Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

CancerTranslMed3387-6022009_164340.pdf

[Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] CTM Cancer Transl Med 2017;3(3):87–95 doi: 10.4103/ctm.ctm_69_16 Cancer Translational Medicine Mini Review Stemness-related Markers in Cancer Wenxiu Zhao1, Yvonne Li2, Xun Zhang1 1 Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; 2Department of Medical Oncology, Dana-Farber Institute and Harvard Medical School, Boston, MA, USA Address for correspondence: Dr. Xun Zhang, Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, BUL457, Boston, MA 02114, USA. E-mail: xzhang5@mgh.harvard.edu Received December 21, 2016; Accepted March 16, 2017 Cancer stem cells (CSCs), with their self‑renewal ability and multilineage differentiation potential, are a critical subpopulation of tumor cells that can drive tumor initiation, growth, and resistance to therapy. Like embryonic and adult stem cells, CSCs express markers that are not expressed in normal somatic cells and are thus thought to contribute toward a “stemness” phenotype. This review summarizes the current knowledge of stemness‑related markers in human cancers, with a particular focus on important transcription factors, protein surface markers, and signaling pathways. Key words: Cancer stem cells, cell surface stemness markers, stemness-related signaling pathways, stemness-related transcriptional factors INTRODUCTION Individual tumors consist of a mixed cell population that difers in function, morphology, and molecular signatures. hese tumors reside in and interact with their microenvironment, which consists of a wide variety of cell types and cellular structures, such as immune cells, ibroblasts, blood vessels, and extracellular matrix. Tumor cells themselves can be of multiple clonal populations, each having accumulated unique molecular alterations over the course of tumor development and growth. In addition, tumor cells that are similar at genetic level may have distinct modes of epigenetic regulation, further increasing the functional heterogeneity. It has been hypothesized that only a small subset of tumor cells are capable of initiating and sustaining tumor growth; they have been termed cancer stem cells (CSCs).1 To date, CSCs have been isolated from many organs and conirmed to have stem cell‑like abilities such as self‑renewal, multilineage diferentiation, and expression of stemness‑related markers;2,3 some of these features are even conirmed by single‑cell analysis.4 hese cells may also play a role in disease recurrence ater treatment and remission. As such, targeting of CSCs is currently an active area of therapeutic development. CSCs are classiied by the expression of stemness‑related markers, which have been identiied in embryonic stem cells (ESCs) and adult stem cells, the two main types of human stem cells. Here, we summarize the current knowledge about molecular markers and pathways that are not only involved in normal stem‑cell maintenance and self‑renewal but also regulate the stemness of CSCs. Investigation of these features may help elucidate the mechanism of CSC‑driven tumorigenesis and lead to novel approaches for CSC‑targeted cancer therapies. STEMNESS‑RELATED TRANSCRIPTIONAL FACTORS IN CANCERS Takahashi and Yamanaka5 showed in 2006 that pluripotent stem cells could be obtained from mouse embryonic ibroblasts by combined expression of four transcriptional factors (TFs) – now named the Yamanaka factors (OCT4, c‑Myc, SOX2, and KLF4). Induced pluripotent stem cells can now be derived from a wide range of somatic cells through the overexpression of a cocktail of TFs6 or a combination of TF expression with chemical compounds.7,8 Moreover, somatic cells can now be directly reprogrammed into entirely diferent cell types9 through the expression of lineage‑speciic sets of transcription factors. Yamanaka’s seminal discovery has introduced the concept that the fate of adult somatic cells can be controlled through TF expression. From another perspective, expression of stem‑cell‑speciic TFs can provide a signature for characterizing cell type as well as indicating their functional roles. here are currently approximately 25 TFs that have been reported to be expressed in stem cells. Of them, OCT4, SOX2, KLF4, Nanog, and SALL4 comprise a core regulatory network for ESC maintenance and self‑renewal. hese TFs are highly expressed in ESCs; in contrast, they are mainly silenced in normal somatic cells, This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms. For reprints contact: reprints@medknow.com How to cite this article: Zhao W, Li Y, Zhang X. Stemness‑related markers in cancer. Cancer Transl Med 2017;3(3):87‑95. © 2017 Cancer Translational Medicine | Published by Wolters Kluwer - Medknow 87 [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer Transl Med 2017;3(3) except in small groups of adult stem‑cell populations. Increasing evidence has shown that embryonic‑speciic TFs are abnormally expressed in human tumor samples,10,11 suggesting the presence of CSCs. Retrospective studies on patient cohorts have also associated TF expression with survival outcomes in speciic tumor types, suggesting that TF expression levels may also be useful for assessing patient prognosis.12 hus, detecting the expression level of these TFs, for example, by immunohistochemistry staining, can aid in tumor diagnosis, classiication, and therapeutic strategies. A summary of these CSC TFs is shown in Table 1. hese TF markers are also classiied by tissue type as shown in Table 2. A few examples are listed here. OCT4 OCT4 expression has been detected in human brain, lung, bladder, ovarian, prostate, renal, testicular tumors, and leukemia,12 by both reverse transcription‑polymerase chain reaction and immunohistochemistry. Furthermore, high expression of OCT4 has been associated with poor prognosis in bladder cancer,13,14 prostate cancer,15 medulloblastoma,16 and esophageal squamous cell carcinoma (ESCC).17 SOX2 SOX2 has been found in brain, breast, lung, liver, prostate, and testicular tumors,12,18,19 and its expression has been correlated with poor prognosis in stage I lung adenocarcinoma,18 squamous cell carcinoma,20,21 gastric carcinoma,22‑24 small cell lung cancer,25‑28 and ovarian carcinoma.29,30 KFL4 KLF4 has been found to be expressed in brain, breast, head and neck, oral, prostate, and testis tumors, as well as in leukemia and myeloma.12 Expression of KLF4 can also be a prognostic predictor for colon cancer31 and head‑neck squamous cell carcinoma.24,32 In addition, nuclear localization of KLF4 has been associated with the aggressive phenotype of early stage of breast cancer,33 as well as worse prognosis in nasopharyngeal34 and oral cancers.53 Nanog Nanog has been shown to be expressed in brain, breast, prostate, colon, liver, and ovarian tumors.40 High expression of Nanog promotes the epithelial‑mesenchymal transition (EMT),41 which Table 1. Stemness‑related transcriptional factor markers in cancer Marker Other names Function in stem cell Characteristics Expressed in tumor types Poor prognosis for tumor types Selected references OCT4 Oct3/4 or POU5F1 Stem‑cell self‑renew and pluripotency maintenance Oct family of POU transcription factor Leukemia, brain, lung, bladder, ovarian, pancreas, prostate, renal, seminoma, testis Esophageal squamous cell carcinoma Medulloblastoma Prostate cancer Bladder cancer 11–16 SOX2 Stem‑cell self‑renew and pluripotency maintenance POU family binder transcription factor Brain, breast, lung, liver, prostate, seminoma, testis Stage I lung adenocarcinoma Squamous cell carcinoma Gastric carcinoma Small cell lung cancer Ovarian carcinoma 11,17–29 KLF4 Stem‑cell self‑renew and pluripotency maintenance Zinc‑inger transcription factor Leukemia, myeloma, brain, breast, head and neck, oral, prostate, testis Breast cancer Nasopharyngeal carcinoma Colon cancer Head and neck squamous cell carcinoma Oral cancer 27,30–34 C‑MYC Stem‑cell self‑renewal Transcription factor and an oncogene Leukemia, lymphoma, myeloma, brain, breast, colon, head and neck, pancreas, prostate, renal, salivary gland, testis Hepatocellular carcinoma Early carcinoma of uterine cervix 11,35–39 Nanog Stem‑cell self‑renew and pluripotency maintenance Transcription factor Brain, breast, prostate, colon, liver, ovarian Breast cancer Colorectal cancer Gastric adenocarcinoma Nonsmall cell lung cancer Ovarian serous carcinoma Liver cancer 40–48 SALL4 Stem‑cell self‑renew and pluripotency maintenance Diferentiation regulation Zinc‑inger transcription factor and an oncogene Leukemia, breast, liver, colon, ovarian, testis Hepatocellular carcinoma Gliomas Myelodysplastic syndromes 49–52 POU: Pit‑Oct‑Unc 88 CD34 Bladder Breast ALDH1A1 ALDH1A1 ALDH1A1 ALDH1A1 CD47 CD49f/ integrin alpha 6 CD166 CD24 CD24 CD44 CD44 CD133 CD133 CD90 CD26 CD38 CD44 CD44 CD47 CD96 Colon Gastric Glioma/ Head and Liver medulloblastoma neck ALDH1A1 CD49f/integrin alpha 6 TNFRSF16 CD27/TNFRSF7 CD24 CD38 CD44 CD44 CD133 CD133 CD15/ Lewis X CD15/Lewis X CD13 CD90/hy1 CD90/hy1 CD44 CD133 Lgr5/ GPR49 EpCAM/ TROP1 BMI‑1 Lgr5/ GPR49 c‑Myc EpCAM/ TROP1 CXCR1/ IL‑8 RA CX3CR1 c‑Myc SALL4 CD24 CD151 CD44 CD44 CD133 CD133 CD133 CD20/MS4A1 CD166 CD19 TRA‑1‑60 (R) CD138Syndecan‑1 ALCAM/ CD166 EpCAM/ TROP1 CXCR4 BMI‑1 EpCAM/ TROP1 CXCR4 BMI‑1 Musashi‑1 Musashi‑1 c‑Myc c‑Myc SOX2 CD24 CD44 Lgr5/ GPR49 CXCR4 BMI‑1 CD49f/ integrin alpha 6 CD117/ c‑kit CD166/ ALCAM BMI‑1 Nestin TIM3 CD133 CD90/ hy1 Aminopeptidase N/ CD13 Pancreatic Prostate Endoglin/ CD24 CD105 CD117/ c‑kit CD123/IL‑3 CEACAM‑6/ R alpha CD66c KLF4 Osteosarcoma Ovarian ALDH1A1 ALDH1A1 CD45 CD117/c‑kit OCT4 Melanoma Myeloma ALDH1A1 CD44 CD29 Lung SOX2 Nestin Nestin c‑Myc SOX2 OCT4 BMI‑1 Nestin SOX2 c‑Myc SOX2 OCT4 KLF4 OCT4 OCT4 KLF4 Nanog Nanog Nanog Nanog Nanog SALL4 SALL4 SALL4 SALL4 SALL4 Rex1 [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Leukemia Cancer stemness markers Table 2. Stemness‑related markers in diferent cancer types 89 [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer Transl Med 2017;3(3) is an important developmental process for cancer cells to obtain stem‑cell characteristics. Nanog has also been associated with poor prognosis in breast,42 colorectal,43,44 gastric,45 lung,46,47 ovarian,48 and liver cancers.54 SALL4 SALL4 expression has been detected in breast, liver, colon, ovarian, and testis cancers and leukemia.49,55 he expression of SALL4 has been studied as a poor prognosis marker in hepatocellular carcinoma,50,51 gliomas,52 and myelodysplastic syndromes.35 C‑Myc C‑Myc is an important TF both in stem cells and cancers. As one of the most studied oncogenes, overexpression of C‑Myc has been shown to cause tumorigenesis in mouse models. Up to 70% of human cancers exhibit c‑Myc overexpression, including brain, breast, colon, head and neck, pancreas, prostate, renal, salivary gland, and testis tumors, as well as leukemia and lymphoma.12,36,37 C‑Myc expression has also been correlated with poor prognosis in hepatocellular carcinoma38 and early carcinoma of uterine cervix.39,56 STEMNESS‑RELATED SURFACE MARKERS IN CANCERS Cell surface proteins provide a feasible way for isolating and studying diferent cell types by low cytometry or magnetic sorting. In addition, they are amenable for speciic targeting, which is useful for disease monitoring and therapeutic delivery. Similar to stemness‑related transcription factors, many surface markers that are highly expressed in stem cells are also expressed in human cancers as TRA‑1‑60, SSEA‑1, EpCAM, ALDH1A1, Lgr5, CD13, CD19, CD20, CD24, CD26, CD27, CD34, CD38, CD44, CD45, CD47, CD49f, CD66c, CD90, CD166, TNFRSF16, CD105, CD133, CD117/c‑kit, CD138, CD151, and CD166. Table 2 describes most of the stemness‑related surface markers and the tumor types they have been found to be expressed in. Among them, CD44 and CD133 are the most widely used markers in CSC research and are also therapeutic targets in cancers. CD44 is a transmembrane glycoprotein that plays diferent roles in cell division, migration, adhesion, and signaling.57 It is normally expressed in both fetal and adult hematopoietic stem cells, and on binding to hyaluronic acid, its primary ligand, CD44 mediates cell‑cell communication and signal transaction. CD44 is highly expressed in many types of cancers including bladder, breast, colon, gastric, glioma, head and neck, osteosarcoma, ovarian, pancreatic, and prostate cancers, as well as leukemia.58,59 CD44 is being studied as a therapeutic target in metastasizing tumors such as breast and colon cancer60,61 and also in leukemia.62 CD133 is another transmembrane glycoprotein and speciically localizes to cellular protrusions. CD133 is reported to be expressed in hematopoietic stem cells, endothelial progenitor cells, glioblastoma, and neuronal and glial stem cells,63,64 and it is also involved in cell growth and development.65 Almost all tumor types can be detected with CD133 expression, and CD133+ tumor cells show stem‑cell‑speciic characteristics such as self‑renewal, diferentiation, and tumor formation in NOD‑SCID mouse model.66 Ater injection into immune‑compromised mice, 90 CD133+ cells also show chemo‑ and radio‑resistance.66 Studies have been performed to use CD133 as a potential therapeutic target in colon cancer,67 ovary cancers,68 and metastatic melanoma.69 CD133 has also been used as a target for drug delivery.70 here are a number of other CSC surface markers that appear to function in speciic types of tumors. For examples, SSEA‑1 has been shown to be expressed in human colonic adenocarcinoma and glioblastoma.71,72 Similarly, TRA‑1‑60 has been associated with prostate tumors.73 Lgr5 has been shown to be expressed in head and neck, colon, and gastric tumors.74,75 CD90 has been detected in high‑grade human glioma,76,77 as well as liver 78 and lung tumors;79,80 while CD117 has been used as a CSC marker in leukemia81,82 and gastrointestinal stromal tumor,83 as well as oral squamous cell carcinomas84,85 and ovarian tumors.86,87 CD117 has been shown to be overexpressed in hepatocellular88 and pancreatic carcinoma.89 CD24 has been used in combination with CD44 in breast cancer cell lines to show that CD44+/CD24‑ cancer cells exhibit drug resistance and invasive properties.90‑92 Studies have also shown that CD24 can be used as an independent prognostic marker nonsmall cell lung cancer93,94 and ovarian cancer.95 OTHER IMPORTANT STEMNESS‑RELATED MARKERS here are a number of stemness‑related markers that are neither TFs nor cell surface proteins, which include aldehyde dehydrogenase (ALDH), Bmi‑1, Nestin, Musashi‑1, TIM‑3, and CXCR. he ubiquitous family of ALDH enzymes catalyzes the irreversible oxidation of cellular aldehydes in the cytoplasm. High activity of ALDH enzymes has been found in ESCs, adult hematopoietic and neural stem cells, as well as CSCs. ALDH activity in CSCs has been attributed to ALDH1A1 expression, which can regulate stem‑cell self‑protection, diferentiation, and population expansion. ALDH has been reported to have prognostic signiicance in head and neck squamous cell96 and ESCC.97 It is also being pursued as a therapeutic target in ovarian98,99 and nonsmall cell lung cancers.100 BMI1 is a protein required for hematopoietic stem‑cell self‑renewal101 and neural stem cells.102 Drug‑induced expression of BMI1 has been shown to enhance stem‑cell populations in head and neck cancer models.103 BMI1 has been reported as a marker for poor prognosis in oligodendroglial tumors104 and breast cancer.105,106 Nestin and Musashi‑1 have been detected in neural stem cells,107 where they both play an important role in stem‑cell self‑renewal and maintenance. Nestin expression has been shown in transformed cells of various human malignancies, correlating with the clinical course of some diseases.108 Furthermore, coexpression of Nestin with other stem‑cell markers was described as a CSC phenotype.109 Nestin was reported as a potential target for tumor angiogenesis.110,111 Musashi‑1 signaling was also detected in hematopoietic stem cells, and it is being investigated as a potential therapeutic target and diagnostic marker for lung cancer.112 Chemokines are small peptide molecules secreted by cells that afect the movement of neighboring cells, thus mediating cellular homing and migration. hey are crucial for normal physiological functions and are found to be dysregulated in cancers. he chemokine CXCL12 (SDF‑1) and its receptor CXCR4 regulate cellular chemotaxis, cell adhesion, survival, proliferation, and gene transcription through multiple divergent pathways. CXCL12/ CXCR4 interactions were shown to play an important role in the [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer stemness markers migration of hematopoietic stem cells.113 CXCR4 is overexpressed in more than twenty cancer types, with discovered roles in tumor growth, invasion, angiogenesis, metastasis, relapse, and therapeutic resistance.114 CXCR4 antagonists have been shown to disrupt tumor‑stromal cell interactions, sensitize cancer cells to cytotoxic drugs, and reduce tumor growth and metastasis. herefore, CXCR4 is considered as a target for therapeutic intervention of lung115,116 and breast cancer.117,118 It has also been used for noninvasive monitoring of disease progression and therapeutic guidance.114 STEMNESS‑RELATED PATHWAYS Stem‑cell maintenance, self‑renewal, and diferentiation pathways are involved in embryonic development and adult tissue homeostasis. Cancers commonly display aberrant activities within these pathways, oten in a cell‑context‑dependent manner. Here, we discuss current evidence for Hedgehog (HH), Notch, JAK/ STAT, phosphatidylinositol‑3‑kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), and Wnt/β‑catenin pathway regulation in CSCs. Hedgehog pathway he HH pathway is a major regulator in vertebrate embryonic development, playing critical roles in stem‑cell maintenance, cell diferentiation, tissue polarity, and cell proliferation, as well as EMT.119 HH ligands (Desert HH, Sonic HH, and Indian HH) bind to Ptch, activating a cascade of downstream signals that lead to the activation and nuclear localization of TFs, consequently followed by expression of genes that are involved in survival, proliferation, and angiogenesis.120 HH signaling has been widely implicated in CSC self‑renewal and cell fate determination120 and is considered a potential therapeutic target in breast cancer and pancreatic cancer.121‑123 control of stem‑cell maintenance in the male germline stem‑cell microenvironment.130,131 Tightly controlled JAK‑STAT signaling is required for stem‑cell maintenance and self‑renewal. Furthermore, JAK‑STAT activity is essential for anchoring the stem cells in their respective niches by regulating diferent adhesion molecules. Phosphatidylinositol‑3‑kinase/Akt/mammalian rapamycin pathway target of he PI3K/Akt and the mTOR signaling pathways are crucial to stem‑cell proliferation, metabolism, and diferentiation. his pathway is oten improperly regulated in human cancers.132 Over 70% of ovarian cancers have active PI3K/Akt/mTOR pathway, making it a therapeutic target in this cancer type.133,134 It is also a therapeutic target for neuroblastoma,135 endometrial cancer,136 and acute myeloid leukemia.137 Wnt/β‑catenin pathway Pathways induced by Wnt ligands are highly evolutionarily conserved. Given their strong conservation in phylogeny, it is not surprising that Wnt pathways also play key roles in regulating stem‑cell diferentiation and pluripotency. Consistently in many tissue types, dysregulation of Wnt pathway has been strongly associated with expansion of stem and/or progenitor cell lineages, as well as carcinogenesis.138 Hence, therapies targeting Wnt pathway may lead to treatment options in hematological malignancies,139 liver cancer,140 and other type of tumors.141 CONCLUSION Notch signaling is a critical part of stem‑cell fate determination and angiogenesis. Notch signaling is predominantly involved in cell‑cell communication between adjacent cells through transmembrane receptors and ligands. In human ESCs, Notch signaling governs cell fate determination in the developing embryo and is required for undiferentiated ESCs to develop all three embryonic germ layers.124 In CSCs, it controls tumor immunity and CSC population maintenance.125,126 Notch signaling is frequently dysregulated in cancers, providing a survival advantage for tumors. In certain tumor types, activation of Notch signaling aids CSCs in maintaining their population in tumors, inducing EMT, and acquiring chemoresistance.127 Notch signaling is a potential target for cancers.128,129 A primary goal of cancer research is to identify mechanisms driving drug resistance, and recent studies have implicated CSCs in intrinsic resistance models. Similar to normal stem cells, the abilities of self‑renewal, maintenance, and diferentiation of CSCs make it serve as a core reservoir for cancer initiation, development, and growth. he overexpression of stem‑cell‑speciic TFs may contribute to the pathologic self‑renewal characteristics of CSCs while the surface molecules mediate interactions between cells and their microenvironment. Other stemness‑related markers and pathways may promote cancer cell proliferation, progression, and metastasis. Our summary of stem cell markers by tissue types and cellular locations in Table 2 and Figure 1 highlights the complex nature of CSC regulation, which appears to utilize diferent pathways in diferent cell or tissue types. his context dependency makes it hard to ind overarching CSC pathways and makers. Understanding the stemness‑related features in cancers will not only provide important knowledge on molecular mechanisms for cancer pathogenesis but also shed new light on the development of efective therapeutic approaches, speciically targeting these stemness‑related features. JAK/STAT pathway Financial support and sponsorship he JAK‑STAT signaling pathway is important in cytokine‑mediated immune responses and known to be involved in many biological processes such as proliferation, apoptosis, and migration, as well as the regulation of stem cells. Cancer cells also show frequent dysregulation of the JAK/STAT. Studies in Drosophila irst implicated JAK‑STAT signaling in the his work was supported in part by NIH (R01CA193520‑01A1) and the Jarislowsky Foundation. Notch pathway Grant Conlicts of interest here are no conlicts of interest. 91 [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer Transl Med 2017;3(3) 17. 18. 19. 20. Figure 1. Categories of cancer stem‑cell markers 21. REFERENCES 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 92 Pardal R, Clarke MF, Morrison SJ. Applying the principles of stem‑cell biology to cancer. Nat Rev Cancer 2003; 3 (12): 895–902. Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol 2013; 15 (4): 338–44. Pattabiraman DR, Weinberg RA. Tackling the cancer stem cells – What challenges do they pose? Nat Rev Drug Discov 2014; 13 (7): 497–512. Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, Cahill DP, Nahed BV, Curry WT, Martuza RL, Louis DN, Rozenblatt‑Rosen O, Suvà ML, Regev A, Bernstein BE. Single‑cell RNA‑seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014; 344 (6190): 1396–401. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult ibroblast cultures by deined factors. Cell 2006; 126 (4): 663–76. Radzisheuskaya A, Silva JC. Do all roads lead to Oct4? he emerging concepts of induced pluripotency. Trends Cell Biol 2014; 24 (5): 275–84. Zhu S, Li W, Zhou H, Wei W, Ambasudhan R, Lin T, Kim J, Zhang K, Ding S. Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell 2010; 7 (6): 651–5. Chen G, Gulbranson DR, Hou Z, Bolin JM, Ruotti V, Probasco MD, Smuga‑Otto K, Howden SE, Diol NR, Propson NE, Wagner R, Lee GO, Antosiewicz‑Bourget J, Teng JM, homson JA. Chemically deined conditions for human iPSC derivation and culture. Nat Methods 2011; 8 (5): 424–9. Kelaini S, Cochrane A, Margariti A. Direct reprogramming of adult cells: avoiding the pluripotent state. Stem Cells Cloning 2014; 7: 19–29. Monk M, Holding C. Human embryonic genes re‑expressed in cancer cells. Oncogene 2001; 20 (56): 8085–91. Zhao W, Ji X, Zhang F, Li L, Ma L. Embryonic stem cell markers. Molecules 2012; 17 (6): 6196–236. Schoenhals M, Kassambara A, De Vos J, Hose D, Moreaux J, Klein B. Embryonic stem cell markers expression in cancers. Biochem Biophys Res Commun 2009; 383 (2): 157–62. Xu K, Zhu Z, Zeng F. Expression and signiicance of Oct4 in bladder cancer. J Huazhong Univ Sci Technolog Med Sci 2007; 27 (6): 675–7. Hatei N, Nouraee N, Parvin M, Ziaee SA, Mowla SJ. Evaluating the expression of oct4 as a prognostic tumor marker in bladder cancer. Iran J Basic Med Sci 2012; 15 (6): 1154–61. de Resende MF, Chinen LT, Vieira S, Jampietro J, da Fonseca FP, Vassallo J, Campos LC, Guimarães GC, Soares FA, Rocha RM. Prognostication of OCT4 isoform expression in prostate cancer. Tumour Biol 2013; 34 (5): 2665–73. Rodini CO, Suzuki DE, Saba‑Silva N, Cappellano A, de Souza JE, Cavalheiro S, Toledo SR, Okamoto OK. Expression analysis of 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. stem cell‑related genes reveal OCT4 as a predictor of poor clinical outcome in medulloblastoma. J Neurooncol 2012; 106 (1): 71–9. Li C, Yan Y, Ji W, Bao L, Qian H, Chen L, Wu M, Chen H, Li Z, Su C. OCT4 positively regulates survivin expression to promote cancer cell proliferation and leads to poor prognosis in esophageal squamous cell carcinoma. PLoS One 2012; 7 (11): e49693. Gillis AJ, Stoop H, Biermann K, van Gurp RJ, Swartzman E, Cribbes S, Ferlinz A, Shannon M, Oosterhuis JW, Looijenga LH. Expression and interdependencies of pluripotency factors LIN28, OCT3/4, NANOG and SOX2 in human testicular germ cells and tumours of the testis. Int J Androl 2011; 34 (4 Pt 2): e160–74. Leis O, Eguiara A, Lopez‑Arribillaga E, Alberdi MJ, Hernandez‑Garcia S, Elorriaga K, Pandiella A, Rezola R, Martin AG. Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene 2012; 31 (11): 1354–65. Wang Q, He W, Lu C, Wang Z, Wang J, Giercksky KE, Nesland JM, Suo Z. Oct3/4 and Sox2 are signiicantly associated with an unfavorable clinical outcome in human esophageal squamous cell carcinoma. Anticancer Res 2009; 29 (4): 1233–41. Forghanifard MM, Ardalan Khales S, Javdani‑Mallak A, Rad A, Farshchian M, Abbaszadegan MR. Stemness state regulators SALL4 and SOX2 are involved in progression and invasiveness of esophageal squamous cell carcinoma. Med Oncol 2014; 31 (4): 922. Li XL, Eishi Y, Bai YQ, Sakai H, Akiyama Y, Tani M, Takizawa T, Koike M, Yuasa Y. Expression of the SRY‑related HMG box protein SOX2 in human gastric carcinoma. Int J Oncol 2004; 24 (2): 257–63. Zhang X, Yu H, Yang Y, Zhu R, Bai J, Peng Z, He Y, Chen L, Chen W, Fang D, Bian X, Wang R. SOX2 in gastric carcinoma, but not Hath1, is related to patients’ clinicopathological features and prognosis. J Gastrointest Surg 2010; 14 (8): 1220–6. Matsuoka J, Yashiro M, Sakurai K, Kubo N, Tanaka H, Muguruma K, Sawada T, Ohira M, Hirakawa K. Role of the stemness factors sox2, oct3/4, and nanog in gastric carcinoma. J Surg Res 2012; 174 (1): 130–5. Li X, Wang J, Xu Z, Ahmad A, Li E, Wang Y, Qin S, Wang Q. Expression of Sox2 and Oct4 and their clinical signiicance in human non‑small‑cell lung cancer. Int J Mol Sci 2012; 13 (6): 7663–75. Chen Y, Huang Y, Huang Y, Chen J, Wang S, Zhou J. he prognostic value of SOX2 expression in non‑small cell lung cancer: a meta‑analysis. PLoS One 2013; 8 (8): e71140. Inoue Y, Matsuura S, Kurabe N, Kahyo T, Mori H, Kawase A, Karayama M, Inui N, Funai K, Shinmura K, Suda T, Sugimura H. Clinicopathological and survival analysis of Japanese patients with resected non‑small‑cell lung cancer harboring NKX2‑1, SETDB1, MET, HER2, SOX2, FGFR1, or PIK3CA gene ampliication. J horac Oncol 2015; 10 (11): 1590–600. Sodja E, Rijavec M, Koren A, Sadikov A, Korošec P, Cufer T. he prognostic value of whole blood SOX2, NANOG and OCT4 mRNA expression in advanced small‑cell lung cancer. Radiol Oncol 2016; 50 (2): 188–96. Ye F, Li Y, Hu Y, Zhou C, Hu Y, Chen H. Expression of Sox2 in human ovarian epithelial carcinoma. J Cancer Res Clin Oncol 2011; 137 (1): 131–7. Pham DL, Scheble V, Bareiss P, Fischer A, Beschorner C, Adam A, Bachmann C, Neubauer H, Boesmueller H, Kanz L, Wallwiener D, Fend F, Lengerke C, Perner S, Fehm T, Staebler A. SOX2 expression and prognostic signiicance in ovarian carcinoma. Int J Gynecol Pathol 2013; 32 (4): 358–67. Patel NV, Ghaleb AM, Nandan MO, Yang VW. Expression of the tumor suppressor Kruppel‑like factor 4 as a prognostic predictor for colon cancer. Cancer Epidemiol Biomarkers Prev 2010; 19 (10): 2631–8. Tai SK, Yang MH, Chang SY, Chang YC, Li WY, Tsai TL, Wang YF, Chu PY, Hsieh SL. Persistent Kruppel‑like factor 4 expression predicts progression and poor prognosis of head and neck squamous cell carcinoma. Cancer Sci 2011; 102 (4): 895–902. Pandya AY, Talley LI, Frost AR, Fitzgerald TJ, Trivedi V, Chakravarthy M, Chhieng DC, Grizzle WE, Engler JA, Krontiras H, [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer stemness markers 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. Bland KI, LoBuglio AF, Lobo‑Ruppert SM, Ruppert JM. Nuclear localization of KLF4 is associated with an aggressive phenotype in early‑stage breast cancer. Clin Cancer Res 2004; 10 (8): 2709–19. Liu Z, Yang H, Luo W, Jiang Q, Mai C, Chen Y, Zhen Y, Yu X, Long X, Fang W. Loss of cytoplasmic KLF4 expression is correlated with the progression and poor prognosis of nasopharyngeal carcinoma. Histopathology 2013; 63 (3): 362–70. Wang F, Guo Y, Chen Q, Yang Z, Ning N, Zhang Y, Xu Y, Xu X, Tong C, Chai L, Cui W. Stem cell factor SALL4, a potential prognostic marker for myelodysplastic syndromes. J Hematol Oncol 2013; 6 (1): 73. Kim J, Orkin SH. Embryonic stem cell‑speciic signatures in cancer: insights into genomic regulatory networks and implications for medicine. Genome Med 2011; 3 (11): 75. Dang CV. MYC on the path to cancer. Cell 2012; 149 (1): 22–35. Wang Y, Wu MC, Sham JS, Zhang W, Wu WQ, Guan XY. Prognostic signiicance of c‑myc and AIB1 ampliication in hepatocellular carcinoma. A broad survey using high‑throughput tissue microarray. Cancer 2002; 95 (11): 2346–52. Riou G, Barrois M, Lê MG, George M, Le Doussal V, Haie C. C‑myc proto‑oncogene expression and prognosis in early carcinoma of the uterine cervix. Lancet 1987; 1 (8536): 761–3. Hart AH, Hartley L, Parker K, Ibrahim M, Looijenga LH, Pauchnik M, Chow CW, Robb L. he pluripotency homeobox gene NANOG is expressed in human germ cell tumors. Cancer 2005; 104 (10): 2092–8. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA. he epithelial‑mesenchymal transition generates cells with properties of stem cells. Cell 2008; 133 (4): 704–15. Bareiss PM, Paczulla A, Wang H, Schairer R, Wiehr S, Kohlhofer U, Rothfuss OC, Fischer A, Perner S, Staebler A, Wallwiener D, Fend F, Fehm T, Pichler B, Kanz L, Quintanilla‑Martinez L, Schulze‑Osthof K, Essmann F, Lengerke C. SOX2 expression associates with stem cell state in human ovarian carcinoma. Cancer Res 2013; 73 (17): 5544–55. Meng HM, Zheng P, Wang XY, Liu C, Sui HM, Wu SJ, Zhou J, Ding YQ, Li J. Over‑expression of Nanog predicts tumor progression and poor prognosis in colorectal cancer. Cancer Biol her 2010; 9 (4): 295–302. Xu F, Dai C, Zhang R, Zhao Y, Peng S, Jia C. Nanog: a potential biomarker for liver metastasis of colorectal cancer. Dig Dis Sci 2012; 57 (9): 2340–6. Lin T, Ding YQ, Li JM. Overexpression of Nanog protein is associated with poor prognosis in gastric adenocarcinoma. Med Oncol 2012; 29 (2): 878–85. Gialmanidis IP, Bravou V, Petrou I, Kourea H, Mathioudakis A, Lilis I, Papadaki H. Expression of Bmi1, FoxF1, Nanog, and gamma‑catenin in relation to hedgehog signaling pathway in human non‑small‑cell lung cancer. Lung 2013; 191 (5): 511–21. Li XQ, Yang XL, Zhang G, Wu SP, Deng XB, Xiao SJ, Liu QZ, Yao KT, Xiao GH. Nuclear beta‑catenin accumulation is associated with increased expression of Nanog protein and predicts poor prognosis of non‑small cell lung cancer. J Transl Med 2013; 11: 114. Lee M, Nam EJ, Kim SW, Kim S, Kim JH, Kim YT. Prognostic impact of the cancer stem cell‑related marker NANOG in ovarian serous carcinoma. Int J Gynecol Cancer 2012; 22 (9): 1489–96. Wang F, Zhao W, Kong N, Cui W, Chai L. he next new target in leukemia: the embryonic stem cell gene SALL4. Mol Cell Oncol 2014; 1 (4): e969169. Oikawa T, Kamiya A, Zeniya M, Chikada H, Hyuck AD, Yamazaki Y, Wauthier E, Tajiri H, Miller LD, Wang XW, Reid LM, Nakauchi H. Sal‑like protein 4 (SALL4), a stem cell biomarker in liver cancers. Hepatology 2013; 57 (4): 1469–83. Masuda S, Suzuki K, Izpisua Belmonte JC. Oncofetal gene SALL4 in aggressive hepatocellular carcinoma. N Engl J Med 2013; 369 (12): 1171–2. Zhang L, Yan Y, Jiang Y, Cui Y, Zou Y, Qian J, Luo C, Lu Y, Wu X. he 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. expression of SALL4 in patients with gliomas: high level of SALL4 expression is correlated with poor outcome. J Neurooncol 2015; 121 (2): 261–8. Chen CJ, Hsu LS, Lin SH, Chen MK, Wang HK, Hsu JD, Lee H, Yeh KT. Loss of nuclear expression of Kruppel‑like factor 4 is associated with poor prognosis in patients with oral cancer. Hum Pathol 2012; 43 (7): 1119–25. Sun C, Sun L, Jiang K, Gao DM, Kang XN, Wang C, Zhang S, Huang S, Qin X, Li Y, Liu YK. NANOG promotes liver cancer cell invasion by inducing epithelial‑mesenchymal transition through NODAL/SMAD3 signaling pathway. Int J Biochem Cell Biol 2013; 45 (6): 1099–108. Chen Q, Qian J, Lin J, Yang J, Li Y, Wang CZ, Chai HY, Chen XX, Qian Z, Ma JC, Zhang M. Expression of SALL4 gene in patients with acute and chronic myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2013; 21 (2): 315–9. (in Chinese) Colombo N, Carinelli S, Colombo A, Marini C, Rollo D, Sessa C; ESMO Guidelines Working Group. Cervical cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‑up. Ann Oncol 2012; 23 Suppl 7: vii27–32. Basakran NS. CD44 as a potential diagnostic tumor marker. Saudi Med J 2015; 36 (3): 273–9. Joshua B, Kaplan MJ, Doweck I, Pai R, Weissman IL, Prince ME, Ailles LE. Frequency of cells expressing CD44, a head and neck cancer stem cell marker: correlation with tumor aggressiveness. Head Neck 2012; 34 (1): 42–9. Palapattu GS, Wu C, Silvers CR, Martin HB, Williams K, Salamone L, Bushnell T, Huang LS, Yang Q, Huang J. Selective expression of CD44, a putative prostate cancer stem cell marker, in neuroendocrine tumor cells of human prostate cancer. Prostate 2009; 69 (7): 787–98. Orian‑Rousseau V. CD44, a therapeutic target for metastasising tumours. Eur J Cancer 2010; 46 (7): 1271–7. Arabi L, Badiee A, Mosafa F, Jaafari MR. Targeting CD44 expressing cancer cells with anti‑CD44 monoclonal antibody improves cellular uptake and antitumor eicacy of liposomal doxorubicin. J Control Release 2015; 220(Pt A): 275–86. Zhang S, Wu CC, Fecteau JF, Cui B, Chen L, Zhang L, Wu R, Rassenti L, Lao F, Weigand S, Kipps TJ. Targeting chronic lymphocytic leukemia cells with a humanized monoclonal antibody speciic for CD44. Proc Natl Acad Sci U S A 2013; 110 (15): 6127–32. Handgretinger R, Gordon PR, Leimig T, Chen X, Buhring HJ, Niethammer D, Kuci S. Biology and plasticity of CD133+ hematopoietic stem cells. Ann N Y Acad Sci 2003; 996: 141–51. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identiication of a cancer stem cell in human brain tumors. Cancer Res 2003; 63 (18): 5821–8. Li Z. CD133: a stem cell biomarker and beyond. Exp Hematol Oncol 2013; 2 (1): 17. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identiication of human brain tumour initiating cells. Nature 2004; 432 (7015): 396–401. Catalano V, Di Franco S, Iovino F, Dieli F, Stassi G, Todaro M. CD133 as a target for colon cancer. Expert Opin her Targets 2012; 16 (3): 259–67. Skubitz AP, Taras EP, Boylan KL, Waldron NN, Oh S, Panoskaltsis‑Mortari A, Vallera DA. Targeting CD133 in an in vivo ovarian cancer model reduces ovarian cancer progression. Gynecol Oncol 2013; 130 (3): 579–87. Rappa G, Fodstad O, Lorico A. he stem cell‑associated antigen CD133 (Prominin‑1) is a molecular therapeutic target for metastatic melanoma. Stem Cells 2008; 26 (12): 3008–17. Smith LM, Nesterova A, Ryan MC, Duniho S, Jonas M, Anderson M, Zabinski RF, Sutherland MK, Gerber HP, Van Orden KL, Moore PA, Ruben SM, Carter PJ. CD133/prominin‑1 is a potential therapeutic target for antibody‑drug conjugates in hepatocellular and gastric cancers. Br J Cancer 2008; 99 (1): 100–9. Mao XG, Zhang X, Xue XY, Guo G, Wang P, Zhang W, Fei Z, Zhen HN, You SW, Yang H. Brain tumor stem‑like cells identiied 93 [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer Transl Med 2017;3(3) 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 94 by neural stem cell marker CD15. Transl Oncol 2009; 2 (4): 247–57. Lin W, Modiano JF, Ito D. Stage‑speciic embryonic antigen (SSEA): determining the expression in canine glioblastoma, melanoma, and mammary cancer cells. J Vet Sci 2017; 18 (1): 101–4. Giwercman A, Andrews PW, Jørgensen N, Müller J, Graem N, Skakkebaek NE. Immunohistochemical expression of embryonal marker TRA‑1‑60 in carcinoma in situ and germ cell tumors of the testis. Cancer 1993; 72 (4): 1308–14. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, Clevers H. Identiication of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007; 449 (7165): 1003–7. Simon E, Petke D, Böger C, Behrens HM, Warneke V, Ebert M, Röcken C. he spatial distribution of LGR5+ cells correlates with gastric cancer progression. PLoS One 2012; 7 (4): e35486. He J, Liu Y, Zhu T, Zhu J, Dimeco F, Vescovi AL, Heth JA, Muraszko KM, Fan X, Lubman DM. CD90 is identiied as a candidate marker for cancer stem cells in primary high‑grade gliomas using tissue microarrays. Mol Cell Proteomics 2012; 11 (6): M111.010744. Parry PV, Engh JA. CD90 is identiied as a marker for cancer stem cells in high‑grade gliomas using tissue microarrays. Neurosurgery 2012; 70 (4): N23–4. Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PW, Lam CT, Poon RT, Fan ST. Signiicance of CD90+ cancer stem cells in human liver cancer. Cancer Cell 2008; 13 (2): 153–66. Kawamura K, Hiroshima K, Suzuki T, Chai K, Yamaguchi N, Shingyoji M, Yusa T, Tada Y, Takiguchi Y, Tatsumi K, Shimada H, Tagawa M. CD90 is a diagnostic marker to diferentiate between malignant pleural mesothelioma and lung carcinoma with immunohistochemistry. Am J Clin Pathol 2013; 140 (4): 544–9. Yan X, Luo H, Zhou X, Zhu B, Wang Y, Bian X. Identiication of CD90 as a marker for lung cancer stem cells in A549 and H446 cell lines. Oncol Rep 2013; 30 (6): 2733–40. Auewarakul CU, Lauhakirti D, Promsuwicha O, Munkhetvit C. C‑kit receptor tyrosine kinase (CD117) expression and its positive predictive value for the diagnosis of hai adult acute myeloid leukemia. Ann Hematol 2006; 85 (2): 108–12. Eren R, Aslan C, Yokuş O, Doğu MH, Suyani E. T‑cell acute lymphoblastic leukemia with co‑expression of CD56, CD34, CD117 and CD33: A case with poor prognosis. Mol Clin Oncol 2016; 5 (2): 331–2. Sarlomo‑Rikala M, Kovatich AJ, Barusevicius A, Miettinen M. CD117: A sensitive marker for gastrointestinal stromal tumors that is more speciic than CD34. Mod Pathol 1998; 11 (8): 728–34. Barth PJ, Schenck zu Schweinsberg T, Ramaswamy A, Moll R. CD34+ ibrocytes, alpha‑smooth muscle antigen‑positive myoibroblasts, and CD117 expression in the stroma of invasive squamous cell carcinomas of the oral cavity, pharynx, and larynx. Virchows Arch 2004; 444 (3): 231–4. Mărgăritescu C, Pirici D, Simionescu C, Stepan A. he utility of CD44, CD117 and CD133 in identiication of cancer stem cells (CSC) in oral squamous cell carcinomas (OSCC). Rom J Morphol Embryol 2011; 52(3 Suppl): 985–93. Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH, Nephew KP. Identiication and characterization of ovarian cancer‑initiating cells from primary human tumors. Cancer Res 2008; 68 (11): 4311–20. Luo L, Zeng J, Liang B, Zhao Z, Sun L, Cao D, Yang J, Shen K. Ovarian cancer cells with the CD117 phenotype are highly tumorigenic and are related to chemotherapy outcome. Exp Mol Pathol 2011; 91 (2): 596–602. Becker G, Schmitt‑Graef A, Ertelt V, Blum HE, Allgaier HP. CD117 (c‑kit) expression in human hepatocellular carcinoma. Clin Oncol (R Coll Radiol) 2007; 19 (3): 204–8. Potti A, Ganti AK, Tendulkar K, Chitajallu S, Sholes K, Koch M, Kargas S. HER‑2/neu and CD117 (C‑kit) overexpression in hepatocellular and pancreatic carcinoma. Anticancer Res 2003; 23 (3B): 2671–4. Sheridan C, Kishimoto H, Fuchs RK, Mehrotra S, Bhat‑Nakshatri P, 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. Turner CH, Goulet R Jr., Badve S, Nakshatri H. CD44+/CD24‑ breast cancer cells exhibit enhanced invasive properties: an early step necessary for metastasis. Breast Cancer Res 2006; 8 (5): R59. Giatromanolaki A, Sivridis E, Fiska A, Koukourakis MI. he CD44+/CD24‑ phenotype relates to ‘triple‑negative’ state and unfavorable prognosis in breast cancer patients. Med Oncol 2011; 28 (3): 745–52. Adamczyk A, Niemiec JA, Ambicka A, Mucha‑Małecka A, Mituś J, Ryś J. CD44/CD24 as potential prognostic markers in node‑positive invasive ductal breast cancer patients treated with adjuvant chemotherapy. J Mol Histol 2014; 45 (1): 35–45. Lee HJ, Choe G, Jheon S, Sung SW, Lee CT, Chung JH. CD24, a novel cancer biomarker, predicting disease‑free survival of non‑small cell lung carcinomas: a retrospective study of prognostic factor analysis from the viewpoint of forthcoming (seventh) new TNM classiication. J horac Oncol 2010; 5 (5): 649–57. Karimi‑Busheri F, Rasouli‑Nia A, Zadorozhny V, Fakhrai H. CD24+/CD38‑ as new prognostic marker for non‑small cell lung cancer. Multidiscip Respir Med 2013; 8 (1): 65. Kristiansen G, Denkert C, Schlüns K, Dahl E, Pilarsky C, Hauptmann S. CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival. Am J Pathol 2002; 161 (4): 1215–21. Qian X, Wagner S, Ma C, Coordes A, Gekeler J, Klussmann JP, Hummel M, Kaufmann AM, Albers AE. Prognostic signiicance of ALDH1A1‑positive cancer stem cells in patients with locally advanced, metastasized head and neck squamous cell carcinoma. J Cancer Res Clin Oncol 2014; 140 (7): 1151–8. Yang L, Ren Y, Yu X, Qian F, Bian BS, Xiao HL, Wang WG, Xu SL, Yang J, Cui W, Liu Q, Wang Z, Guo W, Xiong G, Yang K, Qian C, Zhang X, Zhang P, Cui YH, Bian XW. ALDH1A1 deines invasive cancer stem‑like cells and predicts poor prognosis in patients with esophageal squamous cell carcinoma. Mod Pathol 2014; 27 (5): 775–83. Li H, Bitler BG, Vathipadiekal V, Maradeo ME, Sliker M, Creasy CL, Tummino PJ, Cairns P, Birrer MJ, Zhang R. ALDH1A1 is a novel EZH2 target gene in epithelial ovarian cancer identiied by genome‑wide approaches. Cancer Prev Res (Phila) 2012; 5 (3): 484–91. Condello S, Morgan CA, Nagdas S, Cao L, Turek J, Hurley TD, Matei D. beta‑Catenin‑regulated ALDH1A1 is a target in ovarian cancer spheroids. Oncogene 2015; 34 (18): 2297–308. Liu X, Wang L, Cui W, Yuan X, Lin L, Cao Q, Wang N, Li Y, Guo W, Zhang X, Wu C, Yang J. Targeting ALDH1A1 by disuliram/copper complex inhibits non‑small cell lung cancer recurrence driven by ALDH‑positive cancer stem cells. Oncotarget 2016; 7 (36): 58516–30. Gong H, Zhang YC, Liu WL. Regulatory efects of Bmi‑1 gene on self‑renewal of hematopoietic stem cells‑review. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2006; 14 (2): 413–5. (in Chinese) Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ. Bmi‑1 dependence distinguishes neural stem cell self‑renewal from progenitor proliferation. Nature 2003; 425 (6961): 962–7. Nör C, Zhang Z, Warner KA, Bernardi L, Visioli F, Helman JI, Roesler R, Nör JE. Cisplatin induces Bmi‑1 and enhances the stem cell fraction in head and neck cancer. Neoplasia 2014; 16 (2): 137–46. Häyry V, Tynninen O, Haapasalo HK, Wölfer J, Paulus W, Hasselblatt M, Sariola H, Paetau A, Sarna S, Niemelä M, Wartiovaara K, Nupponen NN. Stem cell protein BMI‑1 is an independent marker for poor prognosis in oligodendroglial tumours. Neuropathol Appl Neurobiol 2008; 34 (5): 555–63. Arnes JB, Collett K, Akslen LA. Independent prognostic value of the basal‑like phenotype of breast cancer and associations with EGFR and candidate stem cell marker BMI‑1. Histopathology 2008; 52 (3): 370–80. Wang Y, Zhe H, Ding Z, Gao P, Zhang N, Li G. Cancer stem cell marker Bmi‑1 expression is associated with basal‑like phenotype and poor survival in breast cancer. World J Surg 2012; 36 (5): 1189–94. Strojnik T, Røsland GV, Sakariassen PO, Kavalar R, Lah T. Neural [Downloaded free from http://www.cancertm.com on Monday, March 12, 2018, IP: 65.96.222.254] Cancer stemness markers 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. stem cell markers, nestin and musashi proteins, in the progression of human glioma: correlation of nestin with prognosis of patient survival. Surg Neurol 2007; 68 (2): 133–43. Neradil J, Veselska R. Nestin as a marker of cancer stem cells. Cancer Sci 2015; 106 (7): 803–11. Wang H, Wang S, Hu J, Kong Y, Chen S, Li L, Li L. Oct4 is expressed in Nestin‑positive cells as a marker for pancreatic endocrine progenitor. Histochem Cell Biol 2009; 131 (5): 553–63. Yamahatsu K, Matsuda Y, Ishiwata T, Uchida E, Naito Z. Nestin as a novel therapeutic target for pancreatic cancer via tumor angiogenesis. Int J Oncol 2012; 40 (5): 1345–57. Matsuda Y, Hagio M, Ishiwata T. Nestin: a novel angiogenesis marker and possible target for tumor angiogenesis. World J Gastroenterol 2013; 19 (1): 42–8. Matsuda Y, Hagio M, Ishiwata T. Musashi1 as a potential therapeutic target and diagnostic marker for lung cancer. Oncotarget 2013; 4 (5): 739–50. Liekens S, Schols D, Hatse S. CXCL12‑CXCR4 axis in angiogenesis, metastasis and stem cell mobilization. Curr Pharm Des 2010; 16 (35): 3903–20. Chatterjee S, Behnam Azad B, Nimmagadda S. he intricate role of CXCR4 in cancer. Adv Cancer Res 2014; 124: 31–82. Otsuka S, Bebb G. he CXCR4/SDF‑1 chemokine receptor axis: a new target therapeutic for non‑small cell lung cancer. J horac Oncol 2008; 3 (12): 1379–83. Wang Z, Sun J, Feng Y, Tian X, Wang B, Zhou Y. Oncogenic roles and drug target of CXCR4/CXCL12 axis in lung cancer and cancer stem cell. Tumour Biol 2016; 37 (7): 8515–28. Epstein RJ. he CXCL12‑CXCR4 chemotactic pathway as a target of adjuvant breast cancer therapies. Nat Rev Cancer 2004; 4 (11): 901–9. Gil M, Seshadri M, Komorowski MP, Abrams SI, Kozbor D. Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast cancer metastases. Proc Natl Acad Sci U S A 2013; 110 (14): E1291–300. Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes Dev 2008; 22 (18): 2454–72. Cochrane CR, Szczepny A, Watkins DN, Cain JE. Hedgehog signaling in the maintenance of cancer stem cells. Cancers (Basel) 2015; 7 (3): 1554–85. Merchant AA, Matsui W. Targeting Hedgehog – A cancer stem cell pathway. Clin Cancer Res 2010; 16 (12): 3130–40. Gould A, Missailidis S. Targeting the hedgehog pathway: the development of cyclopamine and the development of anti‑cancer drugs targeting the hedgehog pathway. Mini Rev Med Chem 2011; 11 (3): 200–13. Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX, Ivy SP. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol 2015; 12 (8): 445–64. Yu X, Zou J, Ye Z, Hammond H, Chen G, Tokunaga A, Mali P, Li YM, Civin C, Gaiano N, Cheng L. Notch signaling activation in human embryonic stem cells is required for embryonic, but not trophoblastic, lineage commitment. Cell Stem Cell 2008; 2 (5): 461–71. 125. Hassan KA, Wang L, Korkaya H, Chen G, Maillard I, Beer DG, Kalemkerian GP, Wicha MS. Notch pathway activity identiies cells with cancer stem cell‑like properties and correlates with worse survival in lung adenocarcinoma. Clin Cancer Res 2013; 19 (8): 1972–80. 126. Abel EV, Kim EJ, Wu J, Hynes M, Bednar F, Proctor E, Wang L, Dziubinski ML, Simeone DM. he Notch pathway is important in maintaining the cancer stem cell population in pancreatic cancer. PLoS One 2014; 9 (3): e91983. 127. Capaccione KM, Pine SR. he Notch signaling pathway as a mediator of tumor survival. Carcinogenesis 2013; 34 (7): 1420–30. 128. Hirose H, Ishii H, Mimori K, Ohta D, Ohkuma M, Tsujii H, Saito T, Sekimoto M, Doki Y, Mori M. Notch pathway as candidate therapeutic target in Her2/Neu/ErbB2 receptor‑negative breast tumors. Oncol Rep 2010; 23 (1): 35–43. 129. Yuan X, Wu H, Xu H, Xiong H, Chu Q, Yu S, Wu GS, Wu K. Notch signaling: an emerging therapeutic target for cancer treatment. Cancer Lett 2015; 369 (1): 20–7. 130. Bausek N. JAK‑STAT signaling in stem cells and their niches in Drosophila. JAKSTAT 2013; 2 (3): e25686. 131. Stine RR, Matunis EL. JAK‑STAT signaling in stem cells. Adv Exp Med Biol 2013; 786: 247–67. 132. Morgensztern D, McLeod HL. PI3K/Akt/mTOR pathway as a target for cancer therapy. Anticancer Drugs 2005; 16 (8): 797–803. 133. Li H, Zeng J, Shen K. PI3K/AKT/mTOR signaling pathway as a therapeutic target for ovarian cancer. Arch Gynecol Obstet 2014; 290 (6): 1067–78. 134. Mabuchi S, Kuroda H, Takahashi R, Sasano T. he PI3K/AKT/ mTOR pathway as a therapeutic target in ovarian cancer. Gynecol Oncol 2015; 137 (1): 173–9. 135. Fulda S. he PI3K/Akt/mTOR pathway as therapeutic target in neuroblastoma. Curr Cancer Drug Targets 2009; 9 (6): 729–37. 136. Slomovitz BM, Coleman RL. he PI3K/AKT/mTOR pathway as a therapeutic target in endometrial cancer. Clin Cancer Res 2012; 18 (21): 5856–64. 137. Dos Santos C, Récher C, Demur C, Payrastre B. he PI3K/Akt/ mTOR pathway: a new therapeutic target in the treatment of acute myeloid leukemia. Bull Cancer 2006; 93 (5): 445–7. 138. Valkenburg KC, Graveel CR, Zylstra‑Diegel CR, Zhong Z, Williams BO. Wnt/beta‑catenin signaling in normal and cancer stem cells. Cancers (Basel) 2011; 3 (2): 2050–79. 139. Ashihara E, Takada T, Maekawa T. Targeting the canonical Wnt/ beta‑catenin pathway in hematological malignancies. Cancer Sci 2015; 106 (6): 665–71. 140. Gedaly R, Galuppo R, Daily MF, Shah M, Maynard E, Chen C, Zhang X, Esser KA, Cohen DA, Evers BM, Jiang J, Spear BT. Targeting the Wnt/beta‑catenin signaling pathway in liver cancer stem cells and hepatocellular carcinoma cell lines with FH535. PLoS One 2014; 9 (6): e99272. 141. Yao H, Ashihara E, Maekawa T. Targeting the Wnt/beta‑catenin signaling pathway in human cancers. Expert Opin her Targets 2011; 15 (7): 873–87. 95