Generic placeholder image

Current Drug Targets

Editor-in-Chief

ISSN (Print): 1389-4501
ISSN (Online): 1873-5592

Review Article

Potential Phytochemicals for Prevention of Familial Breast Cancer with BRCA Mutations

Author(s): Aliasgar Fakruddin Shahiwala* and Gazala Afreen Khan

Volume 24, Issue 6, 2023

Published on: 14 April, 2023

Page: [521 - 531] Pages: 11

DOI: 10.2174/1389450124666230314110800

Price: $65

Abstract

Breast cancer has remained a global challenge and the second leading cause of cancer mortality in women and family history. Hereditary factors are some of the major risk factors associated with breast cancer. Out of total breast cancer cases, 5-10% account only for familial breast cancer, and nearly 50% of all hereditary breast cancer are due to BRCA1/BRCA2 germline mutations. BRCA1/2 mutations play an important role not only in determining the clinical prognosis of breast cancer but also in the survival curves. Since this risk factor is known, a significant amount of the healthcare burden can be reduced by taking preventive measures among people with a known history of familial breast cancer. There is increasing evidence that phytochemicals of nutrients and supplements help in the prevention and cure of BRCA-related cancers by different mechanisms such as limiting DNA damage, altering estrogen metabolism, or upregulating expression of the normal BRCA allele, and ultimately enhancing DNA repair. This manuscript reviews different approaches used to identify potential phytochemicals to mitigate the risk of familial breast cancer with BRCA mutations. The findings of this review can be extended for the prevention and cure of any BRCAmutated cancer after proper experimental and clinical validation of the data.

Graphical Abstract

[1]
Golubnitschaja O, Debald M, Yeghiazaryan K, Kuhn W, Pešta M, Costigliola V. Breast cancer epidemic in the early twenty-first century: evaluation of risk factors, cumulative questionnaires and recommendations for preventive measures. Tumour Biol 2016; 37(10): 12941-57.
[http://dx.doi.org/10.1007/s13277-016-5168-x]
[2]
Kmietowicz Z. Genetic screening could improve breast cancer prevention, study finds. BMJ 2015; 350(apr09 13): h1879.
[http://dx.doi.org/10.1136/bmj.h1879] [PMID: 25858913]
[3]
Mavaddat N, Pharoah PDP, Michailidou K, et al. Prediction of breast cancer risk based on profiling with common genetic variants. J Natl Cancer Inst 2015; 107(5)djv036
[http://dx.doi.org/10.1093/jnci/djv036] [PMID: 25855707]
[4]
Genetic Testing for Breast Cancer. Available from: https://www.nationalbreastcancer.org/genetic-testing-for-breast-cancer
[5]
Rennert G, Bisland-Naggan S, Barnett-Griness O, Bar-Joseph N, Zhang S, Rennert HS. Clinical outcomes of breast cancer in carriers of BRCA1 and BRCA2 mutations. N Engl J Med 2009; 357(2): 115-23.
[6]
Malone KE, Daling JR, Doody DR, et al. Prevalence and predictors of BRCA1 and BRCA2 mutations in a population-based study of breast cancer in white and black American women ages 35 to 64 years. Cancer Res 2006; 66(16): 8297-308.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-0503] [PMID: 16912212]
[7]
Abu-Helalah M, Azab B, Mubaidin R, Ali D, Jafar H, Alshraideh H. BRCA1 and BRCA2 genes mutations among high risk breast cancer patients in Jordan. Sci Rep 2020; 10(1): 17573.
[http://dx.doi.org/10.1038/s41598-020-74250-2]
[8]
Schon K, Tischkowitz M. Clinical implications of germline mutations in breast cancer: TP53. Breast Cancer Res Treat 2018; 167: 417.
[9]
Lalloo F, Evans DG. Familial breast cancer. Clin Genet 2012; 82(2): 105-14.
[http://dx.doi.org/10.1111/j.1399-0004.2012.01859.x] [PMID: 22356477]
[10]
Malkin D, Li FP, Strong LC, et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990; 250(4985): 1233-8.
[http://dx.doi.org/10.1126/science.1978757] [PMID: 1978757]
[11]
Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA -. JAMA 2017; 317(23): 2402-16.
[http://dx.doi.org/10.1001/jama.2017.7112] [PMID: 28632866]
[12]
Schmidt MK, van den Broek AJ, Tollenaar RAEM, et al. Breast cancer survival of BRCA1/BRCA2 mutation carriers in a hospital-based cohort of young women. J Natl Cancer Inst 2017; 109(8): djw329.
[http://dx.doi.org/10.1093/jnci/djw329] [PMID: 28376189]
[13]
Zhu Y, Wu J, Zhang C, Sun S, Zhang J, Liu W. BRCA mutations and survival in breast cancer: An updated systematic review and meta-analysis. Oncotarget 2016; 7(43): 70113-27.
[http://dx.doi.org/10.18632/oncotarget.12158]
[14]
Scott RJ. DNA double strand break repair and its association with inherited predispositions to breast cancer. Hered Cancer Clin Pract 2004; 2(1): 37-43.
[http://dx.doi.org/10.1186/1897-4287-2-1-37]
[15]
MC S. Gene panel testing for hereditary breast cancer. Med J Aust 2016; 204(5): 188-90.
[16]
Peshkin BN, Isaacs C, Finch C, Kent S, Schwartz MD. Tamoxifen as chemoprevention in BRCA1 and BRCA2 mutation carriers with breast cancer: a pilot survey of physicians. J Clin Oncol 2003; 21(23): 4322-8.
[http://dx.doi.org/10.1200/JCO.2003.02.107] [PMID: 14645421]
[17]
Liu RH. Potential synergy of phytochemicals in cancer prevention: mechanism of action. J Nutr 2004; 134(12) (Suppl.): 3479S-85S.
[http://dx.doi.org/10.1093/jn/134.12.3479S] [PMID: 15570057]
[18]
WHO Global report on traditional and complementary medicine. 2019. Available from: http://apps.who.int/bookorders
[19]
Sun J, Chu YF, Wu X, Liu RH. Antioxidant and antiproliferative activities of common fruits. J Agric Food Chem 2002; 50(25): 7449-54.
[http://dx.doi.org/10.1021/jf0207530] [PMID: 12452674]
[20]
Venugopal R, Liu RH. Phytochemicals in diets for breast cancer prevention: The importance of resveratrol and ursolic acid. Food Sci Hum Wellness 2012; 1(1): 1-13.
[http://dx.doi.org/10.1016/j.fshw.2012.12.001]
[21]
Dragsted LO, Strube M, Larsen JC. Cancer-protective factors in fruits and vegetables: biochemical and biological background. Pharmacol Toxicol 1993; 72 (Suppl. 1): 116-35.
[http://dx.doi.org/10.1111/j.1600-0773.1993.tb01679.x] [PMID: 8474974]
[22]
Roy A, Bhatia KS. in silico analysis of plumbagin against cyclin-dependent kinases receptor. Vegetos 2021; 34: 50-6.
[http://dx.doi.org/10.1007/s42535-020-00169-8]
[23]
Garg S, Anand A, Lamba Y, Roy A. Molecular docking analysis of selected phytochemicals against SARS-CoV-2 Mpro receptor. Vegetos 2020; 33: 766-81.
[24]
Meng X-Y, Zhang H-X, Mezei M, Cui M. Molecular Docking: A powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des 2011; 7(2): 146.
[http://dx.doi.org/10.2174/157340911795677602]
[25]
Glide: A new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J Med Chem 2004; 47(7): 1739-49.
[26]
Balogun TA, Buliaminu KD, Chukwudozie OS, Tiamiyu ZA, Idowu TJ. Anticancer Potential of Moringa oleifera on BRCA-1 Gene: Systems Biology. Bioinform Biol Insight 2021; 15: 11779322211010703.
[27]
Singh Bhatia K, Garg S, Anand A, Roy A. Evaluation of different phytochemicals against BRCA2 receptor. Biointer Res Appl Chem 2021; 12(2): 1670-81.
[28]
Exploration and evaluation of bioactive phytocompounds against BRCA proteins by in silico approach. J Biomol Struct Dyn 2021; 39(15): 1-15.
[29]
Couch FJ, Weber BL. Mutations and Polymorphisms in the familial early-onset breast cancer (BRCA1) gene. Hum Mutat 1996; 8(1): 8-18.
[http://dx.doi.org/10.1002/humu.1380080102] [PMID: 8807330]
[30]
Bartek J, Lukas J. DNA damage checkpoints: from initiation to recovery or adaptation. Curr Opin Cell Biol 2007; 19(2): 238-45.
[http://dx.doi.org/10.1016/j.ceb.2007.02.009] [PMID: 17303408]
[31]
Bartek J, Lukas C, Lukas J. Checking on DNA damage in S phase. Nat Rev Mol Cell Biol 2004; 5(10): 792-804.
[http://dx.doi.org/10.1038/nrm1493]
[32]
Shiloh Y. ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer 2003; 3(3): 155-68.
[http://dx.doi.org/10.1038/nrc1011]
[33]
Bartkova J, Hořejší Z, Koed K, Krämer A, Tort F, Zieger K. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 2005; 434(7035): 864-70.
[http://dx.doi.org/10.1038/nature03482]
[34]
Gorgoulis VG, Vassiliou L-VF, Karakaidos P, Zacharatos P, Kotsinas A, Liloglou T. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 2005; 434(7035): 907-13.
[http://dx.doi.org/10.1038/nature03485]
[35]
Tommiska J, Bartkova J, Heinonen M, Hautala L, Kilpivaara O, Eerola H. The DNA damage signalling kinase ATM is aberrantly reduced or lost in BRCA1/BRCA2-deficient and ER/PR/ERBB2-triple-negative breast cancer. Oncogene 2007; 27: 2501-6.
[36]
Sarris E, Saif M, Syrigos K. The Biological Role of PI3K Pathway in Lung Cancer. Pharmaceuticals (Basel) 2012; 5(11): 1236-64.
[http://dx.doi.org/10.3390/ph5111236] [PMID: 24281308]
[37]
Morgan T, Koreckij T, Corey E. Targeted therapy for advanced prostate cancer: inhibition of the PI3K/Akt/mTOR pathway. Curr Cancer Drug Targets 2009; 9(2): 237-49.
[http://dx.doi.org/10.2174/156800909787580999] [PMID: 19275762]
[38]
LC C. Targeting a common collaborator in cancer development. Sci Transl Med 2010; 2(48): 48.
[39]
Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov 2009; 8(8): 627-44.
[http://dx.doi.org/10.1038/nrd2926] [PMID: 19644473]
[40]
Fumarola C, Bonelli MA, Petronini PG, Alfieri RR. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem Pharmacol 2014; 90(3): 197-207.
[http://dx.doi.org/10.1016/j.bcp.2014.05.011] [PMID: 24863259]
[41]
Grunt TW, Mariani GL. Novel approaches for molecular targeted therapy of breast cancer: interfering with PI3K/AKT/mTOR signaling. Curr Cancer Drug Targets 2013; 13(2): 188-204.
[http://dx.doi.org/10.2174/1568009611313020008] [PMID: 23215720]
[42]
Xiang T, Ohashi A, Huang Y, et al. Negative Regulation of AKT Activation by BRCA1. Cancer Res 2008; 68(24): 10040-4.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-3009] [PMID: 19074868]
[43]
Tyagi A, Singh RP, Agarwal C, Siriwardana S, Sclafani RA, Agarwal R. Resveratrol causes Cdc2-tyr15 phosphorylation via ATM/ATR-Chk1/2-Cdc25C pathway as a central mechanism for S phase arrest in human ovarian carcinoma Ovcar-3 cells. Carcinogenesis 2005; 26(11): 1978-87.
[http://dx.doi.org/10.1093/carcin/bgi165] [PMID: 15975956]
[44]
Rashid A, Liu C, Sanli T, Tsiani E, Singh G, Bristow RG. Resveratrol enhances prostate cancer cell response to ionizing radiation. Modulation of the AMPK, Akt and mTOR pathways. Radiat Oncol 2011; 6: 144.
[45]
Lee J-H, Guo Z, Myler LR, Zheng S, Paull TT. Direct activation of ATM by resveratrol under oxidizing conditions. PLoS One 2014; 9(6): e97969.
[46]
Saiko P, Graser G, Giessrigl B, et al. Digalloylresveratrol, a novel resveratrol analog inhibits the growth of human pancreatic cancer cells. Invest New Drugs 2013; 31(5): 1115-24.
[http://dx.doi.org/10.1007/s10637-013-0009-x] [PMID: 23943154]
[47]
Sahu RP, Batra S, Srivastava SK. Activation of ATM/Chk1 by curcumin causes cell cycle arrest and apoptosis in human pancreatic cancer cells. Br J Cancer 2009; 100: 1425-33.
[48]
Lan YH, Wu YC, Wu KW, et al. Death receptor 5-mediated TNFR family signaling pathways modulate γ-humulene-induced apoptosis in human colorectal cancer HT29 cells. Oncol Rep 2011; 25(2): 419-24.
[PMID: 21152878]
[49]
Costantino VV, Mansilla SF, Speroni J, et al. The sesquiterpene lactone dehydroleucodine triggers senescence and apoptosis in association with accumulation of DNA damage markers. PLoS One 2013; 8(1): e53168.
[http://dx.doi.org/10.1371/journal.pone.0053168] [PMID: 23341930]
[50]
Cho JH, Lee JG, Yang YI, et al. Eupatilin, a dietary flavonoid, induces G2/M cell cycle arrest in human endometrial cancer cells. Food Chem Toxicol 2011; 49(8): 1737-44.
[http://dx.doi.org/10.1016/j.fct.2011.04.019] [PMID: 21554918]
[51]
Shyur LF, Lee SH, Chang ST, Lo CP, Kuo YH, Wang SY. Taiwanin A inhibits MCF-7 cancer cell activity through induction of oxidative stress, upregulation of DNA damage checkpoint kinases, and activation of p53 and FasL/Fas signaling pathways. Phytomedicine 2010; 18(1): 16-24.
[http://dx.doi.org/10.1016/j.phymed.2010.06.005] [PMID: 20637573]
[52]
Hsu YL, Chia CC, Chen PJ, Huang SE, Huang SC, Kuo PL. Shallot and licorice constituent isoliquiritigenin arrests cell cycle progression and induces apoptosis through the induction of ATM/p53 and initiation of the mitochondrial system in human cervical carcinoma HeLa cells. Mol Nutr Food Res 2009; 53(7): 826-35.
[http://dx.doi.org/10.1002/mnfr.200800288] [PMID: 19536869]
[53]
Hsu YL, Uen YH, Chen Y, Liang HL, Kuo PL. Tricetin, a dietary flavonoid, inhibits proliferation of human breast adenocarcinoma mcf-7 cells by blocking cell cycle progression and inducing apoptosis. J Agric Food Chem 2009; 57(18): 8688-95.
[http://dx.doi.org/10.1021/jf901053x] [PMID: 19705844]
[54]
Chen CY, Hsu YL, Tsai YC, Kuo PL. Kotomolide A arrests cell cycle progression and induces apoptosis through the induction of ATM/p53 and the initiation of mitochondrial system in human non-small cell lung cancer A549 cells. Food Chem Toxicol 2008; 46(7): 2476-84.
[http://dx.doi.org/10.1016/j.fct.2008.04.016] [PMID: 18511169]
[55]
Fini L, Hotchkiss E, Fogliano V, et al. Chemopreventive properties of pinoresinol-rich olive oil involve a selective activation of the ATM-p53 cascade in colon cancer cell lines. Carcinogenesis 2008; 29(1): 139-46.
[http://dx.doi.org/10.1093/carcin/bgm255] [PMID: 17999988]
[56]
Guo J, Zhao W, Hao W, Ren G, Lu J, Chen X. Cucurbitacin B induces DNA damage, G2/M phase arrest, and apoptosis mediated by reactive oxygen species (ROS) in leukemia K562 cells. Anticancer Agents Med Chem 2014; 14(8): 1146-53.
[http://dx.doi.org/10.2174/1871520614666140601220915] [PMID: 24893803]
[57]
Guo J, Wu G, Bao J, Hao W, Lu J, Chen X. Cucurbitacin B induced ATM-mediated DNA damage causes G2/M cell cycle arrest in a ROS-dependent manner. PLoS One 2014; 9(2)e88140
[http://dx.doi.org/10.1371/journal.pone.0088140] [PMID: 24505404]
[58]
Lee JG, Kim JH, Ahn JH, Lee KT, Baek NI, Choi JH. Jaceosidin, isolated from dietary mugwort (Artemisia princeps), induces G2/M cell cycle arrest by inactivating cdc25C-cdc2 via ATM-Chk1/2 activation. Food Chem Toxicol 2013; 55: 214-21.
[http://dx.doi.org/10.1016/j.fct.2012.12.026] [PMID: 23274058]
[59]
Li-Weber M. Molecular mechanisms and anti-cancer aspects of the medicinal phytochemicals rocaglamides (=flavaglines). Int J Cancer 2015; 137(8): 1791-9.
[http://dx.doi.org/10.1002/ijc.29013] [PMID: 24895251]
[60]
Neumann J, Boerries M, Köhler R, et al. The natural anticancer compound rocaglamide selectively inhibits the G1-S-phase transition in cancer cells through the ATM/ATR-mediated Chk1/2 cell cycle checkpoints. Int J Cancer 2014; 134(8): 1991-2002.
[http://dx.doi.org/10.1002/ijc.28521] [PMID: 24150948]
[61]
Arango D, Parihar A, Villamena FA, et al. Apigenin induces DNA damage through the PKCδ-dependent activation of ATM and H2AX causing down-regulation of genes involved in cell cycle control and DNA repair. Biochem Pharmacol 2012; 84(12): 1571-80.
[http://dx.doi.org/10.1016/j.bcp.2012.09.005] [PMID: 22985621]
[62]
Zhong Y, Krisanapun C, Lee SH, et al. Molecular targets of apigenin in colorectal cancer cells: Involvement of p21, NAG-1 and p53. Eur J Cancer 2010; 46(18): 3365-74.
[http://dx.doi.org/10.1016/j.ejca.2010.07.007] [PMID: 20709524]
[63]
Xie Q, Bai Q, Zou LY, et al. Genistein inhibits DNA methylation and increases expression of tumor suppressor genes in human breast cancer cells. Genes Chromosomes Cancer 2014; 53(5): 422-31.
[http://dx.doi.org/10.1002/gcc.22154] [PMID: 24532317]
[64]
Zhang Z, Wang CZ, Du GJ, et al. Genistein induces G2/M cell cycle arrest and apoptosis via ATM/p53-dependent pathway in human colon cancer cells. Int J Oncol 2013; 43(1): 289-96.
[http://dx.doi.org/10.3892/ijo.2013.1946] [PMID: 23686257]
[65]
Tyagi M, Patro BS, Chattopadhyay S. Mechanism of the malabaricone C-induced toxicity to the MCF-7 cell line. Free Radic Res 2014; 48(4): 466-77.
[http://dx.doi.org/10.3109/10715762.2014.886328] [PMID: 24456233]
[66]
Tyagi M, Bhattacharyya R, Bauri AK, Patro BS, Chattopadhyay S. DNA damage dependent activation of checkpoint kinase-1 and mitogen-activated protein kinase-p38 are required in malabaricone C-induced mitochondrial cell death. Biochim Biophys Acta, Gen Subj 2014; 1840(3): 1014-27.
[http://dx.doi.org/10.1016/j.bbagen.2013.11.020] [PMID: 24291689]
[67]
Tyagi A, Singh RP, Agarwal C, Agarwal R. Silibinin activates p53-caspase 2 pathway and causes caspase-mediated cleavage of Cip1/p21 in apoptosis induction in bladder transitional-cell papilloma RT4 cells: evidence for a regulatory loop between p53 and caspase 2. Carcinogenesis 2006; 27(11): 2269-80.
[http://dx.doi.org/10.1093/carcin/bgl098] [PMID: 16777994]
[68]
Jeong YJ, Cho HJ, Magae J, Lee IK, Park KG, Chang YC. Ascofuranone suppresses EGF-induced HIF-1α protein synthesis by inhibition of the Akt/mTOR/p70S6K pathway in MDA-MB-231 breast cancer cells. Toxicol Appl Pharmacol 2013; 273(3): 542-50.
[http://dx.doi.org/10.1016/j.taap.2013.09.027] [PMID: 24096035]
[69]
Wang Y, Liu Y, Du X, Ma H, Yao J. Berberine reverses doxorubicin resistance by inhibiting autophagy through the PTEN/Akt/mTOR signaling pathway in breast cancer. OncoTargets Ther 2020; 13: 1909-19.
[http://dx.doi.org/10.2147/OTT.S241632] [PMID: 32184626]
[70]
Lu J, Sun D, Gao S, Gao Y, Ye J, Liu P. Cyclovirobuxine D induces autophagy-associated cell death via the Akt/mTOR pathway in MCF-7 human breast cancer cells. J Pharmacol Sci 2014; 125(1): 74-82.
[http://dx.doi.org/10.1254/jphs.14013FP] [PMID: 24758922]
[71]
Shrivastava S, Kulkarni P, Thummuri D, et al. Piperlongumine, an alkaloid causes inhibition of PI3 K/Akt/mTOR signaling axis to induce caspase-dependent apoptosis in human triple-negative breast cancer cells. Apoptosis 2014; 19(7): 1148-64.
[http://dx.doi.org/10.1007/s10495-014-0991-2] [PMID: 24729100]
[72]
Fultang N, Illendula A, Chen B, et al. Strictinin, a novel ROR1-inhibitor, represses triple negative breast cancer survival and migration via modulation of PI3K/AKT/GSK3ß activity. PLoS One 2019; 14(5): e0217789.
[http://dx.doi.org/10.1371/journal.pone.0217789] [PMID: 31150511]
[73]
Guo Y, Pei X. Tetrandrine-induced autophagy in MDA-MB-231 triple-negative breast cancer cell through the inhibition of PI3K/AKT/mTOR signaling. Evid Based Complement Alternat Med 2019; 2019: 7517431.
[74]
Hsieh CJ, Kuo PL, Hou MF, et al. Wedelolactone inhibits breast cancer-induced osteoclastogenesis by decreasing Akt/mTOR signaling. Int J Oncol 2015; 46(2): 555-62.
[http://dx.doi.org/10.3892/ijo.2014.2769] [PMID: 25421824]
[75]
Bratton MR, Martin EC, Elliott S, et al. Glyceollin, a novel regulator of mTOR/p70S6 in estrogen receptor positive breast cancer. J Steroid Biochem Mol Biol 2015; 150: 17-23.
[http://dx.doi.org/10.1016/j.jsbmb.2014.12.014] [PMID: 25771071]
[76]
He X, Wang Y, Zhu J, Orloff M, Eng C. Resveratrol enhances the anti-tumor activity of the mTOR inhibitor rapamycin in multiple breast cancer cell lines mainly by suppressing rapamycin-induced AKT signaling. Cancer Lett 2011; 301(2): 168-76.
[http://dx.doi.org/10.1016/j.canlet.2010.11.012] [PMID: 21168265]
[77]
Tanshinone IIA inhibits HIF-1α and VEGF expression in breast cancer cells via mTOR/p70S6K/RPS6/4E-BP1 signaling pathway. PLoS One 2015; 10(2): e0117440.
[78]
Melkamu T, Zhang X, Tan J, Zeng Y, Kassie F. Alteration of microRNA expression in vinyl carbamate-induced mouse lung tumors and modulation by the chemopreventive agent indole-3-carbinol. Carcinogenesis 2010; 31(2): 252-8.
[http://dx.doi.org/10.1093/carcin/bgp208] [PMID: 19748927]
[79]
Soy isoflavone genistein-mediated downregulation of miR-155 contributes to the anticancer effects of genistein. Nutr Cancer 2016; 68(1): 154-64.
[80]
Zhu H, Wu H, Liu X, Evans BR, Medina DJ, Liu C-G. Role of MicroRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem Pharmacol 2008; 76(5): 582.
[81]
Rodrigues AC, Li X, Radecki L, et al. MicroRNA expression is differentially altered by xenobiotic drugs in different human cell lines. Biopharm Drug Dispos 2011; 32(6): 355-67.
[http://dx.doi.org/10.1002/bdd.764] [PMID: 21796641]
[82]
Iida K, Fukushi J, Matsumoto Y, et al. miR-125b develops chemoresistance in Ewing sarcoma/primitive neuroectodermal tumor. Cancer Cell Int 2013; 13(1): 21.
[http://dx.doi.org/10.1186/1475-2867-13-21] [PMID: 23497288]
[83]
Zhuo L, Liu J, Wang B, Gao M, Huang A. Differential miRNA expression profiles in hepatocellular carcinoma cells and drug-resistant sublines. Oncol Rep 2013; 29(2): 555-62.
[http://dx.doi.org/10.3892/or.2012.2155] [PMID: 23229111]
[84]
Yin W, Wang P, Wang X, Song W, Cui X, Yu H. Identification of microRNAs and mRNAs associated with multidrug resistance of human laryngeal cancer Hep-2 cells. Braz J Med Biol Res 2013; 46(6): 546-.
[http://dx.doi.org/10.1590/1414-431X20131662]
[85]
Du L, Borkowski R, Zhao Z, et al. A high-throughput screen identifies miRNA inhibitors regulating lung cancer cell survival and response to paclitaxel. RNA Biol 2013; 10(11): 1700-13.
[http://dx.doi.org/10.4161/rna.26541] [PMID: 24157646]
[86]
Zhong S, Chen X, Wang D, et al. MicroRNA expression profiles of drug-resistance breast cancer cells and their exosomes. Oncotarget 2016; 7(15): 19601-9.
[http://dx.doi.org/10.18632/oncotarget.7481] [PMID: 26910922]
[87]
Zhong S, Ma T, Zhang X, et al. MicroRNA expression profiling and bioinformatics analysis of dysregulated microRNAs in vinorelbine-resistant breast cancer cells. Gene 2015; 556(2): 113-8.
[http://dx.doi.org/10.1016/j.gene.2014.11.046] [PMID: 25445394]
[88]
Ooi AGL, Sahoo D, Adorno M, Wang Y, Weissman IL, Park CY. MicroRNA-125b expands hematopoietic stem cells and enriches for the lymphoid-balanced and lymphoid-biased subsets. Proc Natl Acad Sci USA 2010; 107(50): 21505-10.
[http://dx.doi.org/10.1073/pnas.1016218107] [PMID: 21118986]
[89]
Qin W, Shi Y, Zhao B, et al. miR-24 regulates apoptosis by targeting the open reading frame (ORF) region of FAF1 in cancer cells. PLoS One 2010; 5(2)e9429
[http://dx.doi.org/10.1371/journal.pone.0009429] [PMID: 20195546]
[90]
Bommer GT, Gerin I, Feng Y, et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol 2007; 17(15): 1298-307.
[http://dx.doi.org/10.1016/j.cub.2007.06.068] [PMID: 17656095]
[91]
Zhang J, Guo H, Qian G, et al. MiR-145, a new regulator of the DNA Fragmentation Factor-45 (DFF45)-mediated apoptotic network. Mol Cancer 2010; 9(1): 211.
[http://dx.doi.org/10.1186/1476-4598-9-211] [PMID: 20687965]
[92]
Körner C, Keklikoglou I, Bender C, Wörner A, Münstermann E, Wiemann S. MicroRNA-31 sensitizes human breast cells to apoptosis by direct targeting of protein kinase C epsilon (PKCepsilon). J Biol Chem 2013; 288(12): 8750-61.
[http://dx.doi.org/10.1074/jbc.M112.414128] [PMID: 23364795]
[93]
Huang N, Wu J, Qiu W, et al. MiR-15a and miR-16 induce autophagy and enhance chemosensitivity of Camptothecin. Cancer Biol Ther 2015; 16(6): 941-8.
[http://dx.doi.org/10.1080/15384047.2015.1040963] [PMID: 25945419]
[94]
Zeng CW, Zhang XJ, Lin KY, et al. Camptothecin induces apoptosis in cancer cells via microRNA-125b-mediated mitochondrial pathways. Mol Pharmacol 2012; 81(4): 578-86.
[http://dx.doi.org/10.1124/mol.111.076794] [PMID: 22252650]
[95]
Wang N, Zhu M, Tsao SW, Man K, Zhang Z, Feng Y. MiR-23a-mediated inhibition of topoisomerase 1 expression potentiates cell response to etoposide in human hepatocellular carcinoma. Mol Cancer 2013; 12(1): 119.
[http://dx.doi.org/10.1186/1476-4598-12-119] [PMID: 24103454]
[96]
Biersack B. Non-coding RNA/microRNA-modulatory dietary factors and natural products for improved cancer therapy and prevention: Alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins. Non-coding RNA Res 2016; 1(1): 51-63.
[97]
Bitarte N, Bandres E, Boni V, et al. MicroRNA-451 is involved in the self-renewal, tumorigenicity, and chemoresistance of colorectal cancer stem cells. Stem Cells 2011; 29(11): 1661-71.
[http://dx.doi.org/10.1002/stem.741] [PMID: 21948564]
[98]
Ruzzo A, Graziano F, Vincenzi B, et al. High let-7a microRNA levels in KRAS-mutated colorectal carcinomas may rescue anti-EGFR therapy effects in patients with chemotherapy-refractory metastatic disease. Oncologist 2012; 17(6): 823-9.
[http://dx.doi.org/10.1634/theoncologist.2012-0081] [PMID: 22584434]
[99]
Boren T, Xiong Y, Hakam A, et al. MicroRNAs and their target messenger RNAs associated with ovarian cancer response to chemotherapy. Gynecol Oncol 2009; 113(2): 249-55.
[http://dx.doi.org/10.1016/j.ygyno.2009.01.014] [PMID: 19237188]
[100]
Gmeiner WH, Reinhold WC, Pommier Y. Genome-wide mRNA and microRNA profiling of the NCI 60 cell-line screen and comparison of FdUMP[10] with fluorouracil, floxuridine, and topoisomerase 1 poisons. Mol Cancer Ther 2010; 9(12): 3105-14.
[http://dx.doi.org/10.1158/1535-7163.MCT-10-0674] [PMID: 21159603]
[101]
Uboldi S, Calura E, Beltrame L, et al. A systems biology approach to characterize the regulatory networks leading to trabectedin resistance in an in vitro model of myxoid liposarcoma. PLoS One 2012; 7(4)e35423
[http://dx.doi.org/10.1371/journal.pone.0035423] [PMID: 22523595]
[102]
Hu H, Li K, Wang X, et al. Set9, NF-κB, and microRNA-21 mediate berberine-induced apoptosis of human multiple myeloma cells. Acta Pharmacol Sin 2013; 34(1): 157-66.
[http://dx.doi.org/10.1038/aps.2012.161] [PMID: 23247593]
[103]
Lo TF, Tsai WC, Chen ST. MicroRNA-21-3p, a berberine-induced miRNA, directly down-regulates human methionine adenosyltransferases 2A and 2B and inhibits hepatoma cell growth. PLoS One 2013; 8(9): e75628.
[http://dx.doi.org/10.1371/journal.pone.0075628] [PMID: 24098708]
[104]
Hagiwara K, Gailhouste L, Yasukawa K, Kosaka N, Ochiya T. A robust screening method for dietary agents that activate tumour-suppressor microRNAs. Sci Rep 2015; 5(1): 14697.
[http://dx.doi.org/10.1038/srep14697] [PMID: 26423775]
[105]
Li H, Xie S, Liu X, et al. Matrine alters microRNA expression profiles in SGC-7901 human gastric cancer cells. Oncol Rep 2014; 32(5): 2118-26.
[http://dx.doi.org/10.3892/or_xxxxxxxx] [PMID: 25174809]
[106]
Wang G, Liu G, Ye Y, Fu Y, Zhang X. Upregulation of miR-34a by diallyl disulfide suppresses invasion and induces apoptosis in SGC-7901 cells through inhibition of the PI3K/Akt signaling pathway. Oncol Lett 2016; 11(4): 2661-7.
[http://dx.doi.org/10.3892/ol.2016.4266] [PMID: 27073535]
[107]
Xiao X, Chen B, Liu X, et al. Diallyl disulfide suppresses SRC/Ras/ERK signaling-mediated proliferation and metastasis in human breast cancer by up-regulating miR-34a. PLoS One 2014; 9(11)e112720
[http://dx.doi.org/10.1371/journal.pone.0112720] [PMID: 25396727]
[108]
Tang H, Kong Y, Guo J, et al. Diallyl disulfide suppresses proliferation and induces apoptosis in human gastric cancer through Wnt-1 signaling pathway by up-regulation of miR-200b and miR-22. Cancer Lett 2013; 340(1): 72-81.
[http://dx.doi.org/10.1016/j.canlet.2013.06.027] [PMID: 23851184]
[109]
Appari M, Babu KR, Kaczorowski A, Gros W, Her I. Sulforaphane, quercetin and catechins complement each other in elimination of advanced pancreatic cancer by miR-let-7 induction and K-ras inhibition. Int J Oncol 2014; 45(4): 1391-400.
[http://dx.doi.org/10.3892/ijo.2014.2539] [PMID: 25017900]
[110]
Liu CM, Peng CY, Liao YW, et al. Sulforaphane targets cancer stemness and tumor initiating properties in oral squamous cell carcinomas via miR-200c induction. J Formos Med Assoc 2017; 116(1): 41-8.
[http://dx.doi.org/10.1016/j.jfma.2016.01.004] [PMID: 26879838]
[111]
Li Q, Yao Y, Eades G, Liu Z, Zhang Y, Zhou Q. Downregulation of miR-140 promotes cancer stem cell formation in basal-like early stage breast cancer. Oncogene 2014; 33(20): 2589-600.
[http://dx.doi.org/10.1038/onc.2013.226] [PMID: 23752191]
[112]
Xiao J, Gong AY, Eischeid AN, et al. miR-141 modulates androgen receptor transcriptional activity in human prostate cancer cells through targeting the small heterodimer partner protein. Prostate 2012; 72(14): 1514-22.
[http://dx.doi.org/10.1002/pros.22501] [PMID: 22314666]
[113]
Izzotti A, Calin GA, Steele VE, et al. Chemoprevention of cigarette smoke-induced alterations of MicroRNA expression in rat lungs. Cancer Prev Res (Phila) 2010; 3(1): 62-72.
[http://dx.doi.org/10.1158/1940-6207.CAPR-09-0202] [PMID: 20051373]
[114]
Pogribny I, Tryndyak V, Ross S, Beland F. Differential expression of microRNAs during hepatocarcinogenesis induced by methyl deficiency in rats. Nutr Rev 2008; 66(S1): 33-5.
[http://dx.doi.org/10.1111/j.1753-4887.2008.00064.x]
[115]
Marsit CJ, Eddy K, Kelsey KT. MicroRNA responses to cellular stress. Cancer Res 2006; 66(22): 10843-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-1894] [PMID: 17108120]
[116]
Kutay H, Bai S, Datta J, et al. Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. J Cell Biochem 2006; 99(3): 671-8.
[http://dx.doi.org/10.1002/jcb.20982] [PMID: 16924677]
[117]
Venturelli S, Sinnberg TW, Berger A, et al. Epigenetic impacts of ascorbate on human metastatic melanoma cells. Front Oncol 2014; 4: 227.
[http://dx.doi.org/10.3389/fonc.2014.00227] [PMID: 25202679]
[118]
Singh B, Ronghe AM, Chatterjee A, Bhat NK, Bhat HK. MicroRNA-93 regulates NRF2 expression and is associated with breast carcinogenesis. Carcinogenesis 2013; 34(5): 1165-72.
[http://dx.doi.org/10.1093/carcin/bgt026] [PMID: 23492819]
[119]
Wang B, Teng Y, Liu Q. MicroRNA-153 Regulates NRF2 Expression and is Associated with Breast Carcinogenesis. Clin Lab 2016; 62(01+02/2016): 39-47.
[http://dx.doi.org/10.7754/Clin.Lab.2015.150518] [PMID: 27012032]
[120]
Fustier P, Corre L, Le , et al. Resveratrol increases BRCA1 and BRCA2 mRNA expression in breast tumour cell lines. Br J Cancer 2003; 89(1): 168.
[http://dx.doi.org/10.1038/sj.bjc.6600983]
[121]
Kim GY, Suh J, Jang J-H, Kim D-H, Park OJ, Park SK. Genistein inhibits proliferation of BRCA1 mutated breast cancer cells: The GPR30-Akt axis as a potential target. J Cancer Prev 2019; 24(4): 197.
[122]
Tommasi C, Pellegrino B, Boggiani D, et al. Biological Role and Clinical Implications of microRNAs in BRCA Mutation Carriers. Front Oncol 2021; 11700853
[http://dx.doi.org/10.3389/fonc.2021.700853] [PMID: 34552867]
[123]
Morales S, Monzo M, Navarro A. Epigenetic regulation mechanisms of microRNA expression. Biomol Concepts 2017; 8(5-6): 203-12.
[http://dx.doi.org/10.1515/bmc-2017-0024] [PMID: 29161231]
[124]
Gasparri ML, Casorelli A, Bardhi E, Besharat AR, Savone D, Ruscito I. Beyond circulating microRNA biomarkers: Urinary microRNAs in ovarian and breast cancer. Tumor Bio 2017; 39(5): 1010428317695525.
[125]
Setti G, Pezzi ME, Viani MV, Pertinhez TA, Cassi D, Magnoni C. Salivary MicroRNA for diagnosis of cancer and systemic diseases: A systematic review. Int J Mol Sci 2020; 21(3): 907.
[http://dx.doi.org/10.3390/ijms21030907]
[126]
Nassar F, Chamandi G, Tfaily M, Zgheib N, Nasr R. Peripheral blood-based biopsy for breast cancer risk prediction and early detection. Front Med (Lausanne) 2020; 7: 28.
[http://dx.doi.org/10.3389/fmed.2020.00028]
[127]
Murria Estal R, Palanca Suela S, de Juan Jiménez I, et al. MicroRNA signatures in hereditary breast cancer. Breast Cancer Res Treat 2013; 142(1): 19-30.
[http://dx.doi.org/10.1007/s10549-013-2723-7] [PMID: 24129975]
[128]
Erturk E, Cecener G, Tezcan G, et al. BRCA mutations cause reduction in miR-200c expression in triple negative breast cancer. Gene 2015; 556(2): 163-9.
[http://dx.doi.org/10.1016/j.gene.2014.11.047] [PMID: 25445393]
[129]
Hagiwara K, Kosaka N, Yoshioka Y, Takahashi R, Takeshita F, Ochiya T. Stilbene derivatives promote Ago2-dependent tumour-suppressive microRNA activity. Sci Rep 2012; 2(1): 314.
[http://dx.doi.org/10.1038/srep00314] [PMID: 22423322]
[130]
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1(1): 12-34.
[http://dx.doi.org/10.1016/j.ncrna.2016.07.001] [PMID: 30159408]
[131]
Gallardo M, Kemmerling U, Aguayo F, Bleak TC, Muñoz JP, Calaf GM. Curcumin rescues breast cells from epithelial-mesenchymal transition and invasion induced by anti-miR-34a. Int J Oncol 2020; 56(2): 480-93. [Internet].
[PMID: 31894298]
[132]
Kronski E, Fiori ME, Barbieri O, et al. miR181b is induced by the chemopreventive polyphenol curcumin and inhibits breast cancer metastasis via down-regulation of the inflammatory cytokines CXCL1 and -2. Mol Oncol 2014; 8(3): 581-95.
[http://dx.doi.org/10.1016/j.molonc.2014.01.005] [PMID: 24484937]
[133]
Alegría-Torres JA, Baccarelli A, Bollati V. Epigenetics and lifestyle. Epigenomics 2011; 3(3): 267-77.
[http://dx.doi.org/10.2217/epi.11.22] [PMID: 22122337]
[134]
Banerjee S, Kaye S. PARP inhibitors in BRCA gene-mutated ovarian cancer and beyond. Curr Oncol Rep 2011; 13(6): 442-9.
[http://dx.doi.org/10.1007/s11912-011-0193-9] [PMID: 21913063]
[135]
Geraets L, Moonen HJJ, Brauers K, Wouters EFM, Bast A, Hageman GJ. Dietary flavones and flavonoles are inhibitors of poly(ADP-ribose)polymerase-1 in pulmonary epithelial cells. J Nutr 2007; 137(10): 2190-5.
[http://dx.doi.org/10.1093/jn/137.10.2190] [PMID: 17884996]
[136]
Chen KC, Sun MF, Chen CYC. in silico investigation of potential PARP-1 inhibitors from traditional chinese medicine. Evidence-based Complement Altern Med 2014.
[137]
Said RS, El-Demerdash E, Nada AS, Kamal MM. Resveratrol inhibits inflammatory signaling implicated in ionizing radiation-induced premature ovarian failure through antagonistic crosstalk between silencing information regulator 1 (SIRT1) and poly(ADP-ribose) polymerase 1 (PARP-1). Biochem Pharmacol 2016; 103: 140-50.
[http://dx.doi.org/10.1016/j.bcp.2016.01.019] [PMID: 26827941]
[138]
Kotsopoulos J, Narod SA. Brief Report: Towards a dietary prevention of hereditary breast cancer. Cancer Causes Control 2005; 16(2): 125-38.
[http://dx.doi.org/10.1007/s10552-004-2593-8] [PMID: 15868454]
[139]
Scalbert A, Andres-Lacueva C, Arita M, et al. Databases on food phytochemicals and their health-promoting effects. J Agric Food Chem 2011; 59(9): 4331-48.
[http://dx.doi.org/10.1021/jf200591d] [PMID: 21438636]
[140]
Kowalska E, Narod SA, Huzarski T, et al. Increased rates of chromosome breakage in BRCA1 carriers are normalized by oral selenium supplementation. Cancer Epidemiol Biomarkers Prev 2005; 14(5): 1302-6.
[http://dx.doi.org/10.1158/1055-9965.EPI-03-0448] [PMID: 15894690]
[141]
Eliassen AH, Liao X, Rosner B, Tamimi RM, Tworoger SS, Hankinson SE. Plasma carotenoids and risk of breast cancer over 20 y of follow-up. Am J Clin Nutr 2015; 101(6): 1197-205.
[http://dx.doi.org/10.3945/ajcn.114.105080] [PMID: 25877493]
[142]
Pool-Zobel B, Bub A, Müller H, Wollowski I, Rechkemmer G. Consumption of vegetables reduces genetic damage in humans: first results of a human intervention trial with carotenoid-rich foods. Carcinogenesis 1997; 18(9): 1847-50.
[http://dx.doi.org/10.1093/carcin/18.9.1847] [PMID: 9328185]
[143]
Nakachi K, Suemasu K, Suga K, Takeo T, Imai K, Higashi Y. Influence of drinking green tea on breast cancer malignancy among Japanese patients. Jpn J Cancer Res 1998; 89(3): 254.
[http://dx.doi.org/10.1111/j.1349-7006.1998.tb00556.x]
[144]
Inoue M, Tajima K, Mizutani M, et al. Regular consumption of green tea and the risk of breast cancer recurrence: follow-up study from the Hospital-based Epidemiologic Research Program at Aichi Cancer Center (HERPACC), Japan. Cancer Lett 2001; 167(2): 175-82.
[http://dx.doi.org/10.1016/S0304-3835(01)00486-4] [PMID: 11369139]
[145]
Wu AH, Yu MC, Tseng CC, Hankin J, Pike MC. Green tea and risk of breast cancer in Asian Americans. Int J Cancer 2003; 106(4): 574-9.
[http://dx.doi.org/10.1002/ijc.11259] [PMID: 12845655]
[146]
Bradlow HL, Telang NT, Sepkovic DW, Osborne MP. Phytochemicals as modulators of cancer risk. Adv Exp Med Biol 1999; 472: 207-21.
[http://dx.doi.org/10.1007/978-1-4757-3230-6_18] [PMID: 10736628]
[147]
Chen P, Li C, Li X, Li J, Chu R, Wang H. Higher dietary folate intake reduces the breast cancer risk: a systematic review and meta-analysis. Br J Cancer 2014; 110(9): 2327-38.
[http://dx.doi.org/10.1038/bjc.2014.155]
[148]
Pan SY, Zhou J, Gibbons L, Morrison H, Wen SW. Antioxidants and breast cancer risk- a population-based case-control study in Canada. BMC Cancer 2011; 11: 372.
[149]
Selenium supplementation reduced oxidative DNA damage in adnexectomized BRCA1 mutations carriers. Cancer Epidemiol Biomarkers Prev 2009; 18(11): 2923-8.
[150]
Seo YR, Kelley MR, Smith ML. Selenomethionine regulation of p53 by a ref1-dependent redox mechanism. Proc Natl Acad Sci USA 2002; 99(22): 14548-53.
[http://dx.doi.org/10.1073/pnas.212319799] [PMID: 12357032]
[151]
Rao AV, Agarwal S. Bioavailability and in vivo antioxidant properties of lycopene from tomato products and their possible role in the prevention of cancer. Nutr Cancer 1998; 31(3): 199-203.
[http://dx.doi.org/10.1080/01635589809514703] [PMID: 9795972]
[152]
Sharoni Y, Girón E, Rise M, Levy J. Effects of lycopene-enriched tomato oleoresin on 7,12-dimethyl-benz[a]anthracene-induced rat mammary tumors. Cancer Detect Prev 1997; 21(2): 118-23.
[PMID: 9101071]
[153]
Nagasawa H, Mitamura T, Sakamoto S, Yamamoto K. Effects of lycopene on spontaneous mammary tumour development in SHN virgin mice. Anticancer Res 1995; 15(4): 1173-8.
[PMID: 7653996]
[154]
Levy J, Bosin E, Feldman B, et al. Lycopene is a more potent inhibitor of human cancer cell proliferation than either α-carotene or β-carotene. Nutr Cancer 1995; 24(3): 257-66.
[http://dx.doi.org/10.1080/01635589509514415] [PMID: 8610045]
[155]
Zhang X, Spiegelman D, Baglietto L, et al. Carotenoid intakes and risk of breast cancer defined by estrogen receptor and progesterone receptor status: A pooled analysis of 18 prospective cohort studies. Am J Clin Nutr 2012; 95(3): 713.
[http://dx.doi.org/10.4016/39352.01]
[156]
Fan S, Meng Q, Auborn K, Carter T, Rosen EM. BRCA1 and BRCA2 as molecular targets for phytochemicals indole-3-carbinol and genistein in breast and prostate cancer cells. Br J Cancer 2006; 94(407)
[http://dx.doi.org/10.1038/sj.bjc.6602935]
[157]
Dietary intake and breast cancer among carriers and noncarriers of BRCA mutations in the Korean Hereditary Breast Cancer Study. Am J Clin Nutr 2013; 98(6): 1493-501.
[158]
Lewis SJ, Harbord RM, Harris R, Smith GD. Meta-analyses of observational and genetic association studies of folate intakes or levels and breast cancer risk. J Natl Cancer Inst 2006; 98(22): 1607-22.
[http://dx.doi.org/10.1093/jnci/djj440] [PMID: 17105984]
[159]
Larsson SC, Giovannucci E, Wolk A. Folate and risk of breast cancer: a meta-analysis. J Natl Cancer Inst 2007; 99(1): 64-76.
[http://dx.doi.org/10.1093/jnci/djk006] [PMID: 17202114]
[160]
Kotsopoulos J, Kim YI, Narod SA. Folate and breast cancer: what about high-risk women? Cancer Causes Control 2012; 23(9): 1405-20.
[http://dx.doi.org/10.1007/s10552-012-0022-y] [PMID: 22767328]
[161]
Kim YI. Folate and colorectal cancer: An evidence-based critical review. Mol Nutr Food Res 2007; 51(3): 267-92.
[http://dx.doi.org/10.1002/mnfr.200600191] [PMID: 17295418]
[162]
Kim SJ, Zuchniak A, Sohn KJ, et al. Plasma folate, vitamin B-6, and vitamin B-12 and breast cancer risk in BRCA1- and BRCA2-mutation carriers: a prospective study. Am J Clin Nutr 2016; 104(3): 671-7.
[http://dx.doi.org/10.3945/ajcn.116.133470] [PMID: 27465373]
[163]
Riboli E. The European prospective investigation into cancer and nutrition: perspectives for cancer prevention. Nestle Nutr Workshop Ser Clin Perform Programme 2000; 4

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