Antiproliferative and palliative activity of flavonoids in colorectal cancer
Tóm tắt
Từ khóa
Tài liệu tham khảo
Merrill, 2011, Risk-adjusted colon and rectal cancer incidence rates in the United States, Dis. Colon Rectum, 54, 1301, 10.1097/DCR.0b013e3182242bd3
Bray, 2013, Global estimates of cancer prevalence for 27 sites in the adult population in 2008, Int. J. Cancer, 132, 1133, 10.1002/ijc.27711
Sung, 2021, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries, CA Cancer J. Clin., 71, 209, 10.3322/caac.21660
Ferlay, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012, Int. J. Cancer, 136, E359, 10.1002/ijc.29210
Humphries, 2008, Colonic crypt organization and tumorigenesis, Nat. Rev. Cancer, 8, 415, 10.1038/nrc2392
Bruce, 1996, The colonic protein fermentation and insulin resistance hypotheses for colon cancer etiology: experimental tests using precursor lesions, Eur. J. Cancer Prev., 5, 41, 10.1097/00008469-199612002-00007
Kinzler, 1996, Lessons from hereditary colorectal cancer, Cell, 87, 159, 10.1016/S0092-8674(00)81333-1
Eom, 2021, Prognostic factors affecting disease-free survival and overall survival in T4 colon cancer, Ann. Coloproctol., 37, 259, 10.3393/ac.2020.00759.0108
Xiong, 2004, Treatment of colorectal cancer metastasis: the role of chemotherapy, Cancer Metastasis Rev., 23, 145, 10.1023/A:1025823315416
Gustavsson, 2015, A review of the evolution of systemic chemotherapy in the management of colorectal cancer, Clin. Colorectal Cancer, 14, 1, 10.1016/j.clcc.2014.11.002
Mahipal, 2016, Role of biologics in first-line treatment of colorectal cancer, J. Oncol. Pract., 12, 1219, 10.1200/JOP.2016.018382
Marín, 2017, De novo biosynthesis of apigenin, luteolin, and eriodictyol in the Actinomycete Streptomyces albus and production improvement by feeding and spore conditioning, Front. Microbiol., 8, 921, 10.3389/fmicb.2017.00921
Redondo-Blanco, 2017, New insights toward colorectal cancer chemotherapy using natural bioactive compounds, Front. Pharmacol., 8, 109, 10.3389/fphar.2017.00109
Hossain, 2021, Natural-derived molecules as a potential adjuvant in chemotherapy: normal cell protectors and cancer cell sensitizers, Anticancer Agents Med. Chem.
Basu, 2020, In vitro and in vivo effects of flavonoids on peripheral neuropathic pain, Molecules, 25, 1171, 10.3390/molecules25051171
Siddiqui, 2021, Flavonoids alleviate peripheral neuropathy induced by anticancer drugs, Cancers, 13
Neveu, 2010, Phenol-Explorer: an online comprehensive database on polyphenol contents in foods, Database: J. Biol. Databases Curation, 2010, 24, 10.1093/database/bap024
Qi, 2020, Apigenin induces apoptosis and counteracts cisplatin-induced chemoresistance via Mcl-1 in ovarian cancer cells, Exp. Ther. Med., 20, 1329, 10.3892/etm.2020.8880
Zhang, 2021, Combined effect of chrysin and apigenin on inhibiting the development and progression of colorectal cancer by suppressing the activity of P38-MAPK/AKT pathway, IUBMB Life, 73, 774, 10.1002/iub.2456
Tong, 2019, Apigenin inhibits epithelial-mesenchymal transition of human colon cancer cells through NF-κB/Snail signaling pathway, Biosci. Rep., 39, 39, 10.1042/BSR20190452
Ai, 2017, Apigenin inhibits colonic inflammation and tumorigenesis by suppressing STAT3-NF-κB signaling, Oncotarget, 8, 100216, 10.18632/oncotarget.22145
Bian, 2020, Inhibitory effects of apigenin on tumor carcinogenesis by altering the gut microbiota, Mediat. Inflamm., 2020, 10.1155/2020/7141970
Imran, 2019, Luteolin, a flavonoid, as an anticancer agent: a review, Biomed. Pharmacother., 112, 10.1016/j.biopha.2019.108612
Ganai, 2021, Anticancer activity of the plant flavonoid luteolin against preclinical models of various cancers and insights on different signalling mechanisms modulated, Phytother. Res., 35, 3509, 10.1002/ptr.7044
Ju, 2007, A critical role of luteolin-induced reactive oxygen species in blockage of tumor necrosis factor-activated nuclear factor-kappaB pathway and sensitization of apoptosis in lung cancer cells, Mol. Pharmacol., 71, 1381, 10.1124/mol.106.032185
Zhang, 2016, Modulatory effect of luteolin on redox homeostasis and inflammatory cytokines in a mouse model of liver cancer, Oncol. Lett., 12, 4767, 10.3892/ol.2016.5291
Baby, 2021, Cogent role of flavonoids as key orchestrators of chemoprevention of hepatocellular carcinoma: a review, J. Food Biochem., 45, e13761, 10.1111/jfbc.13761
Kang, 2017, Luteolin induces apoptotic cell death via antioxidant activity in human colon cancer cells, Int. J. Oncol., 51, 1169, 10.3892/ijo.2017.4091
Zuo, 2018, The dietary flavone luteolin epigenetically activates the Nrf2 pathway and blocks cell transformation in human colorectal cancer HCT116 cells, J. Cell. Biochem., 119, 9573, 10.1002/jcb.27275
Lim, 2017, Naringenin-induced apoptotic cell death in prostate cancer cells is mediated via the PI3K/AKT and MAPK signaling pathways, J. Cell. Biochem., 118, 1118, 10.1002/jcb.25729
Totta, 2004, Mechanisms of Naringenin-induced Apoptotic Cascade in cancer cells: involvement of estrogen receptor α and β signalling., IUBMB Life (Int. Union Biochem. Mol. Biol. Life), 56, 491, 10.1080/15216540400010792
Song, 2016, Naringenin-mediated ATF3 expression contributes to apoptosis in human colon cancer, Biomol. Ther., 24, 140, 10.4062/biomolther.2015.109
Manthey, 2001, Biological properties of citrus flavonoids pertaining to cancer and inflammation, Curr. Med. Chem., 8, 135, 10.2174/0929867013373723
Islam, 2020, The pharmacological and biological roles of eriodictyol, Arch. Pharm. Res., 43, 582, 10.1007/s12272-020-01243-0
Liang, 2017, Eriodictyol 7–O-β-D glucopyranoside from Coreopsis tinctoria Nutt. ameliorates lipid disorders via protecting mitochondrial function and suppressing lipogenesis, Mol. Med. Rep., 16, 1298, 10.3892/mmr.2017.6743
Zhang, 2020, Eriodictyol exerts potent anticancer activity against A549 human lung cancer cell line by inducing mitochondrial-mediated apoptosis, G2/M cell cycle arrest and inhibition of m-TOR/PI3K/Akt signalling pathway, Arch. Med. Sci., 16, 446, 10.5114/aoms.2019.85152
Mariyappan, 2017, Effect of eriodictyol on preneoplastic lesions, oxidative stress and bacterial enzymes in 1,2-dimethyl hydrazine-induced colon carcinogenesis, Toxicol. Res., 6, 678, 10.1039/C7TX00074J
Stevens, 2004, Xanthohumol and related prenylflavonoids from hops and beer: to your good health!, Phytochemistry, 65, 1317, 10.1016/j.phytochem.2004.04.025
Stevens, 1999, Fate of xanthohumol and related prenylflavonoids from hops to beer, J. Agric. Food Chem., 47, 2421, 10.1021/jf990101k
Nookandeh, 2004, Xanthohumol metabolites in faeces of rats, Phytochemistry, 65, 561, 10.1016/j.phytochem.2003.11.016
Nowak, 2020, Pharmacokinetics of xanthohumol in rats of both sexes after oral and intravenous administration of pure xanthohumol and prenylflavonoid extract, Adv. Clin. Exp. Med., 29, 1101, 10.17219/acem/126293
Zhao, 2005, Prenylflavonoids and phloroglucinol derivatives from hops (Humulus lupulus), J. Nat. Prod., 68, 43, 10.1021/np0499113
Sridhar, 2005, Identification of novel angiogenesis inhibitors, Bioorg. Med. Chem. Lett., 15, 4125, 10.1016/j.bmcl.2005.06.001
Wilson, 2003, Isozymes of mammalian hexokinase: structure, subcellular localization and metabolic function, J. Exp. Biol., 206, 2049, 10.1242/jeb.00241
Liu, 2019, Xanthohumol inhibits colorectal cancer cells via downregulation of hexokinases ii-mediated glycolysis, Int. J. Biol. Sci., 15, 2497, 10.7150/ijbs.37481
Jiang, 2018, Anticancer activity and mechanism of xanthohumol: a prenylated flavonoid from hops (Humulus lupulus L.), Front. Pharmacol., 9, 1, 10.3389/fphar.2018.00530
Liu, 2019, JBUON, 24, 2442
Festa, 2011, Xanthohumol induces apoptosis in human malignant glioblastoma cells by increasing reactive oxygen species and activating MAPK pathways, J. Nat. Prod., 74, 2505, 10.1021/np200390x
Hussong, 2005, A safety study of oral xanthohumol administration and its influence on fertility in Sprague Dawley rats, Mol. Nutr. Food Res., 49, 861, 10.1002/mnfr.200500089
Liu, 2020, Xanthohumol protects against Azoxymethane-induced colorectal cancer in Sprague-Dawley rats, Environ. Toxicol., 35, 136, 10.1002/tox.22849
Nurgali, 2018, Editorial: adverse effects of cancer chemotherapy: anything new to improve tolerance and reduce sequelae?, Front. Pharmacol., 9, 245, 10.3389/fphar.2018.00245
Ngai, 2019, TOXview: a novel graphical presentation of cancer treatment toxicity profiles, Acta Oncol., 58, 1138, 10.1080/0284186X.2019.1601256
Davies, 2021, Anti-cancer therapy made easier: a 25-year update, Intern. Med. J., 51, 473, 10.1111/imj.14878
Smith, 2020, An overview of acute gastrointestinal side effects of systemic anti-cancer therapy and their management, Best Pract. Res. Clin. Gastroenterol., 48–49
Li, 2021, Emerging pharmacological and non-pharmacological therapeutics for prevention and treatment of chemotherapy-induced peripheral neuropathy, Cancers, 13
Laforgia, 2021, Peripheral neuropathy under oncologic therapies: a literature review on pathogenetic mechanisms, Int. J. Mol. Sci., 22
Tanay, 2021, A systematic review of behavioural and exercise interventions for the prevention and management of chemotherapy-induced peripheral neuropathy symptoms, J. Cancer Surviv.
Cavaletti, 2019, Toxic neuropathy consortium of the peripheral nerve society chemotherapy-induced peripheral neurotoxicity: a multifaceted, still unsolved issue, J. Peripher. Nerv. Syst., 24, S6
Molina Prats, 2017, The therapeutic effects of apigenin and dexamethasone on 5-fluorouracil-induced oral mucositis - a pilot study using a Syrian hamster model, J. Oral. Pathol. Med., 46, 142, 10.1111/jop.12473
García-Lafuente, 2009, Flavonoids as anti-inflammatory agents: implications in cancer and cardiovascular disease, Inflamm. Res., 58, 537, 10.1007/s00011-009-0037-3
Boeing, 2020, Luteolin prevents irinotecan-induced intestinal mucositis in mice through antioxidant and anti-inflammatory properties, Br. J. Pharmacol., 177, 2393, 10.1111/bph.14987
Vetuschi, 2018, PPAR-γ with its anti-inflammatory and anti-fibrotic action could be an effective therapeutic target in IBD, Eur. Rev. Med. Pharmacol. Sci., 22, 8839
Sadraei, 2017, Anti-inflammatory effect of apigenin and hydroalcoholic extract of Dracocephalum kotschyi on acetic acid-induced colitis in rats, Res. Pharm. Sci., 12, 322, 10.4103/1735-5362.212050
Akinrinde, 2020, Exacerbation of diclofenac-induced gastroenterohepatic damage by concomitant exposure to sodium fluoride in rats: protective role of luteolin, Drug Chem. Toxicol., 1
Dong, 2021, The active ingredients identification and antidiarrheal mechanism analysis of Plantago asiatica L. superfine powder, Front. Pharmacol., 11, 10.3389/fphar.2020.612478
Dou, 2013, Protective effect of naringenin against experimental colitis via suppression of Toll-like receptor 4/NF-κB signalling, Br. J. Nutr., 110, 599, 10.1017/S0007114512005594
Pereira, 2017, Gastroprotective activity of the resin from Virola oleifera, Pharm. Biol., 55, 472, 10.1080/13880209.2016.1251467
Di Carlo, 1993, Inhibition of intestinal motility and secretion by flavonoids in mice and rats: structure-activity relationships, J. Pharm. Pharmacol., 45, 1054, 10.1111/j.2042-7158.1993.tb07180.x
Blandizzi, 2007, Enteric alpha-2 adrenoceptors: pathophysiological implications in functional and inflammatory bowel disorders, Neurochem. Int., 51, 282, 10.1016/j.neuint.2007.05.013
Chamie, 2021, Peripherally acting μ-opioid receptor antagonists in the management of postoperative ileus: a clinical review, J. Gastrointest. Surg., 25, 293, 10.1007/s11605-020-04671-x
Sadraei, 2016, Antidiarrhoeal assessment of hydroalcoholic and hexane extracts of Dracocephalum kotschyi Boiss. and apigenin in mice, Res. Pharm. Sci., 11, 200
Katavic, 2007, Flavonoids as opioid receptor ligands: identification and preliminary structure-activity relationships, J. Nat. Prod., 70, 1278, 10.1021/np070194x
Dou, 2013, Protective effect of naringenin against experimental colitis via suppression of Toll-like receptor 4/NF-κB signalling, Br. J. Nutr., 110, 599, 10.1017/S0007114512005594
Challa, 2015, Surgical animal models of neuropathic pain: pros and cons, Int. J. Neurosci., 125, 170, 10.3109/00207454.2014.922559
Hashemzaei, 2017, Effects of luteolin and luteolin-morphine co-administration on acute and chronic pain and sciatic nerve ligated-induced neuropathy in mice, J. Complement. Integr. Med., 14, 20160066, 10.1515/jcim-2016-0066
Wu, 2014, Quercetin, luteolin and epigallocatechin gallate alleviate TXNIP and NLRP3-mediated inflammation and apoptosis with regulation of AMPK in endothelial cells, Eur. J. Pharmacol., 745, 59, 10.1016/j.ejphar.2014.09.046
Biessels, 2014, Phenotyping animal models of diabetic neuropathy: a consensus statement of the diabetic neuropathy study group of the EASD (Neurodiab), J. Peripher. Nerv. Syst., 19, 77, 10.1111/jns5.12072
Kitada, 2016, Rodent models of diabetic nephropathy: their utility and limitations, Int. J. Nephrol. Renov. Dis., 9, 279, 10.2147/IJNRD.S103784
Singh, 2020, Naringenin ameliorates diabetic neuropathic pain by modulation of oxidative-nitrosative stress, cytokines and MMP-9 levels, Food Funct., 11, 4548, 10.1039/C9FO00881K
Chung, 2019, Antinociceptive and anti-inflammatory effects of the citrus flavanone naringenin, Ci ji yi xue za zhi = Tzu-chi Med. J., 31, 81
Pinheiro, 2012, Antinociceptive effect of the Orbignya speciosa Mart. (Babassu) leaves: evidence for the involvement of apigenin, Life Sci., 91, 293, 10.1016/j.lfs.2012.06.013
Rossato, 2011, Eriodictyol: a flavonoid antagonist of the TRPV1 receptor with antioxidant activity, Biochem. Pharmacol., 81, 544, 10.1016/j.bcp.2010.11.004
Cho, 2012, Neuroprotective and anti-inflammatory effects of flavonoids isolated from Rhus verniciflua in neuronal HT22 and microglial BV2 cell lines, Food Chem. Toxicol., 50, 1940, 10.1016/j.fct.2012.03.052
Lou, 2012, Eriodictyol protects against H(2)O(2)-induced neuron-like PC12 cell death through activation of Nrf2/ARE signaling pathway, Neurochem. Int., 61, 251, 10.1016/j.neuint.2012.05.013
Zhao, 2019, Apigenin attenuates acrylonitrile-induced neuro-inflammation in rats: involved of inactivation of the TLR4/NF-κB signaling pathway, Int. Immunopharmacol., 75, 10.1016/j.intimp.2019.105697
Li, 2015, Naringenin inhibits dendritic cell maturation and has therapeutic effects in a murine model of collagen-induced arthritis, J. Nutr. Biochem., 26, 1467, 10.1016/j.jnutbio.2015.07.016
Shi, 2015, Anti-arthritic activity of luteolin in Freund’s complete adjuvant-induced arthritis in rats by suppressing P2X4 pathway, Chem. Biol. Interact., 226, 82, 10.1016/j.cbi.2014.10.031
Fernández, 2016, Colon microbiota fermentation of dietary prebiotics towards short-chain fatty acids and their roles as anti-inflammatory and antitumour agents: a review, J. Funct. Foods, 25, 511, 10.1016/j.jff.2016.06.032
Fernández, 2015, Healthy effects of prebiotics and their metabolites against intestinal diseases and colorectal cancer, AIMS Microbiol., 1, 48, 10.3934/microbiol.2015.1.48
Liu, 2006, Analysis of P53 mutations and their expression in 56 colorectal cancer cell lines, Proc. Natl. Acad. Sci. USA, 103, 976, 10.1073/pnas.0510146103
He, 2015, Functional repair of p53 mutation in colorectal cancer cells using trans-splicing, Oncotarget, 6, 2034, 10.18632/oncotarget.2988
Pranteda, 2020, The p38 MAPK signaling activation in colorectal cancer upon therapeutic treatments, Int. J. Mol. Sci., 21, 2773, 10.3390/ijms21082773
Alves, 2015, Colorectal cancer-related mutant KRAS alleles function as positive regulators of autophagy, Oncotarget, 6, 30787, 10.18632/oncotarget.5021
Kumar, 2014, KRAS G13D mutation and sensitivity to cetuximab or panitumumab in a colorectal cancer cell line model, Gastrointest. Cancer Res., 7, 23
Liu, 2011, Eriodictyol inhibits RSK2-ATF1 signaling and suppresses EGF-induced neoplastic cell transformation, J. Biol. Chem., 286, 2057, 10.1074/jbc.M110.147306
Jang, 2019, Luteolin shifts oxaliplatin-induced cell cycle arrest at G₀/G₁ to apoptosis in HCT116 human colorectal carcinoma cells, Nutrients, 11, 770, 10.3390/nu11040770