1
|
Palaniappan M, Bohren KM. Purification and biochemical characterization of mutant ligand binding domain of human estrogen receptor α protein. J Steroid Biochem Mol Biol 2025; 251:106763. [PMID: 40245990 DOI: 10.1016/j.jsbmb.2025.106763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/19/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
The acquisition of mutations in the estrogen receptor alpha (ERα) gene (ESR1) is a key driver in the development of resistance to current endocrine therapy in breast cancer. Clinical studies have shown that ESR1 mutations are frequently observed in patients with metastatic ER-positive breast cancer and are associated with poor survival. Activating ESR1 somatic mutations, particularly Y537S and D538G, drive estrogen-independent activities in cell-based studies and these mutant receptors are less sensitive to current endocrine therapies. Here, we describe the bacterial expression and purification of the ligand binding domains of wild-type, Y537S, and D538G human ERα proteins. The biochemical activities of these domains were confirmed by homogeneous time-resolved fluorescence and polar screen ERα competition assays. The wild-type domain binds to coactivator peptides only in the presence of the ligand estradiol, whereas the Y537S or D538G domains bind coactivator peptides spontaneously even without estradiol, with the Y537S domain showing higher affinity. Thermal shift assays showed that the mutations stabilized these domains. Our purified human ERα wild-type, Y537S, and D538G ligand binding domains recapitulate the biological activities ascribed to the full-length proteins and can therefore be used for small molecule screens that seek to discriminate between wild-type and mutant ERα.
Collapse
Affiliation(s)
- Murugesan Palaniappan
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, United States; Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, United States; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, United States.
| | - Kurt M Bohren
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, United States; Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
2
|
Giugliano G, Gajo M, Marforio TD, Zerbetto F, Mattioli EJ, Calvaresi M. Identification of Potential Drug Targets of Calix[4]arene by Reverse Docking. Chemistry 2024; 30:e202400871. [PMID: 38777795 DOI: 10.1002/chem.202400871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
Calixarenes are displaying great potential for the development of new drug delivery systems, diagnostic imaging, biosensing devices and inhibitors of biological processes. In particular, calixarene derivatives are able to interact with many different enzymes and function as inhibitors. By screening of the potential drug target database (PDTD) with a reverse docking procedure, we identify and discuss a selection of 100 proteins that interact strongly with calix[4]arene. We also discover that leucine (23.5 %), isoleucine (11.3 %), phenylalanines (11.3 %) and valine (9.5 %) are the most frequent binding residues followed by hydrophobic cysteines and methionines and aromatic histidines, tyrosines and tryptophanes. Top binders are peroxisome proliferator-activated receptors that already are targeted by commercial drugs, demonstrating the practical interest in calix[4]arene. Nuclear receptors, potassium channel, several carrier proteins, a variety of cancer-related proteins and viral proteins are prominent in the list. It is concluded that calix[4]arene, which is characterized by facile access, well-defined conformational characteristics, and ease of functionalization at both the lower and higher rims, could be a potential lead compound for the development of enzyme inhibitors and theranostic platforms.
Collapse
Affiliation(s)
- Giulia Giugliano
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Margherita Gajo
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Tainah Dorina Marforio
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Francesco Zerbetto
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Edoardo Jun Mattioli
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Matteo Calvaresi
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| |
Collapse
|
3
|
Kazimir A, Götze T, Lönnecke P, Murganić B, Mijatović S, Maksimović-Ivanić D, Hey-Hawkins E. Exploring Raloxifene-Based Metallodrugs: A Versatile Vector Combined with Platinum(II), Palladium(II) and Nickel(II) Dichlorides and Carborates against Triple-Negative Breast Cancer. ChemMedChem 2024; 19:e202400006. [PMID: 38642018 DOI: 10.1002/cmdc.202400006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 04/22/2024]
Abstract
Triple-negative breast cancer (TNBC) poses challenges in therapy due to the absence of target expression such as estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Frequently, the treatment of TNBC involves the combination of several therapeutics. However, an enhanced therapeutic effect can be also achieved within a single molecule. The efficacy of raloxifene can be improved by designing a raloxifene-based hybrid drug bearing a 2,2'-bipyridine moiety (2). Integration of platinum(II), palladium(II), and nickel(II) complexes into this structure dramatically changed the cytotoxicity. The platinum(II) dichloride complex 3 did not demonstrate any activity, while palladium(II) and nickel(II) dichloride complexes 4 and 5 exhibited various cytotoxic behavior towards different types of hormone-receptor positive (HR+) cancer and TNBC cell lines. The replacement of the two chlorido ligands in 3-5 with a dicarbollide (carborate) ion [C2B9H11]2- resulted in reduced activity of compounds 6, 7, and 8. However, the palladacarborane complex 7 demonstrated higher selectivity towards TNBC. Furthermore, the mechanism of action was shifted from cytotoxic to explicitly cytostatic with detectable proliferation arrest and accelerated aging, characterized by senescence-associated phenotype of TNBC cells. This study provides valuable insights into the development of hybrid therapeutics against TNBC.
Collapse
Affiliation(s)
- Aleksandr Kazimir
- new address, Institute for Drug Discovery, Leipzig University, Leipzig, Brüderstraße 34, 04103, Germany
- Faculty of Chemistry and Mineralogy, Leipzig University, Johannisallee 29, Leipzig, 04103, Germany
| | - Tom Götze
- Faculty of Chemistry and Mineralogy, Leipzig University, Johannisallee 29, Leipzig, 04103, Germany
| | - Peter Lönnecke
- Faculty of Chemistry and Mineralogy, Leipzig University, Johannisallee 29, Leipzig, 04103, Germany
| | - Blagoje Murganić
- Institute of Nuclear Sciences "Vinča", University of Belgrade, 12-14 Mike Petrovića Street, Belgrade, 11351, Serbia
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, Belgrade University, Bul. despota Stefana 142, Belgrade, 11060, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, Belgrade University, Bul. despota Stefana 142, Belgrade, 11060, Serbia
| | - Evamarie Hey-Hawkins
- Faculty of Chemistry and Mineralogy, Leipzig University, Johannisallee 29, Leipzig, 04103, Germany
| |
Collapse
|
4
|
Kazimir A, Götze T, Murganić B, Mijatović S, Maksimović-Ivanić D, Hey-Hawkins E. Bipyraloxifene - a modified raloxifene vector against triple-negative breast cancer. RSC Med Chem 2024; 15:1921-1928. [PMID: 38911151 PMCID: PMC11187558 DOI: 10.1039/d4md00051j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/01/2024] [Indexed: 06/25/2024] Open
Abstract
Raloxifene, a selective oestrogen receptor modulator (SERM), has demonstrated efficacy in the prevention and therapy of oestrogen receptor-positive (ER+) breast cancer, with some degree of effectiveness against triple-negative forms. This suggests the presence of oestrogen receptor-independent pathways in raloxifene-mediated anticancer activity. To enhance the potential of raloxifene against the most aggressive breast cancer cells, hybrid molecules combining the drug with a metal chelator moiety have been developed. In this study, we synthetically modified the structure of raloxifene by incorporating a 2,2'-bipyridine (2,2'-bipy) moiety, resulting in [6-methoxy-2-(4-hydroxyphenyl)benzo[b]thiophen-3-yl]-[4-(2,2'-bipyridin-4'-yl-methoxy)phenyl]methanone (bipyraloxifene). We investigated the cytotoxic activity of both raloxifene and bipyraloxifene against ER+ breast adenocarcinomas, glioblastomas, and a triple-negative breast cancer (TNBC) cell line, elucidating their mode of action against TNBC. Bipyraloxifene maintained a mechanism based on caspase-mediated apoptosis but exhibited significantly higher activity and selectivity compared to the original drug, particularly evident in triple-negative stem-like MDA-MB-231 cells.
Collapse
Affiliation(s)
- Aleksandr Kazimir
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University Johannisallee 29 04103 Leipzig Germany
| | - Tom Götze
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University Johannisallee 29 04103 Leipzig Germany
| | - Blagoje Murganić
- Institute of Nuclear Sciences "Vinča", University of Belgrade 12-14 Mike Petrovića Street Belgrade 11351 Serbia
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, Belgrade University Bul. despota Stefana 142 Belgrade 11060 Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, Belgrade University Bul. despota Stefana 142 Belgrade 11060 Serbia
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University Johannisallee 29 04103 Leipzig Germany
| |
Collapse
|
5
|
Mostafa T, Albeir M, Wober J, Abadi A, Salama I, Ahmed NS. Design, synthesis, and in-silico study of novel triarylethylene analogs with dual anti-estrogenic and serotonergic activity. Drug Dev Res 2024; 85:e22127. [PMID: 37877739 DOI: 10.1002/ddr.22127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/08/2023] [Accepted: 10/15/2023] [Indexed: 10/26/2023]
Abstract
Estrogen receptor is an important target in breast cancer. Serotonin receptors (5-HT2A and 5-HT2C , in particular) were investigated for a potential role in development and progression of breast cancer. Ligands that interact with estrogenic receptors influence the emotional state of females. Thus, designing selective estrogen receptor modulator (SERM) analogs with potential serotonergic activity is a plausible approach. The dual ligands can augment cytotoxic effect of SERMs, help in both physical and emotional menopausal symptom relief, enhance cognitive function and support bone health. Herein, we report triarylethylene analogs as potential candidates for treatment of breast cancer. Compound 2e showed (ERα relative β- galactosidase activity = 0.70), 5-HT2A (Ki = 0.97 µM), and 5-HT2C (Ki = 3.86 µM). It was more potent on both MCF-7 (GI50 = 0.27 µM) and on MDA-MB-231 (GI50 = 1.86 µM) compared to tamoxifen (TAM). Compound 4e showed 40 times higher antiproliferative activity on MCF-7 and 15 times on MDA-MBA compared to TAM. Compound 4e had higher average potency than TAM on all nine tested cell line panels. Our in-silico model revealed the binding interactions of compounds 2 and 2e in the three receptors; further structural modifications are suggested to optimize binding to the ERα, 5-HT2A , and 5-HT2C .
Collapse
Affiliation(s)
- Tammy Mostafa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Miriam Albeir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Jannette Wober
- Institute of Zoology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Ashraf Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ismail Salama
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Nermin S Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
6
|
Li M, Zhang J, Chen W, Liu S, Liu X, Ning Y, Cao Y, Zhao Y. Supraphysiologic doses of 17β-estradiol aggravate depression-like behaviors in ovariectomized mice possibly via regulating microglial responses and brain glycerophospholipid metabolism. J Neuroinflammation 2023; 20:204. [PMID: 37679787 PMCID: PMC10485970 DOI: 10.1186/s12974-023-02889-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND 17β-Estradiol (E2) is generally considered neuroprotective in humans. However, the current clinical use of estrogen replacement therapy (ERT) is based on the physiological dose of E2 to treat menopausal syndrome and has limited therapeutic efficacy. The efficacy and potential toxicity of superphysiological doses of ERT for menopausal neurodegeneration are unknown. METHODS In this study, we investigated the effect of E2 with a supraphysiologic dose (0.5 mg/kg, sE2) on the treatment of menopausal mouse models established by ovariectomy. We performed the open field, Y-maze spontaneous alternation, forced swim tests, and sucrose preference test to investigate behavioral alterations. Subsequently, the status of microglia and neurons was detected by immunohistochemistry, HE staining, and Nissl staining, respectively. Real-time PCR was used to detect neuroinflammatory cytokines in the hippocampus and cerebral cortex. Using mass spectrometry proteomics platform and LC-MS/ MS-based metabolomics platform, proteins and metabolites in brain tissues were extracted and analyzed. BV2 and HT22 cell lines and primary neurons and microglia were used to explore the underlying molecular mechanisms in vitro. RESULTS sE2 aggravated depression-like behavior in ovariectomized mice, caused microglia response, and increased proinflammatory cytokines in the cerebral cortex and hippocampus, as well as neuronal damage and glycerophospholipid metabolism imbalance. Subsequently, we demonstrated that sE2 induced the pro-inflammatory phenotype of microglia through ERα/NF-κB signaling pathway and downregulated the expression of cannabinoid receptor 1 in neuronal cells, which were important in the pathogenesis of depression. CONCLUSION These data suggest that sE2 may be nonhelpful or even detrimental to menopause-related depression, at least partly, by regulating microglial responses and glycerophospholipid metabolism.
Collapse
Affiliation(s)
- Ming Li
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Wendi Chen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Shuang Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yunna Ning
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yongzhi Cao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yueran Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
7
|
Bender O, Celik I, Dogan R, Atalay A, Shoman ME, Ali TFS, Beshr EAM, Mohamed M, Alaaeldin E, Shawky AM, Awad EM, Ahmed ASF, Younes KM, Ansari M, Anwar S. Vanillin-Based Indolin-2-one Derivative Bearing a Pyridyl Moiety as a Promising Anti-Breast Cancer Agent via Anti-Estrogenic Activity. ACS OMEGA 2023; 8:6968-6981. [PMID: 36844536 PMCID: PMC9948168 DOI: 10.1021/acsomega.2c07793] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
The structure-based design introduced indoles as an essential motif in designing new selective estrogen receptor modulators employed for treating breast cancer. Therefore, here, a series of synthesized vanillin-substituted indolin-2-ones were screened against the NCI-60 cancer cell panel followed by in vivo, in vitro, and in silico studies. Physicochemical parameters were evaluated with HPLC and SwissADME tools. The compounds demonstrated promising anti-cancer activity for the MCF-7 breast cancer cell line (GI = 6-63%). The compound with the highest activity (6j) was selective for the MCF-7 breast cancer cell line (IC50 = 17.01 μM) with no effect on the MCF-12A normal breast cell line supported by real-time cell analysis. A morphological examination of the used cell lines confirmed a cytostatic effect of compound 6j. It inhibited both in vivo and in vitro estrogenic activity, triggering a 38% reduction in uterine weight induced by estrogen in an immature rat model and hindering 62% of ER-α receptors in in vitro settings. In silico molecular docking and molecular dynamics simulation studies supported the stability of the ER-α and compound 6j protein-ligand complex. Herein, we report that indolin-2-one derivative 6j is a promising lead compound for further pharmaceutical formulations as a potential anti-breast cancer drug.
Collapse
Affiliation(s)
- Onur Bender
- Biotechnology
Institute, Ankara University, 06135 Ankara, Turkey
| | - Ismail Celik
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38280 Kayseri, Turkey
| | - Rumeysa Dogan
- Biotechnology
Institute, Ankara University, 06135 Ankara, Turkey
| | - Arzu Atalay
- Biotechnology
Institute, Ankara University, 06135 Ankara, Turkey
| | - Mai E. Shoman
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Taha F. S. Ali
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Eman A. M. Beshr
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Mahmoud Mohamed
- Department
of Pharmacognosy, College of Clinical Pharmacy, Al Baha University, 65528 Al Baha, Saudi Arabia
| | - Eman Alaaeldin
- Department
of Pharmaceutics, Faculty of Pharmacy, Minia
University, 61519 Minia, Egypt
- Department
of Clinical Pharmacy, Faculty of Pharmacy, Deraya University, 61111 Minia, Egypt
| | - Ahmed M. Shawky
- Science
and Technology Unit (STU), Umm Al-Qura University, 21955 Makkah, Saudi Arabia
- Central
Laboratory for Micro-analysis, Minia University, 61519 Minia, Egypt
| | - Eman M. Awad
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Al-Shaimaa F. Ahmed
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Kareem M. Younes
- Department
of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442 Hail, Saudi Arabia
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, El-Kasr El-Aini Street, ET-11562 Cairo, Egypt
| | - Mukhtar Ansari
- Department
of Clinical Pharmacy, College of Pharmacy, University of Hail, 81442 Hail, Saudi Arabia
| | - Sirajudheen Anwar
- Department
of Pharmacology and Toxicology, College of Pharmacy, University of Hail, 81442 Hail, Saudi Arabia
| |
Collapse
|
8
|
Single crystal investigation, spectroscopic, DFT studies, and in-silico molecular docking of the anticancer activities of acetylacetone coordinated Re(I) tricarbonyl complexes. Inorganica Chim Acta 2023. [DOI: 10.1016/j.ica.2022.121335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Liu Q, Shen Y, Xiong Y, Bai J, Zhang Y, Li C. Comparative Efficacy of AZD9496 and Fulvestrant on the Growth of Pituitary Adenoma via Blocking JAK2/STAT5B Pathway. J Cancer 2023; 14:61-71. [PMID: 36605480 PMCID: PMC9809331 DOI: 10.7150/jca.79726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/26/2022] [Indexed: 01/04/2023] Open
Abstract
Total 158 gonadotropin-type pituitary adenoma tissue specimens were collected and the expression of ESR1 in gonadotropin-type pituitary adenoma and its association with the overall survival of patients were analyzed. Transcriptome-sequencing data containing 79 cases of gonadotropin-type pituitary adenoma was used to search for all ESR1-related genes. KEGG pathway enrichment analysis was performed to identify the altering pathway and targeting genes. The in vitro and in vivo pituitary models were used to evaluate the therapeutic efficacy of estrogen receptor (ER) inhibitors AZD9496 and fulvestrant. The mechanism of AZD9496 and fulvestrant in suppressing pituitary adenoma were also investigated. Low-level ESR1 had longer progression-free survival (PFS) in pituitary adenoma patients. ErbB signaling pathway was discovered as the main enriched pathway. Furthermore, the STAT5B gene was identified as a key ESR-1-related gene. The expression of STAT5B was significantly positively correlated with ESR1 expression in the pituitary adenoma. AZD9496, a novel ER inhibitor, exhibited a potent inhibitory effect on the growth of in vitro and in vivo pituitary adenoma cells, and its efficacy is comparable to the classic ER inhibitor, fulvestrant. Mechanically, the AZD9496 and fulvestrant significantly blocked JAK2/STAT5B pathway in GT1-1 cells and xenograft mice. Our results provide substantial evidence for the subsequent clinical use of AZD9496 in the treatment of patients with pituitary adenoma.
Collapse
Affiliation(s)
- Qian Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Yutao Shen
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Yujia Xiong
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Jiwei Bai
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.,✉ Corresponding authors: Yazhuo Zhang, E-mail address: ; Chuzhong Li, E-mail address:
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China.,✉ Corresponding authors: Yazhuo Zhang, E-mail address: ; Chuzhong Li, E-mail address:
| |
Collapse
|
10
|
Zhaojun C, Lulin T, Xin F, Abdel-Nasser S, Zunguo L, Xiong L. Hydroxy-γ-sanshool from Zanthoxylum bungeanum (prickly ash) induces apoptosis of human colorectal cancer cell by activating P53 and Caspase 8. Front Nutr 2022; 9:914638. [PMID: 35978957 PMCID: PMC9376619 DOI: 10.3389/fnut.2022.914638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Sanshools, long-chain polyunsaturated amides in Zanthoxylum bungeanum (prickly ash), have important bioactivity. The objective was to assess inhibitory effects and molecular mechanisms of sanshools isolated from supercritical fluid (SCF) extract on human colon adenocarcinoma cells (HCT-116) cultured in vitro. Cells were exposed to various concentrations (0, 50, 90, or 130 μM) of sanshools for 24 or 48 h, with assessment of apoptosis and cell cycle arrest as well as regulatory gene and protein expression associated with apoptosis and the cell cycle. Sanshools profoundly inhibited growth of HCT-116 cells, with hydroxy-γ-sanshool (HRS) being the optimal active component (IC50 = 88.01 μM) inhibiting cell proliferation and having no cytotoxic effect to normal cells (IC50 = 481.52 μM) by CCK-8 assay. In HCT-116 cells, HRS inhibited cell growth, induced morphological distortion, and arrested the cell cycle at G1 phase (50.31 ± 4.13% vs. 72.16 ± 8.14% in Control and 130 μM HRS, respectively), and also caused programmed cell death in a dose-dependent manner. The percentage of apoptotic cells were remarkably increased after treated with HRS (6.2, 11.9, 19.8, and 30.7% for 0, 50, 90, and 130 μM, respectively). Moreover, in HCT-116 cells, HRS significantly inhibited mRNA and protein levels of Cyclin D1, CDK4, PCNA, and increased mRNA and protein levels of P21, P53, Fas, and Caspase 8. Furthermore, inhibitors of P53 and Caspase 8 proteins significantly mitigated the HRS-induced cell cycle arrest and apoptosis. In conclusion, our study provides evidence that HRS induced human colorectal cancer cell apoptosis by up-regulating P53 and Caspase 8.
Collapse
Affiliation(s)
- Chen Zhaojun
- College of Food Science, Southwest University, Chongqing, China.,Guizhou Provincial Academy of Agricultural Sciences, Guiyang, China
| | - Tan Lulin
- Guizhou Provincial Academy of Agricultural Sciences, Guiyang, China
| | - Feng Xin
- College of Food Science, Southwest University, Chongqing, China
| | | | - Lei Zunguo
- Guizhou Provincial Academy of Agricultural Sciences, Guiyang, China
| | - Liu Xiong
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
11
|
Optimization of nanoemulsified systems containing lamellar phases for co-delivery of celecoxib and endoxifen to the skin aiming for breast cancer chemoprevention and treatment. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.128901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
Hosfield DJ, Weber S, Li NS, Suavage M, Joiner CF, Hancock GR, Sullivan EA, Ndukwe E, Han R, Cush S, Lainé M, Mader SC, Greene GL, Fanning SW. Stereospecific lasofoxifene derivatives reveal the interplay between estrogen receptor alpha stability and antagonistic activity in ESR1 mutant breast cancer cells. eLife 2022; 11:72512. [PMID: 35575456 PMCID: PMC9177151 DOI: 10.7554/elife.72512] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Chemical manipulation of estrogen receptor alpha ligand binding domain structural mobility tunes receptor lifetime and influences breast cancer therapeutic activities. Selective estrogen receptor modulators (SERMs) extend estrogen receptor alpha (ERα) cellular lifetime/accumulation. They are antagonists in the breast but agonists in the uterine epithelium and/or in bone. Selective estrogen receptor degraders/downregulators (SERDs) reduce ERα cellular lifetime/accumulation and are pure antagonists. Activating somatic ESR1 mutations Y537S and D538G enable resistance to first-line endocrine therapies. SERDs have shown significant activities in ESR1 mutant setting while few SERMs have been studied. To understand whether chemical manipulation of ERα cellular lifetime and accumulation influences antagonistic activity, we studied a series of methylpyrollidine lasofoxifene (Laso) derivatives that maintained the drug’s antagonistic activities while uniquely tuning ERα cellular accumulation. These molecules were examined alongside a panel of antiestrogens in live cell assays of ERα cellular accumulation, lifetime, SUMOylation, and transcriptional antagonism. High-resolution x-ray crystal structures of WT and Y537S ERα ligand binding domain in complex with the methylated Laso derivatives or representative SERMs and SERDs show that molecules that favor a highly buried helix 12 antagonist conformation achieve the greatest transcriptional suppression activities in breast cancer cells harboring WT/Y537S ESR1. Together these results show that chemical reduction of ERα cellular lifetime is not necessarily the most crucial parameter for transcriptional antagonism in ESR1 mutated breast cancer cells. Importantly, our studies show how small chemical differences within a scaffold series can provide compounds with similar antagonistic activities, but with greatly different effects of the cellular lifetime of the ERα, which is crucial for achieving desired SERM or SERD profiles.
Collapse
Affiliation(s)
- David J Hosfield
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Sandra Weber
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Nan-Sheng Li
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Madline Suavage
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Carstyn F Joiner
- Department of Cancer Biology, Loyola University Chicago, Maywood, United States
| | - Govinda R Hancock
- Department of Cancer Biology, Loyola University Chicago, Maywood, United States
| | - Emily A Sullivan
- Department of Cancer Biology, Loyola University Chicago, Maywood, United States
| | - Estelle Ndukwe
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Ross Han
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Sydney Cush
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Muriel Lainé
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Sylvie C Mader
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Geoffrey L Greene
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Sean W Fanning
- Department of Cancer Biology, Loyola University Chicago, Maywood, United States
| |
Collapse
|
13
|
Maximov PY, Fan P, Abderrahman B, Curpan R, Jordan VC. Estrogen Receptor Complex to Trigger or Delay Estrogen-Induced Apoptosis in Long-Term Estrogen Deprived Breast Cancer. Front Endocrinol (Lausanne) 2022; 13:869562. [PMID: 35360069 PMCID: PMC8960923 DOI: 10.3389/fendo.2022.869562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Antiestrogen therapy of breast cancer has been a "gold standard" of treatment of estrogen receptor (ER)-positive breast cancer for decades. Resistance to antiestrogen therapy may develop, however, a vulnerability in long-term estrogen deprived (LTED) breast cancer cells was discovered. LTED breast cancer cells may undergo estrogen-induced apoptosis within a week of treatment with estrogen in vitro. This phenomenon has been also validated in vivo and in the clinic. The molecular ER-mediated mechanism of action of estrogen-induced apoptosis was deciphered, however, the relationship between the structure of estrogenic ligands and the activity of the ER in LTED breast cancer cells remained a mystery until recently. In this review we provide an overview of the structure-activity relationship of various estrogens with different chemical structures and the modulation of estrogen-induced apoptosis in LTED breast cancer cells resistant to antihormone therapy. We provide analysis of evidence gathered over more than a decade of structure-activity relationship studies by our group on the role of the change in the conformation of the estrogen receptor and the biological activities of different classes of estrogens and the receptor as well in LTED breast cancer.
Collapse
Affiliation(s)
- Philipp Y. Maximov
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ping Fan
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Balkees Abderrahman
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ramona Curpan
- Institute of Chemistry, Romanian Academy, Timisoara, Romania
| | - V. Craig Jordan
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: V. Craig Jordan,
| |
Collapse
|
14
|
Vincristine-doxorubicin co-loaded artificial low-density lipoproteins towards solid tumours. Eur J Med Chem 2021; 226:113802. [PMID: 34543934 DOI: 10.1016/j.ejmech.2021.113802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/16/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022]
Abstract
To construct an artificial low-density lipoprotein (aLDL) that highly mimics low-density lipoprotein (LDL) in vivo, and deliver vincristine (VCR) - doxorubicin (DOX) simultaneously, the 100 nm and 35 nm DOX-VCR-aLDLs (DV-aLDLs) were constructed, then the physicochemical characteristics were evaluated. Through in vitro inverse gravity diffusion experiment, the tumour cake and sphere model experiment, draw a conclusion that the diffusion of 35 nm DV-aLDLs was stronger than 100 nm DV-aLDLs, and the tumour retention of 35 nm DV-aLDLs was better than the DV-solution. In addition, the three-dimension (3D) in vivo distribution imaging of aLDLs was performed on HepG-2 tumour-bearing nude mice, followed by the biodistribution and therapeutic efficacy on these xenograft models. Taking advantage of better diffusion capacity in tumour tissue, as well as the synergistic effect of VCR and DOX, the 35 nm DV-aLDL had the strongest efficacy and the lowest toxicity. High entrapment efficiency and stability, both active and passive targeting, making aLDL a potential carrier for tumour-targeted therapy at the same time.
Collapse
|
15
|
Caciolla J, Martini S, Spinello A, Pavlin M, Turrini E, Simonelli F, Belluti F, Rampa A, Bisi A, Fimognari C, Zaffaroni N, Gobbi S, Magistrato A. Balanced dual acting compounds targeting aromatase and estrogen receptor α as an emerging therapeutic opportunity to counteract estrogen responsive breast cancer. Eur J Med Chem 2021; 224:113733. [PMID: 34364162 DOI: 10.1016/j.ejmech.2021.113733] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 02/05/2023]
Abstract
Breast Cancer (BC) is a leading cause of death in women, currently affecting 13% of female population worldwide. First-line clinical treatments against Estrogen Receptor positive (ER+) BC rely on suppressing estrogen production, by inhibiting the aromatase (AR) enzyme, or on blocking estrogen-dependent pro-oncogenic signaling, by targeting Estrogen Receptor (ER) α with selective Modulators/Degraders (SERMs/SERDs). The development of dual acting molecules targeting AR and ERα represents a tantalizing alternative strategy to fight ER + BC, reducing the incidence of adverse effects and resistance onset that limit the effectiveness of these gold-standard therapies. Here, in silico design, synthesis, biological evaluation and an atomic-level characterization of the binding and inhibition mechanism of twelve structurally related drug-candidates enable the discovery of multiple compounds active on both AR and ERα in the sub-μM range. The best drug-candidate 3a displayed a balanced low-nanomolar IC50 towards the two targets, SERM activity and moderate selectivity towards a BC cell line. Moreover, most of the studied compounds reduced ERα levels, suggesting a potential SERD activity. This study dissects the key structural traits needed to obtain optimal dual acting drug-candidates, showing that multitarget compounds may be a viable therapeutic option to counteract ER + BC.
Collapse
Affiliation(s)
- Jessica Caciolla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Silvia Martini
- Fondazione IRCSS Istituto Nazionale Dei Tumori, Via Amadeo 42, 20113, Milano, Italy
| | - Angelo Spinello
- National Research Council of Italy Institute of Materials (CNR-IOM) C/o SISSA, Via Bonomea 265, 34136, Trieste, Italy
| | - Matic Pavlin
- National Research Council of Italy Institute of Materials (CNR-IOM) C/o SISSA, Via Bonomea 265, 34136, Trieste, Italy; Laboratory of Microsensor Structures and Electronics, Faculty of Electrical Engineering, University of Ljubljana, Tržaška Cesta 25, SI-1000 Ljubljana, Slovenia
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso D'Augusto 237, 47921, Rimini, Italy
| | - Federica Simonelli
- National Research Council of Italy Institute of Materials (CNR-IOM) C/o SISSA, Via Bonomea 265, 34136, Trieste, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso D'Augusto 237, 47921, Rimini, Italy
| | - Nadia Zaffaroni
- Fondazione IRCSS Istituto Nazionale Dei Tumori, Via Amadeo 42, 20113, Milano, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy.
| | - Alessandra Magistrato
- National Research Council of Italy Institute of Materials (CNR-IOM) C/o SISSA, Via Bonomea 265, 34136, Trieste, Italy.
| |
Collapse
|
16
|
Horsfall AJ, McDougal DP, Scanlon DB, Bruning JB, Abell AD. Approaches to Introduce Helical Structure in Cysteine-Containing Peptides with a Bimane Group. Chembiochem 2021; 22:2711-2720. [PMID: 34107164 DOI: 10.1002/cbic.202100241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/08/2021] [Indexed: 01/01/2023]
Abstract
An i-i+4 or i-i+3 bimane-containing linker was introduced into a peptide known to target Estrogen Receptor alpha (ERα), in order to stabilise an α-helical geometry. These macrocycles were studied by CD and NMR to reveal the i-i+4 constrained peptide adopts a 310 -helical structure in solution, and an α-helical conformation on interaction with the ERα coactivator recruitment surface in silico. An acyclic bimane-modified peptide is also helical, when it includes a tryptophan or tyrosine residue; but is significantly less helical with a phenylalanine or alanine residue, which indicates such a bimane modification influences peptide structure in a sequence dependent manner. The fluorescence intensity of the bimane appears influenced by peptide conformation, where helical peptides displayed a fluorescence increase when TFE was added to phosphate buffer, compared to a decrease for less helical peptides. This study presents the bimane as a useful modification to influence peptide structure as an acyclic peptide modification, or as a side-chain constraint to give a macrocycle.
Collapse
Affiliation(s)
- Aimee J Horsfall
- ARC Centre of Excellence for Nanoscale Biophotonics (CNBP), University of Adelaide, Adelaide, SA 5005, Australia.,Department of Chemistry, University of Adelaide, Adelaide, SA 5005, Australia.,The Institute for Photonics & Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA 5005, Australia
| | - Daniel P McDougal
- The Institute for Photonics & Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA 5005, Australia.,School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Denis B Scanlon
- Department of Chemistry, University of Adelaide, Adelaide, SA 5005, Australia.,The Institute for Photonics & Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA 5005, Australia
| | - John B Bruning
- The Institute for Photonics & Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA 5005, Australia.,School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew D Abell
- ARC Centre of Excellence for Nanoscale Biophotonics (CNBP), University of Adelaide, Adelaide, SA 5005, Australia.,Department of Chemistry, University of Adelaide, Adelaide, SA 5005, Australia.,The Institute for Photonics & Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
17
|
Liu X, Matsuyama Y, Shimohigashi M, Shimohigashi Y. ERα-agonist and ERβ-antagonist bifunctional next-generation bisphenols with no halogens: BPAP, BPB, and BPZ. Toxicol Lett 2021; 345:24-33. [PMID: 33857583 DOI: 10.1016/j.toxlet.2021.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022]
Abstract
As demonstrated for bisphenol AF (BPAF), the electrostatic halogen bond based on the London dispersion force of halogen atoms was found to be a major driving force of their bifunctional ERα-agonist and ERβ-antagonist activities. Because similar electronic effects are anticipated for hydrocarbon groups (alkyl or aryl groups), we hypothesized that bisphenol compounds consisting of such groups also work bifunctionally. In the present study, we examined bisphenol AP (BPAP), B (BPB), and Z (BPZ). After recognizing their considerably strong receptor binding affinities, we evaluated the abilities of BPAP, BPB, and BPZ to activate ERα and ERβ in a luciferase reporter gene assay. These bisphenols were fully active for ERα but completely inactive for ERβ. When we examined their inhibitory activities for 17β-estradiol in ERβ by two different qualitative and quantitative analytical methods, we found that those bisphenols worked as definite antagonists. Consequently, they were established as bifunctional ERα-agonists and ERβ-antagonists. The present structure-activity analyses revealed that the dispersion force works not only on the halogens but also on the hydrocarbon groups, and that it is a major driving force of bifunctional ERα-agonist and ERβ-antagonist activities.
Collapse
Affiliation(s)
- Xiaohui Liu
- Department of Chemistry, Faculty of Science, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan; Department of Applied Microbial Technology, Faculty of Biotechnology and Life Sciences, Sojo University, Kumamoto, 860-0082, Japan.
| | - Yutaka Matsuyama
- Department of Chemistry, Faculty of Science, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Miki Shimohigashi
- Division of Biology, Department of Earth System of Science, Faculty of Science, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan; Risk Science Research Institute, Ikimatsudai 3-7-5, Nishi-ku, Fukuoka, 819-0044, Japan
| | - Yasuyuki Shimohigashi
- Department of Chemistry, Faculty of Science, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan; Risk Science Research Institute, Ikimatsudai 3-7-5, Nishi-ku, Fukuoka, 819-0044, Japan.
| |
Collapse
|
18
|
Jordan VC. Molecular Mechanism for Breast Cancer Incidence in the Women's Health Initiative. Cancer Prev Res (Phila) 2020; 13:807-816. [DOI: 10.1158/1940-6207.capr-20-0082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/13/2020] [Accepted: 07/10/2020] [Indexed: 11/16/2022]
|
19
|
Maximov PY, Abderrahman B, Hawsawi YM, Chen Y, Foulds CE, Jain A, Malovannaya A, Fan P, Curpan RF, Han R, Fanning SW, Broom BM, Quintana Rincon DM, Greenland JA, Greene GL, Jordan VC. The Structure-Function Relationship of Angular Estrogens and Estrogen Receptor Alpha to Initiate Estrogen-Induced Apoptosis in Breast Cancer Cells. Mol Pharmacol 2020; 98:24-37. [PMID: 32362585 PMCID: PMC7294906 DOI: 10.1124/mol.120.119776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/16/2020] [Indexed: 12/17/2022] Open
Abstract
High-dose synthetic estrogen therapy was the standard treatment of advanced breast cancer for three decades until the discovery of tamoxifen. A range of substituted triphenylethylene synthetic estrogens and diethylstilbestrol were used. It is now known that low doses of estrogens can cause apoptosis in long-term estrogen deprived (LTED) breast cancer cells resistant to antiestrogens. This action of estrogen can explain the reduced breast cancer incidence in postmenopausal women over 60 who are taking conjugated equine estrogens and the beneficial effect of low-dose estrogen treatment of patients with acquired aromatase inhibitor resistance in clinical trials. To decipher the molecular mechanism of estrogens at the estrogen receptor (ER) complex by different types of estrogens-planar [17β-estradiol (E2)] and angular triphenylethylene (TPE) derivatives-we have synthesized a small series of compounds with either no substitutions on the TPE phenyl ring containing the antiestrogenic side chain of endoxifen or a free hydroxyl. In the first week of treatment with E2 the LTED cells undergo apoptosis completely. By contrast, the test TPE derivatives act as antiestrogens with a free para-hydroxyl on the phenyl ring that contains an antiestrogenic side chain in endoxifen. This inhibits early E2-induced apoptosis if a free hydroxyl is present. No substitution at the site occupied by the antiestrogenic side chain of endoxifen results in early apoptosis similar to planar E2 The TPE compounds recruit coregulators to the ER differentially and predictably, leading to delayed apoptosis in these cells. SIGNIFICANCE STATEMENT: In this paper we investigate the role of the structure-function relationship of a panel of synthetic triphenylethylene (TPE) derivatives and a novel mechanism of estrogen-induced cell death in breast cancer, which is now clinically relevant. Our study indicates that these TPE derivatives, depending on the positioning of the hydroxyl groups, induce various conformations of the estrogen receptor's ligand-binding domain, which in turn produces differential recruitment of coregulators and subsequently different apoptotic effects on the antiestrogen-resistant breast cancer cells.
Collapse
Affiliation(s)
- Philipp Y Maximov
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Balkees Abderrahman
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Yousef M Hawsawi
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Yue Chen
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Charles E Foulds
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Antrix Jain
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Anna Malovannaya
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Ping Fan
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Ramona F Curpan
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Ross Han
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Sean W Fanning
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Bradley M Broom
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Daniela M Quintana Rincon
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Jeffery A Greenland
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - Geoffrey L Greene
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| | - V Craig Jordan
- Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan Comprehensive Cancer Center (A.M., C.E.F.), Baylor College of Medicine, Houston, Texas; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (Y.C.); and Coriolan Dragulescu Institute of Chemistry, Romanian Academy, Timisoara, Romania (R.F.C.)
| |
Collapse
|
20
|
Yu E, Xu Y, Shi Y, Yu Q, Liu J, Xu L. Discovery of novel natural compound inhibitors targeting estrogen receptor α by an integrated virtual screening strategy. J Mol Model 2019; 25:278. [PMID: 31463793 DOI: 10.1007/s00894-019-4156-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022]
Abstract
Estrogen receptor (ER) is a nuclear hormone receptor and plays an important role in mediating the cellular effects of estrogen. ER can be classified into two receptors: estrogen receptor alpha (ERα) and beta (ERβ), and the former is expressed in 50~80% of breast tumors and has been extensively investigated in breast cancer for decades. Excessive exposure to estrogen can obviously stimulate the growth of breast cancers primarily mediated by ERα, and thus anti-estrogen therapies by small molecules are of concern to clinicians and pharmaceutical industry in the treatment of ERα-positive breast cancers. Although a series of estrogen receptor modulators have been developed, these drugs can lead to resistance and side effects. Therefore, the development of small molecule inhibitors with high target specificity has been intensified. In this pursuit, an integrated computer-aided virtual screening technique, including molecular docking and pharmacophore model screening, was used to screen traditional Chinese medicine (TCM) databases. The compounds with high docking scores and fit values were subjected to ADME (adsorption, distribution, metabolism, excretion) and toxicity prediction, and ten hits were identified as potential inhibitors targeting ERα. Molecular docking was used to investigate the binding modes between ERα and three most potent hits, and molecular dynamic simulations were chosen to explore the stability of these complexes. The rank of the predicted binding free energies evaluated by MM/GBSA is consistent with the docking score. These novel scaffolds discovered in the present study can be used as critical starting point in the drug discovery process for treating ERα-positive breast cancer. Graphical abstract .
Collapse
Affiliation(s)
- Enguang Yu
- Department of Chinese Surgery, Jiaxing University Affiliated Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Yueping Xu
- Department of Nursing, Jiaxing University Affiliated Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Yanbo Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing University Affiliated Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Qiuyan Yu
- Department of Breast Surgery, Jiaxing University Affiliated Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Jie Liu
- Department of Traditional Chinese Medicine Oncology, Jiaxing University Affiliated Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, Jiangsu, China.
| |
Collapse
|
21
|
Fortini F, Vieceli Dalla Sega F, Caliceti C, Lambertini E, Pannuti A, Peiffer DS, Balla C, Rizzo P. Estrogen-mediated protection against coronary heart disease: The role of the Notch pathway. J Steroid Biochem Mol Biol 2019; 189:87-100. [PMID: 30817989 DOI: 10.1016/j.jsbmb.2019.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/05/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
Estrogen regulates a plethora of biological processes, under physiological and pathological conditions, by affecting key pathways involved in the regulation of cell proliferation, fate, survival and metabolism. The Notch receptors are mediators of communication between adjacent cells and are key determinants of cell fate during development and in postnatal life. Crosstalk between estrogen and the Notch pathway intervenes in many processes underlying the development and maintenance of the cardiovascular system. The identification of molecular mechanisms underlying the interaction between these types of endocrine and juxtacrine signaling are leading to a deeper understanding of physiological conditions regulated by these steroid hormones and, potentially, to novel therapeutic approaches to prevent pathologies linked to reduced levels of estrogen, such as coronary heart disease, and cardiotoxicity caused by hormone therapy for estrogen-receptor-positive breast cancer.
Collapse
Affiliation(s)
| | | | - Cristiana Caliceti
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Pannuti
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Daniel S Peiffer
- Oncology Research Institute, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA; Department of Microbiology and Immunology, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA
| | - Cristina Balla
- Cardiovascular Center, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, RA, Italy; Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
22
|
Fanning SW, Greene GL. Next-Generation ERα Inhibitors for Endocrine-Resistant ER+ Breast Cancer. Endocrinology 2019; 160:759-769. [PMID: 30753408 DOI: 10.1210/en.2018-01095] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/04/2019] [Indexed: 11/19/2022]
Abstract
One in eight women will be diagnosed with breast cancer in their lifetime. Because estrogen receptor-α (ERα) is expressed in ~70% of patients, therapeutic intervention by ERα-targeted endocrine therapies remains the leading strategy to prevent progression and/or metastasis in the adjuvant setting. However, the efficacy of these therapies will be diminished by the development of acquired resistance after prolonged treatment regimens. In preclinical models of endocrine-resistant metastatic breast cancers that retain ERα expression, antiestrogens with improved efficacy and potency can overcome resistance to shrink tumors and prevent metastasis. In particular, selective ER degraders or downregulators, which both antagonize ERα actions and induce its degradation, have demonstrated substantial antitumor efficacy in this setting. In the present review, we have discussed the mechanisms of acquired endocrine resistance in luminal breast cancers and the strategies used by next-generation endocrine therapies to antagonize ERα.
Collapse
Affiliation(s)
- Sean W Fanning
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Geoffrey L Greene
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| |
Collapse
|