Natural and herbal compounds targeting breast cancer, a review based on cancer stem cells

Document Type : Review Article

Authors

1 Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran

2 Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran

Abstract

Cancer stem cells (CSCs) are known as the major reason for therapy resistance. Recently, natural herbal compounds are suggested to have a significant role in inhibiting the breast cancer stem cells (BCSCs). The aim of this study was to explore the effective natural herbal compounds against BCSCs.This review article was designed based on the BCSCs, mechanisms of therapy resistance and natural herbal compounds effective to inhibit their activity. Therefore, Science direct, PubMed and Scopus databases were explored and related original articles were investigated from 2010 to 2019. BCSCs use different mechanisms including special membrane transporters, anti-apoptotic, pro-survival, and self-renewal- related signaling pathways. Natural herbal compounds could disturb these mechanisms, therefore may inhibit or eradicate the BCSCs. Studies show that a broad range of plants, either as a food or medicine, contain anti-cancer agents that phenolic components and their different derivatives share a large quantity. Natural herbal compounds play a pivotal role in the eradication of BCSCs, through the inhibition of biological activities and induction of apoptosis. Although it is necessary to conduct more clinical investigation.

Keywords


1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet‐Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87-108.
2. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144:1941-1953.
3. Siegel R MJ, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9-29.
4. Group EBCTC. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. The Lancet. 2005;365:1687-1717.
5. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, et al. Cancer stem cells—perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006;66:9339-9344.
6. Rasul A, Al-Shawi AA, Malik SA, Anwar H, Rasool B, Razzaq A, et al. Neurada procumbens promotes functions regain in a mouse model of mechanically induced sciatic nerve injury. Pak J Pharm Sci. 2019;32:1761-1766.
7. Rezakhani L, Rashidi Z, Mirzapur P, Khazaei M. Antiproliferatory effects of crab shell extract on breast cancer cell line MCF7. J Breast Cancer. 2014;17:219-225.
8. Liu B, Cheng Y, Liu Q, Bao J-k, Yang J-M. Autophagic pathways as new targets for cancer drug development. Acta Pharm Sinic. 2010;31:1154-1164.
9. Khazaei M, Bozorgi A, Khazaei S, Khademi A. Stem cells in dentistry, sources, and applications. Dent Hypotheses. 2016;7:42-52.
10. Bozorgi A, Khazaei M, Khazaei MR. New findings on breast cancer stem cells: a review. J Breast Cancer. 2015;18:303-312.
11. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.
12. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proceed Natl Acad Sci. 2003;100:3983-3988.
13. Shafee N, Smith CR, Wei S, Kim Y, Mills GB, Hortobagyi GN, et al. Cancer stem cells contribute to cisplatin resistance in Brca1/p53–mediated mouse mammary tumors. Cancer Res. 2008;68:3243-3250.
14. Lee HE, Kim JH, Kim YJ, Choi S, Kim S, Kang E, et al. An increase in cancer stem cell population after primary systemic therapy is a poor prognostic factor in breast cancer. Br J Cancer. 2011;104:1730-1738.
15. Zielske SP, Spalding AC, Wicha MS, Lawrence TS. Ablation of breast cancer stem cells with radiation. Transl Oncol. 2011;4:227-233.
16. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu M-F, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer I. 2008;100:672-679.
17. Sotiropoulou PA, Christodoulou MS, Silvani A, Herold-Mende C, Passarella D. Chemical approaches to targeting drug resistance in cancer stem cells. Drug Discov today. 2014;19:1547-1562.
18. Piggott L, Omidvar N, Pérez SM, Eberl M, Clarkson RW. Suppression of apoptosis inhibitor c-FLIP selectively eliminates breast cancer stem cell activity in response to the anti-cancer agent, TRAIL. Breast Cancer Res. 2011;13:1-15.
19. Yin S, Xu L, Bandyopadhyay S, Sethi S, Reddy KB. Cisplatin and TRAIL enhance breast cancer stem cell death. Int J Oncol. 2011;39:891-898.
20. Korkaya H, Kim G-i, Davis A, Malik F, Henry NL, Ithimakin S, et al. Activation of an IL6 inflammatory loop mediates trastuzumab resistance in HER2+ breast cancer by expanding the cancer stem cell population. Mol Cell. 2012;47:570-584.
21. Wardman P. Chemical radiosensitizers for use in radiotherapy. Clin Oncol. 2007;19:397-417.
22. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, et al. Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell. 2010;140:62-73.
23. Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci. 2012;33:207-214.
24. Muñoz P, Iliou MS, Esteller M. Epigenetic alterations involved in cancer stem cell reprogramming. Mol Oncol. 2012;6:620-636.
25. Vasiliou V, Vasiliou K, Nebert DW. Human ATP-binding cassette (ABC) transporter family. Hum Genomics. 2009;3:281-290.
26. Farnie G, Clarke RB. Mammary stem cells and breast cancer-role of notch signalling. Stem Cell Rev. 2007;3:169-175.
27. Aggarwal BB, Sethi G, Baladandayuthapani V, Krishnan S, Shishodia S. Targeting cell signaling pathways for drug discovery: an old lock needs a new key. J Cell Biochem. 2007;102:580-592.
28. Niida H, Nakanishi M. DNA damage checkpoints in mammals. Mutagenesis. 2006;21:3-9.
29. Selamoglu Z. Biotechnological approaches on anticancer activity of flavonoids-Mini review. Modern Approaches to Drug Design. 2017;1:1.
30. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458:780-783.
31. Weng C-J, Yen G-C. Chemopreventive effects of dietary phytochemicals against cancer invasion and metastasis: phenolic acids, monophenol, polyphenol, and their derivatives. Cancer Treat Rev. 2012;38:76-87.
32. Riaz A, Rasul A, Hussain G, Zahoor MK, Jabeen F, Subhani Z, et al. Astragalin: A bioactive phytochemical with potential therapeutic activities. Adv Phar Sc. 2018;2018:1-15.
33. Miean KH, Mohamed S. Flavonoid (myricetin, quercetin, kaempferol, luteolin, and apigenin) content of edible tropical plants. J Agr Food Chem. 2001;49:3106-112.
34. Ninfali P, Angelino D. Nutritional and functional potential of Beta Vulgaris cicla and rubra. Fitoterapia. 2013;89:188-199.
35. Đorđević S, Cakić M, Amr S. The extraction of apigenin and luteolin from the sage Salvia officinalis L. from Jordan. Facta universitatis-series: Working and Living Enviromental Protection. 2000;1:87-93.
36. Afzal M, Aloriquat G, Alhassan J, Muhammad N. Flavone glycosides from lawsonia innermis. Heterocycles. 1980;14:1973-1976.
37. Khalid H, Abdalla WE, Abdelgadir H, Opatz T, Efferth T. Gems from traditional north-African medicine: Medicinal and aromatic plants from Sudan. Nat Prod Bioprospect. 2012;2:92-103.
38. Kumar S, Sharma A. Apigenin: The anxiolytic constituent of turnera aphrodisiaca. Pharm. 2006;44:84-90.
39. Patel D, Shukla S, Gupta S. Apigenin and cancer chemoprevention: progress, potential and promise. Int J Oncol. 2007;30:233-246.
40. Middleton E, Kandaswami C, Theoharides TC. The effects of plant flavonoids on mammalian cells: Implications for inflammation, heart disease, and cancer. Pharmacol Rev. 2000;52:673-751.
41. Saeed M, Kadioglu O, Khalid H, Sugimoto Y, Efferth T. Activity of the dietary flavonoid, apigenin, against multidrug-resistant tumor cells as determined by pharmacogenomics and molecular docking. J Nutr Biochem. 2015;26:44-56.
42. Bauer D, Redmon N, Mazzio E, Soliman KF. Apigenin inhibits TNFα/IL-1α-induced CCL2 release through IKBK-epsilon signaling in MDA-MB-231 human breast cancer cells. PloS one. 2017;12:1-17.
43. Erdogan S, Doganlar O, Doganlar ZB, Serttas R, Turkekul K, Dibirdik I, et al. The flavonoid apigenin reduces prostate cancer CD44+ stem cell survival and migration through PI3K/Akt/NF-κB signaling. Life Sci. 2016;162:77-86.
44. Huang C, Wei Y-X, Shen M-C, Tu Y-H, Wang C-C, Huang H-C. Chrysin, abundant in morinda citrifolia fruit water–etoac extracts, combined with apigenin synergistically induced apoptosis and inhibited migration in human breast and liver cancer cells. J Agr Food Chem. 2016;64:4235-4245.
45. Seo HS, Ku JM, Choi HS, Woo JK, Lee BH, Kim DS, et al. Apigenin overcomes drug resistance by blocking the signal transducer and activator of transcription 3 signaling in breast cancer cells. Oncol Rep. 2017;38:715-724.
46. Seo H-S, Jo JK, Ku JM, Choi H-S, Choi YK, Woo J-K, et al. Induction of caspase-dependent extrinsic apoptosis by apigenin through inhibition of signal transducer and activator of transcription 3 (STAT3) signalling in HER2-overexpressing BT-474 breast cancer cells. Biosci Rep. 2015;35:1-14.
47. Li YW, Xu J, Zhu GY, Huang ZJ, Lu Y, Li XQ, et al. Apigenin suppresses the stem cell-like properties of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Cell Death Discov. 2018;4:1-9.
48. Sun D, Abraham SN, Beachey EH. Influence of berberine sulfate on synthesis and expression of pap fimbrial adhesin in uropathogenic escherichia coli. Antimicrob Agents Chemother. 1988;32:1274-1277.
49. Hahn FE, Ciak J. Berberine.  Mechanism of action of antimicrobial and antitumor agents: Springer; 1975. p. 577-584.
50. Katiyar SK, Meeran SM, Katiyar N, Akhtar S. p53 cooperates berberine‐induced growth inhibition and apoptosis of non‐small cell human lung cancer cells in vitro and tumor xenograft growth in vivo. Mol Carcinog. 2009;48:24-37.
51. Liu Z, Liu Q, Xu B, Wu J, Guo C, Zhu F, et al. Berberine induces p53-dependent cell cycle arrest and apoptosis of human osteosarcoma cells by inflicting DNA damage. Mutat Res/Fund and Mol M. 2009;662:75-83.
52. Ma X, Zhou J, Zhang C-X, Li X-Y, Li N, Ju R-J, et al. Modulation of drug-resistant membrane and apoptosis proteins of breast cancer stem cells by targeting berberine liposomes. Biomaterials. 2013;34:4452-4465.
53. Zhao Y, Jing Z, Lv J, Zhang Z, Lin J, Cao X, et al. Berberine activates caspase-9/cytochrome c-mediated apoptosis to suppress triple-negative breast cancer cells in vitro and in vivo. Biomed Pharmacother. 2017;95:18-24.
54. Ma W, Zhu M, Zhang D, Yang L, Yang T, Li X, et al. Berberine inhibits the proliferation and migration of breast cancer ZR-75-30 cells by targeting Ephrin-B2. Phytomedicine. 2017;25:45-51.
55. Ahmadiankia N, Moghaddam HK, Mishan MA, Bahrami AR, Naderi-Meshkin H, Bidkhori HR, et al. Berberine suppresses migration of MCF-7 breast cancer cells through down-regulation of chemokine receptors. Iran J Basic Med Sci. 2016;19:125-131.
56. Zhao Y, Jing Z, Li Y, Mao W. Berberine in combination with cisplatin suppresses breast cancer cell growth through induction of DNA breaks and caspase-3-dependent apoptosis. Oncol Rep. 2016;36:567-572.
57. Park CH, Hahm ER, Park S, Kim H-K, Yang CH. The inhibitory mechanism of curcumin and its derivative against β‐catenin/Tcf signaling. FEBS lett. 2005;579:2965-2971.
58. Satoskar R, Shah S, Shenoy S. Evaluation of anti-inflammatory property of curcumin (diferuloyl methane) in patients with postoperative inflammation. Int J Clin Pharmacol Ther  Toxicol. 1986;24:651.
59. Bimonte S, Barbieri A, Palma G, Rea D, Luciano A, D’Aiuto M, et al. Dissecting the role of curcumin in tumour growth and angiogenesis in mouse model of human breast cancer. BioMed Res Int. 2015;2015:1-7.
60. Li X, Wang X, Xie C, Zhu J, Meng Y, Chen Y, et al. Sonic hedgehog and Wnt/beta-catenin pathways mediate curcumin inhibition of breast cancer stem cells. Anti-cancer drugs. 2018;29:208-215.
61. Kakarala M, Brenner DE, Korkaya H, Cheng C, Tazi K, Ginestier C, et al. Targeting breast stem cells with the cancer preventive compounds curcumin and piperine. Breast Cancer Res Treat. 2010;122:777-785.
62. Chung SS, Vadgama JV. Curcumin and epigallocatechin gallate inhibit the cancer stem cell phenotype via down-regulation of STAT3–NFκB signaling. Anticancer Res. 2015;35:39-46.
63. Wei W, Rasul A, Sadiqa A, Sarfraz I, Hussain G, Nageen B, et al. Curcumol: from plant roots to cancer roots. Int J Biol Sci. 2019;15:1600-1609.
64. Zhou Q, Ye M, Lu Y, Zhang H, Chen Q, Huang S, et al. Curcumin improves the tumoricidal effect of mitomycin C by suppressing ABCG2 expression in stem cell-like breast cancer cells. PloS one. 2015;10:1-12.
65. Toden S, Okugawa Y, Jascur T, Wodarz D, Komarova NL, Buhrmann C, et al. Curcumin mediates chemosensitization to 5-fluorouracil through miRNA-induced suppression of epithelial-to-mesenchymal transition in chemoresistant colorectal cancer. Carcinogenesis. 2015;36:355-367.
66. Hu C, Li M, Guo T, Wang S, Huang W, Yang K, et al. Anti-metastasis activity of curcumin against breast cancer via the inhibition of stem cell-like properties and EMT. Phytomedicine. 2019;58:1-32.
67. Hashemzehi M, Behnam-Rassouli R, Hassanian SM, Moradi-Binabaj M, Moradi-Marjaneh R, Rahmani F, et al. Phytosomal-curcumin antagonizes cell growth and migration, induced by thrombin through AMP-kinase in breast cancer. J Cell Biochem. 2018;119:5996-6007.
68. Yuan JD, ZhuGe DL, Tong MQ, Lin MT, Xu XF, Tang X, et al. pH-sensitive polymeric nanoparticles of mPEG-PLGA-PGlu with hybrid core for simultaneous encapsulation of curcumin and doxorubicin to kill the heterogeneous tumour cells in breast cancer. Artif Cells Nanomed Biotechnol. 2018;46:302-313.
69. Dai Z, Nair V, Khan M, Ciolino HP. Pomegranate extract inhibits the proliferation and viability of MMTV-Wnt-1 mouse mammary cancer stem cells in vitro. Oncology Rep. 2010;24:1087-1091.
70. Chen H-S, Bai M-H, Zhang T, Li G-D, Liu M. Ellagic acid induces cell cycle arrest and apoptosis through TGF-β/Smad3 signaling pathway in human breast cancer MCF-7 cells. Int J Oncol. 2015;46:1730-1738.
71. Ahire V, Kumar A, Mishra KP, Kulkarni G. Ellagic acid enhances apoptotic sensitivity of breast cancer cells to γ-radiation. Nutr Cancer. 2017;69:904-910.
72. Cook MT, Liang Y, Besch-Williford C, Goyette S, Mafuvadze B, Hyder SM. Luteolin inhibits progestin-dependent angiogenesis, stem cell-like characteristics, and growth of human breast cancer xenografts. Springerplus. 2015;4:1-16.
73. Wang Z, Wang D, Han S, Wang N, Mo F, Loo TY, et al. Bioactivity-guided identification and cell signaling technology to delineate the lactate dehydrogenase A inhibition effects of spatholobus suberectus on breast cancer. PLoS One. 2013;8:1-12.
74. Chen D, Pamu S, Cui Q, Chan TH, Dou QP. Novel epigallocatechin gallate (EGCG) analogs activate AMP-activated protein kinase pathway and target cancer stem cells. Bioorg Med Chem. 2012;20:3031-3037.
75. Pan X, Zhao B, Song Z, Han S, Wang M. Estrogen receptor-alpha36 is involved in epigallocatechin-3-gallate induced growth inhibition of ER-negative breast cancer stem/progenitor cells. J Pharmacol Sci. 2016;130:85-93.
76. Farabegoli F, Govoni M, Spisni E, Papi A. EGFR inhibition by epigallocatechin-3-gallate and IIF treatments reduces breast cancer cell invasion. Biosci Rep. 2017;37:1-14.
77. El-Rahman SSA, Shehab G, Nashaat H. Epigallocatechin-3-gallate: the prospective targeting of cscs and preventing metastasis of chemically-induced mammary cancer in rats. Am J Med Sci. 2017;354:54-63.
78. Giro-Perafita A, Rabionet M, Planas M, Feliu L, Ciurana J, Ruiz-Martinez S, et al. EGCG-derivative G28 Shows high efficacy inhibiting the mammosphere-forming capacity of sensitive and resistant TNBC models. Molecules. 2019;24;1-15.
79. Jiang D, Rasul A, Batool R, Sarfraz I, Hussain G, Mateen Tahir M, et al. Potential anticancer properties and mechanisms of action of formononetin. Biomed Res Int. 2019;2019:1-11.
80. Montales MTE, Rahal OM, Kang J, Rogers T, Prior RL, Wu X, et al. Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stem-like/progenitor cells. Carcinogenesis. 2012;33:.652-660
81. Montales MTE, Rahal OM, Nakatani H, Matsuda T, Simmen RC. Repression of mammary adipogenesis by genistein limits mammosphere formation of human MCF-7 cells. J Endocrinol. 2013;218:135-149.
82. Liu Y, Zou T, Wang S, Chen H, Su D, Fu X, et al. Genistein-induced differentiation of breast cancer stem/progenitor cells through a paracrine mechanism. Int J Oncol. 2016;48:1063-1072.
83. Rigalli J, Tocchetti G, Arana M, Villanueva S, Catania V, Theile D, et al. The phytoestrogen genistein enhances multidrug resistance in breast cancer cell lines by translational regulation of ABC transporters. Cancer lett. 2016;376:165-172.
84. Fan P, Fan S, Wang H, Mao J, Shi Y, Ibrahim MM, et al. Genistein decreases the breast cancer stem-like cell population through hedgehog pathway. Stem Cell Res Ther. 2013;4:1-10.
85. Paul B, Li Y, Tollefsbol TO. The effects of combinatorial genistein and sulforaphane in breast tumor inhibition: role in epigenetic regulation. Int J mole sci. 2018;19;1-22.
86. Wu C-H, Hong B-H, Ho C-T, Yen G-C. Targeting cancer stem cells in breast cancer: potential anticancer properties of 6-shogaol and pterostilbene. J Agr Food Chem. 2015;63:2432-2441.
87. Lee HS, Seo EY, Kang NE, Kim WK. [6]-Gingerol inhibits metastasis of MDA-MB-231 human breast cancer cells. J Nutr Biochem. 2008;19:313-319.
88. Wang Z, Zhang X, Wang H, Qi L, Lou Y. Neuroprotective effects of icaritin against beta amyloid-induced neurotoxicity in primary cultured rat neuronal cells via estrogen-dependent pathway. Neuroscience. 2007;145:911-922.
89. Wo Yb, Zhu Dy, Hu Y, Wang ZQ, Liu J, Lou YJ. Reactive oxygen species involved in prenylflavonoids, icariin and icaritin, initiating cardiac differentiation of mouse embryonic stem cells. J Cell Biochem. 2008;103:1536-1550.
90. Zhang G, Qin L, Sheng H, Wang X-L, Wang Y-X, Yeung DK-W, et al. A novel semisynthesized small molecule icaritin reduces incidence of steroid-associated osteonecrosis with inhibition of both thrombosis and lipid-deposition in a dose-dependent manner. Bone. 2009;44:345-356.
91. Huang J, Yuan L, Wang X, Zhang T-L, Wang K. Icaritin and its glycosides enhance osteoblastic, but suppress osteoclastic, differentiation and activity in vitro. Life Sci. 2007;81:832-840.
92. Huang X, Zhu D, Lou Y. A novel anticancer agent, icaritin, induced cell growth inhibition, G 1 arrest and mitochondrial transmembrane potential drop in human prostate carcinoma PC-3 cells. Eur J Pharmacol. 2007;564:26-36.
93. Li C, Peng W, Song X, Wang Q, Wang W. Anticancer effect of icaritin inhibits cell growth of colon cancer through reactive oxygen species, Bcl‑2 and cyclin D1/E signaling. Oncol Lett. 2016;12:3537-3542.
94. Guo Y, Zhang X, Meng J, Wang Z-Y. An anticancer agent icaritin induces sustained activation of the extracellular signal-regulated kinase (ERK) pathway and inhibits growth of breast cancer cells. Eur J Pharmacol. 2011;658:114-122.
95. Wang X, Zheng N, Dong J, Wang X, Liu L, Huang J. Estrogen receptor-α36 is involved in icaritin induced growth inhibition of triple-negative breast cancer cells. J Steroid Biochem Mol Biol. 2017;171:318-327.
96. Kang T, Seo J, Oh H, Yoon G, Chae J, Shim J. Licochalcone A suppresses specificity protein 1 as a novel target in human breast cancer cells. J Cell Biochem. 2017;118:4652-4663.
97. Beishline K, Azizkhan-Clifford J. Sp1 and the hallmarks of cancer. FEBS J. 2015;282:224-258.
98. Kwon SJ, Park SY, Kwon GT, Lee KW, Kang Y-H, Choi M-S, et al. Licochalcone E present in licorice suppresses lung metastasis in the 4T1 mammary orthotopic cancer model. Cancer Prev Res. 2013;6:603-613.
99. Empey D, Laitinen L, Young G, Bye C, Hughes D. Comparison of the antitussive effects of codeine phosphate 20 mg, dextromethorphan 30 mg and noscapine 30 mg using citric acid-induced cough in normal subjects. Eur J Clin Pharmacol. 1979;16:393-397.
100. DeBono A, Capuano B, Scammells PJ. Progress toward the development of noscapine and derivatives as anticancer agents. J Med Chem. 2015;58:5699-5727.
101. Sajadian S, Vatankhah M, Majdzadeh M, Kouhsari SM, Ghahremani MH, Ostad SN. Cell cycle arrest and apoptogenic properties of opium alkaloids noscapine and papaverine on breast cancer stem cells. Toxicol Mech Methods. 2015;25:388-395.
102. Doddapaneni R, Patel K, Chowdhury N, Sachdeva M. Noscapine chemosensitization enhances docetaxel anticancer activity and tumor stroma disruption against triple negative breast cancer. AACR; 2016;346:65-73.
103. Quisbert-Valenzuela EO, Calaf GM. Apoptotic effect of noscapine in breast cancer cell lines. International J Oncol. 2016;48:2666-2674.
104. Mahaddalkar T, Manchukonda N, Choudhary S, Cheriyamundath S, Mohanpuria N, Kantevari S, et al. Subtle alterations in microtubule assembly dynamics by Br‐TMB‐noscapine strongly suppress Triple‐negative breast cancer cell viability without mitotic arrest. ChemistrySelect. 2016;1:4313-4319.
105. Muthiah D, Henshaw GK, DeBono AJ, Capuano B, Scammells PJ, Callaghan R. Overcoming p-glycoprotein-mediated drug resistance with noscapine derivatives. Drug Metab Dispos. 2019;47:164-172.
106. Lu LG, Zeng MD, Mao YM, Fang JY, Song YL, Shen ZH, et al. Inhibitory effect of oxymatrine on serum hepatitis B virus DNA in HBV transgenic mice. World J gastroenterol. 2004;10:1176-1179.
107. Zheng P, Niu FL, Liu WZ, Shi Y, Lu LG. Anti-inflammatory mechanism of oxymatrine in dextran sulfate sodium-induced colitis of rats. World J Gastroenterol. 2005;11:4912-4915.
108. Zhang M, Huang J. Recent research progress of anti-tumor mechnism matrine. Zhongguo Zhong Yao Za Zhi. 2004;29:115-118.
109. Jin Y, Hu J, Wang Q, Li Z, Chen Y. Effects of oxymatrine on the apoptosis of human esophageal carcinoma Eca109 cell line and its mechanism. J Huazhong U Sci Med. 2008;28:314-316.
110. Zhang Y, Piao B, Zhang Y, Hua B, Hou W, Xu W, et al. Oxymatrine diminishes the side population and inhibits the expression of β-catenin in MCF-7 breast cancer cells. Med Oncol. 2011;28:99-107.
111. Xie W, Zhang Y, Zhang S, Wang F, Zhang K, Huang Y, et al. Oxymatrine enhanced anti-tumor effects of bevacizumab against triple-negative breast cancer via abating Wnt/beta-catenin signaling pathway. Am J Cancer Res. 2019;9:1796-1814.
112. Chen Y, Chen L, Zhang JY, Chen ZY, Liu TT, Zhang YY, et al. Oxymatrine reverses epithelial-mesenchymal transition in breast cancer cells by depressing alphabeta3 integrin/FAK/PI3K/Akt signaling activation. Onco Targets Ther. 2019;12:6253-6265.
113. Wu J, Cai Y, Li M, Zhang Y, Li H, Tan Z. Oxymatrine promotes S-phase arrest and inhibits cell proliferation of human breast cancer cells in vitro through mitochondria-mediated apoptosis. Biol Pharm Bull. 2017;40:1232-1239.
114. Katiyar SS, Muntimadugu E, Rafeeqi TA, Domb AJ, Khan W. Co-delivery of rapamycin-and piperine-loaded polymeric nanoparticles for breast cancer treatment. Drug Deliv. 2016;23:2608-2616.
115. Talib WH. Regressions of breast carcinoma syngraft following treatment with piperine in combination with thymoquinone. Sci Pharm. 2017;85:1-11.
116. Lai LH, Fu QH, Liu Y, Jiang K, Guo QM, Chen QY, et al. Piperine suppresses tumor growth and metastasis in vitro and in vivo in a 4T1 murine breast cancer model. Acta Pharmacol Sin. 2012;33:523-530.
117. Do MT, Kim HG, Choi JH, Khanal T, Park BH, Tran TP, et al. Antitumor efficacy of piperine in the treatment of human HER2-overexpressing breast cancer cells. Food Chem. 2013;141:2591-2599.
118. Jain S, Meka SRK, Chatterjee K. Engineering a piperine eluting nanofibrous patch for cancer treatment. ACS Biomater Sci Eng. 2016;2:1376-1385.
119. Greenshields AL, Doucette CD, Sutton KM, Madera L, Annan H, Yaffe PB, et al. Piperine inhibits the growth and motility of triple-negative breast cancer cells. Cancer Lett. 2015;357:129-140.
120. Abdelhamed S, Yokoyama S, Refaat A, Ogura K, Yagita H, Awale S, et al. Piperine enhances the efficacy of TRAIL-based therapy for triple-negative breast cancer cells. Anticancer Res. 2014;34:1893-1899.
121. Hagiwara K, Kosaka N, Yoshioka Y, Takahashi RU, Takeshita F, Ochiya T. Stilbene derivatives promote Ago2-dependent tumour-suppressive microRNA activity. Sci Rep. 2012;2:1-9.
122. Mak KK, Wu AT, Lee WH, Chang TC, Chiou JF, Wang LS, et al. Pterostilbene, a bioactive component of blueberries, suppresses the generation of breast cancer stem cells within tumor microenvironment and metastasis via modulating NF‐κB/microRNA 448 circuit. Mol Nutr Food Res. 2013;57:1123-1134.
123. Ignatowicz E, Baer-Dubowska W. Resveratrol, a natural chemopreventive agent against degenerative diseases. Pol J Pharmacol. 2001;53:557-569.
124. Muqbil I, WJ Beck F, Bao B, H Sarkar F, M Mohammad R, M Hadi S, et al. Old wine in a new bottle: the Warburg effect and anticancer mechanisms of resveratrol. Curr Pharm Des. 2012;18:1645-1654.
125. Venkatadri R, Muni T, Iyer AKV, Yakisich JS, Azad N. Role of apoptosis-related miRNAs in resveratrol-induced breast cancer cell death. Cell Death Dis. 2016;7:1-12.
126. Kim YN, Choe SR, Cho KH, Cho DY, Kang J, Park CG, et al. Resveratrol suppresses breast cancer cell invasion by inactivating a RhoA/YAP signaling axis. Exp Mol Med. 2017;49:1-9.
127. Venkatadri R, Iyer AKV, Kaushik V, Azad N. A novel resveratrol–salinomycin combination sensitizes ER-positive breast cancer cells to apoptosis. Pharmacol Rep. 2017;69:788-797.
128. Medina-Aguilar R, Marchat LA, Arechaga Ocampo E, Gariglio P, García Mena J, Villegas Sepúlveda N, et al. Resveratrol inhibits cell cycle progression by targeting Aurora kinase A and Polo-like kinase 1 in breast cancer cells. Oncol Rep. 2016;35:3696-3704.
129. Suh J, Kim DH, Surh YJ. Resveratrol suppresses migration, invasion and stemness of human breast cancer cells by interfering with tumor-stromal cross-talk. Arch Biochem Biophys. 2018;643:62-71.
130. Zhang Y. Cancer-preventive isothiocyanates: measurement of human exposure and mechanism of action. Mutat Res/Fund Mol M. 2004;555:173-190.
131. Singh SV, Singh K. Cancer chemoprevention with dietary isothiocyanates mature for clinical translational research. Carcinogenesis. 2012;33:1833-1842.
132. Li Y, Zhang T. Targeting cancer stem cells with sulforaphane, a dietary component from broccoli and broccoli sprouts. Future Oncol. 2013;9:1097-1103.
133. Li Y, Zhang T, Korkaya H, Liu S, Lee HF, Newman B, et al. Sulforaphane, a dietary component of broccoli/Broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res. 2010;16:2580-2590.
134. Royston KJ, Udayakumar N, Lewis K, Tollefsbol TO. 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;18:1-17.
135. Keshandehghan A, Nikkhah S, Tahermansouri H, Heidari-Keshel S, Gardaneh M. Co-treatment with sulforaphane and nano-metformin molecules accelerates apoptosis in HER2+ breast cancer cells by inhibiting key molecules. Nutr Cancer. 2019:1-14.
136. Burnett JP, Lim G, Li Y, Shah RB, Lim R, Paholak HJ, et al. Sulforaphane enhances the anticancer activity of taxanes against triple negative breast cancer by killing cancer stem cells. Cancer Lett. 2017;394:52-64.
137. Castro NP, Rangel MC, Merchant AS, MacKinnon G, Cuttitta F, Salomon DS, et al. Sulforaphane suppresses the growth of triple-negative breast cancer stem-like cells in vitro and in vivo. Cancer Prev Res. 2019;12:147-158.
138. Huang J, Tao C, Yu Y, Yu F, Zhang H, Gao J, et al. Simultaneous targeting of differentiated breast cancer cells and breast cancer stem cells by combination of docetaxel-and sulforaphane-loaded self-assembled Poly (D, L-lactide-co-glycolide)/Hyaluronic acid block copolymer-based nanoparticles. J Biomed Nanotechnol. 2016;12:1463-1477.
139. Woo CC, Kumar AP, Sethi G, Tan KHB. Thymoquinone: potential cure for inflammatory disorders and cancer. Biochem Pharmacol. 2012;83:443-451.
140. Linjawi SA, Khalil WK, Hassanane MM, Ahmed ES. Evaluation of the protective effect of Nigella sativa extract and its primary active component thymoquinone against DMBA-induced breast cancer in female rats. Arch Med Sci: AMS. 2015;11:220-229.
141. Woo CC, Hsu A, Kumar AP, Sethi G, Tan KHB. Thymoquinone inhibits tumor growth and induces apoptosis in a breast cancer xenograft mouse model: the role of p38 MAPK and ROS. PLoS One. 2013;8:1-14.
142. Sutton KM, Greenshields AL, Hoskin DW. Thymoquinone, a bioactive component of black caraway seeds, causes G1 phase cell cycle arrest and apoptosis in triple-negative breast cancer cells with mutant p53. Nutr Cancer. 2014;66:408-418.
143. Rajput S, Kumar BP, Sarkar S, Das S, Azab B, Santhekadur PK, et al. Targeted apoptotic effects of thymoquinone and tamoxifen on XIAP mediated Akt regulation in breast cancer. PLoS One. 2013;8:1-13.
144. Bashmail HA, Alamoudi AA, Noorwali A, Hegazy GA, G AJ, Choudhry H, et al. Thymoquinone synergizes gemcitabine anti-breast cancer activity via modulating its apoptotic and autophagic activities. Sci Rep. 2018;8:1-11.
145. Kabil NN, Bayraktar R, Kahraman N, Ozpolat B. Thymoquinone inhibits elongation factor 2 kinase signaling axis by inducing tumor suppressor miR-603 in triple negative breast cancer cells. AACR; 2017;2017:1-5.
146. Shanmugam MK, Ahn KS, Hsu A, Woo CC, Yuan Y, Tan KHB, et al. Thymoquinone inhibits bone metastasis of breast cancer cells through abrogation of the CXCR4 signaling axis. Front Pharmacol. 2018;9:1-15.
147. Pasupuleti VR, Sammugam L, Ramesh N, Gan SH. Honey, propolis, and royal jelly: a comprehensive review of their biological actions and health benefits. Oxid Med Cell Longev. 2017;2017:1-21.
148. Nagai T, Inoue R. Preparation and the functional properties of water extract and alkaline extract of royal jelly. Food Chem. 2004;84:181-186.
149. Ramadan MF, Al-Ghamdi A. Bioactive compounds and health-promoting properties of royal jelly: a review. J Funct Foods. 2012;4:39-52.
150. Khazaei M, Ansarian A, Ghanbari E. New findings on biological actions and clinical applications of royal jelly: a review. J Diet Suppl. 2018;15:757-775.
151. Sugiyama T, Takahashi K, Mori H. Royal jelly acid, 10-hydroxy-trans-2-decenoic acid, as a modulator of the innate immune responses. Endocr Metab Immune Dis Drug Targets. 2012;12:368-376.
152. Badr G, Sayed EA, Waly H, Hassan KA, Mahmoud MH, Selamoglu Z. The therapeutic mechanisms of propolis against ccl4 -mediated liver injury by mediating apoptosis of activated hepatic stellate cells and improving the hepatic architecture through PI3K/AKT/mTOR, TGF-beta/Smad2, Bcl2/BAX/P53 and iNOS Signaling Pathways. Cell Physiol Biochem. 2019;53:301-322.
153. Biesalski HK, Dragsted LO, Elmadfa I, Grossklaus R, Muller M, Schrenk D, et al. Bioactive compounds: definition and assessment of activity. Nutrition. 2009;25:1202-1205.
154. Izuta H, Chikaraishi Y, Shimazawa M, Mishima S, Hara H. 10-Hydroxy-2-decenoic acid, a major fatty acid from royal jelly, inhibits VEGF-induced angiogenesis in human umbilical vein endothelial cells. Evid-Based Complement Alternat Med. 2009;6:489-494.
155. Gismondi A, Trionfera E, Canuti L, Di Marco G, Canini A. Royal jelly lipophilic fraction induces antiproliferative effects on SH-SY5Y human neuroblastoma cells. Oncol Rep. 2017;38:1833-1844.
156. Zhang S, Shao Q, Shen Z, Su S. Immunomodulatory response of 4T1 murine breast cancer model to camellia royal jelly. Biomed Res. 2017;28:1223-1230.
157. Nageen B, Sarfraz I, Rasul A, Hussain G, Rukhsar F, Irshad S, et al. Eupatilin: a natural pharmacologically active flavone compound with its wide range applications. J Asian Nat Prod Res. 2020;22:1-16.
158. Sarfraz I, Rasul A, Hussain G, Hussain SM, Ahmad M, Nageen B, et al. Malic enzyme 2 as a potential therapeutic drug target for cancer. IUBMB life. 2018;70:1076-1083.
159. Sharif F, Rasul A, Ashraf A, Hussain G, Younis T, Sarfraz I, et al. Phosphoglycerate mutase 1 in cancer: a promising target for diagnosis and therapy. IUBMB life. 2019;71:1418-1427.