Cancer Stem Cell Hypothesis: Implication for Cancer Prevention and Treatment
Abstract
BACKGROUND: Cancer is a disease of genomic instability, evasion of immune cells, and adaptation of the tumor cells to the changing environment. Genetic heterogeneity caused by tumors and tumor microenvironmental factors forms the basis of aggressive behavior of some cancer cell populations.
CONTENT: Cancers arise in self-renewing cell populations and that the resulting cancers, like their normal organ counterparts, are composed of hierarchically organized cell populations. Self – renewing “cancer stem cells” (CSC) maintain tumor growth and generate the diverse populations constituting the tumor bulk. CSCs in multiple tumor types have been demonstrated to be relatively resistant to radiation and chemotherapy. The clinical relevance of these studies has been supported by neoadjuvant breast cancer trials that demonstrated increases in the proportions of CSCs after therapy. The CSC hypothesis has tremendously important clinical implications.
SUMMARY: In summary, a large and accumulating body of evidence supports the CSC hypothesis, which has important implications for cancer prevention and therapy. The ultimate test of this hypothesis will require clinical trials demonstrating that targeting of these pathways reduces cancer incidence and improves outcomes for patients with cancer.
KEYWORDS: Somatic mutation, tumor heterogeneity, metastasis, epithelial-mesenchymal transition, CSC niche
Full Text:
PDFReferences
Merlo LM, Pepper JW, Reid BJ, Maley CC. Cancer as an evolutionary and ecological process. Nat Rev Cancer. 2006; 6: 924-35, CrossRef.
Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012; 481: 306-13, CrossRef.
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144: 646-74, CrossRef.
Blanpain T. Tracing the cellular origin of cancer. Nat Cell Biol. 2013; 15: 126-34, CrossRef.
Badve S, Nakshatri H. Breast-cancer stem cells: beyond semantics. Lancet Oncol. 2012; 13: e43-8, CrossRef.
Qi Y, Li RM, Kong FM, Li H, Yu JP, Ren XB. How do tumor stem cells actively escape from host immunosurveillance? Biochem Biophys Res Commun. 2012; 420: 699-703, CrossRef.
Magee JA, Piskounova E, Morrison SJ. Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer Cell. 2012; 21: 283-96, CrossRef.
Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997; 3: 730-7, CrossRef.
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: 3983-8, CrossRef.
Gokmen-Polar Y, Badve SS. Cancer stem cells: nonbeliever’s viewpoint. Clin Chem. 2013; 59: 208-10, CrossRef.
Jan M, Snyder TM, Corces-Zimmerman MR, Vyas P, Weissman IL, Quake SR, et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci Transl Med. 2012; 4: 149ra18, CrossRef.
Azizi E, Wicha MS. Cancer stem cells: The evidence accumulates. Clin Chem. 2013; 59: 205-7, CrossRef.
Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012; 488: 522-6, CrossRef.
Driessens G, Beck B, Caauwe A, Simons BD, Blanpain C. Defining the mode of tumour growth by clonal analysis. Nature. 2012; 488: 527-30, CrossRef.
Schepers AG, Snippert HJ, Stange DE, van den Born M, van Es JH, van de Wetering M, et al. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science. 2012; 337: 730-5, CrossRef.
Gilbertson RJ, Graham TA. Cancer: Resolving the stem-cell debate. Nature. 2012; 488: 462-3, CrossRef.
Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011; 17: 313-9, CrossRef.
Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014; 14: 275-91, CrossRef.
Visvader JE, Lindeman GJ. Cancer stem cells: Current status and evolving complexities. Cell Stem Cell. 2012; 10: 717-28, CrossRef.
Matchett KB, Lappin TR. Concise reviews: cancer stem cells: from concept to cure. Stem Cells. 2014; 32: 2563-70, CrossRef.
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: 9339-44, CrossRef.
Nguyen LV, Vanner R, Dirks P, Eaves CJ. Cancer stem cells: an evolving concept. Nat Rev Cancer. 2012; 12: 133-43, CrossRef.
Sarry JE, Murphy K, Perry R, Sanchez PV, Secreto A, Keefer C, et al. Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rcc-deficient mice. J Clin Invest. 2011; 121: 384-95, CrossRef.
Li H, Tang DG. Prostate cancer stem cells and their potential roles in metastasis. J Surg Oncol. 2011; 103: 558-62, CrossRef.
Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007; 67: 1030-7, CrossRef.
Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007; 445: 111-5, CrossRef.
Vermeulen L, Todaro M, de Sousa Mello F, Sprick MR, Kemper K, Perez Alea M, et al. Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc Natl Acad Sci USA. 2008; 105: 13427-32, CrossRef.
Liu S, Ginestier C, Ou SJ, Clouthier SG, Patel SH, Monville F, et al. Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks. Cancer Res. 2011; 71: 614-24, CrossRef.
Charafe-Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, Finetti P, et al. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res. 2009; 69: 1302-13, CrossRef.
Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008; 15: 504-14, CrossRef.
Wang J, Sakariassen PØ, Tsinkalovsky O, Immervoll H, Bøe SO, Svendsen A, et al. CD133 negative glioma cells form tumors in nude rats and give rise to CD133 positive cells. Int J Cancer. 2008; 122: 761-8, CrossRef.
Pollard SM, Yoshikawa K, Clarke ID, Danovi D, Stricker S, Russell R, et al. Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell. 2009; 4: 568-80, CrossRef.
Quintana E, Shackleton M, Foster HR, Fullen DR, Sabel MS, Johnson TM, et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell. 2010; 18: 510-23, CrossRef.
Girouard SD, Murphy GF. Melanoma stem cells: Not rare, but well done. Lab Invest. 2011; 91: 647-64, CrossRef.
Chaffer C, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA, Rodrigues LO, et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci USA. 2011; 108: 7950-5, CrossRef.
Iliopoulos D, Hirsch H, Wang G, Struhl K. Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion. Proc Natl Acad Sci USA. 2011; 108: 1397-402, CrossRef.
Lander AD. The ‘stem cell’ concept: Is it holding us back? J Biol. 2009; 8: 70, CrossRef.
Vermeulen L, Sprick MR, Kemper K, Stassi G, Medema JP. Cancer stem cells: old concepts, new insights. Cell Death Differ. 2008; 15: 947-58, CrossRef.
Vermeulen L, de Sousa e Melo F, Richel DJ, Medema JP. The developing cancer stem-cell model: clinical challenges and opportunities. Lancet Oncol. 2012; 13: e83-9, CrossRef.
Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol. 2013; 15: 338-44, CrossRef.
Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008; 133: 704-15, CrossRef.
Morel AP, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE. 2008; 3: e2888, CrossRef.
Scheel C, Eaton EN, Li SH, Chaffer CL, Reinhardt F, Kah KJ, et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell. 2011; 145: 926-40, CrossRef.
Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T. Opinion: migrating cancer stem cells: an integrated concept of malignant tumour progression. Nat Rev Cancer. 2005; 5: 744-9, CrossRef.
Yang J, Weinberg RA. Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell. 2008; 14: 818-29, CrossRef.
Fuchs E. The tortoise and the hair: slow-cycling cells in the stem cell race. Cell. 2009; 137: 811-9, CrossRef.
Essers MA, Trumpp A. Targeting leukemic stem cells by breaking their dormancy. Mol Oncol. 2010; 4: 443-50, CrossRef.
Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov. 2009; 8: 806-23, CrossRef.
Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006; 444: 756-60, CrossRef.
Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst. 2008; 100: 672-9, CrossRef.
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: 780-3, CrossRef.
O’Hare T, Corbin AS, Druker BJ. Targeted CML therapy: controlling drug resistance, seeking cure. Curr Opin Genet Dev. 2006; 16: 92-9, CrossRef.
Oravecz-Wilson KI, Philips ST, Yilmaz OH, Ames HM, Li L, Crawford BD, et al. Persistence of leukemia-initiating cells in a conditional knockin model of an imatinib-responsive myeloproliferative disorder. Cancer Cell. 2009; 16: 137-48, CrossRef.
Masters JR, Koberle B. Curing metastatic cancer: lessons from testicular germ-cell tumours. Nat Rev Cancer. 2003; 3: 517-25, CrossRef.
Sikic BI. Natural and acquired resistance to cancer therapies. In: Mendelsohn J, Howley PM, Israel MA, Gray JW, Thompson CB, editors. The Molecular Basis of Cancer. 3rd ed. Philadelphia: Saunders/Elsevier; 2008. p.583-92, CrossRef.
Read TA, Fogarty MP, Markant SL, McLendon RE, Wei Z, Ellison DW, et al. Identification of CD15 as a marker for tumor-propagating cells in a mouse model of medulloblastoma. Cancer Cell. 2009; 15: 135-47, CrossRef.
Son MJ, Woolard K, Nam DH, Lee J, Fine HA. SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma. Cell Stem Cell. 2009; 4: 440-52, CrossRef.
Ward RJ, Lee L, Graham K, Satkunendran T, Yoshikawa K, Ling E, et al. Multipotent CD15+ cancer stem cells in patched-1-deficient mouse medulloblastoma. Cancer Res. 2009; 69: 4682-90, CrossRef.
Lathia JD, Gallagher J, Heddleston JM, Wang J, Eyler CE, Macswords J, et al. Integrin alpha 6 regulates glioblastoma stem cells. Cell Stem Cell. 2010; 6: 421-32, CrossRef.
Curtis SJ, Sinkevicius KW, Li D, Lau AN, Roach RR, Zamponi R, et al. Primary tumor genotype is an important determinant in identification of lung cancer propagating cells. Cell Stem Cell. 2010; 7: 127-33, CrossRef.
Meyer MJ, Fleming JM, Lin AF, Hussnain SA, Ginsburg E, Vonderhaar BK. CD44posCD49fhiCD133/2hi defines xenograft-initiating cells in estrogen receptor-negative breast cancer. Cancer Res. 2010; 70: 4624-33, CrossRef.
Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, 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: 555-67, CrossRef.
Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci USA. 2007; 104: 10158-63, CrossRef.
Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007; 1: 313-23, CrossRef.
Curley MD, Therrien VA, Cummings CL, Sergent PA, Koulouris CR, Friel AM, et al. CD133 expression defines a tumor initiating cell population in primary human ovarian cancer. Stem Cells. 2009; 27: 2875-83, CrossRef.
Stewart JM, Shaw PA, Gedye C, Bernardini MQ, Neel BG, Ailles LE. Phenotypic heterogeneity and instability of human ovarian tumor- initiating cells. Proc Natl Acad Sci USA. 2011; 108: 6468-73, CrossRef.
Zhang WC, Shyh-Chang N, Yang H, Rai A, Umashankar S, Ma S, et al. Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis. Cell. 2012; 148: 259-72, CrossRef.
Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008; 9: 582-9, CrossRef.
Brabletz T. To differentiate or not: routes towards metastasis. Nat Rev Cancer. 2012; 12: 425-36, CrossRef.
Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008; 22: 894-907, CrossRef.
Todaro M, Francipane MG, Medema JP, Stassi G. Colon cancer stem cells: promise of targeted therapy. Gastroenterology. 2010; 138: 2151-62, CrossRef.
Xie X, Teknos TN, Pan Q. Are all cancer stem cells created equal? Stem Cell Transl Med. 2014; 3: 1111-5, CrossRef.
Schwitalla S, Fingerle AA, Cammareri P, Nebelsiek T, Göktuna SI, Ziegler PK, et al. Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell. 2013; 152: 25-38, CrossRef.
Ischenko I, Zhi J, Moll UM, Nemajerova A, Petrenko O. Direct reprogramming by oncogenic Ras and Myc. Proc Natl Acad Sci USA. 2013; 110: 3937-42, CrossRef.
Niwa T, Tsukamoto T, Toyoda T, Mori A, Tanaka H, Maekita T, et al. Inflammatory processes triggered by Helicobacter pylori infection cause aberrant DNA methylation in gastric epithelial cells. Cancer Res. 2010; 70: 1430-40, CrossRef.
Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014; 505: 495-501, CrossRef.
Yamada Y, Haga H, Yamada Y. Concise review: dedifferentiation meets cancer development: proof of concept for epigenetic cancer. Stem Cells Transl Med. 2014; 3: 1182-7, CrossRef.
Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008; 40: 499-507, CrossRef.
Folmes CD, Nelson TJ, Martinez-Fernandez A, Arrell DK, Lindor JZ, Dzeja PP, et al. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell Metab. 2011; 14: 264-71, CrossRef.
Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, et al. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature. 2009; 460: 1132-5, CrossRef.
Hochedlinger K, Yamada Y, Beard C, Jaenisch R. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell. 2005; 121: 465-77, CrossRef.
Ohnisi K, Semi K, Yamamoto T, Shimizu M, Tanaka A, Mitsunaga K, et al. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell. 2014; 156: 663-77, CrossRef.
Friedmann-Morvinsky D, Verma IM. Dedifferentiation and reprogramming: origins of cancer stem cells. EMBO Rep. 2014; 25: 244-53, CrossRef.
Riggi N, Suva ML, De Vito C, Provero P, Stehle JC, Baumer K, et al. EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev. 2010; 24: 916-32, CrossRef.
Sarkar A, Hochedlinger K. The sox family of transcription factors: versatile regulators of stem and progenitor cell fate. Cell Stem Cell. 2013; 12: 15-30, CrossRef.
Yamanaka S. Induced pluripotent stem cells: past, present, and future. Cell Stem Cell. 2012; 10: 678-84, CrossRef.
Dang CV. MYC on the path to cancer. Cell. 2012; 149: 22-35, CrossRef.
Morris SA, Daley GQ. A blueprint for engineering cell fate: current technologies to reprogram cell identity. Cell Res. 2013; 23: 33-48, CrossRef.
Daley GQ. Common themes of dedifferentiation in somatic cell reprogramming and cancer. Cold Spring Harb Symp Quant Biol. 2008; 73: 171-4, CrossRef.
Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nature Rev Mol Cell Biol. 2006; 7: 131-42, CrossRef.
Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009; 139: 871-90, CrossRef.
Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009; 119: 1420-8, CrossRef.
Chapman HA. Epithelial-mesenchymal interactions in pulmonary fibrosis. Annu Rev Physiol. 2011; 73: 413-35, CrossRef.
Lamouille S, Xu J, Derynck R. Molecular mechanism of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014; 15: 178-96, CrossRef.
Wanami LS, Chen HY, Peiro S, Garcia de Herreros A, Bachelder RE. Vascular endothelial growth factor-A stimulates Snail expression in breast tumor cells: implications for tumor progression. Exp Cell Res. 2008; 314: 2448-53, CrossRef.
Yang AD, Camp ER, Fan F, Shen L, Gray MJ, Liu W, et al. Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in human pancreatic carcinoma cells. Cancer Res. 2006; 66: 46-51, CrossRef.
Peinado H, Marin F, Cubillo E, Stark HJ, Fusenig N, Nieto MA, et al. Snail and E47 repressors of E-cadherin induce distinct invasive and angiogenic properties in vivo. J Cell Sci. 2004; 117: 2827-39, CrossRef.
Zhang J, Oh KH, Xu H, Margetts PJ. Vascular endothelial growth factor expression in peritoneal mesothelial cells undergoing transdifferentiation. Perit Dial Int. 2008; 28: 497-504, PMID.
Knutson KL, Lu H, Stone B, Reiman JM, Behrens MD, Prosperi CM, et al. Immunoediting of cancers may lead to epithelial mesenchymal transition. J Immunol. 2006; 177: 1526-33, CrossRef.
Moody SE, Sarkisian CJ, Hahn KT, Gunther EJ, Pickup S, Dugan KD, et al. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell. 2002; 2: 451-61, CrossRef.
Yang AD, Fan F, Camp ER, van Buren G, Liu W, Somcio R, et al. Chronic oxaliplatin resistance induces epithelial-to-mesenchymal transition in colorectal cancer cell lines. Clin Cancer Res. 2006; 12: 4147-53, CrossRef.
Kajiyama H, Shibata K, Terauchi M, Yamashita M, Ino K, Nawa A, et al. Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int J Oncol. 2007; 31: 277-83, CrossRef.
Cheng GZ, Chan J, Wang Q, Zhang W, Sun CD, Wang LH. Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. Cancer Res. 2007; 67: 1979-87, CrossRef.
Li QQ, Xu JD, Wang WJ, Cao XX, Chen Q, Tang F, et al. Twist1-mediated adriamycin-induced epithelial-mesenchymal transition relates to multidrug resistance and invasive potential in breast cancer cells. Clin Cancer Res. 2009; 15: 2657-65, CrossRef.
Scheel C, Weinberg RA. Cancer stem cells and epithelial-mesenchymal transition: concepts and molecular links. Semin Cancer Biol. 2012; 22: 396-403, CrossRef.
Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, et al. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007; 11: 259-73, CrossRef.
Vermeulen L, De Sousa E Melo F, van der Heijden M, Cameron K, de Jong JH, Borovski T, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010; 12: 468-76, CrossRef.
Brabletz T, Jung A, Hermann K, Günther K, Hohenberger W, Kirchner T, et al. Nuclear overexpression of the oncoprotein β-catenin in colorectal cancer is localized predominantly at the invasion front. Pathol Res Pract. 1998; 194: 701-4, CrossRef.
Espinoza I, Pochampally R, Xing F, Watabe K, Miele L. Notch signaling: targeting cancer stem cells and epithelial-to-mesenchymal transition. Onco Targets Ther. 2013; 6: 1249-59, CrossRef.
Ma J, Xia J, Miele L, Sarkar FH, Wang Z. Notch signaling pathway in pancreatic cancer progression. Pancreat Disord Ther. 2013; 3: pii: 1000114, CrossRef.
Palagani V, El Khatib M, Kossatz U, Bozko P, Müller MR, Manns MP, et al. Epithelial mesenchymal transition and pancreatic tumor initiating CD44+/EpCAM+ cells are inhibited by γ-secretase inhibitor IX. PLoS ONE. 2012; 7: e46514, CrossRef.
Chua KN, Ma J, Thiery JP. Targeted therapies in control of EMT in carcinoma and fibrosis. Drug Discov Today. 2008; 4: 261-7, CrossRef.
Frederick BA, Helfrich BA, Coldren CD, Zheng D, Chan D, Bunn PA Jr, et al. Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma. Mol Cancer Ther. 2007; 6: 1683-91, CrossRef.
Thomson S, Buck E, Petti F, Griffin G, Brown E, Ramnarine N, et al. Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer Res. 2005; 65: 9455-62, CrossRef.
Yauch RL, Januario T, Eberhard DA, Cavet G, Zhu W, Fu L, et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res. 2005; 11: 8686-98, CrossRef.
Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, et al. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell. 2009; 138: 645-59, CrossRef.
Kreso A, O’Brien CA, van Galen P, Gan OI, Notta F, Brown AM, et al. Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer. Science. 2013; 339: 543-8, CrossRef.
Voog J, Jones DL. Stem cells and the niche: a dynamic duo. Cell Stem Cell. 2010; 6: 103-15, CrossRef.
Korkaya H, Liu S, Wicha MS. Breast cancer stem cells, cytokine networks, and the tumor microenvironment. J Clin Invest. 2011; 121: 3804-9, CrossRef.
Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012; 21: 309-22, CrossRef.
Fessler E, Dijkgraaf FE, De Sousa E Melo F, Medema JP. Cancer stem cell dynamics in tumor progression and metastasis: is the microenvironment to blame? Cancer Lett. 2013; 341, 97-104, CrossRef.
Plaks V, Kong N, Werb Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015; 16: 225-38, CrossRef.
Oskarsson T, Batlle E, Massague J. Metastatic stem cells: sources, niches, and vital pathways. Cell Stem Cell. 2014; 14: 306-21, CrossRef.
Chambers AF, Groom AC, MacDonald IC. Metastasis: Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002; 2: 563-72, CrossRef.
Hüsemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E, et al. Systemic spread is an early step in breast cancer. Cancer Cell . 2008; 13: 58-68, CrossRef.
Kouros-Mehr H, Bechis SK, Slorach EM, Littlepage LE, Egeblad M, Ewald AJ, et al. GATA-3 links tumor differentiation and dissemination in a luminal breast cancer model. Cancer Cell. 2008; 13: 141-52, CrossRef.
Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nature Rev Cancer. 2009; 9: 274-84, CrossRef.
Hess KR, Varadhachary GR, Taylor SH, Wei W, Raber MN, Lenzi R, et al. Metastatic patterns in adenocarcinoma. Cancer. 2006; 106: 1624-33, CrossRef.
Malanchi I, Santamaria-Martinez A, Susanto E, Peng H, Lehr HP, Delaloye JF, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011; 481: 85-9, CrossRef.
Cunha GR. Mesenchymal-epithelial interactions: past, present, and future. Differentiation. 2008; 76: 578-86, CrossRef.
Puri S, Hebrok M. Cellular plasticity within the pancreas: lessons learned from development. Dev Cell. 2010; 18: 342-56, CrossRef.
Wiseman BS, Werb Z. Stromal effects on mammary gland development and breast cancer. Science. 2002; 296: 1046-9, CrossRef.
Egeblad M, Ewald AJ, Askautrud HA, Truitt ML, Welm BE, Bainbridge E, et al. Visualizing stromal cell dynamics in different tumor microenvironments by spinning disk confocal microscopy. Dis Model Mech. 2008; 1: 155-67, CrossRef.
Lin EY, Jones JG, Li P, Zhu L, Whitney KD, Muller WJ, et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J Pathol. 2003; 163: 2113-26, CrossRef.
Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009; 139: 891-906, CrossRef.
Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006; 66: 11238-46, CrossRef.
Provenzano PP, Eliceiri KW, Campbell JM, Inman D, White JG, Keely PJ. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med. 2006; 4: 38, CrossRef.
Egeblad M, Nakasone ES, Werb Z. Tumors as organs: compplex tissue that interface with the entire organism. Dev Cell. 2010; 18: 884-901, CrossRef.
Lee CYF, Diehn M. Mechanism of radioresistance in cancer stem cells. In: L Allan AL, editor. Cancer Stem Cells in Solid Tumors, Stem Cell Biology and Regenerative Medicine. New York: Humana Press; 2011. p.345-60, CrossRef.
Bertolini G, Roz L, Perego P, Tortoreto M, Fontanella E, Gatti L, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci USA. 2009; 106: 16281-6, CrossRef.
Cao L, Bombard J, Cintron K, Sheedy J, Weetall ML, Davis TW. BMI1 as a novel target for drug discovery in cancer. J Cell Biochem. 2011; 112: 2729-41, CrossRef.
Crea F, Serrat MAD, Hurt EM, Thomas SB, Danesi R, Farrar WL. BMI1 silencing enhances docetaxel activity and impairs antioxidant response in prostate cancer. Int J Cancer. 2011; 128: 1946-54, CrossRef.
Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994; 367: 645-8, CrossRef.
Huber GF, Albinger-Hegyi A, Soltermann A, Roessle M, Graf N, Haerle SK, et al. Expression patterns of BMI1 and p16 significantly correlate with overall, disease-specific, and recurrence-free survival in oropharyngeal squamous cell carcinoma. Cancer. 2011; 117: 4659-70, CrossRef.
Yin T, Wei H, Gou S, Shi P, Yang Z, Zhao G, et al. Cancer stem-like cells enriched in Panc-1 spheres possess increased migration ability and resistance to gemcitabine. Int J Mol Sci. 2011; 12: 1595-604, CrossRef.
Raaphorst FM. Deregulated expression of Polycomb-group oncogenes in human malignant lymphomas and epithelial tumors. Hum Mol Genet. 2005; 14: R93-100, CrossRef.
Park IK, Morrison SJ, Clarke MF. Bmi1, stem cells, and senescence regulation. J Clin Invest. 2004; 113: 175-9, CrossRef.
Cui H, Hu B, Li T, Ma J, Alam G, Gunning WT, et al. Bmi-1 is essential for the tumorigenicity of neuroblastoma cells. Am J Pathol. 2007; 170: 1370-8, CrossRef.
Mihic-Probst D, Kuster A, Kilgus S, Bode-Lesniewska B, Ingold-Heppner B, Leung C, et al. Consistent expression of the stem cell renewal factor BMI1 in primary and metastatic melanoma. Int J Cancer. 2007; 121: 1764-70, CrossRef.
Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van Lohuizen M. BMI1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev. 1999; 13: 2678-90, CrossRef.
Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, et al. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008; 68: 4311-20, CrossRef.
Wang E, Bhattacharyya S, Szabolcs A, Rodriguez-Aguayo C, Jennings NB, Lopez-Berestein G, et al. Enhancing chemotherapy response with BMI1 silencing in ovarian cancer. PLoS One. 2011; 6: e17918, CrossRef.
Friedman GK, Gillespie GY. Cancer stem cells and pediatric solid tumors. Cancers. 2011; 3: 298-318, CrossRef.
Lessard J, Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003; 423: 255-60, CrossRef.
Crea F, Danesi R, Farrar WL. Cancer stem cell epigenetic and chemoresistance. Epigenome. 2009; 1: 63-79, CrossRef.
Siddique HR, Saleem M. Role of BMI1, a stem cell factor, in cancer recurrence and chemoresistance: Preclinical and clinical evidences. Stem Cells. 2012; 30: 372-8, CrossRef.
Gilbert CA, Ross AH. Cancer stem cells: Cell culture, markers, and targets for new therapies. J Cell Biochem. 2009; 108: 1031-8, CrossRef.
Yuan S, Wang F, Chen G, Zhang H, Feng L, Wang L, et al. Effective elimination of cancer stem cells by a novel drug combination strategy. Stem Cells. 2013; 31: 23-34, CrossRef.
Frank NY, Schatton T, Frank MH. The therapeutic promise of the cancer stem cell concept. J Clin Invest. 2010; 120: 41-50, CrossRef.
Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002; 2: 48-58, CrossRef.
Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005; 5: 275-84, CrossRef.
Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007; 25: 1315-21, CrossRef.
Costello RT, Mallet F, Gaugler B, Sainty D, Arnoulet C, Gastaut JA, et al. Human acute myeloid leukemia CD34+/CD38– progenitor cells have decreased sensitivity to chemotherapy and Fas-induced apoptosis, reduced immunogenicity, and impaired dendritic cell transformation capacities. Cancer Res. 2000; 60: 4403-11, PMID.
de Grouw EP, Raaijmakers MH, Boezeman JB, van der Reijden BA, van de Locht LT, de Witte TJ, et al. Preferential expression of a high number of ATP binding cassette transporters in both normal and leukemic CD34+CD38-cells. Leukemia. 2006; 20: 750-4, CrossRef.
Ito K, Bernardi R, Morotti A, Matsuoka S, Saglio G, Ikeda Y, et al. PML targeting eradicates quiescent leukaemia-initiating cells. Nature. 2008; 453: 1072-8, CrossRef.
Michor F, Hughes TP, Iwasa Y, Branford S, Shah NP, Sawyers CL, et al. Dynamics of chronic myeloid leukaemia. Nature. 2005; 435: 1267-70, CrossRef.
Viale A, De Franco F, Orleth A, Cambiaghi V, Giuliani V, Bossi D, et al. Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells. Nature. 2009; 457: 51-6, CrossRef.
Wulf GG, Wang RY, Kuehnle I, Weidner D, Marini F, Brenner MK, et al. A leukemic stem cell with intrinsic drug efflux capacity in acute myeloid leukemia. Blood. 2001; 98: 1166-73, CrossRef.
Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, et al. Identification of cells initiating human melanomas. Nature. 2008; 451: 345-9, CrossRef.
Frank NY, Margaryan A, Huang Y, Schatton T, Waaga-Gasser AM, Gasser M, et al. ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res. 2005; 65: 4320-33, CrossRef.
Eramo A, Ricci-Vitiani L, Zeuner A, Pallini R, Lotti F, Sette G, et al. Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ. 2006; 13: 1238-41, CrossRef.
Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007; 131: 1109-23, CrossRef.
Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS One. 2008; 3: e2428, CrossRef.
Phillips TM, McBride WH, Pajonk F. The response of CD24(–/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst. 2006; 98: 1777-85, CrossRef.
Saijo H, Hirohashi Y, Torigoe T, Kochin V, Takahashi H, Sato N. Cytotoxic T lymphocytes: the future of cancer stem cell eradication? Immunotherapy. 2013; 5: 549-51, CrossRef.
Pan Q, Li Q, Liu S, Ning N, Zhang X, Xu Y, et al. Concise review: Targeting cancer stem cells using immunologic approaches. Stem Cells. 2015; 33: 2085-92, CrossRef.
Rasheed ZA, Kowalski J, Smith BD, Matsui W. Concise review: Emerging concepts in clinical targeting of cancer stem cells. Stem Cells. 2011; 29: 883-7, CrossRef.
DOI: https://doi.org/10.18585/inabj.v8i1.190
Copyright (c) 2016 The Prodia Education and Research Institute
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
Indexed by:
The Prodia Education and Research Institute