1
|
Kouider Amar M, Moussa H, Hentabli M. Predicting the anticancer activity of indole derivatives: A novel GP-tree-based QSAR model optimized by ALO with insights from molecular docking and decision-making methods. Comput Biol Med 2025; 189:109988. [PMID: 40058079 DOI: 10.1016/j.compbiomed.2025.109988] [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: 08/31/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Indole derivatives have demonstrated significant potential as anticancer agents; however, the complexity of their structure-activity relationships and the high dimensionality of molecular descriptors present challenges in the drug discovery process. This study addresses these challenges by introducing a modified GP-Tree feature selection algorithm specifically designed for regression tasks and high-dimensional feature spaces. The algorithm effectively identifies relevant descriptors for predicting LogIC50 values, the target variable. Furthermore, the GP-Tree method adeptly balances the selection of both positively and negatively contributing descriptors, enhancing the performance of DT, k-NN, and RF models. Additionally, the SMOGN technique was employed to address class imbalances, expanding the dataset to 1381 instances and enhancing the accuracy of IC50 predictions. Various machine learning models were optimized using probabilistic and nature-inspired algorithms, with the Ant Lion Optimizer (ALO) demonstrating the highest efficacy. The AdaBoost-ALO (ADB-ALO) model outperformed all other models, such as MLR, SVR, ANN, k-NN, DT, XGBoost, and RF, achieving an R2 of 0.9852 across the entire dataset, an RMSE of 0.1470, and a high CCC of 0.9925. SHAP analysis revealed critical descriptors, such as TopoPSA and electronic properties, which are essential for potent anticancer activity. Furthermore, molecular docking, in conjunction with the Weighted Sum Method (WSM), identified promising candidates, particularly N-amide derivatives of indole-benzimidazole-isoxazoles, which exhibit dual inhibition against topoisomerase I and topoisomerase II enzymes. Consequently, this research integrates computational predictions with experimental insights to accelerate the discovery of novel anticancer therapies through the accurate prediction and interpretation of the anti-prostate cancer activity of indole derivatives.
Collapse
Affiliation(s)
- Mohamed Kouider Amar
- Biomaterials and Transport Phenomena Laboratory, Faculty of Technology, University Yahia Fares of Medea, 26000, Medea, Algeria; Laboratory of Quality Control, Physico-Chemical Department, SAIDAL of Medea, Medea, Algeria.
| | - Hamza Moussa
- Département des Sciences Biologiques, Faculté des Sciences de La Nature et de La Vie et des Sciences de La Terre, Université de Bouira, 10000, Bouira, Algeria
| | - Mohamed Hentabli
- Biomaterials and Transport Phenomena Laboratory, Faculty of Technology, University Yahia Fares of Medea, 26000, Medea, Algeria; Laboratory of Quality Control, Physico-Chemical Department, SAIDAL of Medea, Medea, Algeria
| |
Collapse
|
2
|
Sheng X, Yang L, Han JY, Yu X, Cui HL. CuI-Catalyzed Dearomatization/Peroxidation/Cyclization Cascade of Pyrrole-Tethered Indoles. J Org Chem 2025; 90:3639-3652. [PMID: 40014766 DOI: 10.1021/acs.joc.4c02977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
A mild CuI-catalyzed dearomatization/peroxidation/cyclization cascade of pyrrole-tethered indoles has been reached, providing peroxide-incorporated indolizino[8,7-b]indole derivatives in acceptable to good yields (46-76%). Dehydrogenated peroxide can be obtained by the use of a FeCl3/TBHP (tBuOOH)/2,2,2-trifluoroethanol (TFE) system at 50 °C.
Collapse
Affiliation(s)
- Xue Sheng
- Laboratory of Asymmetric Synthesis, College of Chemistry and Environmental Engineering, Chongqing University of Arts and Sciences, 319 Honghe Avenue, Yongchuan, Chongqing 402160, P. R. China
| | - Liu Yang
- Laboratory of Asymmetric Synthesis, College of Chemistry and Environmental Engineering, Chongqing University of Arts and Sciences, 319 Honghe Avenue, Yongchuan, Chongqing 402160, P. R. China
| | - Jia-Yi Han
- Laboratory of Asymmetric Synthesis, College of Chemistry and Environmental Engineering, Chongqing University of Arts and Sciences, 319 Honghe Avenue, Yongchuan, Chongqing 402160, P. R. China
| | - Xin Yu
- Laboratory of Asymmetric Synthesis, College of Chemistry and Environmental Engineering, Chongqing University of Arts and Sciences, 319 Honghe Avenue, Yongchuan, Chongqing 402160, P. R. China
| | - Hai-Lei Cui
- Laboratory of Asymmetric Synthesis, College of Chemistry and Environmental Engineering, Chongqing University of Arts and Sciences, 319 Honghe Avenue, Yongchuan, Chongqing 402160, P. R. China
| |
Collapse
|
3
|
Bakchi B, Devabattula G, Maddipatla S, Singampalli A, Kumar Porna D, Nanduri S, Sharma A, Godugu C, Yaddanapudi VM. New Indole-Based Phenylthiazolyl-2,4-dihydropyrazolones as Tubulin polymerization inhibitors: Multicomponent Synthesis, Cytotoxicity Evaluation, and in silico Studies. ChemMedChem 2025; 20:e202400817. [PMID: 39523484 DOI: 10.1002/cmdc.202400817] [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: 10/17/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
A facile multicomponent synthesis of new indole-based phenylthiazolyl-dihydropyrazolone hybrids, their structural characterization, biological evaluation, and in silico investigations as anticancer agents are reported. Lead molecule 5 i of the series showed potent activity against MCF-7 breast cancer cells with an IC50 of 3.92±0.01 μM while showing minimal toxicity to normal human lung cells (IC50=69.85±3.95 μM). Further studies show that the compound exhibits antiproliferative activity by inducing apoptosis in MCF-7 cancer cells. The wound healing assay indicated impaired cell migration under the concentration-dependent dosage. The lead molecule 5 i also successfully inhibited the tubulin polymerase enzyme with an IC50 of 4.16±0.18 μM. A flow cytometric assay indicated compound 5 i induced apoptosis through G0 phase cell cycle arrest. The binding mode and interactions of the compound with the tubulin were predicted by molecular modelling and calculating binding free energies. These findings explain the current series as a new class of microtubule polymerization inhibitors with anticancer activity suitable for developing anticancer agents targeting tubulin.
Collapse
Affiliation(s)
- Bulti Bakchi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Geetanjali Devabattula
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Sarvan Maddipatla
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Anuradha Singampalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Dileep Kumar Porna
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| |
Collapse
|
4
|
Liu G, Lin W, Zhang K, Chen K, Niu G, Zhu Y, Liu Y, Li P, Li Z, An Y. Elucidating the prognostic and therapeutic significance of TOP2A in various malignancies. Cancer Genet 2024; 288-289:68-81. [PMID: 39454521 DOI: 10.1016/j.cancergen.2024.10.005] [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: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Topoisomerase IIα (TOP2A) is a crucial enzyme that plays a vital role in DNA replication and transcription mechanisms. Dysregulated expression of TOP2A has been associated with various malignancies, including hepatocellular carcinoma, prostate cancer, colon cancer, lung cancer and breast cancer. In this review, we summarized the prognostic relevances of TOP2A in various types of cancer. The increased expression of TOP2A has been linked to resistance to therapy and reduced survival rates. Therefore, evaluating TOP2A levels could assist in identifying patients who may derive advantages from molecular targeted therapy. The amplification of TOP2A has been linked to a positive response to chemotherapy regimens that contain anthracycline. Nevertheless, the overexpression of TOP2A also indicates a heightened likelihood of disease recurrence and unfavorable prognosis. The prognostic significance of TOP2A has been extensively studied in various types of cancer. The increased expression of TOP2A is associated with poor clinical outcomes, indicating its potential as a valuable biomarker for assessing risk and stratifying treatment in these malignancies. However, further investigation is needed to elucidate the underlying mechanisms by which TOP2A influences cancer progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Wenlong Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kangxu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Guanglin Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
5
|
Lin J, Han Y, Li B, Gai W, Wang Z, Wang Q, Teng Y, Li J, Li D. Synthesis and biological evaluation of novel penindolone derivatives as potential antiproliferative agents against SCLC in vitro. Bioorg Med Chem Lett 2024; 110:129877. [PMID: 38964518 DOI: 10.1016/j.bmcl.2024.129877] [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: 04/22/2024] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Small cell lung cancer (SCLC) keeps on the leading cause of cancer mortality world widely, while there is lack of efficient therapeutic drugs especially for the resistant ones. In this work, a compound named penindolone (PND) with new skeleton was found to show weak inhibitory effect (IC50 = 42.5 µM) on H69AR cells (SCLC, adriamycin-resistant) proliferation by screening our in-house compound library. With the aim of improving its low potency, a series of PND derivatives were synthesized and biologically evaluated by the Sulforhodamine B (SRB) assay. Among all tested derivatives, compound 5h possessed higher antiproliferation potency (IC50 = 1.6 µM). Furthermore, preliminary mechanism investigation revealed that 5h was able to induce apoptosis and arrest the cell cycle at G0/G1 phase. These findings suggest that this novel skeleton has expanded the anti-SCLC compound reservoir and provided a new drug lead.
Collapse
Affiliation(s)
- Jiaqi Lin
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Yongqing Han
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Bohan Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Wenrui Gai
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Zhengjie Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Qi Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Yueling Teng
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Sanya Oceanographic Institute, Ocean University of China, Qingdao 266003/Sanya 572025, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China.
| |
Collapse
|
6
|
Santos MB, de Azevedo Teotônio Cavalcanti M, de Medeiros E Silva YMS, Dos Santos Nascimento IJ, de Moura RO. Overview of the New Bioactive Heterocycles as Targeting Topoisomerase Inhibitors Useful Against Colon Cancer. Anticancer Agents Med Chem 2024; 24:236-262. [PMID: 38038012 DOI: 10.2174/0118715206269722231121173311] [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: 07/21/2023] [Revised: 10/14/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer globally, with high mortality. Metastatic CRC is incurable in most cases, and multiple drug therapy can increase patients' life expectancy by 2 to 3 years. Efforts are being made to understand the relationship between topoisomerase enzymes and colorectal cancer. Some studies have shown that higher expression of these enzymes is correlated to a poor prognosis for this type of cancer. One of the primary drugs used in the treatment of CRC is Irinotecan, which can be used in monotherapy or, more commonly, in therapeutic schemes such as FOLFIRI (Fluorouracil, Leucovorin, and Irinotecan) and CAPIRI (Capecitabine and Irinotecan). Like Camptothecin, Irinotecan and other compounds have a mechanism of action based on the formation of a ternary complex with topoisomerase I and DNA providing damage to it, therefore leading to cell death. Thus, this review focused on the principal works published in the last ten years that demonstrate a correlation between the inhibition of different isoforms of topoisomerase and in vitro cytotoxic activity against CRC by natural products, semisynthetic and synthetic compounds of pyridine, quinoline, acridine, imidazoles, indoles, and metal complexes. The results revealed that natural compounds, semisynthetic and synthetic derivatives showed potential in vitro cytotoxicity against several colon cancer cell lines, and this activity was often accompanied by the ability to inhibit both isoforms of topoisomerase (I and II), highlighting that these enzymes can be promising targets for the development of new chemotherapy against CRC. Pyridine analogs were considered the most promising for this study, while the evaluation of the real potential of natural products was limited by the lack of information in their work. Moreover, the complexes, although promising, presented as the main limitation the lack of selectivity.
Collapse
Affiliation(s)
- Mirelly Barbosa Santos
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Misael de Azevedo Teotônio Cavalcanti
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Yvnni Maria Sales de Medeiros E Silva
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Igor José Dos Santos Nascimento
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Departament of Pharmacy, Cesmac University Center, Maceió, Brazil
| | - Ricardo Olimpio de Moura
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| |
Collapse
|
7
|
Zhao W, Zheng XD, Tang PYZ, Li HM, Liu X, Zhong JJ, Tang YJ. Advances of antitumor drug discovery in traditional Chinese medicine and natural active products by using multi-active components combination. Med Res Rev 2023; 43:1778-1808. [PMID: 37183170 DOI: 10.1002/med.21963] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
The antitumor efficacy of Chinese herbal medicines has been widely recognized. Leading compounds such as sterols, glycosides, flavonoids, alkaloids, terpenoids, phenylpropanoids, and polyketides constitute their complex active components. The antitumor monomers derived from Chinese medicine possess an attractive anticancer activity. However, their use was limited by low bioavailability, significant toxicity, and side effects, hindering their clinical applications. Recently, new chemical entities have been designed and synthesized by combining natural drugs with other small drug molecules or active moieties to improve the antitumor activity and selectivity, and reduce side effects. Such a novel conjugated drug that can interact with several vital biological targets in cells may have a more significant or synergistic anticancer activity than a single-molecule drug. In addition, antitumor conjugates could be obtained by combining pharmacophores containing two or more known drugs or leading compounds. Based on these studies, the new drug research and development could be greatly shortened. This study reviews the research progress of conjugates with antitumor activity based on Chinese herbal medicine. It is expected to serve as a valuable reference to antitumor drug research and clinical application of traditional Chinese medicine.
Collapse
Affiliation(s)
- Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiao-Di Zheng
- Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | | | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xue Liu
- Jinan Intellectual Property Protection Center, Jinan, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
8
|
Vazquez DR, Munoz Forti K, Figueroa Rosado MM, Gutierrez Mirabal PI, Suarez-Martinez E, Castro-Rosario ME. Effect of CaS Nanostructures in the Proliferation of Human Breast Cancer and Benign Cells In Vitro. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:10494. [PMID: 37124318 PMCID: PMC10137321 DOI: 10.3390/app122010494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
We report on the effect of naked CaS nanostructures on the proliferation of carcinoma cancer cells and normal fibroblasts in vitro. The CaS nanostructures were prepared via the microwave-mediated decomposition of dimethyl sulfoxide (DMSO) in the presence of calcium acetate Ca ( CH 3 CO 2 ) 2 . Light scattering measurements revealed that dispersions contain CaS nanostructures in the size range of a few Å to about 1 nanometer, and are formed when DMSO is decomposed in the presence of Ca ( CH 3 CO 2 ) 2 . Theoretical calculations at the DFT/B3LYP/DGDZVP level of theory on ( C a S ) n clusters ( n = 1 , 2 , 3 , and 4) are consistent with clusters in this size range. The absorption spectra of the CaS nanostructures are dominated by strong bands in the UV, as well as weaker absorption bands in the visible. We found that a single dose of CaS nanoclusters smaller than 0.8 nm in diameter does not affect the survival and growth rate of normal fibroblasts and inhibits the proliferation rate of carcinoma cells in vitro. Larger CaS nanostructures, approximately (1.1 ± 0.2) nm in diameter, have a similar effect on carcinoma cell proliferation and survival rate. The CaS nanoclusters have little effect on the normal fibroblast cell cycle. Human carcinoma cells treated with CaS nanocluster dispersion exhibited a decreased ability to properly enter the cell cycle, marked by a decrease in cell concentration in the G0/G1 phase in the first 24 h and an increase in cells held in the SubG1 and G0/G1 phases up to 72 h post-treatment. Apoptosis and necrotic channels were found to play significant roles in the death of human carcinoma exposed to the CaS nanoclusters. In contrast, any effect on normal fibroblasts appeared to be short-lived and non-detrimental. The interaction of CaS with several functional groups was further investigated using theoretical calculations. CaS is predicted to interact with thiol ( R-SH ), hydroxide ( R - OH ), amino ( R - NH 2 ), carboxylic acid ( R - COOH ), ammonium ( R-NH 3 + ), and carboxylate ( R-COO - ) functional groups. None of these interactions are predicted to result in the dissociation of CaS. Thermodynamic considerations, on the other hand, are consistent with the dissociation of CaS into Ca 2 + ions and H 2 S in acidic media, both of which are known to cause apoptosis or cell death. Passive uptake and extracellular pH values of carcinoma cells are proposed to result in the observed selectivity of CaS to inhibit cancer cell proliferation with no significant effect on normal fibroblast cells. The results encourage further research with other cell lines in vitro as well as in vivo to translate this nanotechnology into clinical use.
Collapse
Affiliation(s)
- Daniel Rivera Vazquez
- School of Biological and Physical Sciences, Northwestern State University, Natchitoches, LA 71457, USA
- Department of Chemistry, The University of Puerto Rico at Mayaguez, Mayaguez 00680, Puerto Rico, USA
| | - Kevin Munoz Forti
- Department of Biology, The University of Puerto Rico at Ponce, Ponce 00716, Puerto Rico, USA
| | | | - Pura I. Gutierrez Mirabal
- Department of Chemistry, The University of Puerto Rico at Mayaguez, Mayaguez 00680, Puerto Rico, USA
| | - Edu Suarez-Martinez
- Department of Biology, The University of Puerto Rico at Ponce, Ponce 00716, Puerto Rico, USA
| | - Miguel E. Castro-Rosario
- Department of Chemistry, The University of Puerto Rico at Mayaguez, Mayaguez 00680, Puerto Rico, USA
- Correspondence:
| |
Collapse
|
9
|
Mahmoud E, Hayallah AM, Kovacic S, Abdelhamid D, Abdel-Aziz M. Recent progress in biologically active indole hybrids: a mini review. Pharmacol Rep 2022; 74:570-582. [PMID: 35594012 DOI: 10.1007/s43440-022-00370-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 01/06/2023]
Abstract
The indole moiety is one of the most widespread heterocycles found in both natural products and biological systems. Indoles have important biological activities including anticancer, antioxidant, anti-inflammatory, antifungal, anticholinesterase, and antibacterial properties. Scientists are therefore interested in the synthesis of biologically active indole-based hybrids such as indole-coumarin, indole-chalcone, indole-isatin, indole-pyrimidine and so on, with the aim of improving activity, selectivity, and mitigating side effects. This review will discuss the newly synthesized indole-based hybrids along with their biological activity which will be useful in drug discovery and development.
Collapse
Affiliation(s)
- Esraa Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Alaa M Hayallah
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sphinx University, New-Assiut, Egypt
| | - Suzana Kovacic
- Department of Chemistry, Simon Fraser University, British Columbia, Canada
| | - Dalia Abdelhamid
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Mohamad Abdel-Aziz
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
10
|
Synthesis and biological evaluation of xanthone derivatives as anti-cancer agents targeting topoisomerase II and DNA. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02862-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
11
|
Luo H, Chen M, Tian F, He Y, Liu X, Li J. An Efficient Method for the Synthesis of N-Aryl Substituted C1-Functionalized 1,2,3,4-Tetrahydroisoquinolines. HETEROCYCLES 2022. [DOI: 10.3987/com-22-14697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
12
|
Misra SK, Pathak D, Pathak K. Anticancer potential of indole derivatives: an update. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2021-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The heterocyclic indole is one of the most prevalent pharmacophores in nature. It has been a highly privileged scaffold for designing targeted and anticancer therapeutics. Countless fused heterocyclic templates have been developed with diverse physicochemical and biological properties. Due to their versatile ethanobotanical and pharmacological values, indole and its derivatives seek high demand in the chemical and healthcare sectors. Extensive anticancer research has been conducted in this decade to evaluate their efficacy for diverse malignancies. The chapter explores the anticancer activity of natural and synthetic indole derivatives expressed through targeting different biological receptors and enzymes.
Collapse
Affiliation(s)
- Shashi Kiran Misra
- University Institute of Pharmacy, Chhatrapati Shahu Ji Maharaj University , Kanpur , 208026 , India
| | - Devender Pathak
- Faculty of Pharmacy , Uttar Pradesh University of Medical Sciences , Saifai , Etawah , 206130 , Uttar Pradesh , India
| | - Kamla Pathak
- Faculty of Pharmacy , Uttar Pradesh University of Medical Sciences , Saifai , Etawah , 206130 , Uttar Pradesh , India
| |
Collapse
|
13
|
Song F, Bian Y, Liu J, Li Z, Zhao L, Fang J, Lai Y, Zhou M. Indole Alkaloids, Synthetic Dimers and Hybrids with Potential In Vivo Anticancer Activity. Curr Top Med Chem 2021; 21:377-403. [PMID: 32901583 DOI: 10.2174/1568026620666200908162311] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022]
Abstract
Indole, a heterocyclic organic compound, is one of the most promising heterocycles found in natural and synthetic sources since its derivatives possess fascinating structural diversity and various therapeutic properties. Indole alkaloids, synthetic dimers and hybrids could act on diverse targets in cancer cells, and consequently, possess potential antiproliferative effects on various cancers both in vitro and in vivo. Vinblastine, midostaurin, and anlotinib as the representative of indole alkaloids, synthetic dimers and hybrids respectively, have already been clinically applied to treat many types of cancers, demonstrating indole alkaloids, synthetic dimers and hybrids are useful scaffolds for the development of novel anticancer agents. Covering articles published between 2010 and 2020, this review emphasizes the recent development of indole alkaloids, synthetic dimers and hybrids with potential in vivo therapeutic application for cancers.
Collapse
Affiliation(s)
- Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Yunqiang Bian
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Jing Liu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Zhenghua Li
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Li Zhao
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Junman Fang
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Yonghong Lai
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Meng Zhou
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| |
Collapse
|
14
|
A comprehensive overview of β-carbolines and its derivatives as anticancer agents. Eur J Med Chem 2021; 224:113688. [PMID: 34332400 DOI: 10.1016/j.ejmech.2021.113688] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/05/2021] [Accepted: 07/04/2021] [Indexed: 01/13/2023]
Abstract
β-Carboline alkaloids are a family of natural and synthetic products with structural diversity and outstanding antitumor activities. This review summarizes research developments of β-carboline and its derivatives as anticancer agents, which focused on both natural and synthetic monomers as well as dimers. In addition, the structure-activity relationship (SAR) analysis of β-carboline monomers and dimers are summarized and mechanism of action of β-carboline and its derivatives are also presented. A few possible research directions, suggestions and clues for future work on the development of novel β-carboline-based anticancer agents with improved expected activities and lesser toxicity are also provided.
Collapse
|
15
|
Chen X, Wang L, Su X, Luo SY, Tang X, Huang Y. Identification of potential target genes and crucial pathways in small cell lung cancer based on bioinformatic strategy and human samples. PLoS One 2020; 15:e0242194. [PMID: 33186389 PMCID: PMC7665632 DOI: 10.1371/journal.pone.0242194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Small cell lung cancer (SCLC) is a carcinoma of the lungs with strong invasion, poor prognosis and resistant to multiple chemotherapeutic drugs. It has posed severe challenges for the effective treatment of lung cancer. Therefore, searching for genes related to the development and prognosis of SCLC and uncovering their underlying molecular mechanisms are urgent problems to be resolved. This study is aimed at exploring the potential pathogenic and prognostic crucial genes and key pathways of SCLC via bioinformatic analysis of public datasets. Firstly, 117 SCLC samples and 51 normal lung samples were collected and analyzed from three gene expression datasets. Then, 102 up-regulated and 106 down-regulated differentially expressed genes (DEGs) were observed. And then, functional annotation and pathway enrichment analyzes of DEGs was performed utilizing the FunRich. The protein-protein interaction (PPI) network of the DEGs was constructed through the STRING website, visualized by Cytoscape. Finally, the expression levels of eight hub genes were confirmed in Oncomine database and human samples from SCLC patients. It showed that CDC20, BUB1, TOP2A, RRM2, CCNA2, UBE2C, MAD2L1, and BUB1B were upregulated in SCLC tissues compared to paired adjacent non-cancerous tissues. These suggested that eight hub genes might be viewed as new biomarkers for prognosis of SCLC or to guide individualized medication for the therapy of SCLC.
Collapse
Affiliation(s)
- Xiuwen Chen
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Li Wang
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Xiaomin Su
- Department of Immunology, Nankai University School of Medicine, Tianjin, China
| | - Sen-yuan Luo
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Xianbin Tang
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Yugang Huang
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Hubei, China
- * E-mail:
| |
Collapse
|
16
|
Design, synthesis and antitumour evaluation of pyrrolo[1,2-f]-phenanthridine and dibenzo[f,h]pyrrolo[1,2-b]isoquinoline derivatives. Eur J Med Chem 2020; 202:112516. [PMID: 32622270 DOI: 10.1016/j.ejmech.2020.112516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 11/22/2022]
Abstract
A series of 1,2-bis(hydroxymethyl)pyrrolo[1,2-f]phenanthridine derivatives and their alkyl (ethyl and isopropyl) carbamates and 12,13-bis(hydroxymethyl)-9,14-dihydro-dibenzo[f,h]pyrrolo[1,2-b]isoquinoline derivatives were synthesized for antiproliferative evaluation. The preliminary antitumour studies revealed that these two types of bis(hydroxymethyl) derivatives showed significant antitumour activities and were able to inhibit the growth of various human tumour cell lines in vitro. Several of the derivatives were demonstrated to cause DNA interstrand cross-links by an alkaline agarose gel shifting assay. These conjugates were cytotoxic to a variety of cancer cell lines by inducing DNA damage, delaying cell cycle progression in the G2/M phase and triggering apoptosis. Compound 21a, dissolved in a vehicle suitable for intravenous administration, was selected for antitumour studies in animal models. We demonstrated that at a dose that did not cause body weight loss in mice, compound 21a could significantly suppress the growth of tumour xenografts of human lung cancer H460 and colorectal cancer HCT-116 cells in nude mice. Our present results confirm the antitumour activities of these conjugates.
Collapse
|
17
|
Jia Y, Wen X, Gong Y, Wang X. Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur J Med Chem 2020; 200:112359. [PMID: 32531682 DOI: 10.1016/j.ejmech.2020.112359] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
Cancer chemotherapy is frequently hampered by drug resistance, so the resistance to anticancer agents represents one of the major obstacles for the effective cancer treatment. Indole derivatives have the potential to act on diverse targets in cancer cells and exhibit promising activity against drug-resistant cancers. Moreover, some indole-containing compounds such as Semaxanib, Sunitinib, Vinorelbine, and Vinblastine have already been applied in clinics for various kinds of cancer even drug-resistant cancer therapy. Thus, indole derivatives are one of significant resources for the development of novel anti-drug-resistant cancer agents. This review focuses on the recent development of indole derivatives with potential therapeutic application for drug-resistant cancers, and the mechanisms of action, the critical aspects of design as well as structure-activity relationships, covering articles published from 2010 to 2020.
Collapse
Affiliation(s)
- Yanshu Jia
- Chongqing Institute of Engineering, Chongqing, 400056, China
| | - Xiaoyue Wen
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang, Hubei, 443000, China
| | - Yufeng Gong
- The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Xuefeng Wang
- Department of Surgery, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province, 311800, China.
| |
Collapse
|
18
|
Dhuguru J, Skouta R. Role of Indole Scaffolds as Pharmacophores in the Development of Anti-Lung Cancer Agents. Molecules 2020; 25:E1615. [PMID: 32244744 PMCID: PMC7181244 DOI: 10.3390/molecules25071615] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of death in men and women worldwide, affecting millions of people. Between the two types of lung cancers, non-small cell lung cancer (NSCLC) is more common than small cell lung cancer (SCLC). Besides surgery and radiotherapy, chemotherapy is the most important method of treatment for lung cancer. Indole scaffold is considered one of the most privileged scaffolds in heterocyclic chemistry. Indole may serve as an effective probe for the development of new drug candidates against challenging diseases, including lung cancer. In this review, we will focus on discussing the existing indole based pharmacophores in the clinical and pre-clinical stages of development against lung cancer, along with the synthesis of some of the selected anti-lung cancer drugs. Moreover, the basic mechanism of action underlying indole based anti-lung cancer treatment, such as protein kinase inhibition, histone deacetylase inhibition, DNA topoisomerase inhibition, and tubulin inhibition will also be discussed.
Collapse
Affiliation(s)
| | - Rachid Skouta
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA;
| |
Collapse
|
19
|
Indole: A privileged scaffold for the design of anti-cancer agents. Eur J Med Chem 2019; 183:111691. [DOI: 10.1016/j.ejmech.2019.111691] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/21/2022]
|
20
|
Kumari A, Singh RK. Medicinal chemistry of indole derivatives: Current to future therapeutic prospectives. Bioorg Chem 2019; 89:103021. [PMID: 31176854 DOI: 10.1016/j.bioorg.2019.103021] [Citation(s) in RCA: 303] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Indole is a versatile pharmacophore, a privileged scaffold and an outstanding heterocyclic compound with wide ranges of pharmacological activities due to different mechanisms of action. It is an superlative moiety in drug discovery with the sole property of resembling different structures of the protein. Plenty of research has been taking place in recent years to synthesize and explore the various therapeutic prospectives of this moiety. This review summarizes some of the recent effective chemical synthesis (2014-2018) for indole ring. This review also emphasized on the structure-activity relationship (SAR) to reveal the active pharmacophores of various indole analogues accountable for anticancer, anticonvulsant, antimicrobial, antitubercular, antimalarial, antiviral, antidiabetic and other miscellaneous activities which have been investigated in the last five years. The precise features with motives and framework of each research topic is introduced for helping the medicinal chemists to understand the perspective of the context in a better way. This review will definitely offer the platform for researchers to strategically design diverse novel indole derivatives having different promising pharmacological activities with reduced toxicity and side effects.
Collapse
Affiliation(s)
- Archana Kumari
- Rayat-Bahra Institute of Pharmacy, Dist. Hoshiarpur, 146104 Punjab, India
| | - Rajesh K Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126 Punjab, India.
| |
Collapse
|
21
|
Chang SM, Jain V, Chen TL, Patel AS, Pidugu HB, Lin YW, Wu MH, Huang JR, Wu HC, Shah A, Su TL, Lee TC. Design and Synthesis of 1,2-Bis(hydroxymethyl)pyrrolo[2,1-a]phthalazine Hybrids as Potent Anticancer Agents that Inhibit Angiogenesis and Induce DNA Interstrand Cross-links. J Med Chem 2019; 62:2404-2418. [DOI: 10.1021/acs.jmedchem.8b01689] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Anamik Shah
- Center of Excellence in Drug Discovery, Saurashtra University, Rajkot 360005, India
| | | | | |
Collapse
|
22
|
|
23
|
Prasad SS, Reddy NR, Baskaran S. One-Pot Synthesis of Structurally Diverse Iminosugar-Based Hybrid Molecules. J Org Chem 2018; 83:9604-9618. [DOI: 10.1021/acs.joc.8b00748] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Sure Siva Prasad
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | | | - Sundarababu Baskaran
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
24
|
Sun J, Jiang W, Yan CG. Convenient construction of tetrahydrochromeno[4',3':2,3]indolizino[8,7- b]indoles and tetrahydroindolizino[8,7- b]indoles via one-pot domino reaction. RSC Adv 2018; 8:28736-28744. [PMID: 35542494 PMCID: PMC9084333 DOI: 10.1039/c8ra05138k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/06/2018] [Indexed: 12/22/2022] Open
Abstract
The functionalized tetrahydrochromeno[4',3':2,3]indolizino[8,7-b]indoles were conveniently synthesized in high yields by one-pot domino reaction of tryptamines, alkyl propiolates and 2-aryl-3-nitro-2H-chromenes. Under similar conditions, the one-pot reaction of tryptamines, alkyl propiolates and β-nitroalkenes resulted in functionalized tetrahydroindolizino[8,7-b]indoles. The reaction mechanism involved sequential generation of β-enamino ester, Michael addition, Pictet-Spengler reaction and annulation process. The reaction showed high atomic economy and met the goals of sustainable chemistry.
Collapse
Affiliation(s)
- Jing Sun
- College of Chemistry & Chemical Engineering, Yangzhou University Yangzhou 225002 China +86-514-87975244 +86-514-87975531
| | - Wang Jiang
- College of Chemistry & Chemical Engineering, Yangzhou University Yangzhou 225002 China +86-514-87975244 +86-514-87975531
| | - Chao-Guo Yan
- College of Chemistry & Chemical Engineering, Yangzhou University Yangzhou 225002 China +86-514-87975244 +86-514-87975531
| |
Collapse
|
25
|
Indole in the target-based design of anticancer agents: A versatile scaffold with diverse mechanisms. Eur J Med Chem 2018; 150:9-29. [DOI: 10.1016/j.ejmech.2018.02.065] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/13/2018] [Accepted: 02/20/2018] [Indexed: 12/25/2022]
|
26
|
de Almeida SMV, Ribeiro AG, de Lima Silva GC, Ferreira Alves JE, Beltrão EIC, de Oliveira JF, de Carvalho LB, Alves de Lima MDC. DNA binding and Topoisomerase inhibition: How can these mechanisms be explored to design more specific anticancer agents? Biomed Pharmacother 2017; 96:1538-1556. [DOI: 10.1016/j.biopha.2017.11.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
|