Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Review Article

The Synthesis, Structural Modification and Mode of Anticancer Action of Evodiamine: A Review

Author(s): Meixia Fan and Lei Yao*

Volume 17, Issue 3, 2022

Published on: 07 March, 2022

Page: [284 - 296] Pages: 13

DOI: 10.2174/1574892817666211221165739

Price: $65

conference banner
Abstract

Background: Finding novel antitumor reagents from naturally occurring alkaloids is a widely accepted strategy. Evodiamine, a tryptamine indole alkaloid isolated from Evodia rutaecarpa, has a wide range of biological activities, such as anti-tumor, anti-inflammation, and anti-bacteria. Hence, research on the structural modification of evodiamine will facilitate the discovery of new antitumor drugs.

Objective: The recent advances in the synthesis of evodiamine, and studies on the drug design, biological activities, and structure-activity-relationships of its derivatives, published in patents and primary literature, are reviewed in this paper.

Methods: The literature, including patents and follow-up research papers from 2015 to 2020, related to evodiamine is searched in the Scifinder, PubMed, Espacenet, China National Knowledge Infrastructure (CNKI), and Wanfang databases. The keywords are evodiamine, synthesis, modification, anticancer, mechanism.

Results: The synthesis of evodiamine is summarized. Then, structural modifications of evodiamine are described, and the possible modes of action are discussed.

Conclusion: Evodiamine has a 6/5/6/6/6 ring system, and the structural modifications are focused on rings A, D, E, C5, N-13, and N-14. Some compounds show promising anticancer potentials and warrant further study.

Keywords: Antitumor, drug design, evodiamine, mode of action, patent, synthesis, structural modification.

[1]
Kamikado T, Murakoshi S, Tamura S. Structure elucidation and synthesis of alkaloids isolated from fruits of evodia rutaecarpa. Agric Biol Chem 1978; 42(8): 1515-9.
[2]
Jiang J, Hu C. Evodiamine: a novel anti-cancer alkaloid from Evodia rutaecarpa. Molecules 2009; 14(5): 1852-9.
[http://dx.doi.org/10.3390/molecules14051852] [PMID: 19471205]
[3]
Lv Q, Xue Y, Li G, et al. Beneficial effects of evodiamine on P2X(4)-mediated inflammatory injury of human umbilical vein endothelial cells due to high glucose. Int Immunopharmacol 2015; 28(2): 1044-9.
[http://dx.doi.org/10.1016/j.intimp.2015.08.020] [PMID: 26344431]
[4]
Zhang H, Yin L, Lu M, et al. Evodiamine attenuates adjuvant-induced arthritis in rats by inhibiting synovial inflammation and restoring the Th17/Treg balance. J Pharm Pharmacol 2020; 72(6): 798-806.
[http://dx.doi.org/10.1111/jphp.13238] [PMID: 32056223]
[5]
Fan X, Zhu JY, Sun Y, et al. Evodiamine inhibits Zymosan-induced inflammation in vitro and in vivo: inactivation of NF-κB by inhibiting IκBα phosphorylation. Inflammation 2017; 40(3): 1012-27.
[http://dx.doi.org/10.1007/s10753-017-0546-0] [PMID: 28337636]
[6]
Yamashita H, Kusudo T, Takeuchi T, et al. Dietary supplementation with evodiamine prevents obesity and improves insulin resistance in ageing mice. J Funct Foods 2015; 19: 320-9.
[http://dx.doi.org/10.1016/j.jff.2015.09.032]
[7]
Wang L, Wang K, Hou X. Hypolipidemic effect of evodiamine. CN 109288843, 2019.
[8]
Jiang ML, Zhang ZX, Li YZ, Wang XH, Yan W, Gong GQ. Antidepressant-like effect of evodiamine on chronic unpredictable mild stress rats. Neurosci Lett 2015; 588: 154-8.
[http://dx.doi.org/10.1016/j.neulet.2014.12.038] [PMID: 25545553]
[9]
Pan X, Wang M, Wu Y, et al. Identification of active ingredients in Wuzhuyu decoction improving migraine in mice by spectral efficiency association. Mol Med Rep 2015; 12(1): 1524-34.
[http://dx.doi.org/10.3892/mmr.2015.3506] [PMID: 25815775]
[10]
Lin J, Zhang X, Li C, et al. Evodiamine via targeting nNOS and AMPA receptor GluA1 inhibits nitroglycerin-induced migraine- like response. J Ethnopharmacol 2020; 254: 112727.
[http://dx.doi.org/10.1016/j.jep.2020.112727] [PMID: 32147481]
[11]
Li CG, Zeng QZ, Chen MY, et al. Evodiamine augments NLRP3 inflammasome activation and anti-bacterial responses through inducing α-tubulin acetylation. Front Pharmacol 2019; 10: 290.
[http://dx.doi.org/10.3389/fphar.2019.00290] [PMID: 30971927]
[12]
Chen X, Zhou S, Li H. Evodiamine alleviates severe pneumonia induced by methicillin-susceptible Staphylococcus aureus following cytomegalovirus reactivation through suppressing NF-κB and MAPKs. Int J Mol Med 2018; 42(6): 3247-55.
[http://dx.doi.org/10.3892/ijmm.2018.3929] [PMID: 30320369]
[13]
Wang D, Wang C, Liu L, Li S. Protective effects of evodiamine in experimental paradigm of Alzheimer’s disease. Cogn Neurodynamics 2018; 12(3): 303-13.
[http://dx.doi.org/10.1007/s11571-017-9471-z] [PMID: 29765479]
[14]
Wang MX, Lin L, Chen YD, et al. Evodiamine has therapeutic efficacy in ulcerative colitis by increasing Lactobacillus acidophilus levels and acetate production. Pharmacol Res 2020; 159: 104978.
[http://dx.doi.org/10.1016/j.phrs.2020.104978] [PMID: 32485282]
[15]
Zhao Z, Gong S, Wang S, Ma C. Effect and mechanism of evodiamine against ethanol-induced gastric ulcer in mice by suppressing Rho/NF-кB pathway. Int Immunopharmacol 2015; 28(1): 588-95.
[http://dx.doi.org/10.1016/j.intimp.2015.07.030] [PMID: 26225926]
[16]
Jiang XH, Wu QQ, Xiao Y, et al. Evodiamine prevents isoproterenol-induced cardiac fibrosis by regulating endothelial-to mesenchymal transition. Planta Med 2017; 83(9): 761-9.
[http://dx.doi.org/10.1055/s-0042-124044] [PMID: 28010025]
[17]
Li F, Yuan Y, Zhang N, et al. Evodiamine attenuates pressure overload-induced cardiac hypertrophy. Int J Clin Exp Med 2017; 10(7): 10202-13.
[18]
Bai X, Meng H, Ma L, Guo A. Inhibitory effects of evodiamine on human osteosarcoma cell proliferation and apoptosis. Oncol Lett 2015; 9(2): 801-5.
[http://dx.doi.org/10.3892/ol.2014.2791] [PMID: 25621054]
[19]
Meng ZJ, Wu N, Liu Y, et al. Evodiamine inhibits the proliferation of human osteosarcoma cells by blocking PI3K/Akt signaling. Oncol Rep 2015; 34(3): 1388-96.
[http://dx.doi.org/10.3892/or.2015.4084] [PMID: 26135006]
[20]
Han S, Woo JK, Jung Y, et al. Evodiamine selectively targets cancer stem-like cells through the p53-p21-Rb pathway. Biochem Biophys Res Commun 2016; 469(4): 1153-8.
[http://dx.doi.org/10.1016/j.bbrc.2015.12.066] [PMID: 26713361]
[21]
Zhao S, Xu K, Jiang R, et al. Evodiamine inhibits proliferation and promotes apoptosis of hepatocellular carcinoma cells via the Hippo-Yes-Associated Protein signaling pathway. Life Sci 2020; 251: 117424.
[http://dx.doi.org/10.1016/j.lfs.2020.117424] [PMID: 32057900]
[22]
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Lee SJ. Evodiamine suppresses survival, proliferation, migration and epithelial-mesenchymal transition of thyroid carcinoma cells. Anticancer Res 2018; 38(11): 6339-52.
[http://dx.doi.org/10.21873/anticanres.12992] [PMID: 30396956]
[23]
Zhou P, Li XP, Jiang R, et al. Evodiamine inhibits migration and invasion by Sirt1-mediated post-translational modulations in colorectal cancer. Anticancer Drugs 2019; 30(6): 611-7.
[http://dx.doi.org/10.1097/CAD.0000000000000760] [PMID: 30789361]
[24]
Yu H, Jin H, Gong W, Wang Z, Liang H. Pharmacological actions of multi-target-directed evodiamine. Molecules 2013; 18(2): 1826-43.
[http://dx.doi.org/10.3390/molecules18021826] [PMID: 23434865]
[25]
Gavaraskar K, Dhulap S, Hirwani RR. Therapeutic and cosmetic applications of Evodiamine and its derivatives- A patent review. Fitoterapia 2015; 106: 22-35.
[http://dx.doi.org/10.1016/j.fitote.2015.07.019] [PMID: 26255828]
[26]
Tan Q, Zhang J. Evodiamine and its role in chronic diseases. Adv Exp Med Biol 2016; 929: 315-28.
[http://dx.doi.org/10.1007/978-3-319-41342-6_14] [PMID: 27771931]
[27]
Hu X, Li D, Chu C, et al. Antiproliferative effects of alkaloid evodiamine and its derivatives. Int J Mol Sci 2018; 19(11): 3403.
[http://dx.doi.org/10.3390/ijms19113403] [PMID: 30380774]
[28]
Zhao Z, He X, Han W, et al. Genus Tetradium L.: A comprehensive review on traditional uses, phytochemistry, and pharmacological activities. J Ethnopharmacol 2019; 231: 337-54.
[http://dx.doi.org/10.1016/j.jep.2018.11.035] [PMID: 30472402]
[29]
Zhang Y, Wang J, Wang C, et al. Pharmacological basis for the use of evodiamine in Alzheimer’s disease: antioxidation and antiapoptosis. Int J Mol Sci 2018; 19(5): 1527.
[http://dx.doi.org/10.3390/ijms19051527] [PMID: 29883380]
[30]
Li X, Ge J, Zheng Q, Zhang J, Sun R, Liu R. Evodiamine and rutaecarpine from Tetradium ruticarpum in the treatment of liver diseases. Phytomedicine 2020; 68: 153180.
[http://dx.doi.org/10.1016/j.phymed.2020.153180] [PMID: 32092638]
[31]
Sun Q, Xie L, Song J, Li X. Evodiamine: A review of its pharmacology, toxicity, pharmacokinetics and preparation researches. J Ethnopharmacol 2020; 262: 113164.
[http://dx.doi.org/10.1016/j.jep.2020.113164] [PMID: 32738391]
[32]
Hu S, Xu J. Chemical synthesis of dl-evodiamine. Chem World 2007; 48(12): 758.
[33]
Huang G, Roos D, Stadtmüller P, Decker M. A simple heterocyclic fusion reaction and its application for expeditious syntheses of rutaecarpine and its analogs. Tetrahedron Lett 2014; 55(26): 3607-9.
[http://dx.doi.org/10.1016/j.tetlet.2014.04.120]
[34]
Yang Y, Guo H, Wang C, Zhang S. Advances in studies of anti-tumor activity of evodiamine. Chem Res 2011; 22(3): 22-5.
[35]
Pin F, Comesse S, Daïch A. Intramolecular N-aza-amidoalkylation in association with Witkope Winterfeldt oxidation as the key step to synthesize Luotonin-A analogues. Tetrahedron 2011; 67: 5564-71.
[http://dx.doi.org/10.1016/j.tet.2011.05.120]
[36]
Aoki K. Method for producing evodiamine. Patent CN110092786, 2019.
[37]
Wang ZX, Xiang JC, Wang M, Ma JT, Wu YD, Wu AX. One-Pot total synthesis of evodiamine and its analogues through a continuous biscyclization reaction. Org Lett 2018; 20(20): 6380-3.
[http://dx.doi.org/10.1021/acs.orglett.8b02667] [PMID: 30284836]
[38]
Xu W, Qian Y, Zhang W. The chemical synthesis of evodiamine. Zhejiang Chemical Industry 2007; 38(4): 15-6.
[39]
Rao KR, Raghunadh A, Mekala R, et al. Glyoxylic acid in the reaction of isatoic anhydride with amines: a rapid synthesis of 3-(un)substituted quinazolin-4(3H)-ones leading to rutaecarpine and evodiamine. Tetrahedron Lett 2014; 55(43): 6004-6.
[http://dx.doi.org/10.1016/j.tetlet.2014.09.011]
[40]
Zhou X, Ding Y, Huang H. Palladium-catalyzed carbonylative difunctionalization of C=N bond of azaarenes or imines to quinazolinones. Chem Asian J 2020; 15(11): 1678-82.
[http://dx.doi.org/10.1002/asia.202000359] [PMID: 32243085]
[41]
Clemenceau A, Wang Q, Zhu J. Silver nitrate-catalyzed isocyanide insertion/lactamization sequence to imidazolones and quinazolin-4-ones: development and application in natural product synthesis. Org Lett 2017; 19(18): 4872-5.
[http://dx.doi.org/10.1021/acs.orglett.7b02334] [PMID: 28841324]
[42]
Wang S, Li X, Zang J, et al. Palladium-catalyzed multistep tandem carbonylation/N-dealkylation/carbonylation reaction: access to isatoic anhydrides. J Org Chem 2020; 85(4): 2672-9.
[http://dx.doi.org/10.1021/acs.joc.9b02771] [PMID: 31887040]
[43]
Ji F, Wang S. Method for synthesizing evodiamine by three-method process including carbonylation reaction. CN107629051, 2018.
[44]
Wehle S, Espargaró A, Sataté R, Decker M. Investigation into the stability and reactivity of the pentacyclic alkaloid dehydroevodiamine and the benzanalog thereof. Tetrahedron 2016; 72(20): 2535-43.
[http://dx.doi.org/10.1016/j.tet.2016.03.059]
[45]
Nguyen NVT, Lee KR, Lee YJ, et al. Chiral high-performance liquid chromatographic separation of evodiamine enantiomers and rutaecarpine, isolated from Evodiae fructus. J Pharm Biomed Anal 2013; 81-82(7): 151-9.
[http://dx.doi.org/10.1016/j.jpba.2013.04.018] [PMID: 23666252]
[46]
Nakayama A, Kogure N, Kitajima M, Takayama H. Straightforward asymmetric total synthesis of (+)-evodiamine, a major indole alkaloid in herbal medicine “Wu Zhu Yu”. Heterocycles 2008; 76(1): 861-5.
[http://dx.doi.org/10.3987/COM-08-S(N)31]
[47]
Christodoulou MS, Sacchetti A, Ronchetti V, et al. Quinazolinecarboline alkaloid evodiamine as scaffold for targeting topoisomerase I and sirtuins. Bioorg Med Chem 2013; 21(22): 6920-8.
[http://dx.doi.org/10.1016/j.bmc.2013.09.030] [PMID: 24103429]
[48]
Fan J, Yang X, Xu X, Xie Y, Zhang X. Solubility of rutaecarpine and evodiamine in (ethanol + water) mixed solvents at temperatures from (288.2 to328.2) K. J Chem Thermodyn 2015; 83: 85-9.
[http://dx.doi.org/10.1016/j.jct.2014.12.004]
[49]
Zhang W, Guo J, Wang D, et al. Effect of CYP3A inducer/inhibitor on pharmacokinetics of five alkaloids in Evodiae Fructus. Chem Biol Interact 2020; 327: 109146.
[http://dx.doi.org/10.1016/j.cbi.2020.109146] [PMID: 32561335]
[50]
Zhang YT, Zhang DF, Ge NY, et al. Effect of evodiamine on CYP enzymes in rats by a cocktail method. Pharmacology 2016; 97(5-6): 218-23.
[http://dx.doi.org/10.1159/000443178] [PMID: 26872133]
[51]
Song S, Chen Z, Li S, Huang Y, Wan Y, Song H. Design, synthesis and evaluation of N13-substituted evodiamine derivatives against human cancer cell lines. Molecules 2013; 18(12): 15750-68.
[http://dx.doi.org/10.3390/molecules181215750] [PMID: 24352027]
[52]
Li D, Hua H, Li Z, et al. Preparation method of nitrate esters NO- donor evodiamine derivative and their application as anticancer drugs. Patent CN 105622608, 2016.
[53]
Li D, Hua H, Li Z, et al. Preparation of furazan NO-donor evodiamine conjugates useful as anticancer agents. Patent CN105622607, 2016.
[54]
Zhao N, Tian KT, Cheng KG, et al. Antiproliferative activity and apoptosis inducing effects of nitric oxide donating derivatives of evodiamine. Bioorg Med Chem 2016; 24(13): 2971-8.
[http://dx.doi.org/10.1016/j.bmc.2016.05.001] [PMID: 27178387]
[55]
Li D, Hua H, Li Z, et al. Preparation of evodiamine derivatives as antitumor agents. Patent CN107602557, 2018.
[56]
Li D, Hua H, Li Z, et al. Preparation method and application of evodiamine merged chlormethine derivatives having antitumor activity. Patent CN107674076, 2018.
[57]
Hu X, Wang Y, Xue J, et al. Design and synthesis of novel nitrogen mustard-evodiamine hybrids with selective antiproliferative activity. Bioorg Med Chem Lett 2017; 27(22): 4989-93.
[http://dx.doi.org/10.1016/j.bmcl.2017.10.014] [PMID: 29037951]
[58]
Li D, Hua H, Li Z, et al. Preparation of ADT-OH type H2S donor evodiamine derivative for treating cancer. Patent CN108191866, 2018.
[59]
Li D, Hua H, Li Z, et al. ADT-OH-type H2S donor-modified evodiamine derivative, its preparation method and application as antitumor agents. Patent CN108395431, 2018.
[60]
Hu X, Jiao R, Li H, et al. Antiproliferative hydrogen sulfide releasing evodiamine derivatives and their apoptosis inducing properties. Eur J Med Chem 2018; 151: 376-88.
[http://dx.doi.org/10.1016/j.ejmech.2018.04.009] [PMID: 29635169]
[61]
Liang C, Xia J, Song H, Zhou Z, Xue Y, Yao Q. Synthesis, in vitro and in vivo antitumor activity and docking studies of new evodiamine derivatives. J Chem Pharm Res 2014; 6(3): 1161-71.
[62]
Li Z, Dong G, Wang S, et al. Optical evodiamine derivatives: Asymmetric synthesis and antitumor activity. Chin Chem Lett 2015; 26(3): 267-71.
[http://dx.doi.org/10.1016/j.cclet.2014.11.011]
[63]
Chen S, Dong G, Wu S, Liu N, Zhang W, Sheng C. Novel fluorescent probes of 10-hydroxyevodiamine: autophagy and apoptosis-inducing anticancer mechanisms. Acta Pharm Sin B 2019; 9(1): 144-56.
[http://dx.doi.org/10.1016/j.apsb.2018.08.003] [PMID: 30766786]
[64]
Deng JD, Lei S, Jiang Y, et al. A concise synthesis and biological study of evodiamine and its analogues. Chem Commun (Camb) 2019; 55(21): 3089-92.
[http://dx.doi.org/10.1039/C9CC00434C] [PMID: 30785464]
[65]
Wang Z, Tan W, Li J, Deng J, Zhang H. Evodiamine derivative, synthesis method and application in preparing drug for treating tumor, inflammation and obesity. Patent CN109734714, 2019.
[66]
Hu X, Li N, Zhang J, Li J, Yuan J, Zhang X. Method for preparation of Evodiamine derivative 10-aminoevodiamine. Patent CN105175419, 2015.
[67]
Fang K, Dong G, Gong H, et al. Design, synthesis and biological evaluation of E-ring modified evodiamine derivatives as novel antitumor agents. Chin Chem Lett 2014; 25(7): 978-82.
[http://dx.doi.org/10.1016/j.cclet.2014.03.043]
[68]
Wang S, Fang K, Dong G, et al. Scaffold diversity inspired by the natural product evodiamine: discovery of highly potent and multitargeting antitumor agents. J Med Chem 2015; 58(16): 6678-96.
[http://dx.doi.org/10.1021/acs.jmedchem.5b00910] [PMID: 26226379]
[69]
Huang G, Kling B, Darras FH, Heilmann J, Decker M. Identification of a neuroprotective and selective butyrylcholinesterase inhibitor derived from the natural alkaloid evodiamine. Eur J Med Chem 2014; 81: 15-21.
[http://dx.doi.org/10.1016/j.ejmech.2014.05.002] [PMID: 24819955]
[70]
Dong G, Sheng C, Wang S, Miao Z, Yao J, Zhang W. Selection of evodiamine as a novel topoisomerase I inhibitor by structure-based virtual screening and hit optimization of evodiamine derivatives as antitumor agents. J Med Chem 2010; 53(21): 7521-31.
[http://dx.doi.org/10.1021/jm100387d] [PMID: 20942490]
[71]
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Lee SJ. Evodiamine in combination with histone deacetylase inhibitors has synergistic cytotoxicity in thyroid carcinoma cells. Endocrine 2019; 65(1): 110-20.
[http://dx.doi.org/10.1007/s12020-019-01885-1] [PMID: 31102069]
[72]
Dong G, Sheng C, Chen S, et al. Preparation of evodiamine derivatives as anti-tumor agents. Patent CN110066281, 2019.
[73]
He S, Dong G, Wang Z, et al. Discovery of novel multiacting topoisomerase I/II and histone deacetylase inhibitors. ACS Med Chem Lett 2015; 6(3): 239-43.
[http://dx.doi.org/10.1021/ml500327q] [PMID: 25815139]
[74]
Sheng C, Zhang W, Dong G, et al. Preparation method of evodiamine derivative with multi-target points anti-tumor activity and its application in preparation of drug for treating diseases caused by aberrant gene expression. Patent CN105524061, 2016.
[75]
Li X, Wu S, Dong G, et al. Natural product evodiamine with borate trigger unit: discovery of potent antitumor agents against colon cancer. ACS Med Chem Lett 2020; 11(4): 439-44.
[http://dx.doi.org/10.1021/acsmedchemlett.9b00513] [PMID: 32292547]
[76]
Luo H, Sheng C, Lai Z, Chen J, Zhang T. Preparation of evodiamine compounds as inhibitors of phosphodiesterase PDE5. Patent CN107141288, 2017.
[77]
Luo H, Sheng C, Lai Z, Chen J, Zhang T. Preparation of evodiamine compounds as inhibitors of phosphodiesterase PDE5. Patent WO2018188446, 2018.
[78]
Zhang T, Lai Z, Yuan S, et al. Discovery of evodiamine derivatives as highly selective PDE5 inhibitors targeting a unique allosteric pocket. J Med Chem 2020; 63(17): 9828-37.
[http://dx.doi.org/10.1021/acs.jmedchem.0c00983] [PMID: 32794708]
[79]
Guo H, Yang R, Ma J. Amino evodiamine derivative, its preparation method and its application. Patent CN 111620871, 2020.
[80]
Huang Y, Chen S, Wu S, Dong G, Sheng C. Evodiamine-inspired dual inhibitors of histone deacetylase 1 (HDAC1) and topoisomerase 2 (TOP2) with potent antitumor activity. Acta Pharm Sin B 2020; 10(7): 1294-308.
[http://dx.doi.org/10.1016/j.apsb.2019.11.011] [PMID: 32874829]
[81]
Sheng C, Dong G, Huang Y, Wu S, Liu N. Evodiamine derivative with multi-target anti-tumor activity, its preparation method and application. Patent CN108794474, 2018.
[82]
Pang S, Sun C, Gao S, Yang Y, Pan X, Zhang L. Evodiamine derivatives improve cognitive abilities in APPswe/PS1ΔE9 transgenic mouse models of Alzheimer’s disease. Animal Model Exp Med 2020; 3(2): 193-9.
[http://dx.doi.org/10.1002/ame2.12126] [PMID: 32613178]
[83]
Zhang L, Pan X, Yang Y, Zhang L, Dong W, Sun C. Use of oxa-evodiamine derivatives in antioxidant. Patent CN108853113, 2018.
[84]
Tang G, Xiong R, Peng Y, et al. Trimethoxy phenyl-containing evodiamine derivative and its application for treating cancer. Patent CN110483550, 2019.
[85]
Xu J, Qiu Y, Xu S, et al. Process for preparation of evodiamine derivative, and its application. Patent CN107365322, 2017.
[86]
Sun G, Zhang C, Song H, Guo J, Li M, Cao Y. WZY-321, a novel evodiamine analog, inhibits glioma cell growth in an autophagy-associated manner. Oncol Lett 2019; 17(2): 2465-72.
[PMID: 30675312]
[87]
Nie LF, Wang SS, Cao JG, et al. Straightforward synthesis, characterization, and cytotoxicity evaluation of hybrids of natural alkaloid evodiamine/rutaecarpine and thieno[2,3-d]pyrimidinones. J Asian Nat Prod Res 2020; 22(1): 69-82.
[http://dx.doi.org/10.1080/10286020.2018.1540599] [PMID: 30588834]
[88]
Vyawahare NS, Hadambar AA, Chothe AS, Jalnapurkar RR, Bhandare AM, Kathiravan MK. Effect of novel synthetic evodiamine analogue on sexual behavior in male rats. J Chem Biol 2012; 5(1): 35-42.
[http://dx.doi.org/10.1007/s12154-011-0067-5] [PMID: 23049645]
[89]
Wang L, Fang K, Cheng J, et al. Scaffold hopping of natural product evodiamine: discovery of a novel antitumor scaffold with excellent potency against colon cancer. J Med Chem 2020; 63(2): 696-713.
[http://dx.doi.org/10.1021/acs.jmedchem.9b01626] [PMID: 31880942]
[90]
Liang X, Wu Q, Luan S, et al. A comprehensive review of topoisomerase inhibitors as anticancer agents in the past decade. Eur J Med Chem 2019; 171: 129-68.
[http://dx.doi.org/10.1016/j.ejmech.2019.03.034] [PMID: 30917303]
[91]
Sheng C, Miao Z, Zhang W. Chapter 1-Topoisomerase I inhibitors derived from natural products: structure-activity relationships and antitumor potency. Stud Nat Prod Chem 2016; 47: 1-28.
[http://dx.doi.org/10.1016/B978-0-444-63603-4.00001-2]
[92]
Dong G, Wang S, Miao Z, et al. New tricks for an old natural product: discovery of highly potent evodiamine derivatives as novel antitumor agents by systemic structure-activity relationship analysis and biological evaluations. J Med Chem 2012; 55(17): 7593-613.
[http://dx.doi.org/10.1021/jm300605m] [PMID: 22867019]
[93]
Ramsay RR, Popovic-Nikolic MR, Nikolic K, Uliassi E, Bolognesi ML. A perspective on multi-target drug discovery and design for complex diseases. Clin Transl Med 2018; 7(1): 3.
[http://dx.doi.org/10.1186/s40169-017-0181-2] [PMID: 29340951]
[94]
Hemann MT, Lowe SW. The p53-Bcl-2 connection. Cell Death Differ 2006; 13(8): 1256-9.
[http://dx.doi.org/10.1038/sj.cdd.4401962] [PMID: 16710363]
[95]
Liu N, Li Y, Chen G, Ge K. Evodiamine induces reactive oxygen species-dependent apoptosis and necroptosis in human melanoma A-375 cells. Oncol Lett 2020; 20(4): 121.
[http://dx.doi.org/10.3892/ol.2020.11983] [PMID: 32863934]
[96]
Zhao LC, Li J, Liao K, et al. Evodiamine induces apoptosis and inhibits migration of HCT-116 human colorectal cancer cells. Int J Mol Sci 2015; 16(11): 27411-21.
[http://dx.doi.org/10.3390/ijms161126031] [PMID: 26580615]
[97]
Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med 2016; 67(1): 11-28.
[http://dx.doi.org/10.1146/annurev-med-062913-051343] [PMID: 26473415]
[98]
Lin L, Ren L, Wen L, Wang Y, Qi J. Effect of evodiamine on the proliferation and apoptosis of A549 human lung cancer cells. Mol Med Rep 2016; 14(3): 2832-8.
[http://dx.doi.org/10.3892/mmr.2016.5575] [PMID: 27485202]
[99]
Guo XX, Li XP, Zhou P, et al. Evodiamine induces apoptosis in SMMC-7721 and HepG2 cells by suppressing NOD1 signal pathway. Int J Mol Sci 2018; 19(11): 3419.
[http://dx.doi.org/10.3390/ijms19113419] [PMID: 30384473]
[100]
Wei WT, Chen H, Wang ZH, et al. Enhanced antitumor efficacy of gemcitabine by evodiamine on pancreatic cancer via regulating PI3K/Akt pathway. Int J Biol Sci 2012; 8(1): 1-14.
[http://dx.doi.org/10.7150/ijbs.8.1] [PMID: 22211100]
[101]
Lv Z, Zhao D, Liu R, Guo J, Zhang M. Evodiamine inhibits proliferation of human papillary thyroid cancer cell line K1 by regulating of PI3K/Akt signaling pathway. Int J Clin Exp Med 2016; 9(8): 15216-25.
[102]
Wu PK, Becker A, Park JI. Growth inhibitory signaling of the Raf/MEK/ERK pathway. Int J Mol Sci 2020; 21(15): 5436.
[http://dx.doi.org/10.3390/ijms21155436] [PMID: 32751750]
[103]
Wang S, Wang L, Shi Z, Zhong Z, Chen M, Wang Y. Evodiamine synergizes with doxorubicin in the treatment of chemoresistant human breast cancer without inhibiting P-glycoprotein. PLoS One 2014; 9(5): e97512.
[http://dx.doi.org/10.1371/journal.pone.0097512] [PMID: 24830744]
[104]
Shi L, Yang F, Luo F, et al. Evodiamine exerts anti-tumor effects against hepatocellular carcinoma through inhibiting β-catenin-mediated angiogenesis. Tumour Biol 2016; 37(9): 12791-803.
[http://dx.doi.org/10.1007/s13277-016-5251-3] [PMID: 27449032]
[105]
Hu CY, Wu HT, Su YC, Lin CH, Chang CJ, Wu CL. Evodiamine exerts an anti-hepatocellular carcinoma activity through a WWOX-dependent pathway. Molecules 2017; 22(7): 1175.
[http://dx.doi.org/10.3390/molecules22071175] [PMID: 28708106]
[106]
Yang S, Chen J, Tan T, et al. Evodiamine exerts anticancer effects against 143B and MG63 cells through the Wnt/β-Catenin signaling pathway. Cancer Manag Res 2020; 12: 2875-88.
[http://dx.doi.org/10.2147/CMAR.S238093] [PMID: 32425601]
[107]
Kim H, Yu Y, Choi S, et al. Evodiamine eliminates colon cancer stem cells via suppressing Notch and Wnt signaling. Molecules 2019; 24(24): 4520.
[http://dx.doi.org/10.3390/molecules24244520] [PMID: 31835579]
[108]
Jiang ZB, Huang JM, Xie YJ, et al. Evodiamine suppresses non-small cell lung cancer by elevating CD8+ T cells and downregulating the MUC1-C/PD-L1 axis. J Exp Clin Cancer Res 2020; 39(1): 249.
[http://dx.doi.org/10.1186/s13046-020-01741-5] [PMID: 33208183]
[109]
Chen TC, Chien CC, Wu MS, Chen YC. Evodiamine from Evodia rutaecarpa induces apoptosis via activation of JNK and PERK in human ovarian cancer cells. Phytomedicine 2016; 23(1): 68-78.
[http://dx.doi.org/10.1016/j.phymed.2015.12.003] [PMID: 26902409]
[110]
Hwang ST, Um JY, Chinnathambi A, et al. Evodiamine mitigates cellular growth and promotes apoptosis by targeting the c-Met pathway in prostate cancer cells. Molecules 2020; 25(6): 1320.
[http://dx.doi.org/10.3390/molecules25061320] [PMID: 32183146]
[111]
Sun C, Zhang G, Luan S, et al. Evodiamine inhibits the proliferation of leukemia cell line K562 by regulating peroxisome proliferators-activated receptor gamma (PPARγ) pathway. J Recept Signal Transduct Res 2016; 36(4): 422-8.
[http://dx.doi.org/10.3109/10799893.2015.1122040] [PMID: 26671528]
[112]
Feng K, Ren Y, Li R. Combined pharmacophore-guided 3D-QSAR, molecular docking and molecular dynamics studies for evodiamine analogs as DNA topoisomerase I inhibitors. J Taiwan Inst Chem Eng 2017; 78: 81-95.
[http://dx.doi.org/10.1016/j.jtice.2017.06.027]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy