Generic placeholder image

Current Cancer Therapy Reviews

Editor-in-Chief

ISSN (Print): 1573-3947
ISSN (Online): 1875-6301

Review Article

Role of Phytochemicals in the Treatment of Breast Cancer: Natural Swords Battling Cancer Cells

Author(s): Rajni Sawanny, Sheersha Pramanik* and Unnati Agarwal

Volume 17, Issue 3, 2021

Published on: 06 January, 2021

Page: [179 - 196] Pages: 18

DOI: 10.2174/1573394716666210106123255

Price: $65

Abstract

Breast cancer is the most common type of malignancy among ladies (around 30% of newly diagnosed patients every year). To date, various modern treatment modalities for breast cancer, such as radiotherapy, surgical method, hormonal therapy, and chemotherapeutic drug utilisation, are available. However, adverse drug reactions, therapeutic resistance, metastasis, or cancer reoccurrence chances remain the primary causes of mortality for breast cancer patients. To overcome all the potential drawbacks, we need to investigate novel techniques and strategies that are not considered previously to treat breast cancer effectively with safety and efficacy. For centuries, we utilise phytochemicals to treat various diseases because of their safety, low-cost, and least or no side effects. Recently, naturally produced phytochemicals gain immense attention as potential breast cancer therapeutics because of their ideal characteristics; for instance, they operate via modulating molecular pathways associated with cancer growth and progression. The primary mechanism involves inhibition of cell proliferation, angiogenesis, migration, invasion, increasing anti-oxidant status, initiation of the arrest of the cell cycle, and apoptosis. Remedial viability gets effectively enhanced when phytochemicals work as adjuvants with chemotherapeutic drugs. This comprehensive review revolves around the latest chemopreventive, chemotherapeutic, and chemoprotective treatments with their molecular mechanisms to treat breast cancer by utilising phytochemicals such as vinca alkaloids, resveratrol, curcumin, paclitaxel, silibinin, quercetin, genistein, and epigallocatechin gallate. The authors wish to extend the field of phytochemical study for its scientific validity and its druggability.

Keywords: Breast cancer, phytochemicals, anti-cancer agents, mechanism, natural compounds, herbs, cancer therapy.

Graphical Abstract
[1]
Statistics of Breast Cancer In India Available from: https://cytecare.com/blog/statistics-of-breast-cancer/
[2]
U.S. Breast Cancer Statistics Available from: https://www.breastcancer.org/symptoms/understand_bc/statistics
[3]
DeSantis CE, Ma J, Gaudet MM, et al. Breast cancer statistics, 2019. CA Cancer J Clin 2019; 69(6): 438-51.
[http://dx.doi.org/10.3322/caac.21583] [PMID: 31577379]
[4]
Sandhu R, Parker JS, Jones WD, Livasy CA, Coleman WB. Microarray-Based Gene Expression Profiling for Molecular Classification of Breast Cancer and Identification of New Targets for Therapy. Lab Med 2010; 41(6): 364-72.
[http://dx.doi.org/10.1309/LMLIK0VIE3CJK0WD]
[5]
Cazzaniga M, Bonanni B. Breast cancer chemoprevention: old and new approaches. J Biomed Biotechnol 2012; 2012985620
[http://dx.doi.org/10.1155/2012/985620] [PMID: 22851887]
[6]
Cardoso F, Kyriakides S, Ohno S, et al. ESMO Guidelines Committee.. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2019; 30(8): 1194-220.
[http://dx.doi.org/10.1093/annonc/mdz173] [PMID: 31161190]
[7]
Mavrogiannis AV, Kokkinopoulou I, Kontos CK, Sideris DC. Effect of Vinca Alkaloids on the Expression Levels of microRNAs Targeting Apoptosis-related Genes in Breast Cancer Cell Lines. Curr Pharm Biotechnol 2018; 19(13): 1076-86.
[http://dx.doi.org/10.2174/1389201019666181112103204] [PMID: 30417784]
[8]
Zhao YN, Cao YN, Sun J, et al. Anti-breast cancer activity of resveratrol encapsulated in liposomes. J Mater Chem B Mater Biol Med 2020; 8(1): 27-37.
[http://dx.doi.org/10.1039/C9TB02051A] [PMID: 31746932]
[9]
Mittal L, Aryal UK, Camarillo IG, Raman V, Sundararajan R. Effective electrochemotherapy with curcumin in MDA-MB-231-human, triple negative breast cancer cells: A global proteomics study. Bioelectrochemistry 2020; 131107350
[http://dx.doi.org/10.1016/j.bioelechem.2019.107350] [PMID: 31518962]
[10]
Baek JS, Cho CW. A multifunctional lipid nanoparticle for co-delivery of paclitaxel and curcumin for targeted delivery and enhanced cytotoxicity in multidrug resistant breast cancer cells. Oncotarget 2017; 8(18): 30369-82.
[http://dx.doi.org/10.18632/oncotarget.16153] [PMID: 28423731]
[11]
Lamartiniere CA. Protection against breast cancer with genistein: a component of soy. Am J Clin Nutr 2000; 71(6)(Suppl.): 1705S-7S.
[http://dx.doi.org/10.1093/ajcn/71.6.1705S] [PMID: 10837323]
[12]
Si L, Fu J, Liu W, et al. Silibinin inhibits migration and invasion of breast cancer MDA-MB-231 cells through induction of mitochondrial fusion. Mol Cell Biochem 2020; 463(1-2): 189-201.
[http://dx.doi.org/10.1007/s11010-019-03640-6] [PMID: 31612353]
[13]
Karthick V, Panda S, Kumar VG, et al. Quercetin loaded PLGA microspheres induce apoptosis in breast cancer cells. Appl Surf Sci 2019; 487: 211-7.
[http://dx.doi.org/10.1016/j.apsusc.2019.05.047]
[14]
Sheng J, Shi W, Guo H, et al. The Inhibitory Effect of (-)-Epigallocatechin-3-Gallate on Breast Cancer Progression via Reducing SCUBE2 Methylation and DNMT Activity. Molecules 2019; 24(16): 2899.
[http://dx.doi.org/10.3390/molecules24162899] [PMID: 31404982]
[15]
Rajendran P, Ho E, Williams DE, Dashwood RH. Dietary phytochemicals, HDAC inhibition, and DNA damage/repair defects in cancer cells. Clin Epigenetics 2011; 3(1): 4.
[http://dx.doi.org/10.1186/1868-7083-3-4] [PMID: 22247744]
[16]
Bellik Y, Boukraâ L, Alzahrani HA, et al. Molecular mechanism underlying anti-inflammatory and anti-Allergic activities of phytochemicals: An update Molecules 2012; 18(1): 322-53.
[http://dx.doi.org/10.3390/molecules18010322] [PMID: 23271469]
[17]
Tobin G, Kalupahana R, Kulk M. Plant Based Natural Products and Breast Cancer: Considering Multi-Faceted Disease Aspects, Past Successes, and Promising Future InterventionsUsing Old Solutions to New Problems - Natural Drug Discovery in the 21st Century. InTech. USA 2013.
[http://dx.doi.org/10.5772/55794]
[18]
Delmas D, Limagne E, Ghiringhelli F, Aires V. Immune Th17 lymphocytes play a critical role in the multiple beneficial properties of resveratrol. Food Chem Toxicol 2020; 137111091
[http://dx.doi.org/10.1016/j.fct.2019.111091] [PMID: 31883989]
[19]
Fabian C J, Khan SA, Garber JE, et al. Randomized Phase IIB Trial of the Lignan Secoisolariciresinol Diglucoside in Pre-menopausal Women at Increased Risk for Development of Breast Cancer Cancer Prev Res (Phila) 2020; 13(7): 623-34.
[http://dx.doi.org/10.1158/1940-6207.CAPR-20-0050] [PMID: 32312713]
[20]
Saghatelyan T, Tananyan A, Janoyan N, et al. Efficacy and safety of curcumin in combination with paclitaxel in patients with advanced, metastatic breast cancer: A comparative, randomized, double-blind, placebo-controlled clinical trial. Phytomedicine 2020; 70153218
[http://dx.doi.org/10.1016/j.phymed.2020.153218] [PMID: 32335356]
[21]
Tang C, Gong L, Lvzi Xu , Qiu K, Zhang Z, Wan L. Echinacoside inhibits breast cancer cells by suppressing the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 526(1): 170-5.
[http://dx.doi.org/10.1016/j.bbrc.2020.03.050] [PMID: 32201078]
[22]
Desai G, Schelske-Santos M, Nazario CM, et al. Onion and Garlic Intake and Breast Cancer, a Case-Control Study in Puerto Rico. Nutr Cancer 2020; 72(5): 791-800.
[http://dx.doi.org/10.1080/01635581.2019.1651349] [PMID: 31402709]
[23]
Dzimitrowicz A, Bielawska-Pohl A, Pohl P, et al. Application of Oil-in-Water Nanoemulsion Carrying Size-Defined Gold Nanoparticles Synthesised by Non-thermal Plasma for the Human Breast Cancer Cell Lines Migration and Apoptosis. Plasma Chem Plasma Process 2020; 40(4): 1037-62.
[http://dx.doi.org/10.1007/s11090-020-10070-6]
[24]
Potter JD. Cancer prevention: epidemiology and experiment. Cancer Lett 1997; 114(1-2): 7-9.
[http://dx.doi.org/10.1016/S0304-3835(97)04615-6] [PMID: 9103244]
[25]
Hashemi SA, Karami M, Bathaie SZ. Saffron carotenoids change the superoxide dismutase activity in breast cancer: in vitro, in vivo and in silico studies. Int J Biol Macromol 2020; 158: 845-53.
[http://dx.doi.org/10.1016/j.ijbiomac.2020.04.063] [PMID: 32360463]
[26]
Steed KL, Jordan HR, Tollefsbol TO. SAHA and EGCG Promote Apoptosis in Triple-negative Breast Cancer Cells, Possibly Through the Modulation of cIAP2. Anticancer Res 2020; 40(1): 9-26.
[http://dx.doi.org/10.21873/anticanres.13922] [PMID: 31892549]
[27]
England LJ, Levine RJ, Qian C, et al. Glucose tolerance and risk of gestational diabetes mellitus in nulliparous women who smoke during pregnancy. Am J Epidemiol 2004; 160(12): 1205-13.
[http://dx.doi.org/10.1093/aje/kwh340] [PMID: 15583373]
[28]
Rockwell S, Liu Y, Higgins SA. Alteration of the effects of cancer therapy agents on breast cancer cells by the herbal medicine black cohosh. Breast Cancer Res Treat 2005; 90(3): 233-9.
[http://dx.doi.org/10.1007/s10549-004-4260-x] [PMID: 15830136]
[29]
Gutierrez-Pajares JL, Ben Hassen C, Oger C, Galano JM, Durand T, Frank PG. Oxidised products of alpha-linolenic acid negatively regulate cellular survival and motility of breast cancer cells. Biomolecules 2019; 10(1): 50.
[http://dx.doi.org/10.3390/biom10010050] [PMID: 31905626]
[30]
Qin B, Xu B, Ji N, et al. Intake of vitamin D and calcium, sun exposure, and risk of breast cancer subtypes among black women. Am J Clin Nutr 2020; 111(2): 396-405.
[http://dx.doi.org/10.1093/ajcn/nqz302] [PMID: 31826233]
[31]
Jiang X, Liu Y, Zhang G, et al. Aloe-Emodin Induces Breast Tumor Cell Apoptosis through Upregulation of miR-15a/miR-16-1 That Suppresses BCL2. Evid Based Complement Alternat Med 2020; 20205108298
[http://dx.doi.org/10.1155/2020/5108298] [PMID: 32190086]
[32]
Choi WY, Kim G-Y, Lee WH, Choi YH. Sanguinarine, a benzophenanthridine alkaloid, induces apoptosis in MDA-MB-231 human breast carcinoma cells through a reactive oxygen species-mediated mitochondrial pathway. Chemotherapy 2008; 54(4): 279-87.
[http://dx.doi.org/10.1159/000149719] [PMID: 18667818]
[33]
Li Y, Bhuiyan M, Alhasan S, Senderowicz AM, Sarkar FH. Induction of apoptosis and inhibition of c-erbB-2 in breast cancer cells by flavopiridol. Clin Cancer Res 2000; 6(1): 223-9.
[PMID: 10656453]
[34]
Gu H, Rao S, Zhao J, et al. Gambogic acid reduced bcl-2 expression via p53 in human breast MCF-7 cancer cells. J Cancer Res Clin Oncol 2009; 135(12): 1777-82.
[http://dx.doi.org/10.1007/s00432-009-0624-2] [PMID: 19582475]
[35]
Jiang J, Grieb B, Thyagarajan A, Sliva D. Ganoderic acids suppress growth and invasive behavior of breast cancer cells by modulating AP-1 and NF-kappaB signaling. Int J Mol Med 2008; 21(5): 577-84.
[http://dx.doi.org/10.3892/ijmm.21.5.577] [PMID: 18425349]
[36]
Campbell CT, Prince M, Landry GM, Kha V, Kleiner HE. Pro-apoptotic effects of 1′-acetoxychavicol acetate in human breast carcinoma cells. Toxicol Lett 2007; 173(3): 151-60.
[http://dx.doi.org/10.1016/j.toxlet.2007.07.008] [PMID: 17766064]
[37]
Ahmad A, Banerjee S, Wang Z, Kong D, Sarkar FH. Plumbagin-induced apoptosis of human breast cancer cells is mediated by inactivation of NF-kappaB and Bcl-2. J Cell Biochem 2008; 105(6): 1461-71.
[http://dx.doi.org/10.1002/jcb.21966] [PMID: 18980240]
[38]
Murillo G, Peng X, Torres KEO, Mehta RG. Deguelin inhibits growth of breast cancer cells by modulating the expression of key members of the Wnt signaling pathway. Cancer Prev Res (Phila) 2009; 2(11): 942-50.
[http://dx.doi.org/10.1158/1940-6207.CAPR-08-0232] [PMID: 19861542]
[39]
Lee SO, Jeong YJ, Im HG, Kim CH, Chang YC, Lee IS. Silibinin suppresses PMA-induced MMP-9 expression by blocking the AP-1 activation via MAPK signaling pathways in MCF-7 human breast carcinoma cells. Biochem Biophys Res Commun 2007; 354(1): 165-71.
[http://dx.doi.org/10.1016/j.bbrc.2006.12.181] [PMID: 17214970]
[40]
Wishart D S, Feunang YD, Guo AC, et al. DrugBank 5.0: A major update to the DrugBank database for 2018 Nucleic Acids Res 2018; 46(D1): D1074-82.
[http://dx.doi.org/10.1093/nar/gkx1037] [PMID: 29126136]
[41]
Schmid P, Rugo HS, Adams S, et al. IMpassion130 Investigators. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2020; 21(1): 44-59.
[http://dx.doi.org/10.1016/S1470-2045(19)30689-8] [PMID: 31786121]
[42]
Prieto-Vila M, Shimomura I, Kogure A, et al. Quercetin inhibits Lef1 and resensitizes docetaxel-resistant breast cancer cells. Molecules 2020; 25(11): 2576. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7321307/
[http://dx.doi.org/10.3390/molecules25112576] [PMID: 32492961]
[43]
Keshandehghan A, Nikkhah S, Tahermansouri H, Heidari-Keshel S, Gardaneh M. Co-Treatment with Sulforaphane and Nano-Metformin Molecules Accelerates Apoptosis in HER2+ Breast Cancer Cells by Inhibiting Key Molecules. Nutr Cancer 2020; 72(5): 835-48.
[http://dx.doi.org/10.1080/01635581.2019.1655073] [PMID: 31474154]
[44]
Rahman MA, Bulbul MRH, Kabir Y. Plant-based products in cancer prevention and treatment. Elsevier Inc. 2020.
[http://dx.doi.org/10.1016/B978-0-12-816151-7.00013-2]
[45]
Ohishi Y, Mitsuda A, Ejima K, et al. Breast implant-associated anaplastic large-cell lymphoma: first case detected in a Japanese breast cancer patient. Breast Cancer 2020; 27(3): 499-504.
[http://dx.doi.org/10.1007/s12282-020-01064-5] [PMID: 32095988]
[46]
Welt A, Bogner S, Arendt M, et al. Improved survival in metastatic breast cancer: results from a 20-year study involving 1033 women treated at a single comprehensive cancer center. J Cancer Res Clin Oncol 2020; 146(6): 1559-66.
[http://dx.doi.org/10.1007/s00432-020-03184-z] [PMID: 32189107]
[47]
Vernieri C, Prisciandaro M, Nichetti F, et al. Oral capecitabine-vinorelbine is associated with longer overall survival when compared to single-agent capecitabine in patients with hormone receptor-positive advanced breast cancer. Cancers (Basel) 2020; 12(3): 617.
[http://dx.doi.org/10.3390/cancers12030617] [PMID: 32155941]
[48]
Chen L, Guo X, Hu Y, Li L, Liang G, Zhang G. Epigallocatechin-3-gallate sensitises multidrug-resistant oral carcinoma xenografts to vincristine sulfate. FEBS Open Bio 2020; 10(7): 1403-13. Available from: https://pubmed.ncbi.nlm.nih.gov-/32475087/
[http://dx.doi.org/10.1002/2211-5463.12905] [PMID: 32475087]
[49]
Rashid A, Duan X, Gao F, Yang M, Yen A. Roscovitine enhances All-trans retinoic acid (ATRA)-induced leukemia cell differentiation: Novel effects on signaling molecules for a putative Cdk2 inhibitor. Cell Signal 2020; 71109555
[http://dx.doi.org/10.1016/j.cellsig.2020.109555] [PMID: 32032659]
[50]
Dong C, Liu J, Chen SX, et al. Highly robust model of transcription regulator activity predicts breast cancer overall survival BMC Med Genomics 2020; 13(Suppl 5): 49.
[http://dx.doi.org/10.1186/s12920-020-0688-z] [PMID: 32241272]
[51]
Esnaashari SS, Muhammadnejad S, Amanpour S, Amani A. A Combinational Approach Towards Treatment of Breast Cancer: an Analysis of Noscapine-Loaded Polymeric Nanoparticles and Doxorubicin. AAPS PharmSciTech 2020; 21(5): 166.
[http://dx.doi.org/10.1208/s12249-020-01710-3] [PMID: 32504144]
[52]
Jordan MA. Mechanism of action of antitumor drugs that interact with microtubules and tubulin. Curr Med Chem Anticancer Agents 2002; 2(1): 1-17.
[http://dx.doi.org/10.2174/1568011023354290] [PMID: 12678749]
[53]
Bhalla KN. Microtubule-targeted anticancer agents and apoptosis. Oncogene 2003; 22(56): 9075-86.
[http://dx.doi.org/10.1038/sj.onc.1207233] [PMID: 14663486]
[54]
Jordan A, Hadfield JA, Lawrence NJ, McGown AT. Tubulin as a target for anticancer drugs: agents which interact with the mitotic spindle. Med Res Rev 1998; 18(4): 259-96.
[http://dx.doi.org/10.1002/(SICI)1098-1128(199807)18:4<259::AID-MED3>3.0.CO;2-U] [PMID: 9664292]
[55]
Lee C-T, Huang Y-W, Yang C-H, Huang K-S. Drug delivery systems and combination therapy by using vinca alkaloids. Curr Top Med Chem 2015; 15(15): 1491-500.
[http://dx.doi.org/10.2174/1568026615666150414120547] [PMID: 25877096]
[56]
Wang RC, Chen X, Parissenti AM, et al. Sensitivity of docetaxel-resistant MCF-7 breast cancer cells to microtubule-destabilizing agents including vinca alkaloids and colchicine-site binding agents. PLoS One 2017; 12(8)e0182400
[http://dx.doi.org/10.1371/journal.pone.0182400] [PMID: 28787019]
[57]
Yang L, Xu X, Zheng J. Microtubule-associated protein 2 knockdown sensitizes glioma cells to vincristine treatment. Neuroreport 2020; 31(3): 197-204.
[http://dx.doi.org/10.1097/WNR.0000000000001378] [PMID: 31834144]
[58]
Esmaeili-Mahani S, Samandari-Bahraseman MR, Yaghoobi MM. In-Vitro Anti-Proliferative and Pro-Apoptotic Properties of Sutureja Khuzestanica on Human Breast Cancer Cell Line (MCF-7) and Its Synergic Effects with Anticancer Drug Vincristine. Iran J Pharm Res IJPR 2018; 17(1): 343-52. Available from: http://www.ncbi.nlm.nih.gov/pubmed/29755565
[59]
Setthawongsin C, Teewasutrakul P, Tangkawattana S, Techangamsuwan S, Rungsipipat A. Conventional-Vincristine Sulfate vs. Modified Protocol of Vincristine Sulfate and L-Asparaginase in Canine Transmissible Venereal Tumor. Front Vet Sci 2019; 6: 300.
[http://dx.doi.org/10.3389/fvets.2019.00300] [PMID: 31620453]
[60]
Cheuk IWY, Chen J, Siu JMT, et al. Abstract P6-06-16: Resveratrol enhanced chemosensitivity by reversing macrophage polarisation in breast cancerCancer Research. 2020; 80: p. (4 suppl)P6-06-16.
[http://dx.doi.org/10.1158/1538-7445.SABCS19-P6-06-16]
[61]
Gordon KF. Cell Proliferation and viability Inhibition by Resveratrol on Breast Cell Proliferation and viability Inhibition by Resveratrol on Breast Cancer Cell Lines Cancer Cell Lines Available from: https://egrove.olemiss.edu/hon_thesis
[62]
Ferraz da Costa DC, Campos NPC, Santos RA, et al. Resveratrol prevents p53 aggregation in vitro and in breast cancer cells. Oncotarget 2018; 9(49): 29112-22.
[http://dx.doi.org/10.18632/oncotarget.25631] [PMID: 30018739]
[63]
Alobaedi OH, Talib WH, Basheti IA. Antitumor effect of thymoquinone combined with resveratrol on mice transplanted with breast cancer. Asian Pac J Trop Med 2017; 10(4): 400-8.
[http://dx.doi.org/10.1016/j.apjtm.2017.03.026] [PMID: 28552110]
[64]
Wang W, Zhang L, Chen T, et al. Anticancer Effects of Resveratrol-Loaded Solid Lipid Nanoparticles on Human Breast Cancer Cells. Molecules 2017; 22(11): 1814.
[http://dx.doi.org/10.3390/molecules22111814] [PMID: 29068422]
[65]
Venkatadri R, Muni T, Iyer AKV, Yakisich JS, Azad N. Role of apoptosis-related miRNAs in resveratrol-induced breast cancer cell death. Cell Death Dis 2016; 7(2)e2104
[http://dx.doi.org/10.1038/cddis.2016.6] [PMID: 26890143]
[66]
Lv ZD, Liu XP, Zhao WJ, et al. Curcumin induces apoptosis in breast cancer cells and inhibits tumor growth in vitro and in vivo. Int J Clin Exp Pathol 2014; 7(6): 2818-24.
[PMID: 25031701]
[67]
Esfandiarpour-Boroujeni S, Bagheri-Khoulenjani S, Mirzadeh H, Amanpour S. Fabrication and study of curcumin loaded nanoparticles based on folate-chitosan for breast cancer therapy application. Carbohydr Polym 2017; 168: 14-21.
[http://dx.doi.org/10.1016/j.carbpol.2017.03.031] [PMID: 28457434]
[68]
Minafra L, Porcino N, Bravatà V, et al. Radiosensitizing effect of curcumin-loaded lipid nanoparticles in breast cancer cells. Sci Rep 2019; 9(1): 11134.
[http://dx.doi.org/10.1038/s41598-019-47553-2] [PMID: 31366901]
[69]
Kundur S, Prayag A, Selvakumar P, et al. Synergistic anticancer action of quercetin and curcumin against triple-negative breast cancer cell lines. J Cell Physiol 2019; 234(7): 11103-18.
[http://dx.doi.org/10.1002/jcp.27761] [PMID: 30478904]
[70]
Zhang Y, Wang Y, Xue J. Paclitaxel inhibits breast cancer metastasis via suppression of Aurora kinase-mediated cofilin-1 activity. Exp Ther Med 2018; 15(2): 1269-76.
[http://dx.doi.org/10.3892/etm.2017.5588] [PMID: 29434713]
[71]
Guo S. Ultra-thermostable RNA nanoparticles for solubilising and high-yield loading of paclitaxel for breast cancer therapy. Nat Commun 2020; 11(1): 1-11.
[http://dx.doi.org/10.1038/s41467-020-14780-5] [PMID: 31911652]
[72]
Fraguas-Sánchez AI, Fernández-Carballido A, Simancas-Herbada R, Martin-Sabroso C, Torres-Suárez AI. CBD loaded microparticles as a potential formulation to improve paclitaxel and doxorubicin-based chemotherapy in breast cancer. Int J Pharm 2020; 574118916
[http://dx.doi.org/10.1016/j.ijpharm.2019.118916] [PMID: 31811927]
[73]
Chowdhury P, Nagesh PKB, Hatami E, et al. Tannic acid-inspired paclitaxel nanoparticles for enhanced anticancer effects in breast cancer cells. J Colloid Interface Sci 2019; 535: 133-48.
[http://dx.doi.org/10.1016/j.jcis.2018.09.072] [PMID: 30292104]
[74]
Kasiri N, Rahmati M, Ahmadi L, Eskandari N, Motedayyen H. Therapeutic potential of quercetin on human breast cancer in different dimensions Inflammopharmacology 2020; 28(1): 39-62.
[http://dx.doi.org/10.1007/s10787-019-00660-y] [PMID: 31754939]
[75]
Mansourizadeh F, Alberti D, Bitonto V, et al. Efficient synergistic combination effect of Quercetin with Curcumin on breast cancer cell apoptosis through their loading into Apo ferritin cavity. Colloids Surf B Biointerfaces 2020; 191: 110982.
[http://dx.doi.org/10.1016/j.colsurfb.2020.110982] [PMID: 32220813]
[76]
Prieto-Vila M, Shimomura I, Kogure A, et al. Quercetin Inhibits Lef1 and Resensitizes Docetaxel-Resistant Breast Cancer Cells. Molecules 2020; 25(11): 2576.
[http://dx.doi.org/10.3390/molecules25112576] [PMID: 32492961]
[77]
Sadhukhan P, Kundu M, Chatterjee S, et al. Targeted delivery of quercetin via pH-responsive zinc oxide nanoparticles for breast cancer therapy. Mater Sci Eng C 2019; 100: 129-40.
[http://dx.doi.org/10.1016/j.msec.2019.02.096] [PMID: 30948047]
[78]
de Oliveira Pedro R, Hoffmann S, Pereira S, Goycoolea FM, Schmitt CC, Neumann MG. Self-assembled amphiphilic chitosan nanoparticles for quercetin delivery to breast cancer cells. Eur J Pharm Biopharm 2018; 131: 203-10.
[http://dx.doi.org/10.1016/j.ejpb.2018.08.009] [PMID: 30145220]
[79]
Pedro R de O, Pereira S, Goycoolea FM, Schmitt CC, Neumann MG. Self-aggregated nanoparticles of N -dodecyl, N ′-glycidyl(chitosan) as pH-responsive drug delivery systems for quercetin. J Appl Polym Sci 2018; 135(2): 45678.
[http://dx.doi.org/10.1002/app.45678]
[80]
Xu P, Yan F, Zhao Y, et al. Green Tea Polyphenol EGCG Attenuates MDSCs-mediated Immunosuppression through Canonical and Non-Canonical Pathways in a 4T1 Murine Breast Cancer Model. Nutrients 2020; 12(4): 1042.
[http://dx.doi.org/10.3390/nu12041042] [PMID: 32290071]
[81]
Radhakrishnan R, Pooja D, Kulhari H, et al. Bombesin conjugated solid lipid nanoparticles for improved delivery of epigallocatechin gallate for breast cancer treatment. Chem Phys Lipids 2019; 224: 104770.
[http://dx.doi.org/10.1016/j.chemphyslip.2019.04.005] [PMID: 30965023]
[82]
Zan L, Chen Q, Zhang L, Li X. Epigallocatechin gallate (EGCG) suppresses growth and tumorigenicity in breast cancer cells by downregulation of miR-25. Bioengineered 2019; 10(1): 374-82.
[http://dx.doi.org/10.1080/21655979.2019.1657327] [PMID: 31431131]
[83]
Pirouzpanah MB, Sabzichi M, Pirouzpanah S, Chavoshi H, Samadi N. Silibilin-induces apoptosis in breast cancer cells by modulating p53, p21, Bak and Bcl-XL pathways. Asian Pac J Cancer Prev 2015; 16(5): 2087-92.
[http://dx.doi.org/10.7314/APJCP.2015.16.5.2087] [PMID: 25773855]
[84]
Si L, Liu W, Hayashi T, et al. Silibinin-induced apoptosis of breast cancer cells involves mitochondrial impairment. Arch Biochem Biophys 2019; 671: 42-51.
[http://dx.doi.org/10.1016/j.abb.2019.05.009] [PMID: 31085166]
[85]
Jiang M, He K, Qiu T, et al. Tumor-targeted delivery of silibinin and IPI-549 synergistically inhibit breast cancer by remodeling the microenvironment. Int J Pharm 2020; 581: 119239.
[http://dx.doi.org/10.1016/j.ijpharm.2020.119239] [PMID: 32194211]
[86]
Mukund V. Genistein: Its Role in Breast Cancer Growth and Metastasis. Curr Drug Metab 2020; 21(1): 6-10.
[http://dx.doi.org/10.2174/1389200221666200120121919] [PMID: 31987018]
[87]
Mukund V, Saddala MS, Farran B, Mannavarapu M, Alam A, Nagaraju GP. Molecular docking studies of angiogenesis target protein HIF-1α and genistein in breast cancer. Gene 2019; 701: 169-72.
[http://dx.doi.org/10.1016/j.gene.2019.03.062] [PMID: 30930227]
[88]
Paul B, Li Y, Tollefsbol TO. The Effects of Combinatorial Genistein and Sulforaphane in Breast Tumor Inhibition: Role in Epigenetic Regulation. Int J Mol Sci 2018; 19(6): 1754.
[http://dx.doi.org/10.3390/ijms19061754] [PMID: 29899271]
[89]
Pons DG, Vilanova-Llompart J, Gaya-Bover A, et al. The phytoestrogen genistein affects inflammatory-related genes expression depending on the ERα/ERβ ratio in breast cancer cells. Int J Food Sci Nutr 2019; 70(8): 941-9.
[http://dx.doi.org/10.1080/09637486.2019.1597025] [PMID: 30945577]
[90]
Saluzzo J, Hallman KM, Aleck K, et al. The regulation of tumor suppressor protein, p53, and estrogen receptor (ERα) by resveratrol in breast cancer cells. Genes Cancer 2016; 7(11-12): 414-25.
[http://dx.doi.org/10.18632/genesandcancer.125] [PMID: 28191286]
[91]
Kim YN, Choe SR, Cho KH, et al. Resveratrol suppresses breast cancer cell invasion by inactivating a RhoA/YAP signaling axis. Exp Mol Med 2017; 49(2)e296
[http://dx.doi.org/10.1038/emm.2016.151] [PMID: 28232662]
[92]
García-Quiroz J, García-Becerra R, Santos-Cuevas C, et al. Synergistic Antitumorigenic Activity of Calcitriol with Curcumin or Resveratrol is Mediated by Angiogenesis Inhibition in Triple Negative Breast Cancer Xenografts. Cancers (Basel) 2019; 11(11): 1739.
[http://dx.doi.org/10.3390/cancers11111739] [PMID: 31698751]
[93]
Giménez-Bastida JA, Ávila-Gálvez MÁ, Espín JC, González-Sarrías A. Conjugated Physiological Resveratrol Metabolites Induce Senescence in Breast Cancer Cells: Role of p53/p21 and p16/Rb Pathways, and ABC Transporters. Mol Nutr Food Res 2019; 63(22)e1900629
[http://dx.doi.org/10.1002/mnfr.201900629] [PMID: 31441212]
[94]
Javan Maasomi Z, Pilehvar Soltanahmadi Y, Dadashpour M, Alipour Sh, Abolhasani S, Zarghami N. Synergistic anti-cancer effects of silibinin and chrysin in T47D breast cancer cells. Asian Pac J Cancer Prev 2017; 18(5): 1283-7.
[http://dx.doi.org/10.22034/APJCP.2017.18.5.1283] [PMID: 28610415]
[95]
Chavoshi H, Vahedian V, Saghaei S, Pirouzpanah MB, Raeisi M, Samadi N. Adjuvant therapy with silibinin improves the efficacy of paclitaxel and cisplatin in MCF-7 breast cancer cells. Asian Pac J Cancer Prev 2017; 18(8): 2243-7.
[http://dx.doi.org/10.22034/APJCP.2017.18.8.2243] [PMID: 28843263]
[96]
Zadeh MM, Motamed N, Ranji N, Majidi M, Falahi F. Silibinin-induced apoptosis and downregulation of microRNA-21 and MicroRNA-155 in MCF-7 human breast cancer cells. J Breast Cancer 2016; 19(1): 45-52.
[http://dx.doi.org/10.4048/jbc.2016.19.1.45] [PMID: 27066095]
[97]
Jahanafrooz Z, Motameh N, Bakhshandeh B. Comparative evaluation of silibinin effects on cell cycling and apoptosis in human breast cancer MCF-7 and T47D cell lines. Asian Pac J Cancer Prev 2016; 17(5): 2661-5.
[PMID: 27268647]
[98]
Jahanafrooz Z, Motamed N, Bakhshandeh B. Effects of miR-21 downregulation and silibinin treatment in breast cancer cell lines. Cytotechnology 2017; 69(4): 667-80.
[http://dx.doi.org/10.1007/s10616-017-0076-5] [PMID: 28321778]
[99]
Sultan AS, Khalil MI, Sami BM, Alkhuriji AF, Sadek O. Quercetin induces apoptosis in triple-negative breast cancer cells via inhibiting fatty acid synthase and beta-catenin 2017. Available from: www.ijcep.com/
[100]
Ranganathan S, Halagowder D, Sivasithambaram ND. Quercetin suppresses twist to induce apoptosis in MCF-7 breast cancer cells. PLoS One 2015; 10(10)e0141370
[http://dx.doi.org/10.1371/journal.pone.0141370] [PMID: 26491966]
[101]
Zhao X, Wang Q, Yang S, et al. Quercetin inhibits angiogenesis by targeting calcineurin in the xenograft model of human breast cancer. Eur J Pharmacol 2016; 781: 60-8.
[http://dx.doi.org/10.1016/j.ejphar.2016.03.063] [PMID: 27041643]
[102]
Khorsandi L, Orazizadeh M, Niazvand F, Abbaspour MR, Mansouri E, Khodadadi A. Quercetin induces apoptosis and necroptosis in MCF-7 breast cancer cells. Bratisl Lek Listy 2017; 118(2): 123-8.
[http://dx.doi.org/10.4149/BLL_2017_025] [PMID: 28814095]
[103]
Ko YM, Wu TY, Wu YC, Chang FR, Guh JY, Chuang LY. Annonacin induces cell cycle-dependent growth arrest and apoptosis in estrogen receptor-α-related pathways in MCF-7 cells. J Ethnopharmacol 2011; 137(3): 1283-90.
[http://dx.doi.org/10.1016/j.jep.2011.07.056] [PMID: 21840388]
[104]
Pradhan D, Pradhan RK, Tripathy G, Pradhan S. Inhibition of proteasome activity by the dietary flavonoid Quercetin associated with growth inhibition in cultured breast cancer cells and xenografts. J Young Pharm 2015; 7(3): 225-33.
[http://dx.doi.org/10.5530/jyp.2015.3.13]
[105]
Kumar U, Sharma U, Rathi G. Reversal of hypermethylation and reactivation of glutathione S-transferase pi 1 gene by curcumin in breast cancer cell line. Tumour Biol 2017; 39(2)1010428317692258
[http://dx.doi.org/10.1177/1010428317692258] [PMID: 28222671]
[106]
Bimonte S, Barbieri A, Palma G, et al. Dissecting the role of curcumin in tumour growth and angiogenesis in mouse model of human breast cancer. BioMed Res Int 2015; 2015878134
[http://dx.doi.org/10.1155/2015/878134] [PMID: 25879038]
[107]
Colacino JA, McDermott SP, Sartor MA, Wicha MS, Rozek LS. Transcriptomic profiling of curcumin-treated human breast stem cells identifies a role for stearoyl-coa desaturase in breast cancer prevention. Breast Cancer Res Treat 2016; 158(1): 29-41.
[http://dx.doi.org/10.1007/s10549-016-3854-4] [PMID: 27306423]
[108]
Liu Y, Zhou J, Hu Y, Wang J, Yuan C. Curcumin inhibits growth of human breast cancer cells through demethylation of DLC1 promoter. Mol Cell Biochem 2017; 425(1-2): 47-58.
[http://dx.doi.org/10.1007/s11010-016-2861-4] [PMID: 27830358]
[109]
Gallardo M, Calaf GM. Curcumin inhibits invasive capabilities through epithelial mesenchymal transition in breast cancer cell lines. Int J Oncol 2016; 49(3): 1019-27.
[http://dx.doi.org/10.3892/ijo.2016.3598] [PMID: 27573203]
[110]
Geng C, Li J, Ding F, et al. Curcumin suppresses 4-hydroxytamoxifen resistance in breast cancer cells by targeting SLUG/Hexokinase 2 pathway. Biochem Biophys Res Commun 2016; 473(1): 147-53.
[http://dx.doi.org/10.1016/j.bbrc.2016.03.067] [PMID: 27012210]
[111]
Avci CB, Susluer SY, Caglar HO, et al. Genistein-induced mir-23b expression inhibits the growth of breast cancer cells. Contemp Oncol (Pozn) 2015; 19(1): 32-5.
[http://dx.doi.org/10.5114/wo.2014.44121] [PMID: 26199568]
[112]
Donovan MG, Selmin Ol, Doetschman TC, and Romagnolo DF, “Epigenetic Activation of BRCA1 by Genistein in vivo and Triple Negative Breast Cancer Cells Linked to Antagonism toward Aryl Hydrocarbon Receptor. Nutrients 2019; 11(11): 2559.
[http://dx.doi.org/10.3390/nu11112559] [PMID: 31652854]
[113]
Kim GY, Suh J, Jang JH, et al. Genistein Inhibits Proliferation of BRCA1 Mutated Breast Cancer Cells: The GPR30-Akt Axis as a Potential Target. J Cancer Prev 2019; 24(4): 197-207.
[http://dx.doi.org/10.15430/JCP.2019.24.4.197] [PMID: 31950019]
[114]
Zhao Q, Zhao M, Parris AB, Xing Y, Yang X. Genistein targets the cancerous inhibitor of PP2A to induce growth inhibition and apoptosis in breast cancer cells. Int J Oncol 2016; 49(3): 1203-10.
[http://dx.doi.org/10.3892/ijo.2016.3588] [PMID: 27574003]
[115]
Lazzeroni M, Guerrieri-Gonzaga A, Gandini S, et al. A presurgical study of lecithin formulation of green tea extract in women with early breast cancer. Cancer Prev Res (Phila) 2017; 10(6): 363-70.
[http://dx.doi.org/10.1158/1940-6207.CAPR-16-0298] [PMID: 28400479]
[116]
Hong OY, Noh EM, Jang HY, et al. Epigallocatechin gallate inhibits the growth of MDA-MB-231 breast cancer cells via inactivation of the β-catenin signaling pathway. Oncol Lett 2017; 14(1): 441-6.
[http://dx.doi.org/10.3892/ol.2017.6108] [PMID: 28693189]
[117]
Esmaeili MA. Combination of siRNA-directed gene silencing with epigallocatechin-3-gallate (EGCG) reverses drug resistance in human breast cancer cells. J Chem Biol 2015; 9(1): 41-52.
[http://dx.doi.org/10.1007/s12154-015-0144-2] [PMID: 26855680]
[118]
Moradzadeh M, Hosseini A, Erfanian S, Rezaei H. Epigallocatechin-3-gallate promotes apoptosis in human breast cancer T47D cells through down-regulation of PI3K/AKT and Telomerase. Pharmacol Rep 2017; 69(5): 924-8.
[http://dx.doi.org/10.1016/j.pharep.2017.04.008] [PMID: 28646740]
[119]
Nowakowska A, Tarasiuk J. Comparative effects of selected plant polyphenols, gallic acid and epigallocatechin gallate, on matrix metalloproteinases activity in multidrug resistant MCF7/DOX breast cancer cells. Acta Biochim Pol 2016; 63(3): 571-5.
[http://dx.doi.org/10.18388/abp.2016_1256] [PMID: 27231728]
[120]
Hallman K, Aleck K, Quigley M, et al. The regulation of steroid receptors by epigallocatechin-3-gallate in breast cancer cells. Breast Cancer (Dove Med Press) 2017; 9: 365-73.
[http://dx.doi.org/10.2147/BCTT.S131334] [PMID: 28579831]
[121]
Huang CY, Han Z, Li X, Xie HH, Zhu SS. Mechanism of EGCG promoting apoptosis of MCF-7 cell line in human breast cancer. Oncol Lett 2017; 14(3): 3623-7.
[http://dx.doi.org/10.3892/ol.2017.6641] [PMID: 28927122]
[122]
Luchsinger C, Aguilar M, Burgos PV, Ehrenfeld P, Mardones GA. Functional disruption of the Golgi apparatus protein ARF1 sensitizes MDA-MB-231 breast cancer cells to the antitumor drugs Actinomycin D and Vinblastine through ERK and AKT signaling. PLoS One 2018; 13(4)e0195401
[http://dx.doi.org/10.1371/journal.pone.0195401] [PMID: 29614107]
[123]
Rattanaburee T, Tipmanee V, Tedasen A, Thongpanchang T, Graidist P. Inhibition of CSF1R and AKT by (±)-kusunokinin hinders breast cancer cell proliferation. Biomed Pharmacother 2020; 129110361
[http://dx.doi.org/10.1016/j.biopha.2020.110361] [PMID: 32535390]
[124]
Heudel P, Delaloge S, Parent D, et al. Real-world Evaluation of Oral Vinorelbine in the Treatment of Metastatic Breast Cancer: An ESME-MBC Study. Anticancer Res 2020; 40(7): 3905-13.
[http://dx.doi.org/10.21873/anticanres.14381] [PMID: 32620631]
[125]
Reinhorn DJ. Abstract P1-18-33: Taxane versus vinorelbine in combination with pertuzumab and trastuzumab for first-line treatment of patients with metastatic HER2-positive breast cancer - A retrospective two-center studyCancer Research 2020; 8(4): 19-P1-18-33.
[http://dx.doi.org/10.1158/1538-7445.SABCS19-P1-18-33]
[126]
Kamble SS, Gacche RN. Evaluation of anti-breast cancer, anti-angiogenic and anti-oxidant properties of selected medicinal plants. Eur J Integr Med 2019; 25: 13-9.
[http://dx.doi.org/10.1016/j.eujim.2018.11.006]
[127]
Liao WS, Ho Y, Lin YW, et al. Targeting EGFR of triple-negative breast cancer enhances the therapeutic efficacy of paclitaxel- and cetuximab-conjugated nanodiamond nanocomposite. Acta Biomater 2019; 86: 395-405.
[http://dx.doi.org/10.1016/j.actbio.2019.01.025] [PMID: 30660004]
[128]
Zajdel A, Wilczok A, Jelonek K, et al. Cytotoxic Effect of Paclitaxel and Lapatinib Co-Delivered in Polylactide-co-Poly(ethylene glycol) Micelles on HER-2-Negative Breast Cancer Cells. Pharmaceutics 2019; 11(4): 169.
[http://dx.doi.org/10.3390/pharmaceutics11040169] [PMID: 30959904]
[129]
Rodrigues-Ferreira S, Nehlig A, Kacem M, Nahmias C. ATIP3 deficiency facilitates intracellular accumulation of paclitaxel to reduce cancer cell migration and lymph node metastasis in breast cancer patients. Sci Rep 2020; 10(1): 13217.
[http://dx.doi.org/10.1038/s41598-020-70142-7] [PMID: 32764625]
[130]
Kreger BT, Johansen ER, Cerione RA, Antonyak MA. The Enrichment of Survivin in Exosomes from Breast Cancer Cells Treated with Paclitaxel Promotes Cell Survival and Chemoresistance. Cancers (Basel) 2016; 8(12): 111.
[http://dx.doi.org/10.3390/cancers8120111] [PMID: 27941677]
[131]
Wang R, Yang M, Li G, et al. Paclitaxel-betulinic acid hybrid nanosuspensions for enhanced anti-breast cancer activity. Colloids Surf B Biointerfaces 2019; 174: 270-9.
[http://dx.doi.org/10.1016/j.colsurfb.2018.11.029] [PMID: 30469048]
[132]
Shakeri A, Sahebkar A. Opinion Paper: Nanotechnology: A Successful Approach to Improve Oral Bioavailability of Phytochemicals. Recent Pat Drug Deliv Formul 2016; 10(1): 4-6.
[http://dx.doi.org/10.2174/1872211309666150611120724] [PMID: 26063398]
[133]
Cheng Z, Al Zaki A, Hui JZ, Muzykantov VR, Tsourkas A. Multifunctional nanoparticles: cost versus benefit of adding targeting and imaging capabilities Science 2012; 338(6109): 903-10.
[http://dx.doi.org/10.1126/science.1226338] [PMID: 23161990]
[134]
Kumari A, Yadav SK, Pakade YB, Singh B, Yadav SC. Development of biodegradable nanoparticles for delivery of quercetin. Colloids Surf B Biointerfaces 2010; 80(2): 184-92.
[http://dx.doi.org/10.1016/j.colsurfb.2010.06.002] [PMID: 20598513]
[135]
Yu Y-B, Miyashiro H, Nakamura N, Hattori M, Park JC. Effects of triterpenoids and flavonoids isolated from Alnus firma on HIV-1 viral enzymes. Arch Pharm Res 2007; 30(7): 820-6.
[http://dx.doi.org/10.1007/BF02978831] [PMID: 17703732]
[136]
Stewart LK, Soileau JL, Ribnicky D, et al. Quercetin transiently increases energy expenditure but persistently decreases circulating markers of inflammation in C57BL/6J mice fed a high-fat diet. Metabolism 2008; 57(7)(Suppl. 1): S39-46.
[http://dx.doi.org/10.1016/j.metabol.2008.03.003] [PMID: 18555853]
[137]
Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov 2006; 5(6): 493-506.
[http://dx.doi.org/10.1038/nrd2060] [PMID: 16732220]
[138]
Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006; 444(7117): 337-42.
[http://dx.doi.org/10.1038/nature05354] [PMID: 17086191]
[139]
Jang M. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes Science (80- ) 1997; 275(5297): 218-20.
[http://dx.doi.org/10.1126/science.275.5297.218]
[140]
Kumari A, Kumar V, Yadav SK. Plant extract synthesized PLA nanoparticles for controlled and sustained release of quercetin: a green approach. PLoS One 2012; 7(7)e41230
[http://dx.doi.org/10.1371/journal.pone.0041230] [PMID: 22844443]
[141]
Srinivas K, King JW, Howard LR, Monrad JK. Solubility and solution thermodynamic properties of quercetin and quercetin dihydrate in subcritical water. J Food Eng 2010; 100(2): 208-18.
[http://dx.doi.org/10.1016/j.jfoodeng.2010.04.001]
[142]
Detoni CB, Souto GD, da Silva ALM, Pohlmann AR, Guterres SS. Photostability and skin penetration of different E-resveratrol-loaded supramolecular structures. Photochem Photobiol 2012; 88(4): 913-21.
[http://dx.doi.org/10.1111/j.1751-1097.2012.01147.x] [PMID: 22443373]
[143]
Nair KL, Thulasidasan AKT, Deepa G, Anto RJ, Kumar GSV. Purely aqueous PLGA nanoparticulate formulations of curcumin exhibit enhanced anticancer activity with dependence on the combination of the carrier. Int J Pharm 2012; 425(1-2): 44-52.
[http://dx.doi.org/10.1016/j.ijpharm.2012.01.003] [PMID: 22266528]
[144]
Xie X, Tao Q, Zou Y, et al. PLGA nanoparticles improve the oral bioavailability of curcumin in rats: characterizations and mechanisms. J Agric Food Chem 2011; 59(17): 9280-9.
[http://dx.doi.org/10.1021/jf202135j] [PMID: 21797282]
[145]
Teskač K, Kristl J. The evidence for solid lipid nanoparticles mediated cell uptake of resveratrol. Int J Pharm 2010; 390(1): 61-9.
[http://dx.doi.org/10.1016/j.ijpharm.2009.10.011] [PMID: 19833178]
[146]
Wang S, Su R, Nie S, et al. Application of nanotechnology in improving bioavailability and bioactivity of diet-derived phytochemicals. J Nutr Biochem 2014; 25(4): 363-76.
[http://dx.doi.org/10.1016/j.jnutbio.2013.10.002] [PMID: 24406273]
[147]
Mohanty C, Sahoo SK. The in vitro stability and in vivo pharmacokinetics of curcumin prepared as an aqueous nanoparticulate formulation. Biomaterials 2010; 31(25): 6597-611.
[http://dx.doi.org/10.1016/j.biomaterials.2010.04.062] [PMID: 20553984]
[148]
Tsai Y-M, Chien C-F, Lin L-C, Tsai T-H. Curcumin and its nano-formulation: the kinetics of tissue distribution and blood-brain barrier penetration. Int J Pharm 2011; 416(1): 331-8.
[http://dx.doi.org/10.1016/j.ijpharm.2011.06.030] [PMID: 21729743]
[149]
Sanna V, Roggio AM, Siliani S, et al. Development of novel cationic chitosan-and anionic alginate-coated poly(D,L-lactide-co-glycolide) nanoparticles for controlled release and light protection of resveratrol. Int J Nanomedicine 2012; 7: 5501-16.
[PMID: 23093904]
[150]
Sahibzada MUK, Sadiq A, Khan S, et al. Fabrication, characterization and in vitro evaluation of silibinin nanoparticles: an attempt to enhance its oral bioavailability. Drug Des Devel Ther 2017; 11: 1453-64.
[http://dx.doi.org/10.2147/DDDT.S133806] [PMID: 28553075]
[151]
Rowinsky EK, Donehower RC, Jones RJ, Tucker RW. Microtubule changes and cytotoxicity in leukemic cell lines treated with taxol. Cancer Res 1988; 48(14): 4093-100.
[PMID: 2898289]
[152]
Schiff PB, Fant J, Horwitz SB. Promotion of microtubule assembly in vitro by taxol. Nature 1979; 277(5698): 665-7.
[http://dx.doi.org/10.1038/277665a0] [PMID: 423966]
[153]
Smith QR. Brain perfusion systems for studies of drug uptake and metabolism in the central nervous system.Models for assessing drug absorption and metabolism. Springer 1996; pp. 285-307.
[http://dx.doi.org/10.1007/978-1-4899-1863-5_15]
[154]
Liu D, Pearlman E, Diaconu E, et al. Expression of hyaluronidase by tumor cells induces angiogenesis in vivo. Proc Natl Acad Sci USA 1996; 93(15): 7832-7.
[http://dx.doi.org/10.1073/pnas.93.15.7832] [PMID: 8755562]
[155]
Mittapalli RK, Liu X, Adkins CE, et al. Paclitaxel-hyaluronic nanoconjugates prolong overall survival in a preclinical brain metastases of breast cancer model. Mol Cancer Ther 2013; 12(11): 2389-99.
[http://dx.doi.org/10.1158/1535-7163.MCT-13-0132] [PMID: 24002934]

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