1
|
Luo W, Chang G, Lin D, Xie H, Sun H, Li Z, Mo S, Wang R, Wang Y, Zheng Z. 3,3'-((3,4,5-trifluoropHenyl)methylene)bis(4-hydroxy-2H-chromen-2-one) inhibit lung cancer cell proliferation and migration. PLoS One 2024; 19:e0303186. [PMID: 38776295 PMCID: PMC11111047 DOI: 10.1371/journal.pone.0303186] [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: 10/07/2023] [Accepted: 04/19/2024] [Indexed: 05/24/2024] Open
Abstract
Lung cancer is a major public health challenge and, despite therapeutic improvements, is the first leading cause of cancer worldwide. The current cure rate from advanced cancer treatment is excessively low. Therefore, it is of great importance to identify novel, potent and less toxic anticancer agents for the treatment of lung cancer. The aim of our research is to synthesize a new biscoumarin 3,3'-((3,4,5-trifluorop -phenyl)methylene)bis(4-hydroxy-2H-chromen-2-one) (C35) as an anticancer agent. C35 was simply prepared by 4-hydroxycoumarin and 3,4,5-trifluorobenzaldehyde under ethanol and its structure was analyzed by spectroscopic analyses. The anti-proliferation effect of C35 was detected using CCK-8 assay. Migration abilities were measured by Transwell assay. The expression of correlated proteins was determined by Western blot. The results showed that C35 displayed strong cytostatic effects on lung cancer cell proliferation. In addition, C35 possessed a significant inhibition of migration by reducing the expression of matrix metalloproteinases-2 (MMP-2) and MMP-9 in lung cancer cells. Furthermore, C35 treatment suppressed the phosphorylation of p38 in lung cancer cells. Moreover, in vivo experiments were carried out, in which we treated Lewis tumor-bearing C57 mice via intraperitoneal injection of C35. Results showed that C35 inhibited tumor growth in vivo. In conclusion, our study demonstrated the anticancer activity of C35 via suppression of lung cancer cell proliferation and migration, which is possibly involved with the inhibition of the p38 pathway.
Collapse
Affiliation(s)
- Wenhui Luo
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Formula Granule, Guangdong Yifang Pharmaceutical Co., Ltd., Foshan, Guangdong Province, PR China
| | - Guoxin Chang
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Dingmei Lin
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Hongyi Xie
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Huilong Sun
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Zhibin Li
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Shirong Mo
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Ruixue Wang
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Yan Wang
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Zhaoguang Zheng
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| |
Collapse
|
2
|
Ming P, Li J, Yang L, Yu Y, Tang L, Zhou H, Zhang ZY, Zhang GJ. A Drug Molecule-Modified Graphene Field-Effect Transistor Nanosensor for Rapid, Label-Free, and Ultrasensitive Detection of Estrogen Receptor α Protein. Anal Chem 2024; 96:3454-3461. [PMID: 38359782 DOI: 10.1021/acs.analchem.3c04809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Estrogen receptor α (ERα) is an important biomarker in breast cancer diagnosis and treatment. Sensitive and accurate detection of ERα protein expression is crucial in guiding selection of an appropriate therapeutic strategy to improve the effectiveness and prognosis of breast cancer treatment. Herein, we report a liquid-gated graphene field-effect transistor (FET) biosensor that enables rapid, sensitive, and label-free detection of the ERα protein by employing a novel drug molecule as a capture probe. The drug molecule was synthesized and subsequently immobilized onto the sensing surface of the fabricated graphene FET, which was able to distinguish the ERα-positive from the ERα-negative protein. The developed sensor not only demonstrated a low detection limit (LOD: 2.62 fM) but also achieved a fast response to ERα protein samples within 30 min. Moreover, depending on the relationship between the change of dirac point and the ERα protein concentrations, the dissociation constant (Kd) was estimated to be 7.35 ± 0.06 pM, indicating that the drug probe-modified graphene FET had a good affinity with ERα protein. The nanosensor was able to analyze ERα proteins from 36 cell samples lysates. These results show that the graphene FET sensor was able to differentiate between ERα-positive and ERα-negative cells, indicating a promising biosensor for the ultrasensitive and rapid detection of ERα protein without antibody labeling.
Collapse
Affiliation(s)
- Pinghong Ming
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, P. R. China
- Department of Clinical Laboratory, The People's Hospital of Longhua, Shenzhen 518109, P. R. China
| | - Jiahao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, P. R. China
| | - Lu Yang
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, P. R. China
| | - Yi Yu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, P. R. China
| | - Lina Tang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, P. R. China
| | - Haibing Zhou
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, State Key Laboratory of Virology, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, P. R. China
| | - Zhi-Yong Zhang
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing 100871, P. R. China
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, P. R. China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, P. R. China
| |
Collapse
|
3
|
Koley M, Han J, Soloshonok VA, Mojumder S, Javahershenas R, Makarem A. Latest developments in coumarin-based anticancer agents: mechanism of action and structure-activity relationship studies. RSC Med Chem 2024; 15:10-54. [PMID: 38283214 PMCID: PMC10809357 DOI: 10.1039/d3md00511a] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/20/2023] [Indexed: 01/30/2024] Open
Abstract
Many researchers around the world are working on the development of novel anticancer drugs with different mechanisms of action. In this case, coumarin is a highly promising pharmacophore for the development of novel anticancer drugs. Besides, the hybridization of this moiety with other anticancer pharmacophores has emerged as a potent breakthrough in the treatment of cancer to decrease its side effects and increase its efficiency. This review aims to provide a comprehensive overview of the recent development of coumarin derivatives and their application as novel anticancer drugs. Herein, we highlight and describe the largest number of research works reported in this field from 2015 to August 2023, along with their mechanisms of action and structure-activity relationship studies, making this review different from the other review articles published on this topic to date.
Collapse
Affiliation(s)
- Manankar Koley
- CSIR-Central Glass & Ceramic Research Institute Kolkata India
| | - Jianlin Han
- College of Chemical Engineering, Nanjing Forestry University Nanjing China
| | - Vadim A Soloshonok
- Department of Organic Chemistry I, University of the Basque Country San Sebastián Spain
- IKERBASQUE, Basque Foundation for Science Bilbao Spain
| | | | - Ramin Javahershenas
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University Urmia Iran
| | - Ata Makarem
- Institute of Pharmacy, University of Hamburg Hamburg Germany
| |
Collapse
|
4
|
Perumal G, Govindan K, Jayaram A, Sundaramoorthi S, Lin W. Preliminary investigation on biological possessions of Saquinavir‐modified quinoline‐derived azadipeptidomimetics. J CHIN CHEM SOC-TAIP 2023. [DOI: 10.1002/jccs.202200504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Gopi Perumal
- Department of Chemistry Vellore Institute of Technology Vellore India
- Department of Medicinal and Applied Chemistry Kaohsiung Medical University Kaohsiung Taiwan, ROC
| | - Karthick Govindan
- Department of Medicinal and Applied Chemistry Kaohsiung Medical University Kaohsiung Taiwan, ROC
| | - Alageswaran Jayaram
- Department of Medicinal and Applied Chemistry Kaohsiung Medical University Kaohsiung Taiwan, ROC
| | | | - Wei‐Yu Lin
- Department of Medicinal and Applied Chemistry Kaohsiung Medical University Kaohsiung Taiwan, ROC
- Department of Medical Research Kaohsiung Medical University Hospital Kaohsiung Taiwan, ROC
- Drug Development and Value Creation Research Centre Kaohsiung Medical University Kaohsiung Taiwan, ROC
| |
Collapse
|
5
|
Human Estrogen Receptor Alpha Antagonists, Part 3: 3-D Pharmacophore and 3-D QSAR Guided Brefeldin A Hit-to-Lead Optimization toward New Breast Cancer Suppressants. Molecules 2022; 27:molecules27092823. [PMID: 35566172 PMCID: PMC9101642 DOI: 10.3390/molecules27092823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/01/2023] Open
Abstract
The estrogen receptor α (ERα) is an important biological target mediating 17β-estradiol driven breast cancer (BC) development. Aiming to develop innovative drugs against BC, either wild-type or mutated ligand-ERα complexes were used as source data to build structure-based 3-D pharmacophore and 3-D QSAR models, afterward used as tools for the virtual screening of National Cancer Institute datasets and hit-to-lead optimization. The procedure identified Brefeldin A (BFA) as hit, then structurally optimized toward twelve new derivatives whose anticancer activity was confirmed both in vitro and in vivo. Compounds as SERMs showed picomolar to low nanomolar potencies against ERα and were then investigated as antiproliferative agents against BC cell lines, as stimulators of p53 expression, as well as BC cell cycle arrest agents. Most active leads were finally profiled upon administration to female Wistar rats with pre-induced BC, after which 3DPQ-12, 3DPQ-3, 3DPQ-9, 3DPQ-4, 3DPQ-2, and 3DPQ-1 represent potential candidates for BC therapy.
Collapse
|
6
|
Kurtanović N, Tomašević N, Matić S, Mitrović MM, Kostić DA, Sabatino M, Antonini L, Ragno R, Mladenović M. Human estrogen receptor α antagonists, part 2: Synthesis driven by rational design, in vitro antiproliferative, and in vivo anticancer evaluation of innovative coumarin-related antiestrogens as breast cancer suppressants. Eur J Med Chem 2022; 227:113869. [PMID: 34710747 DOI: 10.1016/j.ejmech.2021.113869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/04/2021] [Accepted: 09/21/2021] [Indexed: 12/20/2022]
Abstract
New twelve in silico designed coumarin-based ERα antagonists, namely 3DQ-1a to 3DQ-1е, were synthesized and confirmed as selective ERα antagonists, showing potencies ranging from single-digit nanomolar to picomolar. The hits were confirmed as selective estrogen receptor modulators and validated as antiproliferative agents using MCF-7 breast cancer cell lines exerting from picomolar to low nanomolar potency, at the same time showing no agonistic activity within endometrial cell lines. Their mechanism of action was inspected and revealed to be through the inhibition of the Raf-1/MAPK/ERK signal transduction pathway, preventing hormone-mediated gene expression on either genomic direct or genomic indirect level, and stopping the MCF-7 cells proliferation at G0/G1 phase. In vivo experiments, by means of the per os administration to female Wistar rats with pre-induced breast cancer, distinguished six derivatives, 3DQ-4a, 3DQ-2a, 3DQ-1a, 3DQ-1b, 3DQ-2b, and 3DQ-3b, showing remarkable potency as tumor suppressors endowed with optimal pharmacokinetic profiles and no significant histopathological profiles. The presented data indicate the new compounds as potential candidates to be submitted in clinical trials for breast cancer therapy.
Collapse
Affiliation(s)
- Nezrina Kurtanović
- Kragujevac Center for Computational Biochemistry, Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000, Kragujevac, P.O. Box 60, Serbia
| | - Nevena Tomašević
- Kragujevac Center for Computational Biochemistry, Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000, Kragujevac, P.O. Box 60, Serbia
| | - Sanja Matić
- University of Kragujevac, Institute for Informational Technologies, Jovana Cvijića bb, 34000, Kragujevac, Serbia
| | - Marina M Mitrović
- University of Kragujevac, Faculty of Medical Sciences, Department of Biochemistry, Svetozara Markovića 69, 34000, Kragujevac, Serbia
| | - Danijela A Kostić
- University of Niš, Department of Chemistry, Faculty of Sciences and Mathematics, Višegradska 33, 18000, Niš, Serbia
| | - Manuela Sabatino
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy
| | - Lorenzo Antonini
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy
| | - Rino Ragno
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.
| | - Milan Mladenović
- Kragujevac Center for Computational Biochemistry, Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000, Kragujevac, P.O. Box 60, Serbia.
| |
Collapse
|
7
|
Matos MJ, Uriarte E, Santana L. 3-Phenylcoumarins as a Privileged Scaffold in Medicinal Chemistry: The Landmarks of the Past Decade. Molecules 2021; 26:6755. [PMID: 34771164 PMCID: PMC8587835 DOI: 10.3390/molecules26216755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/27/2022] Open
Abstract
3-Phenylcoumarins are a family of heterocyclic molecules that are widely used in both organic and medicinal chemistry. In this overview, research on this scaffold, since 2010, is included and discussed, focusing on aspects related to its natural origin, synthetic procedures and pharmacological applications. This review paper is based on the most relevant literature related to the role of 3-phenylcoumarins in the design of new drug candidates. The references presented in this review have been collected from multiple electronic databases, including SciFinder, Pubmed and Mendeley.
Collapse
Affiliation(s)
- Maria J Matos
- Centro de Investigação em Química da Universidade do Porto (CIQUP), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eugenio Uriarte
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Instituto de Ciencias Químicas Aplicadas, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Lourdes Santana
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
8
|
Coumarins as Tool Compounds to Aid the Discovery of Selective Function Modulators of Steroid Hormone Binding Proteins. MOLECULES (BASEL, SWITZERLAND) 2021; 26:molecules26175142. [PMID: 34500576 PMCID: PMC8433903 DOI: 10.3390/molecules26175142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 11/20/2022]
Abstract
Steroid hormones play an essential role in a wide variety of actions in the body, such as in metabolism, inflammation, initiating and maintaining sexual differentiation and reproduction, immune functions, and stress response. Androgen, aromatase, and sulfatase pathway enzymes and nuclear receptors are responsible for steroid biosynthesis and sensing steroid hormones. Changes in steroid homeostasis are associated with many endocrine diseases. Thus, the discovery and development of novel drug candidates require a detailed understanding of the small molecule structure–activity relationship with enzymes and receptors participating in steroid hormone synthesis, signaling, and metabolism. Here, we show that simple coumarin derivatives can be employed to build cost-efficiently a set of molecules that derive essential features that enable easy discovery of selective and high-affinity molecules to target proteins. In addition, these compounds are also potent tool molecules to study the metabolism of any small molecule.
Collapse
|
9
|
Ribeiro RFN, Cavadas C, Silva MMC. Small-molecule modulators of the circadian clock: Pharmacological potentials in circadian-related diseases. Drug Discov Today 2021; 26:1620-1641. [PMID: 33781946 DOI: 10.1016/j.drudis.2021.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/20/2021] [Accepted: 03/16/2021] [Indexed: 12/29/2022]
Abstract
Disruption of circadian oscillations has a wide-ranging impact on health, with the potential to induce the development of clock-related diseases. Small-molecule modulators of the circadian clock (SMMCC) target core or noncore clock proteins, modulating physiological effects as a consequence of agonist, inverse agonist, or antagonist interference. These pharmacological modulators are usually identified using chemical screening of large libraries of active compounds. However, target-based screens, chemical optimization, and circadian crystallography have recently assisted in the identification of these compounds. In this review, we focus on established and novel SMMCCs targeting both core and noncore clock proteins, identifying their circadian targets, detailed circadian effects, and specific physiological effects. In addition, we discuss their therapeutic potential for the treatment of diverse clock-related disorders (such as metabolic-associated disorders, autoimmune diseases, mood disorders, and cancer) and as chronotherapeutics. Future perspectives are also considered, such as clinical trials, and potential safety hazards, including those in the absence of clinical trials.
Collapse
Affiliation(s)
- Rodrigo F N Ribeiro
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| | - Maria Manuel C Silva
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
10
|
Al-Warhi T, Sabt A, Elkaeed EB, Eldehna WM. Recent advancements of coumarin-based anticancer agents: An up-to-date review. Bioorg Chem 2020; 103:104163. [DOI: 10.1016/j.bioorg.2020.104163] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022]
|
11
|
Meng Q, Xie B, Ma X, Hu Z, Zhou F, Zhou HB, Dong C. Rational design of ERα targeting hypoxia turn-on fluorescent probes with antiproliferative activity for breast cancer. Chem Commun (Camb) 2020; 56:10493-10496. [PMID: 32776045 DOI: 10.1039/c9cc09754f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The overexpression of estrogen receptor (ER) α is not only closely related to the development of ER+ breast cancer, but is also an important biomarker for clinical diagnosis and treatment. Herein, we report several ERα targeting hypoxia turn-on fluorescent probes with antitumor activity for breast cancer cells. Among them, probes 3 and 5 displayed good ERα targeting ability and favorable hypoxia turn-on response in MCF-7 cells. Moreover, the probes 3 and 5 exhibited good antiproliferative activity towards MCF-7 cells (IC50 = 8.5 μM, 10.3 μM) and a much lower cytotoxicity to normal cells compared with the positive control. It is expected that these novel fluorescent probes may provide useful tools for the theranostics of ER+ breast cancer.
Collapse
Affiliation(s)
- Qiuyu Meng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, State Key Laboratory of Virology, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China.
| | | | | | | | | | | | | |
Collapse
|
12
|
Francisco CS, Francisco CS, Constantino AF, Neto ÁC, Lacerda V. Synthetic Methods Applied in the Preparation of Coumarin-based Compounds. CURR ORG CHEM 2020. [DOI: 10.2174/1385272823666191121150047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Coumarins (2H-chromen-2-ones) are heterocyclic compounds of wide scientific
interest due to their important biological and pharmaceutical properties such as antitumor,
antioxidant, anti-inflammatory and antimicrobial activities as well as enzymatic inhibitors
related to neurodegenerative diseases. Due to their structural variability, this compound
class has been attracting considerable interest in the natural products and synthetic organic
chemistry areas. Coumarins and their derivatives have been prepared by a variety of methods,
including Perkin, Wittig and Reformatsky reactions, Pechmann and Knoevenagel
condensations, and Claisen rearrangement, among others. In the present review we report
the different synthetic methods used in the preparation of coumarin derivatives exploited
in the last ten years (from 2008 to 2018), regarding the research demand for new structural
scaffolds.
Collapse
Affiliation(s)
- Carla S. Francisco
- Programa de Pós-Graduação em Química, Centro de Ciências Exatas, Universidade Federal do Espírito Santo, Campus Goiabeiras, Vitória, Brazil
| | - Cristina S. Francisco
- Programa de Pós-Graduação em Química, Centro de Ciências Exatas, Universidade Federal do Espírito Santo, Campus Goiabeiras, Vitória, Brazil
| | | | - Álvaro Cunha Neto
- Programa de Pós-Graduação em Química, Centro de Ciências Exatas, Universidade Federal do Espírito Santo, Campus Goiabeiras, Vitória, Brazil
| | - Valdemar Lacerda
- Programa de Pós-Graduação em Química, Centro de Ciências Exatas, Universidade Federal do Espírito Santo, Campus Goiabeiras, Vitória, Brazil
| |
Collapse
|
13
|
Design, Synthesis and Biological Evaluation of Novel Coumarin-Based Hydroxamate Derivatives as Histone Deacetylase (Hdac) Inhibitors with Antitumor Activities. Molecules 2019; 24:molecules24142569. [PMID: 31311163 PMCID: PMC6680717 DOI: 10.3390/molecules24142569] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/23/2022] Open
Abstract
A series of novel coumarin-based hydroxamate derivatives were designed and synthesized as histone deacetylase inhibitors (HDACis). Selective compounds showed a potent HDAC inhibition with nM IC50 values, with the best compound (10e) being nearly 90 times more active than vorinostat (SAHA) against HDAC1. Compounds 10e and 11d also increased the levels of acetylated histone H3 and H4, which is consistent with their strong HDAC inhibition. In addition, 10e and 11d displayed a higher potency toward human A549 and Hela cancer cell lines compared with SAHA. Moreover, 10e and 11d significantly arrested A549 cells at the G2/M phase and enhanced apoptosis. Molecular docking studies revealed the possible mode of interaction of compounds 10e and 12a with HDAC1. Our findings suggest that these novel coumarin-based HDAC inhibitors provide a promising scaffold for the development of new potential cancer chemotherapies.
Collapse
|
14
|
Synthesis and biological evaluation of novel 4,7-dihydroxycoumarin derivatives as anticancer agents. Bioorg Med Chem Lett 2019; 29:1819-1824. [DOI: 10.1016/j.bmcl.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/21/2023]
|
15
|
Hu Z, Yang L, Ning W, Tang C, Meng Q, Zheng J, Dong C, Zhou HB. A high-affinity subtype-selective fluorescent probe for estrogen receptor β imaging in living cells. Chem Commun (Camb) 2018; 54:3887-3890. [PMID: 29610818 DOI: 10.1039/c8cc00483h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Estrogen receptor β (ERβ) has recently been identified as a pharmaceutical target in hormone replacement therapy for breast cancers. However, the biological function of ERβ in disease progression remains unclear. A highly ERβ-selective fluorescent probe (FPNM) was discovered exhibiting nanomolar affinity for ERβ with an ERβ/ERα selectivity as high as 80, which allowed specific labeling of intracellular ERβ. Moreover, distinct ERβ dynamics in various cellular bio-settings such as prostate cancer (DU-145) or triple-negative breast cancer (MDA-MB-231) cells were directly observed for the first time viaFPNM staining.
Collapse
Affiliation(s)
- Zhiye Hu
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Hubei Provincial Key Laboratory of Developmentally Originated Disease, State Key Laboratory of Virology, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China.
| | | | | | | | | | | | | | | |
Collapse
|