Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

Novel Therapeutics Against Breast Cancer Stem Cells by Targeting Surface Markers and Signaling Pathways

Author(s): Plabon K. Das, Md. A. Rakib, Jahan A. Khanam, Suja Pillai and Farhadul Islam*

Volume 14, Issue 8, 2019

Page: [669 - 682] Pages: 14

DOI: 10.2174/1574888X14666190628104721

Price: $65

Abstract

Background: Breast cancer remains to be one of the deadliest forms of cancers, owing to the drug resistance and tumor relapse caused by breast cancer stem cells (BCSCs) despite notable advancements in radio-chemotherapies.

Objectives: To find out novel therapeutics against breast cancer stem cells by aiming surface markers and signaling pathways.

Methods: A systematic literature search was conducted through various electronic databases including, Pubmed, Scopus, Google scholar using the keywords "BCSCs, surface markers, signaling pathways and therapeutic options against breast cancer stem cell. Articles selected for the purpose of this review were reviewed and extensively analyzed.

Results: Novel therapeutic strategies include targeting BCSCs surface markers and aberrantly activated signaling pathways or targeting their components, which play critical roles in self-renewal and defense, have been shown to be significantly effective against breast cancer. In this review, we represent a number of ways against BCSCs surface markers and hyper-activated signaling pathways to target this highly malicious entity of breast cancer more effectively in order to make a feasible and useful strategy for successful breast cancer treatment. In addition, we discuss some characteristics of BCSCs in disease progression and therapy resistance.

Conclusion: BCSCs involved in cancer pathogenesis, therapy resistance and cancer recurrence. Thus, it is suggested that a multi-dimensional therapeutic approach by targeting surface markers and aberrantly activated signaling pathways of BCSCs alone or in combination with each other could really be worthwhile in the treatment of breast cancer.

Keywords: Breast cancer, BCSCs, surface markers, signaling pathways, therapeutic options, breast cancer.

[1]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018; 68(1): 7-30.
[http://dx.doi.org/10.3322/caac.21442] [PMID: 29313949]
[2]
Cancer Fact Sheet GLOBOCAN. 2012.International Agency for Research on Cancer http://globocan.iarc.fr/factsheets/cancers/breast.asp
[3]
deRinaldis E, Tutt A, Dontu G. Breast Cancer. Breast Pathol 2011; pp. 352-9.
[4]
Stockler M, Wilcken NR, Ghersi D, Simes RJ. Systematic reviews of chemotherapy and endocrine therapy in metastatic breast cancer. Cancer Treat Rev 2000; 26(3): 151-68.
[http://dx.doi.org/10.1053/ctrv.1999.0161] [PMID: 10814559]
[5]
Gupta PB, Onder TT, Jiang G, et al. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell 2009; 138(4): 645-59.
[http://dx.doi.org/10.1016/j.cell.2009.06.034] [PMID: 19682730]
[6]
Li X, Lewis MT, Huang J, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 2008; 100(9): 672-9.
[http://dx.doi.org/10.1093/jnci/djn123] [PMID: 18445819]
[7]
Palomeras S, Ruiz-Martínez S, Puig T. Targeting breast cancer stem cells to overcome treatment resistance. Molecules 2018; 23(9)E2193
[http://dx.doi.org/10.3390/molecules23092193] [PMID: 30200262]
[8]
Crabtree JS, Miele L. Breast Cancer Stem Cells. Biomedicines 2018; 6(3)E77
[http://dx.doi.org/10.3390/biomedicines6030077] [PMID: 30018256]
[9]
Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer 2005; 5(4): 275-84.
[http://dx.doi.org/10.1038/nrc1590] [PMID: 15803154]
[10]
Croker AK, Goodale D, Chu J, et al. High aldehyde dehydrogenase and expression of cancer stem cell markers selects for breast cancer cells with enhanced malignant and metastatic ability. J Cell Mol Med 2009; 13(8B): 2236-52.
[http://dx.doi.org/10.1111/j.1582-4934.2008.00455.x] [PMID: 18681906]
[11]
Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol 2015; 12(8): 445-64.
[http://dx.doi.org/10.1038/nrclinonc.2015.61] [PMID: 25850553]
[12]
Smalley M, Piggott L, Clarkson R. Breast cancer stem cells: obstacles to therapy. Cancer Lett 2013; 338(1): 57-62.
[http://dx.doi.org/10.1016/j.canlet.2012.04.023] [PMID: 22554712]
[13]
Zang S, Chen F, Dai J, et al. RNAi-mediated knockdown of Notch-1 leads to cell growth inhibition and enhanced chemosensitivity in human breast cancer. Oncol Rep 2010; 23(4): 893-9.
[PMID: 20204271]
[14]
Lawson JC, Blatch GL, Edkins AL. Cancer stem cells in breast cancer and metastasis. Breast Cancer Res Treat 2009; 118(2): 241-54.
[http://dx.doi.org/10.1007/s10549-009-0524-9] [PMID: 19731012]
[15]
Batlle E, Clevers H. Cancer stem cells revisited. Nat Med 2017; 23(10): 1124-34.
[http://dx.doi.org/10.1038/nm.4409] [PMID: 28985214]
[16]
Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003; 100(7): 3983-8.
[http://dx.doi.org/10.1073/pnas.0530291100] [PMID: 12629218]
[17]
Sin WC, Lim CL. Breast cancer stem cells-from origins to targeted therapy. Stem Cell Investig 2017; 4: 96.
[http://dx.doi.org/10.21037/sci.2017.11.03] [PMID: 29270422]
[18]
Collina F, Di Bonito M, Li Bergolis V, et al. Prognostic Value of Cancer Stem Cells Markers in Triple-Negative Breast Cancer. BioMed Res Int 2015.2015158682
[http://dx.doi.org/10.1155/2015/158682] [PMID: 26504780]
[19]
Lee WJ, Kim SC, Yoon JH, et al. Meta-analysis of tumor stem-like breast cancer cells using gene set and network analysis. PLoS One 2016; 11(2)e0148818
[http://dx.doi.org/10.1371/journal.pone.0148818] [PMID: 26870956]
[20]
Vaillant F, Asselin-Labat ML, Shackleton M, Forrest NC, Lindeman GJ, Visvader JE. The mammary progenitor marker CD61/β3 integrin identifies cancer stem cells in mouse models of mammary tumorigenesis. Cancer Res 2008; 68(19): 7711-7.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-1949] [PMID: 18829523]
[21]
Pham PV, Phan NL, Nguyen NT, et al. Differentiation of breast cancer stem cells by knockdown of CD44: Promising differentiation therapy. J Transl Med 2011; 9: 209.
[http://dx.doi.org/10.1186/1479-5876-9-209] [PMID: 22152097]
[22]
Nakanishi T, Ross DD. Breast cancer resistance protein (BCRP/ABCG2): Its role in multidrug resistance and regulation of its gene expression. Chin J Cancer 2012; 31(2): 73-99.
[http://dx.doi.org/10.5732/cjc.011.10320] [PMID: 22098950]
[23]
Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013; 13(10): 714-26.
[http://dx.doi.org/10.1038/nrc3599] [PMID: 24060863]
[24]
Maugeri-Saccà M, Zeuner A, De Maria R. Therapeutic targeting of cancer stem cells. Front Oncol 2011; 1: 10.
[http://dx.doi.org/10.3389/fonc.2011.00010] [PMID: 22655230]
[25]
Muller JM, Chevrier L, Cochaud S, Meunier AC, Chadeneau C. Hedgehog, Notch and Wnt developmental pathways as targets for anti-cancer drugs. Drug Discov Today Dis Mech 2007; 4(4): 285-91.
[http://dx.doi.org/10.1016/j.ddmec.2008.05.009]
[26]
Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature 2013; 501(7467): 328-37.
[http://dx.doi.org/10.1038/nature12624] [PMID: 24048065]
[27]
Zhao J. Cancer stem cells and chemoresistance: The smartest survives the raid. Pharmacol Ther 2016; 160: 145-58.
[http://dx.doi.org/10.1016/j.pharmthera.2016.02.008] [PMID: 26899500]
[28]
Borst P, Jonkers J, Rottenberg S. What makes tumors multidrug resistant? Cell Cycle 2007; 6(22): 2782-7.
[http://dx.doi.org/10.4161/cc.6.22.4936] [PMID: 17998803]
[29]
Cojoc M, Mäbert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin Cancer Biol 2015; 31: 16-27.
[http://dx.doi.org/10.1016/j.semcancer.2014.06.004] [PMID: 24956577]
[30]
Carrasco E, Alvarez PJ, Prados J, et al. Cancer stem cells and their implication in breast cancer. Eur J Clin Invest 2014; 44(7): 678-87.
[http://dx.doi.org/10.1111/eci.12276] [PMID: 24766664]
[31]
Charafe-Jauffret E, Ginestier C, Bertucci F, et al. ALDH1-positive cancer stem cells predict engraftment of primary breast tumors and are governed by a common stem cell program. Cancer Res 2013; 73(24): 7290-300.
[http://dx.doi.org/10.1158/0008-5472.CAN-12-4704] [PMID: 24142344]
[32]
van Oorschot B, Granata G, Di Franco S, et al. Targeting DNA double strand break repair with hyperthermia and DNA-PKcs inhibition to enhance the effect of radiation treatment. Oncotarget 2016; 7(40): 65504-13.
[http://dx.doi.org/10.18632/oncotarget.11798] [PMID: 27602767]
[33]
Yoshida GJ, Saya H. Therapeutic strategies targeting cancer stem cells. Cancer Sci 2016; 107(1): 5-11.
[http://dx.doi.org/10.1111/cas.12817] [PMID: 26362755]
[34]
Czerwinska P, Kaminska B. Regulation of breast cancer stem cell features. Contemp Oncol (Pozn) 2015; 19(1A): A7-A15.
[http://dx.doi.org/10.5114/wo.2014.47126] [PMID: 25691826]
[35]
Diehn M, Cho RW, Lobo NA, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009; 458(7239): 780-3.
[http://dx.doi.org/10.1038/nature07733] [PMID: 19194462]
[36]
Mathieu J, Zhang Z, Zhou W, et al. HIF induces human embryonic stem cell markers in cancer cells. Cancer Res 2011; 71(13): 4640-52.
[http://dx.doi.org/10.1158/0008-5472.CAN-10-3320] [PMID: 21712410]
[37]
Samanta D, Gilkes DM, Chaturvedi P, Xiang L, Semenza GL. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc Natl Acad Sci USA 2014; 111(50): E5429-38.
[http://dx.doi.org/10.1073/pnas.1421438111] [PMID: 25453096]
[38]
Hambardzumyan D, Becher OJ, Rosenblum MK, Pandolfi PP, Manova-Todorova K, Holland EC. PI3K pathway regulates survival of cancer stem cells residing in the perivascular niche following radiation in medulloblastoma in vivo. Genes Dev 2008; 22(4): 436-48.
[http://dx.doi.org/10.1101/gad.1627008] [PMID: 18281460]
[39]
Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol 2011; 8(2): 97-106.
[http://dx.doi.org/10.1038/nrclinonc.2010.196] [PMID: 21151206]
[40]
Teng Y, Wang X, Wang Y, Ma D. Wnt/beta-catenin signaling regulates cancer stem cells in lung cancer A549 cells. Biochem Biophys Res Commun 2010; 392(3): 373-9.
[http://dx.doi.org/10.1016/j.bbrc.2010.01.028] [PMID: 20074550]
[41]
Zhou J, Zhang H, Gu P, Bai J, Margolick JB, Zhang Y. NF-kappaB pathway inhibitors preferentially inhibit breast cancer stem-like cells. Breast Cancer Res Treat 2008; 111(3): 419-27.
[http://dx.doi.org/10.1007/s10549-007-9798-y] [PMID: 17965935]
[42]
Naor D, Sionov RV, Ish-Shalom D. CD44: Structure, function, and association with the malignant process. Adv Cancer Res 1997; 71: 241-319.
[http://dx.doi.org/10.1016/S0065-230X(08)60101-3] [PMID: 9111868]
[43]
Toole BP, Slomiany MG. Hyaluronan, CD44 and Emmprin: partners in cancer cell chemoresistance. Drug Resist Updat 2008; 11(3): 110-21.
[http://dx.doi.org/10.1016/j.drup.2008.04.002] [PMID: 18490190]
[44]
Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med 2006; 12(10): 1167-74.
[http://dx.doi.org/10.1038/nm1483] [PMID: 16998484]
[45]
Thapa R, Wilson GD. The importance of CD44 as a stem cell biomarker and therapeutic target in cancer. Stem Cells Int 2016.20162087204
[http://dx.doi.org/10.1155/2016/2087204] [PMID: 27200096]
[46]
Alkilany AM, Thompson LB, Boulos SP, Sisco PN, Murphy CJ. Gold nanorods: their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Adv Drug Deliv Rev 2012; 64(2): 190-9.
[http://dx.doi.org/10.1016/j.addr.2011.03.005] [PMID: 21397647]
[47]
Zeng C, Toole BP, Kinney SD, Kuo JW, Stamenkovic I. Inhibition of tumor growth in vivo by hyaluronan oligomers. Int J Cancer 1998; 77(3): 396-401.
[http://dx.doi.org/10.1002/(SICI)1097-0215(19980729)77:3<396:AID-IJC15>3.0.CO;2-6] [PMID: 9663602]
[48]
Slomiany MG, Dai L, Bomar PA, et al. Abrogating drug resistance in malignant peripheral nerve sheath tumors by disrupting hyaluronan-CD44 interactions with small hyaluronan oligosaccharides. Cancer Res 2009; 69(12): 4992-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-09-0143] [PMID: 19470767]
[49]
Gilg AG, Tye SL, Tolliver LB, et al. Targeting hyaluronan interactions in malignant gliomas and their drug-resistant multipotent progenitors. Clin Cancer Res 2008; 14(6): 1804-13.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-1228] [PMID: 18347183]
[50]
Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer 2004; 4(7): 528-39.
[http://dx.doi.org/10.1038/nrc1391] [PMID: 15229478]
[51]
Hosono K, Nishida Y, Knudson W, et al. Hyaluronan oligosaccharides inhibit tumorigenicity of osteosarcoma cell lines MG-63 and LM-8 in vitro and in vivo via perturbation of hyaluronan-rich pericellular matrix of the cells. Am J Pathol 2007; 171(1): 274-86.
[http://dx.doi.org/10.2353/ajpath.2007.060828] [PMID: 17591972]
[52]
Tan JX, Wang XY, Su XL, et al. Upregulation of HYAL1 expression in breast cancer promoted tumor cell proliferation, migration, invasion and angiogenesis. PLoS One 2011; 6(7)e22836
[http://dx.doi.org/10.1371/journal.pone.0022836] [PMID: 21829529]
[53]
Zhao L, Yang Y, Zhou P, et al. Targeting CD133high colorectal cancer cells in vitro and in vivo with an asymmetric bispecific antibody. J Immunother 2015; 38(6): 217-28.
[http://dx.doi.org/10.1097/CJI.0000000000000086] [PMID: 26049545]
[54]
Ohlfest JR, Zellmer DM, Panyam J, et al. Immunotoxin targeting CD133(+) breast carcinoma cells. Drug Deliv Transl Res 2013; 3(2): 195-204.
[http://dx.doi.org/10.1007/s13346-012-0066-2] [PMID: 25787984]
[55]
Bostad M, Olsen CE, Peng Q, Berg K, Høgset A, Selbo PK. Light-controlled endosomal escape of the novel CD133-targeting immunotoxin AC133-saporin by photochemical internalization - A minimally invasive cancer stem cell-targeting strategy. J Control Release 2015; 206: 37-48.
[http://dx.doi.org/10.1016/j.jconrel.2015.03.008] [PMID: 25758331]
[56]
Pardee AB, Li YZ, Li CJ. Cancer therapy with beta-lapachone. Curr Cancer Drug Targets 2002; 2(3): 227-42.
[http://dx.doi.org/10.2174/1568009023333854] [PMID: 12188909]
[57]
Costa MP, Feitosa AC, Oliveira FC, et al. Controlled release of nor-β-lapachone by PLGA microparticles: A strategy for improving cytotoxicity against prostate cancer cells. Molecules 2016; 21(7)E873
[http://dx.doi.org/10.3390/molecules21070873] [PMID: 27384551]
[58]
Kee JY, Han YH, Park J, et al. β-lapachone inhibits lung metastasis of colorectal cancer by inducing apoptosis of CT26 cells. Integr Cancer Ther 2017; 16(4): 585-96.
[http://dx.doi.org/10.1177/1534735416681638] [PMID: 27923905]
[59]
Li CJ, Li YZ, Pinto AV, Pardee AB. Potent inhibition of tumor survival in vivo by beta-lapachone plus taxol: Combining drugs imposes different artificial checkpoints. Proc Natl Acad Sci USA 1999; 96(23): 13369-74.
[http://dx.doi.org/10.1073/pnas.96.23.13369] [PMID: 10557327]
[60]
Bey EA, Bentle MS, Reinicke KE, et al. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc Natl Acad Sci USA 2007; 104(28): 11832-7.
[http://dx.doi.org/10.1073/pnas.0702176104] [PMID: 17609380]
[61]
Yang Y, Zhou X, Xu M, et al. β-lapachone suppresses tumour progression by inhibiting epithelial-to-mesenchymal transition in NQO1-positive breast cancers. Sci Rep 2017; 7(1): 2681.
[http://dx.doi.org/10.1038/s41598-017-02937-0] [PMID: 28578385]
[62]
Kim DW, Cho JY. NQO1 is required for β-lapachone-mediated downregulation of breast-cancer stem-cell activity. Int J Mol Sci 2018; 19(12)E3813
[http://dx.doi.org/10.3390/ijms19123813] [PMID: 30513573]
[63]
Glorieux C, Sandoval JM, Dejeans N, et al. Overexpression of NAD(P)H:quinone oxidoreductase 1 (NQO1) and genomic gain of the NQO1 locus modulates breast cancer cell sensitivity to quinones. Life Sci 2016; 145: 57-65.
[http://dx.doi.org/10.1016/j.lfs.2015.12.017] [PMID: 26687450]
[64]
Scatena C, Roncella M, Di Paolo A, et al. Doxycycline, an inhibitor of mitochondrial biogenesis, effectively reduces cancer stem cells (CSCs) in early breast cancer patients: A clinical pilot study. Front Oncol 2018; 8: 452.
[http://dx.doi.org/10.3389/fonc.2018.00452] [PMID: 30364293]
[65]
Zhang L, Xu L, Zhang F, Vlashi E. Doxycycline inhibits the cancer stem cell phenotype and epithelial-to-mesenchymal transition in breast cancer. Cell Cycle 2017; 16(8): 737-45.
[http://dx.doi.org/10.1080/15384101.2016.1241929] [PMID: 27753527]
[66]
Lin CC, Lo MC, Moody RR, Stevers NO, Tinsley SL, Sun D. Doxycycline targets aldehyde dehydrogenase-positive breast cancer stem cells. Oncol Rep 2018; 39(6): 3041-7.
[http://dx.doi.org/10.3892/or.2018.6337] [PMID: 29620216]
[67]
Tin AS, Park AH, Sundar SN, Firestone GL. Essential role of the cancer stem/progenitor cell marker nucleostemin for indole-3-carbinol anti-proliferative responsiveness in human breast cancer cells. BMC Biol 2014; 12: 72.
[http://dx.doi.org/10.1186/s12915-014-0072-6] [PMID: 25209720]
[68]
Moll UM, Petrenko O. The MDM2-p53 interaction. Mol Cancer Res 2003; 1(14): 1001-8.
[PMID: 14707283]
[69]
Wang R, Yang L, Li S, et al. Quercetin Inhibits Breast Cancer Stem Cells via Downregulation of Aldehyde Dehydrogenase 1A1 (ALDH1A1), Chemokine Receptor Type 4 (CXCR4), Mucin 1 (MUC1), and Epithelial Cell Adhesion Molecule (EpCAM). Med Sci Monit 2018; 24: 412-20.
[http://dx.doi.org/10.12659/MSM.908022] [PMID: 29353288]
[70]
Miyoshi Y, Murase K, Saito M, Imamura M, Oh K. Mechanisms of estrogen receptor-α upregulation in breast cancers. Med Mol Morphol 2010; 43(4): 193-6.
[http://dx.doi.org/10.1007/s00795-010-0514-3] [PMID: 21267694]
[71]
Tyson JJ, Baumann WT, Chen C, et al. Dynamic modelling of oestrogen signalling and cell fate in breast cancer cells. Nat Rev Cancer 2011; 11(7): 523-32.
[http://dx.doi.org/10.1038/nrc3081] [PMID: 21677677]
[72]
Guo L, Li F, Wang M, et al. 17β-estradiol regulates the malignancy of cancer stem-like cells derived from the MCF7 cell line partially through Sox2. Oncol Lett 2018; 15(3): 3790-5.
[http://dx.doi.org/10.3892/ol.2018.7796] [PMID: 29467896]
[73]
Battula VL, Shi Y, Evans KW, et al. Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis. J Clin Invest 2012; 122(6): 2066-78.
[http://dx.doi.org/10.1172/JCI59735] [PMID: 22585577]
[74]
Ahmed M, Cheung NK. Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy. FEBS Lett 2014; 588(2): 288-97.
[http://dx.doi.org/10.1016/j.febslet.2013.11.030] [PMID: 24295643]
[75]
Kim SH, Singh SV. Mammary cancer chemoprevention by withaferin A is accompanied by in vivo suppression of self-renewal of cancer stem cells. Cancer Prev Res (Phila) 2014; 7(7): 738-47.
[http://dx.doi.org/10.1158/1940-6207.CAPR-13-0445] [PMID: 24824039]
[76]
Paholak HJ, Stevers NO, Chen H, et al. Elimination of epithelial-like and mesenchymal-like breast cancer stem cells to inhibit metastasis following nanoparticle-mediated photothermal therapy. Biomaterials 2016; 104: 145-57.
[http://dx.doi.org/10.1016/j.biomaterials.2016.06.045] [PMID: 27450902]
[77]
Hunter T. Signalinge 2000 and beyond Cell 2000; 100(1): 113e127.
[78]
Hunter T. The age of crosstalk: phosphorylation, ubiquitination, and beyond Mol Cell 2007; 28(5): 730e738.
[http://dx.doi.org/10.1016/j.molcel.2007.11.019]
[79]
Sever R, Brugge JS. Signal transduction in cancer. Cold Spring Harb Perspect Med 2015; 5(4)a006098
[http://dx.doi.org/10.1101/cshperspect.a006098] [PMID: 25833940]
[80]
Sridharan S, Howard CM, Tilley AMC, et al. Novel and alternative targets against breast cancer stemness to combat chemoresistance. Front Oncol 2019; 9: 1003.
[81]
Nwabo Kamdje AH, Takam Kamga P, Tagne Simo R, et al. Developmental pathways associated with cancer metastasis: Notch, Wnt, and Hedgehog. Cancer Biol Med 2017; 14(2): 109-20.
[82]
Merchant AA, Matsui W. Targeting Hedgehog--a cancer stem cell pathway. Clin Cancer Res 2010; 16(12): 3130-40.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-2846] [PMID: 20530699]
[83]
Harrison H, Farnie G, Howell SJ, et al. Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res 2010; 70(2): 709-18.
[http://dx.doi.org/10.1158/0008-5472.CAN-09-1681] [PMID: 20068161]
[84]
Qiao L, Wong BC. Role of Notch signaling in colorectal cancer. Carcinogenesis 2009; 30(12): 1979-86.
[http://dx.doi.org/10.1093/carcin/bgp236] [PMID: 19793799]
[85]
Pannuti A, Foreman K, Rizzo P, et al. Targeting Notch to target cancer stem cells. Clin Cancer Res 2010; 16(12): 3141-52.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-2823] [PMID: 20530696]
[86]
Wang Z, Ahmad A, Li Y, Azmi AS, Miele L, Sarkar FH. Targeting notch to eradicate pancreatic cancer stem cells for cancer therapy. Anticancer Res 2011; 31(4): 1105-13.
[PMID: 21508353]
[87]
Mazzone M, Selfors LM, Albeck J, et al. Dose-dependent induction of distinct phenotypic responses to Notch pathway activation in mammary epithelial cells. Proc Natl Acad Sci USA 2010; 107(11): 5012-7.
[http://dx.doi.org/10.1073/pnas.1000896107] [PMID: 20194747]
[88]
Baker A, Wyatt D, Bocchetta M, et al. Notch-1-PTEN-ERK1/2 signaling axis promotes HER2+ breast cancer cell proliferation and stem cell survival. Oncogene 2018; 37(33): 4489-504.
[89]
Phillips TM, McBride WH, Pajonk F. The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst 2006; 98(24): 1777-85.
[http://dx.doi.org/10.1093/jnci/djj495] [PMID: 17179479]
[90]
Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat Rev Cancer 2003; 3(10): 756e767.
[http://dx.doi.org/10.1038/nrc1186]
[91]
Reedijk M, Odorcic S, Chang L, et al. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival Cancer Res 2005; 65(18): 8530e8537.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-1069]
[92]
Suman S, Das TP, Damodaran C. Silencing NOTCH signaling causes growth arrest in both breast cancer stem cells and breast cancer cells. Br J Cancer 2013; 109(10): 2587-96.
[http://dx.doi.org/10.1038/bjc.2013.642] [PMID: 24129237]
[93]
Shan NL, Wahler J, Lee HJ, et al. Vitamin D compounds inhibit cancer stem-like cells and induce differentiation in triple negative breast cancer. J Steroid Biochem Mol Biol 2017; 173: 122-9.
[http://dx.doi.org/10.1016/j.jsbmb.2016.12.001] [PMID: 27923595]
[94]
Saeg F, Anbalagan M. Breast cancer stem cells and the challenges of eradication: a review of novel therapies. Stem Cell Investig 2018; 5: 39.
[http://dx.doi.org/10.21037/sci.2018.10.05] [PMID: 30498750]
[95]
Nusse R, Fuerer C, Ching W, et al. Wnt signaling and stem cell control. Cold Spring Harb Symp Quant Biol 2008; 73: 59-66.
[http://dx.doi.org/10.1101/sqb.2008.73.035] [PMID: 19028988]
[96]
MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell 2009; 17(1): 9-26.
[http://dx.doi.org/10.1016/j.devcel.2009.06.016] [PMID: 19619488]
[97]
Brennan KR, Brown AM. Wnt proteins in mammary development and cancer. J Mammary Gland Biol Neoplasia 2004; 9(2): 119-31.
[http://dx.doi.org/10.1023/B:JOMG.0000037157.94207.33] [PMID: 15300008]
[98]
Nagahata T, Shimada T, Harada A, et al. Amplification, upregulation and over-expression of DVL-1, the human counterpart of the Drosophila disheveled gene, in primary breast cancers Cancer Sci 2003; 94(6): 515e518.
[99]
Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis 2008; 4(2): 68-75.
[http://dx.doi.org/10.4161/org.4.2.5851] [PMID: 19279717]
[100]
Zhou QM, Sun Y, Lu YY, Zhang H, Chen QL, Su SB. Curcumin reduces mitomycin C resistance in breast cancer stem cells by regulating Bcl-2 family-mediated apoptosis. Cancer Cell Int 2017; 17: 84.
[http://dx.doi.org/10.1186/s12935-017-0453-3] [PMID: 28959140]
[101]
Wang N, Wang Q, Tang H, et al. Direct inhibition of ACTN4 by ellagic acid limits breast cancer metastasis via regulation of β-catenin stabilization in cancer stem cells. J Exp Clin Cancer Res 2017; 36(1): 172.
[http://dx.doi.org/10.1186/s13046-017-0635-9] [PMID: 29197410]
[102]
Ahmadipour F, Noordin MI, Mohan S, et al. Koenimbin, a natural dietary compound of Murraya koenigii (L) Spreng: Inhibition of MCF7 breast cancer cells and targeting of derived MCF7 breast cancer stem cells (CD44(+)/CD24(-/low)): an in vitro study. Drug Des Devel Ther 2015; 9: 1193-208.
[PMID: 25759564]
[103]
Li Y, Zhang T, Korkaya H, et al. Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res 2010; 16(9): 2580-90.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-2937] [PMID: 20388854]
[104]
Fani S, Dehghan F, Karimian H, et al. Monobenzyltin Complex C1 Induces Apoptosis in MCF-7 Breast Cancer Cells through the Intrinsic Signaling Pathway and through the Targeting of MCF-7-Derived Breast Cancer Stem Cells via the Wnt/β-Catenin Signaling Pathway. PLoS One 2016; 11(8)e0160836
[http://dx.doi.org/10.1371/journal.pone.0160836] [PMID: 27529753]
[105]
Huang C, Chen Y, Liu H, et al. Celecoxib targets breast cancer stem cells by inhibiting the synthesis of prostaglandin E2 and down-regulating the Wnt pathway activity. Oncotarget 2017; 8(70): 115254-69.
[http://dx.doi.org/10.18632/oncotarget.23250] [PMID: 29383157]
[106]
Liu J, Pan S, Hsieh MH, et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc Natl Acad Sci USA 2013; 110(50): 20224-9.
[http://dx.doi.org/10.1073/pnas.1314239110] [PMID: 24277854]
[107]
Kameda C, Tanaka H, Yamasaki A, et al. The Hedgehog pathway is a possible therapeutic target for patients with estrogen receptor-negative breast cancer. Anticancer Res 2009; 29(3): 871-9.
[PMID: 19414322]
[108]
Lee MY, Sun L, Veltmaat JM. Hedgehog and Gli signaling in embryonic mammary gland development. J Mammary Gland Biol Neoplasia 2013; 18(2): 133-8.
[http://dx.doi.org/10.1007/s10911-013-9291-7] [PMID: 23677624]
[109]
Kubo M, Nakamura M, Tasaki A, et al. Hedgehog signaling pathway is a new therapeutic target for patients with breast cancer. Cancer Res 2004; 64(17): 6071-4.
[http://dx.doi.org/10.1158/0008-5472.CAN-04-0416] [PMID: 15342389]
[110]
Sun M, Zhang N, Wang X, et al. Hedgehog pathway is involved in nitidine chloride induced inhibition of epithelial-mesenchymal transition and cancer stem cells-like properties in breast cancer cells. Cell Biosci 2016; 6: 44.
[http://dx.doi.org/10.1186/s13578-016-0104-8] [PMID: 27313840]
[111]
Sims-Mourtada J, Opdenaker LM, Davis J, Arnold KM, Flynn D. Taxane-induced hedgehog signaling is linked to expansion of breast cancer stem-like populations after chemotherapy. Mol Carcinog 2015; 54(11): 1480-93.
[http://dx.doi.org/10.1002/mc.22225] [PMID: 25263583]
[112]
Hui M, Cazet A, Nair R, Watkins DN, O’Toole SA, Swarbrick A. The Hedgehog signalling pathway in breast development, carcinogenesis and cancer therapy. Breast Cancer Res 2013; 15(2): 203.
[http://dx.doi.org/10.1186/bcr3401] [PMID: 23547970]
[113]
Cazet AS, Hui MN, Elsworth BL, et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun 2018; 9(1): 2897.
[http://dx.doi.org/10.1038/s41467-018-05220-6] [PMID: 30042390]
[114]
Ramos A, Camargo FD. The Hippo signaling pathway and stem cell biology. Trends Cell Biol 2012; 22(7): 339-46.
[http://dx.doi.org/10.1016/j.tcb.2012.04.006] [PMID: 22658639]
[115]
Staley BK, Irvine KD. Hippo signaling in Drosophila: Recent advances and insights. Dev Dyn 2012; 241(1): 3-15.
[http://dx.doi.org/10.1002/dvdy.22723] [PMID: 22174083]
[116]
Stanger BZ. Quit your YAPing: a new target for cancer therapy. Genes Dev 2012; 26(12): 1263-7.
[http://dx.doi.org/10.1101/gad.196501.112] [PMID: 22713867]
[117]
Li YW, Xu J, Zhu GY, 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: 105.
[http://dx.doi.org/10.1038/s41420-018-0124-8] [PMID: 30479839]
[118]
Hosea R, Hardiany NS, Ohneda O, Wanandi SI. Glucosamine decreases the stemness of human ALDH+ breast cancer stem cells by inactivating STAT3. Oncol Lett 2018; 16(4): 4737-44.
[http://dx.doi.org/10.3892/ol.2018.9222] [PMID: 30214607]
[119]
Tan XF, Teo WX, Yip GW. In vitro evaluation of candidate gene targets for cancer therapy. Methods Mol Biol 2019; 1974: 21-30.
[http://dx.doi.org/10.1007/978-1-4939-9220-1_2] [PMID: 31098992]
[120]
Jafarlou M, Baradaran B, Saedi TA, et al. An overview of the history, applications, advantages, disadvantages and prospects of gene therapy. J Biol Regul Homeost Agents 2016; 30(2): 315-21.
[PMID: 27358116]
[121]
Hu C, Xu L, Liang S, Zhang Z, Zhang Y, Zhang F. Lentivirus-mediated shRNA targeting Nanog inhibits cell proliferation and attenuates cancer stem cell activities in breast cancer. J Drug Target 2016; 24(5): 422-32.
[122]
Zhang W, Grivennikov SI. Top Notch cancer stem cells by paracrine NF-κB signaling in breast cancer. Breast Cancer Res 2013; 15(5): 316.
[http://dx.doi.org/10.1186/bcr3565] [PMID: 24172068]
[123]
Shim Y, Song JM. Quantum dot nanoprobe-based high-content monitoring of notch pathway inhibition of breast cancer stem cell by capsaicin. Mol Cell Probes 2015; 29(6): 376-81.
[http://dx.doi.org/10.1016/j.mcp.2015.09.004] [PMID: 26384954]
[124]
Koike Y, Ohta Y, Saitoh W, et al. Anti-cell growth and anti-cancer stem cell activities of the non-canonical hedgehog inhibitor GANT61 in triple-negative breast cancer cells. Breast Cancer 2017; 24(5): 683-93.
[http://dx.doi.org/10.1007/s12282-017-0757-0] [PMID: 28144905]

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