1
|
Altharawi A, Alqahtani SM, Aldakhil T, Sharma P, Kumar A, Ramadan MF. A novel green and efficient heterogeneous acid catalyst for the one-pot synthesis of benzopyrazine-aminoimidazole hybrids with antiproliferative potential. RSC Adv 2024; 14:26219-26232. [PMID: 39161442 PMCID: PMC11332591 DOI: 10.1039/d4ra04725g] [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: 06/29/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
A novel, green, efficient, and stable magnetically heterogeneous nanocatalyst was developed by immobilizing butanesulfonic acid (BuSO3H) onto the surface of MFe2O4 magnetic nanoparticles (MNPs). The resulting core-shell structure of the MFe2O4@PDA@BuSO3H nanocatalyst was thoroughly characterized using various analytical techniques, including Fourier-Transform Infrared Spectroscopy (FT-IR), X-ray Diffraction (XRD), Energy-Dispersive X-ray Spectroscopy (EDS), Field Emission Scanning Electron Microscopy (FESEM), Transmission Electron Microscopy (TEM), Vibrating Sample Magnetometry (VSM), and Brunauer-Emmett-Teller (BET) analysis. A nanocatalyst was used to synthesize 2-benzopyrazine-aminoimidazole hybrid derivatives through a domino multicomponent Knoevenagel-condensation-cyclization reaction (5a-p) in an environmentally friendly manner. The resulting compounds were then tested for their anticancer activity against three types of human cancer cells (MCF-7, A549, and U87) using the MTT assay. The experiment showed that the nanocatalyst had excellent catalytic activity, and the synthesized compounds exhibited promising antiproliferative activity. Notably, compounds 5g and 5h, containing a 2-naphthyl ring, showed the highest antiproliferative effects against MCF-7 cells, with IC50 values of 0.03 and 0.32 μM, respectively. Additionally, the activity of compounds 5g and 5h in tubulin polymerization, apoptosis induction, and cell cycle arrest in MCF-7 cells were investigated. The results demonstrated that these compounds effectively induced apoptosis and cell cycle arrest. The binding of representative compounds to the colchicine binding site of tubulin was confirmed through molecular modeling studies.
Collapse
Affiliation(s)
- Ali Altharawi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University Al-Kharj 11942 Saudi Arabia
| | - Safar M Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University Al-Kharj 11942 Saudi Arabia
| | - Taibah Aldakhil
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University Al-Kharj 11942 Saudi Arabia
| | - Pawan Sharma
- Department of Chemistry, School of Sciences, Jain (Deemed-to-be) University Bengaluru Karnataka 560069 India
- Department of Sciences, Vivekananda Global University Jaipur Rajasthan 303012 India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University Gangoh Uttar Pradesh 247341 India
- Department of Pharmacy, Arka Jain University Jamshedpur Jharkhand 831001 India
| | | |
Collapse
|
2
|
Montero V, Montana M, Carré M, Vanelle P. Quinoxaline derivatives: Recent discoveries and development strategies towards anticancer agents. Eur J Med Chem 2024; 271:116360. [PMID: 38614060 DOI: 10.1016/j.ejmech.2024.116360] [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: 12/20/2023] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
Cancer is a leading cause of death and a major health problem worldwide. While many effective anticancer agents are available, most drugs currently on the market are not specific, raising issues like the common side effects of chemotherapy. However, recent research hold promises for the development of more efficient and safer anticancer drugs. Quinoxaline and its derivatives are becoming recognized as a novel class of chemotherapeutic agents with activity against different tumors. The present review compiles and discusses studies concerning the therapeutic potential of the anticancer activity of quinoxaline derivatives, covering articles published between January 2018 and January 2023.
Collapse
Affiliation(s)
- Vincent Montero
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, CEDEX 05, 13385, Marseille, France; AP-HM, Service de Pharmacologie Clinique et Pharmacovigilance, Hôpital de la Timone, Marseille CEDEX 05, 13385, France.
| | - Marc Montana
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, CEDEX 05, 13385, Marseille, France; AP-HM, Oncopharma, Hôpital Nord, Marseille, France
| | - Manon Carré
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm UMR1068, CNRS UMR7258, Aix-Marseille Université UM105, Institut Paoli Calmettes - Faculté de Pharmacie, Marseille, France
| | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, CEDEX 05, 13385, Marseille, France; AP-HM, Service Central de la Qualité et de l'Information Pharmaceutiques, Hôpital Conception, Marseille, 13005, France
| |
Collapse
|
3
|
Liang T, Dong H, Wang Z, Lu L, Song X, Qi J, Zhang Y, Wang J, Du G. Discovery of novel urea derivatives as ferroptosis and autophagy inducer for human colon cancer treatment. Eur J Med Chem 2024; 268:116277. [PMID: 38422700 DOI: 10.1016/j.ejmech.2024.116277] [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: 09/16/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
A series of novel urea derivatives were designed, synthesized and evaluated for their inhibitory activities against HT-29 cells, and structure-activity relationships (SAR) were summarized. Compound 10p stood out from these derivatives, exhibiting the most potent antiproliferative activity. Further biological studies demonstrated that 10p arrested cell cycle at G2/M phase via regulating cell cycle-related proteins CDK1 and Cyclin B1. The underlying molecular mechanisms demonstrated that 10p induced cell death through ferroptosis and autophagy, but not apoptosis. Moreover, 10p-induced ferroptosis and autophagy were both related with accumulation of ROS, but they were independent of each other. Our findings substantiated that 10p combines ferroptosis induction and autophagy trigger in single molecule, making it a potential candidate for colon cancer treatment and is worth further development.
Collapse
Affiliation(s)
- Tingting Liang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China
| | - Haiyang Dong
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China
| | - Zhuangzhuang Wang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China
| | - Lu Lu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China
| | - Xueting Song
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China
| | - Jianguo Qi
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China
| | - Yahong Zhang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China.
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China; Huaihe Hospital of Henan University, Kaifeng, 475004, Henan, China.
| | - Guanhua Du
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, 475004, Henan, China; School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China.
| |
Collapse
|
4
|
Farghaly TA, Alqurashi RM, Masaret GS, Abdulwahab HG. Recent Methods for the Synthesis of Quinoxaline Derivatives and their Biological Activities. Mini Rev Med Chem 2024; 24:920-982. [PMID: 37885112 DOI: 10.2174/0113895575264375231012115026] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 10/28/2023]
Abstract
Quinoxaline derivatives have been incorporated into numerous marketed drugs used for the treatment of various diseases. Examples include glecaprevir (Mavyret), voxilaprevir (Vosevi), Balversa (L01EX16) (erdafitinib), carbadox, XK469R (NSC698215), and becampanel (AMP397). These quinoxaline derivatives exhibit a diverse range of pharmacological activities, including antibacterial, antitubercular, antiviral, anti-HIV, anti-inflammatory, antifungal, anticancer, antiproliferative, antitumor, kinase inhibition, antimicrobial, antioxidant, and analgesic effects. Recognizing the significance of these bioactive quinoxaline derivatives, researchers have dedicated their efforts to developing various synthetic methods for their production. This review aimed to compile the most recent findings on the synthesis and biological properties of quinoxaline derivatives from 2015 to 2023.
Collapse
Affiliation(s)
- Thoraya A Farghaly
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Raghad M Alqurashi
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ghada S Masaret
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hanan Gaber Abdulwahab
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
5
|
Dong H, Lu L, Song X, Li Y, Zhou J, Xu Y, Zhang Y, Qi J, Liang T, Wang J. Design, synthesis and biological evaluation of tetrahydroquinoxaline sulfonamide derivatives as colchicine binding site inhibitors. RSC Adv 2023; 13:30202-30216. [PMID: 37849704 PMCID: PMC10577396 DOI: 10.1039/d3ra05720h] [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: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
Colchicine binding site inhibitors (CBSIs) are potential microtubule targeting agents (MTAs), which can overcome multidrug resistance, improve aqueous solubility and reduce toxicity faced by most MTAs. Novel tetrahydroquinoxaline sulfonamide derivatives were designed, synthesized and evaluated for their antiproliferative activities. The MTT assay results demonstrated that some derivatives exhibited moderate to strong inhibitory activities against HT-29 cell line. Among them, compound I-7 was the most active compound. Moreover, I-7 inhibited tubulin polymerization, disturbed microtubule network, disrupted the formation of mitotic spindle and arrested cell cycle at G2/M phase. However, I-7 didn't induce cell apoptosis. Furthermore, the prediction of ADME demonstrated that I-7 showed favorable physiochemical and pharmacokinetic properties. And the detailed molecular docking confirmed I-7 targeted the site of colchicine through hydrogen and hydrophobic interactions.
Collapse
Affiliation(s)
- Haiyang Dong
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Lu Lu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Xueting Song
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Youkang Li
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jinguang Zhou
- Huaihe Hospital of Henan University Kaifeng 475004 Henan China
| | - Yungen Xu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 211198 China
| | - Yahong Zhang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jianguo Qi
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Tingting Liang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| |
Collapse
|
6
|
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
|