeting Cys530 and enforcing a unique antagonist conformation. H3B-5942 belongs to a class of ERα antagonists referred to as selective estrogen receptor covalent antagonists (SERCA). In vitro comparisons of H3B-5942 with standard-of-care (SoC) and experimental agents confirmed increased antagonist activity across a panel of ERαWT and ERαMUT cell lines. In vivo, H3B-5942 demonstrated significant single-agent antitumor activity in xenograft models representing ERαWT and ERαY537S breast cancer that was superior to fulvestrant. Lastly, H3B-5942 potency can be further improved in combination with CDK4/6 or mTOR inhibitors in both ERαWT and ERαMUT cell lines and/or tumor models. In summary, H3B-5942 belongs to a class of orally available ERα covalent antagonists with an improved profile over SoCs.Significance: Nearly 30% of endocrine therapy-resistant breast cancer metastases harbor constitutively activating mutations in ERα. SERCA H3B-5942 engages C530 of both ERαWT and ERαMUT, promotes a unique antagonist conformation, and demonstrates improved in vitro and in vivo activity over SoC agents. Importantly, single-agent efficacy can be further enhanced by combining with CDK4/6 or mTOR inhibitors. Cancer Discov; 8(9); 1176-93. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 1047.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor Antagonists/administration & dosage , Estrogen Receptor alpha/antagonists & inhibitors , Indazoles/administration & dosage , Mutation , Administration, Oral , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cysteine/antagonists & inhibitors , Drug Screening Assays, Antitumor , Drug Synergism , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Female , Humans , Indazoles/chemistry , Indazoles/pharmacology , MCF-7 Cells , Mice , Protein Conformation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
12.
PLoS One ; 13(5): e0196312, 2018.
Article in English | MEDLINE | ID: mdl-29787591

ABSTRACT

Insulin-like growth factor 1 receptor (IGF-1R) is an important therapeutic target for breast cancer treatment. The alteration in the IGF-1R associated signaling network due to various genetic and environmental factors leads the system towards metastasis. The pharmacophore modeling and logical approaches have been applied to analyze the behaviour of complex regulatory network involved in breast cancer. A total of 23 inhibitors were selected to generate ligand based pharmacophore using the tool, Molecular Operating Environment (MOE). The best model consisted of three pharmacophore features: aromatic hydrophobic (HyD/Aro), hydrophobic (HyD) and hydrogen bond acceptor (HBA). This model was validated against World drug bank (WDB) database screening to identify 189 hits with the required pharmacophore features and was further screened by using Lipinski positive compounds. Finally, the most effective drug, fulvestrant, was selected. Fulvestrant is a selective estrogen receptor down regulator (SERD). This inhibitor was further studied by using both in-silico and in-vitro approaches that showed the targeted effect of fulvestrant in ER+ MCF-7 cells. Results suggested that fulvestrant has selective cytotoxic effect and a dose dependent response on IRS-1, IGF-1R, PDZK1 and ER-α in MCF-7 cells. PDZK1 can be an important inhibitory target using fulvestrant because it directly regulates IGF-1R.


Subject(s)
Antineoplastic Agents/pharmacology , Estradiol/analogs & derivatives , Receptors, Somatomedin/antagonists & inhibitors , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Databases, Pharmaceutical , Drug Evaluation, Preclinical , Estradiol/chemistry , Estradiol/pharmacology , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , Gene Expression/drug effects , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Ligands , MCF-7 Cells , Membrane Proteins , Models, Chemical , Models, Molecular , Receptor, IGF Type 1 , Receptors, Somatomedin/chemistry , Receptors, Somatomedin/genetics , Signal Transduction/drug effects , User-Computer Interface
13.
Bioorg Med Chem ; 26(8): 1638-1642, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29525335

ABSTRACT

The estrogen receptor (ER), a member of the nuclear receptor (NR) family, is involved in the regulation of physiological effects such as reproduction and bone homeostasis. Approximately 70% of human breast cancers are hormone-dependent and ERα-positive, and, thus, ER antagonists are broadly used in breast cancer therapy. We herein designed and synthesized a set of ER antagonists with a 4-heterocycle-4-phenylheptane skeleton.


Subject(s)
Estrogen Receptor Antagonists/pharmacology , Heptanes/pharmacology , Indoles/pharmacology , Pyrroles/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Thiophenes/pharmacology , Dose-Response Relationship, Drug , Drug Design , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Heptanes/chemical synthesis , Heptanes/chemistry , Humans , Indoles/chemical synthesis , Indoles/chemistry , Ligands , MCF-7 Cells , Models, Molecular , Molecular Structure , Pyrroles/chemical synthesis , Pyrroles/chemistry , Receptors, Estrogen/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Tumor Cells, Cultured
14.
Sci Rep ; 8(1): 649, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29330437

ABSTRACT

Somatic mutations of the Estrogen Receptor α (ERα) occur with an up to 40% incidence in ER sensitive breast cancer (BC) patients undergoing prolonged endocrine treatments. These polymorphisms are implicated in acquired resistance, disease relapse, and increased mortality rates, hence representing a current major clinical challenge. Here, multi-microseconds (12.5 µs) molecular dynamics simulations revealed that recurrent ERα polymorphisms (i. e. L536Q, Y537S, Y537N, D538G) (mERα) are constitutively active in their apo form and that they prompt the selection of an agonist (active)-like conformation even upon antagonists binding. Interestingly, our simulations rationalize, for the first time, the efficacy profile of (pre)clinically used Selective Estrogen Receptor Modulators/Downregulators (SERMs/SERDs) against these variants, enlightening, at atomistic level of detail, the key common structural traits needed by drugs able to effectively fight refractory BC types. This knowledge represents a key advancement for mechanism-based therapeutics targeting resistant ERα isoforms, potentially allowing the community to move a step closer to 'precision medicine' calibrated on patients' genetic profiles and disease progression.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Female , Humans , Models, Molecular , Molecular Dynamics Simulation , Polymorphism, Single Nucleotide/drug effects , Protein Structure, Secondary
15.
Pharmacol Ther ; 186: 1-24, 2018 06.
Article in English | MEDLINE | ID: mdl-29289555

ABSTRACT

Breast cancer is the most frequently diagnosed cancer in women, with estrogen receptor positive (ER+) breast cancer making up approximately 75% of all breast cancers diagnosed. Given the dependence on active ER signaling in these tumors, the predominant treatment strategy has been to inhibit various aspects of this pathway including directly antagonizing ER with the use of selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs). Interestingly, the dependence on ER for breast cancer growth is often retained even after progression through several lines of antiestrogen therapy, making ER a bonafide biomarker for this cancer subtype and driving the continued research and development of novel ER-targeted therapeutics to treat this patient population. This, combined with the continuous discovery of mechanisms underlying endocrine resistance, is resulting in a continually evolving treatment landscape for ER+ breast cancer. This review discusses various ER antagonists investigated for the treatment of breast cancer, outlining their pharmacological and tissue-specific mechanisms of action as well as their specified use within the ER+ breast cancer setting. In addition, mechanisms of resistance to SERMs and SERDs, the use of ER antagonists in combination therapy strategies, and the ongoing development of novel drugs are also reviewed in the context of the changing clinical landscape of ER+ breast cancer. Lastly, the role of SERMs and SERDs in non-breast cancer indications is also discussed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/therapeutic use , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor Antagonists/administration & dosage , Estrogen Receptor Antagonists/chemistry , Female , Humans , Molecular Structure , Selective Estrogen Receptor Modulators/administration & dosage , Selective Estrogen Receptor Modulators/chemistry
16.
Eur J Med Chem ; 143: 670-682, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29220789

ABSTRACT

New estrogen receptor α (ERα) antagonists - 3,20-dihydroxy-19-norpregna-1,3,5(10)-trienes containing an additional carbocyclic ring D' at the 16α,17α positions - were synthesized. The effects of the new compounds on the MCF-7 breast cancer cells and ERα activation were investigated. All the steroids studied were synthesized starting from estrone methyl ether. The scaffold of compounds containing the six-membered ring D' was constructed via the Diels-Alder reaction of butadiene with 3-methoxy-19-norpregna-1,3,5(10),16-tetraen-20-one 5. The hydrogenation of primary 16α,17α-cyclohexeno-adduct 7 followed by 3-demethylation (by HBr-AcOH) and reduction of 20-oxo group (by LiAlH4) or in one step by DIBAH gave target mono- and dihydroxy steroids 9-11. The Corey-Chaykovsky reaction of the same 3-methoxy-19-norpregna-1,3,5(10),16-tetraen-20-one 5 gave 16α,17α-methylene-substituted compound. The reaction of the latter with DIBAH immediately yielded 3,20-dihydroxy-16α,17α-methyleno-19-norpregna-1,3,5(10)-triene 13. The same procedures using 3-methoxy-19-norpregna-1,3,5(10),16-tetraen-20-one 5 produced corresponding 3,20-dihydroxy-16,17-19-norpregna-1,3,5(10)-triene 16 and 3,20-dihydroxy-19-norpregna-1,3,5(10),16-tetraene 17. All compounds were fully characterized by 1D and 2D NMR, HRMS, and X-ray diffraction. The molecular docking showed that the target compounds can bind to ER, their binding mode being similar to that of natural estradiol. 16α,17α-Methylene- or unsubstituted compounds exhibit the highest cytotoxicity against MCF-7 cells, being simultaneously relatively weak ERα inhibitors. 3,20-Dihydroxy steroids containing the six-membered ring D' proved to be the most effective ERα inhibitors. These compounds displayed moderate cytotoxicity comparable of that of tamoxifen and showed no toxic effect on MCF-10A normal, nontumorigenic epithelial cells. The new ER antagonists were found to be good candidates for further testing as agents for the treatment and prevention of ERα-positive breast cancers.


Subject(s)
Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Norpregnatrienes/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Humans , MCF-7 Cells , Models, Molecular , Molecular Structure , Norpregnatrienes/chemistry , Structure-Activity Relationship
17.
Neurochem Res ; 42(8): 2246-2256, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28374135

ABSTRACT

Oxabicycloheptene sulfonate (OBHS) is a novel bicyclic core selective estrogen receptor modulator (SERM) with estrogen receptor (ER) antagonistic-activity and anti-inflammatory activity. However, little is known about protective action of OBHS on neurodegenerative disorders. In the present study, OBHS demonstrated a remarkably protective effect against amyloid beta (Aß) induced cytotoxicity via G-protein-coupled estrogen receptor 1 (GPER1) in rat astroglial cell line (C6). The C6 cell death induced by Aß was decreased by OBHS (1 µM) treatment for 45 min. This rapid protective action was blocked by GPER1 specific antagonist or siRNA knockdown. Inhibitors of phosphatidylinositol 3-kinase (PI3k)/Akt and extracellular signal-regulated kinase (ERK) activation also exhibited similar effects as GPER1 antagonist in blocking the protective effects of OBHS. Moreover, the expression of anti-apoptotic protein Bcl-2 was also increased by OBHS as a consequence of the activation of GPER1-PI3K/Akt and ERK pathways. Additionally, the phenyl sulfonate moiety of OBHS played a vital role in producing GPER1's agonist property according to the molecular docking analysis. These findings suggest that OBHS provide protection directed at enhancing glial cell survival through the activation of GPER1, which, in turn, offers a novel insight into the molecular mechanisms behind the potential application of OBHS in treating Alzheimer's disease (AD).


Subject(s)
Amyloid beta-Peptides/toxicity , Estrogen Receptor Antagonists/pharmacology , MAP Kinase Signaling System/physiology , Peptide Fragments/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Estrogen Receptor Antagonists/chemistry , MAP Kinase Signaling System/drug effects , Protein Structure, Secondary , Rats , Receptors, G-Protein-Coupled/agonists
18.
Life Sci ; 170: 115-122, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27939940

ABSTRACT

AIMS: The role of estrogen in embryo implantation in golden hamster (Mesocricetus auratus) is still ambiguous. In order to clarify it, we investigated the spatial distribution and expression of estrogen receptors, ER-α and ER-ß in the uterus of pregnant hamster during peri-implantation period and identified the effect of estrogen receptor antagonist ICI-182,780 on the embryo implantation. MAIN METHODS: We performed in vivo experiments on early pregnant hamsters involving treatment with ICI-182,780, an estrogen receptor antagonist. Immunohistochemistry, western blot analysis and quantitative PCR were employed to evaluate the spatio-temporal distribution and expression of ER-α and ER-ß in the uterus of normal early pregnant and treated hamsters. KEY FINDINGS: Results showed that embryo implantation was completely absent in ICI-182,780 treated uterine horn while, normal implantation occurred in control and vehicle treated horns. Both the receptors were differentially expressed in the uterus of hamster from day 1 (D1) to day7 (D7). In contrast, treated horns without any implantation site showed no trace of any receptors. Protein and mRNA expression of both the receptors were high around the day of implantation while, ER-ß expression was up-regulated on D7 of embryo implantation. P value˂0.05 is considered significant. SIGNIFICANCE: Spatio-temporal expression of ERs in the uterus during peri-implantation period have crucial role for endometrium receptivity and implantation in hamster. Recurrent implantation failure is the devastating problem among the desirable couple and is mainly due to defect in endometrium receptivity. This study may provide a new insight to manage the problem of idiopathic infertility.


Subject(s)
Decidua/metabolism , Embryo Implantation , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Uterus/metabolism , Animals , Cricetinae , Estradiol/analogs & derivatives , Estradiol/chemistry , Estrogen Receptor Antagonists/chemistry , Estrogens/metabolism , Female , Fulvestrant , Gene Expression Profiling , Gene Expression Regulation , Mesocricetus , Pregnancy , Pregnancy, Animal , Time Factors
19.
PLoS One ; 10(5): e0125927, 2015.
Article in English | MEDLINE | ID: mdl-26016941

ABSTRACT

Cryptococcosis is one of the most important invasive fungal infections and is a significant contributor to the mortality associated with HIV/AIDS. As part of our program to repurpose molecules related to the selective estrogen receptor modulator (SERM) tamoxifen as anti-cryptococcal agents, we have explored the structure-activity relationships of a set of structurally diverse SERMs and tamoxifen derivatives. Our data provide the first insights into the structural requirements for the antifungal activity of this scaffold. Three key molecular characteristics affecting anti-cryptococcal activity emerged from our studies: 1) the presence of an alkylamino group tethered to one of the aromatic rings of the triphenylethylene core; 2) an appropriately sized aliphatic substituent at the 2 position of the ethylene moiety; and 3) electronegative substituents on the aromatic rings modestly improved activity. Using a cell-based assay of calmodulin antagonism, we found that the anti-cryptococcal activity of the scaffold correlates with calmodulin inhibition. Finally, we developed a homology model of C. neoformans calmodulin and used it to rationalize the structural basis for the activity of these molecules. Taken together, these data and models provide a basis for the further optimization of this promising anti-cryptococcal scaffold.


Subject(s)
Antifungal Agents/pharmacology , Tamoxifen/pharmacology , Antifungal Agents/chemistry , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacology , Microbial Sensitivity Tests , Molecular Structure , Selective Estrogen Receptor Modulators , Structure-Activity Relationship , Tamoxifen/chemistry
20.
Drug Metab Dispos ; 43(6): 889-97, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25795461

ABSTRACT

We previously reported upregulation of UGT2B15 by 17ß-estradiol in breast cancer MCF7 cells via binding of the estrogen receptor α (ERα) to an estrogen response unit (ERU) in the proximal UGT2B15 promoter. In the present study, we show that this ERα-mediated upregulation was significantly reduced by two ER antagonists (fulvestrant and raloxifene) but was not affected by a third ER antagonist, 4-hydroxytamoxifen (4-OHTAM), a major active tamoxifen (TAM) metabolite. Furthermore, we found that, similar to 17ß-estradiol, 4-OHTAM and endoxifen (another major active TAM metabolite) elevated UGT2B15 mRNA levels, and that this stimulation was significantly abrogated by fulvestrant. Further experiments using 4-OHTAM revealed a critical role for ERα in this regulation. Specifically; knockdown of ERα expression by anti-ERα small interfering RNA reduced the 4-OHTAM-mediated induction of UGT2B15 expression; 4-OHTAM activated the wild-type but not the ERU-mutated UGT2B15 promoter; and chromatin immunoprecipitation assays showed increased ERα occupancy at the UGT2B15 ERU in MCF7 cells upon exposure to 4-OHTAM. Together, these data indicate that both 17ß-estradiol and the antiestrogen 4-OHTAM upregulate UGT2B15 in MCF7 cells via the same ERα-signaling pathway. This is consistent with previous observations that both 17ß-estradiol and TAM upregulate a common set of genes in MCF7 cells via the ER-signaling pathway. As 4-OHTAM is a UGT2B15 substrate, the upregulation of UGT2B15 by 4-OHTAM in target breast cancer cells is likely to enhance local metabolism and inactivation of 4-OHTAM within the tumor. This represents a potential mechanism that may reduce TAM therapeutic efficacy or even contribute to the development of acquired TAM resistance.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drugs, Investigational/pharmacology , Enzyme Induction/drug effects , Estrogen Receptor Antagonists/pharmacology , Glucuronosyltransferase/metabolism , Tamoxifen/analogs & derivatives , Antineoplastic Agents, Hormonal/antagonists & inhibitors , Antineoplastic Agents, Hormonal/metabolism , Breast Neoplasms/metabolism , Drugs, Investigational/chemistry , Drugs, Investigational/metabolism , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Genes, Reporter/drug effects , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/genetics , Humans , MCF-7 Cells , Mutation , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/drug effects , RNA Interference , Response Elements/drug effects , Signal Transduction/drug effects , Substrate Specificity , Tamoxifen/antagonists & inhibitors , Tamoxifen/metabolism , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL