[1]
Senapati, S.; Mahanta, A.K.; Kumar, S.; Maiti, P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct. Target. Ther., 2018, 3, 7.
[2]
Maurya, A.K.; Vinayak, M. Breast cancer stem cell mediated aberrant signaling and epithelial-mesenchymal transition targets: Hope for breast cancer therapy. Int. J. Cancer Oncol., 2016, 3(3), 1-7.
[3]
Maurya, A.K.; Vinayak, M. PI-103 and Quercetin attenuate PI3K-AKT signaling pathway in T- cell lymphoma exposed to hydrogen Peroxide. PLoS One, 2016, 11(8)e0160686
[4]
Das, L.; Vinayak, M. Curcumin modulates glycolytic metabolism and inflammatory cytokines via Nrf 2 in dalton’s lymphoma ascites cells in vivo. Anticancer. Ag Med. Chem., 2018, 18(12), 1779-1791.
[5]
Vinayak, M. Molecular action of herbal antioxidants in regulation of cancer growth: Scope for novel anticancer drugs. Nutr. Cancer, 2018, 70(8), 1199-1209.
[6]
Neuhouser, M.L. Dietary flavonoids and cancer risk: Evidence from human population studies. Nutr. Cancer, 2004, 50(1), 1-7.
[7]
Tang, N.P.; Zhou, B.; Wang, B.; Yu, R.B.; Ma, J. Flavonoids intake and risk of lung cancer: A meta-analysis. Jpn. J. Clin. Oncol., 2009, 39(6), 352-359.
[8]
Cui, Y.; Morgenstern, H.; Greenland, S.; Tashkin, D.P.; Mao, J.T.; Cai, L.; Cozen, W.; Mack, T.M.; Lu, Q.Y.; Zhang, Z.F. Dietary flavonoid intake and lung cancer--a population-based case-control study. Cancer, 2008, 112(10), 2241-2248.
[9]
Forcados, G.E.; James, D.B.; Sallau, A.B.; Muhammad, A.; Mabeta, P. Oxidative stress and carcinogenesis: Potential of phytochemicals in breast cancer therapy. Nutr. Cancer, 2017, 69(3), 365-374.
[10]
Abraham, A.N.; Sharma, T.K.; Bansal, V.; Shukla, R. Phytochemicals as dynamic surface ligands to control nanoparticle-protein interactions. ACS Omega, 2018, 3(2), 2220-2229.
[11]
Maurya, A.K.; Vinayak, M. Modulation of PKC signaling and induction of apoptosis through suppression of reactive oxygen species and tumor necrosis factor receptor 1 (TNFR1): Key role of quercetin in cancer prevention. Tumour Biol., 2015, 36(11), 8913-8924.
[12]
D’Andrea, G. Quercetin: A flavonol with multifaceted therapeutic applications? Fitoterapia, 2015, 106, 256-271.
[13]
Amanzadeh, E.; Esmaeili, A.; Rahgozar, S.; Nourbakhshnia, M. Application of quercetin in neurological disorders: From nutrition to nanomedicine. Rev. Neurosci., 2019, 30(5), 555-572.
[14]
Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.; Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A comprehensive review. Phytother. Res., 2018, 32(11), 2109-2130.
[15]
Maurya, A.K.; Vinayak, M. Anticarcinogenic action of quercetin by downregulation of phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC) via induction of p53 in hepatocellular carcinoma (HepG2) cell line. Mol. Biol. Rep., 2015, 42(9), 1419-1429.
[16]
Kumari, A.; Yadav, S.K.; Pakade, Y.B.; Kumar, V.; Singh, B.; Chaudhary, A.; Yadav, S.C. Nanoencapsulation and characterization of Albizia chinensis isolated antioxidant quercitrin on PLA nanoparticles. Colloids Surf. B Biointerfaces, 2011, 82(1), 224-232.
[17]
Maurya, A.K.; Vinayak, M. Improved synergistic anticancer efficacy of quercetin in combination with PI-103, rottlerin, and G0 6983 against MCF-7 and RAW 264.7 cells. In Vitro Cell. Dev. Biol. Anim., 2019, 55(1), 36-44.
[18]
Kleemann, R.; Verschuren, L.; Morrison, M.; Zadelaar, S.; van Erk, M.J.; Wielinga, P.Y.; Kooistra, T. Anti-inflammatory, anti-proliferative and anti-atherosclerotic effects of quercetin in human in vitro and in vivo models. Atherosclerosis, 2011, 218(1), 44-52.
[19]
Nabavi, S.F.; Russo, G.L.; Daglia, M.; Nabavi, S.M. Role of quercetin as an alternative for obesity treatment: You are what you eat! Food Chem., 2015, 179, 305-310.
[20]
Xu, D.; Hu, M-J.; Wang, Y-Q.; Cui, Y-L. Antioxidant activities of quercetin and its complexes for medicinal application. Molecules, 2019, 24(6), 1123.
[21]
Dueñas, M.; González-Manzano, S.; González-Paramás, A.; Santos-Buelga, C. Antioxidant evaluation of O-methylated metabolites of catechin, epicatechin and quercetin. J. Pharm. Biomed. Anal., 2010, 51(2), 443-449.
[22]
Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: From antioxidant to nutraceutical. Eur. J. Pharmacol., 2008, 585(2-3), 325-337.
[23]
Ganesan, S.; Faris, A.N.; Comstock, A.T.; Wang, Q.; Nanua, S.; Hershenson, M.B.; Sajjan, U.S. Quercetin inhibits rhinovirus replication in vitro and in vivo. Antiviral Res., 2012, 94(3), 258-271.
[24]
Sun, D.; Zhang, W.; Li, N.; Zhao, Z.; Mou, Z.; Yang, E.; Wang, W. Silver nanoparticles-quercetin conjugation to siRNA against drug-resistant Bacillus subtilis for effective gene silencing: In vitro and in vivo. Mater. Sci. Eng. C, 2016, 63, 522-534.
[25]
Rai, M.; Yadav, A.; Gade, A. Silver nanoparticles as a new generation of antimicrobials. Biotechnol. Adv., 2009, 27(1), 76-83.
[26]
Han, Q.; Wang, X.; Cai, S.; Liu, X.; Zhang, Y.; Yang, L.; Wang, C.; Yang, R. Quercetin nanoparticles with enhanced bioavailability as multifunctional agents toward amyloid induced neurotoxicity. J. Mater. Chem. B Mater. Biol. Med., 2018, 6(9), 1387-1393.
[27]
George, D.; Maheswari, P.U.; Begum, K.M.M.S. Synergic formulation of onion peel quercetin loaded chitosan-cellulose hydrogel with green zinc oxide nanoparticles towards controlled release, biocompatibility, antimicrobial and anticancer activity. Int. J. Biol. Macromol., 2019, 132, 784-794.
[28]
Alidadi, H.; Khorsandi, L.; Shirani, M. Effects of quercetin on tubular cell apoptosis and kidney damage in rats induced by titanium dioxide nanoparticles. Malays. J. Med. Sci., 2018, 25(2), 72-81.
[29]
Abdelhalim, M.A.K.; Qaid, H.A.; Al-Mohy, Y.; Al-Ayed, M.S. Effects of quercetin and arginine on the nephrotoxicity and lipid peroxidation induced by gold nanoparticles in vivo. Int. J. Nanomed, 2018, 13, 7765-7770.
[30]
Cruz Dos Santos, S.; Osti Silva, N.; Dos Santos Espinelli, J.B.J.; Germani Marinho, M.A.; Vieira Borges, Z.; Bruzamarello Caon Branco, N.; Faita, F.L.; Meira Soares, B.; Horn, A.P.; Parize, A.L.; Rodrigues de Lima, V. Molecular interactions and physico-chemical characterization of quercetin-loaded magnetoliposomes. Chem. Phys. Lipids, 2019, 218, 22-33.
[31]
Wen, P.; Hu, T.G.; Li, L.; Zong, M.H.; Wu, H. A colon-specific delivery system for quercetin with enhanced cancer prevention based on co-axial electrospinning. Food Funct., 2018, 9(11), 5999-6009.
[32]
Ma, J.J.; Yu, Y.G.; Yin, S.W.; Tang, C.H.; Yang, X.Q. Cellular uptake and intracellular antioxidant activity of Zein/Chitosan nanoparticles incorporated with Quercetin. J. Agric. Food Chem., 2018, 66(48), 12783-12793.
[33]
Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.F.; Flamm, G.W.; Williams, G.M.; Lines, T.C. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem. Toxicol., 2007, 45(11), 2179-2205.
[34]
Reinboth, M.; Wolffram, S.; Abraham, G.; Ungemach, F.R.; Cermak, R. Oral bioavailability of quercetin from different quercetin glycosides in dogs. Br. J. Nutr., 2010, 104(2), 198-203.
[35]
Hirpara, K.V.; Aggarwal, P.; Mukherjee, A.J.; Joshi, N.; Burman, A.C. Quercetin and its derivatives: Synthesis, pharmacological uses with special emphasis on anti-tumor properties and prodrug with enhanced bio-availability. Anticancer. Ag Med. Chem., 2009, 9(2), 138-161.
[36]
Mulholland, P.J.; Ferry, D.R.; Anderson, D.; Hussain, S.A.; Young, A.M.; Cook, J.E.; Hodgkin, E.; Seymour, L.W.; Kerr, D.J. Pre-clinical and clinical study of QC12, a water-soluble, pro-drug of quercetin. Ann. Oncol., 2001, 12(2), 245-248.
[37]
Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of biological importance of Quercetin: A bioactive flavonoid. Pharmacogn. Rev., 2016, 10(20), 84-89.
[38]
Ezzati Nazhad Dolatabadi, J.; Mokhtarzadeh, A.; Ghareghoran, S.M.; Dehghan, G. Synthesis, characterization and antioxidant property of Quercetin-Tb(III) complex. Adv. Pharm. Bull., 2014, 4(2), 101-104.
[39]
Gao, X.; Wang, B.; Wei, X.; Men, K.; Zheng, F.; Zhou, Y.; Zheng, Y.; Gou, M.; Huang, M.; Guo, G.; Huang, N.; Qian, Z.; Wei, Y. Anticancer effect and mechanism of polymer micelle-encapsulated quercetin on ovarian cancer. Nanoscale, 2012, 4(22), 7021-7030.
[40]
Nam, J.S.; Sharma, A.R.; Nguyen, L.T.; Chakraborty, C.; Sharma, G.; Lee, S.S. Application of bioactive quercetin in oncotherapy: From nutrition to nanomedicine. Molecules, 2016, 21(1)E108
[41]
Maurya, A.K.; Vinayak, M. Quercetin regresses Dalton’s lymphoma growth via suppression of PI3K/AKT signaling leading to upregulation of p53 and decrease in energy metabolism. Nutr. Cancer, 2015, 67(2), 354-363.
[42]
Zhu, B.; Yu, L.; Yue, Q. Co-delivery of vincristine and quercetin by nanocarriers for lymphoma combination chemotherapy. Biomed. Pharmacother., 2017, 91, 287-294.
[43]
Maurya, A.K.; Vinayak, M. PI-103 attenuates PI3K-AKT signaling and induces apoptosis in murineT-cell lymphoma. Leuk. Lymphoma, 2017, 58(5), 1153-1161.
[44]
Maurya, A.K.; Vinayak, M. Quercetin attenuates cell survival, inflammation, and angiogenesis via modulation of AKT signaling in murine T-cell lymphoma. Nutr. Cancer, 2017, 69(3), 470-480.
[45]
Rivera Rivera, A.; Castillo-Pichardo, L.; Gerena, Y.; Dharmawardhane, S. Anti-breast cancer potential of Quercetin via the Akt/AMPK/mammalian Target of Rapamycin (mTOR) signaling cascade. PLoS One, 2016, 11(6)e0157251
[46]
Seo, H.S.; Ku, J.M.; Choi, H.S.; Choi, Y.K.; Woo, J.K.; Kim, M.; Kim, I.; Na, C.H.; Hur, H.; Jang, B.H.; Shin, Y.C.; Ko, S.G. Quercetin induces caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells. Oncol. Rep., 2016, 36(1), 31-42.
[47]
Subramaniam, D.; Kaushik, G.; Dandawate, P.; Anant, S. Targeting cancer stem cells for chemoprevention of pancreatic cancer. Curr. Med. Chem., 2018, 25(22), 2585-2594.
[48]
Gavrilas, L.I.; Ionescu, C.; Tudoran, O.; Lisencu, C.; Balacescu, O.; Miere, D. The role of bioactive dietary components in modulating miRNA expression in colorectal cancer. Nutrients, 2016, 8(10)E590
[49]
Saleem, T.H.; Attya, A.M.; Ahmed, E.A.; Ragab, S.M.; Ali Abdallah, M.A.; Omar, H.M. Possible protective effects of quercetin and sodium gluconate against colon cancer induction by dimethylhydrazine in mice. Asian Pac. J. Cancer Prev., 2015, 16(14), 5823-5828.
[50]
Engen, A.; Maeda, J.; Wozniak, D.E.; Brents, C.A.; Bell, J.J.; Uesaka, M.; Aizawa, Y.; Kato, T.A. Induction of cytotoxic and genotoxic responses by natural and novel quercetin glycosides. Mutat. Res. Genet. Toxicol. Environ. Mutagen., 2015, 784-785, 15-22.
[51]
Maurya, A.K.; Vinayak, M. Abstract A07: Decline in the growth of murine T-cell lymphoma via modulation of PI3K signaling pathway: Key role of quercetin and PI-103. Mol. Cancer Ther., 2015, 14(Suppl. 7), A07-A07.
[52]
Chen, L.C.; Chen, Y.C.; Su, C.Y.; Hong, C.S.; Ho, H.O.; Sheu, M.T. Development and characterization of self-assembling lecithin-based mixed polymeric micelles containing quercetin in cancer treatment and an in vivo pharmacokinetic study. Int. J. Nanomedicine, 2016, 11, 1557-1566.
[53]
Del Follo-Martinez, A.; Banerjee, N.; Li, X.; Safe, S.; Mertens-Talcott, S. Resveratrol and quercetin in combination have anticancer activity in colon cancer cells and repress oncogenic microRNA-27a. Nutr. Cancer, 2013, 65(3), 494-504.
[54]
Chan, S-T.; Chuang, C-H.; Lin, Y-C.; Liao, J-W.; Lii, C-K.; Yeh, S-L. Quercetin enhances the antitumor effect of trichostatin A and suppresses muscle wasting in tumor-bearing mice. Food Funct., 2018, 9(2), 871-879.
[55]
Serri, C.; Quagliariello, V.; Iaffaioli, R.V.; Fusco, S.; Botti, G.; Mayol, L.; Biondi, M. Combination therapy for the treatment of pancreatic cancer through hyaluronic acid-decorated nanoparticles loaded with quercetin and gemcitabine: A preliminary in vitro study. J. Cell. Physiol., 2019, 234(4), 4959-4969.
[56]
Flusberg, D.A.; Sorger, P.K. Surviving apoptosis: Life-death signaling in single cells. Trends Cell Biol., 2015, 25(8), 446-458.
[57]
Pobezinskaya, Y.L.; Liu, Z. The role of TRADD in death receptor signaling. Cell Cycle, 2012, 11(5), 871-876.
[58]
Russo, M.; Spagnuolo, C.; Bilotto, S.; Tedesco, I.; Maiani, G.; Russo, G.L.; Russo, M.; Spagnuolo, C.; Bilotto, S.; Tedesco, I.; Maiani, G.; Russo, G.L. Inhibition of protein kinase CK2 by quercetin enhances CD95-mediated apoptotis in a human thymus-derived T cell line. Food Res. Int., 2014, 63(B), 244-251.
[59]
Chou, C.C.; Yang, J.S.; Lu, H.F.; Ip, S.W.; Lo, C.; Wu, C.C.; Lin, J.P.; Tang, N.Y.; Chung, J.G.; Chou, M.J.; Teng, Y.H.; Chen, D.R. Quercetin-mediated cell cycle arrest and apoptosis involving activation of a caspase cascade through the mitochondrial pathway in human breast cancer MCF-7 cells. Arch. Pharm. Res., 2010, 33(8), 1181-1191.
[60]
Haghiac, M.; Walle, T. Quercetin induces necrosis and apoptosis in SCC-9 oral cancer cells. Nutr. Cancer, 2005, 53(2), 220-231.
[61]
Mozhgan Farzami Sepehr, S.B.J.B.H. The Cuscuta kotschyana effects on breast cancer cells line MCF7. J. Med. Plants Res., 2011, 5(27), 6344-6351.
[62]
Niu, G.; Yin, S.; Xie, S.; Li, Y.; Nie, D.; Ma, L.; Wang, X.; Wu, Y. Quercetin induces apoptosis by activating caspase-3 and regulating Bcl-2 and cyclooxygenase-2 pathways in human HL-60 cells. Acta Biochim. Biophys. Sin. (Shanghai), 2011, 43(1), 30-37.
[63]
Hu, J.; Wang, J.; Wang, G.; Yao, Z.; Dang, X. Pharmacokinetics and antitumor efficacy of DSPE-PEG2000 polymeric liposomes loaded with quercetin and temozolomide: Analysis of their effectiveness in enhancing the chemosensitization of drug-resistant glioma cells. Int. J. Mol. Med., 2016, 37(3), 690-702.
[64]
Hu, J.; Yu, Q.; Zhao, F.; Ji, J.; Jiang, Z.; Chen, X.; Gao, P.; Ren, Y.; Shao, S.; Zhang, L.; Yan, M. Protection of quercetin against triptolide-induced apoptosis by suppressing oxidative stress in rat Leydig cells. Chem. Biol. Interact., 2015, 240, 38-46.
[65]
Shen, S.C.; Lee, W.R.; Yang, L.Y.; Tsai, H.H.; Yang, L.L.; Chen, Y.C. Quercetin enhancement of arsenic-induced apoptosis via stimulating ROS-dependent p53 protein ubiquitination in human HaCaT keratinocytes. Exp. Dermatol., 2012, 21(5), 370-375.
[66]
Zhang, J.Y.; Yi, T.; Liu, J.; Zhao, Z.Z.; Chen, H.B. Quercetin induces apoptosis via the mitochondrial pathway in KB and KBv200 cells. J. Agric. Food Chem., 2013, 61(9), 2188-2195.
[67]
Zhang, Q.; Zhao, X.H.; Wang, Z.J. Cytotoxicity of flavones and flavonols to a human esophageal squamous cell carcinoma cell line (KYSE-510) by induction of G2/M arrest and apoptosis. Toxicol. In Vitro, 2009, 23(5), 797-807.
[68]
Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo, Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest and apoptosis in human breast cancer cells by quercetin. Int. J. Oncol., 2001, 19(4), 837-844.
[69]
Lee, Y.K.; Hwang, J.T.; Kwon, D.Y.; Surh, Y.J.; Park, O.J. Induction of apoptosis by quercetin is mediated through AMPKalpha1/ASK1/p38 pathway. Cancer Lett., 2010, 292(2), 228-236.
[70]
Kim, M.C.; Lee, H.J.; Lim, B.; Ha, K.T.; Kim, S.Y.; So, I.; Kim, B.J. Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 channels in AGS cells. Int. J. Mol. Med., 2014, 33(6), 1657-1663.
[71]
Lee, K.H.; Yoo, C.G. Simultaneous inactivation of GSK-3β suppresses quercetin-induced apoptosis by inhibiting the JNK pathway. Am. J. Physiol. Lung Cell. Mol. Physiol., 2013, 304(11), L782-L789.
[72]
Vidya Priyadarsini, R.; Senthil Murugan, R.; Maitreyi, S.; Ramalingam, K.; Karunagaran, D.; Nagini, S. The flavonoid quercetin induces cell cycle arrest and mitochondria-mediated apoptosis in human cervical cancer (HeLa) cells through p53 induction and NF-κB inhibition. Eur. J. Pharmacol., 2010, 649(1-3), 84-91.
[73]
Chen, X.; Dong, X.S.; Gao, H.Y.; Jiang, Y.F.; Jin, Y.L.; Chang, Y.Y.; Chen, L.Y.; Wang, J.H. Suppression of HSP27 increases the antitumor effects of quercetin in human leukemia U937 cells. Mol. Med. Rep., 2016, 13(1), 689-696.
[74]
Mukherjee, A.; Khuda-Bukhsh, A.R. Quercetin down-regulates IL-6/STAT-3 signals to induce mitochondrial-mediated apoptosis in a nonsmall- cell lung-cancer cell line, A549. J. Pharmacopuncture, 2015, 18(1), 19-26.
[75]
Ranganathan, S.; Halagowder, D.; Sivasithambaram, N.D. Quercetin suppresses twist to induce apoptosis in MCF-7 breast cancer cells. PLoS One, 2015, 10(10)e0141370
[76]
Ward, A.B.; Mir, H.; Kapur, N.; Gales, D.N.; Carriere, P.P.; Singh, S. Quercetin inhibits prostate cancer by attenuating cell survival and inhibiting anti-apoptotic pathways. World J. Surg. Oncol., 2018, 16(1), 108.
[77]
Granado-Serrano, A.B.; Martín, M.A.; Bravo, L.; Goya, L.; Ramos, S. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J. Nutr., 2006, 136(11), 2715-2721.
[78]
Walker, E.H.; Pacold, M.E.; Perisic, O.; Stephens, L.; Hawkins, P.T.; Wymann, M.P.; Williams, R.L. Structural determinants of phosphoinositide 3-kinase inhibition by wortmannin, LY294002, quercetin, myricetin, and staurosporine. Mol. Cell, 2000, 6(4), 909-919.
[79]
Datta, S.R.; Dudek, H.; Tao, X.; Masters, S.; Fu, H.; Gotoh, Y.; Greenberg, M.E. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell, 1997, 91(2), 231-241.
[80]
Pap, M.; Cooper, G.M. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-Kinase/Akt cell survival pathway. J. Biol. Chem., 1998, 273(32), 19929-19932.
[81]
Dickey, A.; Schleicher, S.; Leahy, K.; Hu, R.; Hallahan, D.; Thotala, D.K. GSK-3β inhibition promotes cell death, apoptosis, and in vivo tumor growth delay in neuroblastoma Neuro-2A cell line. J. Neurooncol., 2011, 104(1), 145-153.
[82]
Chiang, G.G.; Abraham, R.T. Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase. J. Biol. Chem., 2005, 280(27), 25485-25490.
[83]
Hung, C.M.; Garcia-Haro, L.; Sparks, C.A.; Guertin, D.A. mTOR-dependent cell survival mechanisms. Cold Spring Harb. Perspect. Biol., 2012, 4(12)a008771
[84]
Simpson, D.R.; Mell, L.K.; Cohen, E.E. Targeting the PI3K/AKT/mTOR pathway in squamous cell carcinoma of the head and neck. Oral Oncol., 2015, 51(4), 291-298.
[85]
Mabuchi, S.; Kuroda, H.; Takahashi, R.; Sasano, T. The PI3K/AKT/mTOR pathway as a therapeutic target in ovarian cancer. Gynecol. Oncol., 2015, 137(1), 173-179.
[86]
Martelli, A.M.; Evangelisti, C.; Chiarini, F.; Grimaldi, C.; Cappellini, A.; Ognibene, A.; McCubrey, J.A. The emerging role of the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling network in normal myelopoiesis and leukemogenesis. Biochim. Biophys. Acta, 2010, 1803(9), 991-1002.
[87]
Bai, D.; Ueno, L.; Vogt, P.K. Akt-mediated regulation of NFkappaB and the essentialness of NFkappaB for the oncogenicity of PI3K and Akt. Int. J. Cancer, 2009, 125(12), 2863-2870.
[88]
Choi, K.C.; Chung, W.T.; Kwon, J.K.; Yu, J.Y.; Jang, Y.S.; Park, S.M.; Lee, S.Y.; Lee, J.C. Inhibitory effects of quercetin on aflatoxin B1-induced hepatic damage in mice. Food Chem. Toxicol., 2010, 48(10), 2747-2753.
[89]
Ventura, A.; Kirsch, D.G.; McLaughlin, M.E.; Tuveson, D.A.; Grimm, J.; Lintault, L.; Newman, J.; Reczek, E.E.; Weissleder, R.; Jacks, T. Restoration of p53 function leads to tumour regression in vivo. Nature, 2007, 445(7128), 661-665.
[90]
Moll, U.M.; Petrenko, O. The MDM2-p53 interaction. Mol. Cancer Res., 2003, 1(14), 1001-1008.
[91]
Mayo, L.D.; Donner, D.B. The PTEN, Mdm2, p53 tumor suppressor-oncoprotein network. Trends Biochem. Sci., 2002, 27(9), 462-467.
[92]
Zhao, P.; Mao, J.M.; Zhang, S.Y.; Zhou, Z.Q.; Tan, Y.; Zhang, Y. Quercetin induces HepG2 cell apoptosis by inhibiting fatty acid biosynthesis. Oncol. Lett., 2014, 8(2), 765-769.
[93]
Jia, L.; Huang, S.; Yin, X.; Zan, Y.; Guo, Y.; Han, L. Quercetin suppresses the mobility of breast cancer by suppressing glycolysis through Akt-mTOR pathway mediated autophagy induction. Life Sci., 2018, 208, 123-130.
[94]
Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. Cell Biol., 2007, 8(6), 464-478.
[95]
Graupera, M.; Potente, M. Regulation of angiogenesis by PI3K signaling networks. Exp. Cell Res., 2013, 319(9), 1348-1355.
[96]
Ma, F.; Zhang, L.; Westlund, K.N. Reactive oxygen species mediate TNFR1 increase after TRPV1 activation in mouse DRG neurons. Mol. Pain, 2009, 5, 31.
[97]
Fang, J.; Ding, M.; Yang, L.; Liu, L.Z.; Jiang, B.H. PI3K/PTEN/AKT signaling regulates prostate tumor angiogenesis. Cell. Signal., 2007, 19(12), 2487-2497.
[98]
Sobolewski, C.; Cerella, C.; Dicato, M.; Ghibelli, L.; Diederich, M. The role of cyclooxygenase-2 in cell proliferation and cell death in human malignancies. Int. J. Cell Biol., 2010, 2010215158
[99]
Das, L.; Vinayak, M. Long term effect of curcumin in restoration of tumour suppressor p53 and phase-II antioxidant enzymes via activation of Nrf2 signalling and modulation of inflammation in prevention of cancer. PLoS One, 2015, 10(4)e0124000
[100]
Subbaramaiah, K.; Altorki, N.; Chung, W.J.; Mestre, J.R.; Sampat, A.; Dannenberg, A.J. Inhibition of cyclooxygenase-2 gene expression by p53. J. Biol. Chem., 1999, 274(16), 10911-10915.
[101]
Jeon, J-S.; Kwon, S.; Ban, K.; Kwon Hong, Y.; Ahn, C.; Sung, J-S.; Choi, I. Regulation of the intracellular ROS level is critical for the antiproliferative effect of Quercetin in the hepatocellular carcinoma cell line HepG2. Nutr. Cancer, 2019, 71(5), 861-869.
[102]
Jadhav, N.R.; Nadaf, S.J.; Lohar, D.A.; Ghagare, P.S.; Powar, T.A. Phytochemicals formulated as nanoparticles: Inventions, recent patents and future prospects. Recent Pat. Drug Deliv. Formul., 2017, 11(3), 173-186.
[103]
Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: food sources and bioavailability. Am. J. Clin. Nutr., 2004, 79(5), 727-747.
[104]
Weldin, J.; Jack, R.; Dugaw, K.; Kapur, R.P. Quercetin, an over-the-counter supplement, causes neuroblastoma-like elevation of plasma homovanillic acid. Pediatr. Dev. Pathol., 2003, 6(6), 547-551.
[105]
Konishi, Y.; Zhao, Z.; Shimizu, M. Phenolic acids are absorbed from the rat stomach with different absorption rates. J. Agric. Food Chem., 2006, 54(20), 7539-7543.
[106]
Németh, K.; Plumb, G.W.; Berrin, J.G.; Juge, N.; Jacob, R.; Naim, H.Y.; Williamson, G.; Swallow, D.M.; Kroon, P.A. Deglycosylation by small intestinal epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides in humans. Eur. J. Nutr., 2003, 42(1), 29-42.
[107]
Murota, K.; Terao, J. Quercetin appears in the lymph of unanesthetized rats as its phase II metabolites after administered into the stomach. FEBS Lett., 2005, 579(24), 5343-5346.
[108]
Spencer, J.P.; Kuhnle, G.G.; Williams, R.J.; Rice-Evans, C. Intracellular metabolism and bioactivity of quercetin and its in vivo metabolites. Biochem. J., 2003, 372(Pt 1), 173-181.
[109]
de Boer, V.C.; Dihal, A.A.; van der Woude, H.; Arts, I.C.; Wolffram, S.; Alink, G.M.; Rietjens, I.M.; Keijer, J.; Hollman, P.C. Tissue distribution of quercetin in rats and pigs. J. Nutr., 2005, 135(7), 1718-1725.
[110]
Lee, J.; Mitchell, A.E. Pharmacokinetics of quercetin absorption from apples and onions in healthy humans. J. Agric. Food Chem., 2012, 60(15), 3874-3881.
[111]
Kaşıkcı, M.B.; Bağdatlıoğlu, N. Bioavailability of Quercetin. Curr. Res. Nutr. Food Sci., 2016, 4, 146-151.
[112]
Tran, T.H.; Guo, Y.; Song, D.; Bruno, R.S.; Lu, X. Quercetin-containing self-nanoemulsifying drug delivery system for improving oral bioavailability. J. Pharm. Sci., 2014, 103(3), 840-852.
[113]
Dian, L.; Yu, E.; Chen, X.; Wen, X.; Zhang, Z.; Qin, L.; Wang, Q.; Li, G.; Wu, C. Enhancing oral bioavailability of quercetin using novel soluplus polymeric micelles. Nanoscale Res. Lett., 2014, 9(1), 2406.
[114]
Xu, G.; Shi, H.; Ren, L.; Gou, H.; Gong, D.; Gao, X.; Huang, N. Enhancing the anti-colon cancer activity of quercetin by self-assembled micelles. Int. J. Nanomed, 2015, 10, 2051-2063.
[115]
Matsumura, Y.; Kataoka, K. Preclinical and clinical studies of anticancer agent-incorporating polymer micelles. Cancer Sci., 2009, 100(4), 572-579.
[116]
Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of quercetin bioavailability. J. Nutr. Biochem., 2015, 26(3), 201-210.
[117]
Almeida, A.F.; Borge, G.I.A.; Piskula, M.; Tudose, A.; Tudoreanu, L.; Valentová, K.; Williamson, G.; Santos, C.N. Bioavailability of Quercetin in humans with a focus on interindividual variation. Compr. Rev. Food Sci. Food Saf., 2018, 17, 714-731.
[118]
Mu, Y.; Fu, Y.; Li, J.; Yu, X.; Li, Y.; Wang, Y.; Wu, X.; Zhang, K.; Kong, M.; Feng, C.; Chen, X. Multifunctional quercetin conjugated chitosan nano-micelles with P-gp inhibition and permeation enhancement of anticancer drug. Carbohydr. Polym., 2019, 203, 10-18.
[119]
Ahmad, N.; Ahmad, R.; Naqvi, A.A.; Alam, M.A.; Abdur Rub, R.; Ahmad, F.J. Enhancement of Quercetin oral bioavailability by self-nanoemulsifying drug delivery system and their quantification through ultra high performance liquid chromatography and mass spectrometry in cerebral ischemia. Drug Res. (Stuttg.), 2017, 67(10), 564-575.
[120]
Rezaei-Sadabady, R.; Eidi, A.; Zarghami, N.; Barzegar, A. Intracellular ROS protection efficiency and free radical-scavenging activity of quercetin and quercetin-encapsulated liposomes. Artif. Cells Nanomed. Biotechnol., 2016, 44(1), 128-134.
[121]
Lockhart, J.N.; Stevens, D.M.; Beezer, D.B.; Kravitz, A.; Harth, E. Dual drug delivery of tamoxifen and quercetin: Regulated metabolism for anticancer treatment with nanosponges. J. Control Release., 2015, 220(Pt B), 751-757.
[122]
des Rieux, A.; Fievez, V.; Garinot, M.; Schneider, Y.J.; Préat, V. Nanoparticles as potential oral delivery systems of proteins and vaccines: a mechanistic approach. J. Control. Release, 2006, 116(1), 1-27.
[123]
McClements, D.J.; Decker, E.A.; Park, Y.; Weiss, J. Structural design principles for delivery of bioactive components in nutraceuticals and functional foods. Crit. Rev. Food Sci. Nutr., 2009, 49(6), 577-606.
[124]
Rodriguez-Nogales, C.; Noguera, R.; Patrick, C.; Blanco-Prieto, M.J.J. Therapeutic opportunities in neuroblastoma using nanotechnology. J. Pharmacol. Exper. Ther., 2019. jpet.118.255067.
[125]
Caddeo, C.; Díez-Sales, O.; Pons, R.; Carbone, C.; Ennas, G.; Puglisi, G.; Fadda, A.M.; Manconi, M. Cross-linked chitosan/liposome hybrid system for the intestinal delivery of quercetin. J. Colloid Interface Sci., 2016, 461, 69-78.
[126]
Rahimi, S.; Khoee, S.; Ghandi, M. Preparation and characterization of rod-like chitosan-quinoline nanoparticles as pH-responsive nanocarriers for quercetin delivery. Int. J. Biol. Macromol., 2019, 128, 279-289.
[127]
Barbosa, A.I.; Costa Lima, S.A.; Reis, S. Application of pH-responsive fucoidan/chitosan nanoparticles to improve oral quercetin delivery. Molecules, 2019, 24(2), 346.
[128]
Priprem, S.S.; Na, H-K.; Surh, Y-J.; Chulasiri, M. Effect of formulations of nanosized Quercetin liposomes on COX-2 and NF-kB in MCF-10A Cells. Pharm. Nanotechnol., 2013, 1(1), 26-34.
[129]
Priprem, A.; Watanatorn, J.; Sutthiparinyanont, S.; Phachonpai, W.; Muchimapura, S. Anxiety and cognitive effects of quercetin liposomes in rats. Nanomedicine (Lond.), 2008, 4(1), 70-78.
[130]
Ren, J.; Fang, Z.; Jiang, L.; Du, Q. Quercetin-containing self-assemble proliposome preparation and evaluation. J. Liposome Res., 2017, 27(4), 335-342.
[131]
Zheng, N.G.; Mo, S.J.; Li, J.P.; Wu, J.L. Anti-CSC effects in human esophageal squamous cell carcinomas and Eca109/9706 cells induced by nanoliposomal quercetin alone or combined with CD 133 antiserum. Asian Pac. J. Cancer Prev., 2014, 15(20), 8679-8684.
[132]
Martirosyan, A.; Grintzalis, K.; Polet, M.; Laloux, L.; Schneider, Y.J. Tuning the inflammatory response to silver nanoparticles via quercetin in Caco-2 (co-)cultures as model of the human intestinal mucosa. Toxicol. Lett., 2016, 253, 36-45.
[133]
Mittal, A.K.; Kumar, S.; Banerjee, U.C. Quercetin and gallic acid mediated synthesis of bimetallic (silver and selenium) nanoparticles and their antitumor and antimicrobial potential. J. Colloid Interface Sci., 2014, 431, 194-199.
[134]
Pandey, S.K.; Patel, D.K.; Maurya, A.K.; Thakur, R.; Mishra, D.P.; Vinayak, M.; Haldar, C.; Maiti, P. Controlled release of drug and better bioavailability using poly(lactic acid-co-glycolic acid) nanoparticles. Int. J. Biol. Macromol., 2016, 89, 99-110.
[135]
Suksiriworapong, J.; Sripha, K.; Kreuter, J.; Junyaprasert, V.B. Functionalized (poly(ɛ-caprolactone))2-poly(ethylene glycol) nanoparticles with grafting nicotinic acid as drug carriers. Int. J. Pharm., 2012, 423(2), 562-570.
[136]
Suksiriworapong, J.; Phoca, K.; Ngamsom, S.; Sripha, K.; Moongkarndi, P.; Junyaprasert, V.B. Comparison of poly(ε-caprolactone) chain lengths of poly(ε-caprolactone)-co-d-α-tocopheryl-poly(ethylene glycol) 1000 succinate nanoparticles for enhancement of quercetin delivery to SKBR3 breast cancer cells. Eur. J. Pharm. Biopharm., 2016, 101, 15-24.
[137]
Gupta, A.; Kaur, C.D.; Saraf, S.; Saraf, S. Formulation, characterization, and evaluation of ligand-conjugated biodegradable quercetin nanoparticles for active targeting. Artif. Cells Nanomed. Biotechnol., 2016, 44(3), 960-970.
[138]
Lv, L.; Liu, C.; Chen, C.; Yu, X.; Chen, G.; Shi, Y.; Qin, F.; Ou, J.; Qiu, K.; Li, G. Quercetin and doxorubicin co-encapsulated biotin receptor-targeting nanoparticles for minimizing drug resistance in breast cancer. Oncotarget, 2016, 7(22), 32184-32199.
[139]
Guan, X.; Gao, M.; Xu, H.; Zhang, C.; Liu, H.; Lv, L.; Deng, S.; Gao, D.; Tian, Y. Quercetin-loaded poly (lactic-co-glycolic acid)-d-α-tocopheryl polyethylene glycol 1000 succinate nanoparticles for the targeted treatment of liver cancer. Drug Deliv., 2016, 23(9), 3307-3318.
[140]
Wang, C.; Su, L.; Wu, C.; Wu, J.; Zhu, C.; Yuan, G. RGD peptide targeted lipid-coated nanoparticles for combinatorial delivery of sorafenib and quercetin against hepatocellular carcinoma. Drug Dev. Ind. Pharm., 2016, 42(12), 1938-1944.
[141]
Bishayee, K.; Khuda-Bukhsh, A.R.; Huh, S.O. PLGA-loaded gold-nanoparticles precipitated with Quercetin downregulate HDAC-Akt activities controlling proliferation and activate p53-ROS crosstalk to induce apoptosis in hepatocarcinoma cells. Mol. Cells, 2015, 38(6), 518-527.
[142]
Pimple, S.; Manjappa, A.S.; Ukawala, M.; Murthy, R.S. PLGA nanoparticles loaded with etoposide and quercetin dihydrate individually: in vitro cell line study to ensure advantage of combination therapy. Cancer Nanotechnol., 2012, 3(1-6), 25-36.
[143]
Zhao, J.; Liu, J.; Wei, T.; Ma, X.; Cheng, Q.; Huo, S.; Zhang, C.; Zhang, Y.; Duan, X.; Liang, X.J. Quercetin-loaded nanomicelles to circumvent human castration-resistant prostate cancer in vitro and in vivo. Nanoscale, 2016, 8(9), 5126-5138.
[144]
Pang, X.; Lu, Z.; Du, H.; Yang, X.; Zhai, G. Hyaluronic acid-quercetin conjugate micelles: Synthesis, characterization, in vitro and in vivo evaluation. Colloids Surf. B Biointerfaces, 2014, 123, 778-786.
[145]
Tan, B.J.; Liu, Y.; Chang, K.L.; Lim, B.K.; Chiu, G.N. Perorally active nanomicellar formulation of quercetin in the treatment of lung cancer. Int. J. Nanomed, 2012, 7, 651-661.
[146]
Zhao, L.; Shi, Y.; Zou, S.; Sun, M.; Lil, L.; Zhail, G. Formulation and in vitro evaluation of quercetin loaded polymeric micelles composed of pluronic P123 and D-a-tocopheryl polyethylene glycol succinate. J. Biomed. Nanotechnol., 2011, 7(3), 358-365.
[147]
Khonkarn, R.; Mankhetkorn, S.; Hennink, W.E.; Okonogi, S. PEG-OCL micelles for quercetin solubilization and inhibition of cancer cell growth. Eur. J. Pharm. Biopharm., 2011, 79(2), 268-275.
[148]
Sun, C.; Ding, Y.; Zhou, L.; Shi, D.; Sun, L.; Webster, T.J.; Shen, Y. Noninvasive nanoparticle strategies for brain tumor targeting. Nanomedicine, 2017, 13(8), 2605-2621.
[149]
Lainé, A.L.; Clavreul, A.; Rousseau, A.; Tétaud, C.; Vessieres, A.; Garcion, E.; Jaouen, G.; Aubert, L.; Guilbert, M.; Benoit, J.P.; Toillon, R.A.; Passirani, C. Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules. Nanomedicine, 2014, 10(8), 1667-1677.
[150]
Jain, A.K.; Thanki, K.; Jain, S. Novel self-nanoemulsifying formulation of quercetin: Implications of pro-oxidant activity on the anticancer efficacy. Nanomedicine, 2014, 10(5), 959-969.
[151]
Ghosh, S.; Sarkar, S.; Choudhury, S.T.; Ghosh, T.; Das, N. Triphenyl phosphonium coated nano-quercetin for oral delivery: Neuroprotective effects in attenuating age related global moderate cerebral ischemia reperfusion injury in rats. Nanomedicine , 2017, 13(8), 2439-2450.
[152]
Penalva, R.; González-Navarro, C.J.; Gamazo, C.; Esparza, I.; Irache, J.M. Zein nanoparticles for oral delivery of quercetin: Pharmacokinetic studies and preventive anti-inflammatory effects in a mouse model of endotoxemia. Nanomedicine (Lond.), 2017, 13(1), 103-110.
[153]
Sedaghat Doost, A.; Kassozi, V.; Grootaert, C.; Claeys, M.; Dewettinck, K.; Van Camp, J.; Van der Meeren, P. Self-assembly, functionality, and in-vitro properties of quercetin loaded nanoparticles based on shellac-almond gum biological macromolecules. Int. J. Biol. Macromol., 2019, 129, 1024-1033.
[154]
Fang, J.; Nakamura, H.; Maeda, H. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv. Drug Deliv. Rev., 2011, 63(3), 136-151.
[155]
Patra, A.; Satpathy, S.; Shenoy, A.K.; Bush, J.A.; Kazi, M.; Hussain, M.D. Formulation and evaluation of mixed polymeric micelles of quercetin for treatment of breast, ovarian, and multidrug resistant cancers. Int. J. Nanomedicine, 2018, 13, 2869-2881.
[156]
Men, K.; Duan, X.; Wei, X.W.; Gou, M.L.; Huang, M.J.; Chen, L.J.; Qian, Z.Y.; Wei, Y.Q. Nanoparticle-delivered quercetin for cancer therapy. Anticancer. Agents Med. Chem., 2014, 14(6), 826-832.
[157]
Sharma, G.; Park, J.; Sharma, A.R.; Jung, J.S.; Kim, H.; Chakraborty, C.; Song, D.K.; Lee, S.S.; Nam, J.S. Methoxy poly(ethylene glycol)-poly(lactide) nanoparticles encapsulating quercetin act as an effective anticancer agent by inducing apoptosis in breast cancer. Pharm. Res., 2015, 32(2), 723-735.
[158]
Ahmad, N.; Banala, V.T.; Kushwaha, P.; Karvande, A.; Sharma, S.; Tripathi, A.K.; Verma, A.; Trivedi, R.; Mishra, P.R. Quercetin-loaded solid lipid nanoparticles improve osteoprotective activity in an ovariectomized rat model: A preventive strategy for post-menopausal osteoporosis. RSC Advances, 2016, 6(100), 97613-97628.
[159]
Castangia, I.; Nácher, A.; Caddeo, C.; Merino, V.; Díez-Sales, O.; Catalán-Latorre, A.; Fernàndez-Busquets, X.; Fadda, A.M.; Manconi, M. Therapeutic efficacy of quercetin enzyme-responsive nanovesicles for the treatment of experimental colitis in rats. Acta Biomater., 2015, 13, 216-227.
[160]
Singh, S.K.; Singh, S.; Lillard, J.W., Jr; Singh, R. Drug delivery approaches for breast cancer. Int. J. Nanomedicine, 2017, 12, 6205-6218.
[161]
Gulbake, A.; Jain, S.K. Chitosan: A potential polymer for colon-specific drug delivery system. Expert Opin. Drug Deliv., 2012, 9(6), 713-729.
[162]
Han, L.; Zhao, J.; Zhang, X.; Cao, W.; Hu, X.; Zou, G.; Duan, X.; Liang, X.J. Enhanced siRNA delivery and silencing gold-chitosan nanosystem with surface charge-reversal polymer assembly and good biocompatibility. ACS Nano, 2012, 6(8), 7340-7351.
[163]
Yuan, Y.G.; Peng, Q.L.; Gurunathan, S. Silver nanoparticles enhance the apoptotic potential of gemcitabine in human ovarian cancer cells: combination therapy for effective cancer treatment. Int. J. Nanomedicine, 2017, 12, 6487-6502.
[164]
Yoo, G.; Jeong, S.H.; Ryu, W.I.; Lee, H.; Kim, J.H.; Bae, H.C.; Son, S.W. Gene expression analysis reveals a functional role for the Ag-NPs-induced Egr-1 transcriptional factor in human keratinocytes. Mol. Cell. Toxicol., 2014, 10(2), 149-156.
[165]
Lin, J.; Huang, Z.; Wu, H.; Zhou, W.; Jin, P.; Wei, P.; Zhang, Y.; Zheng, F.; Zhang, J.; Xu, J.; Hu, Y.; Wang, Y.; Li, Y.; Gu, N.; Wen, L. Inhibition of autophagy enhances the anticancer activity of silver nanoparticles. Autophagy, 2014, 10(11), 2006-2020.
[166]
Lin, L.; Xu, Y.; Zhang, S.; Ross, I.M.; Ong, A.C.; Allwood, D.A. Fabrication and luminescence of monolayered boron nitride quantum dots. Small, 2014, 10(1), 60-65.
[167]
AshaRani. P.V.; Low Kah Mun, G.; Hande, M.P.; Valiyaveettil, S. Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano, 2009, 3(2), 279-290.
[168]
Sanpui, P.; Chattopadhyay, A.; Ghosh, S.S. Induction of apoptosis in cancer cells at low silver nanoparticle concentrations using chitosan nanocarrier. ACS Appl. Mater. Interfaces, 2011, 3(2), 218-228.
[169]
Gurunathan, S.; Jeong, J.K.; Han, J.W.; Zhang, X.F.; Park, J.H.; Kim, J.H. Multidimensional effects of biologically synthesized silver nanoparticles in Helicobacter pylori, Helicobacter felis, and human lung (L132) and lung carcinoma A549 cells. Nanoscale Res. Lett., 2015, 10, 35.
[170]
Ortega, F.G.; Fernández-Baldo, M.A.; Fernández, J.G.; Serrano, M.J.; Sanz, M.I.; Diaz-Mochón, J.J.; Lorente, J.A.; Raba, J. Study of antitumor activity in breast cell lines using silver nanoparticles produced by yeast. Int. J. Nanomedicine, 2015, 10, 2021-2031.
[171]
Pandey, S.K.; Patel, D.K.; Maurya, A.K.; Thakur, R.; Mishra, D.P.; Vinayak, M.; Haldar, C.; Maiti, P. Corrigendum to Controlled release of drug and better bioavailability using poly(lactic acid-co-glycolic acid) nanoparticles. Int. J. Biol. Macromol., 2016, 89, 99-110. Int. J. Biol. Macromol., 2018, 114, 1361.
[172]
Zhang, H.; Liu, G.; Zeng, X.; Wu, Y.; Yang, C.; Mei, L.; Wang, Z.; Huang, L. Fabrication of genistein-loaded biodegradable TPGS-b-PCL nanoparticles for improved therapeutic effects in cervical cancer cells. Int. J. Nanomed, 2015, 10, 2461-2473.
[173]
Bernabeu, E.; Gonzalez, L.; Legaspi, M.J.; Moretton, M.A.; Chiappetta, D.A. Paclitaxel-loaded TPGS-b-PCL nanoparticles: In vitro cytotoxicity and cellular uptake in MCF-7 and MDA-MB-231 cells versus mPEG-b-PCL nanoparticles and Abraxane®. J. Nanosci. Nanotechnol., 2016, 16(1), 160-170.
[174]
Bernabeu, E.; Cagel, M.; Lagomarsino, E.; Moretton, M.; Chiappetta, D.A. Paclitaxel: What has been done and the challenges remain ahead. Int. J. Pharm., 2017, 526(1-2), 474-495.
[175]
Zheng, Y.; Chen, H.; Zeng, X.; Liu, Z.; Xiao, X.; Zhu, Y.; Gu, D.; Mei, L. Surface modification of TPGS-b-(PCL-ran-PGA) nanoparticles with polyethyleneimine as a co-delivery system of TRAIL and endostatin for cervical cancer gene therapy. Nanoscale Res. Lett., 2013, 8(1), 161.
[176]
Jiang, L.; Li, X.; Liu, L.; Zhang, Q. Thiolated chitosan-modified PLA-PCL-TPGS nanoparticles for oral chemotherapy of lung cancer. Nanoscale Res. Lett., 2013, 8(1), 66.
[177]
Yadav, M.K.; Maurya, A.K.; Rajput, G.; Manar, K.K.; Vinayak, M.; Drew, M.G.B.; Singh, N. Synthesis, characterization, DNA binding and cleavage activity of homoleptic zinc(II) β-oxodithioester chelate complexes. J. Coord. Chem., 2017, 70(18), 3171-3185.
[178]
Yadav, M.K.; Maurya, A.K.; Rajput, G.; Manar, K.K.; Vinayak, M.; Drew, M.G.B.; Singh, N. New planar trans-copper(II) β-dithioester chelate complexes: Synthesis, characterization, anticancer activity and DNA-binding/cleavage studies. J. Coord. Chem., 2017, 70(4), 565-583.
[179]
Pool, H.; Quintanar, D.; Figueroa, J.D.; Mano, M.C.; Bechara, J.E.H.; Godinez, L.A.; Mendoza, S. Antioxidant effects of quercetin and catechin encapsulated into PLGA nanoparticles. J. Nanomater., 2012, 2012, 12.
[180]
Jain, A.K.; Thanki, K.; Jain, S. Co-encapsulation of tamoxifen and quercetin in polymeric nanoparticles: Implications on oral bioavailability, antitumor efficacy, and drug-induced toxicity. Mol. Pharm., 2013, 10(9), 3459-3474.
[181]
Balakrishnan, S.; Mukherjee, S.; Das, S.; Bhat, F.A.; Raja Singh, P.; Patra, C.R.; Arunakaran, J. Gold nanoparticles-conjugated quercetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated pathway in breast cancer cell lines (MCF-7 and MDA-MB-231). Cell Biochem. Funct., 2017, 35(4), 217-231.
[182]
Sun, M.; Nie, S.; Pan, X.; Zhang, R.; Fan, Z.; Wang, S. Quercetin-nanostructured lipid carriers: Characteristics and anti-breast cancer activities in vitro. Colloids Surf. B Biointerfaces, 2014, 113, 15-24.
[183]
Sarkar, A.; Ghosh, S.; Chowdhury, S.; Pandey, B.; Sil, P.C. Targeted delivery of quercetin loaded mesoporous silica nanoparticles to the breast cancer cells. Biochim. Biophys. Acta, 2016, 1860(10), 2065-2075.
[184]
Aghapour, F.; Moghadamnia, A.A.; Nicolini, A.; Kani, S.N.M.; Barari, L.; Morakabati, P.; Rezazadeh, L.; Kazemi, S. Quercetin conjugated with silica nanoparticles inhibits tumor growth in MCF-7 breast cancer cell lines. Biochem. Biophys. Res. Commun., 2018, 500(4), 860-865.
[185]
Li, J.; Zhang, J.; Wang, Y.; Liang, X.; Wusiman, Z.; Yin, Y.; Shen, Q. Synergistic inhibition of migration and invasion of breast cancer cells by dual docetaxel/quercetin-loaded nanoparticles via Akt/MMP-9 pathway. Int. J. Pharm., 2017, 523(1), 300-309.
[186]
Umrao, S.; Maurya, A.K.; Shukla, V.; Grigoriev, A.; Ahuja, R.; Vinayak, M.; Srivastava, R.R.; Saxena, P.S.; Oh, I.K.; Srivastava, A. Anti-carcinogenic activity of blue fluorescent h-BN quantum dots: As an effective enhancer for dna cleavage activity of anti-cancer drug doxorubicin. Mater. Today Biol., 2019, 1100001
[187]
Cabral, H.; Kataoka, K. Progress of drug-loaded polymeric micelles into clinical studies. J. Control. Release, 2014, 190, 465-476.
[188]
Nishiyama, N.; Matsumura, Y.; Kataoka, K. Development of polymeric micelles for targeting intractable cancers. Cancer Sci., 2016, 107(7), 867-874.
[189]
Soleymani Abyaneh, H.; Vakili, M.R.; Zhang, F.; Choi, P.; Lavasanifar, A. Rational design of block copolymer micelles to control burst drug release at a nanoscale dimension. Acta Biomater., 2015, 24, 127-139.
[190]
Fonge, H.; Huang, H.; Scollard, D.; Reilly, R.M.; Allen, C. Influence of formulation variables on the biodistribution of multifunctional block copolymer micelles. J. Control. Release, 2012, 157(3), 366-374.
[191]
Bae, Y.; Kataoka, K. Intelligent polymeric micelles from functional poly(ethylene glycol)-poly(amino acid) block copolymers. Adv. Drug Deliv. Rev., 2009, 61(10), 768-784.
[192]
Matsukawa, N.; Matsumoto, M.; Shinoki, A.; Hagio, M.; Inoue, R.; Hara, H. Nondigestible saccharides suppress the bacterial degradation of quercetin aglycone in the large intestine and enhance the bioavailability of quercetin glucoside in rats. J. Agric. Food Chem., 2009, 57(20), 9462-9468.
[193]
Miyata, K.; Nishiyama, N.; Kataoka, K. Rational design of smart supramolecular assemblies for gene delivery: Chemical challenges in the creation of artificial viruses. Chem. Soc. Rev., 2012, 41(7), 2562-2574.
[194]
Liu, K.; Chen, W.; Yang, T.; Wen, B.; Ding, D.; Keidar, M.; Tang, J.; Zhang, W. Paclitaxel and quercetin nanoparticles co-loaded in microspheres to prolong retention time for pulmonary drug delivery. Int. J. Nanomed, 2017, 12, 8239-8255.
[195]
Mosqueira, V.C.; Legrand, P.; Gulik, A.; Bourdon, O.; Gref, R.; Labarre, D.; Barratt, G. Relationship between complement activation, cellular uptake and surface physicochemical aspects of novel PEG-modified nanocapsules. Biomaterials, 2001, 22(22), 2967-2979.
[196]
Murakami, M.; Cabral, H.; Matsumoto, Y.; Wu, S.; Kano, M.R.; Yamori, T.; Nishiyama, N.; Kataoka, K. Improving drug potency and efficacy by nanocarrier-mediated subcellular targeting. Sci. Transl. Med., 2011, 3(64), 64ra2.
[197]
Mochida, Y.; Cabral, H.; Kataoka, K. Polymeric micelles for targeted tumor therapy of platinum anticancer drugs. Expert Opin. Drug Deliv., 2017, 14(12), 1423-1438.
[198]
Takahashi, A.; Yamamoto, Y.; Yasunaga, M.; Koga, Y.; Kuroda, J.; Takigahira, M.; Harada, M.; Saito, H.; Hayashi, T.; Kato, Y.; Kinoshita, T.; Ohkohchi, N.; Hyodo, I.; Matsumura, Y. NC-6300, an epirubicin-incorporating micelle, extends the antitumor effect and reduces the cardiotoxicity of epirubicin. Cancer Sci., 2013, 104(7), 920-925.
[199]
Mukai, H.; Kogawa, T.; Matsubara, N.; Naito, Y.; Sasaki, M.; Hosono, A. A first-in-human Phase 1 study of epirubicin-conjugated polymer micelles (K-912/NC-6300) in patients with advanced or recurrent solid tumors. Invest. New Drugs, 2017, 35(3), 307-314.
[200]
Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.; Kimura, M.; Terada, Y.; Kano, M.R.; Miyazono, K.; Uesaka, M.; Nishiyama, N.; Kataoka, K. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat. Nanotechnol., 2011, 6(12), 815-823.
[201]
Chintamani; Tandon, M.; Mishra, A.; Agarwal, U.; Saxena, S. Sentinel lymph node biopsy using dye alone method is reliable and accurate even after neo-adjuvant chemotherapy in locally advanced breast cancer--a prospective study. World J. Surg. Oncol., 2011, 9, 19.
[202]
Ahn, H.K.; Jung, M.; Sym, S.J.; Shin, D.B.; Kang, S.M.; Kyung, S.Y.; Park, J.W.; Jeong, S.H.; Cho, E.K. A phase II trial of Cremorphor EL-free paclitaxel (Genexol-PM) and gemcitabine in patients with advanced non-small cell lung cancer. Cancer Chemother. Pharmacol., 2014, 74(2), 277-282.
[203]
Vega, J.; Ke, S.; Fan, Z.; Wallace, S.; Charsangavej, C.; Li, C. Targeting doxorubicin to epidermal growth factor receptors by site-specific conjugation of C225 to poly(L-glutamic acid) through a polyethylene glycol spacer. Pharm. Res., 2003, 20(5), 826-832.
[204]
Reddy, S.T.; van der Vlies, A.J.; Simeoni, E.; Angeli, V.; Randolph, G.J.; O’Neil, C.P.; Lee, L.K.; Swartz, M.A.; Hubbell, J.A. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat. Biotechnol., 2007, 25(10), 1159-1164.
[205]
Houdaihed, L.; Evans, J.C.; Allen, C. Overcoming the road blocks: Advancement of block copolymer micelles for cancer therapy in the clinic. Mol. Pharm., 2017, 14(8), 2503-2517.
[206]
Lee, K.S.; Chung, H.C. Im, S.A.; Park, Y.H.; Kim, C.S.; Kim, S.B.; Rha, S.Y.; Lee, M.Y.; Ro, J. Multicenter phase II trial of Genexol-PM, a Cremophor-free, polymeric micelle formulation of paclitaxel, in patients with metastatic breast cancer. Breast Cancer Res. Treat., 2008, 108(2), 241-250.
[207]
Kim, J.Y.; Do, Y.R.; Song, H.S.; Cho, Y.Y.; Ryoo, H.M.; Bae, S.H.; Kim, J.G.; Chae, Y.S.; Kang, B.W.; Baek, J.H.; Kim, M.K.; Lee, K.H.; Park, K. Multicenter phase II clinical trial of Genexol-PM® with gemcitabine in advanced biliary tract cancer. Anticancer Res., 2017, 37(3), 1467-1473.
[208]
Sutton, D.; Nasongkla, N.; Blanco, E.; Gao, J. Functionalized micellar systems for cancer targeted drug delivery. Pharm. Res., 2007, 24(6), 1029-1046.
[209]
Matsumura, Y. Preclinical and clinical studies of NK012, an SN-38-incorporating polymeric micelles, which is designed based on EPR effect. Adv. Drug Deliv. Rev., 2011, 63(3), 184-192.
[210]
Hamaguchi, T.; Kato, K.; Yasui, H.; Morizane, C.; Ikeda, M.; Ueno, H.; Muro, K.; Yamada, Y.; Okusaka, T.; Shirao, K.; Shimada, Y.; Nakahama, H.; Matsumura, Y. A phase I and pharmacokinetic study of NK105, a paclitaxel-incorporating micellar nanoparticle formulation. Br. J. Cancer, 2007, 97(2), 170-176.
[211]
Matsumura, Y. The drug discovery by nanomedicine and its clinical experience. Jpn. J. Clin. Oncol., 2014, 44(6), 515-525.
[212]
Wilson, R.H.; Plummer, R.; Adam, J.; Eatock, M.M.; Boddy, A.V.; Griffin, M.; Miller, R.; Matsumura, Y.; Shimizu, T.; Calvert, H. Phase I and pharmacokinetic study of NC-6004, a new platinum entity of cisplatin-conjugated polymer forming micelles. J. Clin. Oncol., 2008, 26(Suppl. 15), 2573-2573.
[213]
Wang, C.; Wu, C.; Zhou, X.; Han, T.; Xin, X.; Wu, J.; Zhang, J.; Guo, S. Enhancing cell nucleus accumulation and DNA cleavage activity of anti-cancer drug via graphene quantum dots. Sci. Rep., 2013, 3, 2852.
[214]
Dolatabadi, J.E.N.; Jamali, A. A.; Hasanzadeh, M.; Omidi, Y. Quercetin delivery into cancer cells with single walled carbon nanotubes. Intl. J. Biosci. Biochem. Bioinform., 2011, 21(a), 25.
[215]
Cirillo, G.; Vittorio, O.; Hampel, S.; Iemma, F.; Parchi, P.; Cecchini, M.; Puoci, F.; Picci, N. Quercetin nanocomposite as novel anticancer therapeutic: Improved efficiency and reduced toxicity. Eur. J. Pharm. Sci., 2013, 49(3), 359-365.
[216]
Wang, Q.; Bao, Y.; Ahire, J.; Chao, Y. Co-encapsulation of biodegradable nanoparticles with silicon quantum dots and quercetin for monitored delivery. Adv. Healthc. Mater., 2013, 2(3), 459-466.
[217]
Zhou, X.; Zhang, Y.; Wang, C.; Wu, X.; Yang, Y.; Zheng, B.; Wu, H.; Guo, S.; Zhang, J. Photo-fenton reaction of graphene oxide: A new strategy to prepare graphene quantum dots for DNA cleavage. ACS Nano, 2012, 6(8), 6592-6599.
[218]
Zhang, L.; Xia, J.; Zhao, Q.; Liu, L.; Zhang, Z. Functional graphene oxide as a nanocarrier for controlled loading and targeted delivery of mixed anticancer drugs. Small, 2010, 6(4), 537-544.
[219]
Zhang, Y.; Wang, J.; Bai, X.; Jiang, T.; Zhang, Q.; Wang, S. Mesoporous silica nanoparticles for increasing the oral bioavailability and permeation of poorly water soluble drugs. Mol. Pharm., 2012, 9(3), 505-513.
[220]
Sapino, S.; Ugazio, E.; Gastaldi, L.; Miletto, I.; Berlier, G.; Zonari, D.; Oliaro-Bosso, S. Mesoporous silica as topical nanocarriers for quercetin: characterization and in vitro studies. Eur. J. Pharm. Biopharm., 2015, 89, 116-125.