1
|
Al-Karmalawy AA, Zeidan MA, Elmaaty AA, Sharaky M, Yassen ASA, Khaleel EF, Eldehna WM, Ashour HF. Design and synthesis of new 1,2,3-triazole derivatives as VEGFR-2/telomerase downregulatory candidates endowed with apoptotic potential for cancer treatment. Bioorg Chem 2025; 156:108159. [PMID: 39823817 DOI: 10.1016/j.bioorg.2025.108159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/15/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
In this current work, we dedicated efforts to designing and synthesizing new 1,2,3-triazole-analogues (5a-d), (6a-d), and (7a-c) to act as dual VEGFR-2 and telomerase inhibitors with promising apoptotic potential. The synthesized analogues were examined against eleven diverse types of cancer cells and two normal cells to assess their ability to inhibit cell growth (GI%). Obviously, compound 7b showed the best average GI% (75.69 %) surpassing the average GI% of Dox (65.79 %). Compound 5d showed the lowest IC50 values (25.86 and 51.91 µM) against HNO-97 and FaDu cancer cells, respectively. Besides, compound 5a exhibited the lowest IC50 value (15.46 µM) against HCT116, whereas compound 6b revealed the lowest IC50 value (31.14 µM) against HuH7. Besides, candidates 5a, 5b, 5d, and 7a showed prominent inhibitory results towards VEGFR-2 protein with decreasing its expression by 0.33, 0.42, 0.38 and 0.26-fold change, respectively. However, compounds 5a, 5b, 5d, and 7a showed promising inhibitory results towards telomerase protein and decreased its expression by 0.60, 0.50, 0.52, and 0.44-fold change, respectively. Additionally, it was clear that compound 5a was able to upregulate the expression of Caspases 3, 8, and 9 proteins by 2.19, 1.83, and 1.62-fold change, respectively. Besides, 5a was able to downregulate the expression of CDK-2, CDK-4, and CDK-6 proteins by 0.50, 0.43, and 0.13-fold change, respectively. Obviously, compound 5a halted the cell cycle at the G1, S, and G2-M phases in HCT116 cells. Subsequently, the synthesized 1,2,3-triazole analogues can be treated as lead VEGFR-2 and telomerase inhibitors with potential apoptotic activity for future optimization and cancer treatment.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt.
| | - Mohamed A Zeidan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt; Medicinal Chemistry Department, Clinical Pharmacy Program, East Port Said National University, Port Said 42526, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Asmaa S A Yassen
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt
| | - Heba F Ashour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
| |
Collapse
|
2
|
Zeidan MA, Ashour HF, Yassen ASA, Abo Elmaaty A, Farag AB, Sharaky M, Abdullah Alzahrani AY, Mughram MHA, Al-Karmalawy AA. Dual EGFR and telomerase inhibitory potential of new triazole tethered Schiff bases endowed with apoptosis: design, synthesis, and biological assessments. RSC Med Chem 2024:d4md00750f. [PMID: 39790121 PMCID: PMC11708207 DOI: 10.1039/d4md00750f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Many cancers have displayed resistance to chemotherapeutic drugs over the past few decades. EGFR has emerged as a leading target for cancer therapy via inhibiting tumor angiogenesis. Besides, studies strongly suggest that blocking telomerase activity could be an effective way to control the growth of certain cancer cells. Based on the fact that multi-target design rationale can afford candidates with greater treatment effectiveness. Besides, it was evidenced that inhibition of human telomerase enhances the effect of some tyrosine kinase inhibitors. So, in the current work, we aimed to design and synthesize novel 1,2,3-triazole-tethered Schiff bases (5a-l) to act as dual EGFR and telomerase inhibitors. Growth inhibition (GI)% was conducted for the synthesized compounds using a panel of eleven cancer cell lines as well as two normal cell lines. Interestingly, compound 5e displayed the highest mean GI% (76.78%) among the investigated compounds surpassing the mean GI% of the reference drug doxorubicin (65.79%). In addition, compound 5g displayed notably the lowest IC50 values (13.31, 13.31, 12.62, and 31.19 μM) for the four utilized cancer cell lines HNO97, HCT116, A375, and HEPG2, respectively. Interestingly, the investigated compounds exhibited significant inhibitory potential to EGFR and telomerase protein expression; in particular, compound 5g recorded inhibitory potentials of 3.45 and 1.31 ng mL-1, respectively. Hence, protein expression of the apoptosis-related proteins was carried out for compound 5g. Pro-apoptotic proteins (caspases 3, 8, and 9) were upregulated by 1.35, 1.55, and 1.51-fold change, respectively. Meanwhile, the anti-apoptotic proteins (CDK-2, CDK-4, and CDK-6) were downregulated by 2.91, 2.01, and 9.15-fold change, respectively, ensuring the apoptotic potential of compound 5g. Accordingly, compound 5g was selected for further investigation of its effects on cell cycle progression in A375 cancer cells. Obviously, compound 5g prompted cell cycle arrest at the G0-G1 phase. Additionally, the investigated compounds showed eligible pharmacokinetic profiles with feasible oral bioavailability. Consequently, the synthesized compounds can be treated as lead multi-target anticancer ligands for future optimization.
Collapse
Affiliation(s)
- Mohamed A Zeidan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt New Damietta 34518 Egypt
| | - Heba F Ashour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University New Galala New Galala 43713 Egypt
| | - Asmaa S A Yassen
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University New Galala New Galala 43713 Egypt
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia 41522 Egypt
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University Port Said 42526 Egypt
- Medicinal Chemistry Department, Clinical Pharmacy Program, East Port said National University Port Said 42526 Egypt
| | - Ayman B Farag
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University 6th of October City Giza 12566 Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University Cairo Egypt
| | | | - Mohammed H Al Mughram
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University Abha 61421 Saudi Arabia
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq Baghdad 10023 Iraq
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt New Damietta 34518 Egypt
| |
Collapse
|
3
|
Chilakala NB, Roy A, Kalia NP, Thumma V, Raju B, Etnoori S, Premalatha K. Design, Synthesis, Evaluation of Antitubercular Activity and Insilco Studies of Novel 1,5-Naphthyridin-2(1H)-One Pendent 1,2,3-Triazoles. Chem Biodivers 2024; 21:e202401491. [PMID: 39167045 DOI: 10.1002/cbdv.202401491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
A library of 1,5-Naphthyridin-2(1H)-one based 1,2,3-triazole analogues (11a-q) were synthesized via series of reactions such as protection, oxidation, cyclization and click chemistry. The new molecules were tested for their antitubercular activity against M. tuberculosis mc26230 and determined the minimum inhibitory concentration (MIC) employing Rifampicin as reference. The 3-cyano and 4-cyano substituted analogues 11e and 11f displayed superior activity with an MIC value of 4.0 μg/ml. Additionally, these potent molecules were tested for determination of their MBC values and ATP depletion assay showed a hopeful relative luminescence. Additionally, determined the MIC of 11e and 11f against multi-drug resistant strains of M. tuberculosis viz. mc2 8243, mc2 8247 and mc2 8259. The cytotoxicity of these two molecules presented no effects on normal cell. The profound results of these two molecules proved them as potential antitubercular agent. Further, molecular docking studies were portrayed against crystal structure of M. tuberculosis dihydrofolate reductase which garnered promising docking scores and binding interactions such as H-bond and hydrophobic. ADME prediction revealed their favorable drug-likeness characteristics.
Collapse
Affiliation(s)
| | - Arnab Roy
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500037, India
| | - Nitin Pal Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500037, India
| | - Vishnu Thumma
- Department of Sciences and Humanities, Matrusri Engineering College, Hyderabad, Telangana, 500059, India
| | - B Raju
- Department of Chemistry, Osmania University, Hyderabad, Telangana, 500007, India
| | - Sharada Etnoori
- Department of Chemistry, Osmania University, Hyderabad, Telangana, 500007, India
| | - K Premalatha
- Department of Chemistry, Osmania University, Hyderabad, Telangana, 500007, India
- Telangana Mahila Viswavidyalayam, Hyderabad, Telangana, 500095, India
| |
Collapse
|
4
|
Huang PF, Fu JL, Peng Y, Tang KW, Liu Y. Electrochemical Oxidative (4 + 2) Cyclization of Anilines and o-Phenylenediamines for the Synthesis of Phenazines. Org Lett 2024; 26:3756-3761. [PMID: 38678581 DOI: 10.1021/acs.orglett.4c00851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Phenazines, crucial constituents of nitrogen-containing heterocycles, widely exist in functional compounds. Herein, we report an anodic oxidative (4 + 2) cyclization between anilines and o-phenylenediamines for the uniform construction of phenazines in a simple undivided cell. Dual C-H amination followed by oxidation represents an outstanding step and atom efficiency. A sequence of phenazines is produced with excellent functional group tolerance at room temperature.
Collapse
Affiliation(s)
- Peng-Fei Huang
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China
| | - Jia-Le Fu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China
| | - Ying Peng
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China
| | - Ke-Wen Tang
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China
| | - Yu Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China
| |
Collapse
|
5
|
Yang H, Zhang D, Yuan Z, Qiao H, Xia Z, Cao F, Lu Y, Jiang F. Novel 4-Aryl-4H-chromene derivative displayed excellent in vivo anti-glioblastoma efficacy as the microtubule-targeting agent. Eur J Med Chem 2024; 267:116205. [PMID: 38350361 DOI: 10.1016/j.ejmech.2024.116205] [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: 12/14/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/15/2024]
Abstract
In this study, a series of novel 4-Aryl-4H-chromene derivatives (D1-D31) were designed and synthesized by integrating quinoline heterocycle to crolibulin template molecule based on the strategy of molecular hybridization. One of these compounds D19 displayed positive antiproliferative activity against U87 cancer cell line (IC50 = 0.90 ± 0.03 μM). Compound D19 was verified as the microtubule-targeting agent through downregulating tubulin related genes of U87 cells, destroying the cytoskeleton of tubulins and interacting with the colchicine-binding site to inhibit the polymerization of tubulins by transcriptome analysis, immune-fluorescence staining, microtubule dynamics and EBI competition assays as well as molecular docking simulations. Moreover, compound D19 induced G2/M phase arrest, resulted in cell apoptosis and inhibited the migration of U87 cells by flow cytometry analysis and wound healing assays. Significantly, compound D19 dose-dependently inhibited the tumor growth of orthotopic glioma xenografts model (GL261-Luc) and effectively prolonged the survival time of mice, which were extremely better than those of positive drug temozolomide (TMZ). Compound D19 exhibited potent in vivo antivascular activity as well as no observable toxicity. Furthermore, the results of in silico simulation studies and P-gp transwell assays verified the positive correlation between compound D19's Blood-Brain Barrier (BBB) permeability and its in vivo anti-GBM activity. Overall, compound D19 can be used as a promising anti-GBM lead compound for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Haoyi Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Dongyu Zhang
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziyang Yuan
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Haishi Qiao
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhuolu Xia
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Cao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Feng Jiang
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Abstract
We disclose herein electro-oxidative synthesis as the general protocol for procuring phenazines under mild reaction conditions. Using aerial oxygen as an oxidant, inexpensive electrolyte, and electrodes, a diverse range of phenazines have been accessed in good yields via the ring contraction of 10,11-dihydro-5H-dibenzo[b,e][1,4]diazepines. In addition, the syntheses of phenazines and diamino phenazines via direct electro-oxidation of dihydrophenazines and electro-dimerization of o-phenylenediamines, respectively, have also been accomplished.
Collapse
Affiliation(s)
- Deepak Sharma
- Department of Chemistry, Indian Institute of Technology Jammu, NH-44, Nagrota Bypass, Jammu 181221, J & K, India
| | - Namrata Kotwal
- Department of Chemistry, Indian Institute of Technology Jammu, NH-44, Nagrota Bypass, Jammu 181221, J & K, India
| | - Pankaj Chauhan
- Department of Chemistry, Indian Institute of Technology Jammu, NH-44, Nagrota Bypass, Jammu 181221, J & K, India
| |
Collapse
|
7
|
Zhang C, Ding Q, Xia Z, Wang H, Jiang F, Lu Y. Novel Chalcone-Phenazine Hybrids Induced Ferroptosis in U87-MG Cells through Activating Ferritinophagy. Chem Biodivers 2023; 20:e202201117. [PMID: 36536551 DOI: 10.1002/cbdv.202201117] [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: 11/24/2022] [Revised: 12/07/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Thirty-seven novel chalcone-phenazine hybrid molecules (C1∼C13 and F1∼F24) with 1,2,3-triazole or ethyl group as linkers were designed and synthesized in this study. Some compounds exhibited selective cytotoxicity against U87-MG cancer cell lines in vitro, in which compound C4 were found to have the best antiproliferative activity. SAR study indicated 1,2,3-triazole group may be crucial for enhancing compounds' cytotoxicity. C4 was verified to induce ferroptosis in U87-MG cells by transcription, lipid peroxidation, lipid ROS assays. Furthermore, C4 was up-regulated LC3-II, degradated FTH1, and then increasing iron resulted in the down-regulation of NCOA4. Together, all above evidences highlighted the potential of compound C4 that triggered ferroptosis by activating ferritinophagy against U87-MG cells.
Collapse
Affiliation(s)
- Chunhua Zhang
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Qifan Ding
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhuolu Xia
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Hengyu Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Jiang
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
8
|
Yang H, Wang H, Feng J, Liao J, Lu Y. Discovery of novel inhibition site centered on 114-bit tryptophan of Thioredoxin reductase 1 through computer-aided drug design. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
9
|
Ding Q, Wang H, Wang Y, Lu Y. A thioredoxin reductase 1 inhibitor pyrano [3,2-a] phenazine inhibits A549 cells proliferation and migration through the induction of reactive oxygen species production. Mol Biol Rep 2022; 49:8835-8845. [PMID: 35780225 DOI: 10.1007/s11033-022-07733-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Thioredoxin reductase 1 (TrxR1) inhibitor, pyrano [3,2-a] phenazine, named CPUL-1, was synthesized with potential anticancer activity. The aim of the present work was to explore the potential anti-proliferative and anti-metastatic ability of CPUL-1 against A549 cancer cell lines in vitro. METHODS AND RESULTS First, Cell Counting Kit-8 (CCK8) assay was used to assess cell proliferation. The A549 cell migration was evaluated by wound healing assay and transwell assay. Second, the epithelial-mesenchymal transition (EMT)-related proteins in A549 cells treated with CPUL-1 were analyzed by western blot methods. Then, TrxR1 enzyme activity assay and reactive oxygen species (ROS) assay were conducted to evaluate the effect of CPUL-1 on TrxR1 inhibition and ROS levels. Finally, western blotting was used to explore the mechanism of CPUL-1. The study results revealed that the ability of cell proliferation and migration was decreased under CPUL-1 treatment. CPUL-1 could distinctly restrain the migration and invasion of A549 cells through inhibiting EMT process. The results of TrxR1 enzyme activity assay, ROS assay and western blotting showed that CPUL-1 influenced EMT via inducing ROS-mediated ERK/JNK signaling by inhibiting TrxR1 enzyme activity. CONCLUSIONS Together, proliferation suppression and anti-metastasis activity of CPUL-1 in A549 cells were demonstrated by all the evidence. Our findings highlight the great potential of phenazine compound CPUL-1 to suppress A549 cells proliferation and metastasis.
Collapse
Affiliation(s)
- Qifan Ding
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hengyu Wang
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying Wang
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Yang Y, Lu Y, Zhang C, Guo Q, Zhang W, Wang T, Xia Z, Liu J, Cheng X, Xi T, Jiang F, Zheng L. Phenazine derivatives attenuate the stemness of breast cancer cells through triggering ferroptosis. Cell Mol Life Sci 2022; 79:360. [PMID: 35690642 PMCID: PMC11072418 DOI: 10.1007/s00018-022-04384-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/03/2022]
Abstract
Breast cancer stem cells (BCSCs) are positively correlated with the metastasis, chemoresistance, and recurrence of breast cancer. However, there are still no drugs targeting BCSCs in clinical using for breast cancer treatment. Here, we tried to screen out small-molecule compounds targeting BCSCs from the phenazine library established by us before. We focused on the compounds without affecting cell viability and screened out three potential compounds (CPUL119, CPUL129, CPUL149) that can significantly attenuate the stemness of breast cancer cells, as evident by the decrease of stemness marker expression, CD44+/CD24- subpopulation, mammary spheroid-formation ability, and tumor-initiating capacity. Additionally, these compounds suppressed the metastatic ability of breast cancer cells in vitro and in vivo. Combined with the transcriptome sequencing analysis, ferroptosis was shown on the top of the most upregulated pathways by CPUL119, CPUL129, and CPUL149, respectively. Mechanistically, we found that these three compounds could trigger ferroptosis by accumulating and sequestering iron in lysosomes through interacting with iron, and by regulating the expression of proteins (IRP2, TfR1, ferritin) engaged in iron transport and storage. Furthermore, inhibition of ferroptosis rescued the suppression of these three compounds on breast cancer cell stemness. This study suggests that CPUL119, CPUL129, and CPUL149 can specifically inhibit the stemness of breast cancer cells through triggering ferroptosis and may be the potential compounds for breast cancer treatment.
Collapse
Affiliation(s)
- Yue Yang
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Yuanyuan Lu
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Chunhua Zhang
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Qianqian Guo
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450003, People's Republic of China
| | - Wenzhou Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450003, People's Republic of China
| | - Ting Wang
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Zhuolu Xia
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Jing Liu
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Xiangyu Cheng
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| | - Feng Jiang
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| | - Lufeng Zheng
- School of Life Science and Technology, School of Engineering, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
11
|
Olyaei A, Sadeghpour M. A review on lawsone-based benzo[ a]phenazin-5-ol: synthetic approaches and reactions. RSC Adv 2022; 12:13837-13895. [PMID: 35541431 PMCID: PMC9082651 DOI: 10.1039/d2ra02139k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/02/2022] [Indexed: 12/05/2022] Open
Abstract
Phenazine systems are an important class of aza-polycyclic compounds that are easily found in nature and isolated as secondary metabolites primarily from Pseudomonas, Streptomyces, and a few other genera from soil or marine habitats. Moreover, various synthetic phenazine analogs are known for their pharmaceutical activities. Among various phenazines, benzo[a]phenazines are structural subunits in a variety of important natural products and have been given special attention due to their unique biological properties in various fields. In this review article, we highlight the synthesis of benzo[a]phenazin-5-ol derivatives from lawsone and benzene-1,2-diamines and their applications for the construction of a variety of five and six membered fused heterocycles such as pyranophenazines, spiropyranophenazines, pyridophenazines, furophenazines, benzochromenophenazines and oxazinophenazines during the period of 1995 to 2021.
Collapse
Affiliation(s)
- Abolfazl Olyaei
- Department of Chemistry, Payame Noor University (PNU) PO BOX 19395-4697 Tehran Iran +98-28-33374081 +98-28-33376366
| | - Mahdieh Sadeghpour
- Department of Chemistry, Takestan Branch, Islamic Azad University Takestan Iran +98-28-35270165 +98-28-35270167
| |
Collapse
|
12
|
Liu J, Ji F, Xia Z, Zhang C, Zhao C, Li Y, Zhou X, Huang D, Chen W, Jiang F. Multifunctional Nanoaggregates Composed of Active CPUL1 and a Triphenylphosphine Derivative for Mitochondria-Targeted Drug Delivery and Cell Imaging. ChemMedChem 2021; 17:e202100632. [PMID: 34750966 DOI: 10.1002/cmdc.202100632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/08/2021] [Indexed: 12/19/2022]
Abstract
We report that active substance (CPUL1) and triphenylphosphine (TPP) derivative could self-assemble into multifunctional nanoaggregates (CPUL1-TPP NAs) through electrostatic and π-π stacking interactions. CPUL1 was wrapped tightly inside the nanoparticles as well as CPUL1 and TPP derivative self-assembled into stable and compact nanoparticles in water. The positive surface charge of CPUL1-TPP NAs made them much easier to be endocytosed to enter cytoplasm, accumulate in the mitochondria and induce cell apoptosis based on their mitochondria targeting ability, fluorescence property and fast cell uptake characteristic, which showed better antitumor efficacy on HUH7 hepatoma cells in vitro than that of free CPUL1.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Fei Ji
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhuolu Xia
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Chunhua Zhang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Changshun Zhao
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Yanfei Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiang Zhou
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Wei Chen
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Jiang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
13
|
Advances in Phenazines over the Past Decade: Review of Their Pharmacological Activities, Mechanisms of Action, Biosynthetic Pathways and Synthetic Strategies. Mar Drugs 2021; 19:md19110610. [PMID: 34822481 PMCID: PMC8620606 DOI: 10.3390/md19110610] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 01/25/2023] Open
Abstract
Phenazines are a large group of nitrogen-containing heterocycles, providing diverse chemical structures and various biological activities. Natural phenazines are mainly isolated from marine and terrestrial microorganisms. So far, more than 100 different natural compounds and over 6000 synthetic derivatives have been found and investigated. Many phenazines show great pharmacological activity in various fields, such as antimicrobial, antiparasitic, neuroprotective, insecticidal, anti-inflammatory and anticancer activity. Researchers continued to investigate these compounds and hope to develop them as medicines. Cimmino et al. published a significant review about anticancer activity of phenazines, containing articles from 2000 to 2011. Here, we mainly summarize articles from 2012 to 2021. According to sources of compounds, phenazines were categorized into natural phenazines and synthetic phenazine derivatives in this review. Their pharmacological activities, mechanisms of action, biosynthetic pathways and synthetic strategies were summarized. These may provide guidance for the investigation on phenazines in the future.
Collapse
|
14
|
Liang T, Sun X, Li W, Hou G, Gao F. 1,2,3-Triazole-Containing Compounds as Anti-Lung Cancer Agents: Current Developments, Mechanisms of Action, and Structure-Activity Relationship. Front Pharmacol 2021; 12:661173. [PMID: 34177578 PMCID: PMC8226129 DOI: 10.3389/fphar.2021.661173] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the most common malignancy and leads to around one-quarter of all cancer deaths. Great advances have been achieved in the treatment of lung cancer with novel anticancer agents and improved technology. However, morbidity and mortality rates remain extremely high, calling for an urgent need to develop novel anti-lung cancer agents. 1,2,3-Triazole could be readily interact with diverse enzymes and receptors in organisms through weak interaction. 1,2,3-Triazole can not only be acted as a linker to tether different pharmacophores but also serve as a pharmacophore. This review aims to summarize the recent advances in 1,2,3-triazole-containing compounds with anti-lung cancer potential, and their structure-activity relationship (SAR) together with mechanisms of action is also discussed to pave the way for the further rational development of novel anti-lung cancer candidates.
Collapse
Affiliation(s)
- Ting Liang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiangyang Sun
- Department of Interventional Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhong Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guihua Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
15
|
Zhong Y, Liu J, Cheng X, Zhang H, Zhang C, Xia Z, Wu Z, Zhang L, Zheng Y, Gao Z, Jiang Z, Wang Z, Huang D, Lu Y, Jiang F. Design, synthesis and biological evaluations of diverse Michael acceptor-based phenazine hybrid molecules as TrxR1 inhibitors. Bioorg Chem 2021; 109:104736. [PMID: 33640630 DOI: 10.1016/j.bioorg.2021.104736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
A series of novel phenazine derivatives (1~27) containing the Michael acceptor scaffolds were designed and synthesized in this study. Some compounds exhibited selective cytotoxicity against Bel-7402 cancer cell line in vitro, in which compound 26 were found to have the best antiproliferative activity. Meanwhile, compound 26 showed no obvious cell toxicity against human normal liver epithelial L02 cells, which means this compound possessed a better safety potential. In the following research, compound 26 was verified to inhibit TrxR1 enzyme activity, ultimately resulting in cellular molecular mechanism events of apoptosis including growth of intracellular ROS level, depletion of reduced Trx1, liberation of ASK1 and up-regulation of p38, respectively. Together, all these evidences implicated that compound 26 acted as the TrxR1 inhibitor against Bel-7402 cells, and could activate apoptosis through the ROS-Trx-ASK1-p38 pathway.
Collapse
Affiliation(s)
- Yucheng Zhong
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Jing Liu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Cheng
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Chunhua Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhuolu Xia
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongxi Wu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yuting Zheng
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhanyu Gao
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhidong Jiang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhixiang Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Dechun Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Feng Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
16
|
Nazeef M, Saquib M, Tiwari SK, Yadav V, Ansari S, Sagir H, Hussain MK, Siddiqui IR. Catalyst Free, Multicomponent Green Approach to Benzo[
a
]chromeno[2,3‐
c
]phenazines Using Glycerol as a Recyclable and Biodegradable Promoting Medium. ChemistrySelect 2020. [DOI: 10.1002/slct.202003732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Mohd Nazeef
- Laboratory of Green Synthesis Department of Chemistry University of Allahabad Allahabad 211002 India
| | - Mohammad Saquib
- Department of Chemistry University of Allahabad Allahabad 211002 India
| | - Saurabh Kumar Tiwari
- Laboratory of Green Synthesis Department of Chemistry University of Allahabad Allahabad 211002 India
| | - Vikas Yadav
- Laboratory of Green Synthesis Department of Chemistry University of Allahabad Allahabad 211002 India
| | - Saif Ansari
- Laboratory of Green Synthesis Department of Chemistry University of Allahabad Allahabad 211002 India
| | - Hozeyfa Sagir
- Department of Chemistry Paliwal P.G. College Shikohabad 283135 India
| | | | - I. R. Siddiqui
- Laboratory of Green Synthesis Department of Chemistry University of Allahabad Allahabad 211002 India
| |
Collapse
|
17
|
Thakur GA, Kadu RK, Patil VR, Thakur PB. Progress in the Synthesis of Oxindole‐Naphthoquinone Molecular Hybrid Scaffolds: A Concise Review. ChemistrySelect 2020. [DOI: 10.1002/slct.202002961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Ganesh A. Thakur
- Department of Chemistry Rayat Shikshan Sanstha's Mahatma Phule Arts Science & Commerce College Panvel (Affiliated to University of Mumbai) Maharashtra 410206 India
| | - Rupashri K. Kadu
- Department of Chemistry Rayat Shikshan Sanstha's Arts Science & Commerce College Mokhada (Affiliated to University of Mumbai Mumbai), District-Palghar, Maharashtra 410206 India
| | - Vishwanath R. Patil
- Department of Chemistry University of Mumbai Santacruz (E) Mumbai, Maharashtra 400 098 India
| | - Pramod B. Thakur
- Department of Chemistry Rayat Shikshan Sanstha's Mahatma Phule Arts Science & Commerce College Panvel (Affiliated to University of Mumbai) Maharashtra 410206 India
| |
Collapse
|
18
|
Mohareb RM, Abouzied AS, Abbas NS. Synthesis and Biological Evaluation of Novel 4,5,6,7-Tetrahydrobenzo[D]-Thiazol-2- Yl Derivatives Derived from Dimedone with Anti-Tumor, C-Met, Tyrosine Kinase and Pim-1 Inhibitions. Anticancer Agents Med Chem 2020; 19:1438-1453. [PMID: 31038076 DOI: 10.2174/1871520619666190416102144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/01/2019] [Accepted: 03/28/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dimedone and thiazole moieties are privileged scaffolds (acting as primary pharmacophores) in many compounds that are useful to treat several diseases, mainly tropical infectious diseases. Thiazole derivatives are a very important class of compounds due to their wide range of pharmaceutical and therapeutic activities. On the other hand, dimedone is used to synthesize many therapeutically active compounds. Therefore, the combination of both moieties through a single molecule to produce heterocyclic compounds will produce excellent anticancer agents. OBJECTIVE The present work reports the synthesis of 47 new substances belonging to two classes of compounds: Dimedone and thiazoles, with the purpose of developing new drugs that present high specificity for tumor cells and low toxicity to the organism. To achieve this goal, our strategy was to synthesize a series of 4,5,6,7-tetrahydrobenzo[d]-thiazol-2-yl derivatives using the reaction of the 2-bromodimedone with cyanothioacetamide. METHODS The reaction of 2-bromodimedone with cyanothioacetamide gave the 4,5,6,7-tetrahydrobenzo[d]- thiazol-2-yl derivative 4. The reactivity of compound 4 towards some chemical reagents was observed to produce different heterocyclic derivatives. RESULTS A cytotoxic screening was performed to evaluate the performance of the new derivatives in six tumor cell lines. Thirteen compounds were shown to be promising toward the tumor cell lines which were further evaluated toward five tyrosine kinases. CONCLUSION The results of antitumor screening showed that many of the tested compounds were of high inhibition towards the tested cell lines. Compounds 6c, 8c, 11b, 11d, 13b, 14b, 15c, 15g, 21b, 21c, 20d and 21d were the most potent compounds toward c-Met kinase and PC-3 cell line. The most promising compounds 6c, 8c, 11b, 11d, 13b, 14b, 15c, 15g, 20c, 20d, 21b, 21c and 21d were further investigated against tyrosine kinase (c-Kit, Flt-3, VEGFR-2, EGFR, and PDGFR). Compounds 6c, 11b, 11d, 14b, 15c, and 20d were selected to examine their Pim-1 kinase inhibition activity the results revealed that compounds 11b, 11d and 15c had high activities.
Collapse
Affiliation(s)
- Rafat M Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Amr S Abouzied
- National Organization for Drug Control & Research, Cairo, Egypt.,Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Nermeen S Abbas
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, A. R., Egypt.,Department of Chemistry, Faculty of Science, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
19
|
Nikoorazm M, Khanmoradi M, Mohammadi M. Guanine‐La complex supported onto SBA‐15: A novel efficient heterogeneous mesoporous nanocatalyst for one‐pot, multi‐component Tandem Knoevenagel condensation–Michael addition–cyclization Reactions. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5504] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Mohsen Nikoorazm
- Department of Chemistry, Faculty of ScienceIlam University P. O. Box 69315516 Ilam Iran
| | - Maryam Khanmoradi
- Department of Chemistry, Faculty of ScienceIlam University P. O. Box 69315516 Ilam Iran
| | - Masoud Mohammadi
- Department of Chemistry, Faculty of ScienceIlam University P. O. Box 69315516 Ilam Iran
| |
Collapse
|
20
|
Thakur A, Singh A, Kaur N, Ojha R, Nepali K. Steering the antitumor drug discovery campaign towards structurally diverse indolines. Bioorg Chem 2020; 94:103436. [DOI: 10.1016/j.bioorg.2019.103436] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
|
21
|
Zhang MC, Gu SH, Liu GP, Li CC, Xu HM, Wu ZX, Ye BP, Lu YY, Huang DC, Wang ZX, Jiang F. Facile Synthesis and Cytotoxicity of Phenazine-Chromene Hybrid Molecules Derived from Phenazine Natural Product. Comb Chem High Throughput Screen 2019; 22:35-40. [PMID: 30848195 DOI: 10.2174/1386207322666190307125015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/12/2019] [Accepted: 02/26/2019] [Indexed: 01/27/2023]
Abstract
AIM AND OBJECTIVE Small molecule targeted drugs can effectively reduce the toxicity and side effects of drugs, and improve the efficacy of drugs by their specific antitumor activity. Hence, the development of small molecular targeted drugs for cancer has important significance. This study was undertaken to design and synthesize novel phenazine-chromene hybrid molecules in order to optimize the structure and improve the efficacy of this kind of hybrids. MATERIALS AND METHODS O-diaminobenzene was used as starting material to synthesize twentyfour heterocyclic compounds designed as hybrid molecules of phenazine and 4H-chromene pharmacophores by facile methods. The structures of the compound were confirmed by 1H NMR, 13C NMR and HRMS. Furthermore, the synthesized compounds were evaluated for in vitro activity against four human cancer cell lines and two non-cancer cell lines by MTT test. RESULTS Some compounds showed strong cytotoxic activities against HepG2 and A549 cancer lines (IC50 = 5-10 µM). Comparing 2i with 2l, the introduction of hydrophilic groups on the phenazine core could not improve the antiproliferative activity significantly. Except 2d and 3c, compounds owning chlorine substituent on the 4H-chromene pharmacophore seemingly contribute to enhance the compounds' antiproliferative activity. Specially, compound 3c showed highest cytotoxicity against A549 cells with IC50 values of 3.3±0.4 µM. Furthermore, all compounds showed low or no cytotoxicity against HUVEC and L02 non-cancer cells in vitro. CONCLUSION Compound 3c may be used as potential lead molecule against A549 cancer cells.
Collapse
Affiliation(s)
- Mei-Chen Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Shu-Hui Gu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Guang-Pan Liu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Chen-Cheng Li
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Han-Mei Xu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.,School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Zhong-Xi Wu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Bo-Ping Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan-Yuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - De-Chun Huang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhi-Xiang Wang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Feng Jiang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China.,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
22
|
Preparation and characterization of new inorganic–organic hybrid catalyst H
3
PMo
12
O
40
/Hyd‐SBA‐15 and its application in the domino multi‐component reaction. Appl Organomet Chem 2019. [DOI: 10.1002/aoc.5201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Xu Z, Zhao SJ, Liu Y. 1,2,3-Triazole-containing hybrids as potential anticancer agents: Current developments, action mechanisms and structure-activity relationships. Eur J Med Chem 2019; 183:111700. [PMID: 31546197 DOI: 10.1016/j.ejmech.2019.111700] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022]
Abstract
Anticancer agents are critical for the cancer treatment, but side effects and the drug resistance associated with the currently used anticancer agents create an urgent need to explore novel drugs with low side effects and high efficacy. 1,2,3-Triazole is privileged building block in the discovery of new anticancer agents, and some of its derivatives have already been applied in clinics or under clinical trials for fighting against cancers. Hybrid molecules occupy an important position in cancer control, and hybridization of 1,2,3-triazole framework with other anticancer pharmacophores may provide valuable therapeutic intervention for the treatment of cancer, especially drug-resistant cancer. This review emphasizes the recent advances in 1,2,3-triazole-containing hybrids with anticancer potential, covering articles published between 2015 and 2019, and the structure-activity relationships, together with mechanisms of action are also discussed.
Collapse
Affiliation(s)
- Zhi Xu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China.
| | - Shi-Jia Zhao
- Wuhan University of Science and Technology, Wuhan, PR China
| | - Yi Liu
- Wuhan University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
24
|
Discovery of indoline derivatives that inhibit esophageal squamous cell carcinoma growth by Noxa mediated apoptosis. Bioorg Chem 2019; 92:103190. [PMID: 31465969 DOI: 10.1016/j.bioorg.2019.103190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/27/2019] [Accepted: 08/08/2019] [Indexed: 11/23/2022]
Abstract
A series of novel indoline derivatives were synthesized and evaluated for antiproliferative activity against four selected cancer cell lines (Hela, A549, HepG2 and KYSE30). Among them, compound 20 displayed the potent inhibition activity against esophageal cancer cells (Kyse30, Kyse450, Kyse510 and EC109). Cellular mechanism studies in esophageal squamous cell carcinoma (ESCC) cells elucidated compound 20 inhibited cell growths in vitro and in vivo, reduced colony formation, arrested cell cycle at M phase, and induced Noxa-dependent apoptosis in ESCC. Importantly, compound 20 was identified as a novel Noxa mediated apoptosis inducer. These results suggested that compound 20 might be a promising anticancer agent with potential for development of further clinical applications.
Collapse
|
25
|
Konwar M, Hazarika R, Ali AA, Chetia M, Khupse ND, Saikia PJ, Sarma D. Benedict's solution/ vitamin C: An alternative catalytic protocol for the synthesis of regioselective-1,4-disubstituted-1H
-1,2,3-triazoles at room temperature. Appl Organomet Chem 2018. [DOI: 10.1002/aoc.4425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Manashjyoti Konwar
- Department of Chemistry; Dibrugarh University; Dibrugarh 786004 Assam India
| | - Roktopol Hazarika
- Department of Chemistry; Dibrugarh University; Dibrugarh 786004 Assam India
| | - Abdul A. Ali
- Department of Chemistry; Dibrugarh University; Dibrugarh 786004 Assam India
| | - Mitali Chetia
- Department of Chemistry; Dibrugarh University; Dibrugarh 786004 Assam India
| | - Nageshwar D. Khupse
- Centre for Materials for Electronics Technology; Pashan Road Pune 411008 India
| | - Prakash J. Saikia
- Analytical Chemistry Division; CSIR-North East Institute of Science & Technology; Jorhat 785006 Assam India
| | - Diganta Sarma
- Department of Chemistry; Dibrugarh University; Dibrugarh 786004 Assam India
| |
Collapse
|
26
|
Discovery of Novel Bromophenol Hybrids as Potential Anticancer Agents through the Ros-Mediated Apoptotic Pathway: Design, Synthesis and Biological Evaluation. Mar Drugs 2017; 15:md15110343. [PMID: 29104274 PMCID: PMC5706033 DOI: 10.3390/md15110343] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 10/18/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023] Open
Abstract
A series of bromophenol hybrids with N-containing heterocyclic moieties were designed, and their anticancer activities against a panel of five human cancer cell lines (A549, Bel7402, HepG2, HCT116 and Caco2) using MTT assay in vitro were explored. Among them, thirteen compounds (17a, 17b, 18a, 19a, 19b, 20a, 20b, 21a, 21b, 22a, 22b, 23a, and 23b) exhibited significant inhibitory activity against the tested cancer cell lines. The structure-activity relationships (SARs) of bromophenol derivatives were discussed. The promising candidate compound 17a could induce cell cycle arrest at G0/G1 phase and induce apoptosis in A549 cells, as well as caused DNA fragmentations, morphological changes and ROS generation by the mechanism studies. Furthermore, compound 17a suppression of Bcl-2 levels (decrease in the expression of the anti-apoptotic proteins Bcl-2 and down-regulation in the expression levels of Bcl-2) in A549 cells were observed, along with activation caspase-3 and PARP, which indicated that compound 17a induced A549 cells apoptosis in vitro through the ROS-mediated apoptotic pathway. These results might be useful for bromophenol derivatives to be explored and developed as novel anticancer drugs.
Collapse
|