Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Mar 27.
Published in final edited form as: Crit Rev Oncog. 2018;23(3-4):189–200. doi: 10.1615/CritRevOncog.2018027501

An Updated Review of Oral Cancer Stem Cells and Their Stemness Regulation

Ki-Hyuk Shin 1,*, Reuben H Kim 1
PMCID: PMC6436811  NIHMSID: NIHMS1014837  PMID: 30311574

Abstract

Cancer stem cells (CSCs; also known as tumor-initiating cells) are a small population of cancer cells that retain characteristics similar to those of normal stem cells. CSCs are known to be responsible for metastasis, drug resistance, and cancer recurrence. Thus, controlling CSCs may provide an effective therapeutic intervention that inhibits tumor growth and aggressiveness. Despite the importance of targeting CSCs in cancer therapy, the detailed nature of oral CSCs remains underexplored. This article reviews the current understanding of oral CSCs, with emphasis on recent advances in novel signaling pathways involved in their stemness regulation.

Keywords: oral cancer, cancer stem cell, stemness pathway

I. INTRODUCTION

Oral/oropharyngeal squamous cell carcinoma (OSCC), a common malignant tumor of the head and neck, is currently the sixth most common cancer worldwide.1 The projected 5-yr survival for OSCC is ~ 50%, and this statistic has remained the same during the past decade. As such, there is an urgent demand for new directions in therapeutics. Risk factors for OSCC development in the elderly are typically associated with a life-long history of cigarette smoking and heavy alcohol consumption.2,3 But recently, the incidence of oral cancer among young adults has been alarmingly elevated,4,5 and a sizeable proportion of cases has been associated with factors other than tobacco and alcohol use.6 Indeed, 34% of the cases in young adults did not show any tobacco/alcohol-related habits7; but rather, changes in sexual behaviors was one of the contributing factors for this increasing incidence of oral cancer in young adults. These data indicate that oral cancer is an emerging public health concern.8 Clinically, well-defined lesions, such as leukoplakia that is histologically classified as dysplastic or nondysplastic leukoplakia, often precede OSCC. Dysplastic leukoplakia is defined as an oral premalignant lesion and is associated with a likely progression to cancer; however, dysplastic leukoplakia is not an accurate predictor of cancer risk.9,10 Early-stage tumors can usually be managed through surgery and radiotherapy, but successful treatment is inversely proportional to extent of disease at the time of treatment. A combination of chemotherapy and radiation therapy, although effective in treating 97% of early-stage tumors, was only 33% effective in treating advanced tumors.11

Recent studies have unveiled and validated the pathophysiologic role of cancer stem cells (CSCs; also called tumor-initiating cells) in long-term sustenance of cancers.12,13 CSCs are small subpopulations of tumor cells that share many molecular similarities to embryonic and normal adult stem cells. CSCs have been isolated from various primary tumors and established cancer cell lines, including OSCC.1420 They play a crucial role in tumorigenicity, metastasis, and recurrence and are thus considered to be the root of the cancer. Therefore, advancing our understanding in the molecular properties and signaling pathways unique to oral CSCs is crucial for developing novel targeted and effective therapies for OSCC. This review focuses on several aspects of oral CSCs and their stemness signaling and also provides current advances in these fields.

II. ISOLATION OF ORAL CSCs

The existence of a CSC population was first reported in leukemic cells.21 The leukemic CSC population was positively stained with CD34 and negatively for CD38 (CD34+/CD38) and became capable of developing leukemia when inoculated onto immunocom-promised mice. Since then, CSC populations have been extensively examined and successfully identified in various solid tumors, such as brain, breast, prostate, colon, pancreas, and head and neck.2225 The presence of subpopulations of oral CSCs was originally suggested by the observation that only a subpopulation of OSCC cells can form an expanding tumor mass.26 Subsequently, Chiou and coworkers demonstrated that a subpopulation of OSCC cells derived from cultured OSCC cell lines possessed properties of both stem cells and aggressive malignant tumors, including self-renewal, tumorigenic potential, migratory ability, and radioresistance.14 Numerous research groups have reported successful isolation of oral CSC populations using various markers.1420,27,28 In general, CSCs in OSCC can be isolated by either cell-surface markers or their unique functional properties.2931 Nevertheless, no single marker and CSC property are capable of specifically isolating oral CSC populations from OSCC cells, suggesting the heterogeneity of CSC populations.32,33 Therefore, identification of additional oral CSC markers and their unique cellular characteristics is warranted.

Many studies have reported differential expression of CD44 on CSCs versus non-CSCs in various solid tumors,34,35 and such CD44+ CSC populations were successfully isolated from head and neck cancer by flow cytometry sorting using the CD44 antibody.25,27,28 CD44, a multifunctional transmembrane glycoprotein, binds to hyaluronan and behaves as a vital surface molecule that can interact with various intrinsic and extrinsic signals to regulate a variety of gene expressions. CD44+ cells can be fractionated from heterogeneous single-cell-prepared cancer cells using CD44 specific antibody labeling followed by flow cytometry sorting.36 These CD44+ cells display discriminative traits of stem cells, including self-renewal capacity, high tumorigenic potential, and metastasis and drug resistance.34,35 When CD44 is experimentally suppressed, CD44+ CSCs diminish their stemness traits, indicating that CD44 expression is imperative for preservation of the CSC phenotype.35

The activity of aldehyde dehydrogenase 1 (ALDH1) has been used as a CSC marker for different types of cancer, including OSCC.3739 ALDH1 is a cytosolic isoenzyme responsible for oxidizing intracellular aldehydes, thus contributing to the oxidation of retinol to retinoic acid in early stem cell differentiation.40 A subpopulation of cancer cells with enriched CSC activity shows high ALDH1 activity (ALDH1HIGH or ALDH1+) compared to the non-CSC population, suggesting that ALDH1+ cells may be a source of CSCs.15,41,42 Moreover, ALDH1HIGH cancer cells display enhanced CSC properties compared to ALDH1low cells.4345 Targeting ALDH1 significantly suppresses multiple CSC properties in human cancer cells.46 However, it remains controversial as to whether CD44 or ALDH1 alone can be defined as a single molecule to identify CSCs. Thus, the combination of ALDH1 and CD44 has been commonly used as a marker for isolating CSCs in head and neck cancer cells.25 Indeed, ALDH1HIGH/CD44+ cancer cells display elevated CSC traits compared to those of ALDH1low/CD44 cancer cells,47,48 indicating that the combination of CSC markers may improve specificity of CSC isolation.

CSCs can be enriched in nonadherent tumor spheres cultured in ultra-low attachment plates to support the undifferentiated growth of self-renewing stem cells.49 The sphere medium is in a serum-free condition supplemented with adequate mitogens, such as basic fibroblast growth factor and epidermal growth factor (EGF).5052 Abundance and growth kinetics of tumor spheres are indicative of self-renewal capacity in a given culture of heterogeneous cancer cells, indicating CSC content. Thus, the tumor sphere-forming assay is a common technique used to isolate CSCs from heterogeneous cancer cell populations by means of the unique functional trait of CSCs. Tumor sphere-forming cells identified in multiple primary tumors and cultured cancer cell lines displayed enhanced CSC characteristics compared to those of corresponding adherent monolayer cells that were considered to be non-CSCs.53 Tumor sphere-forming cells possess high tumorigenicity, metastatic potential, and drug resistance and show the robust expression of stemness factors, suggesting their central role in pathogenesis and progression of cancer.5459 Similarly, tumor spheres derived from OSCC cells display enhanced stem-like properties and express higher levels of pluripotent transcription factors including Nanog, Oct4, KLF4, Lin28, and Sox2, compared with their corresponding adherent monolayer cells.14,52,60,61 Oral tumor sphere-forming cells also express robust expression of CSC specific markers, such as CD44 and ALDH1.51,52,62 They are highly tumorigenic when inoculated into nude mice and maintain their self-renewal capacity for multiple generations.62

III. PROPERTIES AND STEMNESS PATHWAYS OF ORAL CSCs

CSCs share common properties with normal stem cells and have multiple unique properties that maintain tumor growth and aggressiveness. A key feature of CSCs is their self-renewal capacity, which appears to be a driving force for initiating and maintaining tumorigenicity.63 Self-renewal of CSCs can be maintained by several endogenous signaling pathways, such as Notch, Hedgehog, Wnt, B-cell–specific Moloney murine leukemia virus integration site 1 (Bmi1), Pten, Bmp, and TGF-β,6470 which are frequently activated in human cancers.65,71,72 Among these pathways, the roles of Notch and Bmi1 signaling in oral cancer stemness have been extensively documented. Activation of the Notch1 signaling pathway is critical for the maintenance of CSCs and requires binding of its ligands Jagged 1 (JAG1), JAG 2, and δ-like, followed by proteolytic release of the Notch intracellular domain (NICD) and activation of NICD downstream target genes.73 Previously, we reported that prolonged exposure of OSCC cells to the proinflammatory cytokine TNFα enhances self-renewal capacity and tumorigenicity, which is associated with activation of the Notch pathway.50 In TNFα-induced oral cancer stemness, Hes1 is the target of activated Notch1, and its knockdown suppresses self-renewal capacity of TNFα-treated OSCC cells. Hes1 is commonly expressed in most undifferentiated cell types in a developing mouse embryo and has a critical role in maintenance of progenitor cell fate. Hes1-deficient mice displayed premature differentiation, progenitor cell depletion, and consequent lethality.74 Collectively, these findings suggest that the Notch1–Hes1 axis is a novel axis for regulating self-renewal of oral CSCs.

Bmi1, a member of the polycomb group transcription repressor, is implicated in oral cancer.75,76 Recent studies showed a critical role of Bmi1 in maintaining the self-renewal capacity of oral CSCs.77,78 Furthermore, using genetic lineage tracing, the in vivo role of Bmi1 in regulation of oral CSC stemness was clearly demonstrated, including its self-renewal and tumorigenic potential.77

A unique property of CSCs is their metastatic potential.63 Epithelial–mesenchymal transition (EMT) is known to confer migratory potential in cancer cells, and this process has crucial roles in cancer metastasis. EMT is a process by which epithelial cells lose their characteristics to gain the mesenchymal phenotype, thus leading to cell migration and invasion.79,80 During EMT, epithelium-specific protein expressions (e.g., cytokeratins and E-cadherin) are diminished, whereas expressions of mesenchymal-specific proteins (fibronectin, vimentin, and N-Cad) are elevated. Master transcription factors for EMT including SNAIL, TWIST, and LEF-1 have been identified, and their overexpression promoted EMT.81,82 Fractionated CSCs overexpress EMT transcription factors and demonstrate great in vivo metastatic potential compared to that in unfractionated cancer cells, suggesting that CSCs are the major source of the metastatic cancer cell population.83 In addition, other reports also revealed the crucial roles of the zinc-finger E-box–binding homeobox (Zeb) in maintenance of CSC properties and EMT.84 Zeb1 and Zeb2 are significantly increased in head and neck CSCs compared to those in non-CSCs.85 Knockdown of Zeb1 and Zeb2 in head and neck cancer cells decreased their CSC properties such as migration, self-renewal capacity, and expression of stemness markers. Moreover, their suppression inhibited in vivo tumor growth and rate of metastasis to distant sites.85 Conversely, co-overexpression of Zeb1 and Zeb2 enhanced the migration ability of head and neck cancer cells.85

The CSC population can be enriched following chemoradiotherapy, suggesting that therapy results in chemoradioresistance and/or selectively enriches the resistant cell population. Various molecular determinants for CSC chemoradioresistance have been reported. Among these, the roles of adenosine triphosphate (ATP)-binding cassette (ABC) transporters are well documented to be key players in therapy resistance.86 ABC transporters are membrane transporters that can pump various small molecules, for instance anticancer drugs, out of cells at the cost of ATP hydrolysis, thereby resulting in low intracellular drug concentrations. Overexpression of ABC transporters is a common occurrence observed in multidrug resistance in cancer.87 Normal cells and CSCs express high levels of ABC transporters, and overexpression of ABC transporters in cancer cells increased their chemoradioresistance.88 Suppression of ABC transporters increases anticancer drug sensitivity in cancer.89 These reports collectively indicate that ABC transporters are indeed key molecular determinants of CSC chemoradioresistance. Small populations of CSCs possessing high efflux capacity due to increased ABC transporters can be isolated by treatment of cells with Hoechst 33342 dye and then designated as side population (SP). Numerous studies have demonstrated successful isolation of CSCs using this technique, and SP cells harbor a greater capacity for the CSC phenotype than do non-SP cells.90,91 The presence of SP cells in oral cancer has been reported, and oral SP cells, when compared with non-SP cells, possess not only increased anticancer drug resistance but also the stem cell phenotype.9193 Therefore, there is general consensus that CSCs are intrinsically resistant to chemoradiotherapy and contribute to tumor relapse.13

IV. ROLE OF HISTONE DEMETHYLASES IN THE REGULATION OF ORAL CANCER STEMNESS

Emerging evidence has indicated that oral CSCs could be epigenetically regulated by histone demethylases or microRNAs.51,9496 A group of histone demethylases epigenetically modulated gene transcription by removing histone methylation marks.97 As such, histone demethylases have a crucial role in governing gene transcription by altering chromatin accessibility and transcriptional machineries. Compelling evidence indicates that histone demethylases are implicated in various cellular processes, including carcinogenesis, cell fate choices, and cell differentiation.98100 Recently, a growing body of evidence has indicated an important role of histone demethylases, including LSD1, JARID1, KMD3, KDM4, KDM5, KDM6A, KDM6B, and Jumonji domain–containing protein 6 (JMJD6), in the CSC phenotype in multiple cancer types.51,101109

JMJD6 is identified as a novel molecular regulator of oral CSCs.51 JMJD6 is a histone arginine demethylase that preferentially removes methyl groups from dimethylated arginine 2 of histone 3 (H3R2me2) and arginine 3 of histone 4 (H4R3me2),110 thereby enabling dynamic regulation of transcription. JMJD6 also regulates gene expression by modulating RNA splicing,111 suggesting that JMJD6 is a multifaceted regulator of gene expression. Elevated JMJD6 expression has been reported in various human cancers, including breast cancer,112 lung cancer,113 and colon cancer.114 A high expression of JMJD6 protein is also strongly linked to poor prognosis and aggressive behavior in human cancers. The level of JMJD6 in nonmalignant oral epithelial cell lines is much lower than that in OSCC cell lines in vitro.51 JMJD6 is highly expressed in OSCC compared to in normal tissues in vivo, indicating that JMJD6 plays an important part during oral carcinogenesis.

We have found that JMJD6 is enriched in OSCC CSC populations (i.e., tumor spheres and the ALDH1HIGH cell population) compared to that in OSCC non-CSC populations (adherent monolayer cells and the ALDH1low cell population).51 Silencing JMJD6 led to loss of self-renewal capacity, migration ability, and chemoresistance in OSCC cells. It was also reported that knockdown of JMJD6 in invasive breast cancer cell lines decreased cell migration, but its overexpression promoted cellular motility.112 JMJD6 interacts with splicing factor U2AF65 and modulates alternate splicing of the vascular EGF (VEGF) receptor.111 Alternate splicing of the VEGF receptor by U2AF65 promoted endothelial cell migration, and silencing of JMJD6 in endothelial cells led to decreased migration.115 Thus, the effects of JMJD6 on EMT warrants investigation. Conversely, JMJD6 overexpression promotes not only such CSC properties but also the number of CSCs, indicating that JMJD6 is an important regulator of the CSC phenotype and genesis in OSCC. JMJD6 induces robust expression of pluripotent transcription factors and CSC-related genes, such as Oct4, Lin28A, Lin28B, FGF4, Zeb1, Zeb2, Gli1, and Gli3 in OSCC.112 Studies have shown the important role of these genes in acquisition and maintenance of CSC phenotype. For instance, Oct4 is detected in CSCs of various cancers including melanoma, prostate cancer, lung cancer, and oral cancer.14,116,117 Oct4 promotes self-renewal and the tumorigenic potential of melanoma cells.118 Lin28A is up-regulated in the CSC-enriched OSCC population, and overexpression of Lin28A in oral cancer cells increased their proliferation, colony formation, and migration.17 These CSC-related genes are up-regulated by JMJD6 overexpression and down-regulated by JMJD6 knockdown in OSCC cells. These results collectively suggest that JMJD6 enhances oral cancer stemness by modulating various CSC factors.

A recent study has shown a critical role for inflammatory cytokines in the tumor microenvironment and regulation of cancer stemness.119 Inflammatory cytokine TNFα enhances the CSC phenotype of OSCC.50 Multiple cytokines (e.g., TNFα, IL4, etc.) are commonly up-regulated in OSCC CSCs compared to the non-CSC population (our unpublished data). IL4 is known to promote tumor formation by inhibiting apoptosis and enhancing proliferation.120,121 The autocrine production of IL4 by cancer cells confers resistance to chemotherapy-induced cell death122 and promotes tumor growth and metastasis.120 Moreover, the production of IL4 in colon CSCs is responsible for their chemoresistance, which is a key CSC characteristic.123,124 Our study reveals that up-regulation of IL4 is an important event during JMJD6-induced self-renewal in OSCC CSCs. IL4 treatment rescues CSC properties suppressed by JMJD6 knockdown, and JMJD6 up-regulates IL4 by binding to the IL4 promoter region. Because CSCs interact with stromal cells in the tumor microenvironment, involvement of cytokines in this interaction is probable and also a crucial event for cancer progression and propagation. Collectively, these studies conclude that JMJD6 is a novel epigenetic regulator of CSC properties in OSCC by up-regulating IL4, suggesting that the JMJD6-IL4 axis could be an important therapeutic target in OSCC CSCs. Because chemical inhibitors readily inhibit histone demethylases, targeting JMJD6 may reveal a plausible therapeutic modality against oral CSCs.125,126

V. ROLE OF CALCIUM CHANNELS IN REGULATING ORAL CANCER STEMNESS

Recent studies suggested the importance of calcium signaling in regulating oral cancer stemness properties.52,127129 Ca2+ is a universal second messenger that regulates many physiological processes, and its homeostasis is disrupted during carcinogenesis, thereby leading to deregulated cell proliferation, migration, and suppression of apoptosis.130133 In most nonexcitable cells, Ca2+ influx is tightly regulated by the store-operated Ca2+ entry (SOCE) pathway and mediated through store-operated Ca2+ release-activated Ca2+ (CRAC) channels.134 Orai1 is an essential pore subunit of CRAC channels.135137 After stimulation, cells release Ca2+ from the endoplasmic reticulum (ER), followed by extracellular Ca2+ influx through SOCE. SOCE not only refills the depleted ER Ca2+ stores but also provides a direct Ca2+ signal to activate downstream responses, including the nuclear factor of activated T-cells (NFAT) signaling pathway.138,139 Orai1 has been extensively studied in immunology because NFAT is the transcription factor that is necessary for activation, differentiation, and effector functions of T-cells.140 Emerging evidence indicates the crucial role of Orai1 in carcinogenesis.128,141150

Elevated expression of Orai1 is observed in various human cancers, including head and neck cancer,52,128,145,147,150 and strongly linked to poor overall and recurrence-free survival in human cancers.145,150 Orai1 is required for tumor growth.128,142146,151 Our recent study revealed that Orai1 is increased in a stepwise manner during oral carcinogenesis.52 Orai1 is highly expressed in OSCC compared to precancerous and normal tissues in vivo. Precancerous oral epithelial cells express higher Orai1 protein than normal oral epithelial cells. Inhibition of Orai1 function by expressing the dominant negative Orai1 E106Q mutant137 in OSCC cells abolishes SOCE and subsequently inhibits tumorigenic potential in vitro and in vivo. Conversely, ectopic Orai1 expression successfully converts nontumorigenic oral epithelial cells into tumorigenic cells. These findings indicate that Orai1 is required for tumorigenicity of OSCC.

Orai1 is highly expressed in CSC-enriched cell populations, such as tumor spheres and the ALDH1HIGH population of OSCC.52 Furthermore, Orai1 can endow nontumorigenic immortalized oral epithelial cells with self-renewal and concomitantly increases pluripotent transcription and CSC-related factors, including Nanog, Oct4, Sox2, KLF4, Bmi1, Zeb1, and Zeb2. Ectopic Orai1 expression increases the ALDH1+ CSC population in nontumorigenic oral epithelial cells and promotes metastatic potential of OSCC. The latter finding is consistent with other reports showing the importance of Orai1 in the migration ability of invasive breast cancer cells.112 In multiple OSCC cell lines, inhibition of Orai1 led to suppression of CSC properties. Thus, it is hypothesized that Orai1 promotes malignant progression of OSCC by enriching the CSC phenotype. However, the underlying mechanism by which Orai1 regulates oral cancer stemness remains largely unknown.

The main downstream target of Orai1-mediated Ca2+ is NFAT, which is dephosphorylated by a protein phosphatase complex of calmodulin and calcineurin.152,153 This process results in translocation of cytoplasmic NFAT to the nucleus. Once in the nucleus, NFAT binds to target promoter elements and induces gene transcription. Four members of the NFAT family (i.e., NFATc1–c4) that are regulated by Ca2+ signaling have been identified.154 They share a highly conserved DNA-binding domain but depend on interacting partners including other transcription factors and coactivators for their target gene specificity. It is known that NFAT has an important role in tumorigenesis by regulating various target genes involved in cancer development.155,156 For instance, NFATc1 induced the malignant cell growth phenotype in pancreatic cancer cells by up-regulating Myc157 and promoted metastasis of mammalian cancer cells via MMP-2 up-regulation.158160 NFATc2 was overexpressed in multiple cancer types,159,160 and its depletion suppressed the migration/invasion of cancer cells.159 In addition, the crucial role of NFAT in maintenance of CSCs was reported in human cancers, including melanoma and colonic, pancreatic, and lung.161164 In our unpublished study, we found that NFATc3, among four NFAT isoforms, is the dominant isoform in human oral epithelial cells and greatly increases in OSCC compared to normal oral epithelial cells. Interestingly, Orai1-mediated SOCE activates NFATc3.52 Silencing NFATc3 in cells with ectopic Orai1 overexpression in OSCC cells led to suppression of the CSC phenotype. An NFAT chemical inibitor also sucessfully inhibited cancer stemness in cells. These results indicate that NFATc3 is required for the Orai1-induced CSC phenotype, suggesting the funtional role of the Orai1/NFATc3 axis in regulation of oral CSC.

Ca2+ oscillation (spatiotemporal regulation calcium signaling) is more essential than global changes in cytosolic Ca2+ concentration in the context of tumor invasion, growth, and cancer stemness.127129 Ca2+ oscillation is the end result of Orai1-mediated SOCE, and Orai1 is enriched in oral CSCs. Because suppression of Orai1 channel function resulted in complete shutdown of Ca2+ oscillation in OSCC cells, Orai1-mediated Ca2+ oscillation could be a potential selective target for treatment of oral CSCs.

VI. CONCLUSIONS

The pathophysiologic role of CSCs in sustaining and maintaining cancers is increasingly evident in numerous studies. These small subpopulations of tumor cells are responsible for tumorigenicity, metastasis, and recurrence and are thus considered to be the root of the cancer. Our understanding of the molecular signaling pathways specific to CSCs, such as histone modifications and calcium regulations, may provide unique opportunities to develop new strategies in targeting CSCs for cancer treatment.

ACKNOWLEDGMENTS

This work was supported in part by a UCLA School of Dentistry faculty seed grant (K.H.S.) and grant R01DE023348 (R.H.K.) from the National Institute on Dental Craniofacial Research/National Institutes of Health.

ABBREVIATIONS:

ABC

adenosine triphosphate (ATP)-binding cassette

ALDH1

aldehyde dehydrogenase 1

CSC

cancer stem cell

EMT

epithelial–mesenchymal transition

JMJD6

Jumonji domain–containing protein 6

NFAT

nuclear factor of activated T cells

OSCC

oral/oropharyngeal squamous cell carcinoma

SOCE

store-operated Ca2+ entry

SP

side population

REFERENCES

  • 1.Warnakulasuriya S Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 2009;45:309–16. [DOI] [PubMed] [Google Scholar]
  • 2.Lee CH, Lee JM, Wu DC, Hsu HK, Kao EL, Huang HL, Wang TN, Huang MC, Wu MT. Independent and combined effects of alcohol intake, tobacco smoking and betel quid chewing on the risk of esophageal cancer in Taiwan. Int J Cancer. 2005;113:475–82. [DOI] [PubMed] [Google Scholar]
  • 3.Perez-Sayans M, Somoza-Martin JM, Barros-Angueira F, Reboiras-Lopez MD, Gandara Rey JM, Garcia-Garcia A. Genetic and molecular alterations associated with oral squamous cell cancer (review). Oncol Rep. 2009;22:1277–82. [DOI] [PubMed] [Google Scholar]
  • 4.Shiboski CH, Schmidt BL, Jordan RCK. Tongue and tonsil carcinoma: Increasing trends in the US population ages 20–44 years. Cancer Am Cancer Soc. 2005;103:1843–9. [DOI] [PubMed] [Google Scholar]
  • 5.Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao WH, Kim E, Jiang B, Goodman MT, Sibug-Saber M, Cozen W, Liu LH, Lynch CF, Wentzensen N, Jordan RC, Altekruse S, Anderson WF, Rosenberg PS, Gillison ML. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29:4294–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Iype EM, Pandey M, Mathew A, Thomas G, Sebastian P, Nair MK. Squamous cell carcinoma of the tongue among young Indian adults. Neoplasia. 2001;3:273–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sherin N, Simi T, Shameena P, Sudha S. Changing trends in oral cancer. Indian J Cancer. 2008;45:93–6. [DOI] [PubMed] [Google Scholar]
  • 8.Al-Swiahb JN, Chen CH, Chuang HC, Fang FM, Tasi HT, Chien CY. Clinical, pathological and molecular determinants in squamous cell carcinoma of the oral cavity. Future Oncol. 2010;6:837–50. [DOI] [PubMed] [Google Scholar]
  • 9.Warnakulasuriya S, Reibel J, Bouquot J, Dabelsteen E. Oral epithelial dysplasia classification systems: Predictive value, utility, weaknesses and scope for improvement. J Oral Pathol Med. 2008;37:127–33. [DOI] [PubMed] [Google Scholar]
  • 10.Brennan M, Migliorati CA, Lockhart PB, Wray D, Al-Hashimi I, Axell T, Bruce AJ, Carpenter W, Eisenberg E, Epstein JB, Holmstrup P, Jontell M, Nair R, Sasser H, Schifter M, Silverman B, Thongprasom K, Thornhill M, Warnakulasuriya S, van der Waal I. Management of oral epithelial dysplasia: A review. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2007;103 (Suppl):S19 e11–2. [DOI] [PubMed] [Google Scholar]
  • 11.Marcial VA, Pajak TF, Mohiuddin M, Cooper JS, al Sarraf M, Mowry PA, Curran W, Crissman J, Rodriguez M, Velez-Garcia E. Concomitant cisplatin chemotherapy and radiotherapy in advanced mucosal squamous cell carcinoma of the head and neck. Long-term results of the Radiation Therapy Oncology Group study 81–17. Cancer. 1990;66:1861–8. [DOI] [PubMed] [Google Scholar]
  • 12.Beck B, Blanpain C. Unravelling cancer stem cell potential. Nat Rev Cancer. 2013;13:727–38. [DOI] [PubMed] [Google Scholar]
  • 13.Nassar D, Blanpain C. Cancer stem cells: Basic concepts and therapeutic implications. Annu Rev Pathol. 2016;11:47–76. [DOI] [PubMed] [Google Scholar]
  • 14.Chiou SH, Yu CC, Huang CY, Lin SC, Liu CJ, Tsai TH, Chou SH, Chien CS, Ku HH, Lo JF. Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin Cancer Res. 2008;14:4085–95. [DOI] [PubMed] [Google Scholar]
  • 15.Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu SL, Schott A, Hayes D, Birnbaum D, Wicha MS, Dontu G. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Grimm M, Krimmel M, Polligkeit J, Alexander D, Munz A, Kluba S, Keutel C, Hoffmann J, Reinert S, Hoefert S. ABCB5 expression and cancer stem cell hypothesis in oral squamous cell carcinoma. Eur J Cancer. 2012;48:3186–97. [DOI] [PubMed] [Google Scholar]
  • 17.Hayashi S, Tanaka J, Okada S, Isobe T, Yamamoto G, Yasuhara R, Irie T, Akiyama C, Kohno Y, Tachikawa T, Mishima K. Lin28a is a putative factor in regulating cancer stem cell-like properties in side population cells of oral squamous cell carcinoma. Exp Cell Res. 2013;319:1220–8. [DOI] [PubMed] [Google Scholar]
  • 18.Richard V, Sebastian P, Nair MG, Nair SN, Malieckal TT, Santhosh Kumar TR, Pillai MR. Multiple drug resistant, tumorigenic stem-like cells in oral cancer. Cancer Lett. 2013;338:300–16. [DOI] [PubMed] [Google Scholar]
  • 19.Zhang Q, Shi S, Yen Y, Brown J, Ta JQ, Le AD. A subpopulation of CD133+ cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy. Cancer Lett. 2010;289:151–60. [DOI] [PubMed] [Google Scholar]
  • 20.Felthaus O, Ettl T, Gosau M, Driemel O, Brockhoff G, Reck A, Zeitler K, Hautmann M, Reichert TE, Schmalz G, Morsczeck C. Cancer stem cell-like cells from a single cell of oral squamous carcinoma cell lines. Biochem Biophys Res Commun. 2011;407:28–33. [DOI] [PubMed] [Google Scholar]
  • 21.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. [DOI] [PubMed] [Google Scholar]
  • 22.Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030–7. [DOI] [PubMed] [Google Scholar]
  • 23.Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S, Reilly JG, Chandra D, Zhou J, Claypool K, Coghlan L, Tang DG. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene. 2006;25:1696–708. [DOI] [PubMed] [Google Scholar]
  • 24.Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–8. [PubMed] [Google Scholar]
  • 25.Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, Weissman IL, Clarke MF, Ailles LE. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA. 2007;104:973–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mackenzie IC. Growth of malignant oral epithelial stem cells after seeding into organotypical cultures of normal mucosa. J Oral Pathol Med. 2004;33:71–8. [DOI] [PubMed] [Google Scholar]
  • 27.Ghuwalewala S, Ghatak D, Das P, Dey S, Sarkar S, Alam N, Panda CK, Roychoudhury S. CD44highCD24low molecular signature determines the cancer stem cell and EMT phenotype in oral squamous cell carcinoma. Stem Cell Res. 2016;16:405–17. [DOI] [PubMed] [Google Scholar]
  • 28.Todoroki K, Ogasawara S, Akiba J, Nakayama M, Naito Y, Seki N, Kusukawa J, Yano H. CD44v3+/CD24 cells possess cancer stem cell-like properties in human oral squamous cell carcinoma. Int J Oncol. 2016;48:99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Krishnamurthy S, Nor JE. Head and neck cancer stem cells. J Dent Res. 2012;91:334–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shrivastava S, Steele R, Sowadski M, Crawford SE, Varvares M, Ray RB. Identification of molecular signature of head and neck cancer stem-like cells. Sci Rep. 2015;5:7819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pastrana E, Silva-Vargas V, Doetsch F. Eyes wide open: A critical review of sphere-formation as an assay for stem cells. Cell Stem Cell. 2011;8:486–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Mol Cancer. 2017;16:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Eun K, Ham SW, Kim H. Cancer stem cell heterogeneity: Origin and new perspectives on CSC targeting. BMB Rep. 2017;50:117–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wang L, Zuo X, Xie K, Wei D. The role of CD44 and cancer stem cells. Methods Mol Biol. 2018;1692:31–42. [DOI] [PubMed] [Google Scholar]
  • 35.Yan Y, Zuo X, Wei D. Concise review: Emerging role of CD44 in cancer stem cells: A promising biomarker and therapeutic target. Stem Cells Transl Med. 2015;4:1033–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Clay MR, Tabor M, Owen JH, Carey TE, Bradford CR, Wolf GT, Wicha MS, Prince ME. Single-marker identification of head and neck squamous cell carcinoma cancer stem cells with aldehyde dehydrogenase. Head Neck J Sci Spec. 2010;32:1195–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ota N, Ohno J, Seno K, Taniguchi K, Ozeki S. In vitro and in vivo expression of aldehyde dehydrogenase 1 in oral squamous cell carcinoma. Int J Oncol. 2014;44:435–442. [DOI] [PubMed] [Google Scholar]
  • 39.Tomita H, Tanaka K, Tanaka T, Hara A. Aldehyde dehydrogenase 1A1 in stem cells and cancer. Oncotarget. 2016;7:11018–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yoshida A Molecular-genetics of human aldehyde dehydrogenase. Pharmacogenetics. 1992;2:139–47. [DOI] [PubMed] [Google Scholar]
  • 41.Chen YC, Chen YW, Hsu HS, Tseng LM, Huang PI, Lu KH, Chen DT, Tai LK, Yung MC, Chang SC, Ku HH, Chiou SH, Lo WL. Aldehyde dehydrogenase 1 is a putative marker for cancer stem cells in head and neck squamous cancer. Biochem Biophys Res Commun. 2009;385:307–13. [DOI] [PubMed] [Google Scholar]
  • 42.Neumeister V, Agarwal S, Bordeaux J, Camp RL, Rimm DL. In situ identification of putative cancer stem cells by multiplexing ALDH1, CD44, and cytokeratin identifies breast cancer patients with poor prognosis. Am J Pathol. 2010;176:2131–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Clay MR, Tabor M, Owen JH, Carey TE, Bradford CR, Wolf GT, Wicha MS, Prince ME. Single-marker identification of head and neck squamous cell carcinoma cancer stem cells with aldehyde dehydrogenase. Head Neck. 2010;32:1195–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, Weissman IL, Clarke MF, Ailles LE. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA. 2007;104:973–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Prince MEP, Zhou L, Moyer JS, Tao HM, Lu L, Owen J, Egenti M, Zheng F, Chang AE, Xia JC, Wolf G, Wicha MS, Huang S, Ren XB, Li Q. Evaluation of the immunogenicity of ALDHhigh human head and neck squamous cell carcinoma cancer stem cells in vitro. Oral Oncol. 2016;59:30–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rae C, Tesson M, Babich JW, Boyd M, Sorensen A, Mairs RJ. The role of copper in disulfiram-induced toxicity and radiosensitization of cancer cells. J Nucl Med. 2013;54:953–60. [DOI] [PubMed] [Google Scholar]
  • 47.Bourguignon LY, Wong G, Earle C, Chen L. Hyaluronan-CD44v3 interaction with Oct4-Sox2-Nanog promotes miR-302 expression leading to self-renewal, clonal formation, and cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma. J Biol Chem. 2012;287:32800–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Keymoosi H, Gheytanchi E, Asgari M, Shariftabrizi A, Madjd Z. ALDH1 in combination with CD44 as putative cancer stem cell markers are correlated with poor prognosis in urothelial carcinoma of the urinary bladder. Asian Pac J Cancer Prev. 2014;15:2013–20. [DOI] [PubMed] [Google Scholar]
  • 49.Wang Y, Yu Y, Tsuyada A, Ren X, Wu X, Stubblefield K, Rankin-Gee EK, Wang SE. Transforming growth factor-β regulates the sphere-initiating stem cell-like feature in breast cancer through miRNA-181 and ATM. Oncogene. 2011;30:1470–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lee SH, Hong HS, Liu ZX, Kim RH, Kang MK, Park NH, Shin KH. TNFα enhances cancer stem cell-like phenotype via Notch-Hes1 activation in oral squamous cell carcinoma cells. Biochem Biophys Res Commun. 2012;424:58–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lee CR, Lee SH, Rigas NK, Kim RH, Kang MK, Park NH, Shin KH. Elevated expression of JMJD6 is associated with oral carcinogenesis and maintains cancer stemness properties. Carcinogenesis. 2016;37:119–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lee SH, Rigas NK, Lee CR, Bang A, Srikanth S, Gwack Y, Kang MK, Kim RH, Park NH, Shin KH. Orai1 promotes tumor progression by enhancing cancer stemness via NFAT signaling in oral/oropharyngeal squamous cell carcinoma. Oncotarget. 2016;7:43239–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cao L, Zhou Y, Zhai B, Liao J, Xu W, Zhang R, Li J, Zhang Y, Chen L, Qian H, Wu M, Yin Z. Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol. 2011;11:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liu J, Ma L, Xu J, Liu C, Zhang J, Liu J, Chen R, Zhou Y. Spheroid body-forming cells in the human gastric cancer cell line MKN-45 possess cancer stem cell properties. Int J Oncol. 2013;42:453–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Reynolds DS, Tevis KM, Blessing WA, Colson YL, Zaman MH, Grinstaff MW. Breast cancer spheroids reveal a differential cancer stem cell response to chemotherapeutic treatment. Sci Rep. 2017;7:10382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Morata-Tarifa C, Jimenez G, Garcia MA, Entrena JM, Grinan-Lison C, Aguilera M, Picon-Ruiz M, Marchal JA. Low adherent cancer cell subpopulations are enriched in tumorigenic and metastatic epithelial-to-mesenchymal transition-induced cancer stem-like cells. Sci Rep. 2016;6:18772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Grimshaw MJ, Cooper L, Papazisis K, Coleman JA, Bohnenkamp HR, Chiapero-Stanke L, Taylor-Papadimitriou J, Burchell JM. Mammosphere culture of metastatic breast cancer cells enriches for tumorigenic breast cancer cells. Breast Cancer Res. 2008;10:R52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Fujii H, Honoki K, Tsujiuchi T, Kido A, Yoshitani K, Takakura Y. Sphere-forming stem-like cell populations with drug resistance in human sarcoma cell lines. Int J Oncol. 2009;34:1381–6. [PubMed] [Google Scholar]
  • 59.Gibbs CP, Kukekov VG, Reith JD, Tchigrinova O, Suslov ON, Scott EW, Ghivizzani SC, Ignatova TN, Steindler DA. Stem-like cells in bone sarcomas: Implications for tumorigenesis. Neoplasia. 2005;7:967–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Saleem S, Jamshed A, Faisal S, Hussain R, Tahseen M, Loya A, Sutton C. Patterns of cancer cell sphere formation in primary cultures of human oral tongue squamous cell carcinoma and neck nodes. Cancer Cell Int. 2014;14:542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Misuno K, Liu X, Feng S, Hu S. Quantitative proteomic analysis of sphere-forming stem-like oral cancer cells. Stem Cell Res Ther. 2013;4:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chen YK, Huang AH, Lin LM. Sphere-forming-like cells (squamospheres) with cancer stem-like cell traits from VX2 rabbit buccal squamous cell carcinoma. Int J Oral Sci. 2014;6:212–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Beck B, Blanpain C. Unravelling cancer stem cell potential. Nat Rev Cancer. 2013;13:727–38. [DOI] [PubMed] [Google Scholar]
  • 64.Liu SL, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, Suri P, Wicha MS. Hedgehog signaling and BMI-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006;66:6063–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Giles RH, van Es JH, Clevers H. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003;1653:1–24. [DOI] [PubMed] [Google Scholar]
  • 66.O’Brien CA, Kreso A, Jamieson CH. Cancer stem cells and self-renewal. Clin Cancer Res. 2010;16:3113–20. [DOI] [PubMed] [Google Scholar]
  • 67.Hill R, Wu H. Pten, stem cells, and cancer stem cells. J Biol Chem. 2009;284:11755–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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. [DOI] [PubMed] [Google Scholar]
  • 69.Zhang L, Ye Y, Long X, Xiao P, Ren X, Yu J. BMP signaling and its paradoxical effects in tumorigenesis and dissemination. Oncotarget. 2016;7:78206–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mishra L, Shetty K, Tang Y, Stuart A, Byers SW. The role of TGF-β and Wnt signaling in gastrointestinal stem cells and cancer. Oncogene. 2005;24:5775–89. [DOI] [PubMed] [Google Scholar]
  • 71.Evangelista M, Tian H, de Sauvage FJ. The Hedgehog signaling pathway in cancer. Clin Cancer Res. 2006;12:5924–8. [DOI] [PubMed] [Google Scholar]
  • 72.Leong KG, Karsan A. Recent insights into the role of Notch signaling in tumorigenesis. Blood. 2006;107:2223–33. [DOI] [PubMed] [Google Scholar]
  • 73.Yoshida R, Nagata M, Nakayama H, Niimori-Kita K, Hassan W, Tanaka T, Shinohara M, Ito T. The pathological significance of Notch1 in oral squamous cell carcinoma. Lab Invest. 2013;93:1068–81. [DOI] [PubMed] [Google Scholar]
  • 74.Kageyama R, Ohtsuka T, Tomita K. The bHLH gene Hes1 regulates differentiation of multiple cell types. Mol Cells. 2000;10:1–7. [DOI] [PubMed] [Google Scholar]
  • 75.Kim RH, Kang MK, Shin KH, Oo ZM, Han T, Baluda MA, Park NH. Bmi-1 cooperates with human papillomavirus type 16 E6 to immortalize normal human oral keratinocytes. Exp Cell Res. 2007;313:462–72. [DOI] [PubMed] [Google Scholar]
  • 76.Kang MK. Polycomb group proteins: New targets of anti-cancer therapy. Cell Cycle. 2010;9:2704. [DOI] [PubMed] [Google Scholar]
  • 77.Chen D, Wu M, Li Y, Chang I, Yuan Q, Ekimyan-Salvo M, Deng P, Yu B, Yu Y, Dong J, Szymanski JM, Ramadoss S, Li J, Wang CY. Targeting BMI1+ cancer stem cells overcomes chemoresistance and inhibits metastases in squamous cell carcinoma. Cell Stem Cell. 2017;20:621–34, e626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wang Q, Li Z, Wu Y, Huang R, Zhu Y, Zhang W, Wang Y, Cheng J. Pharmacological inhibition of Bmi1 by PTC-209 impaired tumor growth in head neck squamous cell carcinoma. Cancer Cell Int. 2017;17:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 2004;4:118–32. [DOI] [PubMed] [Google Scholar]
  • 80.Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119:1420–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fan F, Samuel S, Evans KW, Lu J, Xia L, Zhou Y, Sceusi E, Tozzi F, Ye XC, Mani SA, Ellis LM. Overexpression of snail induces epithelial-mesenchymal transition and a cancer stem cell-like phenotype in human colorectal cancer cells. Cancer Med. 2012;1:5–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zeisberg M, Neilson EG. Biomarkers for epithelial-mesenchymal transitions. J Clin Invest. 2009;119:1429–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dieter SM, Ball CR, Hoffmann CM, Nowrouzi A, Herbst F, Zavidij O, Abel U, Arens A, Weichert W, Brand K, Koch M, Weitz J, Schmidt M, von Kalle C, Glimm H. Distinct types of tumor-initiating cells form human colon cancer tumors and metastases. Cell Stem Cell. 2011;9:357–65. [DOI] [PubMed] [Google Scholar]
  • 84.Liu ZH, Dai XM, Du B. Hes1: A key role in stemness, metastasis and multidrug resistance. Cancer Biol Ther. 2015;16:353–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chu PY, Hu FW, Yu CC, Tsai LL, Yu CH, Wu BC, Chen YW, Huang PI, Lo WL. Epithelial-mesenchymal transition transcription factor ZEB1/ZEB2 co-expression predicts poor prognosis and maintains tumor-initiating properties in head and neck cancer. Oral Oncol. 2013;49:34–41. [DOI] [PubMed] [Google Scholar]
  • 86.Kathawala RJ, Gupta P, Ashby CR Jr., Chen ZS. The modulation of ABC transporter-mediated multidrug resistance in cancer: A review of the past decade. Drug Resist Updat. 2015;18:1–17. [DOI] [PubMed] [Google Scholar]
  • 87.Moitra K Overcoming multidrug resistance in cancer stem cells. Biomed Res Int. 2015;2015:635745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Shukla S, Ohnuma S, Ambudkar SV. Improving cancer chemotherapy with modulators of ABC drug transporters. Curr Drug Targets. 2011;12:621–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Cui H, Zhang AJ, Chen M, Liu JJ. ABC transporter inhibitors in reversing multidrug resistance to chemotherapy. Curr Drug Targets. 2015;16:1356–71. [DOI] [PubMed] [Google Scholar]
  • 90.Golebiewska A, Brons NH, Bjerkvig R, Niclou SP. Critical appraisal of the side population assay in stem cell and cancer stem cell research. Cell Stem Cell. 2011;8:136–47. [DOI] [PubMed] [Google Scholar]
  • 91.Song J, Chang I, Chen Z, Kang M, Wang CY. Characterization of side populations in HNSCC: Highly invasive, chemoresistant and abnormal wnt signaling. PLoS One. 2010; 5:e11456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yanamoto S, Kawasaki G, Yamada S, Yoshitomi I, Kawano T, Yonezawa H, Rokutanda S, Naruse T, Umeda M. Isolation and characterization of cancer stem-like side population cells in human oral cancer cells. Oral Oncol. 2011;47:855–60. [DOI] [PubMed] [Google Scholar]
  • 93.Zhang P, Zhang Y, Mao L, Zhang Z, Chen W. Side population in oral squamous cell carcinoma possesses tumor stem cell phenotypes. Cancer Lett. 2009;277:227–34. [DOI] [PubMed] [Google Scholar]
  • 94.Liu C, Kelnar K, Liu BG, Chen X, Calhoun-Davis T, Li HW, Patrawala L, Yan H, Jeter C, Honorio S, Wiggins JF, Bader AG, Fagin R, Brown D, Tang DAG. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 2011;17:211–U105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Song SJ, Poliseno L, Song MS, Ala U, Webster K, Ng C, Beringer G, Brikbak NJ, Yuan X, Cantley LC, Richardson AL, Pandolfi PP. MicroRNA-antagonism regulates breast cancer stemness and metastasis via TET-family-dependent chromatin remodeling. Cell. 2013;154:311–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lee SH, Lee CR, Rigas NK, Kim RH, Kang MK, Park NH, Shin KH. Human papillomavirus 16 (HPV16) enhances tumor growth and cancer stemness of HPV-negative oral/oropharyngeal squamous cell carcinoma cells via miR-181 regulation. Papillomavirus Res. 2015;1:116–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kooistra SM, Helin K. Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol. 2012;13:297–311. [DOI] [PubMed] [Google Scholar]
  • 98.Agger K, Cloos PA, Rudkjaer L, Williams K, Andersen G, Christensen J, Helin K. The H3K27me3 demethylase JMJD3 contributes to the activation of the INK4A-ARF locus in response to oncogene- and stress-induced senescence. Genes Dev. 2009;23:1171–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hsia DA, Tepper CG, Pochampalli MR, Hsia EYC, Izumiya C, Huerta SB, Wright ME, Chen HW, Kung HJ, Izumiya Y. KDM8, a H3K36me2 histone demethylase that acts in the cyclin A1 coding region to regulate cancer cell proliferation. Proc Natl Acad Sci USA. 2010;107:9671–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ye L, Fan Z, Yu B, Chang J, Al Hezaimi K, Zhou X, Park NH, Wang CY. Histone demethylases KDM4B and KDM6B promote osteogenic differentiation of human MSCs. Cell Stem Cell. 2012;11:50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Amente S, Lania L, Majello B. The histone LSD1 demethylase in stemness and cancer transcription programs. Biochim Biophys Acta. 2013;1829:981–6. [DOI] [PubMed] [Google Scholar]
  • 102.Harmeyer KM, Facompre ND, Herlyn M, Basu D. JARID1 histone demethylases: Emerging targets in cancer. Trends Cancer. 2017;3:713–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Metzger E, Stepputtis SS, Strietz J, Preca BT, Urban S, Willmann D, Allen A, Zenk F, Iovino N, Bronsert P, Proske A, Follo M, Boerries M, Stickeler E, Xu J, Wallace MB, Stafford JA, Kanouni T, Maurer J, Schule R. KDM4 inhibition targets breast cancer stem-like cells. Cancer Res. 2017;77:5900–12. [DOI] [PubMed] [Google Scholar]
  • 104.Taube JH, Sphyris N, Johnson KS, Reisenauer KN, Nesbit TA, Joseph R, Vijay GV, Sarkar TR, Bhangre NA, Song JJ, Chang JT, Lee MG, Soundararajan R, Mani SA. The H3K27me3-demethylase KDM6A is suppressed in breast cancer stem-like cells, and enables the resolution of bivalency during the mesenchymal-epithelial transition. Oncotarget. 2017;8:65548–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yamamoto S, Tateishi K, Kudo Y, Yamamoto K, Isagawa T, Nagae G, Nakatsuka T, Asaoka Y, Ijichi H, Hirata Y, Otsuka M, Ikenoue T, Aburatani H, Omata M, Koike K. Histone demethylase KDM4C regulates sphere formation by mediating the cross talk between Wnt and Notch pathways in colonic cancer cells. Carcinogenesis. 2013;34:2380–8. [DOI] [PubMed] [Google Scholar]
  • 106.Yan Q, Zhang SM, Meeth K, Micevic G, Bosenberg M. Histone demethylase KDM5B is critical for PI3K-AKT-mTOR signaling and stemness of melanoma. Faseb J. 2017;31(1):468.1. [Google Scholar]
  • 107.Sherry-Lynes MM, Sengupta S, Kulkarni S, Cochran BH. Regulation of the JMJD3 (KDM6B) histone demethylase in glioblastoma stem cells by STAT3. PLoS One. 2017;12(4):e0174775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Li J, Yu B, Deng P, Cheng Y, Yu Y, Kevork K, Ramadoss S, Ding X, Li X, Wang CY. KDM3 epigenetically controls tumorigenic potentials of human colorectal cancer stem cells through Wnt/β-catenin signalling. Nat Commun. 2017;8:15146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ramadoss S, Chen X, Wang CY. Histone demethylase KDM6B promotes epithelial-mesenchymal transition. J Biol Chem. 2012;287:44508–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Chang BS, Chen Y, Zhao YM, Bruick RK. JMJD6 is a histone arginine demethylase. Science. 2007;318:444–7. [DOI] [PubMed] [Google Scholar]
  • 111.Webby CJ, Wolf A, Gromak N, Dreger M, Kramer H, Kessler B, Nielsen ML, Schmitz C, Butler DS, Yates JR, Delahunty CM, Hahn P, Lengeling A, Mann M, Proudfoot NJ, Schofield CJ, Bottger A. JMJD6 catalyses lysyl-hydroxylation of U2AF65, a protein associated with RNA splicing. Science. 2009;325:90–3. [DOI] [PubMed] [Google Scholar]
  • 112.Lee YF, Miller LD, Chan XB, Black MA, Pang B, Ong CW, Salto-Tellez M, Liu ET, Desai KV. JMJD6 is a driver of cellular proliferation and motility and a marker of poor prognosis in breast cancer. Breast Cancer Res. 2012;14(3):R85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhang J, Ni SS, Zhao WL, Dong XC, Wang JL. High expression of JMJD6 predicts unfavorable survival in lung adenocarcinoma. Tumour Biol. 2013;34:2397–401. [DOI] [PubMed] [Google Scholar]
  • 114.Wang F, He L, Huangyang PW, Liang J, Si WZ, Yan RR, Han X, Liu SM, Gui B, Li WJ, Miao D, Jing C, Liu ZH, Pei F, Sun LY, Shang YF. JMJD6 promotes colon carcinogenesis through negative regulation of p53 by hydroxylation. PLoS Biol. 2014;12(3):e1001819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Boeckel JN, Guarani V, Koyanagi M, Roexe T, Lengeling A, Schermuly RT, Gellert P, Braun T, Zeiher A, Dimmeler S. Jumonji domain-containing protein 6 (JMJD6) is required for angiogenic sprouting and regulates splicing of VEGF-receptor 1. Proc Natl Acad Sci USA. 2011;108:3276–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Liu T, Xu FH, Du XL, Lai DM, Liu TJ, Zhao YR, Huang Q, Jiang LZ, Huang WB, Cheng WW, Liu ZX. Establishment and characterization of multi-drug resistant, prostate carcinoma-initiating stem-like cells from human prostate cancer cell lines 22RV1. Mol Cell Biochem. 2010;340:265–73. [DOI] [PubMed] [Google Scholar]
  • 117.Zhang X, Han B, Huang J, Zheng B, Geng Q, Aziz F, Dong Q. Prognostic significance of Oct4 expression in adenocarcinoma of the lung. Japan J Clin Oncol. 2010;40:961–6. [DOI] [PubMed] [Google Scholar]
  • 118.Kumar SM, Liu S, Lu H, Zhang H, Zhang PJ, Gimotty PA, Guerra M, Guo W, Xu X. Acquired cancer stem cell phenotypes through Oct4-mediated dedifferentiation. Oncogene. 2012;31:4898–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chin AR, Wang SE. Cytokines driving breast cancer stemness. Mol Cell Endocrinol. 2014;382:598–602. [DOI] [PubMed] [Google Scholar]
  • 120.Prokopchuk O, Liu Y, Henne-Bruns D, Kornmann M. Interleukin-4 enhances proliferation of human pancreatic cancer cells: Evidence for autocrine and paracrine actions. Br J Cancer. 2005;92:921–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Li BH, Yang MZ, Li PD, Yuan Q, Liu XH, Yuan J, Zhang WH. IL-4/Stat6 activities correlate with apoptosis and metastasis in colon cancer cells. Biochem Bioph Res Commun. 2008;369:554–60. [DOI] [PubMed] [Google Scholar]
  • 122.Conticello C, Pedini F, Zeuner A, Patti M, Zerilli M, Stassi G, Messina A, Peschle C, De Maria R. IL-4 protects tumor cells from anti-CD95 and chemotherapeutic agents via up-regulation of antiapoptotic proteins. J Immunol. 2004;172:5467–77. [DOI] [PubMed] [Google Scholar]
  • 123.Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Lovino F, Tripodo C, Russo A, Gulotta G, Medema JP, Stassi G. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell. 2007;1:389–402. [DOI] [PubMed] [Google Scholar]
  • 124.Di Stefano AB, Iovino F, Lombardo Y, Eterno V, Hoger T, Dieli F, Stassi G, Todaro M. Survivin is regulated by interleukin-4 in colon cancer stem cells. J Cell Physiol. 2010;225:555–61. [DOI] [PubMed] [Google Scholar]
  • 125.Kim MS, Cho HI, Yoon HJ, Ahn YH, Park EJ, Jin YH, Jang YK. JIB-04, a small molecule histone demethylase inhibitor, selectively targets colorectal cancer stem cells by inhibiting the Wnt/β-catenin signaling pathway. Sci Rep. 2018;8:6611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sakaki H, Okada M, Kuramoto K, Takeda H, Watarai H, Suzuki S, Seino S, Seino M, Ohta T, Nagase S, Kurachi H, Kitanaka C. GSKJ4, a selective jumonji H3K27 demethylase inhibitor, effectively targets ovarian cancer stem cells. Anticancer Res. 2015;35:6607–14. [PubMed] [Google Scholar]
  • 127.Zhao W, Wang L, Han H, Jin K, Lin N, Guo T, Chen Y, Cheng H, Lu F, Fang W, Wang Y, Xing B, Zhang Z. 1B50–1, a mAb raised against recurrent tumor cells, targets liver tumor-initiating cells by binding to the calcium channel α2δ1 subunit. Cancer Cell. 2013;23:541–56. [DOI] [PubMed] [Google Scholar]
  • 128.Zhu H, Zhang H, Jin F, Fang M, Huang M, Yang CS, Chen T, Fu L, Pan Z. Elevated Orai1 expression mediates tumor-promoting intracellular Ca2+ oscillations in human esophageal squamous cell carcinoma. Oncotarget. 2014;5(11):3455–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sun J, Lu F, He H, Shen J, Messina J, Mathew R, Wang D, Sarnaik AA, Chang WC, Kim M, Cheng H, Yang S. Stim1- and Orai1-mediated Ca2+ oscillation orchestrates invadopodium formation and melanoma invasion. J Cell Biol. 2014;207:535–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Berridge MJ, Bootman MD, Roderick HL. Calcium signalling: Dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol. 2003;4:517–29. [DOI] [PubMed] [Google Scholar]
  • 131.Monteith GR, McAndrew D, Faddy HM, Roberts-Thomson SJ. Calcium and cancer: Targeting Ca2+ transport. Nat Rev Cancer. 2007;7:519–30. [DOI] [PubMed] [Google Scholar]
  • 132.Roderick HL, Cook SJ. Ca2+ signalling checkpoints in cancer: Remodelling Ca2+ for cancer cell proliferation and survival. Nat Rev Cancer. 2008;8:361–75. [DOI] [PubMed] [Google Scholar]
  • 133.Prevarskaya N, Skryma R, Shuba Y. Calcium in tumour metastasis: New roles for known actors. Nat Rev Cancer. 2011;11:609–18. [DOI] [PubMed] [Google Scholar]
  • 134.Srikanth S, Gwack Y. Molecular regulation of the pore component of CRAC channels, Orai1 In: Murali P, ed. Current topics in membranes. Cambridge, MA: Academic Press; 2013. pp. 181–207. [DOI] [PubMed] [Google Scholar]
  • 135.Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, Tanasa B, Hogan PG, Lewis RS, Daly M, Rao A. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature. 2006;441:179–85. [DOI] [PubMed] [Google Scholar]
  • 136.Prakriya M, Feske S, Gwack Y, Srikanth S, Rao A, Hogan PG. Orai1 is an essential pore subunit of the CRAC channel. Nature. 2006;443:230–3. [DOI] [PubMed] [Google Scholar]
  • 137.Gwack Y, Srikanth S, Feske S, Cruz-Guilloty F, Oh-hora M, Neems DS, Hogan PG, Rao A. Biochemical and functional characterization of Orai proteins. J Biol Chem. 2007;282:16232–43. [DOI] [PubMed] [Google Scholar]
  • 138.Parekh AB. Functional consequences of activating store-operated CRAC channels. Cell Calcium. 2007;42:111–21. [DOI] [PubMed] [Google Scholar]
  • 139.Parekh AB, Putney JW Jr. Store-operated calcium channels. Phys Rev. 2005;85:757–810. [DOI] [PubMed] [Google Scholar]
  • 140.Srikanth S, Gwack Y. Orai1-NFAT signalling pathway triggered by T-cell receptor stimulation. Mol Cells. 2013;35:182–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Monteith GR, McAndrew D, Faddy HM, Roberts-Thomson SJ. Calcium and cancer: Targeting Ca2+ transport. Nat Rev Cancer. 2007;7:519–30. [DOI] [PubMed] [Google Scholar]
  • 142.Flourakis M, Lehen’kyi V, Beck B, Raphael M, Vandenberghe M, Abeele FV, Roudbaraki M, Lepage G, Mauroy B, Romanin C, Shuba Y, Skryma R, Prevarskaya N. Orai1 contributes to the establishment of an apoptosis-resistant phenotype in prostate cancer cells. Cell Death Dis. 2010;1:e75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Yang S, Zhang JJ, Huang XY. Orai1 and STIM1 are critical for breast tumor cell migration and metastasis. Cancer Cell. 2009;15:124–34. [DOI] [PubMed] [Google Scholar]
  • 144.Liu H, Hughes JD, Rollins S, Chen B, Perkins E. Calcium entry via Orai1 regulates glioblastoma cell proliferation and apoptosis. Exp Mol Pathol. 2011;91:753–60. [DOI] [PubMed] [Google Scholar]
  • 145.Kim JH, Lkhagvadorj S, Lee MR, Hwang KH, Chung HC, Jung JH, Cha SK, Eom M. Orai1 and STIM1 are critical for cell migration and proliferation of clear cell renal cell carcinoma. Biochem Biophys Res Commun. 2014;448:76–82. [DOI] [PubMed] [Google Scholar]
  • 146.Lee SH, Rigas NK, Lee CR, Bang A, Srikanth S, Gwack Y, Kang MK, Kim RH, Park NH, Shin KH. Orai1 promotes tumor progression by enhancing cancer stemness via NFAT signaling in oral/oropharyngeal squamous cell carcinoma. Oncotarget. 2016;7(28):43239–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wang L, Hao J, Zhang Y, Yang Z, Cao Y, Lu W, Shu Y, Jiang L, Hu Y, Lv W, Liu Y, Dong P. Orai1 mediates tumor-promoting store-operated Ca2+ entry in human gastrointestinal stromal tumors via c-KIT and the extracellular signal-regulated kinase pathway. Tumour Biol. 2017;39:1010428317691426. [DOI] [PubMed] [Google Scholar]
  • 148.Xia J, Wang H, Huang H, Sun L, Dong S, Huang N, Shi M, Bin J, Liao Y, Liao W. Elevated Orai1 and STIM1 expressions upregulate MACC1 expression to promote tumor cell proliferation, metabolism, migration, and invasion in human gastric cancer. Cancer Lett. 2016;381:31–40. [DOI] [PubMed] [Google Scholar]
  • 149.Deng W, Wang J, Zhang J, Cai J, Bai Z, Zhang Z. Orai1, a direct target of microRNA-519, promotes progression of colorectal cancer via Akt/GSK3β signaling pathway. Dig Dis Sci. 2016;61:1553–60. [DOI] [PubMed] [Google Scholar]
  • 150.Zhan ZY, Zhong LX, Feng M, Wang JF, Liu DB, Xiong JP. Over-expression of Orai1 mediates cell proliferation and associates with poor prognosis in human non-small-cell lung carcinoma. Int J Clin Exp Pathol. 2015;8:5080–8. [PMC free article] [PubMed] [Google Scholar]
  • 151.Lee SJ, Leoni G, Neumann PA, Chun J, Nusrat A, Yun CC. Distinct phospholipase C-β isozymes mediate lysophosphatidic acid receptor 1 effects on intestinal epithelial homeostasis and wound closure. Mol Cell Biol. 2013;33:2016–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Gwack Y, Feske S, Srikanth S, Hogan PG, Rao A. Signalling to transcription: Store-operated Ca2+ entry and NFAT activation in lymphocytes. Cell Calcium. 2007;42:145–56. [DOI] [PubMed] [Google Scholar]
  • 153.Maul-Pavicic A, Chiang SC, Rensing-Ehl A, Jessen B, Fauriat C, Wood SM, Sjoqvist S, Hufnagel M, Schulze I, Bass T, Schamel WW, Fuchs S, Pircher H, McCarl CA, Mikoshiba K, Schwarz K, Feske S, Bryceson YT, Ehl S. Orai1-mediated calcium influx is required for human cytotoxic lymphocyte degranulation and target-cell lysis. Proc Natl Acad Sci USA. 2011;108:3324–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Daniel C, Gerlach K, Väth M, Neurath MF, Weigmann B. Nuclear factor of activated T cells—A transcription factor family as critical regulator in lung and colon cancer. Int J Cancer. 2014;134:1767–75. [DOI] [PubMed] [Google Scholar]
  • 155.Mancini M, Toker A. NFAT proteins: Emerging roles in cancer progression. Nat Rev Cancer. 2009;9:810–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Mognol GP, Carneiro FR, Robbs BK, Faget DV, Viola JP. Cell cycle and apoptosis regulation by NFAT transcription factors: New roles for an old player. Cell Death Dis. 2016;7:e2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Buchholz M, Schatz A, Wagner M, Michl P, Linhart T, Adler G, Gress TM, Ellenrieder V. Overexpression of c-myc in pancreatic cancer caused by ectopic activation of NFATc1 and the Ca2+/calcineurin signaling pathway. EMBO J. 2006;25:3714–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Velupillai P, Sung CK, Tian Y, Dahl J, Carroll J, Bronson R, Benjamin T. Polyoma virus-induced osteosarcomas in in-bred strains of mice: Host determinants of metastasis. PLoS Pathog. 2010;6:e1000733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Liu JF, Zhao SH, Wu SS. Depleting NFAT1 expression inhibits the ability of invasion and migration of human lung cancer cells. Cancer Cell Int. 2013;13:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Chen ZL, Zhao SH, Wang Z, Qiu B, Li BZ, Zhou F, Tan XG, He J. Expression and unique functions of four nuclear factor of activated T cells isoforms in non-small-cell lung cancer. Chin J Cancer. 2011;30:62–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Gerlach K, Daniel C, Lehr HA, Nikolaev A, Gerlach T, Atreya R, Rose-John S, Neurath MF, Weigmann B. Transcription factor NFATc2 controls the emergence of colon cancer associated with IL-6-dependent colitis. Cancer Res. 2012;72:4340–50. [DOI] [PubMed] [Google Scholar]
  • 162.Perotti V, Baldassari P, Molla A, Vegetti C, Bersani I, Maurichi A, Santinami M, Anichini A, Mortarini R. NFATc2 is an intrinsic regulator of melanoma dedifferentiation. Oncogene. 2016;35:2862–72. [DOI] [PubMed] [Google Scholar]
  • 163.Hessmann E, Zhang JS, Chen NM, Hasselluhn M, Liou GY, Storz P, Ellenrieder V, Billadeau DD, Koenig A. NFATc4 regulates Sox9 gene expression in acinar cell plasticity and pancreatic cancer initiation. Stem Cells Int. 2016;2016:5272498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Xiao ZJ, Liu J, Wang SQ, Zhu Y, Gao XY, Tin VP, Qin J, Wang JW, Wong MP. NFATc2 enhances tumor-initiating phenotypes through the NFATc2/SOX2/ALDH axis in lung adenocarcinoma. eLife Digest. 2017;6:e26733. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES