Skip to main content
Molecular Oncology logoLink to Molecular Oncology
. 2017 Jun 19;11(7):878–891. doi: 10.1002/1878-0261.12082

The EMT spectrum and therapeutic opportunities

Dominic C Voon 1,2, Ruby Y Huang 3,4, Rebecca A Jackson 5, Jean P Thiery 5,6,7,
PMCID: PMC5496500  PMID: 28544151

Abstract

Carcinomas are phenotypically arrayed along an epithelial–mesenchymal transition (EMT) spectrum, a developmental program currently exploited to understand the acquisition of drug resistance through a re‐routing of growth factor signaling. This review collates the current approaches employed in developing therapeutics against cancer‐associated EMT, and provides an assessment of their respective strengths and drawbacks. We reflect on the close relationship between EMT and chemoresistance against current targeted therapeutics, with a special focus on the epigenetic mechanisms that link these processes. This prompts the hypothesis that carcinoma‐associated EMT shares a common epigenetic pathway to cellular plasticity as somatic cell reprogramming during tissue repair and regeneration. Indeed, their striking resemblance suggests that EMT in carcinoma is a pathological adaptation of an intrinsic program of cellular plasticity that is crucial to tissue homeostasis. We thus propose a revised approach that targets the epigenetic mechanisms underlying pathogenic EMT to arrest cellular plasticity regardless of upstream cancer‐driving mutations.

Keywords: cancer therapeutics, cellular plasticity, drug discovery, drug resistance, EMT spectrum, epithelial‐mesenchymal transition


Abbreviations

CSCs

cancer stem cells

CTCs

circulating tumor cells

EMP

epithelial–mesenchymal plasticity

EMT

epithelial–mesenchymal transition

HCC

hepatocellular carcinoma

HDAC

histone deacetylases

HDACi

histone deacetylase inhibitor

MET

mesenchymal‐epithelial transition

NSCLC

non‐small cell lung cancer

PHF2

PHD finger protein 2

PKA

protein kinase A

TET1

ten‐eleven translocation 1

1. The EMT spectrum

Recent evidence has advanced and broadened the definition of epithelial–mesenchymal transition (EMT) in human pathologies. While earlier studies relied on the use of key epithelial and mesenchymal markers to detect its aberrant activation during pathogenesis, it now becomes clear that this is a not a simple binary decision to acquire either an epithelial or a mesenchymal state. Rather, pathological EMT manifests dynamic transitional states punctuated by metastable intermediates (Nieto et al., 2016). This review collates the current knowledge of the molecular mechanisms underlying this phenomenon, and discusses current efforts in the deployment and development of therapeutic interventions.

EMT is orchestrated by a core set of transcription factors (EMT‐TFs), each having the ability to drive EMT via largely analogous genetic programs. These include SNAI1/2, TWIST, and ZEB, among others. As reviewed elsewhere, a myriad of growth factor and developmental signals activate these EMT‐TFs (Thiery et al., 2009). However, the precise reasons for why this highly controlled program is aberrantly triggered at times are varied and often obscured. This is compounded by the inherent difficulty in quantifying the extent of the so‐called partial EMT in each disease state – just exactly how stable is metastable? Such complexities present a formidable challenge in rational drug design. Indeed, with such variations, what works in one context or in a particular patient could be futile or harmful in another. Nevertheless, with fresh knowledge and the benefit of hindsight, certain principles have emerged.

Like with other examples of heterogeneity encountered in biology, there is also heterogeneity following the execution of the EMT program. One explanation is that EMT heterogeneity results from a diverse mix of populations undergoing EMT at different rates and downstream to various cues. For example, circulating tumor cells (CTCs) isolated from patients with breast cancer display a spectrum of epithelial–mesenchymal hybrid features (Khoo et al., 2015; Yadavalli et al., 2017; Yu et al., 2013a), the composition of which varies significantly among patients and is greatly dominated by the underlying biology of the primary tumor. Along the clinical course, the epithelial–mesenchymal hybrid features of CTCs continue to evolve, further illustrating that the metastable state itself exists as a dynamic range of equilibrium. With this appreciation of EMT as a spectrum of different states, broader perspectives of how to manipulate the metastable state within each context can thus be provided.

2. EMT drug discovery platforms

At the heart of each drug discovery platform is a cohesive concept. In the development of EMT‐targeting therapeutics, the following approaches have been adopted: (a) killing cells that have undergone EMT and (b) reversing EMT in metastable cells. It is worth noting here that while these approaches share a common purpose, the rationale for each is distinct.

2.1. Targeting EMT‐induced cancer stem cells

In addition to greater chemoresistance, cells that have undergone EMT bear increased stem‐like traits in vitro (Mani et al., 2008; Morel et al., 2008) and in vivo (Guo et al., 2012); this observation raised the hope that targeting EMT could eradicate the rare self‐renewing and multipotent ‘cancer stem cells’ (CSCs) that persist following conventional chemotherapy. EMT is also associated with increased cell migration and resistance to anoikis, properties that are associated with tumor invasion and metastasis. Thus, the specific killing of cells that have undergone EMT is an attractive therapeutic strategy against CSCs.

To date, the most extensive and prominent EMT‐targeting screen was performed on the HMLE series of immortalized human mammary epithelial lines. These lines have been well characterized in studies of cellular transformation (Elenbaas et al., 2001). This model system led to the discovery of the EMT‐induced, tumor‐initiating CSC, typified by their CD44high/CD24low phenotype (Mani et al., 2008; Morel et al., 2008). The production of these cells was shown to be achieved either through the forced expression of EMT‐TFs (SNAI1, TWIST1, and ZEB1) or through a combination of growth factors and RNAi (shEcad) (Mani et al., 2008; Scheel et al., 2011).

A high‐throughput screen in a 384‐well format was conducted using an HMLE derivative line that was induced to undergo EMT by expressing shEcad. This screen identified the selective cytotoxic effects of salinomycin, a potassium ionophore hitherto known as an antibiotic, on the CSC subpopulation >100‐fold relative to paclitaxel (Gupta et al., 2009). Subsequent studies revealed that salinomycin promotes the degradation of the Wnt coreceptor LRP6 (lipoprotein receptor‐related protein 6) by inhibiting its phosphorylation, thereby attenuating Wnt signaling (Lu et al., 2011). The HMLE platform was further deployed in expanded screens identifying other candidate compounds, most notably ML239, which appears to target NF‐κB signaling (Carmody et al., 2012). More recently, a synthetic derivative of salinomycin was shown to kill breast CSCs by sequestering iron in the lysosome, thereby triggering ferroptosis (Mai et al., 2017).

However, despite these advances, there are potential drawbacks to the cytotoxic killing of carcinoma cells undergoing an EMT. First, the endpoint of their transition is often not a permanent mesenchymal state but rather a metastable intermediate state, thus rendering them difficult to target. Indeed, the spectrum of intermediate states exhibited by CTCs (Khoo et al., 2015; Yadavalli et al., 2017; Yu et al., 2013a) likely means that they are not an effective target. Second, cytotoxicity exerts a selective pressure that may hasten the evolution of CSCs into alternative metastable states not sensitive to the drug.

2.2. Reversing EMT in metastable cancer cells

In using an EMT reversal approach, mesenchymal‐like carcinoma cells are reverted to their epithelial‐like (original) phenotype, thereby restricting the (acquired) self‐renewal and invasive properties of these cancer cells. However, few suitable models exist for testing noncytotoxic, EMT‐reversing agents. One platform used the NBT‐II rat bladder carcinoma line to screen for compounds that could reverse growth factor‐induced cell scattering (Chua et al., 2012). Although modest in scale, this screen identified noncytotoxic compounds that target ALK5/TGFβR1, MAPK, Src, and PI3K to reverse the scattering phenotype without impacting cellular proliferation. Two of these compounds, PD0325901 and saracatinib, enhanced mesenchymal‐epithelial transition (MET) when used in combination in non‐small cell lung cancer (NSCLC) lines (Chua et al., 2015). Two other preclinical studies have reported the anti‐EMT activity of Src kinase inhibitors in ovarian and breast carcinoma cell lines (Huang et al., 2013; Vultur et al., 2008).

A mesenchymal derivative of the HMLE cell model has also been used to identify compounds that promote MET (Pattabiraman et al., 2016; Tam et al., 2013). In a high‐throughput screen with a firefly reporter linked to the Cdh1/E‐cadherin, the authors found that forskolin and cholera toxin effectively induced MET by activating protein kinase A (PKA) through elevating intracellular cyclic AMP. This, in turn, activates PHD finger protein 2 (PHF2), which demethylates histone H3K9me2 and H3K9me3 to derepress epithelial markers and permanently reverse EMT driven by epigenetic mechanisms. Importantly, the resultant MET strongly suppresses the tumor‐initiating capacity and increases the drug sensitivity of EMT‐prone carcinoma lines of various tissue origins. A similar platform also utilized an epithelial marker promoter induction (EpI) screen to identify histone deacetylase inhibitors (HDACi) as a potent class of EMT‐reversing agents (Tang et al., 2016; Yun‐Ju Huang and Yo‐Yan Huang, 2016).

An inherent shortcoming of the conventional cell‐based platforms is their inadequacy to model the complex tissue microenvironment in which EMT occurs in vivo. To mimic this, a coculturing system employing modern microfluidics has been developed incorporating tumor spheroids in a three‐dimensional hydrogel scaffold (Aref et al., 2013). This model also allows for assessing the contribution of endothelial cells in the system. One could expect that, with continual advances in methodology, new facets of the EMT process and, therefore, new strategies of intervention will be uncovered.

Several candidate EMT‐reversing agents are already available clinically, such as saracatinib. Initially developed for the treatment of cancer, saracatinib is a dual‐kinase inhibitor, targeting Src and Bcr‐Abl tyrosine kinases. Although saracatinib is well tolerated in humans and showed promising results in animal studies, its efficacy in clinical trials has been disappointing either alone or in combinatorial treatments (Kim et al., 2009; Puls et al., 2011). In view of this, the functionally related focal adhesion kinase (FAK) could be tested for EMT reversal properties, as an inhibitor PF‐00562271 has shown encouraging signs in early clinical trials (Infante et al., 2012).

A further application of these EMT‐reversing inhibitors would be in combination with other drugs to generate synthetic lethality. Along these lines, small chemical inhibitors of various signaling pathways are currently being used in clinical trials for their anti‐EMT activities. Among these, inhibitors targeting the TGF‐β pathway – a classical activator of EMT – have shown the most promise. Of note, the TGF‐β inhibitor, LY2157299 (galunisertib), is in phase II studies against glioblastoma and hepatocellular carcinoma (Brandes et al., 2016; Giannelli et al., 2016; Rodon et al., 2015). Activation of the AXL receptor is reported to aberrantly phosphorylate SMAD3 to induce EMT in hepatocellular carcinoma (HCC) progression in collaboration with TGF‐β (Reichl et al., 2015). As such, the concurrent targeting of AXL and TGF‐β may prove superior to monotherapy aimed at interfering with TGF‐β signaling, and this warrants further investigation, especially given the current availability of AXL inhibitors in the clinic (Antony et al., 2016; Byers et al., 2013; Feneyrolles et al., 2014; Giannelli et al., 2016; Nieto, 2013).

Broadly speaking, inhibitors targeting the major cellular signaling pathways often have an impact on the EMT status of the carcinomas, as these pathways are intimately linked with EMT during development (Thiery et al., 2009; Voon and Thiery, 2017). It is worth noting, too, the potential hazards of reversing EMT in disseminated tumor cells, as MET is already employed by these metastasized cells as a strategy to promote colonization at distal sites (Beerling et al., 2016; Nieto, 2013; Ocana et al., 2012; Tsai et al., 2012). Therefore, precautions should be observed in the use of EMT‐reversing agents in the clinic and only within a clear therapeutic window.

While these drugs may have anti‐EMT activities, they were developed to target cancer‐driving mutations within these pathways (Table 1). In other words, their clinical benefits are seldom benchmarked against their overall contribution to EMT‐associated tumorigenicity and plasticity. Ironically, their inability to completely inhibit EMT may eventually become a driving force behind chemoresistance against these drugs.

Table 1.

A list of clinical trials and drug discovery experiments targeting EMT regulatory components

Disease Tissues Inhibitors Targets Pathway targeted/mechanism Study type References
Fibrosis Kidney Cyclosporin Calcineurin Association of EMT and kidney graft interstitial fibrogenesis Retrospective Hazzan et al. (2011), Hertig et al. (2008)
Cyclosporin Calcineurin Early withdrawal of immunosuppressant did not reduce fibrosis risk in transplant kidneys with EMT features CERTITEM Rostaing et al. (2015)
Cance Bladder Saracatinib c‐Src Attenuated growth and metastasis of transplanted tumors Preclinical Green et al. (2009)
Breast SM16 ALK5/TGFβR1 Reducing spontaneous metastases of established allograft tumors Preclinical Rausch et al. (2009)
Ki26896 ALK5/TGFβR1 Reduced bone metastasis of breast cancer cell line Preclinical Ehata et al. (2007)
1400W, L‐NAME, L‐NMMA iNOS Impairment of HIF‐1α and ER stress/TGF‐β/ATF3,4 crosstalk Preclinical Granados‐Principal et al. (2015)
EW‐7195/7197/7203, IN‐1130 ALK5/TGFβR1 Inhibition of TGF‐β1‐mediated EMT and metastasis of breast cancer Preclinical Park et al. (2011a,b), Son et al. (2014)
Salinomycin LRP6 Identified in high‐throughput screen to show selectivity against CD44high/CD24low mammary cancer stem cells HTS Gupta et al. (2009), Lu et al. (2011)
ML239 NF‐κB pathway Identified in an expanded screen using the same platform as Gupta et al. HTS Carmody et al. (2012)
Colon LY2109761 TGFβRI/II Reduced liver metastases in a metastatic colorectal xenograft model Preclinical Li et al. (2010b,b), Zhang et al. (2009)
Sorafenib/regorafenib SHP1 Activate SHP1 to block TGF‐β‐induced EMT and STAT3 phosphorylation Preclinical Fan et al. (2015, 2016)
Emodin CK2alpha Inhibition of CK2alpha suppressed tumorigenicity and EMT of CRC cells Preclinical Zou et al. (2011)
HNSCC Gefitinib EGFR Gefitinib sensitivity in HNSCC lines is associated with EMT markers Preclinical Frederick et al. (2007)
Gefitinib/saracatinib EGFR/c‐Src Combined targeting of EGFR and c‐Src effectively inhibited HNSCC growth and invasion Preclinical Koppikar et al. (2008)
Cisplatin, cetuximab, and valproic acid HDAC/EGFR HDAC inhibitory activity of valproic acid may offer same benefits as vorinostat in suppressing EGFR expression and reversing EMT Phase II Bruzzese et al. (2011), Caponigro et al. (2016)
HCC Galunisertib TGFβRI Inhibiting TGF‐β signaling restores E‐cadherin expression and diminishes the migratory capacity of HCC cells Phase II Giannelli et al. (2016, 2014)
miR‐216a inhibitor PTEN, SMAD7 miR‐216a/217 targets PTEN and SMAD7 to confer sorafenib resistance Preclinical Xia et al. (2013)
miR‐125 SMAD2/4 Interference of SMAD2/4 to attenuate TGF‐β‐mediated chemoresistance Preclinical Zhou et al. (2015)
Lung Erlotinib EGFR Erlotinib sensitivity in NSCLC lines and xenografts is determined by EMT status Preclinical Thomson et al. (2005)
Erlotinib/PQIP EGFR/IGF‐1R EMT status determines the efficacy of combined blockade of EGFR/IGF‐1R in NSCLC lines and xenografts Preclinical Buck et al. (2008)
Silmitasertib CK2 Inhibition of TGF‐β1 induced EMT in A549 cells Preclinical Kim and Hwan Kim (2013)
Silmitasertib CK2 and FAK–Src–paxillin Blocks micropillar‐induced FAK activation and EMT HTS Kim et al. (2015)
Gefitinib/DN‐30 EGFR/cMET Concurrent suppression of c‐MET significantly increases gefitinib sensitivity in NSCLC cells Preclinical Yano et al. (2008), Zucali et al. (2008)
Gefitinib EGFR Gefitinib sensitivity of NSCLC lines is correlated with the expression of EMT‐associated markers Preclinical Frederick et al. (2007)
Melanoma PLX4032 BRAFV600E Significant regression of metastatic melanoma that carries the V600E BRAF mutation Approved Flaherty et al. (2010)
Ovary ABT‐627 ET‐1/ETAR‐ILK Inhibition of ILK suppressed EMT and tumor growth in a xenograft model Preclinical Rosano et al. (2005)
ZD4054 ETAR/paclitaxel Cotreatment with ZD4054 sensitized ovarian xenograft tumors to paclitaxel Preclinical Rosano et al. (2007)
Saracatinib c‐Src Inhibition of c‐Src restored E‐cadherin expression in ovarian cell lines with intermediate mesenchymal state and attenuated spheroid formation Preclinical Huang et al. (2013)
Pancreas LY2109761 TGFβRI/II Significant reduction in spontaneous abdominal liver metastases in combination with gemcitabine Preclinical Melisi et al. (2008)

2.3. EMT, epigenetics, and chemoresistance

Numerous studies have reported the presence of residual resistant cells following chemotherapy, and these cells have been associated with an EMT phenotype in clinical settings as well as in animal models (Byers et al., 2013; Fischer et al., 2015; Kitai et al., 2016; Manchado et al., 2016; Shao et al., 2014; Zheng et al., 2015). EMT‐associated chemoresistance may also be accompanied with a switch to compensatory pathways, so that carcinoma cells can regain cellular homeostasis (Kitai et al., 2016; Manchado et al., 2016). While the precise basis for the correlation between EMT and cell survival remains obscure, it is likely that intermediate EMT states offer attractive ‘safe havens’ in which cell signaling can be re‐wired to become independent of the targeted pathway. Here, the capacity to shift to an alternate and viable phenotype relies on the cell's EMT‐endowed plasticity, often termed epithelial–mesenchymal plasticity (EMP) (Byers et al., 2013; Nieto, 2013).

It has been proposed that intermediate states represent quasi‐discreet epigenetic states, which are characterized by altered histone modifications on key loci such as E‐cadherin/Cdh1 and miR‐200 (Nieto et al., 2016; Tam and Weinberg, 2013). Accordingly, the same epigenetic machineries that mark these intermediate states are often implicated in the acquisition of chemoresistance. An important class of such histone modifiers are the polycomb group (PcG) repressor complexes, PRC1 and ‐2. During EMT, the PRC2 complex is recruited to the CDH1 promoter by the EMT‐TF SNAI1, whereby it catalyzes the trimethylation of histone H3K27 to repress E‐cadherin expression (Herranz et al., 2008). The same complex is also responsible for the trimethylation and silencing of miR‐200, which gives rise to chemoresistance (Ceppi et al., 2010; Lim et al., 2013; Sato et al., 2017; Tryndyak et al., 2010). PRC1 components, such as BMI1, are considered stem cell factors that support normal stem cells and their transformed counterparts (Park et al., 2004; Valk‐Lingbeek et al., 2004). The upregulation of BMI1 during carcinogenesis was reported to induce EMT and the invasive phenotype, and this was mediated via its cooperative actions with TWIST1 on Cdh1 and INK4A (Song et al., 2009; Yang et al., 2010).

Acetylation is another histone modification associated with EMT and chemoresistance. During cancer metastasis, the histone deacetylases (HDAC) 1 and 2 – as part of the Mi‐2–nucleosome remodeling and deacetylase (NuRD) repressive complex – are recruited by Snail and TWIST to the Cdh1 and Foxa1 promoters, leading to their repression, respectively (von Burstin et al., 2009; Fu et al., 2011; Peinado et al., 2004; Xu et al., 2017). However, various components of the NuRD complex, and specifically the HDACs, will confer drug resistance to cancer cells (Fu et al., 2011; Li et al., 2014; Sakamoto et al., 2016). Consequently, HDAC inhibitors such as vorinostat, mocetinostat, and valproic acid are currently being evaluated as anti‐EMT agents (Bruzzese et al., 2011; Caponigro et al., 2016; Lan et al., 2016; Meidhof et al., 2015; Sakamoto et al., 2016; Schech et al., 2015; Schobert and Biersack, 2017).

A similar correlation between EMT and chemoresistance is also observed for lysine‐specific demethylases, such as LSD1, an emerging class of epigenetic modulators (Bennani‐Baiti, 2012; Lei et al., 2015; Nagasawa et al., 2015). LSD1 modulates gene expression by removing methyl groups on lysine 4 or lysine 9 of histone H3 to repress or activate target promoters, respectively (Shi et al., 2004). In the context of EMT, the induction of EMT in mammary epithelial cells involves the recruitment of LSD1 by SNAI1 to promoters of E‐cadherin, claudin, and cytokeratin family genes, which targets them for repression (Lin et al., 2010a,b). In recent years, the association of LSD1 expression with malignancy, chemoresistance, and poor survival has raised interest into the therapeutic potential of its inhibitors (Lv et al., 2012; Nagasawa et al., 2015; Yu et al., 2013b; Zhao et al., 2012).

In addition to histone modification, DNA methylation patterns are altered during persistent, mutation‐driven EMT during carcinogenesis (McDonald et al., 2011; Tam and Weinberg, 2013). A key mediator of these aberrations appears to be the ten‐eleven translocation 1 (TET1) methylcytosine dioxygenase, which initiates the demethylation of DNA and is associated with tumorigenesis in many cancers (Fu et al., 2014; Song et al., 2013; Sun et al., 2013; Tsai et al., 2014). However, there is opposing evidence as to the role of TET1 in EMT‐induced chemoresistance: TET1 has been reported to promote cisplatin resistance through its induction of EMT in ovarian cancer (Han et al., 2017), but act as a barrier against EMT in mammary epithelial cells by derepressing the miR‐200 promoter (Song et al., 2013).

Finally, it warrants highlighting that the epigenetic states of the EMT intermediates are cooperatively maintained at multiple levels of epigenetic regulation, with all the usual regulatory elements and limitations of a complex network. For example, just as miR‐200 is a target of PRC2‐mediated repression, the PRC2 component Suz12 is conversely targeted by miR‐200 (Iliopoulos et al., 2010; Lim et al., 2013). Moreover, a functional crosstalk between TET1 and NuRD during EMT is also likely, given their cooperation in vitamin C‐induced MET during somatic cell reprogramming (Chen et al., 2013).

2.4. A better mousetrap beyond the EMT spectrum?

From a clinical perspective, the resistance of cancer cells by virtue of their EMT state necessitates targeting the compensatory pathways employed by the cells for their eradication. However, it is just as likely that the very same mechanisms will later give rise to resistance to a new drug. Hence, rather than targeting the ever‐shifting compensatory growth factor pathways, it would seem a better idea to shutdown cellular plasticity. A major obstacle in this approach is that we have an incomplete grasp of the molecular underpinnings of this plasticity. Nevertheless, some cues can be drawn from the field of tissue stem cells, where recent data reveal a genetic program in differentiated cells that promotes cellular plasticity.

Modern lineage tracing studies have demonstrated that some differentiated epithelial cells possess an innate ability to dedifferentiate in vivo, and gain multipotency under specific circumstances (van de Moosdijk et al., 2017; Rios et al., 2016). This phenomenon is most clearly seen during injury and tissue regeneration, but also during inflammation and at certain stages during postnatal development, such as in the mammary gland during pregnancy. Indeed, in specific instances, the induction of stemness is reliant on the coactivation of the EMT program (Guo et al., 2012; Ye et al., 2015). And, although the precise reason for this association is not known, it is clear that the capacity for somatic cell reprogramming – which was dramatically demonstrated in the generation of induced pluripotent stem cells (iPSc) from terminally differentiated fibroblasts – is integral to tissue homeostasis (van Es et al., 2012; Gregorieff et al., 2015; Smith et al., 2016; Takahashi and Yamanaka, 2006; Tetteh et al., 2016). In this light, it is possible that our current investigation of EMT‐associated plasticity and induction would converge on common molecular mechanisms. That is, disease‐associated EMT may be a pathological manifestation of aberrantly activated normal somatic reprogramming of differentiated cells into functional stem cells (Ye et al., 2015).

Such a model of common epigenetic pathways governing EMP and induced pluripotency (iP) indeed has the capacity to accommodate common observations between the two phenomena. A prime example of this would be the role of p53 as a barrier, whereby the loss of its function lowers the threshold for entrance into EMP just as it would enhance the iP efficiency (Ansieau et al., 2008; Austin et al., 2013; Hong et al., 2009; Kawamura et al., 2009; Marion et al., 2009; Mu et al., 2017). A significant part of this is mediated through the p53‐miR‐200 regulatory network, which features prominently in the regulation of EMP and iP (Chang et al., 2011; Hu et al., 2014; Kim et al., 2011; Song et al., 2013). A further common feature is the repressive effects exerted by lineage‐determining transcription factors, such as BRIGHT/ARID3A, RUNX3, GRHL2, and PAX5 (Chung et al., 2016; Hanna et al., 2008; Hikichi et al., 2013; Popowski et al., 2014; Voon et al., 2012). Of relevance, both processes are governed by cell extrinsic factors, such as growth factors (van Es et al., 2012; Lluis et al., 2008; Thiery et al., 2009; Vidal et al., 2014), and intrinsic epigenetics elements, such as the TET/miR‐200 axis (Hu et al., 2014; Song et al., 2013) and the NuRD repressor complex (Chen et al., 2013; Ebrahimi, 2015; Fu et al., 2011; dos Santos et al., 2014).

Despite these parallels, there are obvious differences between the induction of EMP in carcinoma and somatic reprogramming, specifically during the generation of iPSc from fibroblasts. Most notably, the induction of pluripotency in the case of the latter is preceded by MET. It reverts fibroblasts into an epithelial phenotype similar to that of embryonic stem cells (Li et al., 2010b). Consistent with this, pro‐EMT signals like TGF‐β (Ichida et al., 2009; Qin et al., 2014; Vidal et al., 2014), Wnt/β‐catenin (Ho et al., 2013; Lluis et al., 2008), and Hippo (Qin et al., 2012) pathways act as barriers against iP in a context‐specific manner. At the same time, inhibitors of these pathways, such as the aforementioned anti‐EMT TGF‐β inhibitors, strongly enhance the efficiency of somatic reprogramming (Ichida et al., 2009; Maherali and Hochedlinger, 2009). Overall, it seems EMP and iP each require a phenotypic shift along the EMT spectrum (albeit, in opposite directions) toward an intermediate metastable state en route to dedifferentiation and reprogramming. If so, then it is imperative that the innate molecular barriers – such as oxidative and methylation states of the chromatin and their regulators, which safeguard against phenotypic slippage – are thoroughly elucidated. Ultimately, the promise of a plasticity‐centric paradigm is its amenability to the precise targeting of EMT‐associated plasticity in carcinomas irrespective of the upstream driver mutations, and invulnerable to the re‐routing of the signaling circuit observed in current strategies. Accordingly, the development of these next‐generation therapeutics will require discovery platforms that assay the functional output of the involved epigenetic machineries rather than, for example, the activation of a particular marker gene.

3. Concluding remarks

EMT has emerged in recent years to be a major driver of chemoresistance to anticancer therapies in the clinic. This is closely linked to phenotypic plasticity in the form of metastable intermediates over the EMT spectrum. The biological reason for this phenomenon is currently unclear, but it is possible that aberrant EMT in carcinoma cells unlocks an innate dedifferentiation program integral to tissue repair, development, and homeostasis. Importantly, such an engine of plasticity would also fuel tumor heterogeneity, progression, and immune escape. Despite the clear need, targeting EMT in cancer therapy has proven challenging due to conceptual difficulties in the design of viable screens. Conventional screening approaches that focus on interfering with specific molecular interactions are unsuitable or have yielded inconsistent results. In this review, we surveyed the current efforts to develop and deploy anti‐EMT therapeutics and discussed their relative effectiveness. By way of this evaluation, a novel concept is put forth to selectively inhibit low‐order epigenetic mechanisms that promote plasticity. In doing so, the phenotypic flexibility that enables cancer cells to be ‘moving targets’ will be greatly restricted, thereby enhancing the efficacies of current therapeutics.

References

  1. Ansieau S, Bastid J, Doreau A, Morel AP, Bouchet BP, Thomas C, Fauvet F, Puisieux I, Doglioni C, Piccinin S et al (2008) Induction of EMT by twist proteins as a collateral effect of tumor‐promoting inactivation of premature senescence. Cancer Cell 14, 79–89. [DOI] [PubMed] [Google Scholar]
  2. Antony J, Tan TZ, Kelly Z, Low J, Choolani M, Recchi C, Gabra H, Thiery JP, Huang RY (2016) The GAS6‐AXL signaling network is a mesenchymal (Mes) molecular subtype‐specific therapeutic target for ovarian cancer. Sci Signal 9, ra97. [DOI] [PubMed] [Google Scholar]
  3. Aref AR, Huang RY, Yu W, Chua KN, Sun W, Tu TY, Bai J, Sim WJ, Zervantonakis IK, Thiery JP et al (2013) Screening therapeutic EMT blocking agents in a three‐dimensional microenvironment. Integr Biol (Camb) 5, 381–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Austin P, Freeman SA, Gray CA, Gold MR, Vogl AW, Andersen RJ, Roberge M and Roskelley CD (2013) The invasion inhibitor sarasinoside A1 reverses mesenchymal tumor transformation in an E‐cadherin‐independent manner. Mol Cancer Res 11, 530–540. [DOI] [PubMed] [Google Scholar]
  5. Beerling E, Seinstra D, de Wit E, Kester L, van der Velden D, Maynard C, Schafer R, van Diest P, Voest E, van Oudenaarden A et al (2016) Plasticity between epithelial and mesenchymal states unlinks EMT from metastasis‐enhancing stem cell capacity. Cell Rep 14, 2281–2288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bennani‐Baiti IM (2012) Integration of ERalpha‐PELP1‐HER2 signaling by LSD1 (KDM1A/AOF2) offers combinatorial therapeutic opportunities to circumventing hormone resistance in breast cancer. Breast Cancer Res 14, 112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Brandes AA, Carpentier AF, Kesari S, Sepulveda‐Sanchez JM, Wheeler HR, Chinot O, Cher L, Steinbach JP, Capper D, Specenier P et al (2016) A Phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol 18, 1146–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bruzzese F, Leone A, Rocco M, Carbone C, Piro G, Caraglia M, Di Gennaro E and Budillon A (2011) HDAC inhibitor vorinostat enhances the antitumor effect of gefitinib in squamous cell carcinoma of head and neck by modulating ErbB receptor expression and reverting EMT. J Cell Physiol 226, 2378–2390. [DOI] [PubMed] [Google Scholar]
  9. Buck E, Eyzaguirre A, Rosenfeld‐Franklin M, Thomson S, Mulvihill M, Barr S, Brown E, O'Connor M, Yao Y, Pachter J et al (2008) Feedback mechanisms promote cooperativity for small molecule inhibitors of epidermal and insulin‐like growth factor receptors. Cancer Res 68, 8322–8332. [DOI] [PubMed] [Google Scholar]
  10. von Burstin J, Eser S, Paul MC, Seidler B, Brandl M, Messer M, von Werder A, Schmidt A, Mages J, Pagel P et al (2009) E‐cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex. Gastroenterology 137, 361–371, 371.e361–365. [DOI] [PubMed] [Google Scholar]
  11. Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, Shen L, Fan Y, Giri U, Tumula PK et al (2013) An epithelial‐mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res 19, 279–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Caponigro F, Di Gennaro E, Ionna F, Longo F, Aversa C, Pavone E, Maglione MG, Di Marzo M, Muto P, Cavalcanti E et al (2016) Phase II clinical study of valproic acid plus cisplatin and cetuximab in recurrent and/or metastatic squamous cell carcinoma of Head and Neck‐V‐CHANCE trial. BMC Cancer 16, 918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Carmody LC, Germain AR, VerPlank L, Nag PP, Munoz B, Perez JR and Palmer MA (2012) Phenotypic high‐throughput screening elucidates target pathway in breast cancer stem cell‐like cells. J Biomol Screen 17, 1204–1210. [DOI] [PubMed] [Google Scholar]
  14. Ceppi P, Mudduluru G, Kumarswamy R, Rapa I, Scagliotti GV, Papotti M and Allgayer H (2010) Loss of miR‐200c expression induces an aggressive, invasive, and chemoresistant phenotype in non‐small cell lung cancer. Mol Cancer Res 8, 1207–1216. [DOI] [PubMed] [Google Scholar]
  15. Chang CJ, Chao CH, Xia W, Yang JY, Xiong Y, Li CW, Yu WH, Rehman SK, Hsu JL, Lee HH et al (2011) p53 regulates epithelial‐mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol 13, 317–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chen J, Guo L, Zhang L, Wu H, Yang J, Liu H, Wang X, Hu X, Gu T, Zhou Z et al (2013) Vitamin C modulates TET1 function during somatic cell reprogramming. Nat Genet 45, 1504–1509. [DOI] [PubMed] [Google Scholar]
  17. Chua KN, Kong LR, Sim WJ, Ng HC, Ong WR, Thiery JP, Huynh H and Goh BC (2015) Combinatorial treatment using targeted MEK and SRC inhibitors synergistically abrogates tumor cell growth and induces mesenchymal‐epithelial transition in non‐small‐cell lung carcinoma. Oncotarget 6, 29991–30005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chua KN, Sim WJ, Racine V, Lee SY, Goh BC and Thiery JP (2012) A cell‐based small molecule screening method for identifying inhibitors of epithelial‐mesenchymal transition in carcinoma. PLoS One 7, e33183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chung VY, Tan TZ, Tan M, Wong MK, Kuay KT, Yang Z, Ye J, Muller J, Koh CM, Guccione E et al (2016) GRHL2‐miR‐200‐ZEB1 maintains the epithelial status of ovarian cancer through transcriptional regulation and histone modification. Sci Rep 6, 19943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Ebrahimi B (2015) Reprogramming barriers and enhancers: strategies to enhance the efficiency and kinetics of induced pluripotency. Cell Regen (Lond) 4, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Ehata S, Hanyu A, Fujime M, Katsuno Y, Fukunaga E, Goto K, Ishikawa Y, Nomura K, Yokoo H, Shimizu T et al (2007) Ki26894, a novel transforming growth factor‐beta type I receptor kinase inhibitor, inhibits in vitro invasion and in vivo bone metastasis of a human breast cancer cell line. Cancer Sci 98, 127–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Elenbaas B, Spirio L, Koerner F, Fleming MD, Zimonjic DB, Donaher JL, Popescu NC, Hahn WC and Weinberg RA (2001) Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev 15, 50–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. van Es JH, Sato T, van de Wetering M, Lyubimova A, Nee AN, Gregorieff A, Sasaki N, Zeinstra L, van den Born M, Korving J et al (2012) Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat Cell Biol 14, 1099–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Fan LC, Shiau CW, Tai WT, Hung MH, Chu PY, Hsieh FS, Lin H, Yu HC and Chen KF (2015) SHP‐1 is a negative regulator of epithelial‐mesenchymal transition in hepatocellular carcinoma. Oncogene 34, 5252–5263. [DOI] [PubMed] [Google Scholar]
  25. Fan LC, Teng HW, Shiau CW, Tai WT, Hung MH, Yang SH, Jiang JK and Chen KF (2016) Regorafenib (Stivarga) pharmacologically targets epithelial‐mesenchymal transition in colorectal cancer. Oncotarget 7, 64136–64147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Feneyrolles C, Spenlinhauer A, Guiet L, Fauvel B, Dayde‐Cazals B, Warnault P, Cheve G and Yasri A (2014) Axl kinase as a key target for oncology: focus on small molecule inhibitors. Mol Cancer Ther 13, 2141–2148. [DOI] [PubMed] [Google Scholar]
  27. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J et al (2015) Epithelial‐to‐mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 527, 472–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K et al (2010) Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 363, 809–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Frederick BA, Helfrich BA, Coldren CD, Zheng D, Chan D, Bunn PA Jr and Raben D (2007) Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non‐small cell lung carcinoma. Mol Cancer Ther 6, 1683–1691. [DOI] [PubMed] [Google Scholar]
  30. Fu HL, Ma Y, Lu LG, Hou P, Li BJ, Jin WL and Cui DX (2014) TET1 exerts its tumor suppressor function by interacting with p53‐EZH2 pathway in gastric cancer. J Biomed Nanotechnol 10, 1217–1230. [DOI] [PubMed] [Google Scholar]
  31. Fu J, Qin L, He T, Qin J, Hong J, Wong J, Liao L and Xu J (2011) The TWIST/Mi2/NuRD protein complex and its essential role in cancer metastasis. Cell Res 21, 275–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Giannelli G, Mikulits W, Dooley S, Fabregat I, Moustakas A, ten Dijke P, Portincasa P, Winter P, Janssen R, Leporatti S et al (2016) The rationale for targeting TGF‐beta in chronic liver diseases. Eur J Clin Invest 46, 349–361. [DOI] [PubMed] [Google Scholar]
  33. Giannelli G, Villa E and Lahn M (2014) Transforming growth factor‐beta as a therapeutic target in hepatocellular carcinoma. Cancer Res 74, 1890–1894. [DOI] [PubMed] [Google Scholar]
  34. Granados‐Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, Patel T, Rodriguez AA, Cusimano J, Weiss HL et al (2015) Inhibition of iNOS as a novel effective targeted therapy against triple‐negative breast cancer. Breast Cancer Res 17, 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Green TP, Fennell M, Whittaker R, Curwen J, Jacobs V, Allen J, Logie A, Hargreaves J, Hickinson DM, Wilkinson RW et al (2009) Preclinical anticancer activity of the potent, oral Src inhibitor AZD0530. Mol Oncol 3, 248–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Gregorieff A, Liu Y, Inanlou MR, Khomchuk Y and Wrana JL (2015) Yap‐dependent reprogramming of Lgr5(+) stem cells drives intestinal regeneration and cancer. Nature 526, 715–718. [DOI] [PubMed] [Google Scholar]
  37. Guo W, Keckesova Z, Donaher JL, Shibue T, Tischler V, Reinhardt F, Itzkovitz S, Noske A, Zurrer‐Hardi U, Bell G et al (2012) Slug and Sox9 cooperatively determine the mammary stem cell state. Cell 148, 1015–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA and Lander ES (2009) Identification of selective inhibitors of cancer stem cells by high‐throughput screening. Cell 138, 645–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Han X, Zhou Y, You Y, Lu J, Wang L, Hou H, Li J, Chen W, Zhao L and Li X (2017) TET1 promotes cisplatin‐resistance via demethylating the vimentin promoter in ovarian cancer. Cell Biol Int 41, 405–414. [DOI] [PubMed] [Google Scholar]
  40. Hanna J, Markoulaki S, Schorderet P, Carey BW, Beard C, Wernig M, Creyghton MP, Steine EJ, Cassady JP, Foreman R et al (2008) Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell 133, 250–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Hazzan M, Hertig A, Buob D, Copin MC, Noel C, Rondeau E and Dubois‐Xu YC (2011) Epithelial‐to‐mesenchymal transition predicts cyclosporine nephrotoxicity in renal transplant recipients. J Am Soc Nephrol 22, 1375–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Herranz N, Pasini D, Diaz VM, Franci C, Gutierrez A, Dave N, Escriva M, Hernandez‐Munoz I, Di Croce L, Helin K et al (2008) Polycomb complex 2 is required for E‐cadherin repression by the Snail1 transcription factor. Mol Cell Biol 28, 4772–4781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Hertig A, Anglicheau D, Verine J, Pallet N, Touzot M, Ancel PY, Mesnard L, Brousse N, Baugey E, Glotz D et al (2008) Early epithelial phenotypic changes predict graft fibrosis. J Am Soc Nephrol 19, 1584–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Hikichi T, Matoba R, Ikeda T, Watanabe A, Yamamoto T, Yoshitake S, Tamura‐Nakano M, Kimura T, Kamon M, Shimura M et al (2013) Transcription factors interfering with dedifferentiation induce cell type‐specific transcriptional profiles. Proc Natl Acad Sci U S A 110, 6412–6417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Ho R, Papp B, Hoffman JA, Merrill BJ and Plath K (2013) Stage‐specific regulation of reprogramming to induced pluripotent stem cells by Wnt signaling and T cell factor proteins. Cell Rep 3, 2113–2126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, Okita K and Yamanaka S (2009) Suppression of induced pluripotent stem cell generation by the p53‐p21 pathway. Nature 460, 1132–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hu X, Zhang L, Mao SQ, Li Z, Chen J, Zhang RR, Wu HP, Gao J, Guo F, Liu W et al (2014) Tet and TDG mediate DNA demethylation essential for mesenchymal‐to‐epithelial transition in somatic cell reprogramming. Cell Stem Cell 14, 512–522. [DOI] [PubMed] [Google Scholar]
  48. Huang RY, Wong MK, Tan TZ, Kuay KT, Ng AH, Chung VY, Chu YS, Matsumura N, Lai HC, Lee YF et al (2013) An EMT spectrum defines an anoikis‐resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e‐cadherin restoration by a src‐kinase inhibitor, saracatinib (AZD0530). Cell Death Dis 4, e915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ichida JK, Blanchard J, Lam K, Son EY, Chung JE, Egli D, Loh KM, Carter AC, Di Giorgio FP, Koszka K et al (2009) A small‐molecule inhibitor of tgf‐Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell 5, 491–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Iliopoulos D, Lindahl‐Allen M, Polytarchou C, Hirsch HA, Tsichlis PN and Struhl K (2010) Loss of miR‐200 inhibition of Suz12 leads to polycomb‐mediated repression required for the formation and maintenance of cancer stem cells. Mol Cell 39, 761–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Infante JR, Camidge DR, Mileshkin LR, Chen EX, Hicks RJ, Rischin D, Fingert H, Pierce KJ, Xu H, Roberts WG et al (2012) Safety, pharmacokinetic, and pharmacodynamic phase I dose‐escalation trial of PF‐00562271, an inhibitor of focal adhesion kinase, in advanced solid tumors. J Clin Oncol 30, 1527–1533. [DOI] [PubMed] [Google Scholar]
  52. Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya A, Wahl GM and Belmonte JC (2009) Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Khoo BL, Lee SC, Kumar P, Tan TZ, Warkiani ME, Ow SG, Nandi S, Lim CT and Thiery JP (2015) Short‐term expansion of breast circulating cancer cells predicts response to anti‐cancer therapy. Oncotarget 6, 15578–15593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kim J, Choi WJ, Moon SH, Jung J, Park JK, Kim SH and Lee JO (2015) Micropillar arrays as potential drug screens: inhibition of micropillar‐mediated activation of the FAK‐Src‐paxillin signaling pathway by the CK2 inhibitor CX‐4945. Acta Biomater 27, 13–20. [DOI] [PubMed] [Google Scholar]
  55. Kim J and Hwan Kim S (2013) CK2 inhibitor CX‐4945 blocks TGF‐beta1‐induced epithelial‐to‐mesenchymal transition in A549 human lung adenocarcinoma cells. PLoS One 8, e74342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kim LC, Song L and Haura EB (2009) Src kinases as therapeutic targets for cancer. Nat Rev Clin Oncol 6, 587–595. [DOI] [PubMed] [Google Scholar]
  57. Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon YJ, Volinia S, Pineau P, Marchio A, Palatini J, Suh SS et al (2011) p53 regulates epithelial‐mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J Exp Med 208, 875–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kitai H, Ebi H, Tomida S, Floros KV, Kotani H, Adachi Y, Oizumi S, Nishimura M, Faber AC and Yano S (2016) Epithelial‐to‐mesenchymal transition defines feedback activation of receptor tyrosine kinase signaling induced by MEK inhibition in KRAS‐mutant lung cancer. Cancer Discov 6, 754–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Koppikar P, Choi SH, Egloff AM, Cai Q, Suzuki S, Freilino M, Nozawa H, Thomas SM, Gooding WE, Siegfried JM et al (2008) Combined inhibition of c‐Src and epidermal growth factor receptor abrogates growth and invasion of head and neck squamous cell carcinoma. Clin Cancer Res 14, 4284–4291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Lan X, Lu G, Yuan C, Mao S, Jiang W, Chen Y, Jin X and Xia Q (2016) Valproic acid (VPA) inhibits the epithelial‐mesenchymal transition in prostate carcinoma via the dual suppression of SMAD4. J Cancer Res Clin Oncol 142, 177–185. [DOI] [PubMed] [Google Scholar]
  61. Lei ZJ, Wang J, Xiao HL, Guo Y, Wang T, Li Q, Liu L, Luo X, Fan LL, Lin L et al (2015) Lysine‐specific demethylase 1 promotes the stemness and chemoresistance of Lgr5(+) liver cancer initiating cells by suppressing negative regulators of beta‐catenin signaling. Oncogene 34, 3188–3198. [DOI] [PubMed] [Google Scholar]
  62. Li Y, Cao H, Jiao Z, Pakala SB, Sirigiri DN, Li W, Kumar R and Mishra L (2010a) Carcinoembryonic antigen interacts with TGF‐{beta} receptor and inhibits TGF‐{beta} signaling in colorectal cancers. Cancer Res 70, 8159–8168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Li R, Liang J, Ni S, Zhou T, Qing X, Li H, He W, Chen J, Li F, Zhuang Q et al (2010b) A mesenchymal‐to‐epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63. [DOI] [PubMed] [Google Scholar]
  64. Li DQ, Yang Y and Kumar R (2014) MTA family of proteins in DNA damage response: mechanistic insights and potential applications. Cancer Metastasis Rev 33, 993–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lim YY, Wright JA, Attema JL, Gregory PA, Bert AG, Smith E, Thomas D, Lopez AF, Drew PA, Khew‐Goodall Y et al (2013) Epigenetic modulation of the miR‐200 family is associated with transition to a breast cancer stem‐cell‐like state. J Cell Sci 126, 2256–2266. [DOI] [PubMed] [Google Scholar]
  66. Lin T, Ponn A, Hu X, Law BK and Lu J (2010a) Requirement of the histone demethylase LSD1 in Snai1‐mediated transcriptional repression during epithelial‐mesenchymal transition. Oncogene 29, 4896–4904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Lin Y, Wu Y, Li J, Dong C, Ye X, Chi YI, Evers BM and Zhou BP (2010b) The SNAG domain of Snail1 functions as a molecular hook for recruiting lysine‐specific demethylase 1. EMBO J 29, 1803–1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Lluis F, Pedone E, Pepe S and Cosma MP (2008) Periodic activation of Wnt/beta‐catenin signaling enhances somatic cell reprogramming mediated by cell fusion. Cell Stem Cell 3, 493–507. [DOI] [PubMed] [Google Scholar]
  69. Lu D, Choi MY, Yu J, Castro JE, Kipps TJ and Carson DA (2011) Salinomycin inhibits Wnt signaling and selectively induces apoptosis in chronic lymphocytic leukemia cells. Proc Natl Acad Sci U S A 108, 13253–13257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Lv T, Yuan D, Miao X, Lv Y, Zhan P, Shen X and Song Y (2012) Over‐expression of LSD1 promotes proliferation, migration and invasion in non‐small cell lung cancer. PLoS One 7, e35065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Maherali N and Hochedlinger K (2009) Tgfbeta signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and cMyc. Curr Biol 19, 1718–1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Mai TT, Hamaï A, Hienzsch A, Cañeque T, Müller S, Wicinski J, Cabaud O, Leroy C, David A, Acevedo V et al (2017). Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. https://doi.org/10.1038/nchem.2778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Manchado E, Weissmueller S, Morris JPt, Chen CC, Wullenkord R, Lujambio A, de Stanchina E, Poirier JT, Gainor JF, Corcoran RB et al (2016) A combinatorial strategy for treating KRAS‐mutant lung cancer. Nature 534, 647–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M et al (2008) The epithelial‐mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Marion RM, Strati K, Li H, Murga M, Blanco R, Ortega S, Fernandez‐Capetillo O, Serrano M and Blasco MA (2009) A p53‐mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 460, 1149–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. McDonald OG, Wu H, Timp W, Doi A and Feinberg AP (2011) Genome‐scale epigenetic reprogramming during epithelial‐to‐mesenchymal transition. Nat Struct Mol Biol 18, 867–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Meidhof S, Brabletz S, Lehmann W, Preca BT, Mock K, Ruh M, Schuler J, Berthold M, Weber A, Burk U et al (2015) ZEB1‐associated drug resistance in cancer cells is reversed by the class I HDAC inhibitor mocetinostat. EMBO Mol Med 7, 831–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Melisi D, Ishiyama S, Sclabas GM, Fleming JB, Xia Q, Tortora G, Abbruzzese JL and Chiao PJ (2008) LY2109761, a novel transforming growth factor beta receptor type I and type II dual inhibitor, as a therapeutic approach to suppressing pancreatic cancer metastasis. Mol Cancer Ther 7, 829–840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. van de Moosdijk AA, Fu NY, Rios AC, Visvader JE and van Amerongen R (2017) Lineage tracing of mammary stem and progenitor cells. Methods Mol Biol 1501, 291–308. [DOI] [PubMed] [Google Scholar]
  80. Morel AP, Lievre M, Thomas C, Hinkal G, Ansieau S and Puisieux A (2008) Generation of breast cancer stem cells through epithelial‐mesenchymal transition. PLoS One 3, e2888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Mu P, Zhang Z, Benelli M, Karthaus WR, Hoover E, Chen CC, Wongvipat J, Ku SY, Gao D, Cao Z et al (2017) SOX2 promotes lineage plasticity and antiandrogen resistance in TP53‐ and RB1‐deficient prostate cancer. Science 355, 84–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Nagasawa S, Sedukhina AS, Nakagawa Y, Maeda I, Kubota M, Ohnuma S, Tsugawa K, Ohta T, Roche‐Molina M, Bernal JA et al (2015) LSD1 overexpression is associated with poor prognosis in basal‐like breast cancer, and sensitivity to PARP inhibition. PLoS One 10, e0118002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Nieto MA (2013) Epithelial plasticity: a common theme in embryonic and cancer cells. Science 342, 1234850. [DOI] [PubMed] [Google Scholar]
  84. Nieto MA, Huang RY, Jackson RA and Thiery JP (2016) EMT: 2016. Cell 166, 21–45. [DOI] [PubMed] [Google Scholar]
  85. Ocana OH, Corcoles R, Fabra A, Moreno‐Bueno G, Acloque H, Vega S, Barrallo‐Gimeno A, Cano A and Nieto MA (2012) Metastatic colonization requires the repression of the epithelial‐mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724. [DOI] [PubMed] [Google Scholar]
  86. Park CY, Kim DK and Sheen YY (2011a) EW‐7203, a novel small molecule inhibitor of transforming growth factor‐beta (TGF‐beta) type I receptor/activin receptor‐like kinase‐5, blocks TGF‐beta1‐mediated epithelial‐to‐mesenchymal transition in mammary epithelial cells. Cancer Sci 102, 1889–1896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Park IK, Morrison SJ and Clarke MF (2004) Bmi1, stem cells, and senescence regulation. J Clin Invest 113, 175–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Park CY, Son JY, Jin CH, Nam JS, Kim DK and Sheen YY (2011b) EW‐7195, a novel inhibitor of ALK5 kinase inhibits EMT and breast cancer metastasis to lung. Eur J Cancer 47, 2642–2653. [DOI] [PubMed] [Google Scholar]
  89. Pattabiraman DR, Bierie B, Kober KI, Thiru P, Krall JA, Zill C, Reinhardt F, Tam WL, Weinberg RA (2016) Activation of PKA leads to mesenchymal‐to‐epithelial transition and loss of tumor‐initiating ability. Science 351, aad3680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Peinado H, Ballestar E, Esteller M and Cano A (2004) Snail mediates E‐cadherin repression by the recruitment of the Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol 24, 306–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Popowski M, Templeton TD, Lee BK, Rhee C, Li H, Miner C, Dekker JD, Orlanski S, Bergman Y, Iyer VR et al (2014) Bright/Arid3A acts as a barrier to somatic cell reprogramming through direct regulation of Oct4, Sox2, and Nanog. Stem Cell Reports 2, 26–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Puls LN, Eadens M and Messersmith W (2011) Current status of SRC inhibitors in solid tumor malignancies. Oncologist 16, 566–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Qin H, Blaschke K, Wei G, Ohi Y, Blouin L, Qi Z, Yu J, Yeh RF, Hebrok M and Ramalho‐Santos M (2012) Transcriptional analysis of pluripotency reveals the Hippo pathway as a barrier to reprogramming. Hum Mol Genet 21, 2054–2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Qin H, Diaz A, Blouin L, Lebbink RJ, Patena W, Tanbun P, LeProust EM, McManus MT, Song JS and Ramalho‐Santos M (2014) Systematic identification of barriers to human iPSC generation. Cell 158, 449–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Rausch MP, Hahn T, Ramanathapuram L, Bradley‐Dunlop D, Mahadevan D, Mercado‐Pimentel ME, Runyan RB, Besselsen DG, Zhang X, Cheung HK et al (2009) An orally active small molecule TGF‐beta receptor I antagonist inhibits the growth of metastatic murine breast cancer. Anticancer Res 29, 2099–2109. [PMC free article] [PubMed] [Google Scholar]
  96. Reichl P, Dengler M, van Zijl F, Huber H, Fuhrlinger G, Reichel C, Sieghart W, Peck‐Radosavljevic M, Grubinger M and Mikulits W (2015) Axl activates autocrine transforming growth factor‐beta signaling in hepatocellular carcinoma. Hepatology 61, 930–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Rios AC, Fu NY, Cursons J, Lindeman GJ and Visvader JE (2016) The complexities and caveats of lineage tracing in the mammary gland. Breast Cancer Res 18, 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Rodon J, Carducci M, Sepulveda‐Sanchez JM, Azaro A, Calvo E, Seoane J, Brana I, Sicart E, Gueorguieva I, Cleverly A et al (2015) Pharmacokinetic, pharmacodynamic and biomarker evaluation of transforming growth factor‐beta receptor I kinase inhibitor, galunisertib, in phase 1 study in patients with advanced cancer. Invest New Drugs 33, 357–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Rosano L, Di Castro V, Spinella F, Nicotra MR, Natali PG and Bagnato A (2007) ZD4054, a specific antagonist of the endothelin A receptor, inhibits tumor growth and enhances paclitaxel activity in human ovarian carcinoma in vitro and in vivo. Mol Cancer Ther 6, 2003–2011. [DOI] [PubMed] [Google Scholar]
  100. Rosano L, Spinella F, Di Castro V, Nicotra MR, Dedhar S, de Herreros AG, Natali PG and Bagnato A (2005) Endothelin‐1 promotes epithelial‐to‐mesenchymal transition in human ovarian cancer cells. Cancer Res 65, 11649–11657. [DOI] [PubMed] [Google Scholar]
  101. Rostaing L, Hertig A, Albano L, Anglicheau D, Durrbach A, Vuiblet V, Moulin B, Merville P, Hazzan M, Lang P et al (2015) Fibrosis progression according to epithelial‐mesenchymal transition profile: a randomized trial of everolimus versus CsA. Am J Transplant 15, 1303–1312. [DOI] [PubMed] [Google Scholar]
  102. Sakamoto T, Kobayashi S, Yamada D, Nagano H, Tomokuni A, Tomimaru Y, Noda T, Gotoh K, Asaoka T, Wada H et al (2016) A histone deacetylase inhibitor suppresses epithelial‐mesenchymal transition and attenuates chemoresistance in biliary tract cancer. PLoS One 11, e0145985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. dos Santos RL, Tosti L, Radzisheuskaya A, Caballero IM, Kaji K, Hendrich B and Silva JC (2014) MBD3/NuRD facilitates induction of pluripotency in a context‐dependent manner. Cell Stem Cell 15, 102–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Sato H, Shien K, Tomida S, Okayasu K, Suzawa K, Hashida S, Torigoe H, Watanabe M, Yamamoto H, Soh J et al (2017) Targeting the miR‐200c/LIN28B axis in acquired EGFR‐TKI resistance non‐small cell lung cancer cells harboring EMT features. Sci Rep 7, 40847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Schech A, Kazi A, Yu S, Shah P and Sabnis G (2015) Histone deacetylase inhibitor entinostat inhibits tumor‐initiating cells in triple‐negative breast cancer cells. Mol Cancer Ther 14, 1848–1857. [DOI] [PubMed] [Google Scholar]
  106. Scheel C, Eaton EN, Li SH, Chaffer CL, Reinhardt F, Kah KJ, Bell G, Guo W, Rubin J, Richardson AL et al (2011) Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell 145, 926–940. https://doi.org/10.1016/j.cell.2011.04.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Schobert R, Biersack B (2017) Multimodal HDAC inhibitors with improved anticancer activity. Curr Cancer Drug Targets. https://doi.org/10.2174/1568009617666170206102613 [DOI] [PubMed] [Google Scholar]
  108. Shao DD, Xue W, Krall EB, Bhutkar A, Piccioni F, Wang X, Schinzel AC, Sood S, Rosenbluh J, Kim JW et al (2014) KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158, 171–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA and Shi Y (2004) Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953. [DOI] [PubMed] [Google Scholar]
  110. Smith ZD, Sindhu C and Meissner A (2016) Molecular features of cellular reprogramming and development. Nat Rev Mol Cell Biol 17, 139–154. [DOI] [PubMed] [Google Scholar]
  111. Son JY, Park SY, Kim SJ, Lee SJ, Park SA, Kim MJ, Kim SW, Kim DK, Nam JS and Sheen YY (2014) EW‐7197, a novel ALK‐5 kinase inhibitor, potently inhibits breast to lung metastasis. Mol Cancer Ther 13, 1704–1716. [DOI] [PubMed] [Google Scholar]
  112. Song LB, Li J, Liao WT, Feng Y, Yu CP, Hu LJ, Kong QL, Xu LH, Zhang X, Liu WL et al (2009) The polycomb group protein Bmi‐1 represses the tumor suppressor PTEN and induces epithelial‐mesenchymal transition in human nasopharyngeal epithelial cells. J Clin Invest 119, 3626–3636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Song SJ, Poliseno L, Song MS, Ala U, Webster K, Ng C, Beringer G, Brikbak NJ, Yuan X, Cantley LC et al (2013) MicroRNA‐antagonism regulates breast cancer stemness and metastasis via TET‐family‐dependent chromatin remodeling. Cell 154, 311–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Sun M, Song CX, Huang H, Frankenberger CA, Sankarasharma D, Gomes S, Chen P, Chen J, Chada KK, He C et al (2013) HMGA2/TET1/HOXA9 signaling pathway regulates breast cancer growth and metastasis. Proc Natl Acad Sci U S A 110, 9920–9925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Takahashi K and Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676. [DOI] [PubMed] [Google Scholar]
  116. Tam WL, Lu H, Buikhuisen J, Soh BS, Lim E, Reinhardt F, Wu ZJ, Krall JA, Bierie B, Guo W et al (2013) Protein kinase C alpha is a central signaling node and therapeutic target for breast cancer stem cells. Cancer Cell 24, 347–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Tam WL and Weinberg RA (2013) The epigenetics of epithelial‐mesenchymal plasticity in cancer. Nat Med 19, 1438–1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Tang HM, Kuay KT, Koh PF, Asad M, Tan TZ, Chung VY, Lee SC, Thiery JP and Huang RJ (2016) An epithelial marker promoter induction screen identifies histone deacetylase inhibitors to restore epithelial differentiation and abolishes anchorage independence growth in cancers. Cell Death Discov 2, 16041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Tetteh PW, Basak O, Farin HF, Wiebrands K, Kretzschmar K, Begthel H, van den Born M, Korving J, de Sauvage F, van Es JH et al (2016) Replacement of lost Lgr5‐positive stem cells through plasticity of their enterocyte‐lineage daughters. Cell Stem Cell 18, 203–213. [DOI] [PubMed] [Google Scholar]
  120. Thiery JP, Acloque H, Huang RY and Nieto MA (2009) Epithelial‐mesenchymal transitions in development and disease. Cell 139, 871–890. [DOI] [PubMed] [Google Scholar]
  121. Thomson S, Buck E, Petti F, Griffin G, Brown E, Ramnarine N, Iwata KK, Gibson N and Haley JD (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non‐small‐cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer Res 65, 9455–9462. [DOI] [PubMed] [Google Scholar]
  122. Tryndyak VP, Beland FA and Pogribny IP (2010) E‐cadherin transcriptional down‐regulation by epigenetic and microRNA‐200 family alterations is related to mesenchymal and drug‐resistant phenotypes in human breast cancer cells. Int J Cancer 126, 2575–2583. [DOI] [PubMed] [Google Scholar]
  123. Tsai YP, Chen HF, Chen SY, Cheng WC, Wang HW, Shen ZJ, Song C, Teng SC, He C and Wu KJ (2014) TET1 regulates hypoxia‐induced epithelial‐mesenchymal transition by acting as a co‐activator. Genome Biol 15, 513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Tsai JH, Donaher JL, Murphy DA, Chau S and Yang J (2012) Spatiotemporal regulation of epithelial‐mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 22, 725–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Valk‐Lingbeek ME, Bruggeman SW and van Lohuizen M (2004) Stem cells and cancer; the polycomb connection. Cell 118, 409–418. [DOI] [PubMed] [Google Scholar]
  126. Vidal SE, Amlani B, Chen T, Tsirigos A and Stadtfeld M (2014) Combinatorial modulation of signaling pathways reveals cell‐type‐specific requirements for highly efficient and synchronous iPSC reprogramming. Stem Cell Reports 3, 574–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Voon DC and Thiery JP (2017) The emerging roles of RUNX transcription factors in epithelial‐mesenchymal transition. Adv Exp Med Biol 962, 471–489. [DOI] [PubMed] [Google Scholar]
  128. Voon DC, Wang H, Koo JK, Nguyen TA, Hor YT, Chu YS, Ito K, Fukamachi H, Chan SL, Thiery JP et al (2012) Runx3 protects gastric epithelial cells against epithelial‐mesenchymal transition‐induced cellular plasticity and tumorigenicity. Stem Cells 30, 2088–2099. [DOI] [PubMed] [Google Scholar]
  129. Vultur A, Buettner R, Kowolik C, Liang W, Smith D, Boschelli F and Jove R (2008) SKI‐606 (bosutinib), a novel Src kinase inhibitor, suppresses migration and invasion of human breast cancer cells. Mol Cancer Ther 7, 1185–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Xia H, Ooi LL and Hui KM (2013) MicroRNA‐216a/217‐induced epithelial‐mesenchymal transition targets PTEN and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology 58, 629–641. [DOI] [PubMed] [Google Scholar]
  131. Xu Y, Qin L, Sun T, Wu H, He T, Yang Z, Mo Q, Liao L and Xu J (2017) Twist1 promotes breast cancer invasion and metastasis by silencing Foxa1 expression. Oncogene 36, 1157–1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Yadavalli S, Jayaram S, Manda SS, Madugundu AK, Nayakanti DS, Tan TZ, Bhat R, Rangarajan A, Chatterjee A, Gowda H et al (2017) Data‐driven discovery of extravasation pathway in circulating tumor cells. Sci Rep 7, 43710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Yang MH, Hsu DS, Wang HW, Wang HJ, Lan HY, Yang WH, Huang CH, Kao SY, Tzeng CH, Tai SK et al (2010) Bmi1 is essential in Twist1‐induced epithelial‐mesenchymal transition. Nat Cell Biol 12, 982–992. [DOI] [PubMed] [Google Scholar]
  134. Yano S, Wang W, Li Q, Matsumoto K, Sakurama H, Nakamura T, Ogino H, Kakiuchi S, Hanibuchi M, Nishioka Y et al (2008) Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor‐activating mutations. Cancer Res 68, 9479–9487. [DOI] [PubMed] [Google Scholar]
  135. Ye X, Tam WL, Shibue T, Kaygusuz Y, Reinhardt F, Ng Eaton E and Weinberg RA (2015) Distinct EMT programs control normal mammary stem cells and tumour‐initiating cells. Nature 525, 256–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff SJ, Ciciliano JC, Wells MN, Shah AM et al (2013a) Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339, 580–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Yu Y, Wang B, Zhang K, Lei Z, Guo Y, Xiao H, Wang J, Fan L, Lan C, Wei Y et al (2013b) High expression of lysine‐specific demethylase 1 correlates with poor prognosis of patients with esophageal squamous cell carcinoma. Biochem Biophys Res Commun 437, 192–198. [DOI] [PubMed] [Google Scholar]
  138. Yun‐Ju Huang R and Yo‐Yan Huang T (2016) A new dimension in drug discovery: reversing epithelial‐mesenchymal transition (EMT). Cell Death Dis 7, e2417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Zhang B, Halder SK, Zhang S and Datta PK (2009) Targeting transforming growth factor‐beta signaling in liver metastasis of colon cancer. Cancer Lett 277, 114–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Zhao ZK, Yu HF, Wang DR, Dong P, Chen L, Wu WG, Ding WJ and Liu YB (2012) Overexpression of lysine specific demethylase 1 predicts worse prognosis in primary hepatocellular carcinoma patients. World J Gastroenterol 18, 6651–6656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS, Kalluri R (2015) Epithelial‐to‐mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 527, 525–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Zhou JN, Zeng Q, Wang HY, Zhang B, Li ST, Nan X, Cao N, Fu CJ, Yan XL, Jia YL et al (2015) MicroRNA‐125b attenuates epithelial‐mesenchymal transitions and targets stem‐like liver cancer cells through small mothers against decapentaplegic 2 and 4. Hepatology 62, 801–815. [DOI] [PubMed] [Google Scholar]
  143. Zou J, Luo H, Zeng Q, Dong Z, Wu D and Liu L (2011) Protein kinase CK2alpha is overexpressed in colorectal cancer and modulates cell proliferation and invasion via regulating EMT‐related genes. J Transl Med 9, 97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Zucali PA, Ruiz MG, Giovannetti E, Destro A, Varella‐Garcia M, Floor K, Ceresoli GL, Rodriguez JA, Garassino I, Comoglio P et al (2008) Role of cMET expression in non‐small‐cell lung cancer patients treated with EGFR tyrosine kinase inhibitors. Ann Oncol 19, 1605–1612. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular Oncology are provided here courtesy of Wiley

RESOURCES