1
|
Long J, Ye P, Yuan W, Yang Q, Wang Z, Xiao H, Xie Z, Lei X, Yang X, Deng X, Tang G. Research progress of flavonoids targeting estrogen receptor in the treatment of breast cancer. Bioorg Med Chem 2025; 120:118106. [PMID: 39938393 DOI: 10.1016/j.bmc.2025.118106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Breast cancer (BC) stands as the most prevalent malignancy among women. Targeting the estrogen receptor (ER) or ER pathway is one of the important approaches for ER+ BC treatment. As a class of phytoestrogens, flavonoids possess notable anti-tumor properties and hold immense potential in regulating ER signaling. In this review, we reported the recent advances in both in vitro and in vivo studies of flavonoids and their synthetic derivatives targeting the ER signaling pathway, including the target and mechanism of action of these molecules, as well as their structure-activity relationship. Based on the available literature, the beneficial effects of flavonoids as ER targeting agents are promising but they require further in vitro and in vivo studies to enable its translation from bench to bedside. This review will provide valuable guidance and insights for the future development of drugs targeting the ER pathway.
Collapse
Affiliation(s)
- Jianling Long
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Pengju Ye
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Weixi Yuan
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qixian Yang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, China
| | | | - Zhizhong Xie
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyan Yang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Guotao Tang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
2
|
Talagayev V, Chen Y, Doering NP, Obendorf L, Denzinger K, Puls K, Lam K, Liu S, Wolf CA, Noonan T, Breznik M, Knaus P, Wolber G. OpenMMDL - Simplifying the Complex: Building, Simulating, and Analyzing Protein-Ligand Systems in OpenMM. J Chem Inf Model 2025; 65:1967-1978. [PMID: 39933881 PMCID: PMC11863370 DOI: 10.1021/acs.jcim.4c02158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025]
Abstract
Molecular dynamics (MD) simulations have become an essential tool for studying the dynamics of biological systems and exploring protein-ligand interactions. OpenMM is a modern, open-source software toolkit designed for MD simulations. Until now, it has lacked a module dedicated to building receptor-ligand systems, which is highly useful for investigating protein-ligand interactions for drug discovery. We therefore introduce OpenMMDL, an open-source toolkit that enables the preparation and simulation of protein-ligand complexes in OpenMM, along with the subsequent analysis of protein-ligand interactions. OpenMMDL consists of three main components: OpenMMDL Setup, a graphical user interface based on Python Flask to prepare protein and simulation settings, OpenMMDL Simulation to perform MD simulations with consecutive trajectory postprocessing, and finally OpenMMDL Analysis to analyze simulation results with respect to ligand binding. OpenMMDL is not only a versatile tool for analyzing protein-ligand interactions and generating ligand binding modes throughout simulations; it also tracks and clusters water molecules, particularly those exhibiting minimal displacement from their previous coordinates, providing insights into solvent dynamics. We applied OpenMMDL to study ligand-receptor interactions across diverse biological systems, including LDN-193189 and LDN-212854 with ALK2 (kinases), nifedipine and amlodipine in Cav1.1 (ion channels), LSD in 5-HT2B (G-protein coupled receptors), letrozole in CYP19A1 (cytochrome P450 oxygenases), flavin mononucleotide binding the FMN-riboswitch (RNAs), ligand C08 bound to TLR8 (toll-like receptor), and PZM21 bound to MOR (opioid receptor), highlighting distinct functionalities of OpenMMDL. OpenMMDL is publicly available at https://github.com/wolberlab/OpenMMDL.
Collapse
Affiliation(s)
- Valerij Talagayev
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Yu Chen
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Niklas Piet Doering
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Leon Obendorf
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
- Department
of Biology, Chemistry and Pharmacy, Institute
of Biochemistry, Signal Transduction Group, Thielallee 64, 14195 Berlin, Germany
| | - Katrin Denzinger
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Kristina Puls
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Kevin Lam
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Sijie Liu
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Clemens Alexander Wolf
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Theresa Noonan
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Marko Breznik
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Petra Knaus
- Department
of Biology, Chemistry and Pharmacy, Institute
of Biochemistry, Signal Transduction Group, Thielallee 64, 14195 Berlin, Germany
| | - Gerhard Wolber
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| |
Collapse
|
3
|
Ferreira Almeida C, Correia-da-Silva G, Teixeira N, Amaral C. Influence of tumor microenvironment on the different breast cancer subtypes and applied therapies. Biochem Pharmacol 2024; 223:116178. [PMID: 38561089 DOI: 10.1016/j.bcp.2024.116178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Despite the significant improvements made in breast cancer therapy during the last decades, this disease still has increasing incidence and mortality rates. Different targets involved in general processes, like cell proliferation and survival, have become alternative therapeutic options for this disease, with some of them already used in clinic, like the CDK4/6 inhibitors for luminal A tumors treatment. Nevertheless, there is a demand for novel therapeutic strategies focused not only on tumor cells, but also on their microenvironment. Tumor microenvironment (TME) is a very complex and dynamic system that, more than surrounding and supporting tumor cells, actively participates in tumor development and progression. During the last decades, it has become clear that the cellular and acellular components of TME differ between the various breast cancer subtypes and shape the differences regarding their severity and prognosis. The pivotal role of the TME in controlling tumor growth and influencing responses to therapy represents a potential source for novel targets and therapeutic strategies. In this review, we present a description of the multiple therapeutic options used for different breast cancer subtypes, as well as the influence that the TME may exert on the development of the disease and on the response to the distinct therapies, which in some cases may explain their failure by the occurrence of relapses and resistance. Furthermore, the ongoing studies focused on the use of TME components for developing potential cancer treatments are described.
Collapse
Affiliation(s)
- Cristina Ferreira Almeida
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Natércia Teixeira
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|
4
|
Xie Y, Yang L, Wu Y, Zheng H, Gou Q. Adjuvant endocrine therapy in patients with estrogen receptor-low positive breast cancer: A prospective cohort study. Breast 2022; 66:89-96. [PMID: 36209701 PMCID: PMC9551143 DOI: 10.1016/j.breast.2022.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Little is known about the benefits of adjuvant endocrine therapy (ET) in low ER-positive breast cancer (1%-10%) patients. We analyzed the association between ET and breast cancer-specific survival (BCSS) in these patients with respect to the regimen and the duration of ET. METHODS Patients were classified into three groups based on the regimen and duration of ET. The regimens included aromatase inhibitor (AI) monotherapy or sequential tamoxifen followed by an AI (AI/T + AI), or only tamoxifen and no ET. The duration of ET included 2-3 years and >3 years. Multivariate Cox regression analysis was employed to calculate the hazard ratios (HRs) with 95% confidence intervals (CIs). RESULTS Of the 10,696 patients diagnosed with breast cancer between 2010 and 2020, 407 women were identified with ER-low positive disease and met the inclusion criteria. During a median follow-up of 5.2 years, patients who received ET improved BCSS. Of them, those with AI/T + AI had increased BCSS compared to patients without ET, after adjusting for demographics and tumor characteristics, especially in ER-low/HER-2-positive breast cancer. After additional adjustment for treatment mode, the association maintained a similar trend. Patients who received >3 years of ET was associated with a better DFS. There was no significant difference in BCSS between patients with 2-3 years and >3 years of ET. CONCLUSION For ER-low patients, findings suggest that ET with AI/T + AI may be a reasonable treatment alternative. This effect should be assessed in randomized studies.
Collapse
Affiliation(s)
- Yuxin Xie
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China,Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Libo Yang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanqi Wu
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China,Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zheng
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China,Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiheng Gou
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China,Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Radiation Oncology and Head & Neck Oncology Division, Cancer Center, West China Hospital, Sichuan University, Chengdu, China,Corresponding author. Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, 37 Guoxue Xiang, Wuhou District, Chengdu, 610041, China. Tel: +86-28-85422685
| |
Collapse
|
5
|
Caciolla J, Martini S, Spinello A, Belluti F, Bisi A, Zaffaroni N, Magistrato A, Gobbi S. Single-digit nanomolar inhibitors lock the aromatase active site via a dualsteric targeting strategy. Eur J Med Chem 2022; 244:114802. [DOI: 10.1016/j.ejmech.2022.114802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/04/2022]
|
6
|
De Luca M, Occhiuzzi MA, Rizzuti B, Ioele G, Ragno G, Garofalo A, Grande F. Interaction of letrozole and its degradation products with aromatase: chemometric assessment of kinetics and structure-based binding validation. J Enzyme Inhib Med Chem 2022; 37:1600-1609. [PMID: 35635194 PMCID: PMC9176668 DOI: 10.1080/14756366.2022.2081845] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Letrozole is one of the most prescribed drugs for the treatment of breast cancer in post-menopausal women, and it is endowed with selective peripheral aromatase inhibitory activity. The efficacy of this drug is also a consequence of its long-lasting activity, likely due to its metabolic stability. The reactivity of cyano groups in the letrozole structure could, however, lead to chemical derivatives still endowed with residual biological activity. Herein, the chemical degradation process of the drug was studied by coupling multivariate curve resolution and spectrophotometric methodologies in order to assess a detailed kinetic profile. Three main derivatives were identified after drug exposure to different degradation conditions, consisting of acid-base and oxidative environments and stressing light. Molecular docking confirmed the capability of these compounds to accommodate into the active site of the enzyme, suggesting that the sustained inhibitory activity of letrozole may be at least in part attributed to the degradation compounds.
Collapse
Affiliation(s)
- Michele De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | - Bruno Rizzuti
- CNR-NANOTEC, SS Rende (CS), Department of Physics, University of Calabria, Rende, Italy
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Joint Unit GBsC-CSIC-BIFI, University of Zaragoza, Zaragoza, Spain
| | - Giuseppina Ioele
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Gaetano Ragno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Antonio Garofalo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
7
|
Wang Y, Jing F, Wang H. Role of Exemestane in the Treatment of Estrogen-Receptor-Positive Breast Cancer: A Narrative Review of Recent Evidence. Adv Ther 2022; 39:862-891. [PMID: 34989983 DOI: 10.1007/s12325-021-01924-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/14/2021] [Indexed: 11/01/2022]
Abstract
INTRODUCTION Breast cancer (BC) is the most common type of cancer diagnosed among women worldwide with an estimated 2.3 million new cases every year. Almost two-thirds of all patients with BC have estrogen receptor-positive (ER+) tumors. In this review, the clinical evidence of exemestane in different treatment settings in ER+ BC is presented and summarized. SEARCH STRATEGY A search strategy with the keywords "breast cancer [MeSH Terms]" AND "exemestane [Title/Abstract]" was devised and a search was performed in PubMed. RESULTS The efficacy of exemestane in different treatment settings has been established by numerous clinical studies. Exemestane is recommended as an adjuvant treatment in postmenopausal women previously treated with tamoxifen in trials comparing 5 years of tamoxifen with 2-3 years of tamoxifen combined with 2-3 years of exemestane, which proved that treatment with exemestane provided better survival outcomes. Similarly, exemestane could be considered as a safe treatment option for neoadjuvant treatment, prevention of chemotherapy, and treatment of advanced BC either alone or in combination with other targeted therapy drugs in both pre- and postmenopausal women. CONCLUSION Exemestane could be considered as a reasonable therapeutic option in the treatment of ER+ BC at any stage in pre- and postmenopausal women.
Collapse
|
8
|
Shah V, Bhaliya J, Patel GM. In silico docking and ADME study of deketene curcumin derivatives (DKC) as an aromatase inhibitor or antagonist to the estrogen-alpha positive receptor (Erα+): potent application of breast cancer. Struct Chem 2022; 33:571-600. [PMID: 35106036 PMCID: PMC8794617 DOI: 10.1007/s11224-021-01871-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022]
Abstract
Regardless of many extensive studies, hormonal-based breast cancer is the most common cause of cancer-related mortality of females worldwide. Indeed, estrogen receptor-positive (ER +) is the communal subtype in breast cancer. To treat this, three types of medications are typically used: selective estrogen receptor modulators (SERMs), selective estrogen receptor down modulators (SERDMs), and aromatase inhibitors (AIs), all of which directly interact with the activation of the estrogen signaling pathway and its formation. Despite their effectiveness, the development of new treatments is required since clinical efficacy is restricted owing to resistance. As a result, in silico studies for drug discovery are booming over the decades because of their affordability and less time-consuming features. Here, 25 deketene curcumin derivatives have been selected for docking studies through MVD software over the positive type of breast cancer through both the treatment hosts Erα + receptor and aromatase. DKC compounds are used because they have several pharmacological uses, including anti-cancer, anti-diabetic, anti-viral, anti-fungal, and anti-bacterial properties. Moreover, an ADME study was carried out for DKC derivatives that reveal the optimum drug-likeness profile. From 25 derivatives, the results showed a better MolDock score, hydrogen bonding, and steric interaction between compounds DKC-10, DKC-20, and DKC-21 with Erα + and aromatase. Although the study was done on both the treatable path hosts, better results were obtained with Erα + as an antagonist. Therefore, it is proposed that three selected DKC derivatives would be better therapeutic agents against breast cancer.
Collapse
Affiliation(s)
- Vraj Shah
- Department of Chemistry, School of Science, ITM SLS Baroda University, Vadodara, 391510 India
| | - Jaydip Bhaliya
- Department of Chemistry, School of Science, ITM SLS Baroda University, Vadodara, 391510 India
| | - Gautam M. Patel
- Department of Industrial Chemistry, Institute of Science & Technology for Advanced Studies & Research (ISTAR), CVM University, V.V., Nagar, 388120 GJ India
| |
Collapse
|
9
|
Steel TR, Walsh F, Wieczorek-Błauż A, Hanif M, Hartinger CG. Monodentately-coordinated bioactive moieties in multimodal half-sandwich organoruthenium anticancer agents. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213890] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Ammazzalorso A, Agamennone M, De Filippis B, Fantacuzzi M. Development of CDK4/6 Inhibitors: A Five Years Update. Molecules 2021; 26:molecules26051488. [PMID: 33803309 PMCID: PMC7967197 DOI: 10.3390/molecules26051488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/24/2022] Open
Abstract
The inhibition of cyclin dependent kinases 4 and 6 plays a role in aromatase inhibitor resistant metastatic breast cancer. Three dual CDK4/6 inhibitors have been approved for the breast cancer treatment that, in combination with the endocrine therapy, dramatically improved the survival outcomes both in first and later line settings. The developments of the last five years in the search for new selective CDK4/6 inhibitors with increased selectivity, treatment efficacy, and reduced adverse effects are reviewed, considering the small-molecule inhibitors and proteolysis-targeting chimeras (PROTACs) approaches, mainly pointing at structure-activity relationships, selectivity against different kinases and antiproliferative activity.
Collapse
|
11
|
Adhikari N, Baidya SK, Jha T. Effective anti-aromatase therapy to battle against estrogen-mediated breast cancer: Comparative SAR/QSAR assessment on steroidal aromatase inhibitors. Eur J Med Chem 2020; 208:112845. [DOI: 10.1016/j.ejmech.2020.112845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 02/08/2023]
|
12
|
Caciolla J, Bisi A, Belluti F, Rampa A, Gobbi S. Reconsidering Aromatase for Breast Cancer Treatment: New Roles for an Old Target. Molecules 2020; 25:molecules25225351. [PMID: 33207783 PMCID: PMC7696276 DOI: 10.3390/molecules25225351] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 12/31/2022] Open
Abstract
The current therapeutic approach for the treatment of hormone dependent breast cancer includes interference with estrogen receptors via either selective modulators or estrogens deprivation, by preventing their biosynthesis with aromatase inhibitors. Severe side effects and acquired resistance are drawbacks of both drug classes, and the efforts to overcome these issues still allow for research in this field to be animated. This review reports on recent findings that have opened new avenues for reconsidering the role of aromatase enzymes (and estrogen receptors) leading to the possibility of looking at well-known targets in a new perspective.
Collapse
Affiliation(s)
- Jessica Caciolla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
13
|
Verma SK, Ratre P, Jain AK, Liang C, Gupta GD, Thareja S. De novo designing, assessment of target affinity and binding interactions against aromatase: Discovery of novel leads as anti-breast cancer agents. Struct Chem 2020. [DOI: 10.1007/s11224-020-01673-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
14
|
Banjare L, Verma SK, Jain AK, Thareja S. Design and pharmacophoric identification of flavonoid scaffold‐based aromatase inhibitors. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical SciencesGuru Ghasidas Central University Bilaspur Chhattisgarh India
| | - Sant Kumar Verma
- School of Pharmaceutical SciencesGuru Ghasidas Central University Bilaspur Chhattisgarh India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical SciencesGuru Ghasidas Central University Bilaspur Chhattisgarh India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural ProductsCentral University of Punjab Bathinda Punjab India
| |
Collapse
|
15
|
Aromatase inhibitors: Role in postmenopausal breast cancer. Arch Pharm (Weinheim) 2020; 353:e2000081. [DOI: 10.1002/ardp.202000081] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 12/17/2022]
|
16
|
Banjare L, Verma SK, Jain AK, Thareja S. Lead Molecules as Novel Aromatase Inhibitors: In Silico De Novo Designing and Binding Affinity Studies. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190703152659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Aromatase inhibitors emerged as a pivotal moiety to selectively block estrogen production, prevention and treatment of tumour growth in breast cancer. De novo drug design is an alternative approach to blind virtual screening for successful designing of the novel molecule against various therapeutic targets.Objective:In the present study, we have explored the de novo approach to design novel aromatase inhibitors.Method:The e-LEA3D, a computational-aided drug design web server was used to design novel drug-like candidates against the target aromatase. For drug-likeness ADME parameters (molecular weight, H-bond acceptors, H-bond donors, LogP and number of rotatable bonds) of designed molecules were calculated in TSAR software package, geometry optimization and energy minimization was accomplished using Chem Office. Further, molecular docking study was performed in Molegro Virtual Docker (MVD).Results:Among 17 generated molecules using the de novo pathway, 13 molecules passed the Lipinski filter pertaining to their bioavailability characteristics. De novo designed molecules with drug-likeness were further docked into the mapped active site of aromatase to scale up their affinity and binding fitness with the target. Among de novo fabricated drug like candidates (1-13), two molecules (5, 6) exhibited higher affinity with aromatase in terms of MolDock score (-150.650, -172.680 Kcal/mol, respectively) while molecule 8 showed lowest target affinity (-85.588 Kcal/mol).Conclusion:The binding patterns of lead molecules (5, 6) could be used as a pharmacophore for medicinal chemists to explore these molecules for their aromatase inhibitory potential.
Collapse
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| | - Sant Kumar Verma
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| | - Suresh Thareja
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| |
Collapse
|
17
|
Ferreira Almeida C, Oliveira A, João Ramos M, Fernandes PA, Teixeira N, Amaral C. Estrogen receptor-positive (ER +) breast cancer treatment: Are multi-target compounds the next promising approach? Biochem Pharmacol 2020; 177:113989. [PMID: 32330493 DOI: 10.1016/j.bcp.2020.113989] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
Endocrine therapy is currently the main therapeutic approach for estrogen receptor-positive (ER+) breast cancer, the most frequent subtype of breast cancer in women worldwide. For this subtype of tumors, the current clinical treatment includes aromatase inhibitors (AIs) and anti-estrogenic compounds, such as Tamoxifen and Fulvestrant, being AIs the first-line treatment option for post-menopausal women. Moreover, the recent guidelines also suggest the use of these compounds by pre-menopausal women after suppressing ovaries function. However, besides its therapeutic efficacy, the prolonged use of this type of therapies may lead to the development of several adverse effects, as well as, endocrine resistance, limiting the effectiveness of such treatments. In order to surpass this issues and clinical concerns, during the last years, several studies have been suggesting alternative therapeutic approaches, considering the function of aromatase, ERα and ERβ. Here, we review the structural and functional features of these three targets and their importance in ER+ breast cancer treatment, as well as, the current treatment strategies used in clinic, emphasizing the importance of the development of multi-target compounds able to simultaneously modulate these key targets, as a novel and promising therapeutic strategy for this type of cancer.
Collapse
Affiliation(s)
- Cristina Ferreira Almeida
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Ana Oliveira
- UCIBIO.REQUIMTE, Computational Biochemistry Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Maria João Ramos
- UCIBIO.REQUIMTE, Computational Biochemistry Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Pedro A Fernandes
- UCIBIO.REQUIMTE, Computational Biochemistry Laboratory, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|
18
|
Kaur K, Jaitak V. Recent Development in Indole Derivatives as Anticancer Agents for Breast Cancer. Anticancer Agents Med Chem 2020; 19:962-983. [PMID: 30864529 DOI: 10.2174/1871520619666190312125602] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Breast Cancer (BC) is the second most common cause of cancer related deaths in women. Due to severe side effects and multidrug resistance, current therapies like hormonal therapy, surgery, radiotherapy and chemotherapy become ineffective. Also, the existing drugs for BC treatment are associated with several drawbacks such as poor oral bioavailability, non-selectivity and poor pharmacodynamics properties. Therefore, there is an urgent need for the development of more effective and safer anti BC agents. OBJECTIVE This article explored in detail the possibilities of indole-based heterocyclic compounds as anticancer agents with breast cancer as their major target. METHODS Recent literature related to indole derivatives endowed with encouraging anti BC potential is reviewed. With special focus on BC, this review offers a detailed account of multiple mechanisms of action of various indole derivatives: aromatase inhibitor, tubulin inhibitor, microtubule inhibitor, targeting estrogen receptor, DNA-binding mechanism, induction of apoptosis, inhibition of PI3K/AkT/NFkB/mTOR, and HDAC inhibitors, by which these derivatives have shown promising anticancer potential. RESULTS Exhaustive literature survey indicated that indole derivatives are associated with properties of inducing apoptosis and disturbing tubulin assembly. Indoles are also associated with the inhibition of NFkB/mTOR/PI3K/AkT and regulation of estrogen-mediated activity. Furthermore, indole derivatives have been found to modulate critical targets such as topoisomerase and HDAC. These derivatives have shown significant activity against breast cancer cells. CONCLUSION In BC, indole derivatives seem to be quite competent and act through various mechanisms that are well established in case of BC. This review has shown that indole derivatives can further be explored for the betterment of BC chemotherapy. A lot of potential is still hidden which demands to be discovered for upgrading BC chemotherapy.
Collapse
Affiliation(s)
- Kamalpreet Kaur
- Laboratory of Natural Products, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda (Pb) -151001, India
| | - Vikas Jaitak
- Laboratory of Natural Products, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda (Pb) -151001, India
| |
Collapse
|
19
|
Gaber M, Hany M, Mokhtar S, Helmy MW, Elkodairy KA, Elzoghby AO. Boronic-targeted albumin-shell oily-core nanocapsules for synergistic aromatase inhibitor/herbal breast cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110099. [DOI: 10.1016/j.msec.2019.110099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/13/2019] [Accepted: 08/18/2019] [Indexed: 12/28/2022]
|
20
|
Anisiewicz A, Filip-Psurska B, Pawlik A, Nasulewicz-Goldeman A, Piasecki T, Kowalski K, Maciejewska M, Jarosz J, Banach J, Papiernik D, Mazur A, Kutner A, Maier JA, Wietrzyk J. Calcitriol Analogues Decrease Lung Metastasis but Impair Bone Metabolism in Aged Ovariectomized Mice Bearing 4T1 Mammary Gland Tumours. Aging Dis 2019; 10:977-991. [PMID: 31595196 PMCID: PMC6764735 DOI: 10.14336/ad.2018.0921] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022] Open
Abstract
Calcitriol and its analogues are considered drugs supporting the anticancer treatment of breast cancer and preventing the osteoporosis that results from the development of cancer or from chemotherapy or hormone therapy. Following the orthotopic implantation of 4T1 mammary carcinoma cells into aged ovariectomized (OVX) mice, we evaluated the effects of calcitriol and its two analogues, PRI-2191 and PRI-2205, on metastatic spread and bone homeostasis. Calcitriol and its analogues temporarily inhibited the formation of metastases in the lungs. Unexpectedly, only mice treated with calcitriol analogues showed a deterioration of bone-related parameters, such as bone column density, marrow column density and the CaPO4 coefficient. These findings correlated with an increased number of active osteoclasts differentiated from bone marrow-derived macrophages in mice treated with the analogues. Interestingly, in the tumours from mice treated with PRI-2191 and PRI-2205, the expression of Tnfsf11 (RANKL) was increased. On the other hand, osteopontin (OPN) levels in plasma and tumour tissue, as well as TRAC5b levels in tumours, were diminished by calcitriol and its analogues. Despite a similar action of both analogues towards bone metabolism, their impact on vitamin D metabolism differed. In particular, PRI-2191 and calcitriol, not PRI-2205 treatment significantly diminished the levels of both 25(OH)D3 and 24,25(OH)2D3. In conclusion, though there is evident antimetastatic activity in old OVX mice, signs of increased bone metabolism and deterioration of bone mineralization during therapy with calcitriol analogues were observed.
Collapse
Affiliation(s)
- Artur Anisiewicz
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Beata Filip-Psurska
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Agata Pawlik
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Anna Nasulewicz-Goldeman
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Tomasz Piasecki
- 2Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | | | - Magdalena Maciejewska
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Joanna Jarosz
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Joanna Banach
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Diana Papiernik
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Andrzej Mazur
- 4Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, F-63000 Clermont-Ferrand, France
| | - Andrzej Kutner
- 5Pharmaceutical Research Institute, 01-793 Warsaw, Poland
| | - Jeanette A Maier
- 6Università di Milano, Dept. Biomedical and Clinical Sciences, 20157 Milano, Italy
| | - Joanna Wietrzyk
- 1Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| |
Collapse
|
21
|
Click chemistry in silico, docking, quantum chemical calculations, and molecular dynamics simulations to identify novel 1,2,4-triazole-based compounds as potential aromatase inhibitors. SN APPLIED SCIENCES 2019. [DOI: 10.1007/s42452-019-1051-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
22
|
Banjare L, Verma SK, Jain AK, Thareja S. Structure Guided Molecular Docking Assisted Alignment Dependent 3DQSAR Study on Steroidal Aromatase Inhibitors (SAIs) as Anti-breast Cancer Agents. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666181010101024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background:
In spite of the availability of various treatment approaches including
surgery, radiotherapy, and hormonal therapy, the steroidal aromatase inhibitors (SAIs) play a
significant role as chemotherapeutic agents for the treatment of estrogen-dependent breast cancer
with the benefit of reduced risk of recurrence. However, due to greater toxicity and side effects
associated with currently available anti-breast cancer agents, there is emergent requirement to
develop target-specific AIs with safer anti-breast cancer profile.
Methods:
It is challenging task to design target-specific and less toxic SAIs, though the molecular
modeling tools viz. molecular docking simulations and QSAR have been continuing for more than
two decades for the fast and efficient designing of novel, selective, potent and safe molecules
against various biological targets to fight the number of dreaded diseases/disorders. In order to
design novel and selective SAIs, structure guided molecular docking assisted alignment dependent
3D-QSAR studies was performed on a data set comprises of 22 molecules bearing steroidal
scaffold with wide range of aromatase inhibitory activity.
Results:
3D-QSAR model developed using molecular weighted (MW) extent alignment approach
showed good statistical quality and predictive ability when compared to model developed using
moments of inertia (MI) alignment approach.
Conclusion:
The explored binding interactions and generated pharmacophoric features (steric and
electrostatic) of steroidal molecules could be exploited for further design, direct synthesis and
development of new potential safer SAIs, that can be effective to reduce the mortality and
morbidity associated with breast cancer.
Collapse
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| | - Sant Kumar Verma
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| | - Suresh Thareja
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| |
Collapse
|
23
|
Roleira FMF, Varela C, Amaral C, Costa SC, Correia-da-Silva G, Moraca F, Costa G, Alcaro S, Teixeira NAA, Tavares da Silva EJ. C-6α- vs C-7α-Substituted Steroidal Aromatase Inhibitors: Which Is Better? Synthesis, Biochemical Evaluation, Docking Studies, and Structure–Activity Relationships. J Med Chem 2019; 62:3636-3657. [DOI: 10.1021/acs.jmedchem.9b00157] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fernanda M. F. Roleira
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Carla Varela
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Saul C. Costa
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Federica Moraca
- Laboratorio di Chimica Farmaceutica, Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, 88100 Catanzaro, Italy
- Department of Pharmacy, University of Naples “Federico II”, via D. Montesano 49, 80131, Naples, Italy
- Net4Science Academic Spin-Off, “Magna Græcia” University of Catanzaro, “S. Venuta”, Catanzaro, Italy
| | - Giosuè Costa
- Laboratorio di Chimica Farmaceutica, Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, “Magna Græcia” University of Catanzaro, “S. Venuta”, Catanzaro, Italy
| | - Stefano Alcaro
- Laboratorio di Chimica Farmaceutica, Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, “Magna Græcia” University of Catanzaro, “S. Venuta”, Catanzaro, Italy
| | - Natércia A. A. Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Elisiário J. Tavares da Silva
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| |
Collapse
|
24
|
Olvera-Hernández S, Hernández A, Reyes R, Fernández-Guasti A. Establishment of partner preference in male rats: Effect of prenatal letrozole and sexual experience. Horm Behav 2019; 109:56-63. [PMID: 30769015 DOI: 10.1016/j.yhbeh.2019.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 02/05/2019] [Accepted: 02/10/2019] [Indexed: 01/07/2023]
Abstract
Repeated testing for masculine sexual behavior influences female sex preference in males. Males perinatally treated with aromatase inhibitors show male preference, but also copulate with the receptive female. Such copulation modifies sex preference most likely because of its rewarding properties. In this study, we intended to equal the incentive value of both stimuli -in the sex preference test- by using receptive females with vaginal occlusion. Vehicle and letrozole-treated (0.56 μg/kg, gestation days 10-21) males were repeatedly tested for sex preference at 40, 55, 70, 85 and 100 days of age. These ages were selected because males of 40 days are unable to copulate, while by 100 days of age almost all males show the complete repertoire of masculine sexual behavior. At 40 days of age, males of all groups fail to show sex preference and none of them was able to copulate. In controls of 100 days of age all males showed female-sex preference and all intromitted the female. A large proportion (44%) of vehicle-treated males that could not copulate the female showed male preference. Twenty to 30% of the prenatally letrozole treated males also had same-sex preference even if they could copulate; and most of them (67%) had a male preference when copulation was precluded. These data support the idea that copulation is crucial for developing a female preference in control animals. The results suggest that brain changes produced by estrogens along early development and stimuli coming from the partner are essential for shaping sex preference.
Collapse
Affiliation(s)
- Sandra Olvera-Hernández
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, México
| | - Alejandra Hernández
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, México
| | - Rebeca Reyes
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, México
| | - Alonso Fernández-Guasti
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, México.
| |
Collapse
|
25
|
Abstract
The mineralocorticoid aldosterone is an important regulator of blood pressure and electrolyte balance. However, excess aldosterone can be deleterious as a driver of inflammation, vascular remodeling and tissue fibrosis associated with cardiometabolic diseases. Mineralocorticoid receptor antagonists (MRA) and renin-angiotensin-aldosterone system (RAAS) antagonists are current clinical therapies used to antagonize deleterious effects of aldosterone in patients. MRAs compete with aldosterone for binding at its cognate receptor thereby limiting its effect while RAS antagonists reduce aldosterone levels indirectly by blocking the stimulatory effect of angiotensin. Both MRAs and RAS antagonists can result in incomplete inhibition of the harmful effects of excess aldosterone. Aldosterone synthase (AS) inhibitors (ASI) attenuate the production of aldosterone directly and have been proposed as an alternative to MRAs and RAS blockers. Cortisol synthase (CS) is an enzyme closely related to AS and responsible for generating the important glucocorticoid cortisol, required for maintaining critical metabolic and immune responses. The importance of selectivity against CS is shown by early examples of ASIs that were only modestly selective and as such, attenuated cortisol responses when evaluated in patients. Recently, next-generation, highly selective ASIs have been described and are presently being evaluated in the clinic as an alternative to angiotensin and MR antagonists for cardiometabolic disease. Herein we provide a brief review of the challenges associated with discovery of selective ASIs and the transition from the early compounds that paved the way toward the next-generation of highly selective ASIs currently under development.
Collapse
Affiliation(s)
- Steven M Weldon
- Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States.
| | - Nicholas F Brown
- Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, United States
| |
Collapse
|
26
|
Heck AL, Crestani CC, Fernández-Guasti A, Larco DO, Mayerhofer A, Roselli CE. Neuropeptide and steroid hormone mediators of neuroendocrine regulation. J Neuroendocrinol 2018; 30:e12599. [PMID: 29645316 PMCID: PMC6181757 DOI: 10.1111/jne.12599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/27/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
To maintain the health and well-being of all mammals, numerous aspects of physiology are controlled by neuroendocrine mechanisms. These mechanisms ultimately enable communication between neurones and glands throughout the body and are centrally mediated by neuropeptides and/or steroid hormones. A recent session at the International Workshop in Neuroendocrinology highlighted the essential roles of some of these neuropeptide and steroid hormone mediators in the neuroendocrine regulation of stress-, reproduction- and behaviour-related processes. Accordingly, the present review highlights topics presented in this session, including the role of the neuropeptides corticotrophin-releasing factor and gonadotrophin-releasing hormone in stress and reproductive physiology, respectively. Additionally, it details an important role for gonadal sex steroids in the development of behavioural sex preference.
Collapse
Affiliation(s)
- Ashley L. Heck
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO USA 80523
| | - Carlos C. Crestani
- Laboratory of Pharmacology, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil 14800-903
| | | | | | - Artur Mayerhofer
- Biomedical Center, Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), Planegg, Germany 82152
| | - Charles E. Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR USA 97239-3098
| |
Collapse
|
27
|
Shoombuatong W, Schaduangrat N, Nantasenamat C. Towards understanding aromatase inhibitory activity via QSAR modeling. EXCLI JOURNAL 2018; 17:688-708. [PMID: 30190660 PMCID: PMC6123608 DOI: 10.17179/excli2018-1417] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/10/2018] [Indexed: 12/14/2022]
Abstract
Aromatase is a rate-limiting enzyme for estrogen biosynthesis that is overproduced in breast cancer tissue. To block the growth of breast tumors, aromatase inhibitors (AIs) are employed to bind and inhibit aromatase in order to lower the amount of estrogen produced in the body. Although a number of synthetic aromatase inhibitors have been released for clinical use in the treatment of hormone-receptor positive breast cancer, these inhibitors may lead to undesirable side effects (e.g. increased rash, diarrhea and vomiting; effects on the bone, brain and heart) and therefore, the search for novel AIs continues. Over the past decades, there has been an intense effort in employing medicinal chemistry and quantitative structure-activity relationship (QSAR) to shed light on the mechanistic basis of aromatase inhibition. To the best of our knowledge, this article constitutes the first comprehensive review of all QSAR studies of both steroidal and non-steroidal AIs that have been published in the field. Herein, we summarize the experimental setup of these studies as well as summarizing the key features that are pertinent for robust aromatase inhibition.
Collapse
Affiliation(s)
- Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
28
|
Kuruva CS, Gandavaram SP, Kadiam VS, Valluru L, Chamarthi NR. Synthesis of New Heteroaryl α‐Aminophosphonates and Evaluation of Their Cytotoxicity against Human Breast Cancer MCF‐7 Cell Lines. ChemistrySelect 2018. [DOI: 10.1002/slct.201800700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Chandra S. Kuruva
- Department of ChemistrySri Venkateswara University Tirupati 517502 India
| | - Syam P. Gandavaram
- Department of ChemistrySree Vidyanikethan Engineering College, Sree Sainath Nagar Tirupati 517102 India
| | | | | | - Naga R. Chamarthi
- Department of ChemistrySri Venkateswara University Tirupati 517502 India
| |
Collapse
|
29
|
Jayapal JJ, Dhanaraj S. Exemestane loaded alginate nanoparticles for cancer treatment: Formulation and in vitro evaluation. Int J Biol Macromol 2017; 105:416-421. [DOI: 10.1016/j.ijbiomac.2017.07.064] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/26/2017] [Accepted: 07/10/2017] [Indexed: 11/29/2022]
|
30
|
Anisiewicz A, Pawlik A, Filip-Psurska B, Turlej E, Dzimira S, Milczarek M, Gdesz K, Papiernik D, Jarosz J, Kłopotowska D, Kutner A, Mazur A, Wietrzyk J. Unfavorable effect of calcitriol and its low-calcemic analogs on metastasis of 4T1 mouse mammary gland cancer. Int J Oncol 2017; 52:103-126. [PMID: 29115583 PMCID: PMC5743363 DOI: 10.3892/ijo.2017.4185] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022] Open
Abstract
Low vitamin D status is considered as a risk factor for breast cancer and has prognostic significance. Furthermore, vitamin D deficiency increases after adjuvant cancer therapy, which alters bone metabolism increasing the risk of osteoporosis. It is now postulated that vitamin D supplementation in breast cancer treatment delays the recurrence of cancer thereby extending survival. We evaluated the impact of calcitriol and its low-calcemic analogs, PRI-2191 and PRI-2205, on the tumor growth, angiogenesis, and metastasis of 4T1 mouse mammary gland cancer. Gene expression analysis related to cancer invasion/metastasis, real-time PCR, ELISA, western blotting, and histochemical studies were performed. In vitro studies were conducted to compare the effects of calcitriol and its analogs on 4T1 and 67NR cell proliferation and expression of selected proteins. Calcitriol and its analogs increased lung metastasis without influencing the growth of primary tumor. The levels of plasma 17β-estradiol and transforming growth factor β (TGFβ) were found to be elevated after treatment. Moreover, the results showed that tumor blood perfusion improved and osteopontin (OPN) levels increased, whereas vascular endothelial growth factor (VEGF) and TGFβ levels decreased in tumors from treated mice. All the studied treatments resulted in increased collagen content in the tumor tissue in the early step of tumor progression, and calcitriol caused an increase in collagen content in lung tissue. In addition, in vitro proliferation of 4T1 tumor cells was not found to be affected by calcitriol or its analogs in contrast to non-metastatic 67NR cells. Calcitriol and its analogs enhanced the metastatic potential of 4T1 mouse mammary gland cancer by inducing the secretion of OPN probably via host cells. In addition, OPN tumor overexpression prevailed over the decreasing tumor TGFβ level and blood vessel normalization via tumor VEGF deprivation induced by calcitriol and its analogs. Moreover, the increased plasma TGFβ and 17β-estradiol levels contributed to the facilitation of metastatic process.
Collapse
Affiliation(s)
- Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Agata Pawlik
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Eliza Turlej
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Stanisław Dzimira
- Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Katarzyna Gdesz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Diana Papiernik
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Joanna Jarosz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Dagmara Kłopotowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Andrzej Kutner
- Department of Pharmacology, Pharmaceutical Research Institute, 01-793 Warsaw, Poland
| | - Andrzej Mazur
- Université Clermont Auvergne, INRA, UNH, F-63000 Clermont-Ferrand, France
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| |
Collapse
|
31
|
van Dijk M, ter Laak AM, Wichard JD, Capoferri L, Vermeulen NPE, Geerke DP. Comprehensive and Automated Linear Interaction Energy Based Binding-Affinity Prediction for Multifarious Cytochrome P450 Aromatase Inhibitors. J Chem Inf Model 2017; 57:2294-2308. [PMID: 28776988 PMCID: PMC5615371 DOI: 10.1021/acs.jcim.7b00222] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 11/30/2022]
Abstract
Cytochrome P450 aromatase (CYP19A1) plays a key role in the development of estrogen dependent breast cancer, and aromatase inhibitors have been at the front line of treatment for the past three decades. The development of potent, selective and safer inhibitors is ongoing with in silico screening methods playing a more prominent role in the search for promising lead compounds in bioactivity-relevant chemical space. Here we present a set of comprehensive binding affinity prediction models for CYP19A1 using our automated Linear Interaction Energy (LIE) based workflow on a set of 132 putative and structurally diverse aromatase inhibitors obtained from a typical industrial screening study. We extended the workflow with machine learning methods to automatically cluster training and test compounds in order to maximize the number of explained compounds in one or more predictive LIE models. The method uses protein-ligand interaction profiles obtained from Molecular Dynamics (MD) trajectories to help model search and define the applicability domain of the resolved models. Our method was successful in accounting for 86% of the data set in 3 robust models that show high correlation between calculated and observed values for ligand-binding free energies (RMSE < 2.5 kJ mol-1), with good cross-validation statistics.
Collapse
Affiliation(s)
- Marc van Dijk
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | | | - Jörg D. Wichard
- Bayer AG, Pharmaceuticals Division, Müllerstrasse
178, D-13353 Berlin, Germany
| | - Luigi Capoferri
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Nico P. E. Vermeulen
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Daan P. Geerke
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
32
|
Adhikari N, Amin SA, Saha A, Jha T. Combating breast cancer with non-steroidal aromatase inhibitors (NSAIs): Understanding the chemico-biological interactions through comparative SAR/QSAR study. Eur J Med Chem 2017. [DOI: 10.1016/j.ejmech.2017.05.041] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
33
|
Jha T, Adhikari N, Halder AK, Saha A. Ligand- and Structure-Based Drug Design of Non-Steroidal Aromatase Inhibitors (NSAIs) in Breast Cancer. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aromatase is a multienzyme complex overexpressed in breast cancer and responsible for estrogen production. It is the potential target for designing anti-breast cancer drugs. Ligand and Structure-Based Drug Designing approaches (LBDD and SBDD) are involved in development of active and more specific Nonsteroidal Aromatase Inhibitors (NSAIs). Different LBDD and SBDD approaches are presented here to understand their utility in designing novel NSAIs. It is observed that molecules should possess a five or six membered heterocyclic nitrogen containing ring to coordinate with heme portion of aromatase for inhibition. Moreover, one or two hydrogen bond acceptor features, hydrophobicity, and steric factors may play crucial roles for anti-aromatase activity. Electrostatic, van der Waals, and p-p interactions are other important factors that determine binding affinity of inhibitors. HQSAR, LDA-QSAR, GQSAR, CoMFA, and CoMSIA approaches, pharmacophore mapping followed by virtual screening, docking, and dynamic simulation may be effective approaches for designing new potent anti-aromatase molecules.
Collapse
|
34
|
Song Z, Liu Y, Dai Z, Liu W, Zhao K, Zhang T, Hu Y, Zhang X, Dai Y. Synthesis and aromatase inhibitory evaluation of 4-N-nitrophenyl substituted amino-4H-1,2,4-triazole derivatives. Bioorg Med Chem 2016; 24:4723-4730. [DOI: 10.1016/j.bmc.2016.08.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 01/10/2023]
|
35
|
Sobral AF, Amaral C, Correia-da-Silva G, Teixeira N. Unravelling exemestane: From biology to clinical prospects. J Steroid Biochem Mol Biol 2016; 163:1-11. [PMID: 26992705 DOI: 10.1016/j.jsbmb.2016.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/29/2016] [Accepted: 03/13/2016] [Indexed: 11/28/2022]
Abstract
Aromatase inhibitors (AIs) are anti-tumor agents used in clinic to treat hormone-dependent breast cancer. AIs block estrogens biosynthesis by inhibiting the enzyme aromatase, preventing tumor progression. Exemestane, a third-generation steroidal AI, belongs to this class of drugs and is currently used in clinic to treat postmenopausal women, due to its high efficacy and good tolerability. Here, its pharmacological and biological aspects as well as its clinical applications and comparison to other endocrine therapeutic agents, are reviewed. It is also focused the benefits and risks of exemestane, drawbacks to be overcome and aspects to be explored.
Collapse
Affiliation(s)
- Ana Filipa Sobral
- Faculty of Science and Technology, University of Coimbra, Calçada Martim de Freitas 3000-456 Coimbra, Portugal; UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| | - Georgina Correia-da-Silva
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| |
Collapse
|
36
|
Varela CL, Amaral C, Correia-da-Silva G, Costa SC, Carvalho RA, Costa G, Alcaro S, Teixeira NA, Tavares-da-Silva EJ, Roleira FM. Exploring new chemical functionalities to improve aromatase inhibition of steroids. Bioorg Med Chem 2016; 24:2823-31. [DOI: 10.1016/j.bmc.2016.04.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 02/08/2023]
|
37
|
Sashidhara KV, Singh LR, Choudhary D, Arun A, Gupta S, Adhikary S, Palnati GR, Konwar R, Trivedi R. Design, synthesis and in vitro evaluation of coumarin–imidazo[1,2-a]pyridine derivatives against cancer induced osteoporosis. RSC Adv 2016. [DOI: 10.1039/c6ra15674f] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The potential of coumarin–imidazo[1,2-a]pyridine hybrids to prevent bone loss in patients with bone metastases is discussed.
Collapse
Affiliation(s)
- Koneni V. Sashidhara
- Medicinal and Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - L. Ravithej Singh
- Medicinal and Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | | | - Ashutosh Arun
- Endocrinology Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Sampa Gupta
- Medicinal and Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Sulekha Adhikary
- Endocrinology Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Gopala Reddy Palnati
- Medicinal and Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Rituraj Konwar
- Endocrinology Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Ritu Trivedi
- Endocrinology Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| |
Collapse
|
38
|
Yadav MR, Barmade MA, Tamboli RS, Murumkar PR. Developing steroidal aromatase inhibitors-an effective armament to win the battle against breast cancer. Eur J Med Chem 2015; 105:1-38. [DOI: 10.1016/j.ejmech.2015.09.038] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 01/05/2023]
|
39
|
Mast N, Lin JB, Pikuleva IA. Marketed Drugs Can Inhibit Cytochrome P450 27A1, a Potential New Target for Breast Cancer Adjuvant Therapy. Mol Pharmacol 2015; 88:428-36. [PMID: 26082378 PMCID: PMC4551053 DOI: 10.1124/mol.115.099598] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 06/16/2015] [Indexed: 02/06/2023] Open
Abstract
Cytochrome P450 CYP27A1 is the only enzyme in humans converting cholesterol to 27-hydroxycholesterol, an oxysterol of multiple functions, including tissue-specific modulation of estrogen and liver X receptors. Both receptors seem to mediate adverse effects of 27-hydroxycholesterol in breast cancer when the levels of this oxysterol are elevated. The present work assessed druggability of CYP27A1 as a potential antibreast cancer target. We selected 26 anticancer and noncancer medications, most approved by the Food and Drug Administration, and evaluated them first in vitro for inhibition of purified recombinant CYP27A1 and binding to the enzyme active site. Six strong CYP27A1 inhibitors/binders were identified. These were the two antibreast cancer pharmaceuticals anastrozole and fadrozole, antiprostate cancer drug bicalutamide, sedative dexmedetomidine, and two antifungals ravuconazole and posaconazole. Anastrozole was then tested in vivo on mice, which received subcutaneous drug injections for 1 week. Mouse plasma and hepatic 27-hydroxycholesterol levels were decreased 2.6- and 1.6-fold, respectively, whereas plasma and hepatic cholesterol content remained unchanged. Thus, pharmacologic CYP27A1 inhibition is possible in the whole body and individual organs, but does not negatively affect cholesterol elimination. Our results enhance the potential of CYP27A1 as an antibreast cancer target, could be of importance for the interpretation of Femara versus Anastrozole Clinical Evaluation Trial, and bring attention to posaconazole as a potential complementary anti-breast cancer medication. More medications on the US market may have unanticipated off-target inhibition of CYP27A1, and we propose strategies for their identification.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Joseph B Lin
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
40
|
García-Cárdenas N, Olvera-Hernández S, Gómez-Quintanar BN, Fernández-Guasti A. Male rats with same sex preference show high experimental anxiety and lack of anxiogenic-like effect of fluoxetine in the plus maze test. Pharmacol Biochem Behav 2015; 135:128-35. [DOI: 10.1016/j.pbb.2015.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/22/2015] [Accepted: 05/28/2015] [Indexed: 01/04/2023]
|
41
|
Correction: Xie, H.; et al. 3D QSAR studies, pharmacophore modeling and virtual screening on a series of steroidal aromatase inhibitors. Int. J. Mol. Sci. 2014, 15, 20927-20947. Int J Mol Sci 2015; 16:5072-5. [PMID: 25751723 PMCID: PMC4394465 DOI: 10.3390/ijms16035072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 02/16/2015] [Accepted: 02/18/2015] [Indexed: 11/17/2022] Open
|
42
|
Prenatal letrozole produces a subpopulation of male rats with same-sex preference and arousal as well as female sexual behavior. Physiol Behav 2015; 139:403-11. [DOI: 10.1016/j.physbeh.2014.11.060] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/28/2023]
|
43
|
Jha T, Adhikari N, Halder AK, Saha A. Ligand- and Structure-Based Drug Design of Non-Steroidal Aromatase Inhibitors (NSAIs) in Breast Cancer. QUANTITATIVE STRUCTURE-ACTIVITY RELATIONSHIPS IN DRUG DESIGN, PREDICTIVE TOXICOLOGY, AND RISK ASSESSMENT 2015. [DOI: 10.4018/978-1-4666-8136-1.ch011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Aromatase is a multienzyme complex overexpressed in breast cancer and responsible for estrogen production. It is the potential target for designing anti-breast cancer drugs. Ligand and Structure-Based Drug Designing approaches (LBDD and SBDD) are involved in development of active and more specific Nonsteroidal Aromatase Inhibitors (NSAIs). Different LBDD and SBDD approaches are presented here to understand their utility in designing novel NSAIs. It is observed that molecules should possess a five or six membered heterocyclic nitrogen containing ring to coordinate with heme portion of aromatase for inhibition. Moreover, one or two hydrogen bond acceptor features, hydrophobicity, and steric factors may play crucial roles for anti-aromatase activity. Electrostatic, van der Waals, and p-p interactions are other important factors that determine binding affinity of inhibitors. HQSAR, LDA-QSAR, GQSAR, CoMFA, and CoMSIA approaches, pharmacophore mapping followed by virtual screening, docking, and dynamic simulation may be effective approaches for designing new potent anti-aromatase molecules.
Collapse
|
44
|
Vosooghi M, Firoozpour L, Rodaki A, Pordeli M, Safavi M, Ardestani SK, Dadgar A, Asadipour A, Moshafi MH, Foroumadi A. Design, synthesis, docking study and cytotoxic activity evaluation of some novel letrozole analogs. ACTA ACUST UNITED AC 2014; 22:83. [PMID: 25539909 PMCID: PMC4284924 DOI: 10.1186/s40199-014-0083-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/08/2014] [Indexed: 11/23/2022]
Abstract
Background Breast cancer is the most common type of female cancer. One class of hormonal therapy for breast cancer drugs -non steroidal aromatase inhibitors- are triazole analogues. In this work, some derivatives of these drugs was designed and synthesized. All synthesized compounds were evaluated for their cytotoxic activities on breast cancer cell lines (MDA-MB-231, T47D and MCF-7). Methods Our synthetic route for designed compounds started from 4-bromotolunitrile which was reacted with 1H-1,2,4-triazole to afford 4-(4-cyanobenzyl)-1,2,4-triazole. The reaction of later compound with aromatic aldehydes led to formation of the designed compounds. Eleven novel derivatives 1a-k were tested for their cytotoxic activities on three human breast cancer cell lines. Results Among the synthesized compound, 4-[2-(3-chlorophenyl)-1-(1H-1,2,4-triazol-1-yl)ethenyl]benzonitrile (1c) showed the highest activity against MCF-7 and MDA-MB-231 cell lines and 4-[2-(4-methoxyphenyl)-1-(1H-1,2,4-triazol-1-yl)ethenyl]benzonitrile (1 h) exhibited highest activity against T47D cell line. According to cytotoxic activities results, compound 4-[2-(4-dimethylamino)-1-(1H-1,2,4-triazol-1-yl)ethenyl]benzonitrile (1 k) showed comparative activity against T47D and MDA-MB-231 cell lines with compound (1 h) and our reference drug Etoposide. Conclusion In the process of anti-cancer drug discovery, to find new potential anti-breast cancer agents, we designed and synthesized a novel series of letrozole analogs. Cytotoxicity evaluation revealed that compounds (1c) and (1 k) were the most potent compounds with comparative activity with Etoposide. The results revealed that π-π interactions are responsible for the enzyme inhibitions of compounds (1 c) and (1 k).
Collapse
Affiliation(s)
- Mohsen Vosooghi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Loghman Firoozpour
- Drug Design and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Abolfazl Rodaki
- Drug Design and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahboobeh Pordeli
- Institute of Biochemistry and Biophysics, University of Tehran, PO Box 13145-1384, Tehran, Iran.
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran.
| | - Sussan K Ardestani
- Institute of Biochemistry and Biophysics, University of Tehran, PO Box 13145-1384, Tehran, Iran.
| | - Armin Dadgar
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ali Asadipour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medicinal Sciences, Kerman, Iran.
| | - Mohammad Hassan Moshafi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medicinal Sciences, Kerman, Iran.
| | - Alireza Foroumadi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medicinal Sciences, Kerman, Iran. .,Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
3D QSAR studies, pharmacophore modeling and virtual screening on a series of steroidal aromatase inhibitors. Int J Mol Sci 2014; 15:20927-47. [PMID: 25405729 PMCID: PMC4264204 DOI: 10.3390/ijms151120927] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/28/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022] Open
Abstract
Aromatase inhibitors are the most important targets in treatment of estrogen-dependent cancers. In order to search for potent steroidal aromatase inhibitors (SAIs) with lower side effects and overcome cellular resistance, comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) were performed on a series of SAIs to build 3D QSAR models. The reliable and predictive CoMFA and CoMSIA models were obtained with statistical results (CoMFA: q2 = 0.636, r2ncv = 0.988, r2pred = 0.658; CoMSIA: q2 = 0.843, r2ncv = 0.989, r2pred = 0.601). This 3D QSAR approach provides significant insights that can be used to develop novel and potent SAIs. In addition, Genetic algorithm with linear assignment of hypermolecular alignment of database (GALAHAD) was used to derive 3D pharmacophore models. The selected pharmacophore model contains two acceptor atoms and four hydrophobic centers, which was used as a 3D query for virtual screening against NCI2000 database. Six hit compounds were obtained and their biological activities were further predicted by the CoMFA and CoMSIA models, which are expected to design potent and novel SAIs.
Collapse
|
46
|
Xie H, Qiu K, Xie X. Pharmacophore modeling, virtual screening, and 3D-QSAR studies on a series of non-steroidal aromatase inhibitors. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1257-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Ji JZ, Lao KJ, Hu J, Pang T, Jiang ZZ, Yuan HL, Miao JS, Chen X, Ning SS, Xiang H, Guo YM, Yan M, Zhang LY. Discovery of novel aromatase inhibitors using a homogeneous time-resolved fluorescence assay. Acta Pharmacol Sin 2014; 35:1082-92. [PMID: 25047514 PMCID: PMC4125720 DOI: 10.1038/aps.2014.53] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/20/2014] [Indexed: 01/04/2023]
Abstract
AIM Aromatase is an important target for drugs to treat hormone-dependent diseases, including breast cancer. The aim of this study was to develop a homogeneous time-resolved fluorescence (HTRF) aromatase assay suitable for high-throughput screening (HTS). METHODS A 384-well aromatase HTRF assay was established, and used to screen about 7000 compounds from a compound library. Anti-proliferation activity of the hit was evaluated using alamarBlue(R) assay in a hormone-dependent breast cancer cell line T47D. Molecular docking was conducted to elucidate the binding mode of the hit using the Discovery Studio program. RESULTS The Z' value and signal to background (S/B) ratio were 0.74 and 5.4, respectively. Among the 7000 compounds, 4 hits (XHN22, XHN26, XHN27 and triptoquinone A) were found to inhibit aromatase with IC50 values of 1.60±0.07, 2.76±0.24, 0.81±0.08 and 45.8±11.3 μmol /L, respectively. The hits XHN22, XHN26 and XHN27 shared the same chemical scaffold of 4-imidazolyl quinoline. Moreover, the most potent hit XHN27 at 10 and 50 μmol/L inhibited the proliferation of T47D cells by 45.3% and 35.2%, respectively. The docking study revealed that XHN27 docked within the active site of aromatase and might form a hydrogen bond and had a π-cation interaction with amino acid residues of the protein. CONCLUSION XHN27, an imidazolyl quinoline derivative of flavonoid, is a potent aromatase inhibitor with anti-proliferation activity against breast cancer in vitro. The established assay can be used in HTS for discovering novel aromatase inhibitor.
Collapse
Affiliation(s)
- Jin-zi Ji
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Ke-jing Lao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Hu
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Pang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Zhen-zhou Jiang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China
| | - Hao-liang Yuan
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-shan Miao
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Chen
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Shan-shan Ning
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hua Xiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-meng Guo
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Yan
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Lu-yong Zhang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- State Key laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
48
|
Van Asten K, Neven P, Lintermans A, Wildiers H, Paridaens R. Aromatase inhibitors in the breast cancer clinic: focus on exemestane. Endocr Relat Cancer 2014; 21:R31-49. [PMID: 24434719 DOI: 10.1530/erc-13-0269] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Breast cancer is the most prevalent type of cancer in women and responsible for significant female cancer-related mortality worldwide. In the Western world, over 80% of breast cancers are hormone-receptor positive for which endocrine therapy is administered. The main anti-estrogen treatments in use consist of selective estrogen-receptor modulators, such as tamoxifen, and third-generation aromatase inhibitors (AIs), such as exemestane, letrozole, and anastrozole. In this review, the focus will lie on exemestane, its clinical use, and its side-effect profile. Exemestane is the only third-generation steroidal AI. Its efficacy as a first-line treatment in metastatic breast cancer has been demonstrated. Therefore, exemestane could be considered a valid first-line therapeutic option, but it also can be used in second-line or further situations. Exemestane is mostly used as part of sequential adjuvant treatment following tamoxifen, but in this setting it is also active in monotherapy. Furthermore, this AI has been studied in the neoadjuvant setting as presurgical treatment, and even as chemoprevention in high-risk healthy postmenopausal women. It may reverse side effects of tamoxifen, such as endometrial changes and thromboembolic disease but may also cause some inconvenient side effects itself. Additionally, there is a lack of total cross-resistance between exemestane and nonsteroidal AIs as far as their anti-tumoral efficacy is concerned; moreover the two classes of AIs display a nontotal overlapping toxicity profile. Taking together, exemestane can be considered as a useful treatment option at all stages of breast cancer.
Collapse
Affiliation(s)
- Kathleen Van Asten
- KU Leuven, Department of Oncology, Leuven, Belgium University Hospitals Leuven, Department of Gynecology and Obstetrics, Leuven, Belgium University Hospitals Leuven, Department of General Medical Oncology, Leuven, Belgium
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Martin GDA, Narvaez J, Marti A. Synthesis and bioconversions of formestane. JOURNAL OF NATURAL PRODUCTS 2013; 76:1966-1969. [PMID: 24074257 DOI: 10.1021/np400585t] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
In an effort to generate new steroidal aromatase inhibitors, formestane (4-hydroxyandrost-4-ene-3,17-dione) (1) was biotransformed by Rhizopus oryzae to yield the known 4β,5α-dihydroxyandrostane-3,17-dione as the major product (5) and bioconverted by Beauveria bassiana to afford the known reduced 4,17β-dihydroxyandrost-4-en-3-one (6) and 3α,17β-dihydroxy-5β-androstan-4-one (7) and the new 4,11α,17β-trihydroxyandrost-4-en-3-one (8). All the metabolites showed more potent activities than their parent congener in the aromatase and MCF-7 breast cancer assays. The bioactivities and structural elucidation of these metabolites as well as the semisynthesis of formestane (1) from testosterone (2) are reported herein.
Collapse
Affiliation(s)
- Glenroy D A Martin
- Chemistry, Biochemistry and Physics Department, The University of Tampa , 401 West Kennedy Boulevard, Tampa, Florida 33606-1490, United States
| | | | | |
Collapse
|