Skip to main content
Erschienen in: Molecular Cancer 1/2017

Open Access 01.12.2017 | Review

Epigenetics in cancer stem cells

verfasst von: Tan Boon Toh, Jhin Jieh Lim, Edward Kai-Hua Chow

Erschienen in: Molecular Cancer | Ausgabe 1/2017

Abstract

Compelling evidence have demonstrated that bulk tumors can arise from a unique subset of cells commonly termed “cancer stem cells” that has been proposed to be a strong driving force of tumorigenesis and a key mechanism of therapeutic resistance. Recent advances in epigenomics have illuminated key mechanisms by which epigenetic regulation contribute to cancer progression. In this review, we present a discussion of how deregulation of various epigenetic pathways can contribute to cancer initiation and tumorigenesis, particularly with respect to maintenance and survival of cancer stem cells. This information, together with several promising clinical and preclinical trials of epigenetic modulating drugs, offer new possibilities for targeting cancer stem cells as well as improving cancer therapy overall.
Abkürzungen
ABC
ATP-binding cassette
ABCG2
ATP-binding cassette sub-family G member 2
ALL
Acute lymphoblastic leukemia
AML
Acute myeloid leukemia
AML1-ETO
Acute myeloid leukemia protein 1 Eight twenty-one protein
APC
Adenomatous polyposis coli
ASCL1
Achaete-scute family BHLH transcription factor 1
BCC
Basal cell carcinoma
BCL2
B-cell lymphoma 2
BET
Bromodomain and extra-terminal
Bmi1
B-lymphoma Mo-MLV insertion region 1 homolog
CK1
Casein kinase 1
CK19
Cytokeratin 19
CSC
Cancer stem cells
DACT3
Polycomb repressor complex 2, PCR2 Dishevelled-binding antagonist of beta-catenin 3
DKK1
Dickkopf-related protein 1
DNMTs
DNA methyltransferases
DZNep
3-deazaneplanocin A
ECM
Extracellular matrix
EED
Embryonic ectoderm development protein
EMT
Epithelial-to-mesenchymal transition
EpCAM
Epithelial cell adhesion molecule
ESCs
Embryonic stem cells
EZH2
Enhancer of zeste homolog 2
FAD
Flavin adenine dinucleotide
G9a/EHMT2
Euchromatic histone lysine methyltransferase 2
GBM
Glioblastoma multiforme
GLP/EHMT1
Euchromatic histone lysine methyltransferase 1
GNPs
Granule neuron progenitors/precursors
GSK-3β
Glycogen synthase kinase 3 beta
H3K27
Histone H3 lysine 27
H3K36
Histone H3 lysine 36
H3K4
Histone H3 lysine 4
H3K79
Histone H3 lysine 79
H3K9
Histone H3 lysine 9
H4K20
Histone H4 lysine 20
HAT
Histone acetyltransferases
HCC
Hepatocellular carcinoma
HDAC
Histone deacetylase
Hh
Hedgehog
HKMT
Histone lysine methyltransferase
HoxA9
Homeobox A9
ICC
Intrahepatic cholangiocarcinoma
JmjC
Jumonji domain-containing (JmjC)
KDM
Histone lysine demethylase
Kif7
Kinesin family member 7
LDR5/6
Low density lipoprotein receptor-related protein 5/6
LSD
Lysine-specific demethylase
MDR1
Multidrug resistance protein 1
Meis1
Meis homeobox 1
miRNAs
Micro RNAs
MLL
mixed lineage leukemia
MLL2
Mixed lineage leukemia protein 2
MRP1
Multidrug resistance-associated protein 1
NF-κB
Nuclear factor kappa b
NICD
Notch intracellular domain
NKD1
Naked cuticle homolog 1
NMC
NUT midline carcinoma
NSCLC
non-small cell lung cancer
NSCs
Neural stem cells
OCT4
Octamer-binding transcription factor 4
PML-RARα
Promyelocytic leukemia protein retinoic acid receptor alpha
PTCH1
Patched receptor
RBPJ-κ
Recombination signal binding protein for immunoglobulin kappa J region
SAH
S-adenosylhomocysteine hydrolase
SAM
S-adenosyl methionine
SFRP-1
Secreted frizzled-related protein 1
Shh
Sonic hedgehog ligand
SirT1
Sirtuin 1
SMO
Smoothened
SMRT
Nuclear receptor co-repressor 2
SNAIL
Snail family zinc finger 1
SP
Side population
STRAP
Serine-threonine kinase receptor-associated protein
SUFU
Suppressor of fused homolog
SUV39H1
Suppressor of variegation 3–9 homolog 1
SUV39H2
Suppressor of variegation 3–9 homolog 2
Suz12
Suppressor of zeste 12 protein homolog
TCF/LEF
T-cell factor/lymphoid enhancer factor
TGF-β
Transforming growth factor-β
TNBC
Triple-negative breast cancer
TWIST1
Twist-related protein 1
UTX
Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX)
WIF-1
Wnt inhibitory factor 1
ZEB1
Zinc finger E-box-binding homeobox 1
ZEB2
Zinc finger E-box-binding homeobox 2

Background

Advances in genomic and epigenomic research has shaped our understanding of cancer over the past two decades. Rather than merely a perpetuating mass of dysregulated cells growing in an uncontrolled manner, cancer is also defined by the dynamic genetic and epigenetic alterations that contribute to cancer initiation and progression. Since epigenetic changes such as DNA methylation and histone modifications are crucial factors in developmental programming of stem cells to specific lineages of cellular and tissue differentiation, aberrant epigenetic alterations may transform normal stem cells to cancer stem cells with the loss of differentiation capacity and the acquisition of stem-like characteristics. More importantly, epigenetic mechanisms have been shown to be implicated in the observed variability of treatment response. For instance, a small subset of cells has been shown to be resistant to drug therapy in a variety of cancers such as melanoma, gastric, colon and lung cancers as a result of aberrant expression of key epigenetic modifiers. In this review, we will focus our discussion on the epigenetic regulation of CSCs and their impact on tumor-initiation, progression and response to therapies. We will also discuss recent advances in using epigenetic therapy to target cancer stem cells.

Main text

Cancer stem cells (CSCs)

Cancer stem cells (CSCs) define a small, unique subset of cells with self-renewal ability and the capacity to generate the different cell types that constitute the whole tumor [1]. These cells are termed CSCs because of their “stem-like” properties commonly shared with normal tissue stem cells. Such properties include extensive self-renewal ability (symmetrical and asymmetrical) and differentiation capacity. It should be noted that a general capacity to differentiate is not a mandatory feature of CSCs and that the ability of CSCs to differentiate and repopulate the cell types found in the original tumor is of greater significance. More importantly, CSCs should demonstrate potent tumor-initiation capacity. This property is usually demonstrated by injecting limited number of CSCs into an orthotopic in vivo environment to generate the bulk tumor. Nevertheless, the concept of CSC is of significant importance as it highlights the need to eradicate the CSC populations to achieve an effective cure.
The first clear evidence of CSCs being a key tumor-initiating subset of cancer cells was demonstrated in acute myeloid leukemia (AML) where prospective CSCs were isolated using cell surface markers that identify normal haematopoietic stem cells and evaluated for their tumor-initiating properties [2, 3]. Since then, similar identifications of tumor-initiating populations have been identified in multiple solid tumors that includes brain, breast, liver, ovary, prostate, lung, melanoma and colon cancers, by using different cell surface markers or through side population (SP) analysis [411]. For instance, in malignant glioma and medulloblastoma tumors, a putative neural stem cell marker, CD133 has been demonstrated to be adequate and essential to initiate and recapitulate the tumor upon xenotransplantation into immune-compromised mice [5]. However, this finding has been disputed as tumors can also arise from CD133-negative cells in a subset of glioma tumors [12]. In addition, CD133 surface marker expression has been demonstrated to change according to disease state and progression, further complicating its role as a bona fide CSC marker in brain tumors [13, 14]. In liver cancers such as hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC), similar use of cell surface markers such as epithelial cell adhesion molecule (EpCAM), cytokeratin 19 (CK19), CD133, CD90, CD44, CD24, and CD13 has been applied to define a subpopulation of liver cancer cells as CSCs [15]. Importantly, it has recently been shown that these CSC markers are not specific to liver CSCs, and that distinct populations of liver CSCs express different surface markers possibly due to the strong intra- and inter-heterogeneity and varied etiology of liver cancer [16]. As a result, CSC studies have begun to move away from the reliance of cell surface markers to identify tumor-initiating cells and have begun to identify other complementary methods of measuring the functional activities of CSCs that may serve to identify CSCs as well as the molecular mechanisms that regulate CSCs [17].
Currently, the central theme of the CSC model is the ability of a subset of cells at the apex of the hierarchy to propagate tumors and promote tumor progression as compared to the non-tumorigenic cells within the bulk tumor. One of the gold standards to functionally identify CSCs is the capacity of these cells to regenerate a phenotypic copy of the original tumor in an orthotopic transplantation model. Non-CSCs, by definition, lack this ability and fail to generate tumors in the transplantation model. It is important to note that the CSC hierarchy model may not be ubiquitous for all cancers and that some tumorigenic cells are common in certain cancers. It is also important to note that such transplantation assays measure the tumorigenic potential of the cells to form tumors and not their actual fate. For example, alterations in tumorigenic assays carried out by Quintana and colleagues showed that CSC frequency could be increased by changing several experimental parameters such as the use of extracellular matrix (ECM) in the form of matrigel, prolonging the duration for tumor formation, and varying the severity of immune-compromised mice used [18]. This study highlighted that the tumor-initiating capacity may be an artificial consequence of the conditions employed in xenograft mouse models.
While analyzing CSC surface marker expression in primary tumors has been often performed to study the clinical impact of CSCs on tumor progression, more often than not, this has resulted in ambiguous data possibly due to the fact that CSC properties that sustain the primary tumor phenotype are defined by more than just specific marker expression [19, 20]. Analysis of key signalling pathway activity that resembles those functioning in stem-like cells, is more likely to accurately interrogate the clinical contribution of CSCs. An example of such studies was carried out by Lim et al. in BRCA1 mutation-associated breast tumors, where the authors prospectively isolated distinct subpopulations of normal and tumorigenic epithelial cells from BRCA1 mutation heterozygous individuals and found that luminal progenitors were highly represented in BRCA1 mutation-associated breast tumors, more than the stem cell population [21]. This suggests that luminal progenitors are more likely the cells-of-origin for BRCA1 mutation-associated breast tumors, which was later confirmed in a transgenic mouse model study carried out by Molyneux and colleagues [22]. These studies highlight the predictive capability of gene expression mapping of pathway activation rather than specific marker identity. In a separate study, John Dick and colleagues demonstrated that tumor-initiating AML stem cells contribute to disease progression and patient survival outcome, underscoring the importance of functionally defining the CSCs [23]. More importantly, the contribution of CSCs, with preferential activation of core stem cell programs, to patient survival outcome has been demonstrated. The study by Shats et al. showed that a stemness gene signature derived from embryonic stem cells (ESCs) could predict a breast cancer patient cohort sensitive to drugs linked to this signature using a Connectivity Map [24], demonstrating the clinical contribution of CSCs to patient outcome [25]. Collectively, these studies highlight that CSCs that perpetuate tumors are not merely defined by surface marker expression, but more importantly and more accurately by their gene expression profiles and consequent pathway activations.

Epigenetics: normal and cancer stem cells

Epigenetic regulation of the genome is one of the primary means by which genetic code is altered to control cellular developmental hierarchies. Epigenetic mechanisms such as histone modifications, DNA methylation, chromatin remodelling and even changes in noncoding RNAs including miRNAs together govern the epigenome landscape that dictate the outcome of cell fate specification without changes to the DNA sequences. Such changes in the genome is important during normal mammalian development and ESCs differentiation [26]. Importantly, gene expression profiles change during cellular differentiation according to not only a network of transcription factors but also the “epigenomic landscape” of the cell. For the purpose of this review, we will focus our discussions on two primary mechanisms of epigenetic regulation: histone modifications and DNA methylation.
Histone methylation occurs predominantly on lysine (K) and arginine (R) residues and these methylation marks serve as docking sites for histone readers [27]. Both lysine and arginine methylation can occur on both histones and non-histone proteins. The highly conserved histone lysine methylation occurs at three different levels: mono-, di-, and tri-methylation. Such modifications are commonly associated with gene activation or repression, depending on the target histone modification. For instance, histone H3 lysine 4 (H3K4), histone H3 lysine 36 (H3K36), and histone H3 lysine 79 (H3K79) are associated with gene activation whereas histone H3 lysine 9 (H3K9), histone H3 lysine 27 (H3K27) and histone H4 lysine 20 (H4K20) are associated with gene repression. The N-terminal tails of histones frequently undergo other post-translational modifications, which play significant roles in various DNA-templated processes, including transcription [28]. Hence, aberrations in histone modifications can lead to deregulated gene expression as seen in various human disease and malignancies.
DNA methyltransferases (DNMTs) are a class of enzymes involved in transferring a methyl group from S-adenosyl methionine (SAM) to cytosine bases of CpG dinucleotides at gene promoters and regulatory regions [29]. CpG dinucleotides are concentrated in short CpG-rich regions known commonly as “CpG islands”. In humans, CpG islands occupy about 60% of the gene promoters. CpG promoter islands can be methylated during development that results in long-term gene silencing. One classic example of such naturally occurring CpG methylation is the X-chromosome inactivation and the imprinted genes. DNA hypermethylation has also been associated with the silencing of tumor suppressor genes as well as differentiation genes in various cancers [30]. The reduced expression of these genes may then contribute to the formation of CSCs within tumor cell populations [31, 32]. Indeed, the importance of DNA methylation in maintaining CSC properties have been reported in leukemic, lung and colon stem cells [3335]. The accumulation of epigenetic abnormalities has been suggested to be an early event that predisposes these tumor cells to acquire further mutations and genomic instability. This is supported by the fact that epigenetic machinery is crucial for the maintenance of normal stem and progenitor cells and that any epigenetic deregulation can lead to accumulation of cells with increased stemness properties and self-renewal ability, thus giving rise to CSCs.

Key CSC pathways regulated by epigenetic mechanisms

Wnt/β-catenin signaling pathway

The canonical Wnt/β-catenin signaling pathway mediates gene activation through the transcription factor β-catenin. In the absence of Wnt signaling, cytoplasmic β-catenin is inactivated by a degradation complex comprising Adenomatous polyposis coli (APC), Axin, glycogen synthase kinase 3 beta (GSK-3β), and casein kinase 1 (CK1). Phosphorylation by GSK-3β targets β-catenin for ubiquitination and subsequent proteasomal degradation. Upon Wnt ligand binding to Frizzled receptors, the degradation complex is inactivated via low density lipoprotein receptor-related protein 5/6 (LDR5/6) and Dishevelled, allowing for stabilisation of β-catenin. Accumulated β-catenin then translocates into the nucleus, where it associates with T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors to induce transcription of Wnt target genes such as CCND1 and MYC. The Wnt/β-catenin pathway has important functions in normal tissue development and maintenance, as well as in self-renewal and differentiation of CSCs [36, 37]. In fact, the Wnt/β-catenin pathway has been found to be aberrantly activated in a variety of cancers, either via genetic alterations, such as mutations in CTNNB1, APC and AXIN genes [3840], or through epigenetic modulation.
DNA methylation has been linked to aberrant Wnt/β-catenin pathway activation through the enhanced promoter methylation and subsequent silencing of various Wnt inhibitors such as Wnt inhibitory factor 1 (WIF-1), AXIN2, Secreted frizzled-related protein 1 (SFRP-1), and Dickkopf-related protein 1 (DKK1) in breast and colorectal cancers [4143]. In gastric cancer, Yoda et al. showed that aberrant methylation of Wnt negative regulators, including DKK3, Naked cuticle homolog 1 (NKD1) and SFRP1, could lead to activation of Wnt/β-catenin pathway [44]. Deregulation of Wnt/β-catenin pathway in cancer is also mediated by aberrant histone modifications. Decreased acetylation of H3K16 and increased H3K27 trimethylation along with recruitment of Sirtuin 1 (SirT1), enhancer of zeste homolog 2 (EZH2) and suppressor of zeste 12 protein homolog (Suz12) (components of polycomb repressor complex 2, PCR2) to the promoter of DKK1 inhibited the expression of the DKK1 Wnt antagonist (Fig. 1) [45]. In colorectal cancer, Dishevelled-binding antagonist of beta-catenin 3 (DACT3), an antagonist of Dishevelled, was found to be regulated by bivalent histone modifications—activating H3K4me3 and repressive H3K27me3 histone marks—at its locus [46]. This bivalent histone state was associated with decreased DACT3 expression in colorectal cancer cell lines [46]. In addition, methylation of H3K4 at the regulatory element of DKK1 marks the site for binding by the transcription factor Achaete-scute family BHLH transcription factor 1 (ASCL1), resulting in a repressed chromatin configuration [47]. ASCL1-mediated inhibition of DKK1 consequently leads to activation of Wnt signaling, and ASCL1 was found to be crucial for glioblastoma CSC maintenance and tumorigenicity [4749].
Besides alterations in DNA and histones, non-coding RNAs have also been found to act as epigenetic modulators of Wnt/β-catenin signaling. Wang et al. demonstrated that long non-coding RNA of transcription factor 7 (lncTCF7), which is highly upregulated in liver CSCs, is able to induce TCF7 expression by recruiting the Switch/sucrose non-fermentable (SWI/SNF) chromatin remodelling complex to its promoter [50]. This subsequently activates the Wnt pathway, leading to self-renewal of liver CSCs and tumor propagation.

Hedgehog signaling pathway

The Hedgehog (Hh) signaling pathway plays important roles in guiding cell fate during embryonic development and in maintaining adult tissue homeostasis [51, 52]. It also functions in regulating stem and progenitor cell proliferation and maintenance in several tissues [53]. In the absence of sonic hedgehog ligand (Shh), the Patched receptor (PTCH1) prevents activation of Smoothened (SMO), allowing Gli proteins to be sequestered by suppressor of fused homolog (SUFU) and kinesin family member 7 (Kif7). Upon Shh binding to PTCH1, SMO is activated and mediates Hh signaling transduction via release of Gli proteins, which then enter the nucleus and act as transcription factors. Gli1 activates transcription of Hh target genes, Gli2 can both activate and repress transcription, while Gli3 functions as a transcriptional repressor.
The Hh signaling has been implicated in tumorigenesis in various tissues [54]. In basal cell carcinoma (BCC), upregulation of Hh signaling in stem cells in the interfollicular epidermis [55] or within the hair follicle [56] has been reported to contribute to tumor formation. In medulloblastomas, granule neuron progenitors/precursors (GNPs) in the cerebellum that have constitutively active Hh signaling have been identified as cells-of-origin of the tumor [54, 57, 58].
The Hh pathway is activated by genetic mutations in both BCC and medulloblastoma. However, epigenetic mechanisms also play a role in modulating the expression and function of Hh pathway components in various tumors. The chromatin remodelling protein SNF5 directly interacts with Hh signaling effector Gli1 to downregulate expression of Hh target genes. SNF5 is a member of the SWI-SNF complex and inhibits gene expression by altering chromatin structure at Gli1-regulated promoters, which includes genes such as Ptch1 and Gli1 itself. Hence, inactivation of SNF5 would contribute to aberrant Hh signaling activity as seen in human malignant rhabdoid tumors [5961].
In addition, histone deacetylases are also involved in regulating Gli protein function. Gli1 and Gli2 proteins require deacetylation by HDAC1 to be transcriptionally active, and they, in turn, can induce HDAC1 expression through a positive autoregulatory loop [62]. This mechanism is inhibited by E3-ubiquitin ligase complex (comprising Cullin3 and renin, REN)-mediated degradation of HDAC1. However, REN is often deleted in human medulloblastoma [63], resulting in increased levels of HDAC1 and Gli1, and subsequent deregulation of Hh signaling in neural progenitors and tumor cells [62].
Hh pathway can also be epigenetically regulated by aberrant DNA methylation. Studies have found that hypomethylation of Shh promoter leads to enhanced expression of Shh ligand in breast and gastric cancers (Fig. 1) [64, 65]. Indeed, Duan and colleagues reported that promoter hypomethylation allowed nuclear factor kappa b (NF-κB) to bind and activate transcription of Shh, resulting in overexpression of the ligand [66]. Consequently, the upregulation of Hh signaling was able to promote self-renewal and invasiveness in breast cancer cells [66].

Notch signaling pathway

Notch is a transmembrane receptor involved in cell contact-dependent signaling [67]. Binding of ligands Jagged1/2 or Delta1-4 triggers cleavage of Notch intracellular domain (NICD) by γ-secretase and its release into the cytoplasm [68]. NICD then translocates into the nucleus, where it interacts with recombination signal binding protein for immunoglobulin kappa J region (RBPJ-κ) to transcriptionally induce expression of Notch target genes, such as MYC and HES1 [69]. In the inactive state, RBPJ-κ recruits co-repressor complexes to suppress Notch target genes [70].
Notch signaling is an evolutionarily conserved pathway that has important roles in development of various tissues and organs [71]. It also regulates cell proliferation and differentiation across a wide range of cell types and during different stages of cell lineage progression [69]. Furthermore, Notch pathway modulates stem cell differentiation and self-renewal. Importantly, Notch signaling has been shown to be crucial for survival of neural stem cells (NSCs) [72]. In murine intestinal stem cells, loss of B-lymphoma Mo-MLV insertion region 1 homolog (Bmi1), a target of Notch signaling, decreases proliferation and induces cellular differentiation into goblet cells [73]. Deregulation of Notch pathway has been implicated in various tumors such as prostate cancer, breast cancer, lung cancer, colorectal cancer and haematological malignancies [7477]. Recent studies have also reported the role of Notch signaling in breast, colon and oesophageal CSCs [7880].
Epigenetic modifications affecting various components of the Notch pathway have been found to cause aberrations in Notch signaling activity. Overexpression of Notch ligand Jagged2 in multiple myeloma has been associated with enhanced histone acetylation at the JAGGED2 promoter region [81]. Nuclear co-repressors such as nuclear receptor co-repressor 2 (SMRT) normally recruit HDACs to promoter regions to regulate gene expression. However, in multiple myeloma, the decreased levels of nuclear co-repressor SMRT reduces HDAC recruitment to JAGGED2 promoter, resulting in increased transcription of the Notch ligand and subsequent activation of Notch signaling [81]. In addition, Jin et al. reported that serine-threonine kinase receptor-associated protein (STRAP) promotes stemness in colorectal cancer-initiating cells via modulating the Notch pathway [80]. They found that STRAP interacts with EZH2 and SUZ12 of PRC2 complex, inhibiting histone methylation of H3K27 on HES1 and HES5 promoters, leading to gene activation (Fig. 1). This was concordant with the finding that both genes had increased activating (H3K4me3) and decreased repressive (H3K27me3) histone marks in wild-type (WT) cells as compared to STRAP knockdown (KD) cells. Moreover, ectopically expressed HES1 or HES5 was able to rescue the stemness phenotype in STRAP KD cells [80], further demonstrating the significance of Notch signaling in regulating stemness potential in CSCs.

Epigenetic regulation of metastasis and chemoresistance pathways

During tumor progression, metastasis of tumor cells has been linked to the induction of epithelial-to-mesenchymal transition (EMT). EMT is a multi-step process that results in decreased cell-cell adhesion, loss of cell polarity, increased cell motility, and gain of invasive mesenchymal properties [82, 83]. There is evidence that activation of EMT can confer cells with CSC and tumor-initiating properties [84, 85]. It was reported that EMT induction in both immortalised and transformed human mammary epithelial cells resulted in increased expression of CSC markers and mammosphere formation. Moreover, stem-like cells of mammary carcinomas were also found to express markers of EMT [85]. The relationship between EMT and acquisition of stem-like properties in tumor cells suggests that stemness properties may help increase the chances of disseminated tumor cells to successfully metastasize to distant sites [70].
Several signaling pathways involved in embryonic development, such as Wnt, Hedgehog and Notch, have been identified to regulate the EMT process [86, 87]. The transforming growth factor-β (TGF-β) family of cytokines are also known inducers of EMT [88, 89]. Hence, deregulation of these pathways and proteins could activate aberrant EMT induction, resulting in tumor metastasis and contribute to poorer patient prognosis. A hallmark of EMT is the loss of membrane protein E-cadherin, which functions in maintaining cell-cell adhesion [9092]. Loss of E-cadherin can arise from mutations in its encoding gene CDH1, or via mechanisms that regulate its expression and function, including transcriptional repressors Twist-related protein 1 (TWIST1), Snail family zinc finger 1 (SNAIL), Zinc finger E-box-binding homeobox 1 (ZEB1) and Zinc finger E-box-binding homeobox 2 (ZEB2) [93]. Epigenetic mechanisms have also been found to play a dynamic role in silencing E-cadherin expression. For instance, DNA methylation of E-cadherin promoter helps to recruit HDACs to the site, leading to histone deacetylation and transcriptional silencing [94, 95]. In addition, histone methylation of CDH1 promoter by EZH2 and PRC2 complex, which is recruited by Snail1, also represses E-cadherin expression [96, 97].
Micro RNAs (miRNAs) that regulate the EMT pathway are epigenetically regulated as well. MiR-200 family members and miR-205 repress EMT and invasion by directly inhibiting transcription factors ZEB1 and ZEB2 [98100]. Hence, inhibition of these miRNAs would result in increased EMT and metastasis. This is observed in high-grade breast cancers, whereby low levels of miR-200c is correlated with upregulation of EMT and stemness markers [101]. Silencing of miR-200c and miR-205 expression can also occur via enrichment of H3K27me3-mediated chromatin remodelling and DNA methylation, which leads to induction of EMT and CSC phenotype in immortalised human bronchial epithelial cells [102].
Studies have shown that cells with both CSC properties and EMT-like phenotype tend to be more resistant to chemotherapy drugs as compared to other cancer cell populations [103105]. Arumugam et al. demonstrated that pancreatic cancer cell lines with EMT features were resistant to common chemotherapy drugs such as gemcitabine, 5-fluorouracil and cisplatin [106]. Moreover, cells that were resistant to gemcitabine expressed high ZEB1 and low E-cadherin, and acquired greater cell migration ability [106]. Indeed, these findings indicate that epigenetic modulations involved in the gain of CSC and EMT properties would most likely impact tumor cells’ response to therapy.
The increased drug resistance observed in CSCs is commonly mediated by enhanced expression of drug efflux transporters, such as ATP-binding cassette (ABC) family of transporters, which includes ATP-binding cassette sub-family G member 2 (ABCG2), multidrug resistance protein 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1) [17, 107, 108]. These drug transporters utilise ATP in moving drugs out of the cell against its concentration gradient. The expression of these transporters are regulated by various mechanisms and pathways, and their deregulation would result in an enrichment of these proteins and drug efflux capability. Studies have shown that MRP1 expression can be upregulated by Notch signaling, and is responsible for drug resistance in CSCs [109, 110]. Expression of ABCG2 is upregulated upon enrichment of permissive histone modifications such as greater histone H3 acetylation, increased H3K4 tri-methylation and phosphorylation of H3S10, as well as decreased HDAC1 levels [111]. These histone marks along with decreased H3K9 tri-methylation allow RNA polymerase II and chromatin remodelling protein Brahma-related gene 1 (Brg1) to gain access to the promoter and activate transcription of ABCG2 [111]. Collectively, a complex network of signaling pathways that function in modulating the activity of normal stem cells can be susceptible to deregulation as a result of aberrant epigenetic modifications during the course of tumor formation. These abnormal alterations in key signaling pathways contribute to CSC proliferation and maintenance, as well as tumor progression and invasion. Hence, epigenetic regulation of these signalling pathways may serve as potential mechanisms for targeted therapy against CSCs.

Therapeutic intervention using epigenetic modifying drugs

As epigenetic mechanisms have important functions in modulating stem cell properties in cancer cells, targeting components of these epigenetic pathways would help in eradicating both CSCs and the bulk tumor population. Inhibitors of epigenetic modulatory enzymes such as HDACs and DNMTs have been widely studied and many are currently in clinical trials for treatment of a variety of cancers. In addition, deregulation of chromatin remodelling has been associated with tumorigenesis and tumor progression, thus making chromatin remodelling proteins viable targets for small molecule inhibitors as well. Indeed, many of these therapeutic strategies aim to induce differentiation of CSCs and to sensitise these cells to chemotherapy, with the ultimate goal of reducing tumor relapse and improving patient survival. Here, we review the development of various epigenetic therapies designed to target different components of the epigenetic machinery. A summary of these epigenetic drugs and their clinical status can be found in Table 1.
Table 1
Epigenetic modulators in cancer
Drug
Target
Clinical status
Indication
References
DNMT inhibitors
Azacitidine
Inhibit DNMT (act as nucleoside analog)
FDA-approved
MDS
[277, 278]
Decitabine
Inhibit DNMT (act as nucleoside analog)
FDA-approved
MDS
[115, 277]
SGI-110
Inhibit DMNT by incorporating into guanine nucleotide
Phase 3
(NCT02348489
Phase 1/2
(NCT01261312, NCT02197676)
Phase 2
(NCT01752933)
Phase 1/2
(NCT01696032)
AML
MDS, AML
Advanced HCC
Platinum-resistant recurrent ovarian cancer
[279281]
HDAC inhibitors
Vorinostat
Inhibitor of Class I and II HDACs
FDA-approved
Cutaneous T cell lymphoma
[135]
Romidepsin
Inhibitor of Class I HDACs
FDA-approved
Cutaneous T cell lymphoma
[136]
Panobinostat
Pan-HDAC inhibitor
Phase 3
(NCT01034163)
Phase 2/3
(NCT00425555)
Hodgkin’s lymphoma
Cutaneous T cell lymphoma
[282, 283]
Entinostat
Inhibitor of Class I HDACs
Phase 2 (NCT00866333)
Phase 1/2 (NCT01038778)
Hodgkin’s lymphoma
Clear cell renal cell carcinoma, metastatic renal cell cancer
[284]
Belinostat
Inhibitor of Class I and II HDACs
Phase 2
(NCT00357032)
Phase 2
(NCT00274651)
Phase 2
(NCT00301756)
Relapsed/refractory AML or older patients with newly diagnosed AML
Recurrent/refractory cutaneous and peripheral T cell lymphomas
Ovarian cancer
[285287]
Pracinostat
Inhibitor of Class I and II HDACs
Phase 2
(NCT01112384, NCT01075308)
Translocation-associated recurrent/metastatic sarcomas, metastatic prostate cancer
[288, 289]
Givinostat
Inhibitor of Class I and II HDACs
Phase 2
(NCT01761968)
Chronic myeloproliferative neoplasms
[290]
Valproic acid
Inhibitor of Class I and II HDACs
Phase 2
(NCT01900730)
Breast cancer
[163, 291]
HMT inhibitors
EPZ-5676
Inhibit DOT1L methyltransferase (H3K79) activity by competing with SAM
Phase 1
(NCT02141828, NCT01684150)
MLL-fusion leukemia, AML, acute lymphocytic/lymphoblastic leukemia, MDS, myeloproliferative disorders
[182]
DZNep
Inhibit HMT activity of EZH2 via inhibiting S-adenosylhomocysteine (SAH) hydrolase
Not in trial
Breast cancer,
prostate cancer, glioblastoma multiforme (GBM)
[193, 292, 293]
E7438 (EPZ-6438)
Inhibit HMT activity of EZH2 by competing with co-factor S-adenosyl-methionine (SAM)
Phase 2
(NCT02860286, NCT02601950)
Malignant mesothelioma, rhabdoid tumors, synovial sarcoma, epitheloid sarcoma
[196]
GSK2816126 (GSK126)
Inhibit HMT activity of EZH2 by competing with co-factor S-adenosyl-methionine (SAM)
Phase 1
(NCT02082977)
Relapsed/refractory DLBCL, transformed follicular lymphoma, multiple myeloma, non-Hodgkin’s lymphoma, solid tumors
[198, 199]
CPI-1205
Inhibit HMT activity of EZH2 by competing with co-factor S-adenosyl-methionine (SAM)
Phase 1
(NCT02395601)
B-cell lymphoma
[294]
Chaetocin
Inhibit SUV39H1
Not in trial
HCC, multiple myeloma
[216, 218]
BIX01294
Inhibit G9a (substrate-competitive)
Not in trial
Breast cancer, colon cancer
[209, 212]
UNC0638
Inhibit G9a (substrate-competitive)
Preclinical
Breast cancer
[213]
UNC0642
Inhibit G9a (substrate-competitive)
Preclinical
Pancreatic cancer
[214]
HDM inhibitors
Tranylcypromine
Irreversible inhibitor of LSD1
Phase 1
(NCT02273102, NCT02717884)
Phase1/2
(NCT02261779)
AML, MDS
AML
[226]
ORY-1001
Irreversible inhibitor of LSD1
Phase 1
AML
[227]
GSK2879552
Irreversible inhibition of LSD1 activity by modifying its cofactor FAD
Phase 1
(NCT02034123, NCT02177812)
Small cell lung cancer,
AML
[231]
GSK-J4
Inhibit UTX
Not in trial
T-cell acute lymphoblastic leukemia (T-ALL)
[295]
BET inhibitors
I-BET762 (GSK525762A)
Interfere with binding of BET proteins to acetylated histones
Phase 1
(NCT01587703, NCT01943851)
Solid tumors, relapsed refractory haematological malignancies
[243, 244]
JQ1
Interfere with binding of BET proteins to acetylated histones (greatest specificity for BRD3 and BRD4)
Not in trial
NUT midline carcinoma (NMC), multiple myeloma, AML, Burkitt’s lymphoma, DLBCL
[239, 240, 296, 297]
I-BET151 (GSK1210151A)
Interfere with binding of BET proteins to acetylated histones
Not in trial
MLL fusion leukemia, medulloblastoma
[298, 299]
OTX015
Interfere with binding of BET proteins to acetylated histones
Phase 1
(NCT01713582)
Hematological malignancies
[248, 297]
CPI-203
Interfere with binding of BET proteins to acetylated histones
Not in trial
Lymphoma
[300]
CPI-0610
Interfere with binding of BET proteins to acetylated histones
Phase 1
(NCT02157636, NCT01949883, NCT02158858)
Acute leukaemia, MDS, myelodysplastic/myeloproliferative neoplasms, lymphoma, multiple myeloma
[301]

Targeting DNA methylation - DNA methyltransferase (DNMT) inhibitors

Inhibitors of DNA methylation were among the first epigenetic drugs tested for use in treatment of cancer [112]. The most widely studied DNMT inhibitors include azacitidine (5-azacitidine) and decitabine (5-aza-2’-deoxycytidine), which act as analogues of cytosine. These molecules get incorporated into DNA and covalently bond with DNA methyltransferase, thus preventing its function [113, 114] and leading to its degradation [115]. These drugs were initially used as cytotoxic chemotherapeutics in the late 1960s [116], but were found to be highly toxic [117120]. Subsequently, recent studies have discovered that low doses of DNMT inhibitors had greater efficacy in sustaining decreased DNA methylation and associated re-expression of silenced genes in leukemic and epithelial tumor cells [121]. These lower doses were also able to reduce tumorigenicity and target CSC populations within the tumor. In lung cancer, Liu et al. showed that inhibition of DNMT1 was able to decrease proliferation and tumorigenic ability of lung CSCs [34].
Multiple studies have also demonstrated the role of DNMT inhibitors in differentiation therapy. Pinto et al. showed that azacitidine could induce primary AML cells from patients to differentiate into less or non-malignant cells [122, 123]. Prostate cancer derived-CSCs that were treated with decitabine showed decreased expression of stemness genes Octamer-binding transcription factor 4 (OCT40029 and Nanog homeobox (NANOG), leading to overall reduction in tumor growth [124]. In addition, low doses of SGI-110, a newer DNMT inhibitor, was recently reported to be capable of reprogramming ovarian CSCs to a more differentiated state [125]. Treatment with SGI-110 also decreased tumor-initiating ability and re-sensitized these cells to platinum, suggesting a potential use of DNMT inhibitors in combination with other chemotherapeutic agents in preventing recurrence of ovarian cancer [125]. Both azacitidine and decitabine have been approved by the FDA for treatment of myelodysplastic syndrome (MDS) [126]. Clinical trials for other indications such as AML and colorectal cancer are still ongoing. SGI-110 is also in phases of clinical trials for treatment of various cancers such as AML, MDS, liver cancer and platinum-resistant ovarian cancer.

Targeting histone deacetylation - Histone deacetylase(HDAC) inhibitors

An important histone tail modification is acetylation, which is regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs are responsible for adding an acetyl group onto lysine residues of histone tail, which neutralizes the positive charge, resulting in a more “open” chromatin structure [127]. In contrast, HDACs remove the additional acetyl group, leading to increased binding affinity between DNA and histones, which is generally associated with gene repression [128]. Very often, deregulated gene silencing in cancers has been associated with aberrant histone deacetylation. For instance, in leukemia, this can be mediated through aberrant recruitment of HDACs by fusion proteins such as Acute myeloid leukemia protein 1 Eight twenty-one protein (AML1-ETO) and Promyelocytic leukemia protein retinoic acid receptor alpha (PML-RARα), which leads to abnormal gene silencing and subsequent leukemogenesis [129, 130]. Besides, HDACs can also acetylate non-histone proteins, including tumor suppressor p53 and oncogene B-cell lymphoma 2 (BCL2), resulting in inhibition of p53-dependent transcription [131] and upregulation of pro-survival protein, BCL2 [132]. Hence, the use of HDAC inhibitors in returning histone acetylation patterns to a normal state has been found to be effective in inducing apoptosis and differentiation as well as inhibit proliferation of tumor cells [129, 133]. These HDAC inhibitors can be divided mainly into two classes—the pan HDAC inhibitors and the class-specific inhibitors [134], and they all function via chelating the zinc atom in the active site of the enzyme [127].
Two HDAC inhibitors, vorinostat (subseroylanilide hydroxamic acid) and romidepsin (depsipeptide), have been approved for treatment of cutaneous T-cell lymphoma [135, 136]. Both drugs were found to produce durable response and efficacy in patients with cutaneous T-cell lymphoma in Phase 2 multi-center trials [135138]. However, besides cutaneous T-cell lymphoma, monotherapy of vorinostat and romidepsin in treatment of various solid tumors have had little success in clinical trials [139150]. Apart from these two compounds, many other HDAC inhibitors have also been developed and tested in clinical trials, the details of which have been well-reviewed elsewhere [115, 151153]. Monotherapies of these compounds, including panobinostat [154, 155], entinostat [156, 157], belinostat [158, 159] and pracinostat (SB939) [160], are being tested against various haematological malignancies and solid tumors.
Another mechanism of action of HDAC inhibitors for cancer treatment is via differentiation or reprogramming of cancer cells. As therapy resistance is a major hurdle in cancer treatment and is often associated with CSCs and epigenetic control [161], HDAC inhibitors possess the ability to induce differentiation of CSCs from their quiescent state, thereby re-sensitising them to other chemotherapy agents. Valproic acid, an antiepileptic drug, has been found to be a powerful HDAC inhibitor [162]. Gottlicher et al. demonstrated that valproic acid could trigger differentiation of transformed hematopoietic progenitor cells and leukemic blasts from AML patients [162]. Furthermore, Travaglini et al. found that valproic acid was able to epigenetically reprogram breast cancer cells into a more “physiologic” phenotype, thus improving sensitivity to other forms of breast cancer therapy [163]. In addition, entinostat, a selective inhibitor of class I HDACs, was recently reported to reverse EMT phenotype and decrease the population of tumor-initiating cells in triple-negative breast cancer (TNBC) [164]. These tumor-initiating cells possessed CSC properties and were responsible for driving metastasis and drug resistance in TNBC, thus contributing to poor patient prognosis. Hence, this study demonstrated the utility of HDAC inhibitors in preventing CSC invasiveness and tumor metastasis. Overall, these studies demonstrate the potential use of epigenetic modulators towards the differentiation and therapeutic sensitization of CSCs.

Targeting histone methylation – Histone methyltransferase (HMT) inhibitors

A class of enzymes called histone lysine methyltransferases (HKMTs) mediate the addition of a methyl group to the nitrogen atom of the lysine side chain [165]. Despite catalysing a common chemical reaction, this family of HKMTs demonstrate large structural diversity of its active sites, allowing these enzymes to have high substrate specificity [127]. For example, DOT1L (KMT4) is a unique HKMT as it is currently the only known enzyme that methylates lysine 79 of histone H3 (H3K79) [166]. Similarly, methylation of H3K27 is only mediated by the catalytic subunit EZH2 (KMT6) of PRC2 [127]. In contrast, some methylation marks can be catalysed by several proteins, such as H3K9 methylation. These post-translational methylation of histones have important roles in regulation of gene expression, differentiation, DNA damage repair as well as in tumorigenesis [167, 168]. Aberrant histone methylation can be due to gene mutations, over-expression or deregulated control of epigenetic modulatory enzymes involved. Thus, HKMTs are potential therapeutic targets, and the structural differences between members of the family also enable greater selectivity in inhibition of these proteins by small molecule compounds [169].
HKMT inhibitors have only recently gained more attention as cancer therapeutics, resulting in a rapidly increasing number of these small molecule inhibitors being developed [170172]. In fact, several DOT1 like histone H3K79 methyltransferase (DOT1L) and EZH2 inhibitors have progressed to being tested in clinical trials as cancer interventions [173]. H3K79 methylation by DOT1L is associated with transcriptional activation of genes under its regulation [174, 175], and overexpression or aberrant DOT1L activity has been found in cancer, such as leukemia with mixed lineage leukemia (MLL) gene translocation. The MLL fusion protein can recruit DOT1L into a transcription complex, which subsequently methylates H3K79 [176180]. This leads to dysregulation and overexpression of many MLL-target genes, including Homeobox A9 (HoxA9) and Meis homeobox 1 (Meis1), which are key regulators of hematopoietic stem cell differentiation that contributes to leukemogenesis [165]. Therefore, DOT1L is an attractive target for therapy, resulting in the first selective DOT1L inhibitor EPZ-4777 to be synthesised with anti-tumor effects against murine models of MLL-rearranged leukemia [181]. Further optimisation of the drug led to the development of EPZ-5676, the first HKMT inhibitor to enter clinical trials. This compound has been shown to be highly potent and selective for DOT1L. Treatment with EPZ-5676 in a MLL-rearranged leukemia xenograft model showed durable and complete tumor regression [182]. EPZ-5676 is currently under clinical studies (Phase I) for MLL-fusion leukemia, AML, MDS and myeloproliferative disorders.
EZH2 is a member of PRC2, along with proteins embryonic ectoderm development protein (EED) and SUZ12, and is responsible for catalysing H3K27 mono-, di- and tri-methylation [183185]. Overexpression of EZH2 has been found in various cancers of the breast, lung, prostate and haematological malignancies [186191], and is associated with poor disease prognosis. Studies have also shown the role of EZH2 deregulation in tumor progression, metastasis [192, 193] and maintenance of CSC self-renewal properties [194]. In glioblastoma multiforme (GBM), inhibition of EZH2 by S-adenosylhomocysteine hydrolase (SAH) inhibitor 3-deazaneplanocin A (DZNep) was able to reduce self-renewal and tumor-initiating capabilities of GBM CSCs in vivo via affecting transcriptional regulation of oncogene MYC [193]. However, DZNep affects methylation of other histone residues [195], leading to the development of more specific EZH2 inhibitors. The earliest SAM-competitive and selective EZH2 inhibitor to advance into clinical trials for treatment of rhabdoid tumors and lymphomas is EPZ-6438 (E7438) [196, 197]. A more recent drug, GSK2816126 (GSK126) has also entered clinical studies for relapsed/refractory diffuse large B-cell lymphoma (DLBCL), multiple myeloma and transformed follicular lymphoma [198, 199]. Both drugs have shown high potency and selectivity in inhibiting tumor growth in preclinical studies [197, 198, 200].
H3K9 methyltransferases, such as euchromatic histone lysine methyltransferase 2 (G9a/EHMT2) and euchromatic histone lysine methyltransferase 1 (GLP/EHMT1), catalyse mono- and di-methylation of the lysine residue, while tri-methylation of H3K9 is mediated by Suppressor of variegation 3–9 homolog 1 (SUV39H1) and Suppressor of variegation 3–9 homolog 2 (SUV39H2) [201]. Upregulation of G9a activity has been linked to several types of cancer, including ovarian, lung, liver and bladder cancers [202208]. Hence, several substrate-competitive inhibitors of these HKMTs have been developed. BIX-01294 is the first specific inhibitor of G9a and GLP, and studies have reported its ability to decrease H3K9me2 levels in mammalian cells [209211]. Kim et al. reported that BIX-01294 was able to induce cell death in colon and breast cancer cells via EHMT dysfunction [212]. However, due to the increased toxicity levels of BIX-01294 at higher concentrations, the use of this drug is limited. This led to the recent development of a more potent, specific and selective EHMT inhibitor, UNC0638 that was found to decrease local H3K9me2 and DNA methylation levels [213]. Further development generated UNC0642, which possessed better pharmacokinetic properties and higher efficacy in inhibiting colony formation ability of pancreatic adenocarcinoma cells [214].
Methylation of H3K9 by SUV39H1 is associated with silencing of tumor suppressor genes, including E-cadherin and p15INK4B, in AML [215]. Overexpression of SUV39H1 has also been correlated with poor prognosis in multiple myeloma patients [216]. Treatment of multiple myeloma cells with chaetocin, a small molecule inhibitor of SUV39H1 showed anti-tumor effects at low doses of the drug [216]. Similarly, chaetocin was found to decrease H3K9me3 levels and induce differentiation of AML cells at non-toxic doses [217]. Furthermore, chaetocin was able to repress cell proliferation and induce apoptosis in hepatocellular carcinoma (HCC) cultures and xenografts [218], implying a potential tumorigenic role of EHMTs in HCC progression and development.

Targeting histone demethylation - Histone demethylase (HDM) inhibitors

Methylation of lysine on histones is also regulated by histone lysine demethylases (KDMs). This group of epigenetic erasers function in removing the methyl groups from lysine side chains on histones [219, 220]. As proper functioning of both HKMTs and KDMs is required to maintain stable histone methylation levels, small molecule inhibitors have also been developed to target KDMs. KDMs can be grouped into two families - the lysine-specific demethylase (LSD) family and Jumonji domain-containing (JmjC) family [221]. The LSD family are flavin adenine dinucleotide (FAD)-dependent amine oxidase that demethylates mono- and di-methyl lysine residues, while JmjC enzymes utilise 2-oxoglutarate and iron to oxidatively release methyl groups from all three methylation states at lysine residues [172, 222].
Upregulated expression of LSD1 (KDM1A) has been found in various human cancers, including AML, ovarian, lung, bladder and colorectal cancers [223225]. Hence, small molecule inhibitors of LSD1 that target the enzyme cofactor FAD have been developed, the first of which is tranylcypromine [226]. Further studies have led to the synthesis of more selective derivatives of tranylcypromine, such as ORY-1001 [227] and GSK2879552 [228]. They function by irreversibly changing FAD, leading to the formation of a tetracyclic adduct [229]. LSD1 is important for normal hematopoiesis; loss of LSD1 has been found to inhibit differentiation and impair hematopoiesis [230]. This suggests a potential role of aberrant LSD1 activity in affecting stemness properties in tumor cells. The inhibitor ORY-1001 has been shown to decrease the population of AML stem cells and improve survival of mice with acute lymphoblastic leukemia (ALL) in preclinical studies [227, 228]. GSK2879552 has also been found to influence differentiation in small cell lung cancer (SCLC) [231]. These compounds are currently in phase 1 studies for relapsed or refractory AML (ORY-1001) and SCLC (GSK2879552).
Similarly, JmjC demethylases are amenable to pharmacological intervention as well. Ubiquitously transcribed tetratricopeptide repeat X chromosome (UTX), also known as KDM6A, is responsible for demethylating H3K27 [232234], and loss of UTX activity has been found in multiple human malignancies, including multiple myeloma, esophageal squamous cell carcinoma and renal carcinoma [166]. However, no inhibitors of JmjC enzymes have advanced beyond biochemical studies [127]. Nevertheless, as UTX is a component of the mixed lineage leukemia protein 2 (MLL2) H3K4 methyltransferase complex, and interacts with SWI/SNF chromatin remodelling complex [235237], it is still an important epigenetic target and its role in epigenetic modulation still warrants further study.

Targeting epigenetic readers – BET inhibitors

While epigenetic modulatory enzymes are obvious targets for therapy, epigenetic readers are also important components of the epigenetic machinery as they directly or indirectly regulate gene expression. One such group of readers called bromodomain and extra-terminal (BET) proteins modulate gene expression by recognising acetylated histones. Increased BET activities have been associated with NUT midline carcinoma (NMC), glioblastoma and various haematological malignancies, through aberrant transcription of disease-associated genes and oncogenes such as MYC [238]. Hence, BET proteins appear to be attractive therapeutic targets for controlling dysregulated gene expression.
JQ1 is a selective BET inhibitor of BRD family of proteins, including Bromodomain-containing protein 4 (BRD4) [239]. In preclinical studies, JQ1 was able to cause tumor regression in NMC mouse models, inhibit proliferation, induce apoptosis and differentiation in cancer cells [239242]. Another BET inhibitor, I-BET762 (GSK525762A), functions by binding to the acetyl-binding pocket of BET proteins [243, 244]. Studies have shown that I-BET762 treatment was able to induce terminal differentiation of patient-derived malignant cells [245] and activate apoptosis in neuroblastoma and prostate cancer via inhibition of Myc-driven pathways [246, 247]. This compound is currently in phase I trials for solid tumors and relapsed or refractory haematological cancers.
OTX015 is another BET inhibitor that has progressed into clinical trials for various haematological malignancies. This compound has been found to possess anti-proliferative effects via directly influencing MYC expression and activity [248, 249]. Similarly, CPI-0610 has also entered clinical testing for lymphoma, multiple myeloma and myelodysplastic or myeloproliferative neoplasms. I-BET151 is a pan-BET inhibitor, similar to JQ1, and has been found to block proliferation and induce apoptosis in myeloma cells via repressing Myc activity [250]. Antitumor effects have also been observed in NMC, MLL, ALL, lung cancer and brain cancer [238].

Combination therapy with epigenetic modulators

While epigenetic drugs have been tested preclinically and clinically as single agents, further studies have revealed the increased efficacy of these drugs when used in combination with other therapies. One common combination of different epigenetic therapies is that of DNMT and HDAC inhibitors. Pathania et al. reported that combining azacitidine (DNMT inhibitor) and butyrate (HDAC inhibitor) was capable of significantly decreasing breast cancer CSC population [251]. In addition, combination of azacitidine and HDAC inhibitor entinostat at low doses in a phase I/II clinical trial showed sustained and favourable responses in treatment-resistant non-small cell lung cancer (NSCLC) patients [252]. Azacitidine and valproic acid co-treatment was also able to promote tumor regression in Patched mutant mouse models of medulloblastoma [253]. Besides DNMT-HDAC inhibitor combination therapy, studies have demonstrated synergistic effects of other epigenetic drug combinations. For example, inhibiting both EZH2 and G9a histone methyltransferases showed greater efficacy in blocking cell proliferation as compared to single drug treatment [254]. Furthermore, the DOT1L inhibitor EPZ-5676 could interact synergistically with DNA hypomethylating agents, such as azacitidine and decitabine, in MLL-rearranged leukemia cells [255].
In recent years, an increasing number of studies have reported the use of epigenetic drugs in combination with conventional chemotherapeutics, with underlying mechanisms of re-sensitising resistant CSCs to drug treatment, or to prime cancer cells for subsequent therapies [134, 256]. For example, low doses of SGI-110 (DNMT inhibitor) was found to drive ovarian CSCs towards a more differentiated phenotype and sensitise them to platinum treatment [125]. DOT1L inhibitor EPZ-5676 was also able to establish a chromatin state that enhanced the anti-tumor effects of cytarabine or daunorubicin in MLL-rearranged leukemia [255]. Moreover, pre-treatment with azacitidine was demonstrated to prime colon cancer cell lines to irinotecan therapy [257]. Indeed, various combinations have been tested in clinical trials with promising results on drug response and anti-tumor efficacy [258261]. In addition to drug combination synergy, the method of delivery could also improve response to therapy. A recent paper by Li et al. showed that encapsulating decitabine and doxorubicin in nanoparticles was able to better target breast CSCs and inhibit tumor growth [262].
The use of immunotherapy in cancer has made significant progress over the past two decades, with several immunotherapy drugs being approved by the FDA for the treatment of cancer. These drugs function to overcome the mechanisms of immune tolerance that are employed by cancer cells to evade or limit the body’s immune response. These mechanisms include changes in antigen processing and presentation, creation of an immunosuppressive microenvironment, induction of T-cell death and activation of negative immune regulatory pathways [263]. One key receptor involved in the immunoinhibitory pathways is the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), which is expressed on the surface of immune cells and acts as an immune checkpoint. Studies have shown that targeting CTLA-4 receptor induced favourable responses in patients with advanced melanoma [264], and the FDA-approved CTLA-4 inhibitor, Ipilimumab, is now in clinical trials for prostate and lung cancers. Another immune checkpoint involved in tumor immune-resistance is the interaction between programmed cell death-1 (PD-1) and programmed death-ligand 1 (PD-L1) [265]. Specific targeting of PD-1 and PD-L1 has been clinically shown to be very effective in treatment of metastatic cancers and melanomas [266, 267].
However, as most of these immunotherapy strategies are mainly targeted at bulk tumors, which contain more differentiated cells with “differentiation antigens” [268], CSCs (which have a different set of tumor antigens) would not be successfully eradicated. Hence, more effective targeting of the CSC population can be achieved via CSC-specific immunologic approaches, or by combining immunotherapy with epigenetic therapies that induce CSC differentiation and alter surface protein expression. The latter approach would likely improve the overall antitumor efficacy as both CSC and bulk tumor populations can be targeted simultaneously. For instance, the use of DNA hypomethylating agent (5-aza-2-deoxycytidine) in combination with anti-CTLA-4 monoclonal antibody in syngeneic transplantable murine models demonstrated significant reduction in tumor volumes as compared to single agent treatment alone [269]. The improved efficacy of this combination was attributed to the increased CD3+ T-cell infiltration in the combination cohort tumors and a sustained expression of cancer antigens and MHC proteins due to promoter demethylation. Furthermore, combinatorial drug treatment with immune checkpoint inhibitors (anti-CTLA-4 and anti-PD-1) and epigenetic modulators (5-azacytidine and Entinostat) showed remarkable eradication of CT26 colorectal tumors and 4 T1 mammary tumors in more than 80% of the tumor-bearing mice [270]. Importantly, 4 T1 tumor-bearing mice that were given combinatorial treatment did not develop metastases as compared to mice given single agent treatment. These findings demonstrate that epigenetic drugs in combination with immunotherapy can enhance the reversal of immune tolerance in cancer cells, including CSCs.
Another way in which cancer cells evade cytotoxic T-cells is by down-regulating human leukocyte antigen (HLA) to avoid tumor antigen presentation [134]. Hypermethylation of HLA promoters was frequently observed in gastric cancer and esophageal squamous cell cancers [271, 272]. Treatment with DNMT and HDAC inhibitors were found to be capable of reversing this hypermethylation and increasing HLA expression [272275], thus priming these cells for immunotherapy. In addition, Li et al. showed that azacitidine treatment was able to enhance immunomodulatory pathways, such as antigen processing/presentation and interferon signaling, in breast, colorectal and ovarian cancers [276]. These preclinical data highlight the promising potential of combining epigenetic and immunotherapies in improving cancer treatment efficacy, which will be verified in several ongoing clinical trials.

Conclusion

Our understanding of cancer has changed over the last decade with the advances in sequencing technologies and the deciphering of the human genome. It is now clear to us that the tumor genome is complex and heterogeneous and that tumors do not arise from a single clone with a single tumor genome. We have discussed several important aspects and examples of how epigenetic deregulation may drive or promote tumorigenesis and metastasis by alteration of key transcriptomic programs and signaling pathways, especially in CSCs. More importantly, we have provided several evidences that these epigenetic modifiers are targetable and many of these epigenetic modulating drugs have entered clinical trials, and some including azacitidine, decitabine, vorinostat and romidepsin have been approved for various indications by the FDA. We believe that the success of these epigenetic therapeutic trials will provide a promising path to follow.

Acknowledgements

We apologize for not reviewing numerous original and pertinent articles due to space limitations and the scope of this review. We would also like to thank Mr Wong Chun Xi (Mechanobiology Institute, Singapore) for his help in illustrating the figure in this manuscript.

Funding

The authors are supported by the National Research Foundation Cancer Science Institute of Singapore RCE Main Grant, Ministry of Education Academic Research Fund (MOE AcRF Tier 2 (MOE2015-T2-2-126, Seed Fund Grant T1-BSRG 2014–05). This work is funded by the NCIS Yong Siew Yoon Research Grant through donations from the Yong Loo Lin Trust.

Availability of data and materials

Not applicable.

Authors’ Contributions

TBT, JJL and EKC were major contributors in the writing of the manuscript. TBT and EKC designed the manuscript. EKC revised and edited the manuscript. All authors read and approved the final manuscript.

Competing Interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.PubMedCrossRef Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.PubMedCrossRef
2.
Zurück zum Zitat Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–7.PubMedCrossRef Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–7.PubMedCrossRef
3.
Zurück zum Zitat Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645–8.PubMedCrossRef Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645–8.PubMedCrossRef
4.
Zurück zum Zitat Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111–5.PubMedCrossRef Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111–5.PubMedCrossRef
5.
Zurück zum Zitat Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401.PubMedCrossRef Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401.PubMedCrossRef
6.
Zurück zum Zitat Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, et al. Identification of cells initiating human melanomas. Nature. 2008;451:345–9.PubMedPubMedCentralCrossRef Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, et al. Identification of cells initiating human melanomas. Nature. 2008;451:345–9.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–8.PubMedPubMedCentralCrossRef Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–8.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH, Nephew KP. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008;68:4311–20.PubMedPubMedCentralCrossRef Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH, Nephew KP. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008;68:4311–20.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Ma S, Chan KW, Hu L, Lee TK, Wo JY, Ng IO, Zheng BJ, Guan XY. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology. 2007;132:2542–56.PubMedCrossRef Ma S, Chan KW, Hu L, Lee TK, Wo JY, Ng IO, Zheng BJ, Guan XY. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology. 2007;132:2542–56.PubMedCrossRef
10.
Zurück zum Zitat Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005;65:10946–51.PubMedCrossRef Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005;65:10946–51.PubMedCrossRef
11.
Zurück zum Zitat Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15:504–14.PubMedCrossRef Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15:504–14.PubMedCrossRef
12.
Zurück zum Zitat Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, Aigner L, Brawanski A, Bogdahn U, Beier CP. CD133(+) and CD133(−) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007;67:4010–5.PubMedCrossRef Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, Aigner L, Brawanski A, Bogdahn U, Beier CP. CD133(+) and CD133(−) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007;67:4010–5.PubMedCrossRef
13.
Zurück zum Zitat Kemper K, Sprick MR, de Bree M, Scopelliti A, Vermeulen L, Hoek M, Zeilstra J, Pals ST, Mehmet H, Stassi G, Medema JP. The AC133 epitope, but not the CD133 protein, is lost upon cancer stem cell differentiation. Cancer Res. 2010;70:719–29.PubMedCrossRef Kemper K, Sprick MR, de Bree M, Scopelliti A, Vermeulen L, Hoek M, Zeilstra J, Pals ST, Mehmet H, Stassi G, Medema JP. The AC133 epitope, but not the CD133 protein, is lost upon cancer stem cell differentiation. Cancer Res. 2010;70:719–29.PubMedCrossRef
14.
Zurück zum Zitat Zhou F, Cui C, Ge Y, Chen H, Li Q, Yang Z, Wu G, Sun S, Chen K, Gu J, et al. Alpha2,3-Sialylation regulates the stability of stem cell marker CD133. J Biochem. 2010;148:273–80.PubMedCrossRef Zhou F, Cui C, Ge Y, Chen H, Li Q, Yang Z, Wu G, Sun S, Chen K, Gu J, et al. Alpha2,3-Sialylation regulates the stability of stem cell marker CD133. J Biochem. 2010;148:273–80.PubMedCrossRef
15.
Zurück zum Zitat Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, Jia H, Ye Q, Qin LX, Wauthier E, et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009;136:1012–24.PubMedCrossRef Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, Jia H, Ye Q, Qin LX, Wauthier E, et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009;136:1012–24.PubMedCrossRef
16.
Zurück zum Zitat Wilson GS, Hu Z, Duan W, Tian A, Wang XM, McLeod D, Lam V, George J, Qiao L. Efficacy of using cancer stem cell markers in isolating and characterizing liver cancer stem cells. Stem Cells Dev. 2013;22:2655–64.PubMedPubMedCentralCrossRef Wilson GS, Hu Z, Duan W, Tian A, Wang XM, McLeod D, Lam V, George J, Qiao L. Efficacy of using cancer stem cell markers in isolating and characterizing liver cancer stem cells. Stem Cells Dev. 2013;22:2655–64.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Chow EK, Fan LL, Chen X, Bishop JM. Oncogene-specific formation of chemoresistant murine hepatic cancer stem cells. Hepatology. 2012;56:1331–41.PubMedPubMedCentralCrossRef Chow EK, Fan LL, Chen X, Bishop JM. Oncogene-specific formation of chemoresistant murine hepatic cancer stem cells. Hepatology. 2012;56:1331–41.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–8.PubMedPubMedCentralCrossRef Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–8.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, DiMeco F, Olivi A, Eberhart CG. Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells. 2007;25:2524–33.PubMedPubMedCentralCrossRef Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, DiMeco F, Olivi A, Eberhart CG. Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells. 2007;25:2524–33.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Mahesparan R, Molthoff C, Sminia P, Sundlisaeter E, Misra A, Tysnes BB, et al. Angiogenesis-independent tumor growth mediated by stem-like cancer cells. Proc Natl Acad Sci U S A. 2006;103:16466–71.PubMedPubMedCentralCrossRef Sakariassen PO, Prestegarden L, Wang J, Skaftnesmo KO, Mahesparan R, Molthoff C, Sminia P, Sundlisaeter E, Misra A, Tysnes BB, et al. Angiogenesis-independent tumor growth mediated by stem-like cancer cells. Proc Natl Acad Sci U S A. 2006;103:16466–71.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Lim E, Vaillant F, Wu D, Forrest NC, Pal B, Hart AH, Asselin-Labat ML, Gyorki DE, Ward T, Partanen A, et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med. 2009;15:907–13.PubMedCrossRef Lim E, Vaillant F, Wu D, Forrest NC, Pal B, Hart AH, Asselin-Labat ML, Gyorki DE, Ward T, Partanen A, et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med. 2009;15:907–13.PubMedCrossRef
22.
Zurück zum Zitat Molyneux G, Geyer FC, Magnay FA, McCarthy A, Kendrick H, Natrajan R, Mackay A, Grigoriadis A, Tutt A, Ashworth A, et al. BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell. 2010;7:403–17.PubMedCrossRef Molyneux G, Geyer FC, Magnay FA, McCarthy A, Kendrick H, Natrajan R, Mackay A, Grigoriadis A, Tutt A, Ashworth A, et al. BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell. 2010;7:403–17.PubMedCrossRef
23.
Zurück zum Zitat Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med. 2011;17:1086–93.PubMedCrossRef Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P, Metzeler KH, Poeppl A, Ling V, Beyene J, et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med. 2011;17:1086–93.PubMedCrossRef
24.
Zurück zum Zitat Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006;313:1929–35.PubMedCrossRef Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006;313:1929–35.PubMedCrossRef
25.
Zurück zum Zitat Shats I, Gatza ML, Chang JT, Mori S, Wang J, Rich J, Nevins JR. Using a stem cell-based signature to guide therapeutic selection in cancer. Cancer Res. 2011;71:1772–80.PubMedCrossRef Shats I, Gatza ML, Chang JT, Mori S, Wang J, Rich J, Nevins JR. Using a stem cell-based signature to guide therapeutic selection in cancer. Cancer Res. 2011;71:1772–80.PubMedCrossRef
26.
Zurück zum Zitat Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447:425–32.PubMedCrossRef Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447:425–32.PubMedCrossRef
27.
Zurück zum Zitat Stallcup MR. Role of protein methylation in chromatin remodeling and transcriptional regulation. Oncogene. 2001;20:3014–20.PubMedCrossRef Stallcup MR. Role of protein methylation in chromatin remodeling and transcriptional regulation. Oncogene. 2001;20:3014–20.PubMedCrossRef
28.
29.
Zurück zum Zitat Wongtrakoongate P. Epigenetic therapy of cancer stem and progenitor cells by targeting DNA methylation machineries. World J Stem Cells. 2015;7:137–48.PubMedPubMedCentralCrossRef Wongtrakoongate P. Epigenetic therapy of cancer stem and progenitor cells by targeting DNA methylation machineries. World J Stem Cells. 2015;7:137–48.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet. 2007;16 Spec No 1:R50–59.PubMedCrossRef Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet. 2007;16 Spec No 1:R50–59.PubMedCrossRef
31.
Zurück zum Zitat Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, Mohammad HP, Chen W, Daniel VC, Yu W, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39:237–42.PubMedPubMedCentralCrossRef Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, Mohammad HP, Chen W, Daniel VC, Yu W, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39:237–42.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Broske AM, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, Kuhl C, Enns A, Prinz M, Jaenisch R, et al. DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet. 2009;41:1207–15.PubMedCrossRef Broske AM, Vockentanz L, Kharazi S, Huska MR, Mancini E, Scheller M, Kuhl C, Enns A, Prinz M, Jaenisch R, et al. DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat Genet. 2009;41:1207–15.PubMedCrossRef
34.
Zurück zum Zitat Liu CC, Lin JH, Hsu TW, Su K, Li AF, Hsu HS, Hung SC. IL-6 enriched lung cancer stem-like cell population by inhibition of cell cycle regulators via DNMT1 upregulation. Int J Cancer. 2015;136:547–59.PubMed Liu CC, Lin JH, Hsu TW, Su K, Li AF, Hsu HS, Hung SC. IL-6 enriched lung cancer stem-like cell population by inhibition of cell cycle regulators via DNMT1 upregulation. Int J Cancer. 2015;136:547–59.PubMed
35.
Zurück zum Zitat Morita R, Hirohashi Y, Suzuki H, Takahashi A, Tamura Y, Kanaseki T, Asanuma H, Inoda S, Kondo T, Hashino S, et al. DNA methyltransferase 1 is essential for initiation of the colon cancers. Exp Mol Pathol. 2013;94:322–9.PubMedCrossRef Morita R, Hirohashi Y, Suzuki H, Takahashi A, Tamura Y, Kanaseki T, Asanuma H, Inoda S, Kondo T, Hashino S, et al. DNA methyltransferase 1 is essential for initiation of the colon cancers. Exp Mol Pathol. 2013;94:322–9.PubMedCrossRef
36.
Zurück zum Zitat Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, Hein K, Vogt R, Kemler R. Wnt/beta-catenin signaling regulates telomerase in stem cells and cancer cells. Science. 2012;336:1549–54.PubMedCrossRef Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, Hein K, Vogt R, Kemler R. Wnt/beta-catenin signaling regulates telomerase in stem cells and cancer cells. Science. 2012;336:1549–54.PubMedCrossRef
37.
Zurück zum Zitat Myant KB, Cammareri P, McGhee EJ, Ridgway RA, Huels DJ, Cordero JB, Schwitalla S, Kalna G, Ogg EL, Athineos D, et al. ROS production and NF-kappaB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell. 2013;12:761–73.PubMedPubMedCentralCrossRef Myant KB, Cammareri P, McGhee EJ, Ridgway RA, Huels DJ, Cordero JB, Schwitalla S, Kalna G, Ogg EL, Athineos D, et al. ROS production and NF-kappaB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell. 2013;12:761–73.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov. 2006;5:997–1014.PubMedCrossRef Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov. 2006;5:997–1014.PubMedCrossRef
39.
Zurück zum Zitat Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14:1837–51.PubMed Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14:1837–51.PubMed
40.
Zurück zum Zitat Lindvall C, Bu W, Williams BO, Li Y. Wnt signaling, stem cells, and the cellular origin of breast cancer. Stem Cell Rev. 2007;3:157–68.PubMedCrossRef Lindvall C, Bu W, Williams BO, Li Y. Wnt signaling, stem cells, and the cellular origin of breast cancer. Stem Cell Rev. 2007;3:157–68.PubMedCrossRef
41.
Zurück zum Zitat Klarmann GJ, Decker A, Farrar WL. Epigenetic gene silencing in the Wnt pathway in breast cancer. Epigenetics. 2008;3:59–63.PubMedCrossRef Klarmann GJ, Decker A, Farrar WL. Epigenetic gene silencing in the Wnt pathway in breast cancer. Epigenetics. 2008;3:59–63.PubMedCrossRef
42.
Zurück zum Zitat Suzuki H, Watkins DN, Jair KW, Schuebel KE, Markowitz SD, Chen WD, Pretlow TP, Yang B, Akiyama Y, Van Engeland M, et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet. 2004;36:417–22.PubMedCrossRef Suzuki H, Watkins DN, Jair KW, Schuebel KE, Markowitz SD, Chen WD, Pretlow TP, Yang B, Akiyama Y, Van Engeland M, et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet. 2004;36:417–22.PubMedCrossRef
43.
Zurück zum Zitat Koinuma K, Yamashita Y, Liu W, Hatanaka H, Kurashina K, Wada T, Takada S, Kaneda R, Choi YL, Fujiwara SI, et al. Epigenetic silencing of AXIN2 in colorectal carcinoma with microsatellite instability. Oncogene. 2006;25:139–46.PubMed Koinuma K, Yamashita Y, Liu W, Hatanaka H, Kurashina K, Wada T, Takada S, Kaneda R, Choi YL, Fujiwara SI, et al. Epigenetic silencing of AXIN2 in colorectal carcinoma with microsatellite instability. Oncogene. 2006;25:139–46.PubMed
44.
Zurück zum Zitat Yoda Y, Takeshima H, Niwa T, Kim JG, Ando T, Kushima R, Sugiyama T, Katai H, Noshiro H, Ushijima T. Integrated analysis of cancer-related pathways affected by genetic and epigenetic alterations in gastric cancer. Gastric Cancer. 2015;18:65–76.PubMedCrossRef Yoda Y, Takeshima H, Niwa T, Kim JG, Ando T, Kushima R, Sugiyama T, Katai H, Noshiro H, Ushijima T. Integrated analysis of cancer-related pathways affected by genetic and epigenetic alterations in gastric cancer. Gastric Cancer. 2015;18:65–76.PubMedCrossRef
45.
Zurück zum Zitat Hussain M, Rao M, Humphries AE, Hong JA, Liu F, Yang M, Caragacianu D, Schrump DS. Tobacco smoke induces polycomb-mediated repression of Dickkopf-1 in lung cancer cells. Cancer Res. 2009;69:3570–8.PubMedCrossRef Hussain M, Rao M, Humphries AE, Hong JA, Liu F, Yang M, Caragacianu D, Schrump DS. Tobacco smoke induces polycomb-mediated repression of Dickkopf-1 in lung cancer cells. Cancer Res. 2009;69:3570–8.PubMedCrossRef
46.
Zurück zum Zitat Jiang X, Tan J, Li J, Kivimae S, Yang X, Zhuang L, Lee PL, Chan MT, Stanton LW, Liu ET, et al. DACT3 is an epigenetic regulator of Wnt/beta-catenin signaling in colorectal cancer and is a therapeutic target of histone modifications. Cancer Cell. 2008;13:529–41.PubMedPubMedCentralCrossRef Jiang X, Tan J, Li J, Kivimae S, Yang X, Zhuang L, Lee PL, Chan MT, Stanton LW, Liu ET, et al. DACT3 is an epigenetic regulator of Wnt/beta-catenin signaling in colorectal cancer and is a therapeutic target of histone modifications. Cancer Cell. 2008;13:529–41.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Rheinbay E, Suva ML, Gillespie SM, Wakimoto H, Patel AP, Shahid M, Oksuz O, Rabkin SD, Martuza RL, Rivera MN, et al. An aberrant transcription factor network essential for Wnt signaling and stem cell maintenance in glioblastoma. Cell Rep. 2013;3:1567–79.PubMedPubMedCentralCrossRef Rheinbay E, Suva ML, Gillespie SM, Wakimoto H, Patel AP, Shahid M, Oksuz O, Rabkin SD, Martuza RL, Rivera MN, et al. An aberrant transcription factor network essential for Wnt signaling and stem cell maintenance in glioblastoma. Cell Rep. 2013;3:1567–79.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Zhang N, Wei P, Gong A, Chiu WT, Lee HT, Colman H, Huang H, Xue J, Liu M, Wang Y, et al. FoxM1 promotes beta-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell. 2011;20:427–42.PubMedPubMedCentralCrossRef Zhang N, Wei P, Gong A, Chiu WT, Lee HT, Colman H, Huang H, Xue J, Liu M, Wang Y, et al. FoxM1 promotes beta-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell. 2011;20:427–42.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Zheng H, Ying H, Wiedemeyer R, Yan H, Quayle SN, Ivanova EV, Paik JH, Zhang H, Xiao Y, Perry SR, et al. PLAGL2 regulates Wnt signaling to impede differentiation in neural stem cells and gliomas. Cancer Cell. 2010;17:497–509.PubMedPubMedCentralCrossRef Zheng H, Ying H, Wiedemeyer R, Yan H, Quayle SN, Ivanova EV, Paik JH, Zhang H, Xiao Y, Perry SR, et al. PLAGL2 regulates Wnt signaling to impede differentiation in neural stem cells and gliomas. Cancer Cell. 2010;17:497–509.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, et al. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 2015;16:413–25.PubMedCrossRef Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, et al. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 2015;16:413–25.PubMedCrossRef
51.
Zurück zum Zitat Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15:3059–87.PubMedCrossRef Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15:3059–87.PubMedCrossRef
52.
Zurück zum Zitat Ma J, Meng Y, Kwiatkowski DJ, Chen X, Peng H, Sun Q, Zha X, Wang F, Wang Y, Jing Y, et al. Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade. J Clin Invest. 2010;120:103–14.PubMedCrossRef Ma J, Meng Y, Kwiatkowski DJ, Chen X, Peng H, Sun Q, Zha X, Wang F, Wang Y, Jing Y, et al. Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade. J Clin Invest. 2010;120:103–14.PubMedCrossRef
53.
Zurück zum Zitat Beachy PA, Karhadkar SS, Berman DM. Tissue repair and stem cell renewal in carcinogenesis. Nature. 2004;432:324–31.PubMedCrossRef Beachy PA, Karhadkar SS, Berman DM. Tissue repair and stem cell renewal in carcinogenesis. Nature. 2004;432:324–31.PubMedCrossRef
54.
Zurück zum Zitat Teglund S, Toftgard R. Hedgehog beyond medulloblastoma and basal cell carcinoma. Biochim Biophys Acta. 1805;2010:181–208. Teglund S, Toftgard R. Hedgehog beyond medulloblastoma and basal cell carcinoma. Biochim Biophys Acta. 1805;2010:181–208.
55.
Zurück zum Zitat Youssef KK, Van Keymeulen A, Lapouge G, Beck B, Michaux C, Achouri Y, Sotiropoulou PA, Blanpain C. Identification of the cell lineage at the origin of basal cell carcinoma. Nat Cell Biol. 2010;12:299–305.PubMed Youssef KK, Van Keymeulen A, Lapouge G, Beck B, Michaux C, Achouri Y, Sotiropoulou PA, Blanpain C. Identification of the cell lineage at the origin of basal cell carcinoma. Nat Cell Biol. 2010;12:299–305.PubMed
56.
Zurück zum Zitat Sellheyer K. Basal cell carcinoma: cell of origin, cancer stem cell hypothesis and stem cell markers. Br J Dermatol. 2011;164:696–711.PubMedCrossRef Sellheyer K. Basal cell carcinoma: cell of origin, cancer stem cell hypothesis and stem cell markers. Br J Dermatol. 2011;164:696–711.PubMedCrossRef
57.
Zurück zum Zitat Yang ZJ, Ellis T, Markant SL, Read TA, Kessler JD, Bourboulas M, Schuller U, Machold R, Fishell G, Rowitch DH, et al. Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell. 2008;14:135–45.PubMedPubMedCentralCrossRef Yang ZJ, Ellis T, Markant SL, Read TA, Kessler JD, Bourboulas M, Schuller U, Machold R, Fishell G, Rowitch DH, et al. Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell. 2008;14:135–45.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Schuller U, Heine VM, Mao J, Kho AT, Dillon AK, Han YG, Huillard E, Sun T, Ligon AH, Qian Y, et al. Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell. 2008;14:123–34.PubMedPubMedCentralCrossRef Schuller U, Heine VM, Mao J, Kho AT, Dillon AK, Han YG, Huillard E, Sun T, Ligon AH, Qian Y, et al. Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell. 2008;14:123–34.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Biegel JA, Zhou JY, Rorke LB, Stenstrom C, Wainwright LM, Fogelgren B. Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors. Cancer Res. 1999;59:74–9.PubMed Biegel JA, Zhou JY, Rorke LB, Stenstrom C, Wainwright LM, Fogelgren B. Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors. Cancer Res. 1999;59:74–9.PubMed
60.
Zurück zum Zitat Sevenet N, Sheridan E, Amram D, Schneider P, Handgretinger R, Delattre O. Constitutional mutations of the hSNF5/INI1 gene predispose to a variety of cancers. Am J Hum Genet. 1999;65:1342–8.PubMedPubMedCentralCrossRef Sevenet N, Sheridan E, Amram D, Schneider P, Handgretinger R, Delattre O. Constitutional mutations of the hSNF5/INI1 gene predispose to a variety of cancers. Am J Hum Genet. 1999;65:1342–8.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Versteege I, Sevenet N, Lange J, Rousseau-Merck MF, Ambros P, Handgretinger R, Aurias A, Delattre O. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature. 1998;394:203–6.PubMedCrossRef Versteege I, Sevenet N, Lange J, Rousseau-Merck MF, Ambros P, Handgretinger R, Aurias A, Delattre O. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature. 1998;394:203–6.PubMedCrossRef
62.
Zurück zum Zitat Canettieri G, Di Marcotullio L, Greco A, Coni S, Antonucci L, Infante P, Pietrosanti L, De Smaele E, Ferretti E, Miele E, et al. Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation. Nat Cell Biol. 2010;12:132–42.PubMedCrossRef Canettieri G, Di Marcotullio L, Greco A, Coni S, Antonucci L, Infante P, Pietrosanti L, De Smaele E, Ferretti E, Miele E, et al. Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation. Nat Cell Biol. 2010;12:132–42.PubMedCrossRef
63.
Zurück zum Zitat Di Marcotullio L, Ferretti E, De Smaele E, Argenti B, Mincione C, Zazzeroni F, Gallo R, Masuelli L, Napolitano M, Maroder M, et al. REN(KCTD11) is a suppressor of Hedgehog signaling and is deleted in human medulloblastoma. Proc Natl Acad Sci U S A. 2004;101:10833–8.PubMedPubMedCentralCrossRef Di Marcotullio L, Ferretti E, De Smaele E, Argenti B, Mincione C, Zazzeroni F, Gallo R, Masuelli L, Napolitano M, Maroder M, et al. REN(KCTD11) is a suppressor of Hedgehog signaling and is deleted in human medulloblastoma. Proc Natl Acad Sci U S A. 2004;101:10833–8.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Cui W, Wang LH, Wen YY, Song M, Li BL, Chen XL, Xu M, An SX, Zhao J, Lu YY, et al. Expression and regulation mechanisms of Sonic Hedgehog in breast cancer. Cancer Sci. 2010;101:927–33.PubMedCrossRef Cui W, Wang LH, Wen YY, Song M, Li BL, Chen XL, Xu M, An SX, Zhao J, Lu YY, et al. Expression and regulation mechanisms of Sonic Hedgehog in breast cancer. Cancer Sci. 2010;101:927–33.PubMedCrossRef
65.
Zurück zum Zitat Wang LH, Choi YL, Hua XY, Shin YK, Song YJ, Youn SJ, Yun HY, Park SM, Kim WJ, Kim HJ, et al. Increased expression of sonic hedgehog and altered methylation of its promoter region in gastric cancer and its related lesions. Mod Pathol. 2006;19:675–83.PubMedCrossRef Wang LH, Choi YL, Hua XY, Shin YK, Song YJ, Youn SJ, Yun HY, Park SM, Kim WJ, Kim HJ, et al. Increased expression of sonic hedgehog and altered methylation of its promoter region in gastric cancer and its related lesions. Mod Pathol. 2006;19:675–83.PubMedCrossRef
66.
Zurück zum Zitat Duan ZH, Wang HC, Zhao DM, Ji XX, Song M, Yang XJ, Cui W. Cooperatively transcriptional and epigenetic regulation of sonic hedgehog overexpression drives malignant potential of breast cancer. Cancer Sci. 2015;106:1084–91.PubMedPubMedCentralCrossRef Duan ZH, Wang HC, Zhao DM, Ji XX, Song M, Yang XJ, Cui W. Cooperatively transcriptional and epigenetic regulation of sonic hedgehog overexpression drives malignant potential of breast cancer. Cancer Sci. 2015;106:1084–91.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Andersson ER, Lendahl U. Therapeutic modulation of Notch signalling--are we there yet? Nat Rev Drug Discov. 2014;13:357–78.PubMedCrossRef Andersson ER, Lendahl U. Therapeutic modulation of Notch signalling--are we there yet? Nat Rev Drug Discov. 2014;13:357–78.PubMedCrossRef
68.
Zurück zum Zitat Kopan R. Notch signaling. Cold Spring Harb Perspect Biol. 2012;4. Kopan R. Notch signaling. Cold Spring Harb Perspect Biol. 2012;4.
69.
Zurück zum Zitat Andersson ER, Sandberg R, Lendahl U. Notch signaling: simplicity in design, versatility in function. Development. 2011;138:3593–612.PubMedCrossRef Andersson ER, Sandberg R, Lendahl U. Notch signaling: simplicity in design, versatility in function. Development. 2011;138:3593–612.PubMedCrossRef
70.
Zurück zum Zitat Munoz P, Iliou MS, Esteller M. Epigenetic alterations involved in cancer stem cell reprogramming. Mol Oncol. 2012;6:620–36.PubMedCrossRef Munoz P, Iliou MS, Esteller M. Epigenetic alterations involved in cancer stem cell reprogramming. Mol Oncol. 2012;6:620–36.PubMedCrossRef
71.
Zurück zum Zitat Liu J, Sato C, Cerletti M, Wagers A. Notch signaling in the regulation of stem cell self-renewal and differentiation. Curr Top Dev Biol. 2010;92:367–409.PubMedCrossRef Liu J, Sato C, Cerletti M, Wagers A. Notch signaling in the regulation of stem cell self-renewal and differentiation. Curr Top Dev Biol. 2010;92:367–409.PubMedCrossRef
72.
Zurück zum Zitat Androutsellis-Theotokis A, Leker RR, Soldner F, Hoeppner DJ, Ravin R, Poser SW, Rueger MA, Bae SK, Kittappa R, McKay RD. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature. 2006;442:823–6.PubMedCrossRef Androutsellis-Theotokis A, Leker RR, Soldner F, Hoeppner DJ, Ravin R, Poser SW, Rueger MA, Bae SK, Kittappa R, McKay RD. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature. 2006;442:823–6.PubMedCrossRef
73.
Zurück zum Zitat Lopez-Arribillaga E, Rodilla V, Pellegrinet L, Guiu J, Iglesias M, Roman AC, Gutarra S, Gonzalez S, Munoz-Canoves P, Fernandez-Salguero P, et al. Bmi1 regulates murine intestinal stem cell proliferation and self-renewal downstream of Notch. Development. 2015;142:41–50.PubMedCrossRef Lopez-Arribillaga E, Rodilla V, Pellegrinet L, Guiu J, Iglesias M, Roman AC, Gutarra S, Gonzalez S, Munoz-Canoves P, Fernandez-Salguero P, et al. Bmi1 regulates murine intestinal stem cell proliferation and self-renewal downstream of Notch. Development. 2015;142:41–50.PubMedCrossRef
74.
Zurück zum Zitat Yuan X, Wu H, Han N, Xu H, Chu Q, Yu S, Chen Y, Wu K. Notch signaling and EMT in non-small cell lung cancer: biological significance and therapeutic application. J Hematol Oncol. 2014;7:87.PubMedPubMedCentralCrossRef Yuan X, Wu H, Han N, Xu H, Chu Q, Yu S, Chen Y, Wu K. Notch signaling and EMT in non-small cell lung cancer: biological significance and therapeutic application. J Hematol Oncol. 2014;7:87.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Ichida JK, Tcw J, Williams LA, Carter AC, Shi Y, Moura MT, Ziller M, Singh S, Amabile G, Bock C, et al. Notch inhibition allows oncogene-independent generation of iPS cells. Nat Chem Biol. 2014;10:632–9.PubMedPubMedCentralCrossRef Ichida JK, Tcw J, Williams LA, Carter AC, Shi Y, Moura MT, Ziller M, Singh S, Amabile G, Bock C, et al. Notch inhibition allows oncogene-independent generation of iPS cells. Nat Chem Biol. 2014;10:632–9.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Zou J, Li P, Lu F, Liu N, Dai J, Ye J, Qu X, Sun X, Ma D, Park J, Ji C. Notch1 is required for hypoxia-induced proliferation, invasion and chemoresistance of T-cell acute lymphoblastic leukemia cells. J Hematol Oncol. 2013;6:3.PubMedPubMedCentralCrossRef Zou J, Li P, Lu F, Liu N, Dai J, Ye J, Qu X, Sun X, Ma D, Park J, Ji C. Notch1 is required for hypoxia-induced proliferation, invasion and chemoresistance of T-cell acute lymphoblastic leukemia cells. J Hematol Oncol. 2013;6:3.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Gallahan D, Callahan R. The mouse mammary tumor associated gene INT3 is a unique member of the NOTCH gene family (NOTCH4). Oncogene. 1997;14:1883–90.PubMedCrossRef Gallahan D, Callahan R. The mouse mammary tumor associated gene INT3 is a unique member of the NOTCH gene family (NOTCH4). Oncogene. 1997;14:1883–90.PubMedCrossRef
78.
Zurück zum Zitat D’Angelo RC, Ouzounova M, Davis A, Choi D, Tchuenkam SM, Kim G, Luther T, Quraishi AA, Senbabaoglu Y, Conley SJ, et al. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Mol Cancer Ther. 2015;14:779–87.PubMedPubMedCentralCrossRef D’Angelo RC, Ouzounova M, Davis A, Choi D, Tchuenkam SM, Kim G, Luther T, Quraishi AA, Senbabaoglu Y, Conley SJ, et al. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Mol Cancer Ther. 2015;14:779–87.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Wang Z, Da Silva TG, Jin K, Han X, Ranganathan P, Zhu X, Sanchez-Mejias A, Bai F, Li B, Fei DL, et al. Notch signaling drives stemness and tumorigenicity of esophageal adenocarcinoma. Cancer Res. 2014;74:6364–74.PubMedPubMedCentralCrossRef Wang Z, Da Silva TG, Jin K, Han X, Ranganathan P, Zhu X, Sanchez-Mejias A, Bai F, Li B, Fei DL, et al. Notch signaling drives stemness and tumorigenicity of esophageal adenocarcinoma. Cancer Res. 2014;74:6364–74.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Jin L, Vu TT, Datta PK. Abstract 1709: STRAP mediates the stemness of human colorectal cancer cells by epigenetic regulation of Notch pathway. Cancer Res. 2016;76:1709.CrossRef Jin L, Vu TT, Datta PK. Abstract 1709: STRAP mediates the stemness of human colorectal cancer cells by epigenetic regulation of Notch pathway. Cancer Res. 2016;76:1709.CrossRef
81.
Zurück zum Zitat Ghoshal P, Nganga AJ, Moran-Giuati J, Szafranek A, Johnson TR, Bigelow AJ, Houde CM, Avet-Loiseau H, Smiraglia DJ, Ersing N, et al. Loss of the SMRT/NCoR2 corepressor correlates with JAG2 overexpression in multiple myeloma. Cancer Res. 2009;69:4380–7.PubMedCrossRef Ghoshal P, Nganga AJ, Moran-Giuati J, Szafranek A, Johnson TR, Bigelow AJ, Houde CM, Avet-Loiseau H, Smiraglia DJ, Ersing N, et al. Loss of the SMRT/NCoR2 corepressor correlates with JAG2 overexpression in multiple myeloma. Cancer Res. 2009;69:4380–7.PubMedCrossRef
82.
83.
Zurück zum Zitat Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.PubMedCrossRef Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.PubMedCrossRef
84.
Zurück zum Zitat Morel A-P, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of Breast Cancer Stem Cells through Epithelial-Mesenchymal Transition. PLoS One. 2008;3:e2888.PubMedPubMedCentralCrossRef Morel A-P, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of Breast Cancer Stem Cells through Epithelial-Mesenchymal Transition. PLoS One. 2008;3:e2888.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–15.PubMedPubMedCentralCrossRef Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–15.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Dissanayake SK, Wade M, Johnson CE, O’Connell MP, Leotlela PD, French AD, Shah KV, Hewitt KJ, Rosenthal DT, Indig FE, et al. The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition. J Biol Chem. 2007;282:17259–71.PubMedPubMedCentralCrossRef Dissanayake SK, Wade M, Johnson CE, O’Connell MP, Leotlela PD, French AD, Shah KV, Hewitt KJ, Rosenthal DT, Indig FE, et al. The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition. J Biol Chem. 2007;282:17259–71.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Vincan E, Barker N. The upstream components of the Wnt signalling pathway in the dynamic EMT and MET associated with colorectal cancer progression. Clin Exp Metastasis. 2008;25:657–63.PubMedCrossRef Vincan E, Barker N. The upstream components of the Wnt signalling pathway in the dynamic EMT and MET associated with colorectal cancer progression. Clin Exp Metastasis. 2008;25:657–63.PubMedCrossRef
89.
Zurück zum Zitat Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14:818–29.PubMedCrossRef Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14:818–29.PubMedCrossRef
90.
Zurück zum Zitat Dohadwala M, Yang SC, Luo J, Sharma S, Batra RK, Huang M, Lin Y, Goodglick L, Krysan K, Fishbein MC, et al. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res. 2006;66:5338–45.PubMedCrossRef Dohadwala M, Yang SC, Luo J, Sharma S, Batra RK, Huang M, Lin Y, Goodglick L, Krysan K, Fishbein MC, et al. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res. 2006;66:5338–45.PubMedCrossRef
91.
Zurück zum Zitat Dumont N, Wilson MB, Crawford YG, Reynolds PA, Sigaroudinia M, Tlsty TD. Sustained induction of epithelial to mesenchymal transition activates DNA methylation of genes silenced in basal-like breast cancers. Proc Natl Acad Sci U S A. 2008;105:14867–72.PubMedPubMedCentralCrossRef Dumont N, Wilson MB, Crawford YG, Reynolds PA, Sigaroudinia M, Tlsty TD. Sustained induction of epithelial to mesenchymal transition activates DNA methylation of genes silenced in basal-like breast cancers. Proc Natl Acad Sci U S A. 2008;105:14867–72.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Gibbons DL, Lin W, Creighton CJ, Rizvi ZH, Gregory PA, Goodall GJ, Thilaganathan N, Du L, Zhang Y, Pertsemlidis A, Kurie JM. Contextual extracellular cues promote tumor cell EMT and metastasis by regulating miR-200 family expression. Genes Dev. 2009;23:2140–51.PubMedPubMedCentralCrossRef Gibbons DL, Lin W, Creighton CJ, Rizvi ZH, Gregory PA, Goodall GJ, Thilaganathan N, Du L, Zhang Y, Pertsemlidis A, Kurie JM. Contextual extracellular cues promote tumor cell EMT and metastasis by regulating miR-200 family expression. Genes Dev. 2009;23:2140–51.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Avgustinova A, Benitah SA. The epigenetics of tumour initiation: cancer stem cells and their chromatin. Curr Opin Genet Dev. 2016;36:8–15.PubMedCrossRef Avgustinova A, Benitah SA. The epigenetics of tumour initiation: cancer stem cells and their chromatin. Curr Opin Genet Dev. 2016;36:8–15.PubMedCrossRef
94.
Zurück zum Zitat Koizume S, Tachibana K, Sekiya T, Hirohashi S, Shiraishi M. Heterogeneity in the modification and involvement of chromatin components of the CpG island of the silenced human CDH1 gene in cancer cells. Nucleic Acids Res. 2002;30:4770–80.PubMedPubMedCentralCrossRef Koizume S, Tachibana K, Sekiya T, Hirohashi S, Shiraishi M. Heterogeneity in the modification and involvement of chromatin components of the CpG island of the silenced human CDH1 gene in cancer cells. Nucleic Acids Res. 2002;30:4770–80.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Wang Y, Shang Y. Epigenetic control of epithelial-to-mesenchymal transition and cancer metastasis. Exp Cell Res. 2013;319:160–9.PubMedCrossRef Wang Y, Shang Y. Epigenetic control of epithelial-to-mesenchymal transition and cancer metastasis. Exp Cell Res. 2013;319:160–9.PubMedCrossRef
96.
Zurück zum Zitat Cao Q, Yu J, Dhanasekaran SM, Kim JH, Mani RS, Tomlins SA, Mehra R, Laxman B, Cao X, Yu J, et al. Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene. 2008;27:7274–84.PubMedPubMedCentralCrossRef Cao Q, Yu J, Dhanasekaran SM, Kim JH, Mani RS, Tomlins SA, Mehra R, Laxman B, Cao X, Yu J, et al. Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene. 2008;27:7274–84.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Herranz N, Pasini D, Diaz VM, Franci C, Gutierrez A, Dave N, Escriva M, Hernandez-Munoz I, Di Croce L, Helin K, et al. Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor. Mol Cell Biol. 2008;28:4772–81.PubMedPubMedCentralCrossRef Herranz N, Pasini D, Diaz VM, Franci C, Gutierrez A, Dave N, Escriva M, Hernandez-Munoz I, Di Croce L, Helin K, et al. Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor. Mol Cell Biol. 2008;28:4772–81.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10:593–601.PubMedCrossRef Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10:593–601.PubMedCrossRef
99.
Zurück zum Zitat Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem. 2008;283:14910–4.PubMedPubMedCentralCrossRef Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem. 2008;283:14910–4.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22:894–907.PubMedPubMedCentralCrossRef Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22:894–907.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Chang CJ, Chao CH, Xia W, Yang JY, Xiong Y, Li CW, Yu WH, Rehman SK, Hsu JL, Lee HH, et al. p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol. 2011;13:317–23.PubMedPubMedCentralCrossRef Chang CJ, Chao CH, Xia W, Yang JY, Xiong Y, Li CW, Yu WH, Rehman SK, Hsu JL, Lee HH, et al. p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol. 2011;13:317–23.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Tellez CS, Juri DE, Do K, Bernauer AM, Thomas CL, Damiani LA, Tessema M, Leng S, Belinsky SA. EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic silencing are early manifestations during carcinogen-induced transformation of human lung epithelial cells. Cancer Res. 2011;71:3087–97.PubMedPubMedCentralCrossRef Tellez CS, Juri DE, Do K, Bernauer AM, Thomas CL, Damiani LA, Tessema M, Leng S, Belinsky SA. EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic silencing are early manifestations during carcinogen-induced transformation of human lung epithelial cells. Cancer Res. 2011;71:3087–97.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Shah AN, Summy JM, Zhang J, Park SI, Parikh NU, Gallick GE. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol. 2007;14:3629–37.PubMedCrossRef Shah AN, Summy JM, Zhang J, Park SI, Parikh NU, Gallick GE. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol. 2007;14:3629–37.PubMedCrossRef
104.
Zurück zum Zitat Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, Ali S, Abbruzzese JL, Gallick GE, Sarkar FH. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69:2400–7.PubMedPubMedCentralCrossRef Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, Ali S, Abbruzzese JL, Gallick GE, Sarkar FH. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69:2400–7.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Shukla S, Meeran SM. Epigenetics of cancer stem cells: Pathways and therapeutics. Biochim Biophys Acta. 1840;2014:3494–502. Shukla S, Meeran SM. Epigenetics of cancer stem cells: Pathways and therapeutics. Biochim Biophys Acta. 1840;2014:3494–502.
106.
Zurück zum Zitat Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, Gallick GE, Logsdon CD, McConkey DJ, Choi W. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009;69:5820–8.PubMedPubMedCentralCrossRef Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, Gallick GE, Logsdon CD, McConkey DJ, Choi W. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009;69:5820–8.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Wu CP, Calcagno AM, Ambudkar SV. Reversal of ABC drug transporter-mediated multidrug resistance in cancer cells: evaluation of current strategies. Curr Mol Pharmacol. 2008;1:93–105.PubMedPubMedCentralCrossRef Wu CP, Calcagno AM, Ambudkar SV. Reversal of ABC drug transporter-mediated multidrug resistance in cancer cells: evaluation of current strategies. Curr Mol Pharmacol. 2008;1:93–105.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Wang X, Low XC, Hou W, Abdullah LN, Toh TB. Mohd Abdul Rashid M, Ho D, Chow EK: Epirubicin-adsorbed nanodiamonds kill chemoresistant hepatic cancer stem cells. ACS Nano. 2014;8:12151–66.PubMedPubMedCentralCrossRef Wang X, Low XC, Hou W, Abdullah LN, Toh TB. Mohd Abdul Rashid M, Ho D, Chow EK: Epirubicin-adsorbed nanodiamonds kill chemoresistant hepatic cancer stem cells. ACS Nano. 2014;8:12151–66.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Cho S, Lu M, He X, Ee PL, Bhat U, Schneider E, Miele L, Beck WT. Notch1 regulates the expression of the multidrug resistance gene ABCC1/MRP1 in cultured cancer cells. Proc Natl Acad Sci U S A. 2011;108:20778–83.PubMedPubMedCentralCrossRef Cho S, Lu M, He X, Ee PL, Bhat U, Schneider E, Miele L, Beck WT. Notch1 regulates the expression of the multidrug resistance gene ABCC1/MRP1 in cultured cancer cells. Proc Natl Acad Sci U S A. 2011;108:20778–83.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Kim B, Stephen SL, Hanby AM, Horgan K, Perry SL, Richardson J, Roundhill EA, Valleley EM, Verghese ET, Williams BJ, et al. Chemotherapy induces Notch1-dependent MRP1 up-regulation, inhibition of which sensitizes breast cancer cells to chemotherapy. BMC Cancer. 2015;15:634.PubMedPubMedCentralCrossRef Kim B, Stephen SL, Hanby AM, Horgan K, Perry SL, Richardson J, Roundhill EA, Valleley EM, Verghese ET, Williams BJ, et al. Chemotherapy induces Notch1-dependent MRP1 up-regulation, inhibition of which sensitizes breast cancer cells to chemotherapy. BMC Cancer. 2015;15:634.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat To KK, Polgar O, Huff LM, Morisaki K, Bates SE. Histone modifications at the ABCG2 promoter following treatment with histone deacetylase inhibitor mirror those in multidrug-resistant cells. Mol Cancer Res. 2008;6:151–64.PubMedPubMedCentralCrossRef To KK, Polgar O, Huff LM, Morisaki K, Bates SE. Histone modifications at the ABCG2 promoter following treatment with histone deacetylase inhibitor mirror those in multidrug-resistant cells. Mol Cancer Res. 2008;6:151–64.PubMedPubMedCentralCrossRef
112.
113.
Zurück zum Zitat Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008;123:8–13.PubMedCrossRef Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008;123:8–13.PubMedCrossRef
114.
Zurück zum Zitat Juttermann R, Li E, Jaenisch R. Toxicity of 5-aza-2′-deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation. Proc Natl Acad Sci U S A. 1994;91:11797–801.PubMedPubMedCentralCrossRef Juttermann R, Li E, Jaenisch R. Toxicity of 5-aza-2′-deoxycytidine to mammalian cells is mediated primarily by covalent trapping of DNA methyltransferase rather than DNA demethylation. Proc Natl Acad Sci U S A. 1994;91:11797–801.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Ghoshal K, Datta J, Majumder S, Bai S, Kutay H, Motiwala T, Jacob ST. 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal. Mol Cell Biol. 2005;25:4727–41.PubMedPubMedCentralCrossRef Ghoshal K, Datta J, Majumder S, Bai S, Kutay H, Motiwala T, Jacob ST. 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal. Mol Cell Biol. 2005;25:4727–41.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Sorm F, Vesely J. the activity of a new antimetabolite, 5-azacytidine, against lymphoid leukaemia in ak mice. Neoplasma. 1964;11:123–30.PubMed Sorm F, Vesely J. the activity of a new antimetabolite, 5-azacytidine, against lymphoid leukaemia in ak mice. Neoplasma. 1964;11:123–30.PubMed
117.
Zurück zum Zitat Abele R, Clavel M, Dodion P, Bruntsch U, Gundersen S, Smyth J, Renard J, van Glabbeke M, Pinedo HM. The EORTC Early Clinical Trials Cooperative Group experience with 5-aza-2′-deoxycytidine (NSC 127716) in patients with colo-rectal, head and neck, renal carcinomas and malignant melanomas. Eur J Cancer Clin Oncol. 1987;23:1921–4.PubMedCrossRef Abele R, Clavel M, Dodion P, Bruntsch U, Gundersen S, Smyth J, Renard J, van Glabbeke M, Pinedo HM. The EORTC Early Clinical Trials Cooperative Group experience with 5-aza-2′-deoxycytidine (NSC 127716) in patients with colo-rectal, head and neck, renal carcinomas and malignant melanomas. Eur J Cancer Clin Oncol. 1987;23:1921–4.PubMedCrossRef
118.
Zurück zum Zitat Cowan LA, Talwar S, Yang AS. Will DNA methylation inhibitors work in solid tumors? A review of the clinical experience with azacitidine and decitabine in solid tumors. Epigenomics. 2010;2:71–86.PubMedCrossRef Cowan LA, Talwar S, Yang AS. Will DNA methylation inhibitors work in solid tumors? A review of the clinical experience with azacitidine and decitabine in solid tumors. Epigenomics. 2010;2:71–86.PubMedCrossRef
119.
Zurück zum Zitat Clavel M, Monfardini S, Fossa S, Smyth J, Renard J, Kaye SB. 5-Aza-2′-deoxycytidine (NSC 127716) in non-seminomatous testicular cancer. Phase II from the EORTC Early Clinical Trials Cooperative Group and Genito-Urinary Group. Ann Oncol. 1992;3:399–400.PubMedCrossRef Clavel M, Monfardini S, Fossa S, Smyth J, Renard J, Kaye SB. 5-Aza-2′-deoxycytidine (NSC 127716) in non-seminomatous testicular cancer. Phase II from the EORTC Early Clinical Trials Cooperative Group and Genito-Urinary Group. Ann Oncol. 1992;3:399–400.PubMedCrossRef
120.
Zurück zum Zitat Stadler WM, Margolin K, Ferber S, McCulloch W, Thompson JA. A phase II study of depsipeptide in refractory metastatic renal cell cancer. Clin Genitourin Cancer. 2006;5:57–60.PubMedCrossRef Stadler WM, Margolin K, Ferber S, McCulloch W, Thompson JA. A phase II study of depsipeptide in refractory metastatic renal cell cancer. Clin Genitourin Cancer. 2006;5:57–60.PubMedCrossRef
121.
Zurück zum Zitat Tsai HC, Li H, Van Neste L, Cai Y, Robert C, Rassool FV, Shin JJ, Harbom KM, Beaty R, Pappou E, et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell. 2012;21:430–46.PubMedPubMedCentralCrossRef Tsai HC, Li H, Van Neste L, Cai Y, Robert C, Rassool FV, Shin JJ, Harbom KM, Beaty R, Pappou E, et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell. 2012;21:430–46.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Pinto A, Zagonel V, Attadia V, Bullian PL, Gattei V, Carbone A, Monfardini S, Colombatti A. 5-Aza-2′-deoxycytidine as a differentiation inducer in acute myeloid leukaemias and myelodysplastic syndromes of the elderly. Bone Marrow Transplant. 1989;4 Suppl 3:28–32.PubMed Pinto A, Zagonel V, Attadia V, Bullian PL, Gattei V, Carbone A, Monfardini S, Colombatti A. 5-Aza-2′-deoxycytidine as a differentiation inducer in acute myeloid leukaemias and myelodysplastic syndromes of the elderly. Bone Marrow Transplant. 1989;4 Suppl 3:28–32.PubMed
123.
Zurück zum Zitat Zagonel V, Lo Re G, Marotta G, Babare R, Sardeo G, Gattei V, De Angelis V, Monfardini S, Pinto A. 5-Aza-2′-deoxycytidine (Decitabine) induces trilineage response in unfavourable myelodysplastic syndromes. Leukemia. 1993;7 Suppl 1:30–5.PubMed Zagonel V, Lo Re G, Marotta G, Babare R, Sardeo G, Gattei V, De Angelis V, Monfardini S, Pinto A. 5-Aza-2′-deoxycytidine (Decitabine) induces trilineage response in unfavourable myelodysplastic syndromes. Leukemia. 1993;7 Suppl 1:30–5.PubMed
124.
Zurück zum Zitat Tian J, Lee SO, Liang L, Luo J, Huang CK, Li L, Niu Y, Chang C. Targeting the unique methylation pattern of androgen receptor (AR) promoter in prostate stem/progenitor cells with 5-aza-2′-deoxycytidine (5-AZA) leads to suppressed prostate tumorigenesis. J Biol Chem. 2012;287:39954–66.PubMedPubMedCentralCrossRef Tian J, Lee SO, Liang L, Luo J, Huang CK, Li L, Niu Y, Chang C. Targeting the unique methylation pattern of androgen receptor (AR) promoter in prostate stem/progenitor cells with 5-aza-2′-deoxycytidine (5-AZA) leads to suppressed prostate tumorigenesis. J Biol Chem. 2012;287:39954–66.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Wang Y, Cardenas H, Fang F, Condello S, Taverna P, Segar M, Liu Y, Nephew KP, Matei D. Epigenetic targeting of ovarian cancer stem cells. Cancer Res. 2014;74:4922–36.PubMedPubMedCentralCrossRef Wang Y, Cardenas H, Fang F, Condello S, Taverna P, Segar M, Liu Y, Nephew KP, Matei D. Epigenetic targeting of ovarian cancer stem cells. Cancer Res. 2014;74:4922–36.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Kaminskas E, Farrell AT, Wang YC, Sridhara R, Pazdur R. FDA drug approval summary: azacitidine (5-azacytidine, Vidaza) for injectable suspension. Oncologist. 2005;10:176–82.PubMedCrossRef Kaminskas E, Farrell AT, Wang YC, Sridhara R, Pazdur R. FDA drug approval summary: azacitidine (5-azacytidine, Vidaza) for injectable suspension. Oncologist. 2005;10:176–82.PubMedCrossRef
127.
Zurück zum Zitat Finley A, Copeland RA. Small molecule control of chromatin remodeling. Chem Biol. 2014;21:1196–210.PubMedCrossRef Finley A, Copeland RA. Small molecule control of chromatin remodeling. Chem Biol. 2014;21:1196–210.PubMedCrossRef
128.
Zurück zum Zitat de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J. 2003;370:737–49.PubMedPubMedCentralCrossRef de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J. 2003;370:737–49.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Johnstone RW, Licht JD. Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell. 2003;4:13–8.PubMedCrossRef Johnstone RW, Licht JD. Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell. 2003;4:13–8.PubMedCrossRef
130.
Zurück zum Zitat Gelmetti V, Zhang J, Fanelli M, Minucci S, Pelicci PG, Lazar MA. Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO. Mol Cell Biol. 1998;18:7185–91.PubMedPubMedCentralCrossRef Gelmetti V, Zhang J, Fanelli M, Minucci S, Pelicci PG, Lazar MA. Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO. Mol Cell Biol. 1998;18:7185–91.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Luo J, Su F, Chen D, Shiloh A, Gu W. Deacetylation of p53 modulates its effect on cell growth and apoptosis. Nature. 2000;408:377–81.PubMedCrossRef Luo J, Su F, Chen D, Shiloh A, Gu W. Deacetylation of p53 modulates its effect on cell growth and apoptosis. Nature. 2000;408:377–81.PubMedCrossRef
132.
Zurück zum Zitat Duan H, Heckman CA, Boxer LM. Histone deacetylase inhibitors down-regulate bcl-2 expression and induce apoptosis in t(14;18) lymphomas. Mol Cell Biol. 2005;25:1608–19.PubMedPubMedCentralCrossRef Duan H, Heckman CA, Boxer LM. Histone deacetylase inhibitors down-regulate bcl-2 expression and induce apoptosis in t(14;18) lymphomas. Mol Cell Biol. 2005;25:1608–19.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Federico M, Bagella L. Histone deacetylase inhibitors in the treatment of hematological malignancies and solid tumors. J Biomed Biotechnol. 2011;2011:475641.PubMedCrossRef Federico M, Bagella L. Histone deacetylase inhibitors in the treatment of hematological malignancies and solid tumors. J Biomed Biotechnol. 2011;2011:475641.PubMedCrossRef
134.
Zurück zum Zitat Juo YY, Gong XJ, Mishra A, Cui X, Baylin SB, Azad NS, Ahuja N. Epigenetic therapy for solid tumors: from bench science to clinical trials. Epigenomics. 2015;7:215–35.PubMedCrossRef Juo YY, Gong XJ, Mishra A, Cui X, Baylin SB, Azad NS, Ahuja N. Epigenetic therapy for solid tumors: from bench science to clinical trials. Epigenomics. 2015;7:215–35.PubMedCrossRef
135.
Zurück zum Zitat Olsen EA, Kim YH, Kuzel TM, Pacheco TR, Foss FM, Parker S, Frankel SR, Chen C, Ricker JL, Arduino JM, Duvic M. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol. 2007;25:3109–15.PubMedCrossRef Olsen EA, Kim YH, Kuzel TM, Pacheco TR, Foss FM, Parker S, Frankel SR, Chen C, Ricker JL, Arduino JM, Duvic M. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J Clin Oncol. 2007;25:3109–15.PubMedCrossRef
136.
Zurück zum Zitat Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, Zain J, Prince HM, Leonard JP, Geskin LJ, et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol. 2009;27:5410–7.PubMedPubMedCentralCrossRef Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, Zain J, Prince HM, Leonard JP, Geskin LJ, et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J Clin Oncol. 2009;27:5410–7.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Thaler F. Current trends in the development of histone deacetylase inhibitors: a review of recent patent applications. Pharm Pat Anal. 2012;1:75–90.PubMedCrossRef Thaler F. Current trends in the development of histone deacetylase inhibitors: a review of recent patent applications. Pharm Pat Anal. 2012;1:75–90.PubMedCrossRef
138.
Zurück zum Zitat Whittaker SJ, Demierre MF, Kim EJ, Rook AH, Lerner A, Duvic M, Scarisbrick J, Reddy S, Robak T, Becker JC, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol. 2010;28:4485–91.PubMedCrossRef Whittaker SJ, Demierre MF, Kim EJ, Rook AH, Lerner A, Duvic M, Scarisbrick J, Reddy S, Robak T, Becker JC, et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol. 2010;28:4485–91.PubMedCrossRef
139.
Zurück zum Zitat Vansteenkiste J, Van Cutsem E, Dumez H, Chen C, Ricker JL, Randolph SS, Schoffski P. Early phase II trial of oral vorinostat in relapsed or refractory breast, colorectal, or non-small cell lung cancer. Invest New Drugs. 2008;26:483–8.PubMedCrossRef Vansteenkiste J, Van Cutsem E, Dumez H, Chen C, Ricker JL, Randolph SS, Schoffski P. Early phase II trial of oral vorinostat in relapsed or refractory breast, colorectal, or non-small cell lung cancer. Invest New Drugs. 2008;26:483–8.PubMedCrossRef
140.
Zurück zum Zitat Luu TH, Morgan RJ, Leong L, Lim D, McNamara M, Portnow J, Frankel P, Smith DD, Doroshow JH, Wong C, et al. A phase II trial of vorinostat (suberoylanilide hydroxamic acid) in metastatic breast cancer: a California Cancer Consortium study. Clin Cancer Res. 2008;14:7138–42.PubMedPubMedCentralCrossRef Luu TH, Morgan RJ, Leong L, Lim D, McNamara M, Portnow J, Frankel P, Smith DD, Doroshow JH, Wong C, et al. A phase II trial of vorinostat (suberoylanilide hydroxamic acid) in metastatic breast cancer: a California Cancer Consortium study. Clin Cancer Res. 2008;14:7138–42.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Woyach JA, Kloos RT, Ringel MD, Arbogast D, Collamore M, Zwiebel JA, Grever M, Villalona-Calero M, Shah MH. Lack of therapeutic effect of the histone deacetylase inhibitor vorinostat in patients with metastatic radioiodine-refractory thyroid carcinoma. J Clin Endocrinol Metab. 2009;94:164–70.PubMedCrossRef Woyach JA, Kloos RT, Ringel MD, Arbogast D, Collamore M, Zwiebel JA, Grever M, Villalona-Calero M, Shah MH. Lack of therapeutic effect of the histone deacetylase inhibitor vorinostat in patients with metastatic radioiodine-refractory thyroid carcinoma. J Clin Endocrinol Metab. 2009;94:164–70.PubMedCrossRef
142.
Zurück zum Zitat Modesitt SC, Sill M, Hoffman JS, Bender DP. A phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2008;109:182–6.PubMedCrossRef Modesitt SC, Sill M, Hoffman JS, Bender DP. A phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2008;109:182–6.PubMedCrossRef
143.
Zurück zum Zitat Blumenschein Jr GR, Kies MS, Papadimitrakopoulou VA, Lu C, Kumar AJ, Ricker JL, Chiao JH, Chen C, Frankel SR. Phase II trial of the histone deacetylase inhibitor vorinostat (Zolinza, suberoylanilide hydroxamic acid, SAHA) in patients with recurrent and/or metastatic head and neck cancer. Invest New Drugs. 2008;26:81–7.PubMedCrossRef Blumenschein Jr GR, Kies MS, Papadimitrakopoulou VA, Lu C, Kumar AJ, Ricker JL, Chiao JH, Chen C, Frankel SR. Phase II trial of the histone deacetylase inhibitor vorinostat (Zolinza, suberoylanilide hydroxamic acid, SAHA) in patients with recurrent and/or metastatic head and neck cancer. Invest New Drugs. 2008;26:81–7.PubMedCrossRef
144.
Zurück zum Zitat Traynor AM, Dubey S, Eickhoff JC, Kolesar JM, Schell K, Huie MS, Groteluschen DL, Marcotte SM, Hallahan CM, Weeks HR, et al. Vorinostat (NSC# 701852) in patients with relapsed non-small cell lung cancer: a Wisconsin Oncology Network phase II study. J Thorac Oncol. 2009;4:522–6.PubMedPubMedCentralCrossRef Traynor AM, Dubey S, Eickhoff JC, Kolesar JM, Schell K, Huie MS, Groteluschen DL, Marcotte SM, Hallahan CM, Weeks HR, et al. Vorinostat (NSC# 701852) in patients with relapsed non-small cell lung cancer: a Wisconsin Oncology Network phase II study. J Thorac Oncol. 2009;4:522–6.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Galanis E, Jaeckle KA, Maurer MJ, Reid JM, Ames MM, Hardwick JS, Reilly JF, Loboda A, Nebozhyn M, Fantin VR, et al. Phase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study. J Clin Oncol. 2009;27:2052–8.PubMedPubMedCentralCrossRef Galanis E, Jaeckle KA, Maurer MJ, Reid JM, Ames MM, Hardwick JS, Reilly JF, Loboda A, Nebozhyn M, Fantin VR, et al. Phase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study. J Clin Oncol. 2009;27:2052–8.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, Hancox A, Hong JA, Chen GA, Kruchin E, et al. Clinical and molecular responses in lung cancer patients receiving Romidepsin. Clin Cancer Res. 2008;14:188–98.PubMedCrossRef Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, Hancox A, Hong JA, Chen GA, Kruchin E, et al. Clinical and molecular responses in lung cancer patients receiving Romidepsin. Clin Cancer Res. 2008;14:188–98.PubMedCrossRef
147.
Zurück zum Zitat Otterson GA, Hodgson L, Pang H, Vokes EE. Cancer, Leukemia Group B: Phase II study of the histone deacetylase inhibitor Romidepsin in relapsed small cell lung cancer (Cancer and Leukemia Group B 30304). J Thorac Oncol. 2010;5:1644–8.PubMedPubMedCentralCrossRef Otterson GA, Hodgson L, Pang H, Vokes EE. Cancer, Leukemia Group B: Phase II study of the histone deacetylase inhibitor Romidepsin in relapsed small cell lung cancer (Cancer and Leukemia Group B 30304). J Thorac Oncol. 2010;5:1644–8.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Iwamoto FM, Lamborn KR, Kuhn JG, Wen PY, Yung WK, Gilbert MR, Chang SM, Lieberman FS, Prados MD, Fine HA. A phase I/II trial of the histone deacetylase inhibitor romidepsin for adults with recurrent malignant glioma: North American Brain Tumor Consortium Study 03–03. Neuro Oncol. 2011;13:509–16.PubMedPubMedCentralCrossRef Iwamoto FM, Lamborn KR, Kuhn JG, Wen PY, Yung WK, Gilbert MR, Chang SM, Lieberman FS, Prados MD, Fine HA. A phase I/II trial of the histone deacetylase inhibitor romidepsin for adults with recurrent malignant glioma: North American Brain Tumor Consortium Study 03–03. Neuro Oncol. 2011;13:509–16.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Whitehead RP, Rankin C, Hoff PM, Gold PJ, Billingsley KG, Chapman RA, Wong L, Ward JH, Abbruzzese JL, Blanke CD. Phase II trial of romidepsin (NSC-630176) in previously treated colorectal cancer patients with advanced disease: a Southwest Oncology Group study (S0336). Invest New Drugs. 2009;27:469–75.PubMedCrossRef Whitehead RP, Rankin C, Hoff PM, Gold PJ, Billingsley KG, Chapman RA, Wong L, Ward JH, Abbruzzese JL, Blanke CD. Phase II trial of romidepsin (NSC-630176) in previously treated colorectal cancer patients with advanced disease: a Southwest Oncology Group study (S0336). Invest New Drugs. 2009;27:469–75.PubMedCrossRef
150.
Zurück zum Zitat Molife LR, Attard G, Fong PC, Karavasilis V, Reid AH, Patterson S, Riggs Jr CE, Higano C, Stadler WM, McCulloch W, et al. Phase II, two-stage, single-arm trial of the histone deacetylase inhibitor (HDACi) romidepsin in metastatic castration-resistant prostate cancer (CRPC). Ann Oncol. 2010;21:109–13.PubMedCrossRef Molife LR, Attard G, Fong PC, Karavasilis V, Reid AH, Patterson S, Riggs Jr CE, Higano C, Stadler WM, McCulloch W, et al. Phase II, two-stage, single-arm trial of the histone deacetylase inhibitor (HDACi) romidepsin in metastatic castration-resistant prostate cancer (CRPC). Ann Oncol. 2010;21:109–13.PubMedCrossRef
151.
153.
Zurück zum Zitat Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. 2009;27:5459–68.PubMedCrossRef Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. 2009;27:5459–68.PubMedCrossRef
154.
Zurück zum Zitat Tarhini AA, Zahoor H, McLaughlin B, Gooding WE, Schmitz JC, Siegfried JM, Socinski MA, Argiris A. Phase I trial of carboplatin and etoposide in combination with panobinostat in patients with lung cancer. Anticancer Res. 2013;33:4475–81.PubMedPubMedCentral Tarhini AA, Zahoor H, McLaughlin B, Gooding WE, Schmitz JC, Siegfried JM, Socinski MA, Argiris A. Phase I trial of carboplatin and etoposide in combination with panobinostat in patients with lung cancer. Anticancer Res. 2013;33:4475–81.PubMedPubMedCentral
155.
Zurück zum Zitat Cassier PA, Lefranc A, Amela EY, Chevreau C, Bui BN, Lecesne A, Ray-Coquard I, Chabaud S, Penel N, Berge Y, et al. A phase II trial of panobinostat in patients with advanced pretreated soft tissue sarcoma. A study from the French Sarcoma Group. Br J Cancer. 2013;109:909–14.PubMedPubMedCentralCrossRef Cassier PA, Lefranc A, Amela EY, Chevreau C, Bui BN, Lecesne A, Ray-Coquard I, Chabaud S, Penel N, Berge Y, et al. A phase II trial of panobinostat in patients with advanced pretreated soft tissue sarcoma. A study from the French Sarcoma Group. Br J Cancer. 2013;109:909–14.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Witta SE, Jotte RM, Konduri K, Neubauer MA, Spira AI, Ruxer RL, Varella-Garcia M, Bunn Jr PA, Hirsch FR. Randomized phase II trial of erlotinib with and without entinostat in patients with advanced non-small-cell lung cancer who progressed on prior chemotherapy. J Clin Oncol. 2012;30:2248–55.PubMedPubMedCentralCrossRef Witta SE, Jotte RM, Konduri K, Neubauer MA, Spira AI, Ruxer RL, Varella-Garcia M, Bunn Jr PA, Hirsch FR. Randomized phase II trial of erlotinib with and without entinostat in patients with advanced non-small-cell lung cancer who progressed on prior chemotherapy. J Clin Oncol. 2012;30:2248–55.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Yardley DA, Ismail-Khan RR, Melichar B, Lichinitser M, Munster PN, Klein PM, Cruickshank S, Miller KD, Lee MJ, Trepel JB. Randomized phase II, double-blind, placebo-controlled study of exemestane with or without entinostat in postmenopausal women with locally recurrent or metastatic estrogen receptor-positive breast cancer progressing on treatment with a nonsteroidal aromatase inhibitor. J Clin Oncol. 2013;31:2128–35.PubMedPubMedCentralCrossRef Yardley DA, Ismail-Khan RR, Melichar B, Lichinitser M, Munster PN, Klein PM, Cruickshank S, Miller KD, Lee MJ, Trepel JB. Randomized phase II, double-blind, placebo-controlled study of exemestane with or without entinostat in postmenopausal women with locally recurrent or metastatic estrogen receptor-positive breast cancer progressing on treatment with a nonsteroidal aromatase inhibitor. J Clin Oncol. 2013;31:2128–35.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Yeo W, Chung HC, Chan SL, Wang LZ, Lim R, Picus J, Boyer M, Mo FK, Koh J, Rha SY, et al. Epigenetic therapy using belinostat for patients with unresectable hepatocellular carcinoma: a multicenter phase I/II study with biomarker and pharmacokinetic analysis of tumors from patients in the mayo phase II consortium and the cancer therapeutics research group. J Clin Oncol. 2012;30:3361–7.PubMedPubMedCentralCrossRef Yeo W, Chung HC, Chan SL, Wang LZ, Lim R, Picus J, Boyer M, Mo FK, Koh J, Rha SY, et al. Epigenetic therapy using belinostat for patients with unresectable hepatocellular carcinoma: a multicenter phase I/II study with biomarker and pharmacokinetic analysis of tumors from patients in the mayo phase II consortium and the cancer therapeutics research group. J Clin Oncol. 2012;30:3361–7.PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat Dizon DS, Blessing JA, Penson RT, Drake RD, Walker JL, Johnston CM, Disilvestro PA, Fader AN. a phase ii evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a gynecologic oncology group study. Gynecol Oncol. 2012;125:367–71.PubMedPubMedCentralCrossRef Dizon DS, Blessing JA, Penson RT, Drake RD, Walker JL, Johnston CM, Disilvestro PA, Fader AN. a phase ii evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a gynecologic oncology group study. Gynecol Oncol. 2012;125:367–71.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Zorzi AP, Bernstein M, Samson Y, Wall DA, Desai S, Nicksy D, Wainman N, Eisenhauer E, Baruchel S. A phase I study of histone deacetylase inhibitor, pracinostat (SB939), in pediatric patients with refractory solid tumors: IND203 a trial of the NCIC IND program/C17 pediatric phase I consortium. Pediatr Blood Cancer. 2013;60:1868–74.PubMedCrossRef Zorzi AP, Bernstein M, Samson Y, Wall DA, Desai S, Nicksy D, Wainman N, Eisenhauer E, Baruchel S. A phase I study of histone deacetylase inhibitor, pracinostat (SB939), in pediatric patients with refractory solid tumors: IND203 a trial of the NCIC IND program/C17 pediatric phase I consortium. Pediatr Blood Cancer. 2013;60:1868–74.PubMedCrossRef
161.
Zurück zum Zitat Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, McDermott U, Azizian N, Zou L, Fischbach MA, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141:69–80.PubMedPubMedCentralCrossRef Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, McDermott U, Azizian N, Zou L, Fischbach MA, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141:69–80.PubMedPubMedCentralCrossRef
162.
Zurück zum Zitat Gottlicher M, Minucci S, Zhu P, Kramer OH, Schimpf A, Giavara S, Sleeman JP, Lo Coco F, Nervi C, Pelicci PG, Heinzel T. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. Embo j. 2001;20:6969–78.PubMedPubMedCentralCrossRef Gottlicher M, Minucci S, Zhu P, Kramer OH, Schimpf A, Giavara S, Sleeman JP, Lo Coco F, Nervi C, Pelicci PG, Heinzel T. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. Embo j. 2001;20:6969–78.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Travaglini L, Vian L, Billi M, Grignani F, Nervi C. Epigenetic reprogramming of breast cancer cells by valproic acid occurs regardless of estrogen receptor status. Int J Biochem Cell Biol. 2009;41:225–34.PubMedCrossRef Travaglini L, Vian L, Billi M, Grignani F, Nervi C. Epigenetic reprogramming of breast cancer cells by valproic acid occurs regardless of estrogen receptor status. Int J Biochem Cell Biol. 2009;41:225–34.PubMedCrossRef
164.
Zurück zum Zitat Schech A, Kazi A, Yu S, Shah P, Sabnis G. Histone Deacetylase Inhibitor Entinostat Inhibits Tumor-Initiating Cells in Triple-Negative Breast Cancer Cells. Mol Cancer Ther. 2015;14:1848–57.PubMedCrossRef Schech A, Kazi A, Yu S, Shah P, Sabnis G. Histone Deacetylase Inhibitor Entinostat Inhibits Tumor-Initiating Cells in Triple-Negative Breast Cancer Cells. Mol Cancer Ther. 2015;14:1848–57.PubMedCrossRef
165.
Zurück zum Zitat Song Y, Wu F, Wu J. Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J Hematol Oncol. 2016;9:49.PubMedPubMedCentralCrossRef Song Y, Wu F, Wu J. Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J Hematol Oncol. 2016;9:49.PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat Copeland RA, Solomon ME, Richon VM. Protein methyltransferases as a target class for drug discovery. Nat Rev Drug Discov. 2009;8:724–32.PubMedCrossRef Copeland RA, Solomon ME, Richon VM. Protein methyltransferases as a target class for drug discovery. Nat Rev Drug Discov. 2009;8:724–32.PubMedCrossRef
168.
Zurück zum Zitat Bhaumik SR, Smith E, Shilatifard A. Covalent modifications of histones during development and disease pathogenesis. Nat Struct Mol Biol. 2007;14:1008–16.PubMedCrossRef Bhaumik SR, Smith E, Shilatifard A. Covalent modifications of histones during development and disease pathogenesis. Nat Struct Mol Biol. 2007;14:1008–16.PubMedCrossRef
169.
Zurück zum Zitat Basavapathruni A, Jin L, Daigle SR, Majer CR, Therkelsen CA, Wigle TJ, Kuntz KW, Chesworth R, Pollock RM, Scott MP, et al. Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L. Chem Biol Drug Des. 2012;80:971–80.PubMedCrossRef Basavapathruni A, Jin L, Daigle SR, Majer CR, Therkelsen CA, Wigle TJ, Kuntz KW, Chesworth R, Pollock RM, Scott MP, et al. Conformational adaptation drives potent, selective and durable inhibition of the human protein methyltransferase DOT1L. Chem Biol Drug Des. 2012;80:971–80.PubMedCrossRef
170.
Zurück zum Zitat Liu Y, Liu K, Qin S, Xu C, Min J. Epigenetic targets and drug discovery: part 1: histone methylation. Pharmacol Ther. 2014;143:275–94.PubMedCrossRef Liu Y, Liu K, Qin S, Xu C, Min J. Epigenetic targets and drug discovery: part 1: histone methylation. Pharmacol Ther. 2014;143:275–94.PubMedCrossRef
171.
Zurück zum Zitat Liu K, Liu Y, Lau JL, Min J. Epigenetic targets and drug discovery Part 2: Histone demethylation and DNA methylation. Pharmacol Ther. 2015;151:121–40.PubMedCrossRef Liu K, Liu Y, Lau JL, Min J. Epigenetic targets and drug discovery Part 2: Histone demethylation and DNA methylation. Pharmacol Ther. 2015;151:121–40.PubMedCrossRef
172.
Zurück zum Zitat McGrath J, Trojer P. Targeting histone lysine methylation in cancer. Pharmacol Ther. 2015;150:1–22.PubMedCrossRef McGrath J, Trojer P. Targeting histone lysine methylation in cancer. Pharmacol Ther. 2015;150:1–22.PubMedCrossRef
173.
Zurück zum Zitat Copeland RA, Moyer MP, Richon VM. Targeting genetic alterations in protein methyltransferases for personalized cancer therapeutics. Oncogene. 2013;32:939–46.PubMedCrossRef Copeland RA, Moyer MP, Richon VM. Targeting genetic alterations in protein methyltransferases for personalized cancer therapeutics. Oncogene. 2013;32:939–46.PubMedCrossRef
174.
Zurück zum Zitat Steger DJ, Lefterova MI, Ying L, Stonestrom AJ, Schupp M, Zhuo D, Vakoc AL, Kim JE, Chen J, Lazar MA, et al. DOT1L/KMT4 recruitment and H3K79 methylation are ubiquitously coupled with gene transcription in mammalian cells. Mol Cell Biol. 2008;28:2825–39.PubMedPubMedCentralCrossRef Steger DJ, Lefterova MI, Ying L, Stonestrom AJ, Schupp M, Zhuo D, Vakoc AL, Kim JE, Chen J, Lazar MA, et al. DOT1L/KMT4 recruitment and H3K79 methylation are ubiquitously coupled with gene transcription in mammalian cells. Mol Cell Biol. 2008;28:2825–39.PubMedPubMedCentralCrossRef
175.
Zurück zum Zitat Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, Cui K, Roh TY, Peng W, Zhang MQ, Zhao K. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet. 2008;40:897–903.PubMedPubMedCentralCrossRef Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, Cui K, Roh TY, Peng W, Zhang MQ, Zhao K. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet. 2008;40:897–903.PubMedPubMedCentralCrossRef
176.
Zurück zum Zitat Mueller D, Bach C, Zeisig D, Garcia-Cuellar MP, Monroe S, Sreekumar A, Zhou R, Nesvizhskii A, Chinnaiyan A, Hess JL, Slany RK. A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood. 2007;110:4445–54.PubMedPubMedCentralCrossRef Mueller D, Bach C, Zeisig D, Garcia-Cuellar MP, Monroe S, Sreekumar A, Zhou R, Nesvizhskii A, Chinnaiyan A, Hess JL, Slany RK. A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood. 2007;110:4445–54.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Bitoun E, Oliver PL, Davies KE. The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling. Hum Mol Genet. 2007;16:92–106.PubMedCrossRef Bitoun E, Oliver PL, Davies KE. The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling. Hum Mol Genet. 2007;16:92–106.PubMedCrossRef
178.
Zurück zum Zitat Zhang W, Xia X, Reisenauer MR, Hemenway CS, Kone BC. Dot1a-AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner. J Biol Chem. 2006;281:18059–68.PubMedPubMedCentralCrossRef Zhang W, Xia X, Reisenauer MR, Hemenway CS, Kone BC. Dot1a-AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner. J Biol Chem. 2006;281:18059–68.PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Okada Y, Feng Q, Lin Y, Jiang Q, Li Y, Coffield VM, Su L, Xu G, Zhang Y. hDOT1L links histone methylation to leukemogenesis. Cell. 2005;121:167–78.PubMedCrossRef Okada Y, Feng Q, Lin Y, Jiang Q, Li Y, Coffield VM, Su L, Xu G, Zhang Y. hDOT1L links histone methylation to leukemogenesis. Cell. 2005;121:167–78.PubMedCrossRef
180.
Zurück zum Zitat Krivtsov AV, Feng Z, Lemieux ME, Faber J, Vempati S, Sinha AU, Xia X, Jesneck J, Bracken AP, Silverman LB, et al. H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell. 2008;14:355–68.PubMedPubMedCentralCrossRef Krivtsov AV, Feng Z, Lemieux ME, Faber J, Vempati S, Sinha AU, Xia X, Jesneck J, Bracken AP, Silverman LB, et al. H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell. 2008;14:355–68.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Daigle SR, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J, Johnston LD, Scott MP, Smith JJ, Xiao Y, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20:53–65.PubMedPubMedCentralCrossRef Daigle SR, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J, Johnston LD, Scott MP, Smith JJ, Xiao Y, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20:53–65.PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat Daigle SR, Olhava EJ, Therkelsen CA, Basavapathruni A, Jin L, Boriack-Sjodin PA, Allain CJ, Klaus CR, Raimondi A, Scott MP, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood. 2013;122:1017–25.PubMedPubMedCentralCrossRef Daigle SR, Olhava EJ, Therkelsen CA, Basavapathruni A, Jin L, Boriack-Sjodin PA, Allain CJ, Klaus CR, Raimondi A, Scott MP, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood. 2013;122:1017–25.PubMedPubMedCentralCrossRef
183.
Zurück zum Zitat Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298:1039–43.PubMedCrossRef Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298:1039–43.PubMedCrossRef
184.
Zurück zum Zitat Muller J, Hart CM, Francis NJ, Vargas ML, Sengupta A, Wild B, Miller EL, O’Connor MB, Kingston RE, Simon JA. Histone methyltransferase activity of a Drosophila Polycomb group repressor complex. Cell. 2002;111:197–208.PubMedCrossRef Muller J, Hart CM, Francis NJ, Vargas ML, Sengupta A, Wild B, Miller EL, O’Connor MB, Kingston RE, Simon JA. Histone methyltransferase activity of a Drosophila Polycomb group repressor complex. Cell. 2002;111:197–208.PubMedCrossRef
185.
Zurück zum Zitat Kuzmichev A, Nishioka K, Erdjument-Bromage H, Tempst P, Reinberg D. Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes Dev. 2002;16:2893–905.PubMedPubMedCentralCrossRef Kuzmichev A, Nishioka K, Erdjument-Bromage H, Tempst P, Reinberg D. Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes Dev. 2002;16:2893–905.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Wang X, Zhao H, Lv L, Bao L, Wang X, Han S. Prognostic Significance of EZH2 Expression in Non-Small Cell Lung Cancer: A Meta-analysis. Sci Rep. 2016;6:19239.PubMedPubMedCentralCrossRef Wang X, Zhao H, Lv L, Bao L, Wang X, Han S. Prognostic Significance of EZH2 Expression in Non-Small Cell Lung Cancer: A Meta-analysis. Sci Rep. 2016;6:19239.PubMedPubMedCentralCrossRef
187.
Zurück zum Zitat Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RG, Otte AP, Hayes DF, et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A. 2003;100:11606–11.PubMedPubMedCentralCrossRef Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RG, Otte AP, Hayes DF, et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A. 2003;100:11606–11.PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647:21–9.PubMedCrossRef Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647:21–9.PubMedCrossRef
189.
Zurück zum Zitat Li Z, Wang Y, Qiu J, Li Q, Yuan C, Zhang W, Wang D, Ye J, Jiang H, Yang J, Cheng J. The polycomb group protein EZH2 is a novel therapeutic target in tongue cancer. Oncotarget. 2013;4:2532–49.PubMedPubMedCentralCrossRef Li Z, Wang Y, Qiu J, Li Q, Yuan C, Zhang W, Wang D, Ye J, Jiang H, Yang J, Cheng J. The polycomb group protein EZH2 is a novel therapeutic target in tongue cancer. Oncotarget. 2013;4:2532–49.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, Ghosh D, Pienta KJ, Sewalt RG, Otte AP, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419:624–9.PubMedCrossRef Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, Ghosh D, Pienta KJ, Sewalt RG, Otte AP, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419:624–9.PubMedCrossRef
191.
Zurück zum Zitat Weikert S, Christoph F, Kollermann J, Muller M, Schrader M, Miller K, Krause H. Expression levels of the EZH2 polycomb transcriptional repressor correlate with aggressiveness and invasive potential of bladder carcinomas. Int J Mol Med. 2005;16:349–53.PubMed Weikert S, Christoph F, Kollermann J, Muller M, Schrader M, Miller K, Krause H. Expression levels of the EZH2 polycomb transcriptional repressor correlate with aggressiveness and invasive potential of bladder carcinomas. Int J Mol Med. 2005;16:349–53.PubMed
192.
Zurück zum Zitat Shi J, Wang E, Zuber J, Rappaport A, Taylor M, Johns C, Lowe SW, Vakoc CR. The Polycomb complex PRC2 supports aberrant self-renewal in a mouse model of MLL-AF9;Nras(G12D) acute myeloid leukemia. Oncogene. 2013;32:930–8.PubMedCrossRef Shi J, Wang E, Zuber J, Rappaport A, Taylor M, Johns C, Lowe SW, Vakoc CR. The Polycomb complex PRC2 supports aberrant self-renewal in a mouse model of MLL-AF9;Nras(G12D) acute myeloid leukemia. Oncogene. 2013;32:930–8.PubMedCrossRef
193.
Zurück zum Zitat Suva ML, Riggi N, Janiszewska M, Radovanovic I, Provero P, Stehle JC, Baumer K, Le Bitoux MA, Marino D, Cironi L, et al. EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res. 2009;69:9211–8.PubMedCrossRef Suva ML, Riggi N, Janiszewska M, Radovanovic I, Provero P, Stehle JC, Baumer K, Le Bitoux MA, Marino D, Cironi L, et al. EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res. 2009;69:9211–8.PubMedCrossRef
194.
Zurück zum Zitat Bracken AP, Dietrich N, Pasini D, Hansen KH, Helin K. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev. 2006;20:1123–36.PubMedPubMedCentralCrossRef Bracken AP, Dietrich N, Pasini D, Hansen KH, Helin K. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev. 2006;20:1123–36.PubMedPubMedCentralCrossRef
195.
Zurück zum Zitat Miranda TB, Cortez CC, Yoo CB, Liang G, Abe M, Kelly TK, Marquez VE, Jones PA. DZNep is a global histone methylation inhibitor that reactivates developmental genes not silenced by DNA methylation. Mol Cancer Ther. 2009;8:1579–88.PubMedPubMedCentralCrossRef Miranda TB, Cortez CC, Yoo CB, Liang G, Abe M, Kelly TK, Marquez VE, Jones PA. DZNep is a global histone methylation inhibitor that reactivates developmental genes not silenced by DNA methylation. Mol Cancer Ther. 2009;8:1579–88.PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat Knutson SK, Warholic NM, Wigle TJ, Klaus CR, Allain CJ, Raimondi A, Porter Scott M, Chesworth R, Moyer MP, Copeland RA, et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A. 2013;110:7922–7.PubMedPubMedCentralCrossRef Knutson SK, Warholic NM, Wigle TJ, Klaus CR, Allain CJ, Raimondi A, Porter Scott M, Chesworth R, Moyer MP, Copeland RA, et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A. 2013;110:7922–7.PubMedPubMedCentralCrossRef
197.
Zurück zum Zitat Knutson SK, Kawano S, Minoshima Y, Warholic NM, Huang KC, Xiao Y, Kadowaki T, Uesugi M, Kuznetsov G, Kumar N, et al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol Cancer Ther. 2014;13:842–54.PubMedCrossRef Knutson SK, Kawano S, Minoshima Y, Warholic NM, Huang KC, Xiao Y, Kadowaki T, Uesugi M, Kuznetsov G, Kumar N, et al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol Cancer Ther. 2014;13:842–54.PubMedCrossRef
198.
Zurück zum Zitat McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, Liu Y, Graves AP, Della Pietra 3rd A, Diaz E, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012;492:108–12.PubMedCrossRef McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, Liu Y, Graves AP, Della Pietra 3rd A, Diaz E, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012;492:108–12.PubMedCrossRef
199.
Zurück zum Zitat Chen YT, Zhu F, Lin WR, Ying RB, Yang YP, Zeng LH. The novel EZH2 inhibitor, GSK126, suppresses cell migration and angiogenesis via down-regulating VEGF-A. Cancer Chemother Pharmacol. 2016;77:757–65.PubMedCrossRef Chen YT, Zhu F, Lin WR, Ying RB, Yang YP, Zeng LH. The novel EZH2 inhibitor, GSK126, suppresses cell migration and angiogenesis via down-regulating VEGF-A. Cancer Chemother Pharmacol. 2016;77:757–65.PubMedCrossRef
200.
Zurück zum Zitat Van Aller GS, Pappalardi MB, Ott HM, Diaz E, Brandt M, Schwartz BJ, Miller WH, Dhanak D, McCabe MT, Verma SK, et al. Long residence time inhibition of EZH2 in activated polycomb repressive complex 2. ACS Chem Biol. 2014;9:622–9.PubMedCrossRef Van Aller GS, Pappalardi MB, Ott HM, Diaz E, Brandt M, Schwartz BJ, Miller WH, Dhanak D, McCabe MT, Verma SK, et al. Long residence time inhibition of EZH2 in activated polycomb repressive complex 2. ACS Chem Biol. 2014;9:622–9.PubMedCrossRef
201.
Zurück zum Zitat Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, Fukuda M, Takeda N, Niida H, Kato H, Shinkai Y. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 2002;16:1779–91.PubMedPubMedCentralCrossRef Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, Fukuda M, Takeda N, Niida H, Kato H, Shinkai Y. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 2002;16:1779–91.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Huang J, Dorsey J, Chuikov S, Perez-Burgos L, Zhang X, Jenuwein T, Reinberg D, Berger SL. G9a and Glp methylate lysine 373 in the tumor suppressor p53. J Biol Chem. 2010;285:9636–41.PubMedPubMedCentralCrossRef Huang J, Dorsey J, Chuikov S, Perez-Burgos L, Zhang X, Jenuwein T, Reinberg D, Berger SL. G9a and Glp methylate lysine 373 in the tumor suppressor p53. J Biol Chem. 2010;285:9636–41.PubMedPubMedCentralCrossRef
203.
Zurück zum Zitat Kondo Y, Shen L, Ahmed S, Boumber Y, Sekido Y, Haddad BR, Issa JP. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS One. 2008;3:e2037.PubMedPubMedCentralCrossRef Kondo Y, Shen L, Ahmed S, Boumber Y, Sekido Y, Haddad BR, Issa JP. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS One. 2008;3:e2037.PubMedPubMedCentralCrossRef
204.
Zurück zum Zitat Kondo Y, Shen L, Suzuki S, Kurokawa T, Masuko K, Tanaka Y, Kato H, Mizuno Y, Yokoe M, Sugauchi F, et al. Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol Res. 2007;37:974–83.PubMedCrossRef Kondo Y, Shen L, Suzuki S, Kurokawa T, Masuko K, Tanaka Y, Kato H, Mizuno Y, Yokoe M, Sugauchi F, et al. Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol Res. 2007;37:974–83.PubMedCrossRef
205.
Zurück zum Zitat Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, Shiah SG, Chen PS, Jeng YM, Cheng TY, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 2010;70:7830–40.PubMedCrossRef Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, Shiah SG, Chen PS, Jeng YM, Cheng TY, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 2010;70:7830–40.PubMedCrossRef
206.
Zurück zum Zitat Cho HS, Kelly JD, Hayami S, Toyokawa G, Takawa M, Yoshimatsu M, Tsunoda T, Field HI, Neal DE, Ponder BA, et al. Enhanced expression of EHMT2 is involved in the proliferation of cancer cells through negative regulation of SIAH1. Neoplasia. 2011;13:676–84.PubMedPubMedCentralCrossRef Cho HS, Kelly JD, Hayami S, Toyokawa G, Takawa M, Yoshimatsu M, Tsunoda T, Field HI, Neal DE, Ponder BA, et al. Enhanced expression of EHMT2 is involved in the proliferation of cancer cells through negative regulation of SIAH1. Neoplasia. 2011;13:676–84.PubMedPubMedCentralCrossRef
207.
Zurück zum Zitat Hua KT, Wang MY, Chen MW, Wei LH, Chen CK, Ko CH, Jeng YM, Sung PL, Jan YH, Hsiao M, et al. The H3K9 methyltransferase G9a is a marker of aggressive ovarian cancer that promotes peritoneal metastasis. Mol Cancer. 2014;13:189.PubMedPubMedCentralCrossRef Hua KT, Wang MY, Chen MW, Wei LH, Chen CK, Ko CH, Jeng YM, Sung PL, Jan YH, Hsiao M, et al. The H3K9 methyltransferase G9a is a marker of aggressive ovarian cancer that promotes peritoneal metastasis. Mol Cancer. 2014;13:189.PubMedPubMedCentralCrossRef
208.
Zurück zum Zitat Bai K, Cao Y, Huang C, Chen J, Zhang X, Jiang Y. Association of Histone Methyltransferase G9a and Overall Survival After Liver Resection of Patients With Hepatocellular Carcinoma With a Median Observation of 40 Months. Medicine (Baltimore). 2016;95:e2493.CrossRef Bai K, Cao Y, Huang C, Chen J, Zhang X, Jiang Y. Association of Histone Methyltransferase G9a and Overall Survival After Liver Resection of Patients With Hepatocellular Carcinoma With a Median Observation of 40 Months. Medicine (Baltimore). 2016;95:e2493.CrossRef
209.
Zurück zum Zitat Kubicek S, O’Sullivan RJ, August EM, Hickey ER, Zhang Q, Teodoro ML, Rea S, Mechtler K, Kowalski JA, Homon CA, et al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol Cell. 2007;25:473–81.PubMedCrossRef Kubicek S, O’Sullivan RJ, August EM, Hickey ER, Zhang Q, Teodoro ML, Rea S, Mechtler K, Kowalski JA, Homon CA, et al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol Cell. 2007;25:473–81.PubMedCrossRef
210.
Zurück zum Zitat Trojer P, Zhang J, Yonezawa M, Schmidt A, Zheng H, Jenuwein T, Reinberg D. Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins. J Biol Chem. 2009;284:8395–405.PubMedPubMedCentralCrossRef Trojer P, Zhang J, Yonezawa M, Schmidt A, Zheng H, Jenuwein T, Reinberg D. Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins. J Biol Chem. 2009;284:8395–405.PubMedPubMedCentralCrossRef
211.
Zurück zum Zitat Maze I, Covington 3rd HE, Dietz DM, LaPlant Q, Renthal W, Russo SJ, Mechanic M, Mouzon E, Neve RL, Haggarty SJ, et al. Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science. 2010;327:213–6.PubMedPubMedCentralCrossRef Maze I, Covington 3rd HE, Dietz DM, LaPlant Q, Renthal W, Russo SJ, Mechanic M, Mouzon E, Neve RL, Haggarty SJ, et al. Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science. 2010;327:213–6.PubMedPubMedCentralCrossRef
212.
Zurück zum Zitat Kim Y, Kim YS, Kim DE, Lee JS, Song JH, Kim HG, Cho DH, Jeong SY, Jin DH, Jang SJ, et al. BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production. Autophagy. 2013;9:2126–39.PubMedCrossRef Kim Y, Kim YS, Kim DE, Lee JS, Song JH, Kim HG, Cho DH, Jeong SY, Jin DH, Jang SJ, et al. BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production. Autophagy. 2013;9:2126–39.PubMedCrossRef
213.
Zurück zum Zitat Vedadi M, Barsyte-Lovejoy D, Liu F, Rival-Gervier S, Allali-Hassani A, Labrie V, Wigle TJ, Dimaggio PA, Wasney GA, Siarheyeva A, et al. A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells. Nat Chem Biol. 2011;7:566–74.PubMedPubMedCentralCrossRef Vedadi M, Barsyte-Lovejoy D, Liu F, Rival-Gervier S, Allali-Hassani A, Labrie V, Wigle TJ, Dimaggio PA, Wasney GA, Siarheyeva A, et al. A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells. Nat Chem Biol. 2011;7:566–74.PubMedPubMedCentralCrossRef
214.
Zurück zum Zitat Liu F, Barsyte-Lovejoy D, Li F, Xiong Y, Korboukh V, Huang XP, Allali-Hassani A, Janzen WP, Roth BL, Frye SV, et al. Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP. J Med Chem. 2013;56:8931–42.PubMedCrossRef Liu F, Barsyte-Lovejoy D, Li F, Xiong Y, Korboukh V, Huang XP, Allali-Hassani A, Janzen WP, Roth BL, Frye SV, et al. Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP. J Med Chem. 2013;56:8931–42.PubMedCrossRef
215.
Zurück zum Zitat Lakshmikuttyamma A, Scott SA, DeCoteau JF, Geyer CR. Reexpression of epigenetically silenced AML tumor suppressor genes by SUV39H1 inhibition. Oncogene. 2010;29:576–88.PubMedCrossRef Lakshmikuttyamma A, Scott SA, DeCoteau JF, Geyer CR. Reexpression of epigenetically silenced AML tumor suppressor genes by SUV39H1 inhibition. Oncogene. 2010;29:576–88.PubMedCrossRef
216.
Zurück zum Zitat Devin J, Viziteu E, Herviou L, Seckinger A, Camille G, Goldschmidt H, Vincent L, Pasero P, Hose D, Klein B, Moreaux J. Inhibition of SUV39H Methyltransferase As a Potent Therapeutic Target in Multiple Myeloma. Blood. 2015;126:1771. Devin J, Viziteu E, Herviou L, Seckinger A, Camille G, Goldschmidt H, Vincent L, Pasero P, Hose D, Klein B, Moreaux J. Inhibition of SUV39H Methyltransferase As a Potent Therapeutic Target in Multiple Myeloma. Blood. 2015;126:1771.
217.
Zurück zum Zitat Lai YS, Chen JY, Tsai HJ, Chen TY, Hung WC. The SUV39H1 inhibitor chaetocin induces differentiation and shows synergistic cytotoxicity with other epigenetic drugs in acute myeloid leukemia cells. Blood Cancer J. 2015;5:e313.PubMedPubMedCentralCrossRef Lai YS, Chen JY, Tsai HJ, Chen TY, Hung WC. The SUV39H1 inhibitor chaetocin induces differentiation and shows synergistic cytotoxicity with other epigenetic drugs in acute myeloid leukemia cells. Blood Cancer J. 2015;5:e313.PubMedPubMedCentralCrossRef
218.
Zurück zum Zitat Chiba T, Saito T, Yuki K, Zen Y, Koide S, Kanogawa N, Motoyama T, Ogasawara S, Suzuki E, Ooka Y, et al. Histone lysine methyltransferase SUV39H1 is a potent target for epigenetic therapy of hepatocellular carcinoma. Int J Cancer. 2015;136:289–98.PubMedCrossRef Chiba T, Saito T, Yuki K, Zen Y, Koide S, Kanogawa N, Motoyama T, Ogasawara S, Suzuki E, Ooka Y, et al. Histone lysine methyltransferase SUV39H1 is a potent target for epigenetic therapy of hepatocellular carcinoma. Int J Cancer. 2015;136:289–98.PubMedCrossRef
220.
Zurück zum Zitat Hojfeldt JW, Agger K, Helin K. Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discov. 2013;12:917–30.PubMedCrossRef Hojfeldt JW, Agger K, Helin K. Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discov. 2013;12:917–30.PubMedCrossRef
221.
Zurück zum Zitat Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11:384–400.PubMedCrossRef Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11:384–400.PubMedCrossRef
222.
Zurück zum Zitat Klose RJ, Kallin EM, Zhang Y. JmjC-domain-containing proteins and histone demethylation. Nat Rev Genet. 2006;7:715–27.PubMedCrossRef Klose RJ, Kallin EM, Zhang Y. JmjC-domain-containing proteins and histone demethylation. Nat Rev Genet. 2006;7:715–27.PubMedCrossRef
223.
Zurück zum Zitat Harris WJ, Huang X, Lynch JT, Spencer GJ, Hitchin JR, Li Y, Ciceri F, Blaser JG, Greystoke BF, Jordan AM, et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell. 2012;21:473–87.PubMedCrossRef Harris WJ, Huang X, Lynch JT, Spencer GJ, Hitchin JR, Li Y, Ciceri F, Blaser JG, Greystoke BF, Jordan AM, et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell. 2012;21:473–87.PubMedCrossRef
224.
Zurück zum Zitat Konovalov S, Garcia-Bassets I. Analysis of the levels of lysine-specific demethylase 1 (LSD1) mRNA in human ovarian tumors and the effects of chemical LSD1 inhibitors in ovarian cancer cell lines. J Ovarian Res. 2013;6:75.PubMedPubMedCentralCrossRef Konovalov S, Garcia-Bassets I. Analysis of the levels of lysine-specific demethylase 1 (LSD1) mRNA in human ovarian tumors and the effects of chemical LSD1 inhibitors in ovarian cancer cell lines. J Ovarian Res. 2013;6:75.PubMedPubMedCentralCrossRef
225.
Zurück zum Zitat Lynch JT, Harris WJ, Somervaille TC. LSD1 inhibition: a therapeutic strategy in cancer? Expert Opin Ther Targets. 2012;16:1239–49.PubMedCrossRef Lynch JT, Harris WJ, Somervaille TC. LSD1 inhibition: a therapeutic strategy in cancer? Expert Opin Ther Targets. 2012;16:1239–49.PubMedCrossRef
226.
Zurück zum Zitat Schenk T, Chen WC, Gollner S, Howell L, Jin L, Hebestreit K, Klein HU, Popescu AC, Burnett A, Mills K, et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med. 2012;18:605–11.PubMedPubMedCentralCrossRef Schenk T, Chen WC, Gollner S, Howell L, Jin L, Hebestreit K, Klein HU, Popescu AC, Burnett A, Mills K, et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med. 2012;18:605–11.PubMedPubMedCentralCrossRef
227.
Zurück zum Zitat Maes T, Tirapu I, Mascaró C, Ortega A, Estiarte A, Valls N, Castro-Palomino J, Arjol CB, Kurz G. Preclinical characterization of a potent and selective inhibitor of the histone demethylase KDM1A for MLL leukemia. J Clin Oncol. 2013;31:e13543. Maes T, Tirapu I, Mascaró C, Ortega A, Estiarte A, Valls N, Castro-Palomino J, Arjol CB, Kurz G. Preclinical characterization of a potent and selective inhibitor of the histone demethylase KDM1A for MLL leukemia. J Clin Oncol. 2013;31:e13543.
228.
Zurück zum Zitat Mohammad H, Smitheman K, Cusan M, Liu Y, Pappalardi M, Federowicz K, Van Aller G, Kasparec J, Tian X, Suarez D, et al. Inhibition Of LSD1 As a Therapeutic Strategy For The Treatment Of Acute Myeloid Leukemia. Blood. 2013;122:3964.CrossRef Mohammad H, Smitheman K, Cusan M, Liu Y, Pappalardi M, Federowicz K, Van Aller G, Kasparec J, Tian X, Suarez D, et al. Inhibition Of LSD1 As a Therapeutic Strategy For The Treatment Of Acute Myeloid Leukemia. Blood. 2013;122:3964.CrossRef
230.
Zurück zum Zitat Sprussel A, Schulte JH, Weber S, Necke M, Handschke K, Thor T, Pajtler KW, Schramm A, Konig K, Diehl L, et al. Lysine-specific demethylase 1 restricts hematopoietic progenitor proliferation and is essential for terminal differentiation. Leukemia. 2012;26:2039–51.PubMedCrossRef Sprussel A, Schulte JH, Weber S, Necke M, Handschke K, Thor T, Pajtler KW, Schramm A, Konig K, Diehl L, et al. Lysine-specific demethylase 1 restricts hematopoietic progenitor proliferation and is essential for terminal differentiation. Leukemia. 2012;26:2039–51.PubMedCrossRef
231.
Zurück zum Zitat Mohammad HP, Smitheman KN, Kamat CD, Soong D, Federowicz KE, Van Aller GS, Schneck JL, Carson JD, Liu Y, Butticello M, et al. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell. 2015;28:57–69.PubMedCrossRef Mohammad HP, Smitheman KN, Kamat CD, Soong D, Federowicz KE, Van Aller GS, Schneck JL, Carson JD, Liu Y, Butticello M, et al. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell. 2015;28:57–69.PubMedCrossRef
232.
Zurück zum Zitat Lan F, Bayliss PE, Rinn JL, Whetstine JR, Wang JK, Chen S, Iwase S, Alpatov R, Issaeva I, Canaani E, et al. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature. 2007;449:689–94.PubMedCrossRef Lan F, Bayliss PE, Rinn JL, Whetstine JR, Wang JK, Chen S, Iwase S, Alpatov R, Issaeva I, Canaani E, et al. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature. 2007;449:689–94.PubMedCrossRef
233.
Zurück zum Zitat Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rappsilber J, Issaeva I, Canaani E, Salcini AE, Helin K. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature. 2007;449:731–4.PubMedCrossRef Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rappsilber J, Issaeva I, Canaani E, Salcini AE, Helin K. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature. 2007;449:731–4.PubMedCrossRef
234.
Zurück zum Zitat Lee MG, Villa R, Trojer P, Norman J, Yan KP, Reinberg D, Di Croce L, Shiekhattar R. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science. 2007;318:447–50.PubMedCrossRef Lee MG, Villa R, Trojer P, Norman J, Yan KP, Reinberg D, Di Croce L, Shiekhattar R. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science. 2007;318:447–50.PubMedCrossRef
235.
Zurück zum Zitat Issaeva I, Zonis Y, Rozovskaia T, Orlovsky K, Croce CM, Nakamura T, Mazo A, Eisenbach L, Canaani E. Knockdown of ALR (MLL2) reveals ALR target genes and leads to alterations in cell adhesion and growth. Mol Cell Biol. 2007;27:1889–903.PubMedCrossRef Issaeva I, Zonis Y, Rozovskaia T, Orlovsky K, Croce CM, Nakamura T, Mazo A, Eisenbach L, Canaani E. Knockdown of ALR (MLL2) reveals ALR target genes and leads to alterations in cell adhesion and growth. Mol Cell Biol. 2007;27:1889–903.PubMedCrossRef
236.
Zurück zum Zitat Cho YW, Hong T, Hong S, Guo H, Yu H, Kim D, Guszczynski T, Dressler GR, Copeland TD, Kalkum M, Ge K. PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J Biol Chem. 2007;282:20395–406.PubMedPubMedCentralCrossRef Cho YW, Hong T, Hong S, Guo H, Yu H, Kim D, Guszczynski T, Dressler GR, Copeland TD, Kalkum M, Ge K. PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J Biol Chem. 2007;282:20395–406.PubMedPubMedCentralCrossRef
237.
Zurück zum Zitat Miller SA, Mohn SE, Weinmann AS. Jmjd3 and UTX play a demethylase-independent role in chromatin remodeling to regulate T-box family member-dependent gene expression. Mol Cell. 2010;40:594–605.PubMedPubMedCentralCrossRef Miller SA, Mohn SE, Weinmann AS. Jmjd3 and UTX play a demethylase-independent role in chromatin remodeling to regulate T-box family member-dependent gene expression. Mol Cell. 2010;40:594–605.PubMedPubMedCentralCrossRef
238.
Zurück zum Zitat Fu LL, Tian M, Li X, Li JJ, Huang J, Ouyang L, Zhang Y, Liu B. Inhibition of BET bromodomains as a therapeutic strategy for cancer drug discovery. Oncotarget. 2015;6:5501–16.PubMedPubMedCentralCrossRef Fu LL, Tian M, Li X, Li JJ, Huang J, Ouyang L, Zhang Y, Liu B. Inhibition of BET bromodomains as a therapeutic strategy for cancer drug discovery. Oncotarget. 2015;6:5501–16.PubMedPubMedCentralCrossRef
239.
Zurück zum Zitat Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–73.PubMedPubMedCentralCrossRef Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–73.PubMedPubMedCentralCrossRef
240.
Zurück zum Zitat Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146:904–17.PubMedPubMedCentralCrossRef Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146:904–17.PubMedPubMedCentralCrossRef
241.
Zurück zum Zitat Feng Q, Zhang Z, Shea MJ, Creighton CJ, Coarfa C, Hilsenbeck SG, Lanz R, He B, Wang L, Fu X, et al. An epigenomic approach to therapy for tamoxifen-resistant breast cancer. Cell Res. 2014;24:809–19.PubMedPubMedCentralCrossRef Feng Q, Zhang Z, Shea MJ, Creighton CJ, Coarfa C, Hilsenbeck SG, Lanz R, He B, Wang L, Fu X, et al. An epigenomic approach to therapy for tamoxifen-resistant breast cancer. Cell Res. 2014;24:809–19.PubMedPubMedCentralCrossRef
242.
Zurück zum Zitat Rajagopalan V, Vaidyanathan M, Janardhanam VA, Bradner JE. Pre-clinical analysis of changes in intra-cellular biochemistry of glioblastoma multiforme (GBM) cells due to c-Myc silencing. Cell Mol Neurobiol. 2014;34:1059–69.PubMedCrossRef Rajagopalan V, Vaidyanathan M, Janardhanam VA, Bradner JE. Pre-clinical analysis of changes in intra-cellular biochemistry of glioblastoma multiforme (GBM) cells due to c-Myc silencing. Cell Mol Neurobiol. 2014;34:1059–69.PubMedCrossRef
243.
Zurück zum Zitat Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468:1119–23.PubMedCrossRef Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468:1119–23.PubMedCrossRef
244.
Zurück zum Zitat Gosmini R, Nguyen VL, Toum J, Simon C, Brusq JM, Krysa G, Mirguet O, Riou-Eymard AM, Boursier EV, Trottet L, et al. The discovery of I-BET726 (GSK1324726A), a potent tetrahydroquinoline ApoA1 up-regulator and selective BET bromodomain inhibitor. J Med Chem. 2014;57:8111–31.PubMedCrossRef Gosmini R, Nguyen VL, Toum J, Simon C, Brusq JM, Krysa G, Mirguet O, Riou-Eymard AM, Boursier EV, Trottet L, et al. The discovery of I-BET726 (GSK1324726A), a potent tetrahydroquinoline ApoA1 up-regulator and selective BET bromodomain inhibitor. J Med Chem. 2014;57:8111–31.PubMedCrossRef
245.
Zurück zum Zitat Garnier JM, Sharp PP, Burns CJ. BET bromodomain inhibitors: a patent review. Expert Opin Ther Pat. 2014;24:185–99.PubMedCrossRef Garnier JM, Sharp PP, Burns CJ. BET bromodomain inhibitors: a patent review. Expert Opin Ther Pat. 2014;24:185–99.PubMedCrossRef
246.
Zurück zum Zitat Wyce A, Degenhardt Y, Bai Y, Le B, Korenchuk S, Crouthame MC, McHugh CF, Vessella R, Creasy CL, Tummino PJ, Barbash O. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer. Oncotarget. 2013;4:2419–29.PubMedPubMedCentralCrossRef Wyce A, Degenhardt Y, Bai Y, Le B, Korenchuk S, Crouthame MC, McHugh CF, Vessella R, Creasy CL, Tummino PJ, Barbash O. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer. Oncotarget. 2013;4:2419–29.PubMedPubMedCentralCrossRef
247.
Zurück zum Zitat Wyce A, Ganji G, Smitheman KN, Chung CW, Korenchuk S, Bai Y, Barbash O, Le B, Craggs PD, McCabe MT, et al. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models. PLoS One. 2013;8:e72967.PubMedPubMedCentralCrossRef Wyce A, Ganji G, Smitheman KN, Chung CW, Korenchuk S, Bai Y, Barbash O, Le B, Craggs PD, McCabe MT, et al. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models. PLoS One. 2013;8:e72967.PubMedPubMedCentralCrossRef
248.
Zurück zum Zitat Herait PE, Berthon C, Thieblemont C, Raffoux E, Magarotto V, Stathis A, Thomas X, Leleu X, Gomez-Roca C, Odore E, et al. Abstract CT231: BET-bromodomain inhibitor OTX015 shows clinically meaningful activity at nontoxic doses: interim results of an ongoing phase I trial in hematologic malignancies. Cancer Res. 2014;74:CT231.CrossRef Herait PE, Berthon C, Thieblemont C, Raffoux E, Magarotto V, Stathis A, Thomas X, Leleu X, Gomez-Roca C, Odore E, et al. Abstract CT231: BET-bromodomain inhibitor OTX015 shows clinically meaningful activity at nontoxic doses: interim results of an ongoing phase I trial in hematologic malignancies. Cancer Res. 2014;74:CT231.CrossRef
249.
Zurück zum Zitat Bonetti P, Ponzoni M, Tibiletti MG, Stathis A, Heirat P, Zucca E, Bertoni F. 528 The BRD-inhibitor OTX015 Shows Pre-clinical Activity in Diffuse Large B-cell Lymphoma (DLBCL). Eur J Cancer. 2012;48:163. Bonetti P, Ponzoni M, Tibiletti MG, Stathis A, Heirat P, Zucca E, Bertoni F. 528 The BRD-inhibitor OTX015 Shows Pre-clinical Activity in Diffuse Large B-cell Lymphoma (DLBCL). Eur J Cancer. 2012;48:163.
250.
Zurück zum Zitat Chaidos A, Caputo V, Gouvedenou K, Liu B, Marigo I, Chaudhry MS, Rotolo A, Tough DF, Smithers NN, Bassil AK, et al. Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762. Blood. 2014;123:697–705.PubMedCrossRef Chaidos A, Caputo V, Gouvedenou K, Liu B, Marigo I, Chaudhry MS, Rotolo A, Tough DF, Smithers NN, Bassil AK, et al. Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762. Blood. 2014;123:697–705.PubMedCrossRef
251.
Zurück zum Zitat Pathania R, Ramachandran S, Mariappan G, Thakur P, Shi H, Choi J-H, Manicassamy S, Kolhe R, Prasad PD, Sharma S, et al. Combined inhibition of DNMT and HDAC blocks the tumorigenicity of cancer stem-like cells and attenuates mammary tumor growth. Cancer Res. 2016;76:3224–35.PubMedCrossRef Pathania R, Ramachandran S, Mariappan G, Thakur P, Shi H, Choi J-H, Manicassamy S, Kolhe R, Prasad PD, Sharma S, et al. Combined inhibition of DNMT and HDAC blocks the tumorigenicity of cancer stem-like cells and attenuates mammary tumor growth. Cancer Res. 2016;76:3224–35.PubMedCrossRef
252.
Zurück zum Zitat Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, Sebree R, Rodgers K, Hooker CM, Franco N, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1:598–607.PubMedPubMedCentralCrossRef Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, Sebree R, Rodgers K, Hooker CM, Franco N, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1:598–607.PubMedPubMedCentralCrossRef
253.
Zurück zum Zitat Ecke I, Petry F, Rosenberger A, Tauber S, Monkemeyer S, Hess I, Dullin C, Kimmina S, Pirngruber J, Johnsen SA, et al. Antitumor effects of a combined 5-aza-2’deoxycytidine and valproic acid treatment on rhabdomyosarcoma and medulloblastoma in Ptch mutant mice. Cancer Res. 2009;69:887–95.PubMedCrossRef Ecke I, Petry F, Rosenberger A, Tauber S, Monkemeyer S, Hess I, Dullin C, Kimmina S, Pirngruber J, Johnsen SA, et al. Antitumor effects of a combined 5-aza-2’deoxycytidine and valproic acid treatment on rhabdomyosarcoma and medulloblastoma in Ptch mutant mice. Cancer Res. 2009;69:887–95.PubMedCrossRef
254.
Zurück zum Zitat Curry E, Green I, Chapman-Rothe N, Shamsaei E, Kandil S, Cherblanc FL, Payne L, Bell E, Ganesh T, Srimongkolpithak N, et al. Dual EZH2 and EHMT2 histone methyltransferase inhibition increases biological efficacy in breast cancer cells. Clin Epigenetics. 2015;7:84.PubMedPubMedCentralCrossRef Curry E, Green I, Chapman-Rothe N, Shamsaei E, Kandil S, Cherblanc FL, Payne L, Bell E, Ganesh T, Srimongkolpithak N, et al. Dual EZH2 and EHMT2 histone methyltransferase inhibition increases biological efficacy in breast cancer cells. Clin Epigenetics. 2015;7:84.PubMedPubMedCentralCrossRef
255.
Zurück zum Zitat Klaus CR, Iwanowicz D, Johnston D, Campbell CA, Smith JJ, Moyer MP, Copeland RA, Olhava EJ, Scott MP, Pollock RM, et al. DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells. J Pharmacol Exp Ther. 2014;350:646–56.PubMedCrossRef Klaus CR, Iwanowicz D, Johnston D, Campbell CA, Smith JJ, Moyer MP, Copeland RA, Olhava EJ, Scott MP, Pollock RM, et al. DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells. J Pharmacol Exp Ther. 2014;350:646–56.PubMedCrossRef
256.
Zurück zum Zitat Easwaran H, Tsai HC, Baylin SB. Cancer epigenetics: tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol Cell. 2014;54:716–27.PubMedPubMedCentralCrossRef Easwaran H, Tsai HC, Baylin SB. Cancer epigenetics: tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol Cell. 2014;54:716–27.PubMedPubMedCentralCrossRef
257.
Zurück zum Zitat Vatapalli RJ, Guzzetta AA, Fu T, Syed LH, Kwak R, Ahuja N. Improving Sensitivity to Irinotecan Using 5-Azacytidine in Colon Cancer Cell Lines. J Surgical Res. 2013;179:251–252. Vatapalli RJ, Guzzetta AA, Fu T, Syed LH, Kwak R, Ahuja N. Improving Sensitivity to Irinotecan Using 5-Azacytidine in Colon Cancer Cell Lines. J Surgical Res. 2013;179:251–252.
258.
Zurück zum Zitat Fu S, Hu W, Iyer R, Kavanagh JJ, Coleman RL, Levenback CF, Sood AK, Wolf JK, Gershenson DM, Markman M, et al. Phase 1b-2a study to reverse platinum resistance through use of a hypomethylating agent, azacitidine, in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer. Cancer. 2011;117:1661–9.PubMedCrossRef Fu S, Hu W, Iyer R, Kavanagh JJ, Coleman RL, Levenback CF, Sood AK, Wolf JK, Gershenson DM, Markman M, et al. Phase 1b-2a study to reverse platinum resistance through use of a hypomethylating agent, azacitidine, in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer. Cancer. 2011;117:1661–9.PubMedCrossRef
259.
Zurück zum Zitat Appleton K, Mackay HJ, Judson I, Plumb JA, McCormick C, Strathdee G, Lee C, Barrett S, Reade S, Jadayel D, et al. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol. 2007;25:4603–9.PubMedCrossRef Appleton K, Mackay HJ, Judson I, Plumb JA, McCormick C, Strathdee G, Lee C, Barrett S, Reade S, Jadayel D, et al. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol. 2007;25:4603–9.PubMedCrossRef
260.
Zurück zum Zitat Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, Berry WA, Huang T, Nephew KP. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res. 2012;72:2197–205.PubMedPubMedCentralCrossRef Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, Berry WA, Huang T, Nephew KP. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res. 2012;72:2197–205.PubMedPubMedCentralCrossRef
261.
Zurück zum Zitat Ramaswamy B, Fiskus W, Cohen B, Pellegrino C, Hershman DL, Chuang E, Luu T, Somlo G, Goetz M, Swaby R, et al. Phase I-II study of vorinostat plus paclitaxel and bevacizumab in metastatic breast cancer: evidence for vorinostat-induced tubulin acetylation and Hsp90 inhibition in vivo. Breast Cancer Res Treat. 2012;132:1063–72.PubMedCrossRef Ramaswamy B, Fiskus W, Cohen B, Pellegrino C, Hershman DL, Chuang E, Luu T, Somlo G, Goetz M, Swaby R, et al. Phase I-II study of vorinostat plus paclitaxel and bevacizumab in metastatic breast cancer: evidence for vorinostat-induced tubulin acetylation and Hsp90 inhibition in vivo. Breast Cancer Res Treat. 2012;132:1063–72.PubMedCrossRef
262.
Zurück zum Zitat Li SY, Sun R, Wang HX, Shen S, Liu Y, Du XJ, Zhu YH, Jun W. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. J Control Release. 2015;205:7–14.PubMedCrossRef Li SY, Sun R, Wang HX, Shen S, Liu Y, Du XJ, Zhu YH, Jun W. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. J Control Release. 2015;205:7–14.PubMedCrossRef
263.
264.
Zurück zum Zitat Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.PubMedPubMedCentralCrossRef Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23.PubMedPubMedCentralCrossRef
265.
Zurück zum Zitat Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy. Cancer Immunol Immunother. 2005;54:307–14.PubMedCrossRef Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy. Cancer Immunol Immunother. 2005;54:307–14.PubMedCrossRef
266.
Zurück zum Zitat Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54.PubMedPubMedCentralCrossRef Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54.PubMedPubMedCentralCrossRef
267.
Zurück zum Zitat Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65.PubMedPubMedCentralCrossRef Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65.PubMedPubMedCentralCrossRef
268.
Zurück zum Zitat Pan Q, Li Q, Liu S, Ning N, Zhang X, Xu Y, Chang AE, Wicha MS. Concise Review: Targeting Cancer Stem Cells Using Immunologic Approaches. Stem Cells. 2015;33:2085–92.PubMedPubMedCentralCrossRef Pan Q, Li Q, Liu S, Ning N, Zhang X, Xu Y, Chang AE, Wicha MS. Concise Review: Targeting Cancer Stem Cells Using Immunologic Approaches. Stem Cells. 2015;33:2085–92.PubMedPubMedCentralCrossRef
269.
Zurück zum Zitat Covre A, Coral S, Nicolay H, Parisi G, Fazio C, Colizzi F, Fratta E, Di Giacomo AM, Sigalotti L, Natali PG, Maio M. Antitumor activity of epigenetic immunomodulation combined with CTLA-4 blockade in syngeneic mouse models. Oncoimmunology. 2015;4:e1019978.PubMedPubMedCentralCrossRef Covre A, Coral S, Nicolay H, Parisi G, Fazio C, Colizzi F, Fratta E, Di Giacomo AM, Sigalotti L, Natali PG, Maio M. Antitumor activity of epigenetic immunomodulation combined with CTLA-4 blockade in syngeneic mouse models. Oncoimmunology. 2015;4:e1019978.PubMedPubMedCentralCrossRef
270.
Zurück zum Zitat Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, Diaz Jr LA, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A. 2014;111:11774–9.PubMedPubMedCentralCrossRef Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, Diaz Jr LA, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A. 2014;111:11774–9.PubMedPubMedCentralCrossRef
271.
Zurück zum Zitat Qifeng S, Bo C, Xingtao J, Chuanliang P, Xiaogang Z. Methylation of the promoter of human leukocyte antigen class I in human esophageal squamous cell carcinoma and its histopathological characteristics. J Thorac Cardiovasc Surg. 2011;141:808–14.PubMedCrossRef Qifeng S, Bo C, Xingtao J, Chuanliang P, Xiaogang Z. Methylation of the promoter of human leukocyte antigen class I in human esophageal squamous cell carcinoma and its histopathological characteristics. J Thorac Cardiovasc Surg. 2011;141:808–14.PubMedCrossRef
272.
Zurück zum Zitat Ye Q, Shen Y, Wang X, Yang J, Miao F, Shen C, Zhang J. Hypermethylation of HLA class I gene is associated with HLA class I down-regulation in human gastric cancer. Tissue Antigens. 2010;75:30–9.PubMedCrossRef Ye Q, Shen Y, Wang X, Yang J, Miao F, Shen C, Zhang J. Hypermethylation of HLA class I gene is associated with HLA class I down-regulation in human gastric cancer. Tissue Antigens. 2010;75:30–9.PubMedCrossRef
273.
Zurück zum Zitat Kitamura H, Torigoe T, Asanuma H, Honma I, Sato N, Tsukamoto T. Down-regulation of HLA class I antigens in prostate cancer tissues and up-regulation by histone deacetylase inhibition. J Urol. 2007;178:692–6.PubMedCrossRef Kitamura H, Torigoe T, Asanuma H, Honma I, Sato N, Tsukamoto T. Down-regulation of HLA class I antigens in prostate cancer tissues and up-regulation by histone deacetylase inhibition. J Urol. 2007;178:692–6.PubMedCrossRef
274.
Zurück zum Zitat Magner WJ, Kazim AL, Stewart C, Romano MA, Catalano G, Grande C, Keiser N, Santaniello F, Tomasi TB. Activation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitors. J Immunol. 2000;165:7017–24.PubMedCrossRef Magner WJ, Kazim AL, Stewart C, Romano MA, Catalano G, Grande C, Keiser N, Santaniello F, Tomasi TB. Activation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitors. J Immunol. 2000;165:7017–24.PubMedCrossRef
275.
Zurück zum Zitat Mora-Garcia Mde L, Duenas-Gonzalez A, Hernandez-Montes J, De la Cruz-Hernandez E, Perez-Cardenas E, Weiss-Steider B, Santiago-Osorio E, Ortiz-Navarrete VF, Rosales VH, Cantu D, et al. Up-regulation of HLA class-I antigen expression and antigen-specific CTL response in cervical cancer cells by the demethylating agent hydralazine and the histone deacetylase inhibitor valproic acid. J Transl Med. 2006;4:55.PubMedCrossRef Mora-Garcia Mde L, Duenas-Gonzalez A, Hernandez-Montes J, De la Cruz-Hernandez E, Perez-Cardenas E, Weiss-Steider B, Santiago-Osorio E, Ortiz-Navarrete VF, Rosales VH, Cantu D, et al. Up-regulation of HLA class-I antigen expression and antigen-specific CTL response in cervical cancer cells by the demethylating agent hydralazine and the histone deacetylase inhibitor valproic acid. J Transl Med. 2006;4:55.PubMedCrossRef
276.
Zurück zum Zitat Li H, Chiappinelli KB, Guzzetta AA, Easwaran H, Yen RW, Vatapalli R, Topper MJ, Luo J, Connolly RM, Azad NS, et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5:587–98.PubMedPubMedCentralCrossRef Li H, Chiappinelli KB, Guzzetta AA, Easwaran H, Yen RW, Vatapalli R, Topper MJ, Luo J, Connolly RM, Azad NS, et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5:587–98.PubMedPubMedCentralCrossRef
277.
Zurück zum Zitat Azad N, Zahnow CA, Rudin CM, Baylin SB. The future of epigenetic therapy in solid tumours--lessons from the past. Nat Rev Clin Oncol. 2013;10:256–66.PubMedPubMedCentralCrossRef Azad N, Zahnow CA, Rudin CM, Baylin SB. The future of epigenetic therapy in solid tumours--lessons from the past. Nat Rev Clin Oncol. 2013;10:256–66.PubMedPubMedCentralCrossRef
278.
Zurück zum Zitat Silverman LR, Fenaux P, Mufti GJ, Santini V, Hellstrom-Lindberg E, Gattermann N, Sanz G, List AF, Gore SD, Seymour JF. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117:2697–702.PubMedPubMedCentralCrossRef Silverman LR, Fenaux P, Mufti GJ, Santini V, Hellstrom-Lindberg E, Gattermann N, Sanz G, List AF, Gore SD, Seymour JF. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117:2697–702.PubMedPubMedCentralCrossRef
279.
Zurück zum Zitat Griffiths EA, Choy G, Redkar S, Taverna P, Azab M, Karpf AR. SGI-110: DNA Methyltransferase Inhibitor Oncolytic. Drugs Future. 2013;38:535–43.PubMedPubMedCentral Griffiths EA, Choy G, Redkar S, Taverna P, Azab M, Karpf AR. SGI-110: DNA Methyltransferase Inhibitor Oncolytic. Drugs Future. 2013;38:535–43.PubMedPubMedCentral
280.
Zurück zum Zitat Issa JP, Roboz G, Rizzieri D, Jabbour E, Stock W, O’Connell C, Yee K, Tibes R, Griffiths EA, Walsh K, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study. Lancet Oncol. 2015;16:1099–110.PubMedCrossRef Issa JP, Roboz G, Rizzieri D, Jabbour E, Stock W, O’Connell C, Yee K, Tibes R, Griffiths EA, Walsh K, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study. Lancet Oncol. 2015;16:1099–110.PubMedCrossRef
281.
Zurück zum Zitat Fang F, Munck J, Tang J, Taverna P, Wang Y, Miller DF, Pilrose J, Choy G, Azab M, Pawelczak KS, et al. The novel, small-molecule DNA methylation inhibitor SGI-110 as an ovarian cancer chemosensitizer. Clin Cancer Res. 2014;20:6504–16.PubMedPubMedCentralCrossRef Fang F, Munck J, Tang J, Taverna P, Wang Y, Miller DF, Pilrose J, Choy G, Azab M, Pawelczak KS, et al. The novel, small-molecule DNA methylation inhibitor SGI-110 as an ovarian cancer chemosensitizer. Clin Cancer Res. 2014;20:6504–16.PubMedPubMedCentralCrossRef
282.
Zurück zum Zitat Younes A, Sureda A, Ben-Yehuda D, Zinzani PL, Ong TC, Prince HM, Harrison SJ, Kirschbaum M, Johnston P, Gallagher J, et al. Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: results of a phase II study. J Clin Oncol. 2012;30:2197–203.PubMedCrossRef Younes A, Sureda A, Ben-Yehuda D, Zinzani PL, Ong TC, Prince HM, Harrison SJ, Kirschbaum M, Johnston P, Gallagher J, et al. Panobinostat in patients with relapsed/refractory Hodgkin’s lymphoma after autologous stem-cell transplantation: results of a phase II study. J Clin Oncol. 2012;30:2197–203.PubMedCrossRef
283.
Zurück zum Zitat Duvic M, Dummer R, Becker JC, Poulalhon N, Ortiz Romero P, Grazia Bernengo M, Lebbe C, Assaf C, Squier M, Williams D, et al. Panobinostat activity in both bexarotene-exposed and -naive patients with refractory cutaneous T-cell lymphoma: results of a phase II trial. Eur J Cancer. 2013;49:386–94.PubMedCrossRef Duvic M, Dummer R, Becker JC, Poulalhon N, Ortiz Romero P, Grazia Bernengo M, Lebbe C, Assaf C, Squier M, Williams D, et al. Panobinostat activity in both bexarotene-exposed and -naive patients with refractory cutaneous T-cell lymphoma: results of a phase II trial. Eur J Cancer. 2013;49:386–94.PubMedCrossRef
284.
Zurück zum Zitat Batlevi CL, Kasamon Y, Bociek RG, Lee P, Gore L, Copeland A, Sorensen R, Ordentlich P, Cruickshank S, Kunkel L, et al. ENGAGE- 501: phase II study of entinostat (SNDX-275) in relapsed and refractory Hodgkin lymphoma. Haematologica. 2016;101:968–75.PubMedPubMedCentralCrossRef Batlevi CL, Kasamon Y, Bociek RG, Lee P, Gore L, Copeland A, Sorensen R, Ordentlich P, Cruickshank S, Kunkel L, et al. ENGAGE- 501: phase II study of entinostat (SNDX-275) in relapsed and refractory Hodgkin lymphoma. Haematologica. 2016;101:968–75.PubMedPubMedCentralCrossRef
285.
Zurück zum Zitat Kirschbaum MH, Foon KA, Frankel P, Ruel C, Pulone B, Tuscano JM, Newman EM. A phase 2 study of belinostat (PXD101) in patients with relapsed or refractory acute myeloid leukemia or patients over the age of 60 with newly diagnosed acute myeloid leukemia: a California Cancer Consortium Study. Leuk Lymphoma. 2014;55:2301–4.PubMedPubMedCentralCrossRef Kirschbaum MH, Foon KA, Frankel P, Ruel C, Pulone B, Tuscano JM, Newman EM. A phase 2 study of belinostat (PXD101) in patients with relapsed or refractory acute myeloid leukemia or patients over the age of 60 with newly diagnosed acute myeloid leukemia: a California Cancer Consortium Study. Leuk Lymphoma. 2014;55:2301–4.PubMedPubMedCentralCrossRef
286.
Zurück zum Zitat O’Connor OA, Horwitz S, Masszi T, Van Hoof A, Brown P, Doorduijn J, Hess G, Jurczak W, Knoblauch P, Chawla S, et al. Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results of the Pivotal Phase II BELIEF (CLN-19) Study. J Clin Oncol. 2015;33:2492–9.PubMedPubMedCentralCrossRef O’Connor OA, Horwitz S, Masszi T, Van Hoof A, Brown P, Doorduijn J, Hess G, Jurczak W, Knoblauch P, Chawla S, et al. Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results of the Pivotal Phase II BELIEF (CLN-19) Study. J Clin Oncol. 2015;33:2492–9.PubMedPubMedCentralCrossRef
287.
Zurück zum Zitat Mackay HJ, Hirte H, Colgan T, Covens A, MacAlpine K, Grenci P, Wang L, Mason J, Pham PA, Tsao MS, et al. Phase II trial of the histone deacetylase inhibitor belinostat in women with platinum resistant epithelial ovarian cancer and micropapillary (LMP) ovarian tumours. Eur J Cancer. 2010;46:1573–9.PubMedPubMedCentralCrossRef Mackay HJ, Hirte H, Colgan T, Covens A, MacAlpine K, Grenci P, Wang L, Mason J, Pham PA, Tsao MS, et al. Phase II trial of the histone deacetylase inhibitor belinostat in women with platinum resistant epithelial ovarian cancer and micropapillary (LMP) ovarian tumours. Eur J Cancer. 2010;46:1573–9.PubMedPubMedCentralCrossRef
288.
Zurück zum Zitat Chu QS, Nielsen TO, Alcindor T, Gupta A, Endo M, Goytain A, Xu H, Verma S, Tozer R, Knowling M, et al. A phase II study of SB939, a novel pan-histone deacetylase inhibitor, in patients with translocation-associated recurrent/metastatic sarcomas-NCIC-CTG IND 200dagger. Ann Oncol. 2015;26:973–81.PubMedCrossRef Chu QS, Nielsen TO, Alcindor T, Gupta A, Endo M, Goytain A, Xu H, Verma S, Tozer R, Knowling M, et al. A phase II study of SB939, a novel pan-histone deacetylase inhibitor, in patients with translocation-associated recurrent/metastatic sarcomas-NCIC-CTG IND 200dagger. Ann Oncol. 2015;26:973–81.PubMedCrossRef
289.
Zurück zum Zitat Eigl BJ, North S, Winquist E, Finch D, Wood L, Sridhar SS, Powers J, Good J, Sharma M, Squire JA, et al. A phase II study of the HDAC inhibitor SB939 in patients with castration resistant prostate cancer: NCIC clinical trials group study IND195. Invest New Drugs. 2015;33:969–76.PubMedCrossRef Eigl BJ, North S, Winquist E, Finch D, Wood L, Sridhar SS, Powers J, Good J, Sharma M, Squire JA, et al. A phase II study of the HDAC inhibitor SB939 in patients with castration resistant prostate cancer: NCIC clinical trials group study IND195. Invest New Drugs. 2015;33:969–76.PubMedCrossRef
290.
Zurück zum Zitat Rambaldi A, Dellacasa CM, Finazzi G, Carobbio A, Ferrari ML, Guglielmelli P, Gattoni E, Salmoiraghi S, Finazzi MC, Di Tollo S, et al. A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. Br J Haematol. 2010;150:446–55.PubMed Rambaldi A, Dellacasa CM, Finazzi G, Carobbio A, Ferrari ML, Guglielmelli P, Gattoni E, Salmoiraghi S, Finazzi MC, Di Tollo S, et al. A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. Br J Haematol. 2010;150:446–55.PubMed
291.
Zurück zum Zitat Mawatari T, Ninomiya I, Inokuchi M, Harada S, Hayashi H, Oyama K, Makino I, Nakagawara H, Miyashita T, Tajima H, et al. Valproic acid inhibits proliferation of HER2-expressing breast cancer cells by inducing cell cycle arrest and apoptosis through Hsp70 acetylation. Int J Oncol. 2015;47:2073–81.PubMedPubMedCentral Mawatari T, Ninomiya I, Inokuchi M, Harada S, Hayashi H, Oyama K, Makino I, Nakagawara H, Miyashita T, Tajima H, et al. Valproic acid inhibits proliferation of HER2-expressing breast cancer cells by inducing cell cycle arrest and apoptosis through Hsp70 acetylation. Int J Oncol. 2015;47:2073–81.PubMedPubMedCentral
292.
Zurück zum Zitat Tan J, Yang X, Zhuang L, Jiang X, Chen W, Lee PL, Karuturi RK, Tan PB, Liu ET, Yu Q. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 2007;21:1050–63.PubMedPubMedCentralCrossRef Tan J, Yang X, Zhuang L, Jiang X, Chen W, Lee PL, Karuturi RK, Tan PB, Liu ET, Yu Q. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 2007;21:1050–63.PubMedPubMedCentralCrossRef
293.
Zurück zum Zitat Crea F, Hurt EM, Mathews LA, Cabarcas SM, Sun L, Marquez VE, Danesi R, Farrar WL. Pharmacologic disruption of Polycomb Repressive Complex 2 inhibits tumorigenicity and tumor progression in prostate cancer. Mol Cancer. 2011;10:40.PubMedPubMedCentralCrossRef Crea F, Hurt EM, Mathews LA, Cabarcas SM, Sun L, Marquez VE, Danesi R, Farrar WL. Pharmacologic disruption of Polycomb Repressive Complex 2 inhibits tumorigenicity and tumor progression in prostate cancer. Mol Cancer. 2011;10:40.PubMedPubMedCentralCrossRef
294.
Zurück zum Zitat Bradley WD, Arora S, Busby J, Balasubramanian S, Gehling VS, Nasveschuk CG, Vaswani RG, Yuan CC, Hatton C, Zhao F, et al. EZH2 inhibitor efficacy in non-Hodgkin’s lymphoma does not require suppression of H3K27 monomethylation. Chem Biol. 2014;21:1463–75.PubMedCrossRef Bradley WD, Arora S, Busby J, Balasubramanian S, Gehling VS, Nasveschuk CG, Vaswani RG, Yuan CC, Hatton C, Zhao F, et al. EZH2 inhibitor efficacy in non-Hodgkin’s lymphoma does not require suppression of H3K27 monomethylation. Chem Biol. 2014;21:1463–75.PubMedCrossRef
295.
Zurück zum Zitat Kruidenier L, Chung CW, Cheng Z, Liddle J, Che K, Joberty G, Bantscheff M, Bountra C, Bridges A, Diallo H, et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature. 2012;488:404–8.PubMedPubMedCentralCrossRef Kruidenier L, Chung CW, Cheng Z, Liddle J, Che K, Joberty G, Bantscheff M, Bountra C, Bridges A, Diallo H, et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature. 2012;488:404–8.PubMedPubMedCentralCrossRef
296.
Zurück zum Zitat Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S, Mele DA, Bergeron L, Sims 3rd RJ. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A. 2011;108:16669–74.PubMedPubMedCentralCrossRef Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S, Mele DA, Bergeron L, Sims 3rd RJ. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A. 2011;108:16669–74.PubMedPubMedCentralCrossRef
297.
Zurück zum Zitat Chapuy B, McKeown MR, Lin CY, Monti S, Roemer MG, Qi J, Rahl PB, Sun HH, Yeda KT, Doench JG, et al. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell. 2013;24:777–90.PubMedPubMedCentralCrossRef Chapuy B, McKeown MR, Lin CY, Monti S, Roemer MG, Qi J, Rahl PB, Sun HH, Yeda KT, Doench JG, et al. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell. 2013;24:777–90.PubMedPubMedCentralCrossRef
298.
Zurück zum Zitat Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf C, Savitski MM, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 2011;478:529–33.PubMedPubMedCentralCrossRef Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf C, Savitski MM, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 2011;478:529–33.PubMedPubMedCentralCrossRef
299.
Zurück zum Zitat Long J, Li B, Rodriguez-Blanco J, Pastori C, Volmar CH, Wahlestedt C, Capobianco A, Bai F, Pei XH, Ayad NG, Robbins DJ. The BET bromodomain inhibitor I-BET151 acts downstream of smoothened protein to abrogate the growth of hedgehog protein-driven cancers. J Biol Chem. 2014;289:35494–502.PubMedPubMedCentralCrossRef Long J, Li B, Rodriguez-Blanco J, Pastori C, Volmar CH, Wahlestedt C, Capobianco A, Bai F, Pei XH, Ayad NG, Robbins DJ. The BET bromodomain inhibitor I-BET151 acts downstream of smoothened protein to abrogate the growth of hedgehog protein-driven cancers. J Biol Chem. 2014;289:35494–502.PubMedPubMedCentralCrossRef
300.
Zurück zum Zitat Moros A, Rodriguez V, Saborit-Villarroya I, Montraveta A, Balsas P, Sandy P, Martinez A, Wiestner A, Normant E, Campo E, et al. Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma. Leukemia. 2014;28:2049–59.PubMed Moros A, Rodriguez V, Saborit-Villarroya I, Montraveta A, Balsas P, Sandy P, Martinez A, Wiestner A, Normant E, Campo E, et al. Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma. Leukemia. 2014;28:2049–59.PubMed
301.
Zurück zum Zitat Albrecht BK, Gehling VS, Hewitt MC, Vaswani RG, Cote A, Leblanc Y, Nasveschuk CG, Bellon S, Bergeron L, Campbell R, et al. Identification of a Benzoisoxazoloazepine Inhibitor (CPI-0610) of the Bromodomain and Extra-Terminal (BET) Family as a Candidate for Human Clinical Trials. J Med Chem. 2016;59:1330–9.PubMedCrossRef Albrecht BK, Gehling VS, Hewitt MC, Vaswani RG, Cote A, Leblanc Y, Nasveschuk CG, Bellon S, Bergeron L, Campbell R, et al. Identification of a Benzoisoxazoloazepine Inhibitor (CPI-0610) of the Bromodomain and Extra-Terminal (BET) Family as a Candidate for Human Clinical Trials. J Med Chem. 2016;59:1330–9.PubMedCrossRef
Metadaten
Titel
Epigenetics in cancer stem cells
verfasst von
Tan Boon Toh
Jhin Jieh Lim
Edward Kai-Hua Chow
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2017
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-017-0596-9

Weitere Artikel der Ausgabe 1/2017

Molecular Cancer 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.