Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

邮发代号 80-967

2019 Impact Factor: 3.421

Frontiers of Medicine  2023, Vol. 17 Issue (2): 290-303   https://doi.org/10.1007/s11684-022-0956-8
  本期目录
Repurposed benzydamine targeting CDK2 suppresses the growth of esophageal squamous cell carcinoma
Yubing Zhou1,2, Xinyu He1,2, Yanan Jiang1,2,3,4, Zitong Wang1, Yin Yu1,2, Wenjie Wu1,2, Chenyang Zhang1, Jincheng Li1, Yaping Guo1,3, Xinhuan Chen1,3, Zhicai Liu4,6, Jimin Zhao1,3,4,5, Kangdong Liu1,2,3,4,5(), Zigang Dong1,2,3,5()
1. The Pathophysiology Department, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
2. The China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
3. State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, China
4. Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou 450000, China
5. Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, China
6. Oncology Department, The Tumor Hospital of Linzhou City, Linzhou 456500, China
 全文: PDF(4983 KB)   HTML
Abstract

Esophageal squamous cell carcinoma (ESCC) is one of the leading causes of cancer death worldwide. It is urgent to develop new drugs to improve the prognosis of ESCC patients. Here, we found benzydamine, a locally acting non-steroidal anti-inflammatory drug, had potent cytotoxic effect on ESCC cells. Benzydamine could suppress ESCC proliferation in vivo and in vitro. In terms of mechanism, CDK2 was identified as a target of benzydamine by molecular docking, pull-down assay and in vitro kinase assay. Specifically, benzydamine inhibited the growth of ESCC cells by inhibiting CDK2 activity and affecting downstream phosphorylation of MCM2, c-Myc and Rb, resulting in cell cycle arrest. Our study illustrates that benzydamine inhibits the growth of ESCC cells by downregulating the CDK2 pathway.

Key wordsbenzydamine    cyclin-dependent kinase 2    patient-derived xenograft    esophageal squamous cell carcinoma
收稿日期: 2021-12-07      出版日期: 2023-05-26
Corresponding Author(s): Kangdong Liu,Zigang Dong   
 引用本文:   
. [J]. Frontiers of Medicine, 2023, 17(2): 290-303.
Yubing Zhou, Xinyu He, Yanan Jiang, Zitong Wang, Yin Yu, Wenjie Wu, Chenyang Zhang, Jincheng Li, Yaping Guo, Xinhuan Chen, Zhicai Liu, Jimin Zhao, Kangdong Liu, Zigang Dong. Repurposed benzydamine targeting CDK2 suppresses the growth of esophageal squamous cell carcinoma. Front. Med., 2023, 17(2): 290-303.
 链接本文:  
https://academic.hep.com.cn/fmd/CN/10.1007/s11684-022-0956-8
https://academic.hep.com.cn/fmd/CN/Y2023/V17/I2/290
Fig.1  
Fig.2  
Fig.3  
Fig.4  
Fig.5  
Fig.6  
1 Z Yuan, X Wang, X Geng, Y Li, J Mu, F Tan, Q Xue, S Gao, J He. Liquid biopsy for esophageal cancer: is detection of circulating cell-free DNA as a biomarker feasible?. Cancer Commun (Lond) 2021; 41(1): 3–15
https://doi.org/10.1002/cac2.12118 pmid: 33264481
2 CC Abnet, M Arnold, WQ Wei. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology 2018; 154(2): 360–373
https://doi.org/10.1053/j.gastro.2017.08.023 pmid: 28823862
3 YM Yang, P Hong, WW Xu, QY He, B Li. Advances in targeted therapy for esophageal cancer. Signal Transduct Target Ther 2020; 5(1): 229
https://doi.org/10.1038/s41392-020-00323-3 pmid: 33028804
4 M di Pietro, MI Canto, RC Fitzgerald. Endoscopic management of early adenocarcinoma and squamous cell carcinoma of the esophagus: screening, diagnosis, and therapy. Gastroenterology 2018; 154(2): 421–436
https://doi.org/10.1053/j.gastro.2017.07.041 pmid: 28778650
5 ZW Reichenbach, MG Murray, R Saxena, D Farkas, EG Karassik, A Klochkova, K Patel, C Tice, TM Hall, J Gang, HP Parkman, SJ Ward, MP Tétreault, KA Whelan. Clinical and translational advances in esophageal squamous cell carcinoma. Adv Cancer Res 2019; 144: 95–135
https://doi.org/10.1016/bs.acr.2019.05.004 pmid: 31349905
6 YJ Surh. Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer 2003; 3(10): 768–780
https://doi.org/10.1038/nrc1189 pmid: 14570043
7 P Desai, NJ Thumma, PR Wagh, S Zhan, D Ann, J Wang, S Prabhu. Cancer chemoprevention using nanotechnology-based approaches. Front Pharmacol 2020; 11: 323
https://doi.org/10.3389/fphar.2020.00323 pmid: 32317961
8 A Ranjan, S Ramachandran, N Gupta, I Kaushik, S Wright, S Srivastava, H Das, S Srivastava, S Prasad, SK Srivastava. Role of phytochemicals in cancer prevention. Int J Mol Sci 2019; 20(20): 4981
https://doi.org/10.3390/ijms20204981 pmid: 31600949
9 CY Chen, CJ Kuo, YW Lee, F Lam, KW Tam. Benzydamine hydrochloride on postoperative sore throat: a meta-analysis of randomized controlled trials. Can J Anaesth 2014; 61(3): 220–228
https://doi.org/10.1007/s12630-013-0080-y pmid: 24263969
10 EB Faber, N Wang, GI Georg. Review of rationale and progress toward targeting cyclin-dependent kinase 2 (CDK2) for male contraception†. Biol Reprod 2020; 103(2): 357–367
https://doi.org/10.1093/biolre/ioaa107 pmid: 32543655
11 R Fagundes, LK Teixeira. Cyclin E/CDK2: DNA replication, replication stress and genomic instability. Front Cell Dev Biol 2021; 9: 774845
https://doi.org/10.3389/fcell.2021.774845 pmid: 34901021
12 O Tetsu, F McCormick. Proliferation of cancer cells despite CDK2 inhibition. Cancer Cell 2003; 3(3): 233–245
https://doi.org/10.1016/S1535-6108(03)00053-9 pmid: 12676582
13 G Au-Yeung, F Lang, WJ Azar, C Mitchell, KE Jarman, K Lackovic, D Aziz, C Cullinane, RB Pearson, L Mileshkin, D Rischin, AM Karst, R Drapkin, D Etemadmoghadam, DDL Bowtell. Selective targeting of cyclin E1-amplified high-grade serous ovarian cancer by cyclin-dependent kinase 2 and AKT inhibition. Clin Cancer Res 2017; 23(7): 1862–1874
https://doi.org/10.1158/1078-0432.CCR-16-0620 pmid: 27663592
14 S Tadesse, AT Anshabo, N Portman, E Lim, W Tilley, CE Caldon, S Wang. Targeting CDK2 in cancer: challenges and opportunities for therapy. Drug Discov Today 2020; 25(2): 406–413
https://doi.org/10.1016/j.drudis.2019.12.001 pmid: 31839441
15 S Tadesse, EC Caldon, W Tilley, S Wang. Cyclin-dependent kinase 2 inhibitors in cancer therapy: an update. J Med Chem 2019; 62(9): 4233–4251
https://doi.org/10.1021/acs.jmedchem.8b01469 pmid: 30543440
16 D Gfeller, A Grosdidier, M Wirth, A Daina, O Michielin, V Zoete. SwissTargetPrediction: a web server for target prediction of bioactive small molecules. Nucleic Acids Res 2014; 42: W32–W38
https://doi.org/10.1093/nar/gku293 pmid: 24792161
17 A Grosdidier, V Zoete, O Michielin. SwissDock, a protein-small molecule docking web service based on EADock DSS. Nucleic Acids Res 2011; 39: W270–W277
https://doi.org/10.1093/nar/gkr366 pmid: 21624888
18 Z Wang, MA Jensen, JC Zenklusen. A practical guide to The Cancer Genome Atlas (TCGA). Methods Mol Biol 2016; 1418: 111–141
https://doi.org/10.1007/978-1-4939-3578-9_6 pmid: 27008012
19 Y Hu, F Liu, X Jia, P Wang, T Gu, H Liu, T Liu, H Wei, H Chen, J Zhao, R Yang, Y Chen, Z Dong, K Liu. Periplogenin suppresses the growth of esophageal squamous cell carcinoma in vitro and in vivo by targeting STAT3. Oncogene 2021; 40(23): 3942–3958
https://doi.org/10.1038/s41388-021-01817-2 pmid: 33986510
20 Y Jiang, Q Wu, X Yang, J Zhao, Y Jin, K Li, Y Ma, X Chen, F Tian, S Zhao, J Xu, J Lu, X Yin, K Liu, Z Dong. A method for establishing a patient-derived xenograft model to explore new therapeutic strategies for esophageal squamous cell carcinoma. Oncol Rep 2016; 35(2): 785–792
https://doi.org/10.3892/or.2015.4459 pmid: 26718633
21 G Jin, M Yan, K Liu, K Yao, H Chen, C Zhang, Y Yi, K Reddy, DR Gorja, KV Laster, Z Guo, Z Dong. Discovery of a novel dual-target inhibitor against RSK1 and MSK2 to suppress growth of human colon cancer. Oncogene 2020; 39(43): 6733–6746
https://doi.org/10.1038/s41388-020-01467-w pmid: 32963350
22 MG Sürmen, S Sürmen, A Ali, SG Musharraf, N Emekli. Phosphoproteomic strategies in cancer research: a minireview. Analyst (Lond) 2020; 145(22): 7125–7149
https://doi.org/10.1039/D0AN00915F pmid: 32996481
23 J Ma, T Chen, S Wu, C Yang, M Bai, K Shu, K Li, G Zhang, Z Jin, F He, H Hermjakob, Y Zhu. iProX: an integrated proteome resource. Nucleic Acids Res 2019; 47(D1): D1211–D1217
https://doi.org/10.1093/nar/gky869 pmid: 30252093
24 M Kanehisa, M Furumichi, M Tanabe, Y Sato, K Morishima. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 2017; 45(D1): D353–D361
https://doi.org/10.1093/nar/gkw1092 pmid: 27899662
25 SK Burley, HM Berman, GJ Kleywegt, JL Markley, H Nakamura, S Velankar. Protein Data Bank (PDB): the single global macromolecular structure archive. Methods Mol Biol 2017; 1607: 627–641
https://doi.org/10.1007/978-1-4939-7000-1_26 pmid: 28573592
26 O Trott, AJ Olson. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31(2): 455–461
pmid: 19499576
27 H Sung, J Ferlay, RL Siegel, M Laversanne, I Soerjomataram, A Jemal, F Bray. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71(3): 209–249
https://doi.org/10.3322/caac.21660 pmid: 33538338
28 V Cioli, C Corradino, P Scorza Barcellona. Review of pharmacological data on benzydamine. Int J Tissue React 1985; 7(3): 205–213
pmid: 3899969
29 PA Quane, GG Graham, JB Ziegler. Pharmacology of benzydamine. Inflammopharmacology 1998; 6(2): 95–107
https://doi.org/10.1007/s10787-998-0026-0 pmid: 17694367
30 NP Singh, JK Makkar, V Wourms, PM Singh. Topical benzydamine for preventing postoperative sore throat. Anaesthesia 2018; 73(10): 1297
https://doi.org/10.1111/anae.14437 pmid: 30216428
31 H V Worthington. , Clarkson JE, Bryan G, Furness S, Glenny AM, Littlewood A, McCabe MG, Meyer S, Khalid T, Riley P. Interventions for preventing oral mucositis for patients with cancer receiving treatment. Cochrane database Syst Rev 2011; 2011(4): CD000978
32 RM Golsteyn. Cdk1 and Cdk2 complexes (cyclin dependent kinases) in apoptosis: a role beyond the cell cycle. Cancer Lett 2005; 217(2): 129–138
https://doi.org/10.1016/j.canlet.2004.08.005 pmid: 15617830
33 T Tsuji, SB Ficarro, W Jiang. Essential role of phosphorylation of MCM2 by Cdc7/Dbf4 in the initiation of DNA replication in mammalian cells. Mol Biol Cell 2006; 17(10): 4459–4472
https://doi.org/10.1091/mbc.e06-03-0241 pmid: 16899510
34 RYC Poon. Cell cycle control: a system of interlinking oscillators. Methods Mol Biol 2016; 1342: 3–19
https://doi.org/10.1007/978-1-4939-2957-3_1 pmid: 26254915
35 JW Harbour, RX Luo, AD Santi, AA Postigo, DC Dean. Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1. Cell 1999; 98(6): 859–869
https://doi.org/10.1016/S0092-8674(00)81519-6 pmid: 10499802
36 P Hydbring, LG Larsson. Tipping the balance: Cdk2 enables Myc to suppress senescence. Cancer Res 2010; 70(17): 6687–6691
https://doi.org/10.1158/0008-5472.CAN-10-1383 pmid: 20713526
37 P Hydbring, A Castell, LG Larsson. MYC modulation around the CDK2/p27/SKP2 axis. Genes (Basel) 2017; 8(7): 174
https://doi.org/10.3390/genes8070174 pmid: 28665315
38 PL Garcia, AL Miller, KJ Yoon. Patient-derived xenograft models of pancreatic cancer: overview and comparison with other types of models. Cancers (Basel) 2020; 12(5): 1327
https://doi.org/10.3390/cancers12051327 pmid: 32456018
39 S Klöß, S Dehmel, A Braun, MJ Parnham, U Köhl, S Schiffmann. From cancer to immune-mediated diseases and tolerance induction: lessons learned from immune oncology and classical anti-cancer treatment. Front Immunol 2020; 11: 1423
https://doi.org/10.3389/fimmu.2020.01423 pmid: 32733473
40 M Hidalgo, F Amant, AV Biankin, E Budinská, AT Byrne, C Caldas, RB Clarke, Jong S de, J Jonkers, GM Mælandsmo, S Roman-Roman, J Seoane, L Trusolino, A Villanueva. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov 2014; 4(9): 998–1013
https://doi.org/10.1158/2159-8290.CD-14-0001 pmid: 25185190
41 TO Nielsen, SCY Leung, DL Rimm, A Dodson, B Acs, S Badve, C Denkert, MJ Ellis, S Fineberg, M Flowers, HH Kreipe, AV Laenkholm, H Pan, FM Penault-Llorca, MY Polley, R Salgado, IE Smith, T Sugie, JMS Bartlett, LM McShane, M Dowsett, DF Hayes. Assessment of Ki67 in breast cancer: updated recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst 2021; 113(7): 808–819
https://doi.org/10.1093/jnci/djaa201 pmid: 33369635
[1] FMD-22033-OF-LKD_suppl_1 Download
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed