Hairy and Enhancer-of-split related with YRPW motif (Hey) transcription factors are important regulators of stem cell embryogenesis. Clinical relevance shows that they are also highly expressed in malignant carcinoma. Recent studies have highlighted functions for the Hey factors in tumor metastasis, the maintenance of cancer cell self-renewal, as well as proliferation and the promotion of tumor angiogenesis. Pathways that regulate Hey gene expression, such as Notch and TGFβ signaling, are frequently aberrant in numerous cancers. In addition, Hey factors control downstream targets via recruitment of histone deacetylases (HDAC). Targeting these signaling pathways or HDACs may reverse tumor progression and provide clinical benefit for cancer patients. Thus, some small molecular inhibitors or monoclonal antibodies of each of these signaling pathways have been studied in clinical trials. This review focuses on the involvement of Hey proteins in malignant carcinoma progression and provides valuable therapeutic information for anticancer treatment. Mol Cancer Ther; 16(5); 775–86. ©2017 AACR.

This article is featured in Highlights of This Issue, p. 773

Hairy and Enhancer-of-split related with YRPW motif factors (Hey1/2/L), also known as Hairy and Enhancer-of-split related proteins (Hesr), Hairy-related transcriptional factors (HRT), Hes-related repressor proteins (HERP), and cardiovascular helix-loop-helix factors (CHF), belong to the basic helix-loop-helix Orange (bHLH-O) family (1–5). All three Hey genes have been found in developmental tissue, and abnormal expression of these proteins promotes abnormalities in stem cells, even leading to organ defects. Hey proteins can maintain an undifferentiated state of precursor cells by transcriptionally repressing cell fate regulators such as achaete-scute homolog 1 (6). In the developing heart, Hey proteins regulate cardiomyocyte precursor cell differentiation as well as epithelial–mesenchymal transition (EMT) of endocardium cells (7, 8). Since we recognized that cancer cells can monitor and utilize similar physiologic strategies to normal cells and promote tumor progression, for instance, cancer cells can initiate cellular plasticity and/or activate similar signaling pathways as mesenchymal cells, stem cells, or precursor cells do, we have started to realize the significant role played by Hey factors in tumor progression (9, 10). Hey proteins are found to be selectively expressed in malignant tumor tissues, and numerous studies have been undertaken to explain the molecular mechanism governing the Hey proteins in tumorigenesis. The most outstanding feature is that many signaling pathways can potentially confer EMT via Hey factors in malignant carcinomas. In addition, Hey factors not only regulate differentiation, self-renewal, and proliferation of cancer cells, but contribute to tumor neovasculature as well. Accumulating evidence indicates Hey factors lay at the crossroad of tumor progression. However, there are currently very few review articles illustrating the roles of the Hey family in tumorigenesis. The current review explores the functional significance of the Hey family in initiating these processes. We also describe the signaling pathways involved in the control of Hey expression. Small molecular inhibitors or monoclonal antibodies to each of these signaling pathways show promising antitumor or antiangiogenic effect in clinical trials. Here, these promising avenues for cancer treatment are also discussed.

Structure of the Hey family proteins

Hey family members are highly conserved and resemble their homologs, the Hairy and Enhancer of Split (Hes) family, in the four domain structures: basic, helix-loop-helix (HLH), Orange, and two C-terminal motifs. Hey proteins are directly connected to the E-box DNA sequence (CANNTG) via the glycine-rich basic domain (11, 12). The bHLH-O domain serves as a platform for cofactor interaction (3, 13). Despite extensive homology with the Hes family, Hey proteins also have significant features that distinguish them from Hes proteins, namely, the YRPW motif (YHSW for HeyL) and GTEIGAF (GTEVGAF for Hey2) peptides (ref. 1; Fig. 1). Hey proteins have been regarded as transcription inhibitors in the past. They have since been shown to act as transcription activators as well as inhibitors (Table 1). Strikingly, their function seems to be regulated at multiple levels. For instance, nonsynonymous single-nucleotide polymorphism (SNP) naturally occurs at codon 94 of Hey1, which leads to a substitution of a leucine residue by methionine (L94M) in the helix 2 domain. The L94M-mutant Hey1 transforms from an androgen receptor corepressor to androgen receptor coactivator without changing its intrinsic repressive domains (14). The phosphorylation of the Serine-68 residue of Hey1 prevents its enhancement of p53 transcriptional activation but confers p53-activating chemotherapy resistance, whereas wild-type Hey1 stimulates p53 and alters the sensitivity to p53-activating chemotherapy drugs. Interestingly, such posttranscriptional regulation is also observed within Hey2 (15). The dynamic regulation of Hey proteins at pretranscriptional levels, posttranscriptional levels, or their own characteristic structure could partly explain why Hey proteins eliminate one target molecule in certain cancers but activate the same molecule or its analogues in others. It should be noted that the specificity for protein interactions and target molecules of different Hey variants is differential between certain cell types. L94M Hey1 variant strongly interacts with Hey2, whereas Hey1 forms an unstable homodimer with Hey2 (14). There is a potential, unknown Hey1 variant enhancing matrix metallopeptidase 9 (MMP9) expression in osteosarcoma, whereas wild-type Hey1 is unable to bind to the MMP9 promoter itself (16).

Figure 1

Domain structures with percentage identity of Hey2 and HeyL with Hey1. The bHLH domains show the highest conservation among other domains. The Orange domain shows less conservation. Individual Hey proteins potentially recruit selective cofactors via Orange domain and C-terminal motif.

Figure 1

Domain structures with percentage identity of Hey2 and HeyL with Hey1. The bHLH domains show the highest conservation among other domains. The Orange domain shows less conservation. Individual Hey proteins potentially recruit selective cofactors via Orange domain and C-terminal motif.

Close modal
Table 1.

Summary of target genes, cytokine, and transcriptional factors of Hey

TargetsHey proteinsCommentReferences
P53 Hey1, Hey2, HeyL Activation 14, 15 
MMP9 Hey1 Activation 16 
Snai1 Hey1, Hey2, HeyL Activation 26, 30 
IL6 Hey1 Activation 37 
Twist1 Hey1, Hey2 Repression 34 
Snai2 Hey1, Hey2 Repression 34 
Runx2 Hey1 Repression 40, 41 
Col2a1 Hey1 Repression 42 
VEGFR2 Hey1, Hey2 Repression 51, 58, 60, 61 
TargetsHey proteinsCommentReferences
P53 Hey1, Hey2, HeyL Activation 14, 15 
MMP9 Hey1 Activation 16 
Snai1 Hey1, Hey2, HeyL Activation 26, 30 
IL6 Hey1 Activation 37 
Twist1 Hey1, Hey2 Repression 34 
Snai2 Hey1, Hey2 Repression 34 
Runx2 Hey1 Repression 40, 41 
Col2a1 Hey1 Repression 42 
VEGFR2 Hey1, Hey2 Repression 51, 58, 60, 61 

Hey proteins in malignant carcinomas

The levels of Hey factors are strikingly elevated in high-grade glioma, malignant osteosarcoma, high-grade esophageal squamous cell carcinoma, aggressive pancreatic adenocarcinoma rhabdomyosarcoma, as well as colorectal carcinoma (17–23). In these malignant carcinomas, aberrant Hey expression has been associated with poor prognosis, overall survival (OS), tumor grade, chemotherapy resistance, lymphatic metastasis, and vascular proliferative properties (17, 23–25). Taken together, these studies suggest that elevated levels of Hey proteins contribute to tumor progression, and to a certain extent, this is a result of their regulation of the behavior of cancer cells as well as remodeling of the tumor microenvironment (Fig. 2).

Figure 2

Hey proteins in tumorigenesis. Via activating or inactivating cytokines and other transcriptional factors, Hey proteins show their regulation on tumor progression, including cancer metastasis, cancer cells' quiescence maintenance as well as cancer neovasculature.

Figure 2

Hey proteins in tumorigenesis. Via activating or inactivating cytokines and other transcriptional factors, Hey proteins show their regulation on tumor progression, including cancer metastasis, cancer cells' quiescence maintenance as well as cancer neovasculature.

Close modal

The roles of Hey proteins in cancer metastasis

It was first observed that Hey-induced EMT was required in the developing heart (26–29). Subsequently, Hey proteins were found to be involved in tumor metastasis progression. In vitro, Hey1 knockdown inhibited the invasive phenotype of osteosarcoma via downregulation of MMP9 (16). Furthermore, the transfection of Hey1 antisense oligonucleotides blocked EMT through E-cadherin expression, and Smad3 inhibition repressed Hey1-induced EMT phenotype even in the presence of TGFβ (30). Strikingly, interaction between Hey1 and Smad3 has been observed in vitro (31), suggesting a Hey1–Smad3 complex transcriptionally represses E-cadherin. However, in the absence of activated Smad3, Hey1 does not influence EMT promotion, but only acts as a Snai1-initiated EMT marker (30, 32). On the other hand, Snai1, known as an E-cadherin repressor, potentially contributes to this repression process. Snai1 is reduced in Hey1/HeyL double knockouts and Hey2 knockout AV canals, and Snai1 can form a complex with Smad3 to occupy the E-cadherin promoter (26, 33). All these observations hint that Hey1 interacts with Smad3 and may inhibit E-cadherin directly or in a Snai1-Smad3-Hey1 manner. In other situations, Hey proteins promote mesenchymal–epithelial transition (MET). Upon Notch4 induction, Hey proteins promote melanoma MET and are important in promoting metastatic colonization because Hey1 and Hey2 can eliminate Snai2 as well as Twist1 expression via binding to their promoters (34). The different stimuli have a potential influence on Hey function, as TGFβ-induced Hey1 promotes EMT and Notch-induced Hey proteins regulate MET or EMT/MET irrelevant. However, it is more complex than first thought. Forced expression of Hey proteins has no impact on Snai2 or E-cadherin expression in other cell lines (35, 36). Does the paradox happen in different cell types? Evidence from the previous section indicates the nonsynonymous SNP of Hey genes in different cell types will affect different Hey variants' DNA-binding ability as well as protein-interaction specificity. Based on this, we presume that Hey variants affect Snai1/Snai2 expression transcriptionally to mediate EMT/MET in different cell lines. More intensive research is required to fully characterize Hey variants and the posttranscriptional modification of Hey. Also, Hey1 participates in metastatic microenvironment remodeling. Tumor-derived Jagged1 enhances osteoblast secretion of IL6 via Hey1 activation, and, in turn, IL6 confers a proliferative advantage to cancer cells (37). Epithelium-derived Jagged1 activates Hey1 which then promotes metastatic lymphoma cell chemotherapy resistance as well as progression in the tumor perivascular niche (38).

Hey proteins can regulate the differentiation, self-renewal, and proliferation of cancer cells

Hey proteins were identified as one of a few genes specifically expressed during embryogenesis (1, 39). Following this discovery, the potential capacity of the Hey family in sustaining cell quiescence was recognized (6, 40–42). Cancers monitor the quiescence strategy to keep their nondivide state and contribute to tumor progression (10, 43). The upregulation of Hey1 is likely to inhibit differentiation because rhabdomyosarcoma cells with an shRNA antagonizing Hey1 display differentiation morphology changes and the expression of differentiation marker myogenin (22). The introduction of Hey1 into proliferating osteosarcoma increases p53 expression and makes tumor cells stay in a nondividing state through p53-dependent reversible cell-cycle arrest (14). In the context of quiescence, elevated Hey family expression can reflect the undifferentiating property of malignant cancer cells. In addition, the ability of Hey proteins in maintaining self-renewal was investigated. The expression of Hey1 and Hey2 is remarkably higher in cancer stem cells (CSC), also referred as tumor-initiating cells (TIC), than that in non-CSCs (44). Hey1 is supposed to maintain CSCs self-renewal capacity as the silencing of Hey1 dampens malignant tumor-initiating ability as well as tumor growth in vivo and reduces cancer cell sphere formation in vitro (45, 46). In hepatocellular carcinoma, Hey1 upregulation upon c-Met/FRA1 signaling increases the number and the size of TICs spheroids which represent the self-renewal ability of these cells (47). Furthermore, Hey proteins have an effect on cancer proliferation. Hey2 overexpression increases hepatocellular carcinoma cell viability and proliferation (48). HeyL can promote breast cancer initiation through interaction with TGFβ-activated Smad3 (31). Interestingly, HeyL promotes p53-induced cell-cycle arrest which results in suppression of cancer cell proliferation and induction of cancer cell apoptosis in hepatocellular carcinoma (49). The same study also reported that 75% of hepatocellular carcinoma tissues had inaction of HeyL, suggesting that HeyL is a potential tumor suppressor in hepatocellular carcinoma. This is an interesting observation, despite that it was a single study and demonstrated in a small cohort (n = 80), this will require further validation on a larger scale. However, the fact that HeyL differs in one of the key motifs, namely the YHSW motif, from Hey1 and Hey2 which both have the YRPW motif, may be one of the reasons why it acts differently from other Hey proteins. While YRPW appears to be a good target (16), YHSW, at least in hepatocellular carcinoma, may not be. This is clearly a fascinating area to explore, both in research and in clinical settings.

Balance between HeyL and Hey1/Hey2 regulates cancer neovasculature

Genetic studies have highlighted the great influence of Hey proteins in angiogenesis during development or pathologic conditions (27, 50–52). Angiogenesis actively requires a strict hierarchy between sprouting and vascular tubes (53). Previous research suggests a factor acting upstream of Hey, Delta-like 4 (DLL4), is capable of controlling this hierarchy, as the inhibition of DLL4 leads to a hyper-sprouting phenotype following exposure to proangiogenic factors (54–56). Much evidence, however, supports that DLL4′s control on vascular sprouting is via its downstream factors, Hey1/2. It is acknowledged that epithelium with higher VEGFR2 emerges at the tip position and sustained VEGFR2 pathway activation results in excessive sprouting (57–59). Strikingly, Hey1, as well as Hey2, can suppress VEGFR2 expression and eliminate the increased frequency of epithelial cells at the tip position (58, 60, 61). When activated by the bone morphogenetic protein (BMP)/Activin receptor-like kinase (ALK) pathway, Hey1 as well as Hey2 abrogate the hyper-sprouting phenotype and induce tube formation (58, 62). In tumors, the coordinated balance between VEGFR2 and DLL4/Hey is tightly required for tumor cell expansion (63). DLL4/Hey2 overexpression leads to tumor growth by promoting low-density and mature tumor vessels through downregulation of VEGFR2 levels (64). DLL4/Hey blockage leads to VEGFR2 upregulation, which restrains tumor progression by producing hyper-sprouting, thin, fragile, and nonfunctional tumor vessels (56, 65–67). Interestingly, Jagged1-associated Hey upregulation seems to have little effect on low-density and mature tumor vessel phenotype, and Jagged1 promotes tumor-spouting angiogenesis through distinct mechanisms (54, 68, 69). In contrast, HeyL potentially promotes neovascularization. Studies reveal that breast tumor–derived vascular samples exhibit at least 20-fold higher levels of HeyL than normal breast vasculature. The elevated level of HeyL potentially promotes neovasculature by forcing vascular endothelial cells to undergo proliferation and ceasing apoptosis (25). Taken together, this evidence highlights the complexity of Hey in angiogenesis, and drugs targeting DLL4, Jagged1, and ALK1 are promising.

Notch-Hey signaling pathway

The mature heterodimeric Notch receptors are cleaved at two sites once the five ligands (Delta-like 1, 3 and 4, and Jagged 1 and 2) bind to the four membrane-bound Notch receptors (Notch 1, 2, 3, and 4), firstly by a disintegrin and metalloproteinase domain-containing protein 10/17 (ADAM10/17) and secondly by γ-secretase to release the Notch intracellular domain (NICD). In the nucleus, NICD interacts with the CBF1/Suppressor of Hairless/Lag1 (CSL) and recruits coactivators, allowing for transcriptional activation of Hey genes (4, 70, 71). Intriguingly, Notch receptors or Notch ligands show little selectivity for the induction of individual Hey proteins. Aberrant Notch-Hey axis shows great relevance to cancer biology. The Notch-Hey1 signaling pathway is over activated in invasive breast cell lines. Upon Notch inhibition via γ-secretase inhibitors (GSI), their migration and invasion capacity is reduced and this is accompanied by downregulation of Hey proteins (32, 72). The disruption of Notch-Hey1 in stroma bone cells decreases Jagged1-mediated breast tumor growth and bone metastasis (37). In osteosarcoma as well as rhabdomyosarcoma, Notch-Hey inhibition reverses tumor cell proliferative and relieves tumor burden (20, 22). GSI treatment also contributes to depletion of breast CSCs (44). Furthermore, a nonfunctional vascular network which results in tumor growth inhibition emerges when the DLL4-Notch-Hey1/2 pathway is blocked by DLL4 antibodies (56, 67). Thus, because GSIs, anti-Notch receptors, as well as anti-DLL4 are effective in Notch-Hey pathway inhibition, they have been developed into promising preclinical drugs (as summarized in Table 2).

Table 2.

Selected therapeutic inhibitors of Notch signaling, TGFβ signaling, and HDACs

Mechanism of actionAgentBiology targetedClinical benefitsDiseaseStageNCT number
Notch       
Pan-Notch inhibitor RO-4929097 Antitumor SD, PD Metastatic colorectal cancer Phase II NCT01116687 
   SD, PD Recurrent ovarian cancer Phase II NCT01175343 
   SD Pretreated pancreatic adenocarcinoma Phase II NCT01232829 
   PR, SD Metastatic melanoma Phase II NCT01120275 
 MK-0752 Antitumor CR, SD Advanced solid tumors Phase I NCT00106145 
   SD Children CNS malignancies Phase I NCT00572182 
 LY900009 Antitumor SD Advanced cancers Phase I NCT01158404 
 PF-0308414 Antitumor CR, PR, SD Advanced solid tumors Phase I NCT00878189 
Notch1-specific antibody OMP-52M51 Antitumor PR, SD Solid tumors Phase I NCT01778439 
Notch2/3-specific antibody OMP-59R5 Antitumor SD, PD Untreated metastatic pancreatic cancer Phase I NCT01647828 
   PR, SD Untreated small-cell lung cancer Phase I NCT01859741 
DLL4-specific antibody REGN421 Angiogenesis targeting PR, SD Advanced solid tumors Phase I NCT00871559 
 OMP-21M18 Angiogenesis targeting PR Pretreated solid tumors Phase I NCT00744562 
TGFβ       
ALK1-specific antibody ACE-041 Angiogenesis targeting PR, SD Advanced solid tumors Phase I NCT00996957 
 PF-03446962 Angiogenesis targeting PR, SD Pretreated advanced solid tumors Phase I NCT00557856 
   SD Pretreated urothelial cancer Phase II NCT01620970 
   SD Advanced solid tumors Phase I NCT01337050 
ALK4/5/7 antibody LY2157299 Antitumor CR, PR, SD Advanced cancer and glioma Phase I Unavailablea 
   SD Advanced solid tumors Phase I NCT01722825 
HDAC       
Pan-HDAC inhibitors Vorinostat Antitumor  Cutaneous T-cell lymphoma Approved  
 Belinostat Antitumor  Peripheral T-cell lymphoma Approved  
 Panobinostat Antitumor  Multiple myeloma Approved  
 Romidepsin Antitumor  Cutaneous T-cell lymphoma Approved  
Mechanism of actionAgentBiology targetedClinical benefitsDiseaseStageNCT number
Notch       
Pan-Notch inhibitor RO-4929097 Antitumor SD, PD Metastatic colorectal cancer Phase II NCT01116687 
   SD, PD Recurrent ovarian cancer Phase II NCT01175343 
   SD Pretreated pancreatic adenocarcinoma Phase II NCT01232829 
   PR, SD Metastatic melanoma Phase II NCT01120275 
 MK-0752 Antitumor CR, SD Advanced solid tumors Phase I NCT00106145 
   SD Children CNS malignancies Phase I NCT00572182 
 LY900009 Antitumor SD Advanced cancers Phase I NCT01158404 
 PF-0308414 Antitumor CR, PR, SD Advanced solid tumors Phase I NCT00878189 
Notch1-specific antibody OMP-52M51 Antitumor PR, SD Solid tumors Phase I NCT01778439 
Notch2/3-specific antibody OMP-59R5 Antitumor SD, PD Untreated metastatic pancreatic cancer Phase I NCT01647828 
   PR, SD Untreated small-cell lung cancer Phase I NCT01859741 
DLL4-specific antibody REGN421 Angiogenesis targeting PR, SD Advanced solid tumors Phase I NCT00871559 
 OMP-21M18 Angiogenesis targeting PR Pretreated solid tumors Phase I NCT00744562 
TGFβ       
ALK1-specific antibody ACE-041 Angiogenesis targeting PR, SD Advanced solid tumors Phase I NCT00996957 
 PF-03446962 Angiogenesis targeting PR, SD Pretreated advanced solid tumors Phase I NCT00557856 
   SD Pretreated urothelial cancer Phase II NCT01620970 
   SD Advanced solid tumors Phase I NCT01337050 
ALK4/5/7 antibody LY2157299 Antitumor CR, PR, SD Advanced cancer and glioma Phase I Unavailablea 
   SD Advanced solid tumors Phase I NCT01722825 
HDAC       
Pan-HDAC inhibitors Vorinostat Antitumor  Cutaneous T-cell lymphoma Approved  
 Belinostat Antitumor  Peripheral T-cell lymphoma Approved  
 Panobinostat Antitumor  Multiple myeloma Approved  
 Romidepsin Antitumor  Cutaneous T-cell lymphoma Approved  

NOTE: The unapproved structures of the compounds are available in supplementary data.

Abbreviation: CNS, central nervous system

aReference 121.

γ-Secretase inhibitors

Various preclinical trials show that GSIs have strong antitumor effects (73, 74). When treated with MK-0752 in phase I studies, clinical benefits such as complete response (CR) and prolonged stable disease (SD) were observed (75–78). However, patients present no objective responses to monotherapy of RO-4929097 in phase II clinical trials of solid tumors (79–82). Clinical indication of GSIs is still controversial, as a portion of cancer patients experienced SD during RO-4929097 or MK-0752 therapy, 1 advanced thyroid cancer patient achieved CR, and 71.4% (5/7) desmoid tumor patients had a partial response (PR) when they received another GSI, PF-0308414 (83). The most prominent and dose-limiting toxicity of GSIs is gastrointestinal (GI) events including diarrhea, vomiting, and nausea. This GI toxicity is likely based on the mechanism that inhibition of Notch signaling abrogates the undifferentiated state of intestinal crypt progenitor cells and results in differentiation into goblet cells (84). To reverse GI events, some investigators use glucocorticoid or tamoxifen therapy which potentially protects the intestine from goblet cell metaplasia (85, 86). Besides, the adverse events are scheduled dependent. The once-per-week dosing schedule of MK-0752 shows less severe GI events, as well as fatigue, than intermittent dosing for 3 to 7 days or continuous daily dosing group and the once-per-week group also achieved substantial Notch signaling inhibition (75). With glucocorticoid therapy and intermittent schedule, cancer patients are more tolerant to higher GSIs exposure and may associate with better outcomes. However, it is worth considering that GSIs have an off-target effect as γ-secretase cleaves more than 90 substrates (87). Strikingly, two types of GSIs reduce Notch1 but not Notch4 activity, suggesting some GSIs are receptors specific (88). In addition, different GSIs enjoy quite inequivalent pharmacokinetics. LY900009 is cleared by oxidation and amide hydrolysis, and its renal clearance is low, while semagacestat, an analogue of LY900009, mostly depends on renal clearance (89, 90). RO4929097 is cleared by autoinduction of cytochrome P450 family 3 subfamily A polypeptide 4 (CYP3A4), indicating that combination RO4929097 therapy with antitumor agents metabolized by CYP3A4 might show limit clinical utility (91). Furthermore, intravenous GSIs are under development (ref. 92; chemical structures of the unproved GSIs are available in Supplementary Data: Supplementary Figs. S1–S4).

Anti-Notch receptor antibodies

As GSIs are pan-Notch inhibitors, several antibodies were launched to block Notch receptors more specifically by binding with their extracellular-negative regulator region or ligand-binding domain. Preclinical data show antitumor, antiangiogenesis effects and decreasing CSCs frequency following treatment with these receptor-specific antibodies (93–95). Based on the success of Notch-specific antibodies, OMP-52M51 (anti-Notch1) and OMP-59R5 (anti-Notch2/3) have been studied in clinical trials. In a phase I study in solid tumors, the best response to OMP-52M51 was 2 patients with adenoid cystic carcinomas: one achieved PR; the other had SD for 290 days and SD was also observed in other tumors (96). Untreated metastatic pancreatic cancer patients only present SD, whereas 75% (6/8) of untreated extensive-stage small-cell lung cancer patients achieve PR to OMP-59R5 monotherapy (97, 98). Anti-Notch receptor antibodies are attractive, as they still function even in Notch receptors carrying mutations, and some of these tumors carrying mutations may be highly sensitive to these antibodies (93).

Anti-DLL4 monoclonal antibodies

Considering the great importance that DLL4 exerts on tumor vessel formation, targeting DLL4 was used to target tumor angiogenesis in preclinical studies, and several DLL4-blocking monoclonal antibodies have also been used to target Notch-mediated tumor angiogenesis in clinical trials (64, 99). SD and PR were noted in 41% of patients with advanced solid tumors when treated with REGN421 (Enoticumab), a DLL4 monoclonal antibody, in a phase I trial (100). OMP-21M18 (Demcizumab), another anti-DLL4 monoclonal antibody, showed antitumor effect, and 40% of patients with solid tumors responded with a reduction in tumor size (101). Although promising and well tolerated, severe adverse events, including hemangiomas, bleeding episodes, increased levels of troponin I, and ventricular dysfunction, were observed. In addition, targeting Jagged1 seems to exhibit an alternative therapeutic strategy which requires further clinical data (102, 103).

Agents in preclinical stage

Other agents blocking Notch signaling are also under development. Soluble decoys, which are Notch receptor extracellular domains or Notch ligands fused with or without human IgG, compete with endogenous ligands and inhibit Notch signaling activation. Notch1 decoys consisting of certain EGF-like repeats can interrupt Jagged-class–induced Notch uniquely and show antiangiogenesis as well as antitumor effects with limited adverse events in vivo (68). Soluble DLL1 or Jagged1 decoys can also block Notch signaling successfully (104, 105). Also, cell-permeable peptides interact with NICD-CSL and form a transcriptionally repressive complex which halts leukemia cell proliferation (106, 107).

TGFβ-Hey signaling pathway

Recent evidence has documented that TGFβ signaling induces Hey protein in a Notch-independent manner or through canonical Notch. Upon TGFβ1 activation, initiation of Hey is conducted by Smad3/Smad4 complex binding to Hey promoters at Smad-binding element core repeat (SCR) positions, and Hey gene activation is still observed when canonical Notch is abrogated by GSI (30). BMP9 protein activates Smad1/5/8 and directly stimulates Hey expression via a noncanonical Notch signaling pathway when it binds to TGFβ type I receptor-ALK1 receptor (58, 62, 108). On the other hand, activation of the Hey family can be enhanced by synergy between TGF-β/BMP and Notch signaling. Smads physically interact with Notch-dependent NICD and synergistically activate transcription of Hey1, Hey2, and HeyL, when Smads are activated by BMP-ALK5/6 or TGFβ1 treatment (30, 109, 110). As with the Notch pathway, TGFβ signaling is often elevated in tumors and contributes to tumor progression. Subsequent studies have indicated a crucial role for TGFβ in EMT initiation, and tumors break free from their neighboring tissue to undergo metastasis through TGFβ-induced EMT (111). Smad3 is of significant importance for Hey1-induced EMT as Smad3 is an integral molecule for repressing E-cadherin. In addition to metastasis, TGFβ pathway activation has been linked to tumor angiogenesis. Upon BMP9 treatment, the ALK1-Hey signaling pathway forces epithelial cells to remain in a stalk cell state, resulting in tube induction and mature vessel phenotype (58). If the ALK1-Hey signaling pathway is abrogated through addition of the ALK1 inhibitor, K02288, a hyper-sprouting phenotype is induced in vitro and angiogenesis is disrupted in vivo (112). Thus, TGFβ receptor inhibitors, which are potentially antitumor as well as antiangiogenesis drugs, have been applied in preclinical trials (as summarized in Table 2).

ALK1 blockers

Several ALK1 inhibitors have been studied in clinical trials. ACE-041 (Dalantercept), another ALK1 blocker, was tested in squamous cell carcinoma, non–small cell lung cancer, and intestinal adenocarcinoma and displayed antitumor activity in phase II clinical trial (113). No responses or PR to PF-03446962, an antibody targeting ALK1, was observed in hepatocellular carcinoma, urothelial cancer, colorectal cancer, malignant pleural mesothelioma, and other solid tumors (114–116). Three patients with metastatic hepatocellular carcinoma, metastatic clear cell renal carcinoma, and KRAS-mutant non–small cell lung cancer showed PR to PF-03446962 in another phase I trial (117). SD was observed among these four studies. Although only a very small part of patients have PR to anti-ALK1, further research is required into anti-ALK1. PR and SD were observed in portions of patients who still had lesions and cancer progression following VEGFR tyrosine kinase inhibitor (TKI) treatment. The combination of VEGFR TKI and ACE-041 results in a promising antiangiogenesis effect with marked tumor vascular disruption in xenograft models (118).

ALK5 inhibitor

LY2157299 (galunisertib), a small molecular inhibitor targeting the TGFβ receptor I, was originally developed as an ALK5 inhibitor and proved to complement ALK4/7 inhibitors (119). LY2157299 exerts an anti-invasive effect rather than antiproliferative effect on hepatocellular carcinoma cells via repression of Smad2 and Smad3 phosphorylation (120). A total of 24.3% of patients with glioma had either CR or PR to LY2157299, and 26.7% showed SD to LY2157299 in a phase I trial (121). Interestingly, 80% of low-grade glioma patients with isocitrate dehydrogenase mutation received clinical benefits in this study, when given LY2157299 treatment. In addition, LY2157299 is well tolerated and safe without adverse cardiac events. However, LY2157299 shows limited antitumor effects in pancreatic tumors (122).

Hey mediates histone deacetylases

The mechanisms through which Hey factors regulate their downstream effectors might also provide promising strategies for anticancer treatment. Hey factors are known to repress the expression of their target genes through recruitment of cofactors (123). Through Hey-mediated transcriptional repression, cancer cells maintain their undifferentiation state. Hey1 transcriptionally represses myogenin expression to sustain embryonal rhabdomyosarcoma cells in an immature state (22). Heterodimers between Hes1 and Hey factors potentially silence achaete-scute homolog 1, which results in the maintenance of an undifferentiated state of tumors (124–126). Histone deacetylases (HDAC) are potentially involved in the repressive effects of Hey factors, as treatment with trichostatin A, a pan class I and II HDAC inhibitor, partially abrogates the repressive effect of Hey factors (127–129). It has been suggested that Hey factors can use their bHLH domain to recruit the mSin3-NCoR-HDAC1 complex or associate with SIRT1, a member of NAD+-dependent HDACs, and induce transcriptional repression (11, 127). Further research indicates that Hey-HDACs complexes reduced target gene expression by downregulation of histone H3 lysine 27 acetylation (H3K27ac), which represents active transcription (130). Conversely, the inhibition of HDACs can lead to accumulation of acetylated histones and results in active transcription of target genes which are expected to cause tumor differentiation and induction of apoptosis (131, 132). Because the expression of HDACs is required for tumor cell survival and maintenance of an undifferentiated state, HDAC inhibitors might provide a new antitumor strategy. However, the application of HDAC inhibitors remains paradoxical and should be studied in different types of cancer. The silencing of HDAC1 and/or HDAC2 can give rise to hematologic malignancy initiation (133). Knockout of HDAC3 impairs genome stability as well as integrity and results in hepatocellular cancer (134).

HDAC inhibitors

Five HDAC inhibitors have been approved for T-cell lymphoma treatment, vorinostat (MK0683), belinostat (PXD-101), panobinostat (LBH-589), and romidepsin (FK-228), by the FDA, and chidamide (CS055/HBI-8000) approved in China (ref. 135; as summarized in Table 2). These highlight the impact of HDAC inhibitors as antitumor agents. A great number of HDAC inhibitors are currently in testing in different phases of trials, either combined with other antitumor chemotherapeutics or as monotherapies. A phase II study indicates that entinostat (SNDX-275/MS-275), an inhibitor of HDAC 1 and 3, brings clinical benefits (PR, CR, and SD) to 24% of Hodgkin lymphoma patients, and the median progression-free survival (PFS) as well as OS of these patients was 5.5 months and 25.1 months, respectively (136). Entinostat also shows antitumor effect in several clinical trials (137, 138). In estrogen receptor–positive breast cancer, the combination of exemestane with entinostat improves median PFS to 4.3 months and median OS to 28.1 months, whereas median PFS and OS is 2.3 and 19.8 months, respectively, in the exemestane plus placebo group (139). Other HDAC inhibitors, such as ITF2357, CHR-3996, and JNJ-26481585, have been studied and show promising antitumor effect (140–142).

Combination of therapies

The combination of therapies targeting TGFβ, HDACs, and Notch pathways requires thorough investigation regarding their cross-talk in specific cancer settings. For example, Notch and TGFβ have synergetic carcinogenic effects in lung carcinoma, head and neck squamous, esophageal adenocarcinoma, renal cell carcinoma, thyroid carcinoma, and breast cancer (31, 143–146). Because both TGFβ and Notch signaling can activate Hey, the simultaneous inhibition of both pathways might result in better outcomes than blockade of either individually. Interestingly, inhibition of both Notch and TGFβ cannot increase the synergetic effects on inhibition of cancer cell migration, but additional blockage of Notch attenuates cancer cell proliferation in TGFβ-treated cells (145). This highlights that combination therapies may affect more than one angle. Besides, combination of ALK1 inhibitors and GSI shows promise in targeting tumor angiogenesis, as inhibition of both signaling pathways further abolishes angiogenesis when compared with the inhibition of each alone (58). However, there is little clinical trial data about the combination of Notch and TGFβ inhibitors, and further insightful studies are required. In another instance, targeting both Hey levels and Hey activity concomitantly might prove advantageous in cancer treatment. As an example, Hey proteins exert their influence on tumor cells by recruiting HDACs; when combining GSI and vorinostat, glioma and melanoma cells show a decreased viability (147).

Another strategy is to combine molecular-targeted drugs with classical chemotherapies. The combination of GSIs, HDAC inhibitors, or TGFβ inhibitors with cytotoxic agents results in a more effective therapy since the inhibition of these pathways has been observed to enhance cancer cell lines sensitive to chemotherapy (148–150). Some clinical trials have also established the efficacy of combination therapies. For example, when combined GSIs with cytotoxic chemotherapy, clinical benefits, such as PR and prolonged SD, are observed in solid cancer patients (73, 151, 152). Encouraging antitumor activity is noticed in a Notch2/3-specific antibody study. Treatment OMP-59R5 with etoposide/cisplatin or nab-paclitaxel/gemcitabine shows 100% (3/3) PR in small cell lung cancer and 35% (9/26) PR and 35% (9/26) SD in untreated metastasis pancreatic cancer, respectively (97, 98). HDAC inhibitors in combination with classical chemotherapy also lead to a stronger antitumor effect. For instance, 64% thymoma and thymic carcinoma patients show objective response to belinostat plus cisplatin, doxorubicin, and cyclophosphamide, whereas vorinostat combined with fludarabine, mitoxantrone, and dexamethasone results in a 77.8% overall response rate in relapsed or refractory mantle cell lymphoma (153, 154). However, the combination of HDAC inhibitors with chemotherapy may lead to unacceptable toxicity and on times is less efficient (155–157).

Perspective in selectively targeting Hey proteins and bHLH factors

Because different tumors tend to upregulate Hey proteins via distinct pathways, targeting Hey proteins directly may bring about a higher response rate than blocking these pathways individually. Besides, targeting Hey proteins themselves may result in fewer side effects because the target genes of Notch, TGFβ, and HDAC signaling pathways will be unaffected. To target Hey, we have to understand the mechanism of action of Hey. There are two possible mechanisms of transcriptional regulation mediated by Hey proteins. The first mechanism is E-box–dependent transcriptional regulation. Hey proteins bind to E-box via the basic domain and form a functional complex with other cofactors via HLH domain. A domain located between amino acids 47 and 122 is necessary (11, 158). The second mechanism is E-box independent. Hey interacts with DNA-binding proteins via the HLH-O domain and performs as a cofactor. The critical domains locate in amino acids 47 to 76 and 111 to 291, which stride over bHLH and Orange domains (61, 159). Based on these, some small molecular inhibitors to antagonize Hey–DNA interaction and Hey–cofactor interaction might be promising. Dimer inhibitors from natural compounds were reported to disrupt the Hey homolog Hes1 dimerization (160). It is still possible to isolate small-molecule inhibitors targeting Hey. In addition, mutagenesis of Hes1 amino acid sequence in the basic domain does not decrease its dimerization-forming ability, but abrogates its transcriptional function (161, 162). Thus, we may construct high structural compatible Hey-dominant–negative peptides which can form inert complexes with Hey and block the three critical functional domains of Hey to disrupt their protein–protein and DNA–protein interfaces. The most successful example is designing stabilized, cell-permeable peptides which bind with NICD–CSL complex and prevent mastermind-like-1 interfacing to antagonize leukemia proliferation (107).

Human bHLH transcription factors contain over 200 members and can be divided into five classes based on phylogenetic analysis (163). Hey transcriptional factors belong to clade B, and other transcriptional factors, such as Twist1-2 (clade A), MyoD (clade C), Max (clade D), Myc (clade E), and hypoxia-induced factor (HIF, clade E), are also bHLH factors. From the mechanistic inhibitory action of bHLH, the bHLH inhibitors can be summarized into the following groups: preventing dimerization, preventing DNA binding, and preventing bHLH factors expression (164). For example, some small-molecule inhibitors were isolated to specifically inhibit Myc-Max dimerization and block the binding of Myc-Max to DNA without affecting other structure-like bHLH factor dimerization (165, 166). By using Myc bHLH-Zip domain fragments, researchers also discovered local conformational changes and formation of hydrophobic cavities at the specific peptide sequences of the fragments upon binding with these small-molecule inhibitors (167). This makes it possible to design specific inhibitors simply through protein sequence analysis because the small-molecule binding sites have certain peptide sequence criteria. Also, HIF dimer inhibitors as well as HIF DNA-binding inhibitors have been reported (168, 169). In addition, dominant negative peptides mimicking the HLH domain show a significant impact on E2A dimerization (170). Peptides of MyoD which have a high affinity for Id1 can interrupt MyoD–Id1 interaction and exhibit antitumor effects in vitro (171).

Hey proteins, a subfamily of mammalian bHLH-O transcriptional factors, have been highly investigated in several research studies since they have been found to be overexpressed in aggressive tumors. Previous work has focused on their transcriptional repressive roles in the maintenance of the undifferentiated state. More recently, studies reveal novel characteristics of Hey proteins in the regulation of cancer metastasis and their influence on angiogenesis. This article offers insight into the significant roles of Hey proteins in tumorigenesis. Alternatively, therapeutic agents able to reverse aberrant Notch, TGFβ, and HDACs levels have been evaluated in clinical trials, but treatment-associated toxicities are also observed. Targeting Hey factors may represent an opportunity for higher response rates but fewer side effects than treatment with GSIs, TGFβ blockers, and HDAC inhibitors. Attention should be drawn to the Hey family in drug design, and studies must be carried out to analyze outcomes using Hey-specific inhibitors in the future.

No potential conflicts of interest were disclosed.

The authors acknowledge Cancer Research Wales and Cardiff University China Medical Scholarship for supporting the study.

C. Gong was supported by grants from the Natural Science Foundation of China (81272893, 81472466, and 81672594); National Science Foundation of Guangdong Province (2014A03036003, 2014A030310378, 2014A020212059, 2015A030313172, 2015B050501004, 2016A050502018, and 2016A030313237); China Scholarship Council (No. 201606385034), and Elite Young Scholars Program of Sun Yat-sen Memorial Hospital (Y201401). E. Song was supported by the Natural Science Foundation of China (81490750, 81230060, 81442009, and 81621004); Key R & D Program of Ministry of Science and Technology of China (2016YFC1302300), Sun Yat-sen University Innovative Drug R & D Project, National Science Foundation of Guangdong Province (S2012030006287); Guangzhou Science Technology and Innovation Commission (201508020008, 201508020249, and 201504301341501); and Guangdong Science and Technology Department (2015B050501004). Parts of this work were supported by grants to E. Song obtained from Translational medicine public platform of Guangdong Province (4202037); Guangdong Department of Science & Technology Translational Medicine Center grant (2011A080300002); Grant KLB09001 from the Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University; and Grant (2013) 163 from Key Laboratory of Malignant Tumor Molecular Mechanism and Translational Medicine of Guangzhou Bureau of Science and Information Technology.

1
Leimeister
C
,
Externbrink
A
,
Klamt
B
,
Gessler
M
. 
Hey genes: a novel subfamily of hairy-and Enhancer of split related genes specifically expressed during mouse embryogenesis
.
Mech Dev
1999
;
85
:
173
7
.
2
Kokubo
H
,
Lun
Y
,
Johnson
RL
. 
Identification and expression of a novel family of bHLH cDNAs related to Drosophila hairy and enhancer of split
.
Biochem Biophys Res Commun
1999
;
260
:
459
65
.
3
Nakagawa
O
,
Nakagawa
M
,
Richardson
JA
,
Olson
EN
,
Srivastava
D
. 
HRT1, HRT2, and HRT3: a new subclass of bHLH transcription factors marking specific cardiac, somitic, and pharyngeal arch segments
.
Dev Biol
1999
;
216
:
72
84
.
4
Iso
T
,
Sartorelli
V
,
Chung
G
,
Shichinohe
T
,
Kedes
L
,
Hamamori
Y
. 
HERP, a new primary target of notch regulated by ligand binding
.
Mol Cell Biol
2001
;
21
:
6071
9
.
5
Sakata
Y
,
Kamei
CN
,
Nakagami
H
,
Bronson
R
,
Liao
JK
,
Chin
MT
. 
Ventricular septal defect and cardiomyopathy in mice lacking the transcription factor CHF1/Hey2
.
Proc Natl Acad Sci
2002
;
99
:
16197
202
.
6
Sakamoto
M
,
Hirata
H
,
Ohtsuka
T
,
Bessho
Y
,
Kageyama
R
. 
The basic helix-loop-helix genes Hesr1/Hey1 and Hesr2/Hey2 regulate maintenance of neural precursor cells in the brain
.
J Biol Chem
2003
;
278
:
44808
15
.
7
Fischer
A
,
Leimeister
C
,
Winkler
C
,
Schumacher
N
,
Klamt
B
,
Elmasri
H
, et al
Hey bHLH factors in cardiovascular development
.
Cold Spring Harb Symp Quant Biol
2002
;
67
:
63
70
.
8
Fischer
A
,
Gessler
M
. 
Hey genes in cardiovascular development
.
Trends Cardiovasc Med
2003
;
13
:
221
6
.
9
Lamouille
S
,
Xu
J
,
Derynck
R
. 
Molecular mechanisms of epithelial–mesenchymal transition
.
Nat Rev Mol Cell Biol
2014
;
15
:
178
96
.
10
Sang
L
,
Roberts
JM
,
Coller
HA
. 
Hijacking HES1: how tumors co-opt the anti-differentiation strategies of quiescent cells
.
Trends Mol Med
2010
;
16
:
17
26
.
11
Iso
T
,
Sartorelli
V
,
Poizat
C
,
Iezzi
S
,
Wu
H-Y
,
Chung
G
, et al
HERP, a novel heterodimer partner of HES/E (spl) in Notch signaling
.
Mol Cell Biol
2001
;
21
:
6080
9
.
12
Heisig
J
,
Weber
D
,
Englberger
E
,
Winkler
A
,
Kneitz
S
,
Sung
W-K
, et al
Target gene analysis by microarrays and chromatin immunoprecipitation identifies HEY proteins as highly redundant bHLH repressors
.
PLoS Genet
2012
;
8
:
e1002728
.
13
Davis
RL
,
Turner
DL
. 
Vertebrate hairy and Enhancer of split related proteins: transcriptional repressors regulating cellular differentiation and embryonic patterning
.
Oncogene
2001
;
20
:
8342
57
.
14
Villaronga
MA
,
Lavery
DN
,
Bevan
CL
,
Llanos
S
,
Belandia
B
. 
HEY1 Leu94Met gene polymorphism dramatically modifies its biological functions
.
Oncogene
2010
;
29
:
411
20
.
15
López-Mateo
I
,
Arruabarrena-Aristorena
A
,
Artaza-Irigaray
C
,
López
JA
,
Calvo
E
,
Belandia
B
. 
HEY1 functions are regulated by its phosphorylation at Ser-68
.
Biosci Rep
2016
;
36
:
e00343
.
16
Tsuru
A
,
Setoguchi
T
,
Matsunoshita
Y
,
Nagao-Kitamoto
H
,
Nagano
S
,
Yokouchi
M
, et al
Hairy/enhancer-of-split related with YRPW motif protein 1 promotes osteosarcoma metastasis via matrix metallopeptidase 9 expression
.
Br J Cancer
2015
;
112
:
1232
40
.
17
Forghanifard
MM
,
Taleb
S
,
Abbaszadegan
MR
. 
Notch signaling target genes are directly correlated to esophageal squamous cell carcinoma tumorigenesis
.
Pathol Oncol Res
2015
;
21
:
463
7
.
18
Gaetani
P
,
Hulleman
E
,
Levi
D
,
Quarto
M
,
Scorsetti
M
,
Helin
K
, et al
Expression of the transcription factor HEY1 in glioblastoma: a preliminary clinical study
.
Tumori
2010
;
96
:
97
.
19
El Hindy
N
,
Keyvani
K
,
Pagenstecher
A
,
Dammann
P
,
Sandalcioglu
IE
,
Sure
U
, et al
Implications of Dll4-Notch signaling activation in primary glioblastoma multiforme
.
Neurooncology
2013
;
15
:
1366
78
.
20
Engin
F
,
Bertin
T
,
Ma
O
,
Jiang
MM
,
Wang
L
,
Sutton
RE
, et al
Notch signaling contributes to the pathogenesis of human osteosarcomas
.
Hum Mol Genet
2009
;
18
:
1464
70
.
21
Mullendore
ME
,
Koorstra
J-B
,
Li
Y-M
,
Offerhaus
GJ
,
Fan
X
,
Henderson
CM
, et al
Ligand-dependent Notch signaling is involved in tumor initiation and tumor maintenance in pancreatic cancer
.
Clin Cancer Res
2009
;
15
:
2291
301
.
22
Belyea
BC
,
Naini
S
,
Bentley
RC
,
Linardic
CM
. 
Inhibition of the Notch-Hey1 axis blocks embryonal rhabdomyosarcoma tumorigenesis
.
Clin Cancer Res
2011
;
17
:
7324
36
.
23
Candy
PA
,
Phillips
MR
,
Redfern
AD
,
Colley
SM
,
Davidson
JA
,
Stuart
LM
, et al
Notch-induced transcription factors are predictive of survival and 5-fluorouracil response in colorectal cancer patients
.
Br J Cancer
2013
;
109
:
1023
30
.
24
Hulleman
E
,
Quarto
M
,
Vernell
R
,
Masserdotti
G
,
Colli
E
,
Kros
JM
, et al
A role for the transcription factor HEY1 in glioblastoma
.
J Cell Mol Med
2009
;
13
:
136
46
.
25
Parker
BS
,
Argani
P
,
Cook
BP
,
Liangfeng
H
,
Chartrand
SD
,
Zhang
M
, et al
Alterations in vascular gene expression in invasive breast carcinoma
.
Cancer Res
2004
;
64
:
7857
66
.
26
Fischer
A
,
Steidl
C
,
Wagner
TU
,
Lang
E
,
Jakob
PM
,
Friedl
P
, et al
Combined loss of Hey1 and HeyL causes congenital heart defects because of impaired epithelial to mesenchymal transition
.
Circ Res
2007
;
100
:
856
63
.
27
Kokubo
H
,
Miyagawa-Tomita
S
,
Nakazawa
M
,
Saga
Y
,
Johnson
RL
. 
Mouse hesr1 and hesr2 genes are redundantly required to mediate Notch signaling in the developing cardiovascular system
.
Dev Biol
2005
;
278
:
301
9
.
28
Rutenberg
JB
,
Fischer
A
,
Jia
H
,
Gessler
M
,
Zhong
TP
,
Mercola
M
. 
Developmental patterning of the cardiac atrioventricular canal by Notch and Hairy-related transcription factors
.
Development
2006
;
133
:
4381
90
.
29
Luna-Zurita
L
,
Prados
B
,
Grego-Bessa
J
,
Luxan
G
,
del Monte
G
,
Benguria
A
, et al
Integration of a Notch-dependent mesenchymal gene program and Bmp2-driven cell invasiveness regulates murine cardiac valve formation
.
J Clin Invest
2010
;
120
:
3493
507
.
30
Zavadil
J
,
Cermak
L
,
Soto‐Nieves
N
,
Böttinger
EP
. 
Integration of TGF‐β/Smad and Jagged1/Notch signalling in epithelial‐to‐mesenchymal transition
.
EMBO J
2004
;
23
:
1155
65
.
31
Han
L
,
Diehl
A
,
Nguyen
NK
,
Korangath
P
,
Teo
W
,
Cho
S
, et al
The Notch pathway inhibits TGFbeta signaling in breast cancer through HEYL-mediated crosstalk
.
Cancer Res
2014
;
74
:
6509
18
.
32
Bolos
V
,
Mira
E
,
Martinez-Poveda
B
,
Luxan
G
,
Canamero
M
,
Martinez
AC
, et al
Notch activation stimulates migration of breast cancer cells and promotes tumor growth
.
Breast Cancer Res
2013
;
15
:
R54
.
33
Vincent
T
,
Neve
EPA
,
Johnson
JR
,
Kukalev
A
,
Rojo
F
,
Albanell
J
, et al
A SNAIL1-SMAD3/4 transcriptional repressor complex promotes TGF-β mediated epithelial-mesenchymal transition
.
Nat Cell Biol
2009
;
11
:
943
50
.
34
Bonyadi Rad
E
,
Hammerlindl
H
,
Wels
C
,
Popper
U
,
Ravindran Menon
D
,
Breiteneder
H
, et al
Notch4 signaling induces a mesenchymal-epithelial-like transition in melanoma cells to suppress malignant behaviors
.
Cancer Res
2016
;
76
:
1690
7
.
35
Leong
KG
,
Niessen
K
,
Kulic
I
,
Raouf
A
,
Eaves
C
,
Pollet
I
, et al
Jagged1-mediated Notch activation induces epithelial-to-mesenchymal transition through Slug-induced repression of E-cadherin
.
J Exp Med
2007
;
204
:
2935
48
.
36
Niessen
K
,
Fu
Y
,
Chang
L
,
Hoodless
PA
,
Mcfadden
D
,
Karsan
A
. 
Slug is a direct Notch target required for initiation of cardiac cushion cellularization
.
J Cell Biol
2008
;
182
:
315
25
.
37
Sethi
N
,
Dai
X
,
Winter
CG
,
Kang
Y
. 
Tumor-derived JAGGED1 promotes osteolytic bone metastasis of breast cancer by engaging notch signaling in bone cells
.
Cancer Cell
2011
;
19
:
192
205
.
38
Cao
Z
,
Ding
BS
,
Guo
P
,
Lee
SB
,
Butler
JM
,
Casey
SC
, et al
Angiocrine factors deployed by tumor vascular niche induce B cell lymphoma invasiveness and chemoresistance
.
Cancer Cell
2014
;
25
:
350
65
.
39
Leimeister
C
,
Schumacher
N
,
Steidl
C
,
Gessler
M
. 
Analysis of HeyL expression in wild-type and Notch pathway mutant mouse embryos
.
Mech Dev
2000
;
98
:
175
8
.
40
Zamurovic
N
,
Cappellen
D
,
Rohner
D
,
Susa
M
. 
Coordinated activation of notch, Wnt, and transforming growth factor-beta signaling pathways in bone morphogenic protein 2-induced osteogenesis. Notch target gene Hey1 inhibits mineralization and Runx2 transcriptional activity
.
J Biol Chem
2004
;
279
:
37704
15
.
41
Hilton
MJ
,
Tu
X
,
Wu
X
,
Bai
S
,
Zhao
H
,
Kobayashi
T
, et al
Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation
.
Nat Med
2008
;
14
:
306
14
.
42
Grogan
SP
,
Olee
T
,
Hiraoka
K
,
Lotz
MK
. 
Repression of chondrogenesis through binding of notch signaling proteins HES-1 and HEY-1 to N-box domains in the COL2A1 enhancer site
.
Arthritis Rheum
2008
;
58
:
2754
63
.
43
Yeh
AC
,
Ramaswamy
S
. 
Mechanisms of cancer cell dormancy—another hallmark of cancer?
Cancer Res
2015
;
75
:
5014
22
.
44
Yamamoto
M
,
Taguchi
Y
,
Ito-Kureha
T
,
Semba
K
,
Yamaguchi
N
,
Inoue
J
. 
NF-kappaB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype
.
Nat Commun
2013
;
4
:
2299
.
45
Zhu
P
,
Wang
Y
,
Du
Y
,
He
L
,
Huang
G
,
Zhang
G
, et al
C8orf4 negatively regulates self-renewal of liver cancer stem cells via suppression of NOTCH2 signalling
.
Nat Commun
2015
;
6
:
7122
.
46
Wu
HC
,
Lin
YC
,
Liu
CH
,
Chung
HC
,
Wang
YT
,
Lin
YW
, et al
USP11 regulates PML stability to control Notch-induced malignancy in brain tumours
.
Nat Commun
2014
;
5
:
3214
.
47
Lau
EY
,
Lo
J
,
Cheng
BY
,
Ma
MK
,
Lee
JM
,
Ng
JK
, et al
Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-Met/FRA1/HEY1 signaling
.
Cell Rep
2016
;
15
:
1175
89
.
48
Wu
DC
,
Zhang
MF
,
Su
SG
,
Fang
HY
,
Wang
XH
,
He
D
, et al
HEY2, a target of miR-137, indicates poor outcomes and promotes cell proliferation and migration in hepatocellular carcinoma
.
Oncotarget
2016
;
7
:
38052
63
.
49
Kuo
KK
,
Jian
SF
,
Li
YJ
,
Wan
SW
,
Weng
CC
,
Fang
K
, et al
Epigenetic inactivation of transforming growth factor‐β1 target gene HEYL, a novel tumor suppressor, is involved in the P53‐induced apoptotic pathway in hepatocellular carcinoma
.
Hepatol Res
2015
;
45
:
782
93
.
50
Fischer
A
,
Schumacher
N
,
Maier
M
,
Sendtner
M
,
Gessler
M
. 
The Notch target genes Hey1 and Hey2 are required for embryonic vascular development
.
Genes Dev
2004
;
18
:
901
11
.
51
Li
B
,
Tang
SB
,
Hu
J
,
Gao
Y
,
Zhang
G
,
Lin
SF
, et al
Protective effects of transcription factor HESR1 on retinal vasculature
.
Microvasc Res
2006
;
72
:
146
52
.
52
Adepoju
O
,
Wong
A
,
Kitajewski
A
,
Tong
K
,
Boscolo
E
,
Bischoff
J
, et al
Expression of HES and HEY genes in infantile hemangiomas
.
Vasc Cell
2011
;
3
:
19
.
53
Tung
JJ
,
Tattersall
IW
,
Kitajewski
J
. 
Tips, stalks, tubes: Notch-mediated cell fate determination and mechanisms of tubulogenesis during angiogenesis
.
Cold Spring Harbor Perspect Med
2012
;
2
:
a006601
.
54
Benedito
R
,
Roca
C
,
Sörensen
I
,
Adams
S
,
Gossler
A
,
Fruttiger
M
, et al
The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis
.
Cell
2009
;
137
:
1124
35
.
55
Liu
Z
,
Fan
F
,
Wang
A
,
Zheng
S
,
Lu
Y
. 
Dll4-Notch signaling in regulation of tumor angiogenesis
.
J Cancer Res Clin Oncol
2014
;
140
:
525
36
.
56
Noguera-Troise
I
,
Daly
C
,
Papadopoulos
NJ
,
Coetzee
S
,
Boland
P
,
Gale
NW
, et al
Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis
.
Nature
2006
;
444
:
1032
7
.
57
Jakobsson
L
,
Franco
CA
,
Bentley
K
,
Collins
RT
,
Ponsioen
B
,
Aspalter
IM
, et al
Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting
.
Nat Cell Biol
2010
;
12
:
943
53
.
58
Larrivee
B
,
Prahst
C
,
Gordon
E
,
del Toro
R
,
Mathivet
T
,
Duarte
A
, et al
ALK1 signaling inhibits angiogenesis by cooperating with the Notch pathway
.
Dev Cell
2012
;
22
:
489
500
.
59
Boareto
M
,
Jolly
MK
,
Ben-Jacob
E
,
Onuchic
JN
. 
Jagged mediates differences in normal and tumor angiogenesis by affecting tip-stalk fate decision
.
Proc Natl Acad Sci
2015
;
112
:
E3836
E44
.
60
Taylor
KL
,
Henderson
AM
,
Hughes
CCW
. 
Notch activation during endothelial cell network formation in vitro targets the basic HLH transcription factor HESR-1 and downregulates VEGFR-2/KDR expression
.
Microvasc Res
2002
;
64
:
372
83
.
61
Holderfield
MT
,
Henderson Anderson
AM
,
Kokubo
H
,
Chin
MT
,
Johnson
RL
,
Hughes
CC
. 
HESR1/CHF2 suppresses VEGFR2 transcription independent of binding to E-boxes
.
Biochem Biophys Res Commun
2006
;
346
:
637
48
.
62
Ricard
N
,
Ciais
D
,
Levet
S
,
Subileau
M
,
Mallet
C
,
Zimmers
TA
, et al
BMP9 and BMP10 are critical for postnatal retinal vascular remodeling
.
Blood
2012
;
119
:
6162
71
.
63
Siemerink
MJ
,
Klaassen
I
,
Van Noorden
CJ
,
Schlingemann
RO
. 
Endothelial tip cells in ocular angiogenesis: potential target for anti-angiogenesis therapy
.
J Histochem Cytochem
2013
;
61
:
101
15
.
64
Li
JL
,
Sainson
RC
,
Shi
W
,
Leek
R
,
Harrington
LS
,
Preusser
M
, et al
Delta-like 4 Notch ligand regulates tumor angiogenesis, improves tumor vascular function, and promotes tumor growth in vivo
.
Cancer Res
2007
;
67
:
11244
53
.
65
Patel
NS
,
Li
JL
,
Generali
D
,
Poulsom
R
,
Cranston
DW
,
Harris
AL
. 
Up-regulation of delta-like 4 ligand in human tumor vasculature and the role of basal expression in endothelial cell function
.
Cancer Res
2005
;
65
:
8690
7
.
66
Ridgway
J
,
Zhang
G
,
Wu
Y
,
Stawicki
S
,
Liang
W-C
,
Chanthery
Y
, et al
Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis
.
Nature
2006
;
444
:
1083
7
.
67
Djokovic
D
,
Trindade
A
,
Gigante
J
,
Pinho
M
,
Harris
AL
,
Duarte
A
. 
Incomplete Dll4/Notch signaling inhibition promotes functional angiogenesis supporting the growth of skin papillomas
.
BMC Cancer
2015
;
15
:
608
.
68
Kangsamaksin
T
,
Murtomaki
A
,
Kofler
NM
,
Cuervo
H
,
Chaudhri
RA
,
Tattersall
IW
, et al
NOTCH decoys that selectively block DLL/NOTCH or JAG/NOTCH disrupt angiogenesis by unique mechanisms to inhibit tumor growth
.
Cancer Discov
2015
;
5
:
182
97
.
69
Qiu
XX
,
Chen
L
,
Wang
CH
,
Lin
ZX
,
Chen
BJ
,
You
N
, et al
The vascular Notch ligands delta-like ligand 4 (DLL4) and Jagged1 (JAG1) have opposing correlations with microvascularization but uniform prognostic effect in primary glioblastoma: a preliminary study
.
World Neurosurg
2015
;
88
:
447
58
.
70
Nakagawa
O
,
McFadden
DG
,
Nakagawa
M
,
Yanagisawa
H
,
Hu
T
,
Srivastava
D
, et al
Members of the HRT family of basic helix–loop–helix proteins act as transcriptional repressors downstream of Notch signaling
.
Proc Natl Acad Sci
2000
;
97
:
13655
60
.
71
Vinson
KE
,
George
DC
,
Fender
AW
,
Bertrand
FE
,
Sigounas
G
. 
The Notch pathway in colorectal cancer
.
Int J Cancer
2015
;
138
:
1835
42
.
72
Chen
J
,
Imanaka
N
,
Griffin
J
. 
Hypoxia potentiates Notch signaling in breast cancer leading to decreased E-cadherin expression and increased cell migration and invasion
.
Br J Cancer
2010
;
102
:
351
60
.
73
Schott
AF
,
Landis
MD
,
Dontu
G
,
Griffith
KA
,
Layman
RM
,
Krop
I
, et al
Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors
.
Clin Cancer Res
2013
;
19
:
1512
24
.
74
Luistro
L
,
He
W
,
Smith
M
,
Packman
K
,
Vilenchik
M
,
Carvajal
D
, et al
Preclinical profile of a potent gamma-secretase inhibitor targeting notch signaling with in vivo efficacy and pharmacodynamic properties
.
Cancer Res
2009
;
69
:
7672
80
.
75
Krop
I
,
Demuth
T
,
Guthrie
T
,
Wen
PY
,
Mason
WP
,
Chinnaiyan
P
, et al
Phase I pharmacologic and pharmacodynamic study of the gamma secretase (Notch) inhibitor MK-0752 in adult patients with advanced solid tumors
.
J Clin Oncol
2012
;
30
:
2307
13
.
76
Fouladi
M
,
Stewart
CF
,
Olson
J
,
Wagner
LM
,
Onar-Thomas
A
,
Kocak
M
, et al
Phase I trial of MK-0752 in children with refractory CNS malignancies: a pediatric brain tumor consortium study
.
J Clin Oncol
2011
;
29
:
3529
34
.
77
Piha-Paul
SA
,
Munster
PN
,
Hollebecque
A
,
Argilés
G
,
Dajani
O
,
Cheng
JD
, et al
Results of a phase 1 trial combining ridaforolimus and MK-0752 in patients with advanced solid tumours
.
Eur J Cancer
2015
;
51
:
1865
73
.
78
Hoffman
LM
,
Fouladi
M
,
Olson
J
,
Daryani
VM
,
Stewart
CF
,
Wetmore
C
, et al
Phase I trial of weekly MK-0752 in children with refractory central nervous system malignancies: a pediatric brain tumor consortium study
.
Childs Nerv Syst
2015
;
31
:
1283
9
.
79
Diaz-Padilla
I
,
Wilson
MK
,
Clarke
BA
,
Hirte
HW
,
Welch
SA
,
Mackay
HJ
, et al
A phase II study of single-agent RO4929097, a gamma-secretase inhibitor of Notch signaling, in patients with recurrent platinum-resistant epithelial ovarian cancer: a study of the Princess Margaret, Chicago and California phase II consortia
.
Gynecol Oncol
2015
;
137
:
216
22
.
80
Jesus-Acosta
AD
,
Laheru
D
,
Maitra
A
,
Arcaroli
J
,
Rudek
MA
,
Dasari
A
, et al
A phase II study of the gamma secretase inhibitor RO4929097 in patients with previously treated metastatic pancreatic adenocarcinoma
.
Invest New Drugs
2014
;
32
:
739
45
.
81
Lee
SM
,
James Moon
MS
,
Do
BGR
,
Tarek Chidiac
MD
,
Flaherty
LE
,
Zha
Y
, et al
Phase 2 study of RO4929097, a gamma-secretase inhibitor, in metastatic melanoma: SWOG 0933
.
Cancer
2015
;
121
:
432
40
.
82
Strosberg
JR
,
Yeatman
T
,
Weber
J
,
Coppola
D
,
Schell
MJ
,
Gang
H
, et al
A phase II study of RO4929097 in metastatic colorectal cancer
.
Eur J Cancer
2012
;
48
:
997
1003
.
83
Messersmith
WA
,
Shapiro
GI
,
Cleary
JM
,
Jimeno
A
,
Dasari
A
,
Huang
B
, et al
A phase I, dose-finding study in patients with advanced solid malignancies of the oral gamma-secretase inhibitor PF-03084014
.
Clin Cancer Res
2015
;
21
:
60
7
.
84
van Es
JH
,
van Gijn
ME
,
Riccio
O
,
van den Born
M
,
Vooijs
M
,
Begthel
H
, et al
Notch/γ-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells
.
Nature
2005
;
435
:
959
63
.
85
Samon
JB
,
Castillo-Martin
M
,
Hadler
M
,
Ambesi-Impiobato
A
,
Paietta
E
,
Racevskis
J
, et al
Preclinical analysis of the γ-secretase inhibitor PF-03084014 in combination with glucocorticoids in T-cell acute lymphoblastic leukemia
.
Mol Cancer Ther
2012
;
11
:
1565
75
.
86
Backus
K
. 
Crosstalk between PKCα and Notch-4 in endocrine-resistant breast cancer cells
.
Oncogenesis
2013
;
2
:
e60
.
87
Langosch
D
,
Steiner
H
. 
Substrate processing in intramembrane proteolysis by γ-secretase – the role of protein dynamics
.
Biol Chem
2016
Dec 28. [Epub ahead of print]
.
88
Harrison
H
,
Farnie
G
,
Howell
SJ
,
Rock
RE
,
Stylianou
S
,
Brennan
KR
, et al
Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor
.
Cancer Res
2010
;
70
:
709
18
.
89
Pant
S
,
Jones
SF
,
Kurkjian
CD
,
Infante
JR
,
Moore
KN
,
Burris
HA
, et al
A first-in-human phase I study of the oral Notch inhibitor, LY900009, in patients with advanced cancer
.
Eur J Cancer
2016
;
56
:
1
9
.
90
Yi
P
,
Hadden
C
,
Kulanthaivel
P
,
Calvert
N
,
Annes
W
,
Brown
T
, et al
Disposition and metabolism of semagacestat, a γ-secretase inhibitor, in humans
.
Drug Metab Dispos
2010
;
38
:
554
65
.
91
Tolcher
AW
,
Messersmith
WA
,
Mikulski
SM
,
Papadopoulos
KP
,
Kwak
EL
,
Gibbon
DG
, et al
Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors
.
J Clin Oncol
2012
;
30
:
2348
53
.
92
Knoechel
B
,
Bhatt
A
,
Pan
L
,
Pedamallu
CS
,
Severson
E
,
Gutierrez
A
, et al
Complete hematologic response of early T-cell progenitor acute lymphoblastic leukemia to the γ-secretase inhibitor BMS-906024: genetic and epigenetic findings in an outlier case
.
Cold Spring Harbor Mol Case Studies
2015
;
1
:
a000539
.
93
Wallace
B
,
Wang
M
,
Muriel
C
,
Cain
J
,
Cancilla
B
,
Shah
J
, et al
Novel NOTCH3 activating mutations identified in tumors sensitive to OMP-59R5, a monoclonal antibody targeting the Notch2 and Notch3 receptors [abstract]
. In:
Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6–10; Washington, DC
.
Philadelphia (PA)
:
AACR
.
Abstract nr 213
.
94
Asteamézaga
M
,
Zhang
N
,
Lineberger
JE
,
Arnold
BA
,
Toner
TJ
,
Gu
M
, et al
Characterization of Notch1 antibodies that inhibit signaling of both normal and mutated Notch1 receptors
.
PLoS One
2010
;
5
:
e9094
.
95
Yen
WC
,
Fischer
MM
,
Axelrod
F
,
Bond
C
,
Cain
J
,
Cancilla
B
, et al
Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency
.
Clin Cancer Res
2015
;
21
:
2084
95
.
96
Munster
P
,
Eckhardt
SG
,
Patnaik
A
,
Shields
AF
,
Tolcher
AW
,
Davis
SL
, et al
Safety and preliminary efficacy results of a first-in-human phase I study of the novel cancer stem cell (CSC) targeting antibody brontictuzumab (OMP-52M51, anti-Notch1) administered intravenously to patients with certain advanced solid tumors
.
Mol Cancer Ther
2015
;
14 (12 Suppl 2)
:
C42
.
97
Bendell
J
,
Cohn
A
,
Smith
L
,
Strickler
J
,
Gluck
W
,
Schmidt
W
, et al
688P Final results of a phase 1B of OMP-59R5 (anti-notch2/3/stem cell antibody) in combination with nab-paclitaxel and gemcitabine (NAB-P+ GEM) in patients (PTS) with untreated metastatic pancreatic cancer (MPC): Alpine study
.
Ann Oncol
2014
;
25
:
iv233
iv4
.
98
Pietanza
M
,
Spira
A
,
Jotte
R
,
Gadgeel
S
,
Mita
A
,
Liu
S
, et al
1473P Phase 1B trial of anti-notch 2/3 antibody OMP-59R5 in combination with etoposide and cisplatin (EP) in patients (PTS) with untreated extensive-stage small-cell lung cancer (ED-SCLC): the pinnacle study
.
Ann Oncol
2014
;
25
:
iv514
iv5
.
99
Scehnet
JS
,
Jiang
W
,
Kumar
SR
,
Krasnoperov
V
,
Trindade
A
,
Benedito
R
, et al
Inhibition of Dll4-mediated signaling induces proliferation of immature vessels and results in poor tissue perfusion
.
Blood
2007
;
109
:
4753
60
.
100
Chiorean
EG
,
Lorusso
P
,
Strother
RM
,
Diamond
JR
,
Younger
A
,
Messersmith
WA
, et al
A Phase I first-in-human study of enoticumab (REGN421), a fully human delta-like ligand 4 (Dll4) monoclonal antibody in patients with advanced solid tumors
.
Clin Cancer Res
2015
;
21
:
2695
703
.
101
Smith
DC
,
Eisenberg
PD
,
Manikhas
G
,
Chugh
R
,
Gubens
MA
,
Stagg
RJ
, et al
A phase I dose escalation and expansion study of the anticancer stem cell agent demcizumab (anti-DLL4) in patients with previously treated solid tumors
.
Clin Cancer Res
2014
;
20
:
6295
303
.
102
Steg
AD
,
Katre
AA
,
Goodman
B
,
Han
HD
,
Nick
AM
,
Stone
RL
, et al
Targeting the notch ligand JAGGED1 in both tumor cells and stroma in ovarian cancer
.
Clin Cancer Res
2011
;
17
:
5674
85
.
103
Chen
JY
,
Li
CF
,
Chu
PY
,
Lai
YS
,
Chen
CH
,
Jiang
SS
, et al
Lysine demethylase 2A promotes stemness and angiogenesis of breast cancer by upregulating Jagged1
.
Oncotarget
2014
;
7
:
27689
710
.
104
Varnum-Finney
B
,
Wu
L
,
Yu
M
,
Brashem-Stein
C
,
Staats
S
,
Flowers
D
, et al
Immobilization of Notch ligand, Delta-1, is required for induction of notch signaling
.
J Cell Sci
2000
;
113
:
228
41
.
105
Small
D
,
Kovalenko
D
,
Kacer
D
,
Liaw
L
,
Landriscina
M
,
Di
SC
, et al
Soluble Jagged 1 represses the function of its transmembrane form to induce the formation of the Src-dependent chord-like phenotype
.
J Biol Chem
2001
;
276
:
32022
30
.
106
Weng
AP
,
Nam
Y
,
Wolfe
MS
,
Pear
WS
,
Griffin
JD
,
Blacklow
SC
, et al
Growth Suppression of Pre-T acute lymphoblastic leukemia cells by inhibition of notch signaling
.
Mol Cell Biol
2003
;
23
:
655
64
.
107
Moellering
RE
,
Cornejo
M
,
Davis
TN
,
Del Bianco
C
,
Aster
JC
,
Blacklow
SC
, et al
Direct inhibition of the NOTCH transcription factor complex
.
Nature
2009
;
462
:
182
8
.
108
Sharff
KA
,
Song
W-X
,
Luo
X
,
Tang
N
,
Luo
J
,
Chen
J
, et al
Hey1 basic helix-loop-helix protein plays an important role in mediating BMP9-induced osteogenic differentiation of mesenchymal progenitor cells
.
J Biol Chem
2009
;
284
:
649
59
.
109
Fu
Y
,
Chang
A
,
Chang
L
,
Niessen
K
,
Eapen
S
,
Setiadi
A
, et al
Differential regulation of transforming growth factor β signaling pathways by Notch in human endothelial cells
.
J Biol Chem
2009
;
284
:
19452
62
.
110
Itoh
F
,
Itoh
S
,
Goumans
MJ
,
Valdimarsdottir
G
,
Iso
T
,
Dotto
GP
, et al
Synergy and antagonism between Notch and BMP receptor signaling pathways in endothelial cells
.
EMBO J
2004
;
23
:
541
51
.
111
Xu
J
,
Lamouille
S
,
Derynck
R
. 
TGF-beta-induced epithelial to mesenchymal transition
.
Cell Res
2009
;
19
:
156
72
.
112
Kerr
G
,
Sheldon
H
,
Chaikuad
A
,
Alfano
I
,
Delft
FV
,
Bullock
AN
, et al
A small molecule targeting ALK1 prevents Notch cooperativity and inhibits functional angiogenesis
.
Angiogenesis
2015
;
18
:
209
17
.
113
Bendell
JC
,
Gordon
MS
,
Hurwitz
HI
,
Jones
SF
,
Mendelson
DS
,
Blobe
GC
, et al
Safety, pharmacokinetics, pharmacodynamics, and antitumor activity of dalantercept, an activin receptor-like kinase-1 ligand trap, in patients with advanced cancer
.
Clin Cancer Res
2014
;
20
:
480
9
.
114
Simonelli
M
,
Zucali
P
,
Santoro
A
,
Thomas
MB
,
Braud
FGD
,
Borghaei
H
, et al
Phase I study of PF-03446962, a fully human monoclonal antibody against activin receptor–like kinase 1 in patients with hepatocellular carcinoma
.
Ann Oncol
2016
;
27
:
1782
7
.
115
Necchi
A
,
Giannatempo
P
,
Mariani
L
,
Farè
E
,
Raggi
D
,
Pennati
M
, et al
PF-03446962, a fully-human monoclonal antibody against transforming growth-factor β (TGFβ) receptor ALK1, in pre-treated patients with urothelial cancer: an open label, single-group, phase 2 trial
.
Invest New Drugs
2014
;
32
:
555
60
.
116
Toshihiko
D
,
Lee
KH
,
Kim
TM
,
Ohtsu
A
,
Kim
TY
,
Ikeda
M
, et al
A phase I study of the human anti-activin receptor-like kinase 1 antibody PF-03446962 in Asian patients with advanced solid tumors
.
Cancer Med
2016
;
5
:
1454
63
.
117
Goff
LW
,
Cohen
RB
,
Berlin
J
,
De Braud
FG
,
Lyshchik
A
,
Noberasco
C
, et al
A phase I study of the anti-activin receptor-like kinase 1 (ALK-1) monoclonal antibody PF-03446962 in patients with advanced solid tumors
.
Clin Cancer Res
2016
;
22
:
2146
54
.
118
Wang
X
,
Solban
N
,
Khanna
P
,
Callea
M
,
Song
J
,
Alsop
DC
, et al
Inhibition of ALK1 signaling with dalantercept combined with VEGFR TKI leads to tumor stasis in renal cell carcinoma
.
Oncotarget
2016
;
7
:
41857
69
.
119
Akhurst
RJ
,
Hata
A
. 
Targeting the TGFβ signalling pathway in disease
.
Nat Rev Drug Discov
2012
;
11
:
790
811
.
120
Serova
M
,
Tijeras-Raballand
A
,
Santos
CD
,
Albuquerque
M
,
Paradis
V
,
Neuzillet
C
, et al
Effects of TGF-beta signalling inhibition with galunisertib (LY2157299) in hepatocellular carcinoma models and inex vivowhole tumor tissue samples from patients
.
Oncotarget
2015
;
6
:
348
54
.
121
Rodon
J
,
Carducci
MA
,
Azaro
A
,
Calvo
E
,
Seoane
J
,
Braña
I
, et al
First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma
.
Clin Cancer Res
2015
;
21
:
553
60
.
122
Fujiwara
Y
,
Nokihara
H
,
Yamada
Y
,
Yamamoto
N
,
Sunami
K
,
Utsumi
H
, et al
Phase 1 study of galunisertib, a TGF-beta receptor I kinase inhibitor, in Japanese patients with advanced solid tumors
.
Cancer Chemother Pharmacol
2015
;
76
:
1
10
.
123
David
W
,
Cornelia
W
,
Manfred
G
. 
Hey bHLH transcription factors
.
Curr Topics Dev Biol
2014
;
110
:
285
315
.
124
Fischer
A
,
Gessler
M
. 
Delta-Notch–and then? Protein interactions and proposed modes of repression by Hes and Hey bHLH factors
.
Nucleic Acids Res
2007
;
35
:
4583
96
.
125
Ball
DW
. 
Achaete-scute homolog-1 and Notch in lung neuroendocrine development and cancer
.
Cancer Lett
2004
;
204
:
159
69
.
126
Axelson
H
. 
The Notch signaling cascade in neuroblastoma: role of the basic helix-loop-helix proteins HASH-1 and HES-1
.
Cancer Lett
2004
;
204
:
171
8
.
127
Takata
T
,
Ishikawa
F
. 
Human Sir2-related protein SIRT1 associates with the bHLH repressors HES1 and HEY2 and is involved in HES1- and HEY2-mediated transcriptional repression
.
Biochem Biophys Res Commun
2003
;
301
:
250
7
.
128
Gould
F
,
Harrison
SM
,
Hewitt
EW
,
Whitehouse
A
. 
Kaposi's sarcoma-associated herpesvirus RTA promotes degradation of the Hey1 repressor protein through the ubiquitin proteasome pathway
.
J Virol
2009
;
83
:
6727
38
.
129
Lavery
DN
,
Villaronga
MA
,
Walker
MM
,
Patel
A
,
Belandia
B
,
Bevan
CL
. 
Repression of androgen receptor activity by HEYL, a third member of the Hairy/Enhancer-of-split-related family of Notch effectors
.
J Biol Chem
2011
;
286
:
17796
808
.
130
Weber
D
,
Heisig
J
,
Kneitz
S
,
Wolf
E
,
Eilers
M
,
Gessler
M
. 
Mechanisms of epigenetic and cell-type specific regulation of Hey target genes in ES cells and cardiomyocytes
.
J Mol Cell Cardiol
2015
;
79
:
79
88
.
131
West
AC
,
Johnstone
RW
. 
New and emerging HDAC inhibitors for cancer treatment
.
J Clin Invest
2014
;
124
:
30
9
.
132
Falkenberg
KJ
,
Johnstone
RW
. 
Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders
.
Nat Rev Drug Discov
2014
;
13
:
673
91
.
133
Dovey
OM
,
Foster
CT
,
Conte
N
,
Edwards
SA
,
Edwards
JM
,
Singh
R
, et al
Histone deacetylase 1 and 2 are essential for normal T-cell development and genomic stability in mice
.
Blood
2013
;
121
:
1335
44
.
134
Bhaskara
S
,
Knutson
SG
,
Chandrasekharan
MB
,
Wilson
AJ
,
Zheng
S
,
Yenamandra
A
, et al
Hdac3 is essential for the maintenance of chromatin structure and genome stability
.
Cancer Cell
2010
;
18
:
436
47
.
135
Suresh
P
,
Devaraj
V
,
Srinivas
NR
,
Mullangi
R
. 
Review of bioanalytical assays for the quantitation of various HDAC inhibitors such as vorinostat, belonistat, panobinostat, romidepsin and chidamine
.
Biomed Chromatogr
2017
;
31
:
e3807
.
136
Batlevi
CL
,
Kasamon
Y
,
Bociek
RG
,
Lee
P
,
Gore
L
,
Copeland
A
, et al
ENGAGE-501: phase 2 study of entinostat (SNDX-275) in relapsed and refractory Hodgkin lymphoma
.
Haematologica
2016
;
101
:
968
75
.
137
Ruiz
R
,
Raez
LE
,
Rolfo
C
. 
Entinostat (SNDX-275) for the treatment of non-small cell lung cancer
.
Expert Opin Investig Drugs
2015
;
24
:
1101
9
.
138
Knipstein
J
,
Gore
L
. 
Entinostat for treatment of solid tumors and hematologic malignancies
.
Expert Opin Investig Drugs
2011
;
20
:
1455
67
.
139
Yardley
DA
,
Ismail-Khan
RR
,
Melichar
B
,
Lichinitser
M
,
Munster
PN
,
Klein
PM
, et al
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
.
140
Galli
M
,
Salmoiraghi
S
,
Golay
J
,
Gozzini
A
,
Crippa
C
,
Pescosta
N
, et al
A phase II multiple dose clinical trial of histone deacetylase inhibitor ITF2357 in patients with relapsed or progressive multiple myeloma
.
Ann Hematol
2010
;
89
:
185
90
.
141
Venugopal
B
,
Baird
R
,
Kristeleit
RS
,
Plummer
R
,
Cowan
R
,
Stewart
A
, et al
A phase I study of quisinostat (JNJ-26481585), an oral hydroxamate histone deacetylase inhibitor with evidence of target modulation and antitumor activity, in patients with advanced solid tumors
.
Clin Cancer Res
2013
;
19
:
4262
72
.
142
Banerji
U
,
van Doorn
L
,
Papadatos-Pastos
D
,
Kristeleit
R
,
Debnam
P
,
Tall
M
, et al
A phase I pharmacokinetic and pharmacodynamic study of CHR-3996, an oral class I selective histone deacetylase inhibitor in refractory solid tumors
.
Clin Cancer Res
2012
;
18
:
2687
94
.
143
Ohnuki
H
,
Jiang
K
,
Wang
D
,
Salvucci
O
,
Kwak
H
,
Sánchez-Martín
D
, et al
Tumor-infiltrating myeloid cells activate Dll4/Notch/TGF-β signaling to drive malignant progression
.
Cancer Res
2014
;
74
:
2038
49
.
144
Mendelson
J
,
Song
S
,
Li
Y
,
Maru
DM
,
Mishra
B
,
Davila
M
, et al
Dysfunctional transforming growth factor-β signaling with constitutively active Notch signaling in Barrett's esophageal adenocarcinoma
.
Cancer
2012
;
117
:
3691
702
.
145
Sjölund
J
,
Boström
AK
,
Lindgren
D
,
Manna
S
,
Moustakas
A
,
Ljungberg
B
, et al
The Notch and TGF-β signaling pathways contribute to the aggressiveness of clear cell renal cell carcinoma
.
PLoS One
2011
;
6
:
e23057
.
146
Zhang
J
,
Wang
Y
,
Li
D
,
Jing
S
. 
Notch and TGF-β/Smad3 pathways are involved in the interaction between cancer cells and cancer-associated fibroblasts in papillary thyroid carcinoma
.
Tumor Biol
2014
;
35
:
379
85
.
147
Manigat
L
,
Purow
B
. 
DDEL-16 synergistic combination of an HDAC inhibitor (HDACi) and a Notch inhibitor versus glioblastoma and melanoma cells
.
Neuro-Oncol
2015
;
17
:
v76
v7
.
148
Gilbert
CA
,
Daou
MC
,
Moser
RP
,
Ross
AH
. 
Gamma-secretase inhibitors enhance temozolomide treatment of human gliomas by inhibiting neurosphere repopulation and xenograft recurrence
.
Cancer Res
2010
;
70
:
6870
9
.
149
Fuino
L
,
Bali
P
,
Wittmann
S
,
Donapaty
S
,
Guo
F
,
Yamaguchi
H
, et al
Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells to trastuzumab, taxotere, gemcitabine, and epothilone B
.
Mol Cancer Ther
2003
;
2
:
971
84
.
150
Ren
XF
,
Mu
LP
,
Jiang
YS
,
Wang
L
,
Ma
JF
. 
LY2109761 inhibits metastasis and enhances chemosensitivity in osteosarcoma MG-63 cells
.
Eur Rev Med Pharmacol Sci
2015
;
19
:
1182
90
.
151
Loconte
NK
,
Razak
ARA
,
Ivy
P
,
Tevaarwerk
A
,
Leverence
R
,
Kolesar
J
, et al
A multicenter phase 1 study of γ -secretase inhibitor RO4929097 in combination with capecitabine in refractory solid tumors
.
Invest New Drugs
2014
;
33
:
169
76
.
152
Richter
S
,
Bedard
PL
,
Chen
EX
,
Clarke
BA
,
Tran
B
,
Hotte
SJ
, et al
A phase I study of the oral gamma secretase inhibitor R04929097 in combination with gemcitabine in patients with advanced solid tumors (PHL-078/CTEP 8575)
.
Invest New Drugs
2014
;
32
:
243
9
.
153
Shin
DY
,
Kim
SJ
,
Yoon
DH
,
Yong
P
,
Kong
JH
,
Kim
JA
, et al
Results of a phase II study of vorinostat in combination with intravenous fludarabine, mitoxantrone, and dexamethasone in patients with relapsed or refractory mantle cell lymphoma: an interim analysis
.
Cancer Chemother Pharmacol
2016
;
77
:
1
9
.
154
Thomas
A
,
Rajan
A
,
Szabo
E
,
Tomita
Y
,
Carter
CA
,
Scepura
B
, et al
A Phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study
.
Clin Cancer Res
2014
;
20
:
5392
402
.
155
Matulonis
U
,
Berlin
S
,
Lee
H
,
Whalen
C
,
Obermayer
E
,
Penson
R
, et al
Phase I study of combination of vorinostat, carboplatin, and gemcitabine in women with recurrent, platinum-sensitive epithelial ovarian, fallopian tube, or peritoneal cancer
.
Cancer Chemother Pharmacol
2015
;
76
:
417
23
.
156
Akerley
W
,
Mccoy
J
,
Hesketh
PJ
,
Goodwin
JW
,
Bearden
JD
,
Atkins
JN
, et al
Gemcitabine and irinotecan for patients with untreated extensive stage small cell lung cancer: SWOG 0119
.
J Thorac Oncol
2007
;
2
:
526
30
.
157
Yoo
C
,
Ryu
MH
,
Na
YS
,
Ryoo
BY
,
Lee
CW
,
Kang
YK
. 
Vorinostat in combination with capecitabine plus cisplatin as a first-line chemotherapy for patients with metastatic or unresectable gastric cancer: phase II study and biomarker analysis
.
Br J Cancer
2016
;
114
:
1185
90
.
158
Fischer
A
,
Klattig
J
,
Kneitz
B
,
Diez
H
,
Maier
M
,
Holtmann
B
, et al
Hey basic helix-loop-helix transcription factors are repressors of GATA4 and GATA6 and restrict expression of the GATA target gene ANF in fetal hearts
.
Mol Cell Biol
2005
;
25
:
8960
70
.
159
Sun
J
,
Kamei
CN
,
Layne
MD
,
Jain
MK
,
Liao
JK
,
Lee
ME
, et al
Regulation of myogenic terminal differentiation by the hairy-related transcription factor CHF2
.
J Biol Chem
2001
;
276
:
18591
6
.
160
Arai
MA
,
Masada
A
,
Ohtsuka
T
,
Kageyama
R
,
Ishibashi
M
. 
The first Hes1 dimer inhibitors from natural products
.
Bioorg Med Chem Lett
2009
;
19
:
5778
81
.
161
Müller
P
,
Kietz
S
,
Gustafsson
J
,
Ström
A
. 
The Anti-estrogenic effect of all-trans-retinoic acid on the breast cancer cell line MCF-7 is dependent on HES-1 expression
.
J Biol Chem
2002
;
277
:
28376
9
.
162
Danza
G
,
Di Serio
C
,
Rosati
F
,
Lonetto
G
,
Sturli
N
,
Kacer
D
, et al
Notch signaling modulates hypoxia-induced neuroendocrine differentiation of human prostate cancer cells
.
Mol Cancer Res
2012
;
10
:
230
8
.
163
Skinner
MK
,
Rawls
A
,
Wilsonrawls
J
,
Roalson
EH
. 
Basic helix-loop-helix transcription factor gene family phylogenetics and nomenclature
.
Differentiation
2010
;
80
:
1
8
.
164
Tsigelny
IF
,
Kouznetsova
VL
,
Pingle
SC
,
Kesari
S
. 
bHLH Transcription factors inhibitors for cancer therapy: general features for in silico drug design
.
Curr Med Chem
2014
;
21
:
3227
43
.
165
Yin
X
,
Giap
C
,
Lazo
JS
,
Prochownik
EV
. 
Low molecular weight inhibitors of Myc–Max interaction and function
.
Oncogene
2003
;
22
:
6151
9
.
166
Kiessling
A
,
Sperl
B
,
Hollis
A
,
Eick
D
,
Berg
T
. 
Selective inhibition of c-Myc/Max dimerization and DNA binding by small molecules
.
Chem Biol
2006
;
13
:
745
51
.
167
Hammoudeh
DI
,
Follis
AV
,
Prochownik
EV
,
Metallo
SJ
. 
Multiple independent binding sites for small-molecule inhibitors on the oncoprotein c-Myc
.
J Am Chem Soc
2009
;
131
:
7390
401
.
168
Scheuermann
TH
,
Li
Q
,
Ma
HW
,
Key
J
,
Zhang
L
,
Chen
R
, et al
Allosteric inhibition of hypoxia inducible factor-2 with small molecules
.
Nat Chem Biol
2013
;
9
:
271
6
.
169
Lee
KA
,
Zhang
H
,
Qian
DZ
,
Rey
S
,
Liu
JO
,
Semenza
GL
. 
Acriflavine inhibits HIF-1 dimerization, tumor growth, and vascularization
.
Proc Natl Acad Sci U S A
2009
;
106
:
17910
5
.
170
Ghosh
I
,
Chmielewski
J
. 
A β-sheet peptide inhibitor of E47 dimerization and DNA binding
.
Chem Biol
1998
;
5
:
439
45
.
171
Chen
CH
,
Kuo
SC
,
Huang
LJ
,
Hsu
MH
,
Lung
FDT
. 
Affinity of synthetic peptide fragments of MyoD for Id1 protein and their biological effects in several cancer cells
.
J Pept Sci
2010
;
16
:
231
41
.