Abstract
Head and neck cancer presents primarily as head and neck squamous cell carcinoma (HNSCC), a debilitating malignancy fraught with high morbidity, poor survival rates, and limited treatment options. Mounting evidence indicates that the Wnt/β-catenin signaling pathway plays important roles in the pathobiology of HNSCC. Wnt/β-catenin signaling affects multiple cellular processes that endow cancer cells with the ability to maintain and expand immature stem-like phenotypes, proliferate, extend survival, and acquire aggressive characteristics by adopting mesenchymal traits. A central component of canonical Wnt signaling is β-catenin, which balances its role as a structural component of E-cadherin junctions with its function as a transcriptional coactivator of numerous target genes. Recent genomic characterization of head and neck cancer revealed that while β-catenin is not frequently mutated in HNSCC, its activity is unchecked by more common mutations in genes encoding upstream regulators of β-catenin, NOTCH1, FAT1, and AJUBA. Wnt/β-catenin signaling affects a wide range epigenetic and transcriptional activities, mediated by the interaction of β-catenin with different transcription factors and transcriptional coactivators and corepressors. Furthermore, Wnt/β-catenin functions in a network with many signaling and metabolic pathways that modulate its activity. In addition to its effects on tumor epithelia, β-catenin activity regulates the tumor microenvironment by regulating extracellular matrix remodeling, fibrotic processes, and immune response. These multifunctional oncogenic effects of β-catenin make it an attractive bona fide target for HNSCC therapy.
Keywords: carcinoma, epigenomics, stem cells, signal transduction, cancer microenvironment, therapeutics
Introduction
The Wnt signaling pathway plays critical roles in development, adult homeostasis, and cancer (reviewed in Zhan et al. 2017). This signaling cascade comprises distinct branches: the Wnt/β-catenin or the canonical Wnt signaling pathway, the planar cell polarity (Wnt-PCP) pathway, and the Wnt-calcium signaling pathway (Gordon and Nusse 2006). To date, most studies have focused on the Wnt/β-catenin branch, revealing its prominent roles in diverse cellular processes that underlie human cancers (reviewed in Nusse and Clevers 2017).
Deregulated Wnt/β-catenin activity has been associated with diverse cancer types (reviewed in Nusse and Clevers 2017). Mounting evidence from cellular and animal models coupled with genomic and molecular characterization of head and neck cancers has added this malignancy to the list of tumors affected by Wnt/β-catenin signaling. To date, several excellent reviews have described the involvement of the Wnt/β-catenin pathway in the pathobiology of head and neck cancer (reviewed in Castilho and Gutkind 2014; Cancer Genome Atlas 2015; Beck and Golemis 2016). This review summarizes seminal discoveries and recent advances in Wnt/β-catenin signaling in cancer while highlighting their relevance to head and neck squamous cell carcinoma (HNSCC), and it describes current strategies to inhibit Wnt/β-catenin signaling.
Wnt/β-catenin Signaling in Head and Neck Cancer
Head and neck cancer is a complex malignancy involving multiple tissue sites with distinct biology, extensive intra- and intertumor heterogeneity, and poorly understood intercellular communication (Hedberg et al. 2016; Puram et al. 2017). On the genomic level, HNSCC is characterized by somatic alterations grouped into human papillomavirus (HPV)–positive and HPV-negative tumors with distinct epidemiologic, molecular, and clinical features (reviewed in Cancer Genome Atlas 2015; Beck and Golemis 2016).
Mounting evidence indicates that Wnt/β-catenin signaling plays pivotal roles in the pathobiology of HPV-negative and HPV-positive HNSCC (Yang et al. 2006; Chang et al. 2013; reviewed in Castilho and Gutkind 2014; Gonzalez-Moles et al. 2014; Cancer Genome Atlas 2015). Increased basal cell expression of Wnt1 and Wnt pathway activation by epigenetic alterations of Wnt inhibitory factors, SRFP, WIF, and DKK3, has been reported (Pannone et al. 2010). Augmented tissue levels of β-catenin in HNSCC have been aligned with its increased transcriptional activity (Rampias et al. 2010; Hu et al. 2015), and inappropriate stabilization of β-catenin has been correlated with de-differentiation and poor prognosis (Hu et al. 2015; Padhi et al. 2015). Notably, HNSCC has been shown to rely on high Wnt/β-catenin activity to promote epigenetic changes associated with open chromatin structure and induction of stem cell gene signatures (Wend et al. 2013).
While mutations in β-catenin are infrequent in HNSCC, alterations in genes encoding upstream regulators, NOTCH1, FAT1, and AJUBA, converge on β-catenin signaling and enhance its nuclear activity (Cancer Genome Atlas 2015; Beck and Golemis 2016). To date, mutations in AJUBA and FAT1 have been almost exclusively associated with HPV-negative tumors and linked to the loss of epithelial differentiation programs. In HPV-positive disease, deregulated Wnt/β-catenin signaling has been linked to viral oncoproteins E6- and E7- (E6/E7)–mediated activation of β-catenin (reviewed in Castilho and Gutkind 2014; Rampias and Psyrri 2014). Specifically, oncoprotein E6 has been shown to drive nuclear translocation of β-catenin through the epidermal growth factor receptor (EGFR)–dependent mechanism in oropharyngeal squamous cell carcinoma (OPSCC) (Hu et al. 2015). Inhibition of E6 expression with small interfering RNA and downregulation of EGFR activity with erlotinib abrogated nuclear localization of β-catenin and EGFR phosphorylation concomitant with reduced invasive properties of HNSCC-positive cell lines in vitro (Hu et al. 2015). Also, E6/E7 have been reported to induce nuclear translocation of β-catenin by inhibiting the Siah E3 ubiquitin ligase protein (Rampias et al. 2010). Nonetheless, more studies are needed to better understand the role of Wnt/β-catenin signaling in HPV-positive HNSCC, as they have lagged behind HPV-negative disease due to limited tumor specimens and associated clinical data (Cancer Genome Atlas 2015; Beck and Golemis 2016).
Wnt/β-catenin Signaling: Components and Regulation
The Wnt/β-catenin pathway is pivotal in controlling cell fate determination, cell proliferation, cell survival, and differentiation (reviewed in Nusse and Clevers 2017). A central effector of Wnt signaling is β-catenin that, under conditions of epithelial tissue homeostasis, is maintained at low levels in the cytoplasm while being prominently localized to E-cadherin junctional complexes or adherens junctions (AJs). Steady-state levels of cytoplasmic β-catenin are controlled by a complex of proteins that include adenomatous polyposis coli (APC), the scaffold protein Axin1, glycogen synthase kinase 3β (GSK3β), casein kinase 1α (CK1α), protein phosphatase 2A (PP2A), and additional components that sequester excess cytoplasmic β-catenin and target it for ubiquitination and proteosomal degradation via the β-TrCP, an E3 ubiquitin ligase subunit (Fig. 1). The Wnt/β-catenin pathway is activated by the binding of the Wnt ligand to a 7-pass transmembrane Frizzled (Fzd) receptor and its coreceptor, the low-density lipoprotein receptor-related protein 6 (Lrp6) or Lrp5. This results in a conformational change and phosphorylation of Lrp5/6, leading to the sequestration of Axin 1 and inactivation of the destruction complex. In the absence of the destruction complex, the newly synthesized unphosphorylated β-catenin accumulates in the cytoplasm and translocates to the nucleus, where it displaces repressors from the promoters of target genes and interacts with transcription factors to drive their expression. The most prominent transcriptional partner of β-catenin is the T-cell factor (TCF) and lymphoid enhancer-binding factor (LEF1) family of DNA-binding proteins that together induce the expression genes encoding myc, cyclin D1, axin 2, and survivin, among others.
Transcriptional activity of β-catenin is determined by its cellular abundance and nuclear translocation and by interactions with transcription factors and transcriptional repressors and coactivators. β-Catenin affects diverse cellular functions through its complex protein structure that consists of 12 repeats of approximately 40 amino acids long, named armadillo repeats that together form a single, rigid protein domain, the armadillo (ARM) domain. Phosphorylation of the N-terminal region of β-catenin facilitates its binding to β-TrCP E3 ubiquitin ligase that targets β-catenin for degradation by the proteasome (Fig. 1). In contrast, the C-terminal region interacts with different binding partners and is critical for Wnt signaling. For some of these transcriptional partners, interactions have been mapped to specific domains of β-catenin in functional studies (Fig. 2) (reviewed in Valenta et al. 2012).
In addition to binding Tcf/Lef, β-catenin directly interacts with other transcription factors to affect gene expression and tumor cell behavior (Fig. 2A). Under limiting oxygen conditions, hypoxia-induced factor 1α (HIF1α) is upregulated in abundance and competes with Tcf for β-catenin binding. Furthermore, increased oxidative stress is accompanied by enhanced levels of reactive oxygen species (ROS), leading to upregulation of forkhead transcription factors FOXO that also compete with Tcf for β-catenin. The significance of these Lef/Tcf-independent transcription partners of β-catenin has not been extensively explored in HNSCC, although hypoxia plays an important role in the pathobiology of this malignancy and in treatment resistance (Gammon and Mackenzie 2016; reviewed in Valenta et al. 2012).
Nuclear β-catenin has been shown to interact with numerous coactivators for which its binding domains have been mapped and give rise to distinct functional outcomes (Fig. 2B). Additional key binding partners include Bcl9, shown to promote early stages of intestinal cancer by controlling a subset of β-catenin target genes with roles in epithelial-to-mesenchymal transition (EMT) and invasion (Brembeck et al. 2011). Also, Kindlin2 binds to β-catenin and promotes Axin2-Snail target gene expression critical for cancer stem cells (CSCs), EMT, invasion, and metastases (Yu et al. 2012). Moreover, β-catenin interacts with Klf4 and c-myc to regulate telomerase reverse transcriptase (TERT) gene expression required for telomere maintenance in cancer (Hoffmeyer et al. 2012). Nuclear β-catenin also interacts with the effectors of the Hippo pathway YAP1 and TAZ to regulate the expression of genes associated with cancer, including SOX2, SNAI2, and BIRC5 (Rosenbluh et al. 2012). Notably, β-catenin has a central role in the epigenetic modification of chromatin (reviewed in Mosimann et al. 2009). Nuclear β-catenin interacts with the histone acetyltransferase cAMP-responsive element binding protein (CBP) to maintain a pluripotent state in embryonic tissues (Ma et al. 2005). In HNSCC, the β-catenin/CBP complex has been shown to recruit histone methyltransferase and myeloid/lymphoid or mixed-lineage leukemia 1 (MLL1) to methylate H3K4me3 and activate stem cell–related genes (Wend et al. 2013). Another coactivating partner of β-catenin is p300, a histone acetyltransferase closely related to CBP but with distinct patterns of gene activation (Ma et al. 2005) (Figs. 1 and 2B).
Because of its central roles in diverse cellular processes, levels of β-catenin are highly regulated. Besides the destruction complex, cytoplasmic levels of β-catenin are regulated by YAP1 and TAZ that target it for degradation by β-TrCP E3 ubiquitin ligase (Azzolin et al. 2014). Another level of regulation of β-catenin is imposed by E-cadherin via recycling of E-cadherin/β-catenin complexes from the cell membrane, a process likely to augment the cytoplasmic pool of β-catenin (Heuberger and Birchmeier 2010) (Fig. 1). Furthermore, β-catenin regulates its own abundance by affecting the expression Wnt/β-catenin pathway components, including its antagonists Dickkopf-1 (Dkk-1) and E3 ubiquitin ligases ZNRF3 and RNF43 that induce endocytosis of Wnt receptors (Gonzalez-Sancho et al. 2005; Hao et al. 2012; Koo et al. 2012).
Wnt/β-catenin signaling is dynamically regulated through posttranslational modifications, such as palmitoylation and glycosylation, which affect conformation, secretion, stability, and activity of its components (Komekado et al. 2007). Wnt secretion is also regulated by Wntless, a G protein–coupled receptor GPR177 of the retromer complex, through Wntless-containing exosomes (McGough and Vincent 2016). Also, the activity of the Wnt/β-catenin pathway is modulated through the function of its inhibitors and agonists, such as its antagonist, Dickkopf-1 (Dkk-1), Wnt inhibitory factor (WIF1), and secreted Frizzled-related proteins (SFRP) (Gonzalez-Sancho et al. 2005; Katoh 2005). In addition, R-spondins (RSPO1–4) synergize with Wnt to activate β-catenin-mediated signaling by forming complexes with their receptors LGR4–6 that promote clearance of the RNF43/ZNRF3 E3 ligases to increase Wnt receptor levels to potentiate Wnt signaling (Carmon et al. 2011; Hao et al. 2012; Koo et al. 2012).
As a master regulatory pathway, Wnt/β-catenin signaling cross-talks with other signaling pathways. In mammary and intestinal tumors, Wnt/β-catenin collaborates with TGFβ on a transcriptional level, promoting EMT and fibrosis (Labbe et al. 2007). Also, Wnt/β-catenin collaborates with Notch during development and tumorigenesis (Kwon et al. 2011). Many HNSCC HPV-negative tumors harbor inactivating mutations in NOTCH1, which are associated with enhanced β-catenin activity (Duan et al. 2006; Cancer Genome Atlas 2015; Beck and Golemis 2016). Moreover, β-catenin interacts with the Hippo pathway and its effectors, transcriptional coactivators YAP/TAZ. The Hippo tumor suppressor pathway inhibits the oncogenic activities of YAP/TAZ through their retention in the cytosol, and hyperactivation of YAP/TAZ contributes to aggressive carcinomas, including HNSCC (Hiemer et al. 2015). Another prominent signaling pathway that interacts with Wnt/β-catenin signaling is the EGFR signaling cascade (Hu and Li 2010). The Wnt/β-catenin and EGFR signaling pathways have the ability to transactivate one another through Fzd receptors and via the PI3K/Akt or the ERK pathways (Hu and Li 2010). Furthermore, EGFR has been shown to form a complex with β-catenin in the cytoplasm (Schroeder et al. 2002) and to regulate its localization and stability in HNSCC (Lee et al. 2010). Last, PIK3CA is frequently amplified or harbors activating mutations in HNSCC (Lui et al. 2013). In colon cancer, PI3K and Wnt/β-catenin pathways converge at the level of the Akt substrate FOXO3a to induce IQ motif-containing GTPase activating protein IQGAP2 that inhibits E-cadherin adhesion and promotes cell migration and metastatic potential (Tenbaum et al. 2012).
Similar to other carcinomas, HNSCC cells undergo dramatic changes in their metabolic programs (reviewed in Cairns et al. 2011). During tumor progression, metabolic reprogramming is critical for the maintenance of CSCs and for communication between the tumor epithelia and the stroma (Porporato et al. 2014; Luo and Wicha 2015). As oxygen becomes limiting, cells stabilize HIF1α to upregulate glucose transporters and glycolytic enzymes to promote glycolysis, and β-catenin is diverted from Tcf to enhance transcriptional activity of HIF1α (Mazumdar et al. 2010).
One of the key metabolic processes that interacts with Wnt/β-catenin signaling is the protein N-glycosylation pathway, which modifies protumorigenic proteins with N-glycans (Liu et al. 2013) (reviewed in Varelas et al. 2014). Wnt/β-catenin signaling controls protein N-glycosylation through transcriptional regulation of the DPAGT1 gene at the first committed step in the lipid-linked assembly pathway in the endoplasmic reticulum (ER). Aberrant induction of DPAGT1 by β-catenin promotes a positive feedback loop with Wnt/β-catenin signaling that represses E-cadherin adhesion in HNSCC.
Wnt/β-catenin Signaling in Cancer Stem Cells
Maintenance of tissue homeostasis and repair depends on the activity of tissue-specific stem cells that display the capacity for long-term self-renewal and ability to give rise to 1 or more differentiated cell lineages. Cancer tissues, including HNSCC, also contain small subpopulations of cells with stem-like properties, or CSCs that give rise to tumors with hierarchal organization (Prince et al. 2007; Wend et al. 2013).
An acknowledged function of β-catenin is in the regulation of cell fates in development and cancer (reviewed in Holland et al. 2013). β-Catenin controls asymmetric cell division, a mechanism that generates cells capable of renewing themselves and committed progenitor cells that differentiate into a specified cell type by promoting unequal distribution of Dvl, Fzd, Axin, and APC in the cytoplasm of the mother cell (Lien and Fuchs 2014). The decision to divide or differentiate is controlled by a stem cell niche formed by extrinsic factors, including Wnt ligands, and by cells that produce them. The niche provides signals that promote stem cell phenotypes in multiple epithelial stem cell compartments during development (Yang et al. 2017). In HNSCC, endothelial cells have been shown to secrete EGF that promotes stem cell–like phenotypes, indicating that such a scenario is likely at play in this malignancy (Zhang et al. 2014).
In HNSCC, CSCs were first characterized by the expression of the CD44 surface marker and the Bmi1 oncogene (Prince et al. 2007). These cells were also shown to be marked by high aldehyde dehydrogenase activity, by the expression of c-Met and SOX2, and by the ability to efflux vital dyes and grow under nonadherent conditions as tumor spheres. Moreover, HNSCC CSCs had the ability to seed tumors at low numbers in nude mice and to drive cell expansion (Prince et al. 2007; Clay et al. 2010; Krishnamurthy et al. 2010; Wend et al. 2013). Also, HNSCC CSCs have been aligned with mesenchymal properties, resistance to treatments, and tumor recurrence (Nör et al. 2014; Chen et al. 2017).
To date, multiple components of Wnt/β-catenin signaling have been associated with CSCs. For instance, the R-spondin receptor Lgr5 is a potential CSC marker in intestinal stem cells that can promote tumor growth when APC is deleted (Schepers et al. 2012). In colon adenoma, Lgr5-positive cells can give rise to Lgr5-positive cells and other cell types. Furthermore, RAC1 is required for expansion of the Lgr5 population after APC loss (Myant et al. 2013). RAC1 activation drives ROS production and activates NF-κB signaling, which then enhances Wnt signaling (Myant et al. 2013).
Significantly, HNSCC CSCs were also characterized by an aberrant activation of Wnt/β-catenin activity (Wend et al. 2013; Lee et al. 2014). Recent studies provide evidence that a central role of β-catenin signaling in CSCs derives from its control of epigenetic changes that define distinct chromatin states (reviewed in Mosimann et al. 2009). Specifically, β-catenin promotes H3K4me3 by recruitment of CBP (Parker et al. 2008) and histone methyltransferase, MLL, to induce active chromatin and maintain immature cell phenotypes (Sierra et al. 2006). Indeed, increased interaction of β-catenin with CBP underlies HNSCC CSCs and disease progression (Zhou et al. 2012; Wend et al. 2013; Lenz and Kahn 2014; Chan et al. 2015). This ability to promote aggressive properties is specific to CBP, as recruitment of the p300 histone acetyltransferase by β-catenin drives cellular differentiation (Li et al. 2007).
Wnt/β-catenin Signaling in E-cadherin Adhesion and EMT
In addition to its signaling function, β-catenin serves as a major structural component of E-cadherin–mediated multiprotein complexes, or AJs that function in maintaining epithelial cell-cell adhesion and cell polarity. AJs are powerful tumor suppressors, in part, by inhibiting Wnt/β-catenin signaling (Birchmeier 1994). The major components of AJs include members of the catenin family: β-catenin, α-catenin, and p120 catenin. The C-terminal region of E-cadherin binds β-catenin, and E-cadherin/β-catenin complexes recruit α-catenin to facilitate interaction with the actin cytoskeleton, while the juxtamembrane domain of E-cadherin binds p120 catenin that regulates its turnover. Loss of either α-catenin or p120 from AJs leads to reduced cell-cell adhesion and the development of high-grade cancers (Benjamin and Nelson 2008; Kourtidis et al. 2015). In addition, in HNSCC, β-catenin indirectly inhibits AJs by inducing the expression of the N-glycosylation gene, DPAGT1, leading to their extensive N-glycosylation and destabilization (reviewed in Varelas et al. 2014). Moreover, aberrant activation of β-catenin in HNSCC is associated with mutations in FAT1, a member of the cadherin superfamily (reviewed in Cancer Genome Atlas 2015; Beck and Golemis 2016).
Inappropriate downregulation of E-cadherin is associated with EMT, a developmental program that involves loss of intercellular adhesion and apical-basal polarity, reorganization of cytoskeleton, and increased expression of vimentin, N-cadherin, and fibronectin concomitant with acquisition of mesenchymal traits (Kalluri and Weinberg 2009). Similar to other carcinomas, HNSCC cells adopt EMT to drive progression to advanced disease (Pectasides et al. 2014). Wnt/β-catenin signaling regulates key transcription factors responsible for EMT, including SNAI2 and the zinc finger E-box binding homeobox 1 protein (ZEB1), among others (Baum et al. 2008). Our own studies identified β-catenin–dependent increased expression of collagen triple helix containing 1 (CTHRC1) to be associated with enhanced HNSCC cell migration through collaboration with protein N-glycosylation and induction of the Wnt/PCP pathway (Liu et al. 2013). In addition, important regulators of EMT are microRNAs (miRNAs) that are frequently dysregulated via Wnt/β-catenin signaling (reviewed in Ghahhari and Babashah 2015). In HNSCC, deregulation of miRNA200b and miRNA15b has been linked to EMT and advanced disease (Sun et al. 2012).
Wnt/β-catenin Signaling in the Tumor Microenvironment
The tumor microenvironment (TME) undergoes dynamic alterations with cancer progression to advanced disease and contributes to cancer cell plasticity. Numerous studies highlight contributions of distinct components of the TME to HNSCC tumorigenesis (reviewed in Koontongkaew 2013). The TME includes the extracellular matrix (ECM), a noncellular structure that provides physical support and elasticity to tissues and transmits signals to epithelia by serving as ligands for receptors and regulates availability of growth factors and morphogens, such as Wnts. Many downstream targets of β-catenin are components of the ECM, including laminin, a key protein in the basement membrane, and lysyl oxidase and fibronectin that reside in the interstitial matrix, as well as invasion-associated genes such as MMP-9, MMP-74, and CD44 ligands (reviewed in Bonnans et al. 2014).
The TME also includes the stroma with fibroblasts and cells of the innate and adaptive immune systems with important roles in cancer progression. Wnt/β-catenin signaling is recognized for its inhibitory effects on innate and adaptive immune systems in cancer (Swafford and Manicassamy 2015), which are associated with immune tolerance, a major mechanism underlying failed host antitumor immune responses (reviewed in Pai et al. 2017). β-Catenin inhibits dendritic cells via activation of the ATF3 repressor activity, which downregulates the CCL4 chemokine required for the recruitment of CD103+ dendritic cells to the tumor (Swafford and Manicassamy 2015). Impaired recruitment of dendritic cells leads to diminished activation of CD8+ T cells. Indeed, β-catenin signaling has been aligned with non–T cell–inflamed tumors by promoting Treg persistence and activity, thus suppressing adaptive responses by reducing CD8+ T-cell proliferation, activation, and effector functions (Spranger and Gajewski 2015).
Wnt/β-catenin Signaling as a Therapeutic Target for HNSCC
Due to the important roles of β-catenin as a structural component of AJs and as a regulator of CSCs in cell renewal during homeostasis, the benefits of targeting Wnt/β-catenin signaling have been controversial (Kahn 2014). Nonetheless, Wnt/β-catenin signaling has been increasingly recognized as an attractive target for HNSCC therapy (Aminuddin and Ng 2016). To date, a number of Wnt inhibitors have been developed and tested for antitumor properties in preclinical models, with some moving on to clinical trials (Table). A key member of the Wnt/β-catenin cascade is the acyl-transferase Porcupine, for which inhibitors have been developed, including IWP (Dodge et al. 2012), LGK974 (Kulak et al. 2015), ETC-159 (Madan et al. 2016), and C59 (Proffitt et al. 2013). LGK974 has been shown to inhibit HNSCC growth and metastasis using the chick chorioallantoic membrane (CAM) assay (Rudy et al. 2016). In addition, compounds targeting extracellular Wnt ligands and their receptors are undergoing evaluation in clinical trials (reviewed in Zhan et al. 2017). They include OMP-54F28, a fusion Fzd8-Fc decoy receptor, and antibodies against Fzd-7 (OMP18R5) and R-spondin3 (OMP131R10), as well as Wnt5a mimetic (Foxy-5), among others (Table). Also, inhibitors of tankyrase-mediated adenosine diphosphate (ADP)–ribosylation that increase Axin levels are in development, including XAV939 (Huang et al. 2009) and IWR (Kulak et al. 2015), although they have not moved on to clinical trials.
Table.
Compound | Target | Reference | Trial Phase; Identifier | Tumor Sites |
---|---|---|---|---|
IWP | Porcupine | Dodge et al. 2012 | None | NA |
LGK974 | Porcupine | Kulak et al. 2015 | 1/2; NCT02649530 | Metastatic colorectal cancer with Wnt pathway mutations; HNSCC with Notch receptor mutations |
ETC-159 | Porcupine | Madan et al. 2016 | 1; NCT02521844 | R-spondin fusion-positive colorectal carcinoma |
C59 | Porcupine | Proffitt et al. 2013 | None | NA |
OMP-54F28 (Ipafricept) |
FZD8-Fc Decoy receptor Targets CSCs |
OncoMed 2014 (Clinicaltrials.gov) | 1; NCT02092363 NCT02050178 |
Hepatocellular and pancreatic ductal carcinoma, ovarian cancer |
OMP18R5 | Anti-FZD7 antibody | Gurney et al. 2012 | 1; NCT01957007 NCT02005315 NCT01973309 |
Non–small lung cancer, pancreatic ductal adenocarcinoma, breast cancer |
OMP131R10 | Anti–R-spondin 3 antibody | OncoMed 2015 (Clinicaltrials.gov) | 1; NCT02482441 | Metastatic colorectal cancer (RSPO3 positive) |
Foxy-5 | Wnt5a mimetic | WntResearch 2016 (Clinicaltrials.gov) | 1; NCT02655952 | Breast, colorectal, and prostate cancer |
XAV939 | Tankyrase | Huang et al. 2009 | None | NA |
IWR | Tankyrase | Kulak et al. 2015 | None | NA |
ICG-001 | β-Catenin–CBP interaction |
Emami et al. 2004
Wend et al. 2013 |
None | NA |
PRI-724, second-generation ICG-001 derivative | β-catenin–CBP interaction | Prism Pharma 2012 (Clinicaltrials.gov) | 1/2; NCT01606579 NCT02413853 NCT01764477 |
Myelogenous leukemia; colorectal and pancreatic adenocarcinoma |
E7386, third-generation ICG-001 derivative | β-Catenin–CBP interaction | Eisai, Inc. 2017 (Clinicaltrials.gov) | 1; NCT032664 | Advanced neoplasms; planned expansion to HNSCC |
CBP, cAMP-responsive element binding protein; CSC, cancer stem cell; HNSCC, head and neck squamous cell carcinoma; NA, not applicable.
Increased insights into the mechanisms of β-catenin function in the nucleus have provided support for its potential to serve as a druggable target (Aminuddin and Ng 2016). Recently, a small-molecule inhibitor of nuclear β-catenin activity, axitinib, has been shown to downregulate nuclear β-catenin abundance through stabilization of ubiquitin ligase SHPRH (SNF2, histone linker, PHD, and RNIG finger domain-containing helicase) (Qu et al. 2016). Our own ongoing investigations have shown that a small-molecule inhibitor of the interaction between β-catenin and CBP in the nucleus, ICG-001, suppresses HNSCC CSCs and tumor growth in preclinical models (Kartha et al., unpublished data, 2018). Furthermore, an active metabolite of a closely related compound to ICG-001, PRI-724, is undergoing clinical trials in patients with leukemia and colorectal and pancreatic cancer. A third-generation derivative of ICG-001, E7386, is currently in phase I clinical trials in patients with HNSCC (Table). Importantly, inhibitors of the β-catenin/CBP axis are likely not only to target CSCs but also to intercept oncogenic activities in the tumor microenvironment, as β-catenin activity has been associated with the fibrotic process and immune exhaustion. Thus, it is likely that targeting the β-catenin–CBP interaction will have intratumoral inhibitory effects, as proposed in Figure 3. Last, inhibiting oncogenic pathways in combination with Wnt/β-catenin signaling offers improvement over current monotherapies and standard radiation therapy/chemotherapy for the treatment of HNSCC. In particular, combination therapies targeting the β-catenin/CBP axis and an immune checkpoint blockade may enhance the deletion of aggressive CSCs while increasing anticancer immune cell function (Fig. 3).
Future Directions
Mounting evidence indicates that Wnt/β-catenin signaling is at the nexus of both tumor epithelial and stromal deregulation, including the ECM and immune dysfunction that collectively contribute to this malignancy. The complexity and dynamic nature of Wnt/β-catenin signaling represent a challenge to the design of therapeutic strategies that target the oncogenic functions of β-catenin while not affecting its structural contributions. Future studies focused on decoding the biology of stem cells and their lineages and evolving niches, including control of pluripotency and epigenetic regulation, as well as understanding how cellular and metabolic plasticity dictate interpretation of signals in the context of Wnt/β-catenin activities, will improve our understanding of this pathway in HNSCC. Indeed, recent single-cell sequencing of HNSCC has revealed a complex ecosystem with diverse cell populations exhibiting distinct gene signatures with different tumor-promoting potential (Puram et al. 2017). Future studies will continue to leverage computational and system biology methodologies to map signaling networks and cellular metabolism in different tumor cell populations and apply emerging technologies in genome and epigenome editing to elucidate causal relationships between chromatin organization and regulatory processes across the genome that are deregulated by β-catenin and its colluding partners in HNSCC. Also, comprehensive characterization of gene expression changes at the protein level, including alterations in posttranslational modifications (i.e., lipidation, glycosylation) and protein-protein interactions, will contribute new insights into how to interfere with unchecked Wnt/β-catenin transcriptional signals to improve HNSCC therapy.
Author Contributions
K.A. Alamoud, contributed to data acquisition and analysis, drafted the manuscript; M.A. Kukuruzinska, contributed to conception and design, critically revised the manuscript. Both authors gave final approval and agree to be accountable for all aspects of the work.
Footnotes
This work was supported by the Boston University Evans Center for Interdisciplinary Biomedical Research Affinity Research Collaborative on Etiology and Pathogenesis in Oral Cancer (9950000118) to M.A.K.
The authors declare no potential conflicts of interest with respect to the authorship and/or publication of this article.
References
- Aminuddin A, Ng PY. 2016. Promising druggable target in head and neck squamous cell carcinoma: Wnt signaling. Front Pharmacol. 7:244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Azzolin L, Panciera T, Soligo S, Enzo E, Bicciato S, Dupont S, Bresolin S, Frasson C, Basso G, Guzzardo V, et al. 2014. YAP/TAZ incorporation in the beta-catenin destruction complex orchestrates the Wnt response. Cell. 158(1):157–170. [DOI] [PubMed] [Google Scholar]
- Baum B, Settleman J, Quinlan MP. 2008. Transitions between epithelial and mesenchymal states in development and disease. Semin Cell Dev Biol. 19(3):294–308. [DOI] [PubMed] [Google Scholar]
- Beck TN, Golemis EA. 2016. Genomic insights into head and neck cancer. Cancers Head Neck. 1(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benjamin JM, Nelson WJ. 2008. Bench to bedside and back again: molecular mechanisms of alpha-catenin function and roles in tumorigenesis. Semin Cancer Biol. 18(1):53–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birchmeier W, Behrens J. 1994. Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta. 1198(1):11–26. [DOI] [PubMed] [Google Scholar]
- Bonnans C, Chou J, Werb Z. 2014. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 15(12):786–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brembeck FH, Wiese M, Zatula N, Grigoryan T, Dai Y, Fritzmann J, Birchmeier W. 2011. BCL9-2 promotes early stages of intestinal tumor progression. Gastroenterology. 141(4):1359–1370. [DOI] [PubMed] [Google Scholar]
- Cairns RA, Harris IS, Mak TW. 2011. Regulation of cancer cell metabolism. Nat Rev Cancer. 11(2):85–95. [DOI] [PubMed] [Google Scholar]
- Cancer Genome Atlas Network. 2015. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517(7536):576–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carmon KS, Gong X, Lin Q, Thomas A, Liu Q. 2011. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc Natl Acad Sci USA. 108(28):11452–11457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castilho RM, Gutkind JS. 2014. The Wnt/beta-catenin signaling circuitry in head and neck cancer. In: Burtness B, Golemis EA. editors. Molecular determinants of head and neck cancer. New York: Springer. p. 199–214. [Google Scholar]
- Chan KC, Chan LS, Ip JC, Lo C, Yip TT, Ngan RK, Wong RN, Lo KW, Ng WT, Lee AW, et al. 2015. Therapeutic targeting of CBP/β-catenin signaling reduces cancer stem-like population and synergistically suppresses growth of EBV-positive nasopharyngeal carcinoma cells with cisplatin. Sci Rep. 5:9979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang HW, Lee YS, Nam HY, Han MW, Kim HJ, Moon SY, Jeon H, Park JJ, Carey TE, Chang SE, et al. 2013. Knockdown of β-catenin controls both apoptotic and autophagic cell death through LKB1/AMPK signaling in head and neck squamous cell carcinoma cell lines. Cell Signal. 25(4):839–847. [DOI] [PubMed] [Google Scholar]
- Chen D, Wu M, Li Y, Chang I, Yuan Q, Ekimyan-Salvo M, Deng P, Yu B, Yu Y, Dong J, et al. 2017. Targeting BMI1+ cancer stem cells overcomes chemoresistance and inhibits metastases in squamous cell carcinoma. Cell Stem Cell. 20(5):621–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clay MR, Tabor M, Owen JH, Carey TE, Bradford CR, Wolf GT, Wicha MS, Prince ME. 2010. Single-marker identification of head and neck squamous cell carcinoma cancer stem cells with aldehyde dehydrogenase. Head Neck. 32(9):1195–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodge ME, Moon J, Tuladhar R, Lu J, Jacob LS, Zhang LS, Shi H, Wang X, Moro E, Mongera A, et al. 2012. Diverse chemical scaffolds support direct inhibition of the membrane-bound O-acyltransferase porcupine. J Biol Chem. 287(27):23246–23254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan L, Yao J, Wu X, Fan M. 2006. Growth suppression induced by notch1 activation involves Wnt-beta-catenin down-regulation in human tongue carcinoma cells. Biol Cell. 98(8):479–490. [DOI] [PubMed] [Google Scholar]
- Emami KH, Nguyen C, Ma H, Kim DH, Jeong KW, Eguchi M, Moon RT, Teo JL, Kim HY, Moon SH, et al. 2004. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription [corrected]. Proc Natl Acad Sci USA. 101(34):12682–12687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gammon L, Mackenzie IC. 2016. Roles of hypoxia, stem cells and epithelial-mesenchymal transition in the spread and treatment resistance of head and neck cancer. J Oral Pathol Med. 45(2):77–82. [DOI] [PubMed] [Google Scholar]
- Ghahhari NM, Babashah S. 2015. Interplay between microRNAS and WNT/β-catenin signalling pathway regulates epithelial-mesenchymal transition in cancer. Eur J Cancer. 51(12):1638–1649. [DOI] [PubMed] [Google Scholar]
- Gonzalez-Moles MA, Ruiz-Avila I, Gil-Montoya JA, Plaza-Campillo J, Scully C. 2014. Beta-catenin in oral cancer: an update on current knowledge. Oral Oncol. 50(9):818–824. [DOI] [PubMed] [Google Scholar]
- Gonzalez-Sancho JM, Aguilera O, Garcia JM, Pendas-Franco N, Pena C, Cal S, Garcia de, Herreros A, Bonilla F, Munoz A. 2005. The Wnt antagonist DICKKOPF-1 gene is a downstream target of beta-catenin/TCF and is downregulated in human colon cancer. Oncogene. 24(6):1098–1103. [DOI] [PubMed] [Google Scholar]
- Gordon MD, Nusse R. 2006. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 281(32):22429–22433. [DOI] [PubMed] [Google Scholar]
- Gurney A, Axelrod F, Bond CJ, Cain J, Chartier C, Donigan L, Fischer M, Chaudhari A, Ji M, Kapoun AM, et al. 2012. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc Natl Acad Sci USA. 109(29):11717–11722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hao HX, Xie Y, Zhang Y, Charlat O, Oster E, Avello M, Lei H, Mickanin C, Liu D, Ruffner H, et al. 2012. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature. 485(7397):195–200. [DOI] [PubMed] [Google Scholar]
- Hedberg ML, Goh G, Chiosea SI, Bauman JE, Freilino ML, Zeng Y, Wang L, Diergaarde BB, Gooding WE, Lui VW, et al. 2016. Genetic landscape of metastatic and recurrent head and neck squamous cell carcinoma. J Clin Invest. 126(4):1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heuberger J, Birchmeier W. 2010. Interplay of cadherin-mediated cell adhesion and canonical Wnt signaling. Cold Spring Harb Perspect Biol. 2(2):a002915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hiemer SE, Zhang L, Kartha VK, Packer TS, Almershed M, Noonan V, Kukuruzinska M, Bais MV, Monti S, Varelas X. 2015. A YAP/TAZ-regulated molecular signature is associated with oral squamous cell carcinoma. Mol Cancer Res. 13(6):957–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, Hein K, Vogt R, Kemler R. 2012. Wnt/beta-catenin signaling regulates telomerase in stem cells and cancer cells. Science. 336(6088):1549–1554. [DOI] [PubMed] [Google Scholar]
- Holland JD, Klaus A, Garratt AN, Birchmeier W. 2013. Wnt signaling in stem and cancer stem cells. Curr Opin Cell Biol. 25(2):254–264. [DOI] [PubMed] [Google Scholar]
- Hu T, Li C. 2010. Convergence between Wnt-β-catenin and EGFR signaling in cancer. Mol Cancer. 9:236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Z, Muller S, Qian G, Xu J, Kim S, Chen Z, Jiang N, Wang D, Zhang H, Saba NF, et al. 2015. Human papillomavirus 16 oncoprotein regulates the translocation of beta-catenin via the activation of epidermal growth factor receptor. Cancer. 121(2):214–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang SM, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, Charlat O, Wiellette E, Zhang Y, Wiessner S, et al. 2009. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 461(7264):614–620. [DOI] [PubMed] [Google Scholar]
- Kahn M. 2014. Can we safely target the wnt pathway? Nat Rev Drug Discov. 13(7):513–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalluri R, Weinberg RA. 2009. The basics of epithelial-mesenchymal transition. J Clin Invest. 119(6):1420–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katoh M. 2005. Comparative genomics on SFRP2 orthologs. Oncol Rep. 14(3):783–787. [PubMed] [Google Scholar]
- Komekado H, Yamamoto H, Chiba T, Kikuchi A. 2007. Glycosylation and palmitoylation of Wnt-3a are coupled to produce an active form of Wnt-3a. Genes Cells. 12(4):521–534. [DOI] [PubMed] [Google Scholar]
- Koo BK, Spit M, Jordens I, Low TY, Stange DE, van de Wetering M, van Es JH, Mohammed S, Heck AJ, Maurice MM, et al. 2012. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature. 488(7413):665–669. [DOI] [PubMed] [Google Scholar]
- Koontongkaew S. 2013. The tumor microenvironment contribution to development, growth, invasion and metastasis of head and neck squamous cell carcinomas. J Cancer. 4(1):66–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kourtidis A, Ngok SP, Pulimeno P, Feathers RW, Carpio LR, Baker TR, Carr JM, Yan IK, Borges S, Perez EA, et al. 2015. Distinct E-cadherin-based complexes regulate cell behaviour through miRNA processing or Src and p120 catenin activity. Nat Cell Biol. 17(9):1145–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnamurthy S, Dong Z, Vodopyanov D, Imai A, Helman JI, Prince ME, Wicha MS, Nor JE. 2010. Endothelial cell-initiated signaling promotes the survival and self-renewal of cancer stem cells. Cancer Res. 70(23):9969–9978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulak O, Chen H, Holohan B, Wu X, He H, Borek D, Otwinowski Z, Yamaguchi K, Garofalo LA, Ma Z, et al. 2015. Disruption of Wnt/β-catenin signaling and telomeric shortening are inextricable consequences of tankyrase inhibition in human cells. Mol Cell Biol. 35(14):2425–2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon C, Cheng P, King IN, Andersen P, Shenje L, Nigam V, Srivastava D. 2011. Notch post-translationally regulates β-catenin protein in stem and progenitor cells. Nat Cell Biol. 13(10):1244–1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Labbe E, Lock L, Letamendia A, Gorska AE, Gryfe R, Gallinger S, Moses HL, Attisano L. 2007. Transcriptional cooperation between the transforming growth factor-beta and Wnt pathways in mammary and intestinal tumorigenesis. Cancer Res. 67(1):75–84. [DOI] [PubMed] [Google Scholar]
- Lee CH, Hung HW, Hung PH, Shieh YS. 2010. Epidermal growth factor receptor regulates beta-catenin location, stability, and transcriptional activity in oral cancer. Mol Cancer. 9:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SH, Koo BS, Kim JM, Huang S, Rho YS, Bae WJ, Kang HJ, Kim YS, Moon JH, Lim YC. 2014. Wnt/beta-catenin signalling maintains self-renewal and tumourigenicity of head and neck squamous cell carcinoma stem-like cells by activating Oct4. J Pathol. 234(1):99–107. [DOI] [PubMed] [Google Scholar]
- Lenz HJ, Kahn M. 2014. Safely targeting cancer stem cells via selective catenin coactivator antagonism. Cancer Sci. 105(9):1087–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Sutter C, Parker DS, Blauwkamp T, Fang M, Cadigan KM. 2007. CBP/p300 are bimodal regulators of Wnt signaling. EMBO J. 26(9):2284–2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lien WH, Fuchs E. 2014. Wnt some lose some: transcriptional governance of stem cells by Wnt/beta-catenin signaling. Genes Dev. 28(14):1517–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu G, Sengupta PK, Jamal B, Yang HY, Bouchie MP, Lindner V, Varelas X, Kukuruzinska MA. 2013. N-glycosylation induces the CTHRC1 protein and drives oral cancer cell migration. J Biol Chem. 288(28):20217–20227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, et al. 2013. Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov. 3(7):761–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo M, Wicha MS. 2015. Metabolic plasticity of cancer stem cells. Oncotarget. 6(34):35141–35142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma H, Nguyen C, Lee KS, Kahn M. 2005. Differential roles for the coactivators CBP and p300 on TCF/beta-catenin-mediated survivin gene expression. Oncogene. 24(22):3619–3631. [DOI] [PubMed] [Google Scholar]
- Madan B, Ke Z, Harmston N, Ho SY, Frois AO, Alam J, Jeyaraj DA, Pendharkar V, Ghosh K, Virshup IH, et al. 2016. Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene. 35(17):2197–2207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazumdar J, O’Brien WT, Johnson RS, LaManna JC, Chavez JC, Klein PS, Simon MC. 2010. O2 regulates stem cells through Wnt/β-catenin signalling. Nat Cell Biol. 12(10):1007–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGough IJ, Vincent JP. 2016. Exosomes in developmental signalling. Development. 143(14):2482–2493. [DOI] [PubMed] [Google Scholar]
- Mosimann C, Hausmann G, Basler K. 2009. Beta-catenin hits chromatin: regulation of Wnt target gene activation. Nat Rev Mol Cell Biol. 10(4):276–286. [DOI] [PubMed] [Google Scholar]
- Myant KB, Cammareri P, McGhee EJ, Ridgway RA, Huels DJ, Cordero JB, Schwitalla S, Kalna G, Ogg EL, Athineos D, et al. 2013. ROS production and NF-kB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell. 12(6):761–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nör C, Zhang Z, Warner KA, Bernardi L, Visioli F, Helman JI, Roesler R, Nör JE. 2014. Cisplatin induces Bmi-1 and enhances the stem cell fraction in head and neck cancer. Neoplasia. 16(2):137-146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nusse R, Clevers H. 2017. Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169(6):985–999. [DOI] [PubMed] [Google Scholar]
- Padhi S, Saha A, Kar M, Ghosh C, Adhya A, Baisakh M, Mohapatra N, Venkatesan S, Hande MP, Banerjee B. 2015. Clinico-pathological correlation of β-catenin and telomere dysfunction in head and neck squamous cell carcinoma patients. J Cancer. 6(2):192–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, Barroso-Sousa R, Kaplan JB, Chae YK, Giles FJ. 2017. Wnt/beta-catenin pathway: modulating anticancer immune response. J Hematol Oncol. 10(1):101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pannone G, Bufo P, Santoro A, Franco R, Aquino G, Longo F, Botti G, Serpico R, Cafarelli B, Abbruzzese A, et al. 2010. Wnt pathway in oral cancer: epigenetic inactivation of Wnt-inhibitors. Oncol Rep. 24(4):1035–1041. [DOI] [PubMed] [Google Scholar]
- Parker DS, Ni YY, Chang JL, Li J, Cadigan KM. 2008. Wingless signaling induces widespread chromatin remodeling of target loci. Mol Cell Biol. 28(5):1815–1828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pectasides E, Rampias T, Sasaki C, Perisanidis C, Kouloulias V, Burtness B, Zaramboukas T, Rimm D, Fountzilas G, Psyrri A. 2014. Markers of epithelial to mesenchymal transition in association with survival in head and neck squamous cell carcinoma (HNSCC). PLoS One. 9(4):e94273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Porporato PE, Payen VL, Pérez-Escuredo J, De Saedeleer CJ, Danhier P, Copetti T, Dhup S, Tardy M, Vazeille T, Bouzin C, et al. 2014. A mitochondrial switch promotes tumor metastasis. Cell Rep. 8(3):754–766. [DOI] [PubMed] [Google Scholar]
- Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, Weissman IL, Clarke MF, Ailles LE. 2007. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA. 104(3):973–978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Proffitt KD, Madan B, Ke Z, Pendharkar V, Ding L, Lee MA, Hannoush RN, Virshup DM. 2013. Pharmacological inhibition of the Wnt acyltransferase PORCN prevents growth of WNT-driven mammary cancer. Cancer Res. 73(2):502–507. [DOI] [PubMed] [Google Scholar]
- Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, Rodman C, Luo CL, Mroz EA, Emerick KS, et al. 2017. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell. 171(7):1611–1624, e1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qu Y, Gharbi N, Yuan X, Olsen JR, Blicher P, Dalhus B, Brokstad KA, Lin B, Oyan AM, Zhang W, et al. 2016. Axitinib blocks Wnt/β-catenin signaling and directs asymmetric cell division in cancer. Proc Natl Acad Sci USA. 113(33):9339–9344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rampias T, Boutati E, Pectasides E, Sasaki C, Kountourakis P, Weinberger P, Psyrri A. 2010. Activation of Wnt signaling pathway by human papillomavirus E6 and E7 oncogenes in HPV16-positive oropharyngeal squamous carcinoma cells. Mol Cancer Res. 8(3):433–443. [DOI] [PubMed] [Google Scholar]
- Rampias T, Psyrri A. 2014. Human papillomavirus (HPV)–positive head and neck cancer and the wnt signaling pathway. In: Burtness B, Golemis EA. editors. Molecular determinants of head and. neck cancer. New York: Springer. p. 215–225. [Google Scholar]
- Rosenbluh J, Nijhawan D, Cox AG, Li X, Neal JT, Schafer EJ, Zack TI, Wang X, Tsherniak A, Schinzel AC, et al. 2012. Beta-catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell. 151(7):1457–1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rudy SF, Brenner JC, Harris JL, Liu J, Che J, Scott MV, Owen JH, Komarck CM, Graham MP, Bellile EL, et al. 2016. In vivo Wnt pathway inhibition of human squamous cell carcinoma growth and metastasis in the chick chorioallantoic model. J Otolaryngol Head Neck Surg. 45:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schepers AG, Snippert HJ, Stange DE, van den Born M, van Es JH, van de, Wetering M, Clevers H. 2012. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science. 337(6095):730–735. [DOI] [PubMed] [Google Scholar]
- Schroeder JA, Adriance MC, McConnell EJ, Thompson MC, Pockaj B, Gendler SJ. 2002. ErbB-β-catenin complexes are associated with human infiltrating ductal breast and murine mammary tumor virus (MMTV)-Wnt-1 and MMTV-c-Neu transgenic carcinomas. J Biol Chem. 277(25):22692–22698. [DOI] [PubMed] [Google Scholar]
- Sierra J, Yoshida T, Joazeiro CA, Jones KA. 2006. The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev. 20(5):586–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spranger S, Gajewski TF. 2015. A new paradigm for tumor immune escape: β-catenin-driven immune exclusion. J Immunother Cancer. 3:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun L, Yao Y, Liu B, Lin Z, Lin L, Yang M, Zhang W, Chen W, Pan C, Liu Q, et al. 2012. MiR-200b and miR-15b regulate chemotherapy-induced epithelial-mesenchymal transition in human tongue cancer cells by targeting BMI1. Oncogene. 31(4):432–445. [DOI] [PubMed] [Google Scholar]
- Swafford D, Manicassamy S. 2015. Wnt signaling in dendritic cells: its role in regulation of immunity and tolerance. Discov Med. 19(105):303–310. [PMC free article] [PubMed] [Google Scholar]
- Tenbaum SP, Ordóñez-Morán P, Puig I, Chicote I, Arqués O, Landolfi S, Fernández Y, Herance JR, Gispert JD, Mendizabal L, et al. , et al. 2012. β-Catenin confers resistance to PI3K and AKT inhibitors and subverts FOXO3a to promote metastasis in colon cancer. Nat Med. 18(6):892–901. [DOI] [PubMed] [Google Scholar]
- Valenta T, Hausmann G, Basler K. 2012. The many faces and functions of beta-catenin. EMBO J. 31(12):2714–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varelas X, Bouchie MP, Kukuruzinska MA. 2014. Protein N-glycosylation in oral cancer: dysregulated cellular networks among DPAGT1, E-cadherin adhesion and canonical Wnt signaling. Glycobiology. 24(7):579–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wend P, Fang L, Zhu Q, Schipper JH, Loddenkemper C, Kosel F, Brinkmann V, Eckert K, Hindersin S, Holland JD, et al. 2013. Wnt/beta-catenin signalling induces MLL to create epigenetic changes in salivary gland tumours. EMBO J. 32(14):1977–1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang F, Zeng Q, Yu G, Li S, Wang CY. 2006. Wnt/beta-catenin signaling inhibits death receptor-mediated apoptosis and promotes invasive growth of HNSCC. Cell Signal. 18(5):679–687. [DOI] [PubMed] [Google Scholar]
- Yang H, Adam RC, Ge Y, Hua ZL, Fuchs E. 2017. Epithelial-mesenchymal micro-niches govern stem cell lineage choices. Cell. 169(3):483–496.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu Y, Wu J, Wang Y, Zhao T, Ma B, Liu Y, Fang W, Zhu WG, Zhang H. 2012. Kindlin 2 forms a transcriptional complex with β–catenin and TCF4 to enhance Wnt signalling. EMBO Rep. 13(8):750–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan T, Rindtorff N, Boutros M. 2017. Wnt signaling in cancer. Oncogene. 36(11):1461–1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z, Dong Z, Lauxen IS, Filho MS, Nör JE. 2014. Endothelial cell–secreted EGF induces epithelial to mesenchymal transition and endows head and neck cancer cells with stem-like phenotype. Cancer Res. 74(10):2869–2881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou B, Liu Y, Kahn M, Ann DK, Han A, Wang H, Nguyen C, Flodby P, Zhong Q, Krishnaveni MS, et al. 2012. Interactions between β-catenin and transforming growth factor–β signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)–binding protein (CBP). J Biol Chem. 287(10):7026–7038. [DOI] [PMC free article] [PubMed] [Google Scholar]