Bioactive Compounds: Multi-Targeting Silver Bullets for Preventing and Treating Breast Cancer
Abstract
:1. Bioactive Compounds in Cancer
2. Withaferin A, a Steroidal Lactone from Ashwagandha
3. Honokiol, a Polyphenol from Magnolia
4. Benzyl Isothiocyanate, an Isothiocynate from Cruciferous Vegetables
5. Resveratrol, a Polyphenolic Phytoestrogen
6. Curcumin, the Golden Spice
7. Genistein, a Phytoestrogen from Soy
8. Epigallocatechin-3-Gallate (EGCG), a Green Tea Polyphenol
9. Bioactive Compounds Based Anti-Cancer Drugs in Clinical Trials
10. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Sauter, E.R. Breast Cancer Prevention: Current Approaches and Future Directions. Eur. J. Breast Health 2018, 14, 64–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doyle, L.A.; Yang, W.; Abruzzo, L.V.; Krogmann, T.; Gao, Y.; Rishi, A.K.; Ross, D.D. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc. Natl. Acad. Sci. USA 1998, 95, 15665–15670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ju, J.; Zhu, A.J.; Yuan, P. Progress in targeted therapy for breast cancer. Chronic Dis. Transl. Med. 2018, 4, 164–175. [Google Scholar] [CrossRef]
- Parton, M.; Dowsett, M.; Smith, I. Studies of apoptosis in breast cancer. BMJ 2001, 322, 1528–1532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fridlender, M.; Kapulnik, Y.; Koltai, H. Plant derived substances with anti-cancer activity: From folklore to practice. Front. Plant Sci. 2015, 6, 799. [Google Scholar] [CrossRef] [PubMed]
- Biesalski, H.K.; Dragsted, L.O.; Elmadfa, I.; Grossklaus, R.; Muller, M.; Schrenk, D.; Walter, P.; Weber, P. Bioactive compounds: Definition and assessment of activity. Nutrition 2009, 25, 1202–1205. [Google Scholar] [CrossRef]
- Gali-Muhtasib, H.; Hmadi, R.; Kareh, M.; Tohme, R.; Darwiche, N. Cell death mechanisms of plant-derived anticancer drugs: Beyond apoptosis. Apoptosis 2015, 20, 1531–1562. [Google Scholar] [CrossRef]
- Korkina, L.; Kostyuk, V. Biotechnologically produced secondary plant metabolites for cancer treatment and prevention. Curr. Pharm. Biotechnol. 2012, 13, 265–275. [Google Scholar] [CrossRef]
- Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef]
- Teiten, M.H.; Gaascht, F.; Dicato, M.; Diederich, M. Anticancer bioactivity of compounds from medicinal plants used in European medieval traditions. Biochem. Pharmacol. 2013, 86, 1239–1247. [Google Scholar] [CrossRef]
- George, B.P.; Abrahamse, H. Increased Oxidative Stress Induced by Rubus Bioactive Compounds Induce Apoptotic Cell Death in Human Breast Cancer Cells. Oxidative Med. Cell. Longev. 2019, 2019, 6797921. [Google Scholar] [CrossRef] [PubMed]
- Shin, S.A.; Moon, S.Y.; Kim, W.Y.; Paek, S.M.; Park, H.H.; Lee, C.S. Structure-Based Classification and Anti-Cancer Effects of Plant Metabolites. Int. J. Mol. Sci. 2018, 19, 2651. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Khor, T.O.; Shu, L.; Su, Z.-Y.; Fuentes, F.; Lee, J.-H.; Kong, A.-N.T. Plants vs. Cancer: A Review on Natural Phytochemicals in Preventing and Treating Cancers and Their Druggability. Anti-Cancer Agents Med. Chem. 2012, 12, 1281–1305. [Google Scholar] [CrossRef]
- Cianciosi, D.; Varela-Lopez, A.; Forbes-Hernandez, T.Y.; Gasparrini, M.; Afrin, S.; Reboredo-Rodriguez, P.; Zhang, J.; Quiles, J.L.; Nabavi, S.F.; Battino, M.; et al. Targeting molecular pathways in cancer stem cells by natural bioactive compounds. Pharmacol. Res. 2018, 135, 150–165. [Google Scholar] [CrossRef]
- Wang, Y.; Li, Y.; Liu, X.; Cho, W.C. Genetic and epigenetic studies for determining molecular targets of natural product anticancer agents. Curr. Cancer Drug Targets 2013, 13, 506–518. [Google Scholar] [CrossRef]
- Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.D.C. Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies. Int. J. Mol. Sci. 2019, 20, 2313. [Google Scholar] [CrossRef]
- Aumeeruddy, M.Z.; Mahomoodally, M.F. Combating breast cancer using combination therapy with 3 phytochemicals: Piperine, sulforaphane, and thymoquinone. Cancer 2019, 125, 1600–1611. [Google Scholar] [CrossRef]
- Fu, B.; Wang, N.; Tan, H.Y.; Li, S.; Cheung, F.; Feng, Y. Multi-Component Herbal Products in the Prevention and Treatment of Chemotherapy-Associated Toxicity and Side Effects: A Review on Experimental and Clinical Evidences. Front. Pharmacol. 2018, 9, 1394. [Google Scholar] [CrossRef] [Green Version]
- Kura, B.; Parikh, M.; Slezak, J.; Pierce, G.N. The Influence of Diet on MicroRNAs that Impact Cardiovascular Disease. Molecules 2019, 24, 1509. [Google Scholar] [CrossRef]
- Mitra, S.; Dash, R. Natural Products for the Management and Prevention of Breast Cancer. Evid. Based Complement. Altern. Med. 2018, 2018, 8324696. [Google Scholar] [CrossRef]
- Kaileh, M.; Vanden Berghe, W.; Heyerick, A.; Horion, J.; Piette, J.; Libert, C.; De Keukeleire, D.; Essawi, T.; Haegeman, G. Withaferin a strongly elicits IkappaB kinase beta hyperphosphorylation concomitant with potent inhibition of its kinase activity. J. Biol. Chem. 2007, 282, 4253–4264. [Google Scholar] [CrossRef] [PubMed]
- Stan, S.D.; Hahm, E.R.; Warin, R.; Singh, S.V. Withaferin A causes FOXO3a- and Bim-dependent apoptosis and inhibits growth of human breast cancer cells in vivo. Cancer Res. 2008, 68, 7661–7669. [Google Scholar] [CrossRef] [PubMed]
- Misra, L.; Mishra, P.; Pandey, A.; Sangwan, R.S.; Sangwan, N.S.; Tuli, R. Withanolides from Withania somnifera roots. Phytochemistry 2008, 69, 1000–1004. [Google Scholar] [CrossRef] [PubMed]
- Stan, S.D.; Zeng, Y.; Singh, S.V. Ayurvedic medicine constituent withaferin a causes G2 and M phase cell cycle arrest in human breast cancer cells. Nutr. Cancer 2008, 60 (Suppl. 1), 51–60. [Google Scholar] [CrossRef]
- Nagalingam, A.; Kuppusamy, P.; Singh, S.V.; Sharma, D.; Saxena, N.K. Mechanistic elucidation of the antitumor properties of withaferin a in breast cancer. Cancer Res. 2014, 74, 2617–2629. [Google Scholar] [CrossRef]
- Hahm, E.R.; Lee, J.; Kim, S.H.; Sehrawat, A.; Arlotti, J.A.; Shiva, S.S.; Bhargava, R.; Singh, S.V. Metabolic alterations in mammary cancer prevention by withaferin A in a clinically relevant mouse model. J. Natl. Cancer Inst. 2013, 105, 1111–1122. [Google Scholar] [CrossRef]
- Hahm, E.R.; Moura, M.B.; Kelley, E.E.; Van Houten, B.; Shiva, S.; Singh, S.V. Withaferin A-induced apoptosis in human breast cancer cells is mediated by reactive oxygen species. PLoS ONE 2011, 6, e23354. [Google Scholar] [CrossRef]
- Hahm, E.R.; Singh, S.V. Withaferin A-induced apoptosis in human breast cancer cells is associated with suppression of inhibitor of apoptosis family protein expression. Cancer Lett. 2013, 334, 101–108. [Google Scholar] [CrossRef] [Green Version]
- Widodo, N.; Priyandoko, D.; Shah, N.; Wadhwa, R.; Kaul, S.C. Selective killing of cancer cells by Ashwagandha leaf extract and its component Withanone involves ROS signaling. PLoS ONE 2010, 5, e13536. [Google Scholar] [CrossRef]
- Liu, X.; Li, Y.; Ma, Q.; Wang, Y.; Song, A.L. Withaferin-A Inhibits Growth of Drug-Resistant Breast Carcinoma by Inducing Apoptosis and Autophagy, Endogenous Reactive Oxygen Species (ROS) Production, and Inhibition of Cell Migration and Nuclear Factor kappa B (Nf-kappaB)/Mammalian Target of Rapamycin (m-TOR) Signalling Pathway. Med. Sci. Monit. 2019, 25, 6855–6863. [Google Scholar] [CrossRef]
- Yco, L.P.; Mocz, G.; Opoku-Ansah, J.; Bachmann, A.S. Withaferin A Inhibits STAT3 and Induces Tumor Cell Death in Neuroblastoma and Multiple Myeloma. Biochem. Insights 2014, 7, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Sehrawat, A.; Samanta, S.K.; Hahm, E.R.; Croix, C.; Watkins, S.; Singh, S.V. Withaferin A-mediated apoptosis in breast cancer cells is associated with alterations in mitochondrial dynamics. Mitochondrion 2019, 47, 282–293. [Google Scholar] [CrossRef] [PubMed]
- Muniraj, N.; Siddharth, S.; Nagalingam, A.; Walker, A.; Woo, J.; Gyorffy, B.; Gabrielson, E.; Saxena, N.K.; Sharma, D. Withaferin A inhibits lysosomal activity to block autophagic flux and induces apoptosis via energetic impairment in breast cancer cells. Carcinogenesis 2019, 40, 1110–1120. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Mukerji, R.; Samadi, A.K.; Cohen, M.S. Down-regulation of estrogen receptor-alpha and rearranged during transfection tyrosine kinase is associated with withaferin a-induced apoptosis in MCF-7 breast cancer cells. BMC Complement. Altern. Med. 2011, 11, 84. [Google Scholar] [CrossRef]
- Hahm, E.R.; Lee, J.; Huang, Y.; Singh, S.V. Withaferin a suppresses estrogen receptor-alpha expression in human breast cancer cells. Mol. Carcinog. 2011, 50, 614–624. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Singh, S.V. Mammary cancer chemoprevention by withaferin A is accompanied by in vivo suppression of self-renewal of cancer stem cells. Cancer Prev. Res. 2014, 7, 738–747. [Google Scholar] [CrossRef] [PubMed]
- Royston, K.J.; Udayakumar, N.; Lewis, K.; Tollefsbol, T.O. A Novel Combination of Withaferin A and Sulforaphane Inhibits Epigenetic Machinery, Cellular Viability and Induces Apoptosis of Breast Cancer Cells. Int. J. Mol. Sci. 2017, 18, 1092. [Google Scholar] [CrossRef]
- Kakar, S.S.; Ratajczak, M.Z.; Powell, K.S.; Moghadamfalahi, M.; Miller, D.M.; Batra, S.K.; Singh, S.K. Withaferin a alone and in combination with cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS ONE 2014, 9, e107596. [Google Scholar] [CrossRef]
- Kakar, S.S.; Jala, V.R.; Fong, M.Y. Synergistic cytotoxic action of cisplatin and withaferin A on ovarian cancer cell lines. Biochem. Biophys. Res. Commun. 2012, 423, 819–825. [Google Scholar] [CrossRef] [Green Version]
- Hahm, E.R.; Lee, J.; Abella, T.; Singh, S.V. Withaferin A inhibits expression of ataxia telangiectasia and Rad3-related kinase and enhances sensitivity of human breast cancer cells to cisplatin. Mol. Carcinog. 2019, 58, 2139–2148. [Google Scholar] [CrossRef]
- Fried, L.E.; Arbiser, J.L. Honokiol, a multifunctional antiangiogenic and antitumor agent. Antioxid Redox Signal. 2009, 11, 1139–1148. [Google Scholar] [CrossRef]
- Amblard, F.; Govindarajan, B.; Lefkove, B.; Rapp, K.L.; Detorio, M.; Arbiser, J.L.; Schinazi, R.F. Synthesis, cytotoxicity, and antiviral activities of new neolignans related to honokiol and magnolol. Bioorg. Med. Chem. Lett. 2007, 17, 4428–4431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, N.; Song, Y.; Zhang, W.; Wang, W.; Chen, J.; Wong, A.W.; Roberts, A. Evaluation of the in vitro and in vivo genotoxicity of magnolia bark extract. Regul. Toxicol. Pharmacol. RTP 2007, 49, 154–159. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Zhang, X.; Cui, W.; Zhang, X.; Li, N.; Chen, J.; Wong, A.W.; Roberts, A. Evaluation of short-term and subchronic toxicity of magnolia bark extract in rats. Regul. Toxicol. Pharmacol. RTP 2007, 49, 160–171. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.J.; Lee, Y.M.; Lee, C.K.; Jung, J.K.; Han, S.B.; Hong, J.T. Therapeutic applications of compounds in the Magnolia family. Pharmacol. Ther. 2011, 130, 157–176. [Google Scholar] [CrossRef] [PubMed]
- Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
- Ikai, T.; Akao, Y.; Nakagawa, Y.; Ohguchi, K.; Sakai, Y.; Nozawa, Y. Magnolol-induced apoptosis is mediated via the intrinsic pathway with release of AIF from mitochondria in U937 cells. Biol. Pharm. Bull. 2006, 29, 2498–2501. [Google Scholar] [CrossRef]
- Rasul, A.; Yu, B.; Khan, M.; Zhang, K.; Iqbal, F.; Ma, T.; Yang, H. Magnolol, a natural compound, induces apoptosis of SGC-7901 human gastric adenocarcinoma cells via the mitochondrial and PI3K/Akt signaling pathways. Int. J. Oncol. 2012, 40, 1153–1161. [Google Scholar] [CrossRef]
- Fong, W.F.; Tse, A.K.; Poon, K.H.; Wang, C. Magnolol and honokiol enhance HL-60 human leukemia cell differentiation induced by 1,25-dihydroxyvitamin D3 and retinoic acid. Int. J. Biochem. Cell Biol. 2005, 37, 427–441. [Google Scholar] [CrossRef]
- Lee, S.J.; Cho, Y.H.; Park, K.; Kim, E.J.; Jung, K.H.; Park, S.S.; Kim, W.J.; Moon, S.K. Magnolol elicits activation of the extracellular signal-regulated kinase pathway by inducing p27KIP1-mediated G2/M-phase cell cycle arrest in human urinary bladder cancer 5637 cells. Biochem. Pharmacol. 2008, 75, 2289–2300. [Google Scholar] [CrossRef]
- Park, J.B.; Lee, M.S.; Cha, E.Y.; Lee, J.S.; Sul, J.Y.; Song, I.S.; Kim, J.Y. Magnolol-induced apoptosis in HCT-116 colon cancer cells is associated with the AMP-activated protein kinase signaling pathway. Biol. Pharm. Bull. 2012, 35, 1614–1620. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Bi, Y.; Yang, C.; Yang, J.; Jiang, Y.; Meng, F.; Yu, B.; Khan, M.; Ma, T.; Yang, H. Magnolol induces apoptosis in MCF-7 human breast cancer cells through G2/M phase arrest and caspase-independent pathway. Pharmazie 2013, 68, 755–762. [Google Scholar] [PubMed]
- Bai, X.; Cerimele, F.; Ushio-Fukai, M.; Waqas, M.; Campbell, P.M.; Govindarajan, B.; Der, C.J.; Battle, T.; Frank, D.A.; Ye, K.; et al. Honokiol, a small molecular weight natural product, inhibits angiogenesis in vitro and tumor growth in vivo. J. Biol. Chem. 2003, 278, 35501–35507. [Google Scholar] [CrossRef] [PubMed]
- Nagalingam, A.; Arbiser, J.L.; Bonner, M.Y.; Saxena, N.K.; Sharma, D. Honokiol activates AMP-activated protein kinase in breast cancer cells via an LKB1-dependent pathway and inhibits breast carcinogenesis. Breast Cancer Res. 2012, 14, R35. [Google Scholar] [CrossRef] [PubMed]
- Wolf, I.; O’Kelly, J.; Wakimoto, N.; Nguyen, A.; Amblard, F.; Karlan, B.Y.; Arbiser, J.L.; Koeffler, H.P. Honokiol, a natural biphenyl, inhibits in vitro and in vivo growth of breast cancer through induction of apoptosis and cell cycle arrest. Int. J. Oncol. 2007, 30, 1529–1537. [Google Scholar] [CrossRef]
- Garcia, A.; Zheng, Y.; Zhao, C.; Toschi, A.; Fan, J.; Shraibman, N.; Brown, H.A.; Bar-Sagi, D.; Foster, D.A.; Arbiser, J.L. Honokiol suppresses survival signals mediated by Ras-dependent phospholipase D activity in human cancer cells. Clin. Cancer Res. 2008, 14, 4267–4274. [Google Scholar] [CrossRef]
- Hou, W.; Chen, L.; Yang, G.; Zhou, H.; Jiang, Q.; Zhong, Z.; Hu, J.; Chen, X.; Wang, X.; Yuan, Y.; et al. Synergistic antitumor effects of liposomal honokiol combined with adriamycin in breast cancer models. Phytother. Res. 2008, 22, 1125–1132. [Google Scholar] [CrossRef]
- Tse, A.K.; Wan, C.K.; Shen, X.L.; Yang, M.; Fong, W.F. Honokiol inhibits TNF-alpha-stimulated NF-kappaB activation and NF-kappaB-regulated gene expression through suppression of IKK activation. Biochem. Pharmacol. 2005, 70, 1443–1457. [Google Scholar] [CrossRef]
- Samarajeewa, N.U.; Yang, F.; Docanto, M.M.; Sakurai, M.; McNamara, K.M.; Sasano, H.; Fox, S.B.; Simpson, E.R.; Brown, K.A. HIF-1alpha stimulates aromatase expression driven by prostaglandin E2 in breast adipose stroma. Breast Cancer Res. 2013, 15, R30. [Google Scholar] [CrossRef]
- Lan, K.L.; Lan, K.H.; Sheu, M.L.; Chen, M.Y.; Shih, Y.S.; Hsu, F.C.; Wang, H.M.; Liu, R.S.; Yen, S.H. Honokiol inhibits hypoxia-inducible factor-1 pathway. Int. J. Radiat. Biol. 2011, 87, 579–590. [Google Scholar] [CrossRef]
- Avtanski, D.B.; Nagalingam, A.; Bonner, M.Y.; Arbiser, J.L.; Saxena, N.K.; Sharma, D. Honokiol inhibits epithelial-mesenchymal transition in breast cancer cells by targeting signal transducer and activator of transcription 3/Zeb1/E-cadherin axis. Mol. Oncol. 2014, 8, 565–580. [Google Scholar] [CrossRef] [PubMed]
- Sengupta, S.; Nagalingam, A.; Muniraj, N.; Bonner, M.Y.; Mistriotis, P.; Afthinos, A.; Kuppusamy, P.; Lanoue, D.; Cho, S.; Korangath, P.; et al. Activation of tumor suppressor LKB1 by honokiol abrogates cancer stem-like phenotype in breast cancer via inhibition of oncogenic Stat3. Oncogene 2017, 36, 5709–5721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W.D.; Shang, Y.; Li, Y.; Chen, S.Z. Honokiol inhibits breast cancer cell metastasis by blocking EMT through modulation of Snail/Slug protein translation. Acta Pharmacol. Sin. 2019, 40, 1219–1227. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Zang, C.; Emde, A.; Planas-Silva, M.D.; Rosche, M.; Kuhnl, A.; Schulz, C.O.; Elstner, E.; Possinger, K.; Eucker, J. Anti-tumor effect of honokiol alone and in combination with other anti-cancer agents in breast cancer. Eur. J. Pharmacol. 2008, 591, 43–51. [Google Scholar] [CrossRef]
- Xie, L.; Jiang, F.; Zhang, X.; Alitongbieke, G.; Shi, X.; Meng, M.; Xu, Y.; Ren, A.; Wang, J.; Cai, L.; et al. Honokiol sensitizes breast cancer cells to TNF-alpha induction of apoptosis by inhibiting Nur77 expression. Br. J. Pharmacol. 2016, 173, 344–356. [Google Scholar] [CrossRef]
- Avtanski, D.B.; Nagalingam, A.; Bonner, M.Y.; Arbiser, J.L.; Saxena, N.K.; Sharma, D. Honokiol activates LKB1-miR-34a axis and antagonizes the oncogenic actions of leptin in breast cancer. Oncotarget 2015, 6, 29947–29962. [Google Scholar] [CrossRef] [Green Version]
- Avtanski, D.B.; Nagalingam, A.; Kuppusamy, P.; Bonner, M.Y.; Arbiser, J.L.; Saxena, N.K.; Sharma, D. Honokiol abrogates leptin-induced tumor progression by inhibiting Wnt1-MTA1-beta-catenin signaling axis in a microRNA-34a dependent manner. Oncotarget 2015, 6, 16396–16410. [Google Scholar] [CrossRef]
- Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef]
- Kagara, N.; Huynh, K.T.; Kuo, C.; Okano, H.; Sim, M.S.; Elashoff, D.; Chong, K.; Giuliano, A.E.; Hoon, D.S. Epigenetic regulation of cancer stem cell genes in triple-negative breast cancer. Am. J. Pathol. 2012, 181, 257–267. [Google Scholar] [CrossRef]
- Chang, M.T.; Lee, S.P.; Fang, C.Y.; Hsieh, P.L.; Liao, Y.W.; Lu, M.Y.; Tsai, L.L.; Yu, C.C.; Liu, C.M. Chemosensitizing effect of honokiol in oral carcinoma stem cells via regulation of IL-6/Stat3 signaling. Environ. Toxicol. 2018, 33, 1105–1112. [Google Scholar] [CrossRef]
- Gao, X.; Patel, M.G.; Bakshi, P.; Sharma, D.; Banga, A.K. Enhancement in the Transdermal and Localized Delivery of Honokiol Through Breast Tissue. AAPS PharmSciTech 2018, 19, 3501–3511. [Google Scholar] [CrossRef] [PubMed]
- Verhoeven, D.T.; Goldbohm, R.A.; van Poppel, G.; Verhagen, H.; van den Brandt, P.A. Epidemiological studies on brassica vegetables and cancer risk. Cancer Epidemiol. Biomark. Prev. 1996, 5, 733–748. [Google Scholar]
- Kelloff, G.J.; Boone, C.W.; Crowell, J.A.; Steele, V.E.; Lubet, R.A.; Doody, L.A.; Malone, W.F.; Hawk, E.T.; Sigman, C.C. New agents for cancer chemoprevention. J. Cell. Biochem. Suppl. 1996, 26, 1–28. [Google Scholar] [CrossRef]
- Conaway, C.C.; Yang, Y.M.; Chung, F.L. Isothiocyanates as cancer chemopreventive agents: Their biological activities and metabolism in rodents and humans. Curr. Drug Metab. 2002, 3, 233–255. [Google Scholar] [CrossRef] [PubMed]
- Shapiro, T.A.; Fahey, J.W.; Wade, K.L.; Stephenson, K.K.; Talalay, P. Human metabolism and excretion of cancer chemoprotective glucosinolates and isothiocyanates of cruciferous vegetables. Cancer Epidemiol. Biomark. Prev. 1998, 7, 1091–1100. [Google Scholar]
- Ambrosone, C.B.; McCann, S.E.; Freudenheim, J.L.; Marshall, J.R.; Zhang, Y.; Shields, P.G. Breast cancer risk in premenopausal women is inversely associated with consumption of broccoli, a source of isothiocyanates, but is not modified by GST genotype. J. Nutr. 2004, 134, 1134–1138. [Google Scholar] [CrossRef]
- Singh, S.V.; Singh, K. Cancer chemoprevention with dietary isothiocyanates mature for clinical translational research. Carcinogenesis 2012, 33, 1833–1842. [Google Scholar] [CrossRef] [Green Version]
- Rao, C.V. Benzyl isothiocyanate: Double trouble for breast cancer cells. Cancer Prev. Res. 2013, 6, 760–763. [Google Scholar] [CrossRef]
- Tseng, E.; Scott-Ramsay, E.A.; Morris, M.E. Dietary organic isothiocyanates are cytotoxic in human breast cancer MCF-7 and mammary epithelial MCF-12A cell lines. Exp. Biol. Med. 2004, 229, 83–842. [Google Scholar] [CrossRef]
- Nakamura, Y.; Miyoshi, N. Cell death induction by isothiocyanates and their underlying molecular mechanisms. BioFactors 2006, 26, 123–134. [Google Scholar] [CrossRef]
- Jeong, Y.J.; Cho, H.J.; Chung, F.L.; Wang, X.; Hoe, H.S.; Park, K.K.; Kim, C.H.; Chang, H.W.; Lee, S.R.; Chang, Y.C. Isothiocyanates suppress the invasion and metastasis of tumors by targeting FAK/MMP-9 activity. Oncotarget 2017, 8, 63949–63962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Warin, R.; Xiao, D.; Arlotti, J.A.; Bommareddy, A.; Singh, S.V. Inhibition of human breast cancer xenograft growth by cruciferous vegetable constituent benzyl isothiocyanate. Mol. Carcinog. 2010, 49, 500–507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hecht, S.S. Inhibition of carcinogenesis by isothiocyanates. Drug Metab. Rev. 2000, 32, 395–411. [Google Scholar] [CrossRef] [PubMed]
- Kim, E.J.; Hong, J.E.; Eom, S.J.; Lee, J.Y.; Park, J.H. Oral administration of benzyl-isothiocyanate inhibits solid tumor growth and lung metastasis of 4T1 murine mammary carcinoma cells in BALB/c mice. Breast Cancer Res. Treat. 2011, 130, 61–71. [Google Scholar] [CrossRef] [PubMed]
- Warin, R.; Chambers, W.H.; Potter, D.M.; Singh, S.V. Prevention of mammary carcinogenesis in MMTV-neu mice by cruciferous vegetable constituent benzyl isothiocyanate. Cancer Res. 2009, 69, 9473–9480. [Google Scholar] [CrossRef]
- Yu, R.; Mandlekar, S.; Harvey, K.J.; Ucker, D.S.; Kong, A.N. Chemopreventive isothiocyanates induce apoptosis and caspase-3-like protease activity. Cancer Res. 1998, 58, 402–408. [Google Scholar]
- Xiao, D.; Vogel, V.; Singh, S.V. Benzyl isothiocyanate-induced apoptosis in human breast cancer cells is initiated by reactive oxygen species and regulated by Bax and Bak. Mol. Cancer Ther. 2006, 5, 2931–2945. [Google Scholar] [CrossRef]
- Xiao, D.; Powolny, A.A.; Singh, S.V. Benzyl isothiocyanate targets mitochondrial respiratory chain to trigger reactive oxygen species-dependent apoptosis in human breast cancer cells. J. Biol. Chem. 2008, 283, 30151–30163. [Google Scholar] [CrossRef]
- Kang, L.; Ding, L.; Wang, Z.Y. Isothiocyanates repress estrogen receptor alpha expression in breast cancer cells. Oncol. Rep. 2009, 21, 185–192. [Google Scholar]
- Xiao, D.; Bommareddy, A.; Kim, S.H.; Sehrawat, A.; Hahm, E.R.; Singh, S.V. Benzyl isothiocyanate causes FoxO1-mediated autophagic death in human breast cancer cells. PLoS ONE 2012, 7, e32597. [Google Scholar] [CrossRef]
- Sehrawat, A.; Singh, S.V. Benzyl isothiocyanate inhibits epithelial-mesenchymal transition in cultured and xenografted human breast cancer cells. Cancer Prev. Res. 2011, 4, 1107–1117. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Singh, S.V. Role of Kruppel-like Factor 4-p21(CIP1) Axis in Breast Cancer Stem-like Cell Inhibition by Benzyl Isothiocyanate. Cancer Prev. Res. 2019, 12, 125–134. [Google Scholar] [CrossRef] [PubMed]
- Xie, B.; Zhao, L.; Guo, L.; Liu, H.; Fu, S.; Fan, W.; Lin, L.; Chen, J.; Wang, B.; Fan, L.; et al. Benzyl isothiocyanate suppresses development and metastasis of murine mammary carcinoma by regulating the Wnt/betacatenin pathway. Mol. Med. Rep. 2019, 20, 1808–1818. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Singh, S.V. p53-Independent apoptosis by benzyl isothiocyanate in human breast cancer cells is mediated by suppression of XIAP expression. Cancer Prev. Res. 2010, 3, 718–726. [Google Scholar] [CrossRef] [PubMed]
- Xie, B.; Nagalingam, A.; Kuppusamy, P.; Muniraj, N.; Langford, P.; Gyorffy, B.; Saxena, N.K.; Sharma, D. Benzyl Isothiocyanate potentiates p53 signaling and antitumor effects against breast cancer through activation of p53-LKB1 and p73-LKB1 axes. Sci. Rep. 2017, 7, 40070. [Google Scholar] [CrossRef] [Green Version]
- Saxena, N.K.; Vertino, P.M.; Anania, F.A.; Sharma, D. leptin-induced growth stimulation of breast cancer cells involves recruitment of histone acetyltransferases and mediator complex to CYCLIN D1 promoter via activation of Stat3. J. Biol. Chem. 2007, 282, 13316–13325. [Google Scholar] [CrossRef]
- Saxena, N.K.; Taliaferro-Smith, L.; Knight, B.B.; Merlin, D.; Anania, F.A.; O’Regan, R.M.; Sharma, D. Bidirectional crosstalk between leptin and insulin-like growth factor-I signaling promotes invasion and migration of breast cancer cells via transactivation of epidermal growth factor receptor. Cancer Res. 2008, 68, 9712–9722. [Google Scholar] [CrossRef]
- Knight, B.B.; Oprea-Ilies, G.M.; Nagalingam, A.; Yang, L.; Cohen, C.; Saxena, N.K.; Sharma, D. Survivin upregulation, dependent on leptin-EGFR-Notch1 axis, is essential for leptin-induced migration of breast carcinoma cells. Endocr. Relat. Cancer 2011, 18, 413–428. [Google Scholar] [CrossRef]
- Yan, D.; Avtanski, D.; Saxena, N.K.; Sharma, D. Leptin-induced epithelial-mesenchymal transition in breast cancer cells requires beta-catenin activation via Akt/GSK3- and MTA1/Wnt1 protein-dependent pathways. J. Biol. Chem. 2012, 287, 8598–8612. [Google Scholar] [CrossRef]
- Saxena, N.K.; Sharma, D. Multifaceted leptin network: The molecular connection between obesity and breast cancer. J. Mammary Gland. Biol. Neoplasia 2013, 18, 309–320. [Google Scholar] [CrossRef]
- Taliaferro-Smith, L.; Nagalingam, A.; Knight, B.B.; Oberlick, E.; Saxena, N.K.; Sharma, D. Integral role of PTP1B in adiponectin-mediated inhibition of oncogenic actions of leptin in breast carcinogenesis. Neoplasia 2013, 15, 23–38. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Nagalingam, A.; Saxena, N.K.; Singh, S.V.; Sharma, D. Benzyl isothiocyanate inhibits oncogenic actions of leptin in human breast cancer cells by suppressing activation of signal transducer and activator of transcription 3. Carcinogenesis 2011, 32, 359–367. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Sehrawat, A.; Singh, S.V. Dietary chemopreventative benzyl isothiocyanate inhibits breast cancer stem cells in vitro and in vivo. Cancer Prev. Res. 2013, 6, 782–790. [Google Scholar] [CrossRef] [PubMed]
- Sahu, R.P.; Srivastava, S.K. The role of STAT-3 in the induction of apoptosis in pancreatic cancer cells by benzyl isothiocyanate. J. Natl. Cancer Inst. 2009, 101, 176–193. [Google Scholar] [CrossRef] [PubMed]
- Yeh, Y.T.; Hsu, Y.N.; Huang, S.Y.; Lin, J.S.; Chen, Z.F.; Chow, N.H.; Su, S.H.; Shyu, H.W.; Lin, C.C.; Huang, W.T.; et al. Benzyl isothiocyanate promotes apoptosis of oral cancer cells via an acute redox stress-mediated DNA damage response. Food Chem. Toxicol. 2016, 97, 336–345. [Google Scholar] [CrossRef] [PubMed]
- Pezzuto, J.M. Grapes and human health: A perspective. J. Agric. Food Chem. 2008, 56, 6777–6784. [Google Scholar] [CrossRef] [PubMed]
- Rimando, A.M.; Kalt, W.; Magee, J.B.; Dewey, J.; Ballington, J.R. Resveratrol, pterostilbene, and piceatannol in vaccinium berries. J. Agric. Food Chem. 2004, 52, 4713–4719. [Google Scholar] [CrossRef]
- Sales, J.M.; Resurreccion, A.V. Resveratrol in peanuts. Crit. Rev. Food Sci. Nutr. 2014, 54, 734–770. [Google Scholar] [CrossRef]
- Sun, A.Y.; Simonyi, A.; Sun, G.Y. The “French Paradox” and beyond: Neuroprotective effects of polyphenols. Free Radic. Biol. Med. 2002, 32, 314–318. [Google Scholar] [CrossRef]
- Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef]
- Bhat, K.P.; Lantvit, D.; Christov, K.; Mehta, R.G.; Moon, R.C.; Pezzuto, J.M. Estrogenic and antiestrogenic properties of resveratrol in mammary tumor models. Cancer Res. 2001, 61, 7456–7463. [Google Scholar] [PubMed]
- Dong, Z. Molecular mechanism of the chemopreventive effect of resveratrol. Mutat. Res. 2003, 523–524, 145–150. [Google Scholar] [CrossRef]
- Le Corre, L.; Chalabi, N.; Delort, L.; Bignon, Y.J.; Bernard-Gallon, D.J. Resveratrol and breast cancer chemoprevention: Molecular mechanisms. Mol. Nutr. Food Res. 2005, 49, 462–471. [Google Scholar] [CrossRef] [PubMed]
- Kundu, J.K.; Surh, Y.J. Cancer chemopreventive and therapeutic potential of resveratrol: Mechanistic perspectives. Cancer Lett. 2008, 269, 243–261. [Google Scholar] [CrossRef]
- Shukla, Y.; Singh, R. Resveratrol and cellular mechanisms of cancer prevention. Ann. N. Y. Acad. Sci. 2011, 1215, 1–8. [Google Scholar] [CrossRef]
- Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef]
- Pozo-Guisado, E.; Merino, J.M.; Mulero-Navarro, S.; Lorenzo-Benayas, M.J.; Centeno, F.; Alvarez-Barrientos, A.; Fernandez-Salguero, P.M. Resveratrol-induced apoptosis in MCF-7 human breast cancer cells involves a caspase-independent mechanism with downregulation of Bcl-2 and NF-kappaB. Int. J. Cancer 2005, 115, 74–84. [Google Scholar] [CrossRef]
- Alkhalaf, M.; El-Mowafy, A.; Renno, W.; Rachid, O.; Ali, A.; Al-Attyiah, R. Resveratrol-induced apoptosis in human breast cancer cells is mediated primarily through the caspase-3-dependent pathway. Arch. Med. Res. 2008, 39, 162–168. [Google Scholar] [CrossRef]
- Chen, F.P.; Chien, M.H. Phytoestrogens induce apoptosis through a mitochondria/caspase pathway in human breast cancer cells. Climacteric 2014, 17, 385–392. [Google Scholar] [CrossRef]
- Sareen, D.; Darjatmoko, S.R.; Albert, D.M.; Polans, A.S. Mitochondria, calcium, and calpain are key mediators of resveratrol-induced apoptosis in breast cancer. Mol. Pharmacol. 2007, 72, 1466–1475. [Google Scholar] [CrossRef]
- Nguyen, T.H.; Mustafa, F.B.; Pervaiz, S.; Ng, F.S.; Lim, L.H. ERK1/2 activation is required for resveratrol-induced apoptosis in MDA-MB-231 cells. Int. J. Oncol. 2008, 33, 81–92. [Google Scholar] [CrossRef] [PubMed]
- Tsai, J.H.; Hsu, L.S.; Lin, C.L.; Hong, H.M.; Pan, M.H.; Way, T.D.; Chen, W.J. 3,5,4’-Trimethoxystilbene, a natural methoxylated analog of resveratrol, inhibits breast cancer cell invasiveness by downregulation of PI3K/Akt and Wnt/beta-catenin signaling cascades and reversal of epithelial-mesenchymal transition. Toxicol. Appl. Pharmacol. 2013, 272, 746–756. [Google Scholar] [CrossRef] [PubMed]
- Castillo-Pichardo, L.; Dharmawardhane, S.F. Grape polyphenols inhibit Akt/mammalian target of rapamycin signaling and potentiate the effects of gefitinib in breast cancer. Nutr. Cancer 2012, 64, 1058–1069. [Google Scholar] [CrossRef] [PubMed]
- Fu, Y.; Chang, H.; Peng, X.; Bai, Q.; Yi, L.; Zhou, Y.; Zhu, J.; Mi, M. Resveratrol inhibits breast cancer stem-like cells and induces autophagy via suppressing Wnt/beta-catenin signaling pathway. PLoS ONE 2014, 9, e102535. [Google Scholar] [CrossRef]
- Kotha, A.; Sekharam, M.; Cilenti, L.; Siddiquee, K.; Khaled, A.; Zervos, A.S.; Carter, B.; Turkson, J.; Jove, R. Resveratrol inhibits Src and Stat3 signaling and induces the apoptosis of malignant cells containing activated Stat3 protein. Mol. Cancer Ther. 2006, 5, 621–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- She, Q.B.; Bode, A.M.; Ma, W.Y.; Chen, N.Y.; Dong, Z. Resveratrol-induced activation of p53 and apoptosis is mediated by extracellular-signal-regulated protein kinases and p38 kinase. Cancer Res. 2001, 61, 1604–1610. [Google Scholar]
- Hsieh, T.C.; Wong, C.; John Bennett, D.; Wu, J.M. Regulation of p53 and cell proliferation by resveratrol and its derivatives in breast cancer cells: An in silico and biochemical approach targeting integrin alphavbeta3. Int. J. Cancer 2011, 129, 2732–2743. [Google Scholar] [CrossRef]
- Chatterjee, B.; Ghosh, K.; Kanade, S.R. Resveratrol modulates epigenetic regulators of promoter histone methylation and acetylation that restores BRCA1, p53, p21(CIP1) in human breast cancer cell lines. BioFactors 2019. [Google Scholar] [CrossRef]
- Gehm, B.D.; McAndrews, J.M.; Chien, P.Y.; Jameson, J.L. Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc. Natl. Acad. Sci. USA 1997, 94, 14138–14143. [Google Scholar] [CrossRef] [Green Version]
- Bowers, J.L.; Tyulmenkov, V.V.; Jernigan, S.C.; Klinge, C.M. Resveratrol acts as a mixed agonist/antagonist for estrogen receptors alpha and beta. Endocrinology 2000, 141, 3657–3667. [Google Scholar] [CrossRef]
- Jin, X.; Wei, Y.; Liu, Y.; Lu, X.; Ding, F.; Wang, J.; Yang, S. Resveratrol promotes sensitization to Doxorubicin by inhibiting epithelial-mesenchymal transition and modulating SIRT1/beta-catenin signaling pathway in breast cancer. Cancer Med. 2019, 8, 1246–1257. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Jiang, H.; Chen, Y.; Ren, F. Resveratrol chemosensitizes adriamycin-resistant breast cancer cells by modulating miR-122-5p. J. Cell. Biochem. 2019, 120, 16283–16292. [Google Scholar] [CrossRef] [PubMed]
- Hallajian, F.; Ghasmi, M.; Abedi, S.M.; Behzadi, R.; Hayati, E.; Sadeghzadeh, N.; Rezazadeh, F.; Karimi, H. Evaluation of the Effect of Resveratrol and Doxorubicin on (99m)Tc-MIBI Uptake in Breast Cancer Cell Xenografts in Mice. Cancer Biother. Radiopharm. 2018, 33, 403–410. [Google Scholar] [CrossRef] [PubMed]
- Sinha, D.; Biswas, J.; Sung, B.; Aggarwal, B.B.; Bishayee, A. Chemopreventive and chemotherapeutic potential of curcumin in breast cancer. Curr. Drug Targets 2012, 13, 1799–1819. [Google Scholar] [CrossRef] [PubMed]
- Basnet, P.; Skalko-Basnet, N. Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 2011, 16, 4567–4598. [Google Scholar] [CrossRef] [PubMed]
- Kumar, A.; Chetia, H.; Sharma, S.; Kabiraj, D.; Talukdar, N.C.; Bora, U. Curcumin Resource Database. Database 2015, 2015, bav070. [Google Scholar] [CrossRef] [Green Version]
- Ramachandran, C.; You, W. Differential sensitivity of human mammary epithelial and breast carcinoma cell lines to curcumin. Breast Cancer Res. Treat. 1999, 54, 269–278. [Google Scholar] [CrossRef]
- Choudhuri, T.; Pal, S.; Agwarwal, M.L.; Das, T.; Sa, G. Curcumin induces apoptosis in human breast cancer cells through p53-dependent Bax induction. FEBS Lett. 2002, 512, 334–340. [Google Scholar] [CrossRef]
- Lv, Z.D.; Liu, X.P.; Zhao, W.J.; Dong, Q.; Li, F.N.; Wang, H.B.; Kong, B. Curcumin induces apoptosis in breast cancer cells and inhibits tumor growth in vitro and in vivo. Int. J. Clin. Exp. Pathol. 2014, 7, 2818–2824. [Google Scholar]
- Fan, H.; Liang, Y.; Jiang, B.; Li, X.; Xun, H.; Sun, J.; He, W.; Lau, H.T.; Ma, X. Curcumin inhibits intracellular fatty acid synthase and induces apoptosis in human breast cancer MDA-MB-231 cells. Oncol. Rep. 2016, 35, 2651–2656. [Google Scholar] [CrossRef] [Green Version]
- Chang, L.C.; Hsieh, M.T.; Yang, J.S.; Lu, C.C.; Tsai, F.J.; Tsao, J.W.; Chiu, Y.J.; Kuo, S.C.; Lee, K.H. Effect of bis(hydroxymethyl) alkanoate curcuminoid derivative MTH-3 on cell cycle arrest, apoptotic and autophagic pathway in triple-negative breast adenocarcinoma MDA-MB-231 cells: An in vitro study. Int. J. Oncol. 2018, 52, 67–76. [Google Scholar] [CrossRef] [PubMed]
- Ravindran, J.; Prasad, S.; Aggarwal, B.B. Curcumin and cancer cells: How many ways can curry kill tumor cells selectively? AAPS J. 2009, 11, 495–510. [Google Scholar] [CrossRef] [PubMed]
- Hu, S.; Xu, Y.; Meng, L.; Huang, L.; Sun, H. Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells. Exp. Ther. Med. 2018, 16, 1266–1272. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.K.; Lee, W.S.; Hwang, J.T.; Kwon, D.Y.; Surh, Y.J.; Park, O.J. Curcumin exerts antidifferentiation effect through AMPKalpha-PPAR-gamma in 3T3-L1 adipocytes and antiproliferatory effect through AMPKalpha-COX-2 in cancer cells. J. Agric. Food Chem. 2009, 57, 305–310. [Google Scholar] [CrossRef]
- Guan, F.; Ding, Y.; Zhang, Y.; Zhou, Y.; Li, M.; Wang, C. Curcumin Suppresses Proliferation and Migration of MDA-MB-231 Breast Cancer Cells through Autophagy-Dependent Akt Degradation. PLoS ONE 2016, 11, e0146553. [Google Scholar] [CrossRef]
- Ren, M.; Wang, Y.; Wu, X.; Ge, S.; Wang, B. Curcumin synergistically increases effects of beta-interferon and retinoic acid on breast cancer cells in vitro and in vivo by up-regulation of GRIM-19 through STAT3-dependent and STAT3-independent pathways. J. Drug Target. 2017, 25, 247–254. [Google Scholar] [CrossRef]
- Somasundaram, S.; Edmund, N.A.; Moore, D.T.; Small, G.W.; Shi, Y.Y.; Orlowski, R.Z. Dietary curcumin inhibits chemotherapy-induced apoptosis in models of human breast cancer. Cancer Res. 2002, 62, 3868–3875. [Google Scholar]
- Goel, A.; Aggarwal, B.B. Curcumin, the golden spice from Indian saffron, is a chemosensitizer and radiosensitizer for tumors and chemoprotector and radioprotector for normal organs. Nutr. Cancer 2010, 62, 919–930. [Google Scholar] [CrossRef]
- Aggarwal, B.B.; Shishodia, S.; Takada, Y.; Banerjee, S.; Newman, R.A.; Bueso-Ramos, C.E.; Price, J.E. Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB pathway in breast cancer cells and inhibits lung metastasis of human breast cancer in nude mice. Clin. Cancer Res. 2005, 11, 7490–7498. [Google Scholar] [CrossRef]
- Zhang, P.; Lai, Z.L.; Chen, H.F.; Zhang, M.; Wang, A.; Jia, T.; Sun, W.Q.; Zhu, X.M.; Chen, X.F.; Zhao, Z.; et al. Curcumin synergizes with 5-fluorouracil by impairing AMPK/ULK1-dependent autophagy, AKT activity and enhancing apoptosis in colon cancer cells with tumor growth inhibition in xenograft mice. J. Exp. Clin. Cancer Res. CR 2017, 36, 190. [Google Scholar] [CrossRef]
- Calaf, G.M.; Ponce-Cusi, R.; Carrion, F. Curcumin and paclitaxel induce cell death in breast cancer cell lines. Oncol. Rep. 2018, 40, 2381–2388. [Google Scholar] [CrossRef] [PubMed]
- Dayton, A.; Selvendiran, K.; Meduru, S.; Khan, M.; Kuppusamy, M.L.; Naidu, S.; Kalai, T.; Hideg, K.; Kuppusamy, P. Amelioration of doxorubicin-induced cardiotoxicity by an anticancer-antioxidant dual-function compound, HO-3867. J. Pharmacol. Exp. Ther. 2011, 339, 350–357. [Google Scholar] [CrossRef] [PubMed]
- Sen, G.S.; Mohanty, S.; Hossain, D.M.; Bhattacharyya, S.; Banerjee, S.; Chakraborty, J.; Saha, S.; Ray, P.; Bhattacharjee, P.; Mandal, D.; et al. Curcumin enhances the efficacy of chemotherapy by tailoring p65NFkappaB-p300 cross-talk in favor of p53-p300 in breast cancer. J. Biol. Chem. 2011, 286, 42232–42247. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.C.; Lai, Y.A.; Lin, Y.C.; Ma, J.W.; Huang, L.F.; Yang, N.S.; Ho, C.T.; Kuo, S.C.; Way, T.D. Curcumin suppresses doxorubicin-induced epithelial-mesenchymal transition via the inhibition of TGF-beta and PI3K/AKT signaling pathways in triple-negative breast cancer cells. J. Agric. Food Chem. 2013, 61, 11817–11824. [Google Scholar] [CrossRef] [PubMed]
- Duan, J.; Mansour, H.M.; Zhang, Y.; Deng, X.; Chen, Y.; Wang, J.; Pan, Y.; Zhao, J. Reversion of multidrug resistance by co-encapsulation of doxorubicin and curcumin in chitosan/poly(butyl cyanoacrylate) nanoparticles. Int. J. Pharm. 2012, 426, 193–201. [Google Scholar] [CrossRef] [PubMed]
- Guo, O.; Li, X.; Yang, Y.; Wei, J.; Zhao, Q.; Luo, F.; Qian, Z. Enhanced 4T1 breast carcinoma anticancer activity by co-delivery of doxorubicin and curcumin with core-shell drug-carrier based on heparin modified poly(L-lactide) grafted polyethylenimine cationic nanoparticles. J. Biomed. Nanotechnol. 2014, 10, 227–237. [Google Scholar] [CrossRef]
- Minafra, L.; Porcino, N.; Bravata, V.; Gaglio, D.; Bonanomi, M.; Amore, E.; Cammarata, F.P.; Russo, G.; Militello, C.; Savoca, G.; et al. Radiosensitizing effect of curcumin-loaded lipid nanoparticles in breast cancer cells. Sci. Rep. 2019, 9, 11134. [Google Scholar] [CrossRef]
- Harini, L.; Srivastava, S.; Gnanakumar, G.P.; Karthikeyan, B.; Ross, C.; Krishnakumar, V.; Kannan, V.R.; Sundar, K.; Kathiresan, T. An ingenious non-spherical mesoporous silica nanoparticle cargo with curcumin induces mitochondria-mediated apoptosis in breast cancer (MCF-7) cells. Oncotarget 2019, 10, 1193–1208. [Google Scholar] [CrossRef] [Green Version]
- Saleh, T.; Soudi, T.; Shojaosadati, S.A. Aptamer functionalized curcumin-loaded human serum albumin (HSA) nanoparticles for targeted delivery to HER-2 positive breast cancer cells. Int. J. Biol. Macromol. 2019, 130, 109–116. [Google Scholar] [CrossRef]
- Wong, K.E.; Ngai, S.C.; Chan, K.G.; Lee, L.H.; Goh, B.H.; Chuah, L.H. Curcumin Nanoformulations for Colorectal Cancer: A Review. Front. Pharmacol. 2019, 10, 152. [Google Scholar] [CrossRef]
- Feng, T.; Wei, Y.; Lee, R.J.; Zhao, L. Liposomal curcumin and its application in cancer. Int. J. Nanomed. 2017, 12, 6027–6044. [Google Scholar] [CrossRef] [PubMed]
- Pavese, J.M.; Krishna, S.N.; Bergan, R.C. Genistein inhibits human prostate cancer cell detachment, invasion, and metastasis. Am. J. Clin. Nutr. 2014, 100 (Suppl. 1), 431S–436S. [Google Scholar] [CrossRef]
- Lee, J.Y.; Kim, H.S.; Song, Y.S. Genistein as a Potential Anticancer Agent against Ovarian Cancer. J. Tradit. Complementary Med. 2012, 2, 96–104. [Google Scholar] [CrossRef] [Green Version]
- Ziegler, R.G. Phytoestrogens and breast cancer. Am. J. Clin. Nutr. 2004, 79, 183–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rietjens, I.; Louisse, J.; Beekmann, K. The potential health effects of dietary phytoestrogens. Br. J. Pharmacol. 2017, 174, 1263–1280. [Google Scholar] [CrossRef] [PubMed]
- Vitale, D.C.; Piazza, C.; Melilli, B.; Drago, F.; Salomone, S. Isoflavones: Estrogenic activity, biological effect and bioavailability. Eur. J. Drug Metab. Pharm. 2013, 38, 15–25. [Google Scholar] [CrossRef]
- Wu, Q.; Yang, Y.; Yu, J.; Jin, N. Soy isoflavone extracts stimulate the growth of nude mouse xenografts bearing estrogen-dependent human breast cancer cells (MCF-7). J. Biomed. Res. 2012, 26, 44–52. [Google Scholar] [CrossRef] [Green Version]
- Hsieh, C.Y.; Santell, R.C.; Haslam, S.Z.; Helferich, W.G. Estrogenic effects of genistein on the growth of estrogen receptor-positive human breast cancer (MCF-7) cells in vitro and in vivo. Cancer Res. 1998, 58, 3833–3838. [Google Scholar]
- Liu, R.; Yu, X.; Chen, X.; Zhong, H.; Liang, C.; Xu, X.; Xu, W.; Cheng, Y.; Wang, W.; Yu, L.; et al. Individual factors define the overall effects of dietary genistein exposure on breast cancer patients. Nutr. Res. 2019, 67, 1–16. [Google Scholar] [CrossRef]
- Wu, A.H.; Yu, M.C.; Tseng, C.C.; Pike, M.C. Epidemiology of soy exposures and breast cancer risk. Br. J. Cancer 2008, 98, 9–14. [Google Scholar] [CrossRef] [Green Version]
- Ahn, S.Y.; Jo, M.S.; Lee, D.; Baek, S.E.; Baek, J.; Yu, J.S.; Jo, J.; Yun, H.; Kang, K.S.; Yoo, J.E.; et al. Dual effects of isoflavonoids from Pueraria lobata roots on estrogenic activity and anti-proliferation of MCF-7 human breast carcinoma cells. Bioorg. Chem. 2019, 83, 135–144. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.J.; Xie, M.Y.; Kluxen, F.M.; Diel, P. Genistein modulates the anti-tumor activity of cisplatin in MCF-7 breast and HT-29 colon cancer cells. Arch. Toxicol. 2014, 88, 625–635. [Google Scholar] [CrossRef] [PubMed]
- Shim, H.Y.; Park, J.H.; Paik, H.D.; Nah, S.Y.; Kim, D.S.; Han, Y.S. Genistein-induced apoptosis of human breast cancer MCF-7 cells involves calpain-caspase and apoptosis signaling kinase 1-p38 mitogen-activated protein kinase activation cascades. Anticancer Drugs 2007, 18, 649–657. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Duan, Y.; Zhang, X.; Ye, Y.; Ge, B.; Chen, J. Genistein induces apoptosis by the inactivation of the IGF-1R/p-Akt signaling pathway in MCF-7 human breast cancer cells. Food Funct. 2015, 6, 995–1000. [Google Scholar] [CrossRef]
- Fan, P.; Fan, S.; Wang, H.; Mao, J.; Shi, Y.; Ibrahim, M.M.; Ma, W.; Yu, X.; Hou, Z.; Wang, B.; et al. Genistein decreases the breast cancer stem-like cell population through Hedgehog pathway. Stem Cell Res. Ther. 2013, 4, 146. [Google Scholar] [CrossRef]
- Liu, X.; Sun, C.; Jin, X.; Li, P.; Ye, F.; Zhao, T.; Gong, L.; Li, Q. Genistein enhances the radiosensitivity of breast cancer cells via G(2)/M cell cycle arrest and apoptosis. Molecules 2013, 18, 13200–13217. [Google Scholar] [CrossRef]
- Dave, B.; Eason, R.R.; Till, S.R.; Geng, Y.; Velarde, M.C.; Badger, T.M.; Simmen, R.C. The soy isoflavone genistein promotes apoptosis in mammary epithelial cells by inducing the tumor suppressor PTEN. Carcinogenesis 2005, 26, 1793–1803. [Google Scholar] [CrossRef]
- Katdare, M.; Osborne, M.; Telang, N.T. Soy isoflavone genistein modulates cell cycle progression and induces apoptosis in HER-2/neu oncogene expressing human breast epithelial cells. Int. J. Oncol. 2002, 21, 809–815. [Google Scholar] [CrossRef]
- Ye, D.; Li, Z.; Wei, C. Genistein inhibits the S-phase kinase-associated protein 2 expression in breast cancer cells. Exp. Ther. Med. 2018, 15, 1069–1075. [Google Scholar] [CrossRef]
- Xue, J.P.; Wang, G.; Zhao, Z.B.; Wang, Q.; Shi, Y. Synergistic cytotoxic effect of genistein and doxorubicin on drug-resistant human breast cancer MCF-7/Adr cells. Oncol. Rep. 2014, 32, 1647–1653. [Google Scholar] [CrossRef] [Green Version]
- Satoh, H.; Nishikawa, K.; Suzuki, K.; Asano, R.; Virgona, N.; Ichikawa, T.; Hagiwara, K.; Yano, T. Genistein, a soy isoflavone, enhances necrotic-like cell death in a breast cancer cell treated with a chemotherapeutic agent. Res. Commun. Mol. Pathol. Pharmacol. 2003, 113–114, 149–158. [Google Scholar]
- Li, Y.; Ahmed, F.; Ali, S.; Philip, P.A.; Kucuk, O.; Sarkar, F.H. Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells. Cancer Res. 2005, 65, 6934–6942. [Google Scholar] [CrossRef] [PubMed]
- Imai, K.; Suga, K.; Nakachi, K. Cancer-preventive effects of drinking green tea among a Japanese population. Prev. Med. 1997, 26, 769–775. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Tse, L.A.; Chan, W.C.; Kwok, C.H.; Leung, S.L.; Wu, C.; Yu, W.C.; Yu, I.T.; Yu, C.H.; Wang, F.; et al. Evaluation of breast cancer risk associated with tea consumption by menopausal and estrogen receptor status among Chinese women in Hong Kong. Cancer Epidemiol. 2016, 40, 73–78. [Google Scholar] [CrossRef] [PubMed]
- Wu, A.H.; Yu, M.C.; Tseng, C.C.; Hankin, J.; Pike, M.C. Green tea and risk of breast cancer in Asian Americans. Int. J. Cancer 2003, 106, 574–579. [Google Scholar] [CrossRef] [PubMed]
- Xiang, L.P.; Wang, A.; Ye, J.H.; Zheng, X.Q.; Polito, C.A.; Lu, J.L.; Li, Q.S.; Liang, Y.R. Suppressive Effects of Tea Catechins on Breast Cancer. Nutrients 2016, 8, 458. [Google Scholar] [CrossRef]
- Zeng, L.; Holly, J.M.; Perks, C.M. Effects of physiological levels of the green tea extract epigallocatechin-3-gallate on breast cancer cells. Front. Endocrinol. 2014, 5, 61. [Google Scholar] [CrossRef]
- Thangapazham, R.L.; Singh, A.K.; Sharma, A.; Warren, J.; Gaddipati, J.P.; Maheshwari, R.K. Green tea polyphenols and its constituent epigallocatechin gallate inhibits proliferation of human breast cancer cells in vitro and in vivo. Cancer Lett. 2007, 245, 232–241. [Google Scholar] [CrossRef]
- Deguchi, H.; Fujii, T.; Nakagawa, S.; Koga, T.; Shirouzu, K. Analysis of cell growth inhibitory effects of catechin through MAPK in human breast cancer cell line T47D. Int. J. Oncol. 2002, 21, 1301–1305. [Google Scholar] [CrossRef]
- 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, 924–928. [Google Scholar] [CrossRef]
- Roy, A.M.; Baliga, M.S.; Katiyar, S.K. Epigallocatechin-3-gallate induces apoptosis in estrogen receptor-negative human breast carcinoma cells via modulation in protein expression of p53 and Bax and caspase-3 activation. Mol. Cancer Ther. 2005, 4, 81–90. [Google Scholar] [PubMed]
- Tang, Y.; Zhao, D.Y.; Elliott, S.; Zhao, W.; Curiel, T.J.; Beckman, B.S.; Burow, M.E. Epigallocatechin-3 gallate induces growth inhibition and apoptosis in human breast cancer cells through survivin suppression. Int. J. Oncol. 2007, 31, 705–711. [Google Scholar] [CrossRef] [PubMed]
- Baliga, M.S.; Meleth, S.; Katiyar, S.K. Growth inhibitory and antimetastatic effect of green tea polyphenols on metastasis-specific mouse mammary carcinoma 4T1 cells in vitro and in vivo systems. Clin. Cancer Res. 2005, 11, 1918–1927. [Google Scholar] [CrossRef] [PubMed]
- Hsuuw, Y.D.; Chan, W.H. Epigallocatechin gallate dose-dependently induces apoptosis or necrosis in human MCF-7 cells. Ann. N. Y. Acad. Sci. 2007, 1095, 428–440. [Google Scholar] [CrossRef] [PubMed]
- Hsu, Y.C.; Liou, Y.M. The anti-cancer effects of (-)-epigallocatechin-3-gallate on the signaling pathways associated with membrane receptors in MCF-7 cells. J. Cell. Physiol. 2011, 226, 2721–2730. [Google Scholar] [CrossRef]
- Mittal, A.; Pate, M.S.; Wylie, R.C.; Tollefsbol, T.O.; Katiyar, S.K. EGCG down-regulates telomerase in human breast carcinoma MCF-7 cells, leading to suppression of cell viability and induction of apoptosis. Int. J. Oncol. 2004, 24, 703–710. [Google Scholar] [CrossRef]
- 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, 374–382. [Google Scholar] [CrossRef] [Green Version]
- Radhakrishnan, R.; Pooja, D.; Kulhari, H.; Gudem, S.; Ravuri, H.G.; Bhargava, S.; Ramakrishna, S. Bombesin conjugated solid lipid nanoparticles for improved delivery of epigallocatechin gallate for breast cancer treatment. Chem. Phys. Lipids 2019, 224, 104770. [Google Scholar] [CrossRef]
- Hajipour, H.; Hamishehkar, H.; Nazari Soltan Ahmad, S.; Barghi, S.; Maroufi, N.F.; Taheri, R.A. Improved anticancer effects of epigallocatechin gallate using RGD-containing nanostructured lipid carriers. Artif. Cells Nanomed. Biotechnol. 2018, 46, 283–292. [Google Scholar] [CrossRef] [Green Version]
- Zeng, L.; Yan, J.; Luo, L.; Ma, M.; Zhu, H. Preparation and characterization of (-)-Epigallocatechin-3-gallate (EGCG)-loaded nanoparticles and their inhibitory effects on Human breast cancer MCF-7 cells. Sci. Rep. 2017, 7, 45521. [Google Scholar] [CrossRef]
- Saklani, A.; Kutty, S.K. Plant-derived compounds in clinical trials. Drug Discov. Today 2008, 13, 161–171. [Google Scholar] [CrossRef] [PubMed]
- Chengappa, K.N.R.; Brar, J.S.; Gannon, J.M.; Schlicht, P.J. Adjunctive Use of a Standardized Extract of Withania somnifera (Ashwagandha) to Treat Symptom Exacerbation in Schizophrenia: A Randomized, Double-Blind, Placebo-Controlled Study. J. Clin. Psychiatry 2018, 79. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, A.V.; Martinez, M.; Stamos, M.J.; Moyer, M.P.; Planutis, K.; Hope, C.; Holcombe, R.F. Results of a phase I pilot clinical trial examining the effect of plant-derived resveratrol and grape powder on Wnt pathway target gene expression in colonic mucosa and colon cancer. Cancer Manag. Res. 2009, 1, 25–37. [Google Scholar] [PubMed]
- Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef] [PubMed]
- Kris-Etherton, P.M.; Hecker, K.D.; Bonanome, A.; Coval, S.M.; Binkoski, A.E.; Hilpert, K.F.; Griel, A.E.; Etherton, T.D. Bioactive compounds in foods: Their role in the prevention of cardiovascular disease and cancer. Am. J. Med. 2002, 113 (Suppl. 9B), 71S–88S. [Google Scholar] [CrossRef]
- Liu, R.H. Dietary bioactive compounds and their health implications. J. Food Sci. 2013, 78 (Suppl. 1), A18–A25. [Google Scholar] [CrossRef]
Bioactive Compound | Clinical Trials | Phase | ClinicalTrials.Gov Identifier |
---|---|---|---|
Withania somnifera (Sensoril or Ashwagandha) | A randomized study conducted for schizophrenia | Interventional | NCT01793935 |
Curcumin and Ashwagandha extract | To study the safety and efficacy of Curcumin and Ashwagandha extract in osteosarcoma. | Phase 1 and Phase 2 | NCT00689195 |
WFA | To evaluate the cognitive abilities in persons with bipolar disorder and to study the residual mood/anxiety symptoms | Phase 3 | NCFT00761761 |
WFA | To study the safety and efficacy of the drug in patients having generalized anxiety disorder | Phase 2 | NCT01311180 |
Resveratrol | Colon cancer patients were randomly given resveratrol at different concentration to study Wnt signaling pathway. | Phase 1 | NCT00256334 |
Resveratrol | To study the effect of Notch signaling in neuroendocrine tumor treated with Resveratrol. | Interventional | NCT01476592 |
Resveratrol | To study the effect of resveratrol in colorectal cancer and also studied for tolerability, target tissue levels and pharmacodynamics. | Phase 1 | NCT00433576 |
Resveratrol | A randomized trial to determine the disposition and characterize dietary polyphenols in normal and breast cancer patient. | Interventional | NCT03482401 |
Resveratrol | A randomized double-blind study, resveratrol was given for two weeks in non-diabetic obese subjects. | Phase 2 | NCT02247596 |
Isothiocyanate | A randomized clinical trial, to study phenethyl isothiocyanate in lung cancer patients. | Phase 2 | NCT00691132 |
Isothiocyanate | A randomized phase II study, patients received broccoli sprout extract through oral three times daily for 8 weeks to breast cancer patients, ductal carcinoma in situ and/or atypical ductal hyperplasia | Phase 2 | NCT00843167 PMID:26511489 PMID:26329135 |
Curcumin | A randomized, double-blind, placebo-controlled study of curcumin for the prevention of acute radiation-induced dermatitis during postoperative radiotherapy for breast cancer. | Phase 2 | NCT01042938 |
Curcumin | A randomized A randomized study treating docetaxel alone or together with curcumin in HER2 negative patients with metastatic breast cancer | Phase 2 | NCT00852332 |
Genistein | A randomized study of genistein treatment in high risk of breast cancer patients. | Phase 2 | NCT00290758 PMID: 22307566 |
Genistein | A randomized, phase 1 trial of genistein in preventing breast or endometrial cancer | Phase 1 | NCT00099008 PMID:18446090 |
EGCG | A randomized phase 1 study, to find the effect of catechin extract in hormone negative stage I-III breast cancer patients | Phase 1 | NCT00516243 |
EGCG | A phase 2 trial, to study the effect of green tea extract in breast cancer | Phase 2 | NCT00917735 PMID 30926986, 28747487, 27806972, 26701796, 26581683 |
Bioactive Compounds | Models | Specific Mechanism of Action | References |
---|---|---|---|
Withaferin A (WFA) | Human breast cancer, liver cancer | ↑ PARP ↑expression of Bim-S and Bim EL ↓expression of Bcl2 ↑ generation of ROS ↑Bax and Bak ↓expression of XIAP G2/M arrest, ↓ PCNA and ↑apoptosis | [22,27,28,33,37] |
Honokiol (HNK) | Human breast cancer | ↑G2/M arrest ↑ AMPK | [52,54] |
Benzyl Isothiocyanate (BITC) | Human breast cancer | ↑ROS ↑Increased caspase 3 ↓mTOR ↓XIAP | [87,88,90,94] |
Resveratrol | Human breast cancer | ↓Bcl-2, ↓Bcl-XL, ↑Bax ↑Increased caspase 3 | [117,118] |
Curcumin | Human breast cancer | ↓Bcl-2, ↑Bax, ↑Caspase 3, ↑AMPK, ↓AKT ↓pAKT/↓mTOR ↑PARP ↑Caspase3 and ↑caspase 7 | [139,143,145,151] |
Genistein | Human breast cancer | ↑Caspase7, p38MAPK ↓Bcl2 ↑ATM/Chk2/Cdc25C/Cdc2 | [173,174,176] |
Epigallocatechin-3-gallate (EGCG) | Human breast cancer | ↓Cdc ↑Bax, Bcl2, ↑ADP ribose, ↑Caspase 3 ↑hTERT | [189,190,191,196] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Muniraj, N.; Siddharth, S.; Sharma, D. Bioactive Compounds: Multi-Targeting Silver Bullets for Preventing and Treating Breast Cancer. Cancers 2019, 11, 1563. https://doi.org/10.3390/cancers11101563
Muniraj N, Siddharth S, Sharma D. Bioactive Compounds: Multi-Targeting Silver Bullets for Preventing and Treating Breast Cancer. Cancers. 2019; 11(10):1563. https://doi.org/10.3390/cancers11101563
Chicago/Turabian StyleMuniraj, Nethaji, Sumit Siddharth, and Dipali Sharma. 2019. "Bioactive Compounds: Multi-Targeting Silver Bullets for Preventing and Treating Breast Cancer" Cancers 11, no. 10: 1563. https://doi.org/10.3390/cancers11101563
APA StyleMuniraj, N., Siddharth, S., & Sharma, D. (2019). Bioactive Compounds: Multi-Targeting Silver Bullets for Preventing and Treating Breast Cancer. Cancers, 11(10), 1563. https://doi.org/10.3390/cancers11101563