DOI QR코드

DOI QR Code

Ginsengenin derivatives synthesized from 20(R)-panaxotriol: Synthesis, characterization, and antitumor activity targeting HIF-1 pathway

  • Guo, Hong-Yan (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Xing, Yue (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Sun, Yu-Qiao (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Liu, Can (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Xu, Qian (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Shang, Fan-Fan (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Zhang, Run-Hui (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Jin, Xue-Jun (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Chen, Fener (Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University) ;
  • Lee, Jung Joon (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Kang, Dongzhou (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Shen, Qing-Kun (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University) ;
  • Quan, Zhe-Shan (Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University)
  • 투고 : 2021.07.02
  • 심사 : 2022.03.08
  • 발행 : 2022.11.01

초록

Background: Ginseng possesses antitumor effects, and ginsenosides are considered to be one of its main active chemical components. Ginsenosides can further be hydrolyzed to generate secondary saponins, and 20(R)-panaxotriol is an important sapogenin of ginsenosides. We aimed to synthesize a new ginsengenin derivative from 20(R)-panaxotriol and investigate its antitumor activity in vivo and in vitro. Methods: Here, 20(R)-panaxotriol was selected as a precursor and was modified into its derivatives. The new products were characterized by 1H-NMR, 13C-NMR and HR-MS and evaluated by molecular docking, MTT, luciferase reporter assay, western blotting, immunofluorescent staining, colony formation assay, EdU labeling and immunofluorescence, apoptosis assay, cells migration assay, transwell assay and in vivo antitumor activity assay. Results: The derivative with the best antitumor activity was identified as 6,12-dihydroxy-4,4,8,10,14-pentamethyl-17-(2,6,6-trimethyltetrahydro-2H-pyran-2-yl)hexadecahydro-1H-cyclopenta[a]phenanthren-3-yl(tert-butoxycarbonyl)glycinate (A11). The focus of this research was on the antitumor activity of the derivatives. The efficacy of the derivative A11 (IC50 < 0.3 µM) was more than 100 times higher than that of 20(R)- panaxotriol (IC50 > 30 µM). In addition, A11 inhibited the protein expression and nuclear accumulation of the hypoxia-inducible factor HIF-1α in HeLa cells under hypoxic conditions in a dose-dependent manner. Moreover, A11 dose-dependently inhibited the proliferation, migration, and invasion of HeLa cells, while promoting their apoptosis. Notably, the inhibition by A11 was more significant than that by 20(R)-panaxotriol (p < 0.01) in vivo. Conclusion: To our knowledge, this is the first study to report the production of derivative A11 from 20(R)-panaxotriol and its superior antitumor activity compared to its precursor. Moreover, derivative A11 can be used to further study and develop novel antitumor drugs.

키워드

과제정보

This work was supported by the National Natural Science Foundation of China, China (No.81960626, 82060628), Doctoral Research Startup Fundation of Yanbian University (No. 602020087), Higher Education Discipline Innovation Project, China (111 Project, D18012), and Jilin Provincial Department of Education Science and Technology Research Project, China (NO. JJKH20220559KJ).

참고문헌

  1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin 2011;61:69-90. https://doi.org/10.3322/caac.20107
  2. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359-86. https://doi.org/10.1002/ijc.29210
  3. Hait WN. Anticancer drug development: the grand challenges. Nat Rev Drug Discov 2010;9:253-4. https://doi.org/10.1038/nrd3144
  4. Arbyn M, Weiderpass E, Bruni L, de Sanjose S, Saraiya M, Ferlay J, Bray F. Estimates of incidence and mortality of cervical cancer in 2018: a worldwide analysis. Lancet Global Health 2020;8:e191-203. https://doi.org/10.1016/S2214-109X(19)30482-6
  5. Powis G, Kirkpatrick L. Hypoxia inducible factor-1alpha as a cancer drug target. Mol Cancer Ther 2004;3:647-54. https://doi.org/10.1158/1535-7163.647.3.5
  6. Vaupel Peter, Mayer Arnulf, Hockel M. Tumor hypoxia and malignant progression. Methods Enzymol 2004;381:335-54. https://doi.org/10.1016/S0076-6879(04)81023-1
  7. Semenza GL. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 2010;29:625-34. https://doi.org/10.1038/onc.2009.441
  8. Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol 2006;70:1469-80. https://doi.org/10.1124/mol.106.027029
  9. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer 2003;3:721-32. https://doi.org/10.1038/nrc1187
  10. Pientka FK, Hu J, Schindler SG, Brix B, Thiel A, Johren O, Fandrey J, BerchnerPfannschmidt U, Depping R. Oxygen sensing by the prolyl-4-hydroxylase PHD2 within the nuclear compartment and the influence of compartmentalisation on HIF-1 signalling. J Cell Sci 2012;25:5168-76.
  11. Li MY, Mi C, Wang KS, Wang Z, Zuo HX, Piao LX, Xu GH, Li X, Ma J, Jin X. Shikonin suppresses proliferation and induces cell cycle arrest through the inhibition of hypoxia-inducible factor-1alpha signaling. Chem Biol Interact 2017;274:58-67. https://doi.org/10.1016/j.cbi.2017.06.029
  12. Lu Y, Li Y, Wang Z, Xie S, Wang Q, Lei X, Ruan Y, Li J. Downregulation of RGMA by HIF-1A/miR-210-3p axis promotes cell proliferation in oral squamous cell carcinoma. Biochem Pharmacol 2019;112:108608.
  13. Yao J, Man S, Dong H, Yang L, Ma L, Gao W. Combinatorial treatment of Rhizoma Paridis saponins and sorafenib overcomes the intolerance of sorafenib. J Steroid Biochem Mol Biol 2018;183:159-66. https://doi.org/10.1016/j.jsbmb.2018.06.010
  14. Zhu X, Shen H, Yin X, Yang M, Wei H, Chen Q, Feng F, Liu Y, Xu W, Li Y. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J Exp Clin Cancer Res 2019;38:81. https://doi.org/10.1186/s13046-019-1095-1
  15. Choi KJ, Baik IH, Ye SK, Lee YH. Molecular targeted therapy for hepatocellular carcinoma: present status and future directions. Biol Pharm Bull 2015;38: 986-91. https://doi.org/10.1248/bpb.b15-00231
  16. Wu J, Joseph SO, Muggia FM. Targeted therapy: its status and promise in selected solid tumors part I: areas of major impact. Oncology (Williston Park) 2012;26:936-43.
  17. Hodges TW, Hossain CF, Kim YP, Zhou YD, Nagle DG. Molecular-targeted antitumor agents: the Saururus cernuus dineolignans manassantin B and 4-Odemethylmanassantin B are potent inhibitors of hypoxia-activated HIF-1. J Nat Prod 2004;67:767-71. https://doi.org/10.1021/np030514m
  18. Yuan SM. Potential cardioprotective effects of Ginseng preparations. Pak J Pharm Sci 2015;28:963-8.
  19. Attele AS, Wu JA, Yuan C-S. Ginseng pharmacology. Biochem Pharmacol 1999;58:1685-93. https://doi.org/10.1016/S0006-2952(99)00212-9
  20. Kim JH. Pharmacological and medical applications of Panax ginseng and ginsenosides: a review for use in cardiovascular diseases. J Ginseng Res 2018;42:264-9. https://doi.org/10.1016/j.jgr.2017.10.004
  21. Shibata S. Chemistry and cancer preventing activities of ginseng saponins and some related triterpenoid compounds. J Kor Med Sci 2001;16(Suppl):S28-37. https://doi.org/10.3346/jkms.2001.16.S.S28
  22. Park JD, Rhee DK, Lee YH. Biological activities and chemistry of saponins from Panax ginseng C. A. Meyer. Phytochem Rev 2005;4:159-75. https://doi.org/10.1007/s11101-005-2835-8
  23. Wei Y, Ma C-M, Hattori M. Anti-HIV protease triterpenoids from the acid hydrolysate of Panax ginseng. Phytochem Lett 2009;2:63-6. https://doi.org/10.1016/j.phytol.2008.12.001
  24. Yu R, Zhang Y, Xu ZQ, Wang JJ, Chen BC, Jin H. Potential antitumor effects of panaxatriol against DU-15 human prostate cancer cells is mediated via mitochondrial mediated apoptosis, inhibition of cell migration and sub-G1 cell cycle arrest. J Buon 2018;23:200-4.
  25. Li JL, Ding P, Jiang B, Yang M, Ren J, Song Y, Chen G. Biotransformation of 20(R)-panaxatriol by the fungus Aspergillus flavus link AS 3.3950. Nat Prod Res 2019;33:1393-8. https://doi.org/10.1080/14786419.2017.1416380
  26. Sheng C, Zhang W. New lead structures in antifungal drug discovery. Curr Med Chem 2011;18:733-66. https://doi.org/10.2174/092986711794480113
  27. Neu HC, Fu KP. Cefatrizine activity compared with that of other cephalosporins. Antimicrob Agents Chemother 1979;15:209-12. https://doi.org/10.1128/AAC.15.2.209
  28. Soltis MJ, Yeh HJ, Cole KA, Whittaker N, Wersto RP, Kohn EC. Identification and characterization of human metabolites of CAI [5-amino-1-1(4'-chlorobenzoyl3,5-dichlorobenzyl)-1,2,3-triazole- 4-carboxamide). Drug Metab Dispos 1996;24:799-806.
  29. Higashitani F, Hyodo A, Ishida N, Inoue M, Mitsuhashi S. Inhibition of betalactamases by tazobactam and in-vitro antibacterial activity of tazobactam combined with piperacillin. J Antimicrob Chemother 1990;25:567-74. https://doi.org/10.1093/jac/25.4.567
  30. Zhang G-R, Ren Y, Yin XM, Quan ZS. Synthesis and evaluation of the anticonvulsant activities of new 5-substitued-[1,2,4]triazolo[4,3-a]quinoxalin4(5H)-one derivatives. Lett Drug Des Discov 2018;15:406-13. https://doi.org/10.2174/1570180814666170619094408
  31. Liu X-J, Zhang H-J, Quan Z-S. Synthesis and evaluation of the anticonvulsant activities of 2,3-dihydrophthalazine-1,4-dione derivatives. Med Chem Res 2017;26:1935-46. https://doi.org/10.1007/s00044-017-1896-8
  32. Ren Y, Shen QK, Ding MM, Yin XM, Quan ZS. Synthesis and anticonvulsant activities of 4-Alkoxyl-[1,2,4]Triazolo[4,3-a]Quinoxaline derivatives. Lat Am J Pharm 2016;35:2169-75.
  33. Liu CF, Zhang HJ, Quan ZS. Synthesis and anticonvulsant activity of novel 3-(2- (4H-1,2,4-triazol-4-yl)ethyl)-1-alkyl-1H-indole derivatives. Lett Drug Des Discov 2016;13:833-9. https://doi.org/10.2174/1570180813666160324173717
  34. Zhang H-J, Wang S-B, Wen X, Li J-Z, Quan Z-S. Design, synthesis, and evaluation of the anticonvulsant and antidepressant activities of pyrido[2,3-d]pyrimidine derivatives. Med Chem Res 2016;25:1287-98. https://doi.org/10.1007/s00044-016-1559-1
  35. Pang L, Liu CY, Gong GH, Quan ZS. Synthesis, in vitro and in vivo biological evaluation of novel lappaconitine derivatives as potential anti-inflammatory agents. Acta Pharm Sin B 2020;10:628-45. https://doi.org/10.1016/j.apsb.2019.09.002
  36. Pan F-J, Wang S-B, Liu D-C, Gong G-H, Quan Z-S. Synthesis of 4-Phenylthieno [2,3-e][1,2,4]triazolo[4,3-a]pyrimidine-5(4H)-one derivatives and evaluation of their anti-inflammatory activity. Lett Drug Des Dis 2015;13:141-8. https://doi.org/10.2174/1570180812666150630184439
  37. Zhang T-Y, Li C, Li Y-R, Li X-Z, Sun L-P, Zheng C-J, Piao H-R. Synthesis and antimicrobial evaluation of aminoguanidine and 3-amino- 1,2,4-triazole derivatives as potential antibacterial agents. Lett Drug Des Dis 2016;13: 1063-75. https://doi.org/10.2174/1570180813666160819151239
  38. Zhang HJ, Zhang GR, Piao HR, Quan ZS. Synthesis and characterisation of celastrol derivatives as potential anticancer agents. J Enzyme Inhib Med Chem 2017;33:190-8.
  39. Shen QK, Deng H, Wang SB, Tian YS, Quan ZS. Synthesis, and evaluation of in vitro and in vivo anticancer activity of 14-substituted oridonin analogs: a novel and potent cell cycle arrest and apoptosis inducer through the p53- MDM2 pathway. Eur J Med Chem 2019;173:15-31. https://doi.org/10.1016/j.ejmech.2019.04.005
  40. Luan T, Cao LH, Deng H, Shen QK, Tian YS, Quan ZS. Design and synthesis of C19 isosteviol derivatives as potent and highly selective antiproliferative agents. Molecules 2018;24:121. https://doi.org/10.3390/molecules24010121
  41. De P, Baltas M, Bedos-Belval F. Cinnamic acid derivatives as anticancer agentsa review. Curr Med Chem 2011;18:1672-703. https://doi.org/10.2174/092986711795471347
  42. Li QF, Shi SL, Liu QR, Tang J, Song J, Liang Y. Anticancer effects of ginsenoside Rg1, cinnamic acid, and tanshinone IIA in osteosarcoma MG-63 cells: nuclear matrix downregulation and cytoplasmic trafficking of nucleophosmin. Int J Biochem Cell Biol 2008;40:1918-29. https://doi.org/10.1016/j.biocel.2008.01.031
  43. Ghosh AK, Brindisi M. Organic carbamates in drug design and medicinal chemistry. J Med Chem 2015;58:2895-940. https://doi.org/10.1021/jm501371s
  44. Kamal A, Kumar BA, Suresh P, Juvekar A, Zingde S. Synthesis of 4beta-carbamoyl epipodophyllotoxins as potential antitumour agents. Bioorg Med Chem 2011;19:2975-9. https://doi.org/10.1016/j.bmc.2011.03.030
  45. Liu XK, Ye BJ, Wu Y, Lin ZH, Zhao YQ, Piao HR. Synthesis and anti-tumor evaluation of panaxadiol derivatives. Eur J Med Chem 2011;46:1997-2002. https://doi.org/10.1016/j.ejmech.2011.02.022
  46. Wang Z, Li MY, Zhang ZH, Zuo HX, Wang JY, Xing Y, Ri M, Jin HL, Jin CH, Xu GH, Piao LX, Jiang CG, Ma J, Jin X. Panaxadiol inhibits programmed cell deathligand 1 expression and tumour proliferation via hypoxia-inducible factor (HIF)-1alpha and STAT3 in human colon cancer cells. Pharmacol Res 2020;155:104727. https://doi.org/10.1016/j.phrs.2020.104727
  47. Fan HY, Wang XK, Li X, Ji K, Du SH, Liu Y, Kong LL, Xu JC, Yang GQ, Chen DQ, Qi D. Curcumin, as a pleiotropic agent, improves doxorubicin-induced nephrotic syndrome in rats. J Ethnopharmacol 2020;250:112502. https://doi.org/10.1016/j.jep.2019.112502
  48. Yang Y, Guan D, Lei L, Lu J, Liu JQ, Yang G, Yan C, Zhai R, Tian J, Bi Y, Fu F, Wang H. H6, a novel hederagenin derivative, reverses multidrug resistance in vitro and in vivo. Toxicol Appl Pharmacol 2018;341:98-105. https://doi.org/10.1016/j.taap.2018.01.015
  49. Jin HR, Jin SZ, Cai XF, Li D, Wu X, Nan JX, Lee JJ, Jin X. Cryptopleurine targets NF-kappaB pathway, leading to inhibition of gene products associated with cell survival, proliferation, invasion, and angiogenesis. PLoS One 2012;7: e40355. https://doi.org/10.1371/journal.pone.0040355
  50. Dai ZC, Chen YF, Zhang M, Li SK, Yang TT, Shen L, Wang JX, Qian SS, Zhu HL, Ye YH. Synthesis and antifungal activity of 1,2,3-triazole phenylhydrazone derivatives. Org Biomol Chem 2015;13:477-86. https://doi.org/10.1039/C4OB01758G
  51. Jia Z, Zhu Q. 'Click' assembly of selective inhibitors for MAO-A. Bioorg Med Chem Lett 2010;20:6222-5. https://doi.org/10.1016/j.bmcl.2010.08.104
  52. Meldal M, Tornoe CW. Cu-catalyzed azide-alkyne cycloaddition. Chem Rev 2008;108:2952-3015. https://doi.org/10.1021/cr0783479
  53. Abdel-Atty MM, Farag NA, Kassab SE, Serya RAT, Abouzid KAM. Design, synthesis, 3D pharmacophore, QSAR, and docking studies of carboxylic acid derivatives as Histone Deacetylase inhibitors and cytotoxic agents. Bioorg Chem 2014;57:65-82. https://doi.org/10.1016/j.bioorg.2014.08.006
  54. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer 2011;11:393-410. https://doi.org/10.1038/nrc3064
  55. Lei KF, Wu ZM, Huang CH. Impedimetric quantification of the formation process and the chemosensitivity of cancer cell colonies suspended in 3D environment. Biosens Bioelectron 2015;74:878-85. https://doi.org/10.1016/j.bios.2015.07.060
  56. Zhou B, Zhan H, Tin L, Liu S, Xu J, Dong Y, Li X, Wu L, Guo W. TUFT1 regulates metastasis of pancreatic cancer through HIF1-Snail pathway induced epithelial-mesenchymal transition. Cancer Lett 2016;382:11-20. https://doi.org/10.1016/j.canlet.2016.08.017
  57. Umezu T, Tadokoro H, Azuma K, Yoshizawa S, Ohyashiki K, Ohyashiki JH. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014;124:3748-57.