1
|
Ding SY, Yang YX, Liu C, Quan XY, Zhao ZH, Jin CH. Synthesis and biological evaluation of sulfonamide derivatives containing imidazole moiety as ALK5 inhibitors. Mol Divers 2025; 29:2143-2156. [PMID: 39212874 DOI: 10.1007/s11030-024-10973-y] [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: 07/05/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Four series of sulfonamide derivatives (13a-b, 14a-d, 15a-b, and 16a-d) were synthesized and evaluated for their activin receptor-like kinase 5 (ALK5) inhibitory activities. Of these, compounds 13b (IC50 = 0.130 μM) and 15a (IC50 = 0.130 μM) showed the highest inhibitory activities against ALK5 kinase, with activities similar to the positive control LY-2157299. Notably, we discovered that introduction of sulfonamide group at the 2-position of the central imidazole ring significantly increased ALK5 inhibitory activity. Compounds 13b and 15a did not show toxicity in A549 cells up to the maximum concentration of 50 μM, and effectively inhibited TGF-β1-induced Smad-signaling and cell motility in A549 cells. The results indicate that compounds 13b and 15a are worth of further development as anticancer agents.
Collapse
Affiliation(s)
- Shu-Yan Ding
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Yu-Xuan Yang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Chuang Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Xu-Yin Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Zi-Han Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
2
|
Li WX, Lu YF, Wang F, Ai B, Jin SB, Li S, Xu GH, Jin CH. Application of 18β-glycyrrhetinic acid in the structural modification of natural products: a review. Mol Divers 2025; 29:739-781. [PMID: 38683490 DOI: 10.1007/s11030-024-10864-2] [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: 02/04/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
18β-Glycyrrhetinic acid (GA) is an oleane-type pentacyclic triterpene saponin obtained from glycyrrhizic acid by removing 2 glucuronic acid groups. GA and its analogues are active substances of glycyrrhiza aicd, with similar structure and important pharmacological effects such as anti-inflammatory, anti-diabetes, anti-tumor and anti-fibrosis. Although GA combined compounds are in the clinical trial stages, its application potential is severely restricted by its low bioavailability, water solubility and membrane permeability. In this article, synthetic methods and structure-activity relationships (SARs) of GA derivatives from 2018 to present are reviewed based on pharmacological activity. It is hoped that this review can provide reference for the future development of potential GA preclinical candidate compounds, and furnish ideas for the development of pentacyclic triterpenoid lead compounds.
Collapse
Affiliation(s)
- Wan-Xin Li
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Ye-Fang Lu
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Fei Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Bing Ai
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Sheng-Bo Jin
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Siqi Li
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Guang-Hua Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China.
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
3
|
Han LY, Sun JX, Liu C, Ai B, Piao MG, Zhang C, Quan JS, Jin CH. Synthesis and antimicrobial activity evaluation of pyrazole derivatives containing imidazothiadiazole moiety. Future Med Chem 2025; 17:157-170. [PMID: 39723690 PMCID: PMC11749441 DOI: 10.1080/17568919.2024.2444868] [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: 09/02/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
AIM The purpose of this work was to investigate the antimicrobial activity of pyrazole derivatives (21a - i and 23a - o) synthesized. MATERIALS & METHODS The pyrazole derivatives were synthesized, molecular docked and tested for their antimicrobial activity, cytotoxicity, and hemolysis rate. RESULTS Most of the target compounds showed high selective inhibitory activity against multi-drug resistance compared with other strains. Of these, compounds 21c (MIC = 0.25 µg/mL) and 23 h (MIC = 0.25 µg/mL) showed the strongest antibacterial activity, which was 4-flods than that of the positive control compound gatifloxacin (MIC = 1 µg/mL). Furthermore, compound 23 h showed no cytotoxicity to human LO2 cells, and did not show hemolysis even at ultra-high concentration. CONCLUSION These results prompted that these compounds are valuable for further development as antimicrobial agents.
Collapse
Affiliation(s)
- Lan-Ying Han
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Jing-Xin Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Chuang Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Bing Ai
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Ming-Guan Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Changhao Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Ji-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| |
Collapse
|
4
|
Yang YX, Guo J, Liu C, Nan JX, Wu YL, Jin CH. Synthesis of amide derivatives containing the imidazole moiety and evaluation of their anti-cardiac fibrosis activity. Arch Pharm (Weinheim) 2024; 357:e2400131. [PMID: 38678538 DOI: 10.1002/ardp.202400131] [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: 02/18/2024] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Three series of N-{[4-([1,2,4]triazolo[1,5-α]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl]methyl}acetamides (14a-d, 15a-n, and 16a-f) were synthesized and evaluated for activin receptor-like kinase 5 (ALK5) inhibitory activities in an enzymatic assay. The target compounds showed high ALK5 inhibitory activity and selectivity. The half maximal inhibitory concentration (IC50) for phosphorylation of ALK5 of 16f (9.1 nM), the most potent compound, was 2.7 times that of the clinical candidate EW-7197 (vactosertib) and 14 times that of the clinical candidate LY-2157299. The selectivity index of 16f against p38α mitogen-activated protein kinase was >109, which was much higher than that of positive controls (EW-7197: >41, and LY-2157299: 4). Furthermore, a molecular docking study provided the interaction modes between the target compounds and ALK5. Compounds 14c, 14d, and 16f effectively inhibited the protein expression of α-smooth muscle actin (α-SMA), collagen I, and tissue inhibitor of metalloproteinase 1 (TIMP-1)/matrix metalloproteinase 13 (MMP-13) in transforming growth factor-β-induced human umbilical vein endothelial cells. Compounds 14c and 16f showed especially high activity at low concentrations, which suggests that these compounds could inhibit myocardial cell fibrosis. Compounds 14c, 14d, and 16f are potential preclinical candidates for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Yu-Xuan Yang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Jia Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Chuang Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Ji-Xing Nan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, China
| | - Yan-Ling Wu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, China
| |
Collapse
|
5
|
Mansour MA, Hassan GS, Serya RAT, Jaballah MY, Abouzid KAM. Advances in the discovery of activin receptor-like kinase 5 (ALK5) inhibitors. Bioorg Chem 2024; 147:107332. [PMID: 38581966 DOI: 10.1016/j.bioorg.2024.107332] [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: 02/12/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Activin receptor‑like kinase-5 (ALK5) is an outstanding member of the transforming growth factor-β (TGF-β) family. (TGF-β) signaling pathway integrates pleiotropic proteins that regulate various cellular processes such as growth, proliferation, and differentiation. Dysregulation within the signaling pathway can cause variety of diseases, such as fibrosis, cardiovascular disease, and especially cancer, rendering ALK5 a potential drug target. Hence, various small molecules have been designed and synthesized as potent ALK5 inhibitors. In this review, we shed light on the current ATP-competitive inhibitors of ALK5 through diverse heterocyclic based scaffolds that are in clinical or pre-clinical phases of development. Moreover, we focused on the binding interactions of the compounds to the ATP binding site and the structure-activity relationship (SAR) of each scaffold, revealing new scopes for designing novel candidates with enhanced selectivity and metabolic profiles.
Collapse
Affiliation(s)
- Mai A Mansour
- Pharmaceutical Chemistry Department, School of Pharmacy, Badr University in Cairo, Egypt.
| | - Ghaneya S Hassan
- Pharmaceutical Chemistry Department, School of Pharmacy, Badr University in Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Egypt
| | - Rabah A T Serya
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Maiy Y Jaballah
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Egypt.
| |
Collapse
|
6
|
Meng YQ, Ren J, Sun JX, Guo FY, Min JZ, Nan JX, Quan JS, Lian LH, Jin CH. Synthesis and anti-liver fibrosis activity of imidazole and thiazole compounds containing amino acids. Eur J Med Chem 2024; 269:116311. [PMID: 38508118 DOI: 10.1016/j.ejmech.2024.116311] [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: 10/14/2023] [Revised: 01/27/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
Four series of imidazoles (15a-g, 20c, and 20d) and thiazoles (18a-g, 22a, and 22b) possessing various amino acids were synthesized and evaluated for activin receptor-like kinase 5 (ALK5) inhibitory activities in an enzymatic assay. Among them, compounds 15g and 18c showed the highest inhibitory activity against ALK5, with IC50 values of 0.017 and 0.025 μM, respectively. Compounds 15g and 18c efficiently inhibited extracellular matrix (ECM) deposition in TGF-β-induced hepatic stellate cells (HSCs), and eventually suppressed HSC activation. Moreover, compound 15g showed a good pharmacokinetic (PK) profile with a favorable half-life (t1/2 = 9.14 h). The results indicated that these compounds exhibited activity targeting ALK5 and may have potential in the treatment of liver fibrosis; thus they are worthy of further study.
Collapse
Affiliation(s)
- Yu-Qing Meng
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Jie Ren
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Jing-Xin Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Fang-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Jun-Zhe Min
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Ji-Xing Nan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Ji-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| | - Li-Hua Lian
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
7
|
Yang L, Jiao YX, Quan YH, Li MY, Huang XY, Jin JZ, Li S, Quan JS, Jin CH. Synthesis and Antimicrobial Activity Evaluation of Pyridine Derivatives Containing Imidazo[2,1-b][1,3,4]Thiadiazole Moiety. Chem Biodivers 2024; 21:e202400135. [PMID: 38425248 DOI: 10.1002/cbdv.202400135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/02/2024]
Abstract
Four series of novel pyridine derivatives (17 a-i, 18 a-i, 19 a-e, and 20 a-e) were synthesized and their antimicrobial activities were evaluated. Of all the target compounds, almost half target compounds showed moderate or high antibacterial activity. The 4-F substituted compound 17 d (MIC=0.5 μg/mL) showed the highest antibacterial activity, its activity was twice the positive control compound gatifloxacin (MIC=1.0 μg/mL). For fungus ATCC 9763, the activities of compounds 17 a and 17 d are equivalent to the positive control compound fluconazole (MIC=8 μg/mL). Furthermore, compounds 17 a and 17 d showed little cytotoxicity to human LO2 cells, and did not show hemolysis even at ultra-high concentration (200 μM). The results indicate that these compounds are valuable for further development as antibacterial and antifungal agents.
Collapse
Affiliation(s)
- Liu Yang
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Yu-Xin Jiao
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Yan-Hua Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Ming-Yu Li
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Xin-Yi Huang
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Jun-Zheng Jin
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Siqi Li
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Ji-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| |
Collapse
|
8
|
Zhang J, Yu J, Liu M, Xie Z, Lei X, Yang X, Huang S, Deng X, Wang Z, Tang G. Small-molecule modulators of tumor immune microenvironment. Bioorg Chem 2024; 145:107251. [PMID: 38442612 DOI: 10.1016/j.bioorg.2024.107251] [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: 12/13/2023] [Revised: 02/13/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
In recent years, tumor immunotherapy, aimed at increasing the activity of immune cells and reducing immunosuppressive effects, has attracted wide attention. Among them, immune checkpoint blocking (ICB) is the most commonly explored therapeutic approach. All approved immune checkpoint inhibitors (ICIs) are clinically effective monoclonal antibodies (mAbs). Compared with biological agents, small-molecule drugs have many unique advantages in tumor immunotherapy. Therefore, they also play an important role. Immunosuppressive signals such as PD-L1, IDO1, and TGF-β, etc. overexpressed in tumor cells form the tumor immunosuppressive microenvironment. In addition, the efficacy of multi-pathway combined immunotherapy has also been reported and verified. Here, we mainly reviewed the mechanism of tumor immunotherapy, analyzed the research status of small-molecule modulators, and discussed drug candidates' structure-activity relationship (SAR). It provides more opportunities for further research to design more immune small-molecule modulators with novel structures.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jia Yu
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Meijing Liu
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyan Yang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Sheng Huang
- Jiuzhitang Co., Ltd, Changsha, Hunan 410007, China
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
9
|
Xu WB, Li S, Zheng CJ, Yang YX, Zhang C, Jin CH. Synthesis and Evaluation of Imidazole Derivatives Bearing Imidazo[2,1-b] [1,3,4]thiadiazole Moiety as Antibacterial Agents. Med Chem 2024; 20:40-51. [PMID: 37767798 DOI: 10.2174/0115734064248204230919074743] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/19/2023] [Accepted: 07/27/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Drug-resistant infections kill hundreds of thousands of people globally every year. In previous work, we found that tri-methoxy- and pyridine-substituted imidazoles show strong antibacterial activities. OBJECTIVE The aim of this work was to investigate the antibacterial activities and bacterial resistances of imidazoles bearing an aromatic heterocyclic, alkoxy, or polycyclic moiety on the central ring. METHODS Three series of 2-cyclopropyl-5-(5-(6-methylpyridin-2-yl)-2-substituted-1H-imidazol-4- yl)-6-phenylimidazo[2,1-b][1,3,4]thiadiazoles (13a-e, 14a-d, and 15a-f) were synthesized and their antibacterial activity was evaluated. The structures were confirmed by their 1H NMR, 13C NMR, and HRMS spectra. All the synthesized compounds were screened against Gram-positive, Gramnegative, and multidrug-resistant bacterial strains. RESULTS More than half of the compounds showed moderate or strong antibacterial activity. Among them, compound 13e (MICs = 1-4 μg/mL) showed the strongest activity against Gram-positive and drug-resistant bacteria as well as high selectivity against Gram-negative bacteria. Furthermore, it showed no cytotoxicity against HepG2 cells, even at 100 μM, and no hemolysis at 20 μM. CONCLUSION These results indicate that compound 13e is excellent candicate for further study as a potential antibacterial agent.
Collapse
Affiliation(s)
- Wen-Bo Xu
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji 133002, P.R. China
| | - Siqi Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P.R. China
| | - Chang-Ji Zheng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P.R. China
| | - Yu-Xuan Yang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P.R. China
| | - Changhao Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P.R. China
| | - Cheng-Hua Jin
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji 133002, P.R. China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P.R. China
| |
Collapse
|
10
|
Qi JD, Meng YQ, Sun J, Li WX, Zhai HX, Zhang C, Quan J, Jin CH. Synthesis and antimicrobial activity evaluation of pyrazole derivatives containing the imidazo[2,1-b][1,3,4]thiadiazole moiety. Arch Pharm (Weinheim) 2023:e2300110. [PMID: 37328442 DOI: 10.1002/ardp.202300110] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
Four series of novel pyrazole derivatives (compounds 17a-m, 18a-m, 19a-g, and 20a-g) were synthesized, and their antibacterial and antifungal activities were evaluated. Most of the target compounds (17a-m, 18k-m, and 19b-g) showed strong antifungal activity and high selectivity relative to both Gram-positive and Gram-negative bacteria. Among them, compounds 17l (minimum inhibitory concentration [MIC] = 0.25 µg/mL) and 17m (MIC = 0.25 µg/mL) showed the strongest antifungal activity, being 2- and 4-fold more active than the positive controls gatifloxacin and fluconazole, respectively. In particular, compound 17l showed little cytotoxicity against human LO2 cells and did not exhibit hemolysis at ultrahigh concentrations, as did the positive control compounds gatifloxacin and fluconazole. These results indicate that these compounds are valuable for further development as antifungal agents.
Collapse
Affiliation(s)
- Jun-Da Qi
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Yu-Qing Meng
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, China
| | - Jingxin Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Wan-Xin Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Hou-Xiang Zhai
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Changhao Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
| | - Jishan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, China
| |
Collapse
|
11
|
Yedla P, Babalghith AO, Andra VV, Syed R. PROTACs in the Management of Prostate Cancer. Molecules 2023; 28:molecules28093698. [PMID: 37175108 PMCID: PMC10179857 DOI: 10.3390/molecules28093698] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
Cancer treatments with targeted therapy have gained immense interest due to their low levels of toxicity and high selectivity. Proteolysis-Targeting Chimeras (PROTACs) have drawn special attention in the development of cancer therapeutics owing to their unique mechanism of action, their ability to target undruggable proteins, and their focused target engagement. PROTACs selectively degrade the target protein through the ubiquitin-proteasome system, which describes a different mode of action compared to conventional small-molecule inhibitors or even antibodies. Among different cancer types, prostate cancer (PC) is the most prevalent non-cutaneous cancer in men. Genetic alterations and the overexpression of several genes, such as FOXA1, AR, PTEN, RB1, TP53, etc., suppress the immune response, resulting in drug resistance to conventional drugs in prostate cancer. Since the progression of ARV-110 (PROTAC for PC) into clinical phases, the focus of research has quickly shifted to protein degraders targeting prostate cancer. The present review highlights an overview of PROTACs in prostate cancer and their superiority over conventional inhibitors. We also delve into the underlying pathophysiology of the disease and explain the structural design and linkerology strategies for PROTAC molecules. Additionally, we touch on the various targets for PROTAC in prostate cancer, including the androgen receptor (AR) and other critical oncoproteins, and discuss the future prospects and challenges in this field.
Collapse
Affiliation(s)
- Poornachandra Yedla
- Department of Pharmacogenomics, Institute of Translational Research, Asian Healthcare Foundation, Asian Institute of Gastroenterology Hospitals, Gachibowli, Hyderabad 500082, India
| | - Ahmed O Babalghith
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Vindhya Vasini Andra
- Department of Medical Oncology, Omega Hospitals, Gachibowli, Hyderabad 500032, India
| | - Riyaz Syed
- Department of Chemiinformatics, Centella Scientific, JHUB, Jawaharlal Nehru Technological University, Hyderabad 500085, India
| |
Collapse
|
12
|
Kang BN, Kang HJ, Kim S, Lee J, Lee J, Jeong HJ, Jeon S, Shin Y, Yoon C, Han C, Seo J, Yun J. Synthesis and biological evaluation of N-(3-fluorobenzyl)-4-(1-(methyl-d 3)-1H-indazol-5-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-amine as a novel, potent ALK5 receptor inhibitor. Bioorg Med Chem Lett 2023; 85:129205. [PMID: 36858078 DOI: 10.1016/j.bmcl.2023.129205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Specific inhibition of ALK5 provides a novel method for controlling the development of cancers and fibrotic diseases. In this work, a novel series of N-(3-fluorobenzyl)-4-(1-(methyl-d3)-1H-indazol-5-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-amine (11), a potential clinical candidate, was synthesized by strategic incorporation of deuterium at potential metabolic soft spots and identified as ALK5 inhibitors. This compound has a low potential for CYP-mediated drug-drug interactions as a CYP450 inhibitor (IC50 = >10 μM) and showed potent inhibitory effects in cellular assay (IC50 = 3.5 ± 0.4 nM). The pharmacokinetic evaluation of 11 in mice demonstrated moderate clearance (29.0 mL/min/kg) and also revealed high oral bioavailability in mice (F = 67.6%).
Collapse
Affiliation(s)
- Byung-Nam Kang
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea; Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hong-Jun Kang
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Sunjoo Kim
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Jungwoo Lee
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Jinwoo Lee
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Hee-Jin Jeong
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Seeun Jeon
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Youngdo Shin
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Cheolhwan Yoon
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea
| | - Cheolkyu Han
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea; Department of Medical and Bioscience, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Jeongbeob Seo
- BiSiChem, 3F, Pangyo-ro, 255 beon-gil 74, Bundang-gu, Seongnam 13486, Republic of Korea.
| | - Jaesook Yun
- Department of Chemistry, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
13
|
Chen G, Xu H, Xu T, Ding W, Zhang G, Hua Y, Wu Y, Han X, Xie L, Liu B, Zhou Y. Calycosin reduces myocardial fibrosis and improves cardiac function in post-myocardial infarction mice by suppressing TGFBR1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154277. [PMID: 35752078 DOI: 10.1016/j.phymed.2022.154277] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Excessive myocardial fibrosis is the pathological basis of heart failure following myocardial infarction (MI). Although calycosin improves cardiac function, its effect on cardiac fibrosis and cardiac function after MI in mice and its precise mechanism remain unclear. PURPOSE Here, we firstly investigated the effects of calycosin on cardiac fibrosis and ventricular function in mice after MI and the role of transforming growth factor-beta receptor 1 (TGFBR1) signaling in the amelioration of cardiac fibrosis and ventricular function. METHODS In vivo effects of calycosin on cardiac structure and function in mice with MI induced by left anterior descending coronary artery ligation were determined by hematoxylin and eosin staining, Masson trichrome staining, and echocardiography. The molecular mechanism of the interaction between TGFBR1 and calycosin was investigated using molecular docking, molecular dynamics (MD) simulation, surface plasmon resonance imaging (SPRi), immunohistochemistry, and western blotting (WB). Subsequently, cardiac-specific Tgfbr1 knockout mice were used to verify the effects of calycosin. The effect of calycosin on primary cardiac fibroblasts (CFs) proliferation and collagen deposition was detected using cell counting (CCK-8), EdU assay, and WB in vitro. CFs infected with an adenovirus that encodes TGFBR1 were used to verify the effects of calycosin. RESULTS In vivo, calycosin attenuated myocardial fibrosis and cardiac dysfunction following MI in a dose-dependent pattern. Calycosin-TGFBR1 complex was found to have a binding energy of -9.04 kcal/mol based on molecular docking. In addition, calycosin bound steadily in the cavity of TGFBR1 during the MD simulation. Based on SPRi results, the solution equilibrium dissociation constant for calycosin and TGFBR1 was 5.11 × 10-5 M. Calycosin inhibited the expression of TGFBR1, Smad2/3, collagen I, and collagen III. The deletion of TGFBR1 partially counteracted these effects. In vitro, calycosin suppressed CFs proliferation and collagen deposition after TGF-β1 stimulation by suppressing the TGFBR1 signaling pathway. The suppressive effects of calycosin were partially rescued by overexpression of TGFBR1. CONCLUSION Calycosin attenuates myocardial fibrosis and cardiac dysfunction following MI in mice in vivo via suppressing the TGFBR1 signaling pathway. Calycosin suppresses CFs proliferation and collagen deposition induced by TGF-β1 via inhibition of the TGFBR1 signaling pathway in vitro.
Collapse
Affiliation(s)
- Guanghong Chen
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Honglin Xu
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Tong Xu
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Wenjun Ding
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China
| | - Guoyong Zhang
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Yue Hua
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Yuting Wu
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Xin Han
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Lingpeng Xie
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Bin Liu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China.
| | - Yingchun Zhou
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
14
|
Yang L, Xu WB, Sun L, Zhang C, Jin CH. SAR analysis of heterocyclic compounds with monocyclic and bicyclic structures as antifungal agents. ChemMedChem 2022; 17:e202200221. [PMID: 35475328 DOI: 10.1002/cmdc.202200221] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Indexed: 11/12/2022]
Abstract
Infections caused by eukaryotic organisms, such as fungi, are generally more difficult to treat than bacterial infections. With the widespread use of antifungal drugs in humans and plants, resistance and toxicity have emerged. Therefore, it is desirable to develop new antifungal drugs with low toxicity that are not susceptible to the development of resistance. This review presents a summary of the past 2017 to 2021 years of research on heterocyclic compounds as antifungal agents for use in humans and plants, focusing on the structure-activity relationships (SAR) of these compounds. This review may provide ideas and data for designing and developing new antifungal drugs with fewer side effects compared with currently available drugs.
Collapse
Affiliation(s)
- Liu Yang
- Yanbian University, College of Pharmacy, CHINA
| | - Wen Bo Xu
- Yanbian University, College of Pharmacy, CHINA
| | | | | | - Cheng Hua Jin
- Yanbian University, College of Pharmacy, Gongyuan, 133002, Yanji, CHINA
| |
Collapse
|