1
|
Altwaijry NA, Omar MA, Mohamed HS, Mounier MM, Afifi AH, Srour AM. Design, synthesis, molecular docking and anticancer activity of benzothiazolecarbohydrazide-sulfonate conjugates: insights into ROS-induced DNA damage and tubulin polymerization inhibition. RSC Adv 2025; 15:5895-5905. [PMID: 39990814 PMCID: PMC11843914 DOI: 10.1039/d4ra07810a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/07/2025] [Indexed: 02/25/2025] Open
Abstract
A series of novel benzothiazolecarbohydrazide-sulfonate conjugates 6a-l were designed, synthesized, and then assessed as potential antiproliferative agents in three distinct human cancer cell lines: MCF-7 (breast cancer), HCT-116 (colon cancer), and PC3 (prostate cancer), along with a normal cell line (BJ-1). The reference standard used was 5-fluorouracil. The results obtained reveal that the newly synthesized analogs demonstrate varying degrees of cytotoxicity against the targeted cell lines; however, compounds 6i and 6e exhibited the highest efficacy against MCF-7, HCT-116, and PC3 cells with IC50 values of 78.8 ± 2.6, 81.4 ± 1.9, and 90.6 ± 2.7 μM, respectively, compared to an IC50 of 78.4 ± 4.2 μM for 5-FU in MCF-7 cells, 29.2 ± 1.7 μM in HCT-116 cells and >200 μM in PC3 cells. Moreover, the most potent compounds demonstrated acceptable safety profiles when evaluated aganist BJ-1 cells. Consequently, compound 6i, which possesses no cytotoxicity towards BJ-1 cells and displays promising anticancer activity, was further investigated for its impact on tubulin polymerization compared to control MCF-7 cells, 210.3 and 632.9 pg ml-1, respectively. Compound 6i was found to significantly elevate the ROS levels in treated cancer cells, resulting in an 8.3-fold increase in DNA fragmentation compared to untreated cells. Additionally, it raised the percentage of accumulated cells in the G2 phase from 6.85% to 18.27% in MCF-7 cells. A molecular docking technique was conducted to elucidate the binding energy, binding pose, and binding interactions of compound 6i, revealing a strong fit within the active sites of the tubulin-colchicine binding site (CBS). This study provides valuable insights into the design and synthesis of novel anticancer agents targeting tubulin polymerization.
Collapse
Affiliation(s)
- Najla A Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University Riyadh Saudi Arabia
| | - Mohamed A Omar
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Hanaa S Mohamed
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Marwa M Mounier
- Department of Pharmacognosy, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Ahmed H Afifi
- Department of Pharmacognosy, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Aladdin M Srour
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| |
Collapse
|
2
|
Zheng M, Liu G, Han Y, Qian P, Wu M, Xiang M, Zhou Y. Synthesis, biological evaluation and mechanism study of a novel indole-pyridine chalcone derivative as antiproliferative agent against tumor cells through dual targeting tubulin and HK2. Eur J Med Chem 2025; 282:117058. [PMID: 39571460 DOI: 10.1016/j.ejmech.2024.117058] [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: 08/28/2024] [Revised: 11/03/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024]
Abstract
Chalcones have the characteristics of simple structure, easy synthesis and potent anti-tumor activity. Herein, a small library of fifty-five novel indole-chalcone derivatives were rationally designed and facilely synthesized. Consequently, their antiproliferative activity was systematically evaluated. Among which, compound 26 exhibited the most potent antiproliferative activity, with IC50 value of 0.764 μM against MD-MBA-231 cells. Moreover, it displayed a 5-fold selectivity compared with normal human cells. Further investigation revealed that compound 26 bound at the colchicine binding site of tubulin, disrupted their fibrous structure, thereby blocking the progression of the cell cycle and inducing apoptosis. Molecular docking and cellular thermal shift assay (CETSA) experiments further demonstrated that compound 26 could specifically bind to hexokinase 2 (HK2) and inhibit its activity, leading to impaired mitochondrial function and hindered mitochondrial respiration. Based on the quantitative structure-activity relationship study, further structure modifications were performed. Employing biotin probe pull-down assays, we demonstrated that compound 26 exerted its antiproliferative activity through a dual targeting mechanism, which simultaneously disrupted microtubule function and inhibited HK2 activity. Taken together, these results highlighted that compound 26 might be a promising antiproliferative agent for human cancer therapy.
Collapse
Affiliation(s)
- Mengzhu Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Guangyuan Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Yawei Han
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Pengyu Qian
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Mingze Wu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Ming Xiang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China.
| | - Yirong Zhou
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
3
|
Chowdhary S, Preeti, Shekhar, Gupta N, Kumar R, Kumar V. Advances in chalcone-based anticancer therapy: mechanisms, preclinical advances, and future perspectives. Expert Opin Drug Discov 2024; 19:1417-1437. [PMID: 39621431 DOI: 10.1080/17460441.2024.2436908] [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: 06/17/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
INTRODUCTION Cancer remains a leading cause of death worldwide with traditional treatments like chemotherapy, and radiotherapy becoming less effective due to multidrug resistance (MDR). This highlights the necessity for novel chemotherapeutics like chalcone-based compounds, which demonstrate broad anti-cancer properties and target multiple pathways. These compounds hold promise for improving cancer treatment outcomes compared to existing therapies. AREAS COVERED This review provides a comprehensive synopsis of the recent literature (2018-2024) for anti-proliferative/anti-cancer activity of chalcones. It includes the identification of potential targets, their mechanisms of action, and possible modes of binding. Additionally, chalcone derivatives in preclinical trials are also discussed. EXPERT OPINION Chalcones mark a significant stride in anticancer therapies due to their multifaceted approach in targeting various cellular pathways. Their ability to simultaneously target multiple pathways enables them to overcome drug resistance as compared to traditional therapies. With well-defined mechanisms of action, these compounds can serve as lead molecules for designing new, more promising treatments. Continued progress in synthesis and structural optimization, along with promising results from preclinical trials, offers hope for the development of more potent molecules, heralding a new era in cancer therapeutics.
Collapse
Affiliation(s)
| | - Preeti
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Shekhar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Nikita Gupta
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Rajesh Kumar
- Department of Physics, Lovely Professional University, Phagwara, India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
4
|
Zhang YL, Sun SJ, Zeng L. Biological effects and mechanisms of dietary chalcones: latest research progress, future research strategies, and challenges. Food Funct 2024; 15:10582-10599. [PMID: 39392421 DOI: 10.1039/d4fo03618b] [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: 10/12/2024]
Abstract
Dietary plants are an indispensable part of the human diet, and the various natural active compounds they contain, especially polyphenols, polysaccharides, and amino acids, have always been a hot topic of research among nutritionists. As precursors to polyphenolic substances in dietary plants, chalcones are not only widely distributed but also possess a variety of biological activities due to their unique structure. However, there has not yet been a comprehensive article summarizing the biological activities and mechanisms of dietary chalcones. This review began by discussing the dietary sources and bioavailability of chalcones, providing a comprehensive description of their biological activities and mechanisms of action in antioxidation, anti-inflammation, anti-tumor, and resistance to pathogenic microbes. Additionally, based on the latest research findings, some future research strategies and challenges for dietary chalcones have been proposed, including computer-aided design and molecular docking, targeted biosynthesis and derivative design, interactions between the gut microbiota and chalcones, as well as clinical research. It is expected that this review will contribute to supplementing the scientific understanding of dietary chalcones and promoting their practical application and the development of new food products.
Collapse
Affiliation(s)
- Yun Liang Zhang
- Department of Pharmacy, Shaoyang University, Shaoyang, Hunan 422000, China.
- Southwest Hunan Research Center of Engineering for Development and Utilization of Traditional Chinese Medicine, School of Pharmacy, Shaoyang University, Shaoyang, Hunan 422000, China
| | - Shuang Jiao Sun
- Department of Pharmacy, Shaoyang University, Shaoyang, Hunan 422000, China.
- Southwest Hunan Research Center of Engineering for Development and Utilization of Traditional Chinese Medicine, School of Pharmacy, Shaoyang University, Shaoyang, Hunan 422000, China
| | - Li Zeng
- Department of Pharmacy, Shaoyang University, Shaoyang, Hunan 422000, China.
- Southwest Hunan Research Center of Engineering for Development and Utilization of Traditional Chinese Medicine, School of Pharmacy, Shaoyang University, Shaoyang, Hunan 422000, China
| |
Collapse
|
5
|
Wang H, Zhu J, Zhang Q, Tang J, Huang X. Current scenario of chalcone hybrids with antibreast cancer therapeutic applications. Arch Pharm (Weinheim) 2024; 357:e2300640. [PMID: 38227398 DOI: 10.1002/ardp.202300640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Breast cancer, an epithelial malignant tumor that occurs in the terminal ducts of the breast, is the most common female malignancy. Currently, approximately 70%-80% of breast cancer with early-stage, nonmetastatic disorder is curable, but the emergency of drug resistance often leads to treatment failure. Moreover, advanced breast cancer with distant organ metastases is incurable with the available therapeutics, creating an urgent demand to explore novel antibreast cancer agents. Chalcones, the precursors for flavonoids and isoflavonoids, exhibit promising activity against various breast cancer hallmarks, inclusive of proliferation, angiogenesis, invasion, metastasis, inflammation, stemness, and regulation of cancer epigenetics, representing useful scaffolds for the discovery of novel antibreast cancer chemotherapeutic candidates. In particular, chalcone hybrids could act on two or more different biological targets simultaneously with more efficacy, lower toxicity, and less susceptibility to resistance. Accordingly, there is a huge scope for application of chalcone hybrids to tackle the present difficulties in breast cancer therapy. This review outlines the chalcone hybrids with antibreast cancer potential developed from 2018. The structure-activity relationships as well as mechanisms of action are also discussed to shed light on the development of more effective and multitargeted chalcone candidates.
Collapse
Affiliation(s)
- Huan Wang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Juanying Zhu
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Qianru Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Tang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Yakkala PA, Rahaman S, Soukya PSL, Begum SA, Kamal A. An update on the development on tubulin inhibitors for the treatment of solid tumors. Expert Opin Ther Targets 2024; 28:193-220. [PMID: 38618889 DOI: 10.1080/14728222.2024.2341630] [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: 12/29/2023] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Microtubules play a vital role in cancer therapeutics. They are implicated in tumorigenesis, thus inhibiting tubulin polymerization in cancer cells, and have now become a significant target for anticancer drug development. A plethora of drug molecules has been crafted to influence microtubule dynamics and presently, numerous tubulin inhibitors are being investigated. This review discusses the recently developed inhibitors including natural products, and also examines the preclinical and clinical data of some potential molecules. AREA COVERED The current review article summarizes the development of tubulin inhibitors while detailing their specific binding sites. It also discusses the newly designed inhibitors that may be useful in the treatment of solid tumors. EXPERT OPINION Microtubules play a crucial role in cellular processes, especially in cancer therapy where inhibiting tubulin polymerization holds promise. Ongoing trials signify a commitment to revolutionizing cancer treatment and exploring targeted therapies. Challenges in microtubule modulation, like resistance and off-target effects, demand focused efforts, emphasizing combination therapies and personalized treatments. Beyond microtubules, promising avenues in cancer research include immunotherapy, genomic medicine, CRISPR gene editing, liquid biopsies, AI diagnostics, and stem cell therapy, showcasing a holistic approach for future advancements.
Collapse
Affiliation(s)
- Prasanna Anjaneyulu Yakkala
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shaik Rahaman
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - P S Lakshmi Soukya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad, India
| | - Sajeli Ahil Begum
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad, India
| | - Ahmed Kamal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad, India
- Department of Environment, Forests, Science & Technology, Telangana State Council of Science & Technology, Hyderabad, India
| |
Collapse
|
7
|
Abouelenein MG, El-Rashedy AA, Awad HM, El Farargy AF, Nassar IF, Nassrallah A. Synthesis, molecular modeling Insights, and anticancer assessment of novel polyfunctionalized Pyridine congeners. Bioorg Chem 2023; 141:106910. [PMID: 37871393 DOI: 10.1016/j.bioorg.2023.106910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023]
Abstract
The present study describes synthesizing a novel series of polyfunctionalized pyridine congeners 1-18 and assessed for cytotoxic efficacies versus HCT-116, MCF-7, and HepG-2 among one non-cancerous BJ-1 human normal cell. Most compounds were precisely potent anticancer candidate drugs. The molecular impact of the most active compounds 9, 10, 11, 13, 15, and 17 was evaluated after MCF-7 treatment. The gene expression of pro- and ant-apoptosis markers P53, Bax, Caspase-3 and Bcl-2 as well as VEGFR-2 and HER2 were determined. Compounds 13 and 15 induced upregulation of pro-apoptosis of P53, Bax, Caspase-3 and downregulation of anti-apoptosis Bcl-2 gene. However, compound 15 showed higher effect compared to 13 and respective control. Moreover, a slight reduction in HER2 gene expression was detected due to compound 15 treatment, while VEGFR-2 gene was upregulated. In agreement, the immunoblotting analysis showed higher accumulation of P53, Bax, Caspase-3 proteins and of decrease the Bcl-2 protein levels. Furthermore, docking studies united with molecular dynamic simulation exposed compounds 13 and 15 fitting in the middle of the active site at the interface linking the ATP binding site and the allosteric hydrophobic binding pocket. Finally, we performed Petra/Osiris/ Molinspiration (POM) analysis for the newly synthesized compounds. The evaluation of primary in silico parameters revealed significant differences among individual polyfunctionalized pyridine compounds, highlighting the most promising candidates. These preliminary results may help in coordinating and initiating other research projects focused on polyfunctionalized pyridine compounds, especially those with predicted bioactivity, low toxicity, optimal ADME parameters, and promising perspectives.
Collapse
Affiliation(s)
- Mohamed G Abouelenein
- Chemistry Department, Faculty of Science, Menofia University, Shebin El-Koam, Menofia, Egypt.
| | - Ahmed A El-Rashedy
- Natural and Microbial Products Department, National Research Center (NRC), Egypt
| | - Hanem M Awad
- Department of Tanning Materials and Leather Technology, Chemical Industries Research Institute, National Research Centre (NRC), Egypt
| | - Ahmed F El Farargy
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Ibrahim F Nassar
- Faculty of Specific Education, Ain Shams University, Abassia, Cairo, Egypt
| | - Amr Nassrallah
- Basic Applied Science Institute, Egypt-Japan University of Science and Technology (E-JUST) P.O. Box 179, New Borg El-Arab City Postal Code 21934, Alexandria, Egypt; Biochemistry Department, Faculty of Agriculture, Cairo University, 12613 Giza, Egypt
| |
Collapse
|
8
|
Meng HW, Shen ZB, Meng XS, Leng-Wei, Yin ZQ, Wang XR, Zou TF, Liu ZG, Wang TX, Zhang S, Chen YL, Yang XX, Li QS, Duan YJ. Novel flavonoid 1,3,4-oxadiazole derivatives ameliorate MPTP-induced Parkinson's disease via Nrf2/NF-κB signaling pathway. Bioorg Chem 2023; 138:106654. [PMID: 37300959 DOI: 10.1016/j.bioorg.2023.106654] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/20/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder with a complex etiology. Neuroinflammation and oxidative stress are important factors driving the progression of PD. It has been reported that 1,3,4-oxadiazole and flavone derivatives have numerous biological functions, especially in the aspect of anti-inflammatory and antioxidant. Based on the strategy of pharmacodynamic combination, we introduced 1,3,4-oxadiazole moiety into the flavonoid backbone, designed and synthesized a series of novel flavonoid 1,3,4-oxadiazole derivatives. Further, we evaluated their toxicity, anti-inflammatory and antioxidant activities using BV2 microglia. Following a comprehensive analysis, compound F12 showed the best pharmacological activity. In vivo, we induced the classical PD animal model by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) into C57/BL6J mice. Our results showed that compound F12 ameliorated MPTP-induced dysfunction in mice. Further, compound F12 reduced oxidative stress by promoting the nucleation of nuclear factor erythroid 2-related factor 2 (Nrf2) and decreased the inflammatory response by inhibiting the nuclear translocation of nuclear factor-κB (NF-κB) in vivo and in vitro. Meanwhile, compound F12 inhibited the mitochondrial apoptotic pathway to rescue microglia inflammation-mediated loss of dopaminergic neurons. In conclusion, compound F12 reduced oxidative stress and inflammation and could be as a potential agent for PD treatment.
Collapse
Affiliation(s)
- Hua-Wen Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhen-Bao Shen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xian-She Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Leng-Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ze-Qun Yin
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xue-Rui Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ting-Feng Zou
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhi-Gang Liu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tian-Xiang Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuan-Li Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiao-Xiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Qing-Shan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Ya-Jun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China; Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
9
|
Chalcones: Promising therapeutic agents targeting key players and signaling pathways regulating the hallmarks of cancer. Chem Biol Interact 2023; 369:110297. [PMID: 36496109 DOI: 10.1016/j.cbi.2022.110297] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The need for innovative anticancer treatments with high effectiveness and low toxicity is urgent due to the development of malignancies that are resistant to chemotherapeutic agents and the poor specificity of existing anticancer treatments. Chalcones are 1,3-diaryl-2-propen-1-ones, which are the precursors for flavonoids and isoflavonoids. Chalcones are readily available from a wide range of natural resources and consist of very basic chemical scaffolds. Because the ease with which the synthesis it allows for the production of several chalcone derivatives. Various in-vitro and in-vivo studies indicate that naturally occurring and synthetic chalcone derivatives exhibit promising biological activities against cancer hallmarks such as proliferation, angiogenesis, invasion, metastasis, inflammation, stemness, and regulation of cancer epigenetics. According to their structure and functional groups, chalcones derivatives and their hybrid compounds exert a broad range of biological activities through targeting key elements and signaling molecules relevant to cancer progression. This review will provide valuable insights into the latest updates of chalcone groups as anticancer agents and extensively discuss their underlying molecular mechanisms of action.
Collapse
|
10
|
An update on the recent advances and discovery of novel tubulin colchicine binding inhibitors. Future Med Chem 2023; 15:73-95. [PMID: 36756851 DOI: 10.4155/fmc-2022-0212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Microtubules, formed by α- and β-tubulin heterodimer, are considered as a major target to prevent the proliferation of tumor cells. Microtubule-targeted agents have become increasingly effective anticancer drugs. However, due to the relatively sophisticated chemical structure of taxane and vinblastine, their application has faced numerous obstacles. Conversely, the structure of colchicine binding site inhibitors (CBSIs) is much easier to be modified. Moreover, CBSIs have strong antiproliferative effect on multidrug-resistant tumor cells and have become the mainstream research orientation of microtubule-targeted agents. This review focuses mainly on the recent advances of CBSIs during 2017-2022, attempts to depict their biological activities to analyze the structure-activity relationships and offers new perspectives for designing next generation of novel CBSIs.
Collapse
|
11
|
Theoretical and molecular mechanistic investigations of novel (3-(furan-2-yl)pyrazol-4-yl) chalcones against lung carcinoma cell line (A549). NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:719-736. [PMID: 36469109 PMCID: PMC10042774 DOI: 10.1007/s00210-022-02344-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/16/2022] [Indexed: 12/07/2022]
Abstract
AbstractA new chalcone series has been developed that may be useful in the treatment of lung cancer. The new series was confirmed by the different spectral tools. MTT assay was used to detect the cytotoxic effect of the novel chalcones against lung cancer cell line (A549). Molecular docking studies were performed on the most two effective chalcones 7b and 7c. Different molecular techniques were utilized to study the activity and the effect of two chalcones 7b and 7c on apoptosis of A549 cell line.
Collapse
|
12
|
Rajendran G, Bhanu D, Aruchamy B, Ramani P, Pandurangan N, Bobba KN, Oh EJ, Chung HY, Gangadaran P, Ahn BC. Chalcone: A Promising Bioactive Scaffold in Medicinal Chemistry. Pharmaceuticals (Basel) 2022; 15:1250. [PMID: 36297362 PMCID: PMC9607481 DOI: 10.3390/ph15101250] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chalcones are a class of privileged scaffolds with high medicinal significance due to the presence of an α,β-unsaturated ketone functionality. Numerous functional modifications of chalcones have been reported, along with their pharmacological behavior. The present review aims to summarize the structures from natural sources, synthesis methods, biological characteristics against infectious and non-infectious diseases, and uses of chalcones over the past decade, and their structure-activity relationship studies are detailed in depth. This critical review provides guidelines for the future design and synthesis of various chalcones. In addition, this could be highly supportive for medicinal chemists to develop more promising candidates for various infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Gayathri Rajendran
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Deepu Bhanu
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Baladhandapani Aruchamy
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Prasanna Ramani
- Dhanvanthri Laboratory, Department of Sciences, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
- Center of Excellence in Advanced Materials & Green Technologies (CoE–AMGT), Amrita School of Engineering, Amrita Vishwa Vidyapeetham, Coimbatore 641112, India
| | - Nanjan Pandurangan
- Department of Sciences, Amrita School of Arts and Sciences, Mysuru Campus, Amrita Vishwa Vidyapeetham, Mysuru 570026, India
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California (San Francisco), San Francisco, CA 94143, USA
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| |
Collapse
|
13
|
Synthesis and Spectroscopic Characterization of Nicotinaldehyde Based Derivatives: SC-XRD, Linear and Non-Linear Optical Studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
14
|
Patil SV, Gejji SP, Malkhede DD. Design and synthesis of piezochromic materials exploring intermolecular charge transfer: chalconoids bound to the p-sulfonatocalix[6]arene macrocycle. Phys Chem Chem Phys 2022; 24:17809-17823. [PMID: 35848925 DOI: 10.1039/d2cp01483a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Solid-state systems composed of chalconoid encapsulated within p-sulfonatocalix[6]arene (SCX6) scaffolds that exhibit mechanochromism and thermochromism have been developed. An introduction of a supramolecular host promises a variety of applications in diverse areas, which makes them fascinating. Largely hydrogen bonding as well as π···π interactions are responsible for the host-guest complexation. The complex shows partial encapsulation of the guest with one of the phenyl rings of chalcone (guest) is held inside the SCX6 cavity, whilst other phenyl rings that exclude the cavity are hydrogen-bonded to sulfonate portals of the host. The hydrogen bonding conducing such complexation triggers proton transfer engendering a mechanochromic switch. The complexes are further characterized by a variety of experiments such as cyclic voltammetry (CV), steady-state fluorescence, vibrational spectroscopy, and 1H or 2D NMR (NOESY) spectroscopy experiments. Detailed structure furnished through the NMR shows deshielding of the Ha-e (guest) protons whereas, the hydroxyl protons from the host experience shielding as evidenced from the 1H NMR spectra. These inferences have further been corroborated through the density functional theory. Electrochemical investigations suggested an irreversible one-electron transfer in the host-guest binding. The characteristic 'frequency shift' for the intense carbonyl vibration in the infrared spectra, which can be correlated to the kinetic energy density parameter, G(r), in the quantum theory of atoms in molecules (QTAIM).
Collapse
Affiliation(s)
- Sanhita V Patil
- Department of Chemistry, Savitribai Phule Pune University, Pune, 411007, India.
| | - Shridhar P Gejji
- Department of Chemistry, Savitribai Phule Pune University, Pune, 411007, India.
| | - Dipalee D Malkhede
- Department of Chemistry, Savitribai Phule Pune University, Pune, 411007, India.
| |
Collapse
|
15
|
Urbonavičius A, Fortunato G, Ambrazaitytė E, Plytninkienė E, Bieliauskas A, Milišiūnaitė V, Luisi R, Arbačiauskienė E, Krikštolaitytė S, Šačkus A. Synthesis and Characterization of Novel Heterocyclic Chalcones from 1-Phenyl-1 H-pyrazol-3-ol. Molecules 2022; 27:3752. [PMID: 35744875 PMCID: PMC9227189 DOI: 10.3390/molecules27123752] [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: 05/10/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
An efficient synthetic route to construct diverse pyrazole-based chalcones from 1-phenyl-1H-pyrazol-3-ols bearing a formyl or acetyl group on the C4 position of pyrazole ring, employing a base-catalysed Claisen-Schmidt condensation reaction, is described. Isomeric chalcones were further reacted with N-hydroxy-4-toluenesulfonamide and regioselective formation of 3,5-disubstituted 1,2-oxazoles was established. The novel pyrazole-chalcones and 1,2-oxazoles were characterized by an in-depth analysis of NMR spectral data, which were obtained through a combination of standard and advanced NMR spectroscopy techniques.
Collapse
Affiliation(s)
- Arminas Urbonavičius
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
- Institute of Synthetic Chemistry, Kaunas University of Technology, K. Baršausko g. 59, 51423 Kaunas, Lithuania;
| | - Graziana Fortunato
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, Via E. Orabona 4, 70125 Bari, Italy;
| | - Emilija Ambrazaitytė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
| | - Elena Plytninkienė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
- Institute of Synthetic Chemistry, Kaunas University of Technology, K. Baršausko g. 59, 51423 Kaunas, Lithuania;
| | - Aurimas Bieliauskas
- Institute of Synthetic Chemistry, Kaunas University of Technology, K. Baršausko g. 59, 51423 Kaunas, Lithuania;
| | - Vaida Milišiūnaitė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
- Institute of Synthetic Chemistry, Kaunas University of Technology, K. Baršausko g. 59, 51423 Kaunas, Lithuania;
| | - Renzo Luisi
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, Via E. Orabona 4, 70125 Bari, Italy;
| | - Eglė Arbačiauskienė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
| | - Sonata Krikštolaitytė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
| | - Algirdas Šačkus
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Pl. 19, 50254 Kaunas, Lithuania; (A.U.); (G.F.); (E.A.); (E.P.); (V.M.); (S.K.)
- Institute of Synthetic Chemistry, Kaunas University of Technology, K. Baršausko g. 59, 51423 Kaunas, Lithuania;
| |
Collapse
|
16
|
Tawfik HO, Shaldam MA, Nocentini A, Salem R, Almahli H, Al-Rashood ST, Supuran CT, Eldehna WM. Novel 3-(6-methylpyridin-2-yl)coumarin-based chalcones as selective inhibitors of cancer-related carbonic anhydrases IX and XII endowed with anti-proliferative activity. J Enzyme Inhib Med Chem 2022; 37:1043-1052. [PMID: 35437108 PMCID: PMC9037210 DOI: 10.1080/14756366.2022.2056734] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Carbonic anhydrases (CAs) are one of the promising targets for the development of anticancer agents. CA isoforms are implicated in various physiological processes and are expressed in both normal and cancerous cells. Thus, non-isoform selective inhibitors are associated with several side effects. Consequently, designing selective inhibitors towards cancer-related hCA IX/XII rather than the ubiquitous cytosolic isozymes hCA I and II is the main research objective in the field. Herein, a new series of 3-(6-methylpyridin-2-yl)coumarin derivatives 3 and 5a–o was designed and synthesised. The CA inhibition activities for the synthesised coumarins were analysed on isoforms hCA I, II, IX, and XII. Interestingly, both cancer-linked isoforms hCA IX/XII were inhibited by the prepared coumarins with inhibition constants ranging from sub- to low-micromolar range, whereas hCA I and II isoforms haven’t been inhibited up to 100 µM. Furthermore, the target coumarins were assessed for their antitumor activity on NCI-59 human cancer types.
Collapse
Affiliation(s)
- Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Alessio Nocentini
- Section of Pharmaceutical and Nutraceutical Sciences, Department of NEUROFARBA, University of Florence, Polo Scientifico, Firenze, Italy
| | - Rofaida Salem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Hadia Almahli
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Sara T Al-Rashood
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Claudiu T Supuran
- Section of Pharmaceutical and Nutraceutical Sciences, Department of NEUROFARBA, University of Florence, Polo Scientifico, Firenze, Italy
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
17
|
Ebenezer O, Shapi M, Tuszynski JA. A Review of the Recent Developments of Molecular Hybrids Targeting Tubulin Polymerization. Int J Mol Sci 2022; 23:4001. [PMID: 35409361 PMCID: PMC8999808 DOI: 10.3390/ijms23074001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
Microtubules are cylindrical protein polymers formed from αβ-tubulin heterodimers in the cytoplasm of eukaryotic cells. Microtubule disturbance may cause cell cycle arrest in the G2/M phase, and anomalous mitotic spindles will form. Microtubules are an important target for cancer drug action because of their critical role in mitosis. Several microtubule-targeting agents with vast therapeutic advantages have been developed, but they often lead to multidrug resistance and adverse side effects. Thus, single-target therapy has drawbacks in the effective control of tubulin polymerization. Molecular hybridization, based on the amalgamation of two or more pharmacophores of bioactive conjugates to engender a single molecular structure with enhanced pharmacokinetics and biological activity, compared to their parent molecules, has recently become a promising approach in drug development. The practical application of combined active scaffolds targeting tubulin polymerization inhibitors has been corroborated in the past few years. Meanwhile, different designs and syntheses of novel anti-tubulin hybrids have been broadly studied, illustrated, and detailed in the literature. This review describes various molecular hybrids with their reported structural-activity relationships (SARs) where it is possible in an effort to generate efficacious tubulin polymerization inhibitors. The aim is to create a platform on which new active scaffolds can be modeled for improved tubulin polymerization inhibitory potency and hence, the development of new therapeutic agents against cancer.
Collapse
Affiliation(s)
- Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Michael Shapi
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Jack A. Tuszynski
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada
- DIMEAS, Politecnico di Torino, 10129 Turin, Italy
| |
Collapse
|
18
|
Albratty M, Ahmad Alhazmi H. Novel pyridine and pyrimidine derivatives as promising anticancer agents: A review. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
19
|
Hashem HE, Nath A, Kumer A. Synthesis, molecular docking, molecular dynamic, quantum calculation, and antibacterial activity of new Schiff base-metal complexes. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
20
|
Ramesh D, Sarkar D, Joji A, Singh M, Mohanty AK, G Vijayakumar B, Chatterjee M, Sriram D, Muthuvel SK, Kannan T. First-in-class pyrido[2,3-d]pyrimidine-2,4(1H,3H)-diones against leishmaniasis and tuberculosis: Rationale, in vitro, ex vivo studies and mechanistic insights. Arch Pharm (Weinheim) 2022; 355:e2100440. [PMID: 35106845 DOI: 10.1002/ardp.202100440] [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: 11/08/2021] [Revised: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 11/06/2022]
Abstract
Pyrido[2,3-d]pyrimidine-2,4(1H,3H)-diones were synthesized, for the first time, from indole chalcones and 6-aminouracil, and their ability to inhibit leishmaniasis and tuberculosis (Tb) infections was evaluated. The in vitro antileishmanial activity against promastigotes of Leishmania donovani revealed exceptional activities of compounds 3, 12 and 13, with IC50 values ranging from 10.23 ± 1.50 to 15.58 ± 1.67 µg/ml, which is better than the IC50 value of the standard drug pentostam of 500 μg/ml. The selectivity of the compounds towards Leishmania parasites was evaluated via ex vivo studies in Swiss albino mice. The efficiency of these compounds against Tb infection was then evaluated using the in vitro anti-Tb microplate Alamar Blue assay. Five compounds, 3, 7, 8, 9 and 12, showed MIC100 values against the Mycobacterium tuberculosis H37 Rv strain at 25 µg/ml, and compound 20 yielded an MIC100 value of 50 µg/ml. Molecular modelling of these compounds highlighted interactions with binding sites of dihydrofolate reductase, pteridine reductase and thymidylate kinase, thus establishing the rationale of their pharmacological activity against both pathogens, which is consistent with the in vitro results. From the above results, it is clear that compounds 3 and 12 are promising lead candidates for Leishmania and Mycobacterium infections and may be promising for coinfections.
Collapse
Affiliation(s)
- Deepthi Ramesh
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Deblina Sarkar
- Department of Pharmacology, Institute of Post Graduate Medical Education & Research (IPGME&R), Kolkata, West Bengal, India
| | - Annu Joji
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Monica Singh
- Department of Pharmacy, Birla Institute of Technology & Science Pilani, Hyderabad, India
| | - Amaresh K Mohanty
- Department of Bioinformatics, Pondicherry University, Kalapet, Puducherry, India
| | | | - Mitali Chatterjee
- Department of Pharmacology, Institute of Post Graduate Medical Education & Research (IPGME&R), Kolkata, West Bengal, India
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology & Science Pilani, Hyderabad, India
| | - Suresh K Muthuvel
- Department of Bioinformatics, Pondicherry University, Kalapet, Puducherry, India
| | | |
Collapse
|
21
|
Liu W, He M, Li Y, Peng Z, Wang G. A review on synthetic chalcone derivatives as tubulin polymerisation inhibitors. J Enzyme Inhib Med Chem 2021; 37:9-38. [PMID: 34894980 PMCID: PMC8667932 DOI: 10.1080/14756366.2021.1976772] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Microtubules play an important role in the process of cell mitosis and can form a spindle in the mitotic prophase of the cell, which can pull chromosomes to the ends of the cell and then divide into two daughter cells to complete the process of mitosis. Tubulin inhibitors suppress cell proliferation by inhibiting microtubule dynamics and disrupting microtubule homeostasis. Thereby inducing a cell cycle arrest at the G2/M phase and interfering with the mitotic process. It has been found that a variety of chalcone derivatives can bind to microtubule proteins and disrupt the dynamic balance of microtubules, inhibit the proliferation of tumour cells, and exert anti-tumour effects. Consequently, a great number of studies have been conducted on chalcone derivatives targeting microtubule proteins. In this review, synthetic or natural chalcone microtubule inhibitors in recent years are described, along with their structure-activity relationship (SAR) for anticancer activity.
Collapse
Affiliation(s)
- Wenjing Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China.,Teaching and Research Section of Natural Medicinal Chemistry, School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Min He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China.,Teaching and Research Section of Natural Medicinal Chemistry, School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yongjun Li
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Zhiyun Peng
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Guangcheng Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| |
Collapse
|
22
|
Lin X, Zeng C, Pang X, Zhu C, Liu C, Yang X, Hu Y, Fang Z, Guo K. Electrochemical Synthesis of Selenium‐substituted Benzoxazine
via
Radical Initiated Cyclization. ChemElectroChem 2021. [DOI: 10.1002/celc.202101013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Xinxin Lin
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
| | - Cuilian Zeng
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
| | - Xinyan Pang
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
| | - Chenlong Zhu
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
| | - Chengkou Liu
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
| | - Xiaobing Yang
- Department: China Biology and Medicine Department Institution: Jiangsu Industrial Technology Research Institute Nanjing 210031 P.R. China
| | - Yujing Hu
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
| | - Zheng Fang
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
- Department: State Key Laboratory of Materials-Oriented Chemical Engineering Institution: Nanjing Tech University Address 2 30 Puzhu Rd S. Nanjing 211816 China
| | - Kai Guo
- Department: College of Biotechnology and Pharmaceutical Engineering Institution: Nanjing Tech University 30 Puzhu Rd S. Nanjing 211816 China
- Department: State Key Laboratory of Materials-Oriented Chemical Engineering Institution: Nanjing Tech University Address 2 30 Puzhu Rd S. Nanjing 211816 China
| |
Collapse
|
23
|
Constantinescu T, Lungu CN. Anticancer Activity of Natural and Synthetic Chalcones. Int J Mol Sci 2021; 22:11306. [PMID: 34768736 PMCID: PMC8582663 DOI: 10.3390/ijms222111306] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022] Open
Abstract
Cancer is a condition caused by many mechanisms (genetic, immune, oxidation, and inflammatory). Anticancer therapy aims to destroy or stop the growth of cancer cells. Resistance to treatment is theleading cause of the inefficiency of current standard therapies. Targeted therapies are the most effective due to the low number of side effects and low resistance. Among the small molecule natural compounds, flavonoids are of particular interest for theidentification of new anticancer agents. Chalcones are precursors to all flavonoids and have many biological activities. The anticancer activity of chalcones is due to the ability of these compounds to act on many targets. Natural chalcones, such as licochalcones, xanthohumol (XN), panduretin (PA), and loncocarpine, have been extensively studied and modulated. Modification of the basic structure of chalcones in order to obtain compounds with superior cytotoxic properties has been performed by modulating the aromatic residues, replacing aromatic residues with heterocycles, and obtaining hybrid molecules. A huge number of chalcone derivatives with residues such as diaryl ether, sulfonamide, and amine have been obtained, their presence being favorable for anticancer activity. Modification of the amino group in the structure of aminochalconesis always favorable for antitumor activity. This is why hybrid molecules of chalcones with different nitrogen hetercycles in the molecule have been obtained. From these, azoles (imidazole, oxazoles, tetrazoles, thiazoles, 1,2,3-triazoles, and 1,2,4-triazoles) are of particular importance for the identification of new anticancer agents.
Collapse
Affiliation(s)
- Teodora Constantinescu
- Department of Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University, 400012 Cluj-Napoca, Romania
| | - Claudiu N. Lungu
- Department of Surgery, Country Emergency Hospital Braila, 810249 Braila, Romania
| |
Collapse
|
24
|
Aguilar-Morales CM, Araujo-Huitrado JG, López-Hernández Y, Contreras-Celedón C, Islas-Jácome A, Granados-López AJ, Solorio-Alvarado CR, López JA, Chacón-García L, Cortés-García CJ. A One-Pot Six-Component Reaction for the Synthesis of 1,5-Disubstituted Tetrazol-1,2,3-Triazole Hybrids and Their Cytotoxic Activity against the MCF-7 Cell Line. Molecules 2021; 26:6104. [PMID: 34684686 PMCID: PMC8541533 DOI: 10.3390/molecules26206104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/06/2021] [Accepted: 10/06/2021] [Indexed: 11/24/2022] Open
Abstract
A high-order multicomponent reaction involving a six-component reaction to obtain the novel linked 1,5-disubstituted tetrazole-1,2,3-triazole hybrids in low to moderate yield is described. This one-pot reaction is carried out under a cascade process consisting of three sequential reactions: Ugi-azide, bimolecular nucleophilic substitution (SN2), and copper-catalyzed alkyne-azide reaction (CuAAC), with high atom and step-economy due the formation of six new bonds (one C-C, four C-N, and one N-N). Thus, the protocol developed offers operational simplicity, mild reaction conditions, and structural diversity. Finally, to evaluate the antitumoral potential of the synthetized molecules, a proliferation study was performed in the breast cancer (BC) derived cell line MCF-7. The hybrid compounds showed several degrees of cell proliferation inhibition with a remarkable effect in those compounds with cyclohexane and halogens in their structures. These compounds represent potential drug candidates for breast cancer treatment. However, additionally assays are needed to elucidate their complete effect over the cellular hallmarks of cancer.
Collapse
Affiliation(s)
- Cesia M. Aguilar-Morales
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, Morelia C.P. 58033, Mexico; (C.M.A.-M.); (C.C.-C.)
| | - Jorge Gustavo Araujo-Huitrado
- Laboratorio de microRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas C.P. 98066, Mexico; (J.G.A.-H.); (A.J.G.-L.); (J.A.L.)
| | - Yamilé López-Hernández
- Laboratorio de Metabolómica y Proteómica, CONACyT-Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas C.P. 98066, Mexico;
| | - Claudia Contreras-Celedón
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, Morelia C.P. 58033, Mexico; (C.M.A.-M.); (C.C.-C.)
| | - Alejandro Islas-Jácome
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Ciudad de Mexico C.P. 09340, Mexico;
| | - Angelica Judith Granados-López
- Laboratorio de microRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas C.P. 98066, Mexico; (J.G.A.-H.); (A.J.G.-L.); (J.A.L.)
| | - Cesar R. Solorio-Alvarado
- Departamento de Química, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Campus Guanajuato, Noria Alta S/N, Guanajuato C.P. 36050, Mexico;
| | - Jesús Adrián López
- Laboratorio de microRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas C.P. 98066, Mexico; (J.G.A.-H.); (A.J.G.-L.); (J.A.L.)
| | - Luis Chacón-García
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, Morelia C.P. 58033, Mexico; (C.M.A.-M.); (C.C.-C.)
| | - Carlos J. Cortés-García
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, Morelia C.P. 58033, Mexico; (C.M.A.-M.); (C.C.-C.)
| |
Collapse
|
25
|
Ouyang Y, Li J, Chen X, Fu X, Sun S, Wu Q. Chalcone Derivatives: Role in Anticancer Therapy. Biomolecules 2021; 11:894. [PMID: 34208562 PMCID: PMC8234180 DOI: 10.3390/biom11060894] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
Chalcones (1,3-diaryl-2-propen-1-ones) are precursors for flavonoids and isoflavonoids, which are common simple chemical scaffolds found in many naturally occurring compounds. Many chalcone derivatives were also prepared due to their convenient synthesis. Chalcones as weandhetic analogues have attracted much interest due to their broad biological activities with clinical potentials against various diseases, particularly for antitumor activity. The chalcone family has demonstrated potential in vitro and in vivo activity against cancers via multiple mechanisms, including cell cycle disruption, autophagy regulation, apoptosis induction, and immunomodulatory and inflammatory mediators. It represents a promising strategy to develop chalcones as novel anticancer agents. In addition, the combination of chalcones and other therapies is expected to be an effective way to improve anticancer therapeutic efficacy. However, despite the encouraging results for their response to cancers observed in clinical studies, a full description of toxicity is required for their clinical use as safe drugs for the treatment of cancer. In this review, we will summarize the recent advances of the chalcone family as potential anticancer agents and the mechanisms of action. Besides, future applications and scope of the chalcone family toward the treatment and prevention of cancer are brought out.
Collapse
Affiliation(s)
- Yang Ouyang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.O.); (J.L.); (X.C.); (X.F.)
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.O.); (J.L.); (X.C.); (X.F.)
| | - Xinyue Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.O.); (J.L.); (X.C.); (X.F.)
| | - Xiaoyu Fu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.O.); (J.L.); (X.C.); (X.F.)
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Y.O.); (J.L.); (X.C.); (X.F.)
| |
Collapse
|
26
|
Rioux B, Pinon A, Gamond A, Martin F, Laurent A, Champavier Y, Barette C, Liagre B, Fagnère C, Sol V, Pouget C. Synthesis and biological evaluation of chalcone-polyamine conjugates as novel vectorized agents in colorectal and prostate cancer chemotherapy. Eur J Med Chem 2021; 222:113586. [PMID: 34116328 DOI: 10.1016/j.ejmech.2021.113586] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 12/27/2022]
Abstract
The aim of this study was to synthesize chalcone-polyamine conjugates in order to enhance bioavailability and selectivity of chalcone core towards cancer cells, using polyamine-based vectors. Indeed, it is well-known that polyamine transport system is upregulated in tumor cells. 3',4,4',5'-tetramethoxychalcone was selected as parent chalcone since it was found to be an efficient anti-proliferative agent on various cancer cells. A series of five chalcone-polyamine conjugates was obtained using the 4-bromopropyloxy-3',4',5'-trimethoxychalcone as a key intermediate. Chalcone core and polyamine tails were fused through an amine bond. These conjugates were found to possess a marked in vitro antiproliferative effect against colorectal (HT-29 and HCT-116) and prostate cancer (PC-3 and DU-145) cell lines. The most active conjugate (compound 8b) was then chosen for further biological evaluations to elucidate mechanisms responsible for its antiproliferative activity. Investigations on cell cycle distribution revealed that this conjugate can prevent the proliferation of human colorectal and prostate cancer cells by blocking the cell cycle at the G1 and G2 phase, respectively. Flow cytometry analysis revealed a sub-G1 peak, characteristic of apoptotic cell population and our inquiries highlighted apoptosis induction at early and later stages through several pro-apoptotic markers. Therefore, this chalcone-N1-spermidine conjugate could be considered as a promising agent for colon and prostatic cancer adjuvant therapy.
Collapse
Affiliation(s)
- Benjamin Rioux
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Aline Pinon
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Aurélie Gamond
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Frédérique Martin
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Aurélie Laurent
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Yves Champavier
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France; Université de Limoges, BISCEm NMR Platform, GEIST, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Caroline Barette
- Université Grenoble Alpes, CEA, INSERM, IRIG, BGE U1038, Genetics & Chemogenomics, 17 Avenue des Martyrs, Grenoble, 38054, France
| | - Bertrand Liagre
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Catherine Fagnère
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Vincent Sol
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France
| | - Christelle Pouget
- Université de Limoges, Laboratoire PEIRENE EA 7500, Faculté de Pharmacie, 2 Rue Du Dr Marcland, 87025, Limoges Cedex, France.
| |
Collapse
|
27
|
Rahimzadeh Oskuei S, Mirzaei S, Reza Jafari-Nik M, Hadizadeh F, Eisvand F, Mosaffa F, Ghodsi R. Design, synthesis and biological evaluation of novel imidazole-chalcone derivatives as potential anticancer agents and tubulin polymerization inhibitors. Bioorg Chem 2021; 112:104904. [PMID: 33933802 DOI: 10.1016/j.bioorg.2021.104904] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 12/30/2022]
Abstract
Novel imidazole-chalcone derivatives were designed and synthesized as tubulin polymerization inhibitors and anticancer agents. The antiproliferative activity of the imidazole-chalcone was assessed on some human cancer cell lines including A549 (adenocarcinoma human alveolar basal epithelial cells), MCF-7 (human breast cancer cells), MCF-7/MX (mitoxantrone resistant human breast cancer cells), and HEPG2 (human hepatocellular carcinoma cells). Generally, the imidazole-chalcone derivatives exhibited more cytotoxicity on A549 cancer cells in comparison to the other three cell lines, among them compounds 9j' and 9g showed significant cytotoxicity with IC50 values ranging from 7.05 to 63.43 μM against all the four human cancer cells. The flow cytometry analysis of A549 cancer cells treated with 9g and 9j' displayed that these compounds induced cell cycle arrest at the G2/M phase at low concentrations and increased the number of apoptotic cells (cells in subG1 phase) at higher concentrations. They have also inhibited tubulin polymerization similar to combretastatin A-4 (CA-4). Annexin V binding staining assay in A549 cancer cells revealed that compound 9j' induced apoptosis (early and late). Finally, molecular docking studies of 9j' into the colchicine-binding site of tubulin presented the probable interactions of these compounds with tubulin.
Collapse
Affiliation(s)
- Sara Rahimzadeh Oskuei
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Salimeh Mirzaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Reza Jafari-Nik
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhad Eisvand
- Department of Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
28
|
Khan E. Pyridine Derivatives as Biologically Active Precursors; Organics and Selected Coordination Complexes. ChemistrySelect 2021. [DOI: 10.1002/slct.202100332] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ezzat Khan
- Department of Chemistry University of Malakand, Chakdara 18800, Lower Dir Khyber Pakhtunkhwa Pakistan
- Department of Chemistry, College of Science University of Bahrain Sakhir 32038 Bahrain
| |
Collapse
|
29
|
Dandawate P, Ahmed K, Padhye S, Ahmad A, Biersack B. Anticancer Active Heterocyclic Chalcones: Recent Developments. Anticancer Agents Med Chem 2021; 21:558-566. [PMID: 32628595 DOI: 10.2174/1871520620666200705215722] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/27/2020] [Accepted: 04/07/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chalcones are structurally simple compounds that are easily accessible by synthetic methods. Heterocyclic chalcones have gained the interest of scientists due to their diverse biological activities. The anti-tumor activities of heterocyclic chalcones are especially remarkable and the growing number of publications dealing with this topic warrants an up-to-date compilation. METHODS Search for antitumor active heterocyclic chalcones was carried out using Pubmed and Scifinder as common web-based literature searching tools. Pertinent and current literature was covered from 2015/2016 to 2019. Chemical structures, biological activities and modes of action of anti-tumor active heterocyclic chalcones are summarized. RESULTS Simply prepared chalcones have emerged over the last years with promising antitumor activities. Among them, there are a considerable number of tubulin polymerization inhibitors. But there are also new chalcones targeting special enzymes such as histone deacetylases or with DNA-binding properties. CONCLUSION This review provides a summary of recent heterocyclic chalcone derivatives with distinct antitumor activities.
Collapse
Affiliation(s)
- Prasad Dandawate
- Interdisciplinary Science and Technology Research Academy, Abeda Inamdar Senior College, University of Pune, 2390-B, K.B. Hidayatullah Road, Pune 411001, India
| | - Khursheed Ahmed
- Department of Chemistry, Abeda Inamdar Senior College, University of Pune, 2390-B, K.B. Hidayatullah Road, Pune 411001, India
| | - Subhash Padhye
- Interdisciplinary Science and Technology Research Academy, Abeda Inamdar Senior College, University of Pune, 2390-B, K.B. Hidayatullah Road, Pune 411001, India
| | - Aamir Ahmad
- University of Alabama at Birmingham, 9th Ave South, Birmingham AL 33294, United States
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| |
Collapse
|
30
|
Hou H, Sun Y, Pan Y, Yu H, Han Y, Shi Y, Yan C, Zhu S. Visible-Light Mediated Diarylselenylative Cyclization of 1,6-Enynes. J Org Chem 2021; 86:1273-1280. [PMID: 33283502 DOI: 10.1021/acs.joc.0c02529] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We herein described a selenylative cyclization reaction of enynes by the utilization of diselenides as radical sources. The visible-light irradiation of the reaction mixture enables the generation of the selenium atom radical to trigger the radical addition/cyclization/selenation sequences. Both terminal alkyne and internal alkyne derived 1,6-enynes were tested and suitable for the current synthetic protocol, delivering various kinds of selenium-containing cycles in good yields.
Collapse
Affiliation(s)
- Hong Hou
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yue Sun
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Pan
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Huaguang Yu
- Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, School of Chemical and Environmental Engineering, Jianghan University, Wuhan 430056, China
| | - Ying Han
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yaocheng Shi
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Chaoguo Yan
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Shaoqun Zhu
- School of Chemistry & Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
31
|
Hagras M, Mandour AA, Mohamed EA, Elkaeed EB, Gobaara IMM, Mehany ABM, Ismail NSM, Refaat HM. Design, synthesis, docking study and anticancer evaluation of new trimethoxyphenyl pyridine derivatives as tubulin inhibitors and apoptosis inducers. RSC Adv 2021; 11:39728-39741. [PMID: 35494162 PMCID: PMC9044549 DOI: 10.1039/d1ra07922k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/02/2021] [Indexed: 12/05/2022] Open
Abstract
Microtubules have become an appealing target for anticancer drug development including mainly colchicine binding site inhibitors (CBSIs). A new series of novel trimethoxypyridine derivatives were designed and synthesized as tubulin targeting agents. In vitro anti-proliferative activities of the tested compounds compared to colchicine against hepatocellular carcinoma (HepG-2), colorectal carcinoma (HCT-116), and breast cancer (MCF-7) was carried out. Most of compounds showed significant cytotoxic activities. Compounds Vb, Vc, Vf, Vj and VI showed superior anti-proliferative activities to colchicine. Where compound VI showed IC50 values of 4.83, 3.25 and 6.11 μM compared to colchicine (7.40, 9.32, 10.41 μM) against HCT 116, HepG-2 and MCF-7, respectively. The enzymatic activity against tubulin enzyme was carried out for the compounds that showed high anti-proliferative activity. Also, compound VI exhibited the highest tubulin polymerization inhibitory effect with an IC50 value of 8.92 nM compared to colchicine (IC50 value = 9.85 nM). Compounds Vb, Vc, Vf, Vj, & VIIIb showed promising activities with IC50 values of 22.41, 17.64, 20.39, 10.75, 31.86 nM, respectively. Cell cycle and apoptosis test for compound VI against HepG-2 cells, indicated that compound VI can arrest cell cycle at G2/M phase, and can cause apoptosis at pre-G1 phase, with high apoptotic effect 18.53%. Molecular docking studies of the designed compounds confirmed the essential hydrogen bonding with CYS241 beside the hydrophobic interaction at the binding site compared to reference compounds which assisted in the prediction of the structure requirements for the detected antitumor activity. Interaction of compounds VI (IC50 = 8.92 nM) (A) and Vj (IC50 = 10.75 nM) (B) with key amino acids of CBS.![]()
Collapse
Affiliation(s)
- Mohamed Hagras
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Asmaa A. Mandour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo 11835, Egypt
| | - Esraa A. Mohamed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo 11835, Egypt
| | - Eslam B. Elkaeed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | | | - Ahmed B. M. Mehany
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Nasser S. M. Ismail
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo 11835, Egypt
| | - Hanan M. Refaat
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo 11835, Egypt
| |
Collapse
|
32
|
Álvarez R, Aramburu L, Gajate C, Vicente-Blázquez A, Mollinedo F, Medarde M, Peláez R. Methylsulfanylpyridine based diheteroaryl isocombretastatin analogs as potent anti-proliferative agents. Eur J Med Chem 2020; 209:112933. [PMID: 33328100 DOI: 10.1016/j.ejmech.2020.112933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/30/2020] [Accepted: 10/08/2020] [Indexed: 01/30/2023]
Abstract
Isocombretastatins are the not isomerizable 1,1-diarylethene isomers of combretastatins. Both families of antimitotics are poorly soluble and new analogs with improved water solubility are needed. The ubiquitous 3,4,5-trimethoxyphenyl ring and most of its replacements contribute to the solubility problem. 39 new compounds belonging to two series of isocombretastatin analogs with 2-chloro-6-methylsulfanyl-4-pyridinyl or 2,6-bis(methylsulfanyl)-4-pyridinyl moieties replacing the 3,4,5-trimethoxyphenyl have been synthesized and their antimitotic activity and aqueous solubility have been studied. We show here that 2-chloro-6-methylsulfanylpyridines are more successful replacements than 2,6-bis(methylsulfanyl)pyridines, giving highly potent tubulin inhibitors and cytotoxic compounds with improved water solubilities. The optimal combination is with indole rings carrying polar substitutions at the three position. The resulting diheteroaryl isocombretastatin analogs showed potent cytotoxic activity against human cancer cell lines caused by tubulin inhibition, as shown by in vitro tubulin polymerization inhibitory assays, cell cycle analysis, and confocal microscopy studies. Cell cycle analysis also showed apoptotic responses following G2/M arrest after treatment. Conformational analysis and docking studies were applied to propose binding modes of the compounds at the colchicine site of tubulin and were in good agreement with the observed SAR. 2-Chloro-6-methylsulfanylpyridines represent a new and successful trimethoxyphenyl ring substitution for the development of improved colchicine site ligands.
Collapse
Affiliation(s)
- Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de La Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain.
| | - Laura Aramburu
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de La Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain.
| | - Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), E-28040, Madrid, Spain.
| | - Alba Vicente-Blázquez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), E-28040, Madrid, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de La Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain.
| | - Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), E-28040, Madrid, Spain.
| | - Manuel Medarde
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de La Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain.
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de La Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain.
| |
Collapse
|
33
|
Akram M, Adeel M, Khalid M, Tahir MN, Aliabad HAR, Ullah MA, Iqbal J, Braga AA. Highly efficient one pot palladium-catalyzed synthesis of 3,5-bis (arylated) pyridines: Comparative experimental and DFT studies. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Shuai W, Li X, Li W, Xu F, Lu L, Yao H, Yang L, Zhu H, Xu S, Zhu Z, Xu J. Design, synthesis and anticancer properties of isocombretapyridines as potent colchicine binding site inhibitors. Eur J Med Chem 2020; 197:112308. [DOI: 10.1016/j.ejmech.2020.112308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
|
35
|
Gao F, Huang G, Xiao J. Chalcone hybrids as potential anticancer agents: Current development, mechanism of action, and structure-activity relationship. Med Res Rev 2020; 40:2049-2084. [PMID: 32525247 DOI: 10.1002/med.21698] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022]
Abstract
The continuous emergency of drug-resistant cancers and the low specificity of anticancer agents have been the major challenges in the control and treatment of cancer, making an urgent need to develop novel anticancer agents with high efficacy. Chalcones, precursors of flavonoids and isoflavonoids, exhibit structural heterogeneity and can act on various drug targets. Chalcones which demonstrated potential in vitro and in vivo activity against both drug-susceptible and drug-resistant cancers, are useful templates for the development of novel anticancer agents. Hybridization of chalcone moiety with other anticancer pharmacophores could provide the hybrids which have the potential to overcome drug resistance and improve the specificity, so it represents a promising strategy to develop novel anticancer agents. This review emphasizes the development, the mechanisms of action as well as structure-activity relationships of chalcone hybrids with potential therapeutic application for many cancers in recent 10 years.
Collapse
Affiliation(s)
- Feng Gao
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jiaqi Xiao
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
36
|
Ultrasounds-mediated 10-seconds synthesis of chalcones as potential farnesyltransferase inhibitors. Bioorg Med Chem Lett 2020; 30:127149. [PMID: 32247731 DOI: 10.1016/j.bmcl.2020.127149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 11/23/2022]
Abstract
A broad range of chalcones and derivatives were easily and rapidly synthesized, following Claisen-Schmidt condensation of (hetero)aryl ketones and (hetero)aryl aldehydes using a ultrasound probe. A comparison was made with classical magnetic stirring experiments, and an optimization study was realized, showing lithium hydroxide to be the best basic catalyst of the studied condensations. By-products of the reactions (β-hydroxy-ketone, diketones, and cyclohexanols) were also isolated. All compounds were evaluated in vitro for their ability to inhibit human farnesyltransferase, a protein implicated in cancer and rare diseases and on the NCI-60 cancer cell lines panel. Molecules showed inhibitory activity on the target protein and cytostatic effect on different cell lines with particular activity against MCF7, breast cancer cells.
Collapse
|
37
|
Deliberate-Characterization for Ni(II)-Schiff Base Complexes: Promising In-Vitro Anticancer Feature that Matched MOE Docking-Approach. J Inorg Organomet Polym Mater 2020. [DOI: 10.1007/s10904-020-01503-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
38
|
Álvarez R, Aramburu L, Gajate C, Vicente-Blázquez A, Mollinedo F, Medarde M, Peláez R. Potent colchicine-site ligands with improved intrinsic solubility by replacement of the 3,4,5-trimethoxyphenyl ring with a 2-methylsulfanyl-6-methoxypyridine ring. Bioorg Chem 2020; 98:103755. [PMID: 32200330 DOI: 10.1016/j.bioorg.2020.103755] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 12/27/2022]
Abstract
Colchicine site antimitotic agents typically suffer from low aqueous solubilities and are formulated as phosphate prodrugs of phenolic groups. These hydroxyl groups are the aim of metabolic transformations leading to resistance. There is an urgent need for more intrinsically soluble analogues lacking these hydroxyl groups. The 3,4,5-trimethoxyphenyl ring of combretastatin A-4 is a liability in terms of solubility but it is considered essential for high cytotoxic and tubulin polymerization inhibitory (TPI) activity. We have synthesized 36 new analogues of combretastatin A-4 replacing the trimethoxyphenyl moiety with more polar pyridine based moieties, measured their aqueous solubility, and studied their anti-proliferative effects against 3 human cancer cell lines. We show here that pyridine rings can be successful replacements for the trimethoxyphenyl ring, resulting in potent and more soluble analogues. The more straightforward replacement, a 2,6-dimethoxypyridine ring led to inactive analogues, but a 2-methoxy-6-methylsulfanylpyridine moiety led to active analogues when combined with different B rings. This replacement led to potent cytotoxic activity against sensitive human cancer cell lines due to tubulin inhibition, as shown by cell cycle analysis, confocal microscopy, and tubulin polymerization inhibitory activity studies. Cell cycle analysis also showed apoptotic responses following treatment. Docking studies suggested binding at the colchicine site of tubulin and provided a good agreement with the observed SAR. A 2-methoxy-6-methylsulfanylpyridine moiety is a good trimethoxyphenyl ring replacement for the development of new colchicine site ligands.
Collapse
Affiliation(s)
- Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| | - Laura Aramburu
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| | - Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), E-28040 Madrid, Spain.
| | - Alba Vicente-Blázquez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), E-28040 Madrid, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| | - Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), E-28040 Madrid, Spain.
| | - Manuel Medarde
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| |
Collapse
|
39
|
Abdel‐Latif E, Alashhab RE, El‐Demerdash A, Ismail MA. Utilization of 5‐Chloro‐2‐(cyanoacetamido)pyridines in the Synthesis of Biologically Active Heterocyclic Hybrids. ChemistrySelect 2020. [DOI: 10.1002/slct.201904051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ehab Abdel‐Latif
- Chemistry Department, Faculty of ScienceMansoura University Mansoura 35516 Egypt
| | - Rabia E. Alashhab
- Chemistry Department, Faculty of ScienceMansoura University Mansoura 35516 Egypt
| | - Amr El‐Demerdash
- Chemistry Department, Faculty of ScienceMansoura University Mansoura 35516 Egypt
| | - Mohamed A. Ismail
- Chemistry Department, Faculty of ScienceMansoura University Mansoura 35516 Egypt
| |
Collapse
|
40
|
Chen Z, Lai H, Hou L, Chen T. Rational design and action mechanisms of chemically innovative organoselenium in cancer therapy. Chem Commun (Camb) 2020; 56:179-196. [PMID: 31782422 DOI: 10.1039/c9cc07683b] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Organo-seleno compounds (org-Se) have been widely used in antitumor, antiviral, and antiinflammatory therapy; antioxidation and other biological fields. As such, they have made an important contribution to overcoming various kinds of diseases, and researchers are increasingly attracted to org-Se's synthesis and functional design. This review is mainly focused on the design and synthesis of various kinds of org-Se, followed by their anticancer mechanisms such as the mitochondria mediated pathway induced by ROS, death receptor mediated pathways involving p53 phosphorylation, and the activation of the AMPK pathway to promote apoptosis. Org-Se also serves as a sensitizer in chemotherapy and radiotherapy, and an antagonist against the cytotoxic effects induced by chemotherapeutic agents. Finally, we will summarize the development of cancer-targeted org-Se containing complexes, and nanotechnology-based org-Se for anticancer application. This review could provide information for the future design of chemically innovative org-Se with anticancer potential, and shed light on the discovery of nanomaterial-based pharmaceuticals to improve drug development and formation.
Collapse
Affiliation(s)
- Zhen Chen
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | | | | | | |
Collapse
|
41
|
Malebari AM, Fayne D, Nathwani SM, O'Connell F, Noorani S, Twamley B, O'Boyle NM, O'Sullivan J, Zisterer DM, Meegan MJ. β-Lactams with antiproliferative and antiapoptotic activity in breast and chemoresistant colon cancer cells. Eur J Med Chem 2020; 189:112050. [PMID: 31954879 DOI: 10.1016/j.ejmech.2020.112050] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/20/2019] [Accepted: 01/08/2020] [Indexed: 12/31/2022]
Abstract
A series of novel 1,4-diaryl-2-azetidinone analogues of combretastatin A-4 (CA-4) have been designed, synthesised and evaluated in vitro for antiproliferative activity, antiapoptotic activity and inhibition of tubulin polymerisation. Glucuronidation of CA-4 by uridine 5-diphosphoglucuronosyl transferase enzymes (UGTs) has been identified as a mechanism of resistance in cancer cells. Potential sites of ring B glucuronate conjugation are removed by replacing the B ring meta-hydroxy substituent of selected series of β-lactams with alternative substituents e.g. F, Cl, Br, I, CH3. The 3-phenyl-β-lactam 11 and 3-hydroxy-β-lactam 46 demonstrate improved activity over CA-4 in CA-4 resistant HT-29 colon cancer cells (IC50 = 9 nM and 3 nM respectively compared with IC50 = 4.16 μM for CA-4), while retaining potency in MCF-7 breast cancer cells (IC50 = 17 nM and 22 nM respectively compared with IC50 = for 4 nM for CA-4). Compound 46 binds at the colchicine site of tubulin, and strongly inhibits tubulin assembly at micromolar concentrations comparable to CA-4. In addition, compound 46 induced mitotic arrest at low concentration in both cell lines MCF-7 and HT-29 together with downregulation of expression of antiapoptotic proteins Mcl-1, Bcl-2 and survivin in MCF-7 cells. These novel antiproliferative and antiapoptotic β-lactams are potentially useful scaffolds in the development of tubulin-targeting agents for the treatment of breast cancers and chemoresistant colon cancers.
Collapse
Affiliation(s)
- Azizah M Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia; School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland.
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Seema M Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Fiona O'Connell
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Dublin 2, Ireland
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Niamh M O'Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Dublin 2, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, Ireland
| |
Collapse
|
42
|
Abumelha HM. Synthesis and antioxidant assay of new nicotinonitrile analogues clubbed thiazole, pyrazole and/or pyridine ring systems. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.3820] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Hana M.A. Abumelha
- Department of Chemistry, Faculty of SciencePrincess Nourah Bint Abdulrahman University Riyadh Saudi Arabia
| |
Collapse
|
43
|
A novel chalcone derivative as Nrf2 activator attenuates learning and memory impairment in a scopolamine-induced mouse model. Eur J Med Chem 2020; 185:111777. [DOI: 10.1016/j.ejmech.2019.111777] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/01/2019] [Accepted: 10/09/2019] [Indexed: 01/06/2023]
|
44
|
The Masked Polar Group Incorporation (MPGI) Strategy in Drug Design: Effects of Nitrogen Substitutions on Combretastatin and Isocombretastatin Tubulin Inhibitors. Molecules 2019; 24:molecules24234319. [PMID: 31779228 PMCID: PMC6930638 DOI: 10.3390/molecules24234319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 01/14/2023] Open
Abstract
Colchicine site ligands suffer from low aqueous solubility due to the highly hydrophobic nature of the binding site. A new strategy for increasing molecular polarity without exposing polar groups—termed masked polar group incorporation (MPGI)—was devised and applied to nitrogenated combretastatin analogues. Bulky ortho substituents to the pyridine nitrogen hinder it from the hydrophobic pocket while increasing molecular polarity. The resulting analogues show improved aqueous solubilities and highly potent antiproliferative activity against several cancer cell lines of different origin. The more potent compounds showed moderate tubulin polymerization inhibitory activity, arrested the cell cycle of treated cells at the G2/M phase, and subsequently caused apoptotic cell death represented by the cells gathered at the subG0/G1 population after 48 h of treatment. Annexin V/Propidium Iodide (PI) double-positive cells observed after 72 h confirmed the induction of apoptosis. Docking studies suggest binding at the colchicine site of tubulin in a similar way as combretastatin A4, with the polar groups masked by the vicinal substituents. These results validate the proposed strategy for the design of colchicine site ligands and open a new road to increasing the aqueous solubility of ligands binding in apolar environments.
Collapse
|