Acessibilidade / Reportar erro

Enhancer in cancer pathogenesis and treatment

Abstract

Enhancers are essential cis-acting regulatory elements that determine cell identity and tumor progression. Enhancer function is dependent on the physical interaction between the enhancer and its target promoter inside its local chromatin environment. Enhancer reprogramming is an important mechanism in cancer pathogenesis and can be driven by both cis and trans factors. Super enhancers are acquired at oncogenes in numerous cancer types and represent potential targets for cancer treatment. BET and CDK inhibitors act through mechanisms of enhancer function and have shown promising results in therapy for various types of cancer. Genome editing is another way to reprogram enhancers in cancer treatment. The relationship between enhancers and cancer has been revised by several authors in the past few years, which mainly focuses on the mechanisms by which enhancers can impact cancer. Here, we emphasize SE’s role in cancer pathogenesis and the new therapies involving epigenetic regulators (BETi and CDKi). We suggest that understanding mechanisms of activity would aid clinical success for these anti-cancer agents.

Keywords:
Enhancer; super-enhancer; enhancer reprogramming; cancer; BETi

Characteristics and types of enhancers

Characteristics of enhancers

Enhancers are orientation-independent cis-acting regulatory elements that increase transcription activity from a distant promoter. Enhancer regions have higher DNA accessibility and nucleosomes in enhancer regions have signature histone modifications such as H3K4me1 and H3K4me2 and are usually depleted of H3K4me3 (Kundaje et al., 2015Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, Heravi-Moussavi A, Kheradpour P, Zhang Z, Wang J, Ziller MJ et al. (2015) Integrative analysis of 111 reference human epigenomes. Nature 518:317-330.). There are four enhancer-activation states: inactive, primed, poised and active. Inactive enhancers are buried in compact heterochromatin and have no transcription factor association. Primed enhancers are bound by transcription factors and are inside Dnase I hypersensitive open chromatin, but still need further signal or cofactors binding to exert active enhancer function. Poised enhancers are mostly found in embryonic stem cells and are primed enhancers with repressive histone modifications, such as H3K27me3 (Rada-Iglesias et al., 2011Rada-Iglesias A, Bajpai R, Swigut T, Brugmann SA, Flynn RA and Wysocka J (2011) A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470:279-283.). Active enhancers are marked by H3K27Ac. They are actively transcribed into enhancer RNA (eRNA) by RNA polymerase II and function to boost target gene expression.

Types of enhancers

There are mainly two types of enhancers depending on their activation stimuli and function: cell type-specific enhancers and signal-dependent enhancers (also called inducible enhancers). Cell type-specific enhancers represent a large proportion of all enhancers. In a recent study, researchers identified active enhancers across 10 human tissues, and most of them are tissue-specific enhancers (Xiong et al., 2018Xiong L, Kang R, Ding R, Kang W, Zhang Y, Liu W, Huang Q, Meng J and Guo Z (2018) Genome-wide identification and characterization of enhancers across 10 human tissues. Int J Biol Sci 14:1321-1332.). Enhancer-target networks and enhancer RNA profiles are robust identifying features for different cell and tissue lineages(Cao et al., 2017Cao Q, Anyansi C, Hu X, Xu L, Xiong L, Tang W, Mok MTS, Cheng C, Fan X, Gerstein M et al. (2017) Reconstruction of enhancer-target networks in 935 samples of human primary cells, tissues and cell lines. Nat Genet 49:1428-1436.; Tu et al., 2021Tu YH, Juan HF and Huang HC (2021) Identification of cell states using super-enhancer RNA. BMC Genomics 22:787.). All different cell types in the human body contain the same genome, and one of the vital factors that determines cell type-specific gene expression is cell type-specific enhancers. Although mammalian genomes contain millions of potential enhancers, only a small percentage is active in any given cell type. For a specific gene locus such as T-cell acute lymphocytic leukaemia 1 (TAL1), several developmental enhancers have been identified and different choices and combinations of these enhancers are used for different cell types (Heinz et al., 2015Heinz S, Romanoski CE, Benner C and Glass CK (2015) The selection and function of cell type-specific enhancers. Nat Rev Mol Cell Biol 16:144-154.). The -3.8kb (upstream) and +19kb (downstream) enhancers drive TAL1 expression in human umbilical vein endothelial cells and hematopoietic stem and progenitor cells (Sánchez et al., 1999Sánchez M, Göttgens B, Sinclair AM, Stanley M, Begley CG, Hunter S and Green AR (1999) An SCL 3’ enhancer targets developing endothelium together with embryonic and adult haematopoietic progenitors. Development 126:3891-3904.; Göttgens et al., 2004Göttgens B, Broccardo C, Sanchez MJ, Deveaux S, Murphy G, Göthert JR, Kotsopoulou E, Kinston S, Delaney L, Piltz S et al. (2004) The scl +18/19 stem cell enhancer is not required for hematopoiesis: identification of a 5’ bifunctional hematopoietic-endothelial enhancer bound by Fli-1 and Elf-1. Mol Cell Biol 24:1870-1883.), and the +51kb enhancer is essential for TAL1 transcription in K562 erythroid cells (Delabesse et al., 2005Delabesse E, Ogilvy S, Chapman MA, Piltz SG, Gottgens B and Green AR (2005) Transcriptional regulation of the SCL locus: Identification of an enhancer that targets the primitive erythroid lineage in vivo. Mol Cell Biol 25:5215-5225.). These enhancers are activated according to the cell’s specific developmental stage and environmental stimuli and work to boost the expression of cell identity genes.

Pioneer TFs, lineage-dependent TFs (LDTFs), and signal-dependent TFs (SDTFs) work collaboratively to select and activate inactive and poised enhancers and establish lineage-specific gene expression (Heinz et al., 2010Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre C, Singh H and Glass CK (2010) Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38:576-589.). Chromatin remodelers and histone modifiers are also important players in the activation of cell-type specific enhancers (Park et al., 2021Park YK, Lee JE, Yan Z, McKernan K, O’Haren T, Wang W, Peng W and Ge K (2021) Interplay of BAF and MLL4 promotes cell type-specific enhancer activation. Nat Commun 12:1630.). There are two types of mechanisms by which LDTFs and SDTFs work together to select and activate cell type-specific enhancers (Heinz et al., 2015Heinz S, Romanoski CE, Benner C and Glass CK (2015) The selection and function of cell type-specific enhancers. Nat Rev Mol Cell Biol 16:144-154.). For one mechanism, there is a hierarchical relationship between LDTFs and SDTFs binding, where LDTFs act as pioneer factors that initially select enhancers and the binding of SDTFs can further induce the enhancer activity. For the other mechanism, SDTFs contribute directly to enhancer selection through collaborative interactions with LDTFs.

While cell type-specific enhancers play a vital part in cell-type determination, some enhancers serve as main regulators of gene expression in response to various acute signaling pathways, where signal-dependent transcription factors preferentially bind to enhancers (Tan et al., 2023Tan Y, Yao L, Gamliel A, Nair SJ, Taylor H, Ohgi K, Aggarwal AK and Rosenfeld MG (2023) Signal-induced enhancer activation requires Ku70 to read topoisomerase1-DNA covalent complexes. Nat Struct Mol Biol 30:148-158.). These enhancers belong to signal-dependent enhancers. Examples of signal-dependent enhancers include hormone-responsive enhancers (Shlyueva et al., 2014Shlyueva D, Stelzer C, Gerlach D, Yáñez-Cuna JO, Rath M, Boryń Ł M, Arnold CD and Stark A (2014) Hormone-responsive enhancer-activity maps reveal predictive motifs, indirect repression, and targeting of closed chromatin. Mol Cell 54:180-192.; Hoffman et al., 2022Hoffman JA, Trotter KW, Day CR, Ward JM, Inoue K, Rodriguez J and Archer TK (2022) Multimodal regulatory elements within a hormone-specific super enhancer control a heterogeneous transcriptional response. Mol Cell 82:803-815.e805.), virus-inducible enhancers (Thanos et al., 1993Thanos D, Du W and Maniatis T (1993) The high mobility group protein HMG I(Y) is an essential structural component of a virus-inducible enhancer complex. Cold Spring Harb Symp Quant Biol 58:73-81., 1996Thanos D (1996) Mechanisms of transcriptional synergism of eukaryotic genes. The interferon-beta paradigm. Hypertension 27:1025-1029.), metal-responsive enhancers (Karin et al., 1987Karin M, Haslinger A, Heguy A, Dietlin T and Cooke T (1987) Metal-responsive elements act as positive modulators of human metallothionein-IIA enhancer activity. Mol Cell Biol 7:606-613.; Westin and Schaffner, 1988Westin G and Schaffner W (1988) A zinc-responsive factor interacts with a metal-regulated enhancer element (MRE) of the mouse metallothionein-I gene. EMBO J 7:3763-3770.), and NF-kappa B and cytokine-inducible enhancers (Collins et al., 1995Collins T, Read MA, Neish AS, Whitley MZ, Thanos D and Maniatis T (1995) Transcriptional regulation of endothelial cell adhesion molecules: NF-kappa B and cytokine-inducible enhancers. FASEB J 9:899-909.).

Super-enhancer

Super-enhancer (SE) is the term used to describe clusters of active enhancers that are in a high density in a genomic region. Super-enhancers have the function of regulating genes essential for cell identity determination. SEs are enriched with more TFs, Mediator complexes, and RNA Pol II molecules than typical enhancers, which results in higher transcriptional activity (Yoshino and Suzuki, 2022Yoshino S and Suzuki HI (2022) The molecular understanding of super-enhancer dysregulation in cancer. Nagoya J Med Sci 84:216-229.). BRD4, p300, CDK7, CDK9, and MED1 (Mediator Complex Subunit 1) are all important factors that characterize SEs (Khan and Zhang, 2019Khan A and Zhang X (2019) Integrative modeling reveals key chromatin and sequence signatures predicting super-enhancers. Sci Rep 9:2877.). High concentrations of transcription factors, co-activators (BRD4 and p300), and RNA polymerase II forms transcriptional condensates to drive the interaction between promoter and enhancer. SE has been implicated in the pathogenesis of various types of cancer. SEs are extremely sensitive to perturbations by drugs (Bradner et al., 2017Bradner JE, Hnisz D and Young RA (2017) Transcriptional addiction in cancer. Cell 168:629-643.). A small change in concentrations of SE components causes drastic changes in SE-associated gene expression (Lovén et al., 2013Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.). This has been utilized when exploring potential therapy to treat cancer and will be discussed in detail in part 4.1 targeting mediators of super-enhancer function.

Molecular mechanism of enhancer function

Polymerase II and eRNA

Polymerase II is recruited to active enhancers and produces short transcripts. Pol II is then transferred from enhancers to promoters to initiate transcription at the target gene (Gibbons et al., 2022Gibbons MD, Fang Y, Spicola AP, Linzer N, Jones SM, Johnson BR, Li L, Xie M and Bungert J (2022) Enhancer-mediated formation of nuclear transcription initiation domains. Int J Mol Sci 23:9290.). SEs are characterized by abundant association with Pol II and are most sensitive to interference with Pol II function. Inhibition of Pol II function through CDK7 could be utilized in cancer therapy and is discussed in detail in part 4.1.2 CDK7 inhibitors.

The transcripts that come from the transcription of enhancers are called enhancer RNA (eRNA). Most eRNAs are short transcripts (around 500bp) that are non-polyadenylated and unspliced (Andersson et al., 2014Andersson R, Refsing Andersen P, Valen E, Core LJ, Bornholdt J, Boyd M, Heick Jensen T and Sandelin A (2014b) Nuclear stability and transcriptional directionality separate functionally distinct RNA species. Nat Commun 5:5336.a,b). Only a small number of eRNAs are long (several kb in size) that are polyadenylated (Koch et al., 2011Koch F, Fenouil R, Gut M, Cauchy P, Albert TK, Zacarias-Cabeza J, Spicuglia S, de la Chapelle AL, Heidemann M, Hintermair C et al. (2011) Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat Struct Mol Biol 18:956-963.). eRNA production is predictive of active enhancer function (Melgar et al., 2011Melgar MF, Collins FS and Sethupathy P (2011) Discovery of active enhancers through bidirectional expression of short transcripts. Genome Biol 12:R113.; Andersson et al., 2014aAndersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, Boyd M, Chen Y, Zhao X, Schmidl C, Suzuki T et al. (2014a) An atlas of active enhancers across human cell types and tissues. Nature 507:455-461.; Core et al., 2014Core LJ, Martins AL, Danko CG, Waters CT, Siepel A and Lis JT (2014) Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers. Nat Genet 46:1311-1320.; Henriques et al., 2018Henriques T, Scruggs BS, Inouye MO, Muse GW, Williams LH, Burkholder AB, Lavender CA, Fargo DC and Adelman K (2018) Widespread transcriptional pausing and elongation control at enhancers. Genes Dev 32:26-41.) and eRNA level correlates with the transcriptional activity of their target gene (Kim et al., 2010Kim TK, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara-Haley K, Kuersten S et al. (2010) Widespread transcription at neuronal activity-regulated enhancers. Nature 465:182-187.). Transcription from enhancers can be unidirectional (Koch et al., 2011Koch F, Fenouil R, Gut M, Cauchy P, Albert TK, Zacarias-Cabeza J, Spicuglia S, de la Chapelle AL, Heidemann M, Hintermair C et al. (2011) Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat Struct Mol Biol 18:956-963.) but is mostly bidirectional (Kim et al., 2010Kim TK, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara-Haley K, Kuersten S et al. (2010) Widespread transcription at neuronal activity-regulated enhancers. Nature 465:182-187.). eRNA is typically unstable (Lubas et al., 2015Lubas M, Andersen PR, Schein A, Dziembowski A, Kudla G and Jensen TH (2015) The human nuclear exosome targeting complex is loaded onto newly synthesized RNA to direct early ribonucleolysis. Cell Rep 10:178-192.), so it is not always detectable even when the enhancer is functional (Andersson et al., 2014aAndersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, Boyd M, Chen Y, Zhao X, Schmidl C, Suzuki T et al. (2014a) An atlas of active enhancers across human cell types and tissues. Nature 507:455-461.; Mikhaylichenko et al., 2018Mikhaylichenko O, Bondarenko V, Harnett D, Schor IE, Males M, Viales RR and Furlong EEM (2018) The degree of enhancer or promoter activity is reflected by the levels and directionality of eRNA transcription. Genes Dev 32:42-57.). There has been evidence that eRNA might be contributing to enhancer function through several mechanisms, including increasing chromatin accessibility (Mousavi et al., 2013Mousavi K, Zare H, Dell’orso S, Grontved L, Gutierrez-Cruz G, Derfoul A, Hager GL and Sartorelli V (2013) eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol Cell 51:606-617.), recruitment of cofactors (Kaikkonen et al., 2013Kaikkonen MU, Spann NJ, Heinz S, Romanoski CE, Allison KA, Stender JD, Chun HB, Tough DF, Prinjha RK, Benner C et al. (2013) Remodeling of the enhancer landscape during macrophage activation is coupled to enhancer transcription. Mol Cell 51:310-325.; Bose et al., 2017Bose DA, Donahue G, Reinberg D, Shiekhattar R, Bonasio R and Berger SL (2017) RNA binding to CBP stimulates histone acetylation and transcription. Cell 168:135-149.e122.), maintenance of transcription factor binding (Sigova et al., 2015Sigova AA, Abraham BJ, Ji X, Molinie B, Hannett NM, Guo YE, Jangi M, Giallourakis CC, Sharp PA and Young RA (2015) Transcription factor trapping by RNA in gene regulatory elements. Science 350:978-981.), enhancer-promoter contact (Li et al., 2013Li W, Notani D, Ma Q, Tanasa B, Nunez E, Chen AY, Merkurjev D, Zhang J, Ohgi K, Song X et al. (2013) Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498:516-520.), and phase separation (Nair et al., 2019Nair SJ, Yang L, Meluzzi D, Oh S, Yang F, Friedman MJ, Wang S, Suter T, Alshareedah I, Gamliel A et al. (2019) Phase separation of ligand-activated enhancers licenses cooperative chromosomal enhancer assembly. Nat Struct Mol Biol 26:193-203.).

Promoter-enhancer interaction

The function of an active enhancer is dependent on the physical interaction between the enhancer and its target promoter. Several models have been proposed for enhancer-promoter communication, including tracking, chaining, and looping (the loop extrusion model) (Furlong and Levine, 2018Furlong EEM and Levine M (2018) Developmental enhancers and chromosome topology. Science 361:1341-1345.). In the tracking model, Pol II binds to an enhancer through interaction with transcription factors and tracks along the chromatin, pulling the enhancer with it until it reaches its target promoter. In the chaining model, TFs bound to the enhancer oligomerize and form a chain to interact with the target promoter. In principle, the tracking and chaining model could only work in short-range interactions, and the most widely accepted model of action is the loop extrusion model. The loop extrusion model incorporates looping and tracking. In the loop extrusion model, cohesin complexes form tripartite rings around chromatin and translocate along the chromatin fiber in opposite directions, therefore actively extruding a progressively larger chromatin loop until they are stopped by CTCF boundary elements (Fudenberg et al., 2016Fudenberg G, Imakaev M, Lu C, Goloborodko A, Abdennur N and Mirny LA (2016) Formation of chromosomal domains by loop Extrusion. Cell Rep 15:2038-2049.). The chromatin loop, formed between the enhancer and its target promoter, is called an enhancer-promoter loop. The enhancer-promoter loop provides the structural basis for enhancer function. There are many cofactors that are involved in the enhancer-promoter loop, such as CTCF, cohesin, BRD4, the Mediator complex, RNA Polymerase II, chromatin modifiers, transcription factors, pioneer factors, and transcription coactivators. It is important to note that some evidence shows that some regulatory elements might have both enhancer and promoter functions, and transcription initiation and transcriptional enhancement may not be mutually exclusive functions for a specific regulatory element (Andersson and Sandelin, 2020Andersson R and Sandelin A (2020) Determinants of enhancer and promoter activities of regulatory elements. Nat Rev Genet 21:71-87.).

TAD

Since enhancers can be as much as 1Mb away from their interacting promoters (Furlong and Levine, 2018Furlong EEM and Levine M (2018) Developmental enhancers and chromosome topology. Science 361:1341-1345.), their interaction is based on the 3D organization of the genome (Robson et al., 2019Robson MI, Ringel AR and Mundlos S (2019) Regulatory landscaping: How enhancer-promoter communication is sculpted in 3D. Mol Cell 74:1110-1122.; van Steensel and Furlong, 2019van Steensel B and Furlong EEM (2019) The role of transcription in shaping the spatial organization of the genome. Nat Rev Mol Cell Biol 20:327-337.). Enhancers work in the context of chromatin domains and preferentially interact with promoters that are in the same topological associating domains (TAD) rather than a nearby TAD (Symmons et al., 2014Symmons O, Uslu VV, Tsujimura T, Ruf S, Nassari S, Schwarzer W, Ettwiller L and Spitz F (2014) Functional and topological characteristics of mammalian regulatory domains. Genome Res 24:390-400.). Disruption of TADs could cause improper enhancer-promoter interactions that result in pathogenic phenotypes (Lupiáñez et al., 2015Lupiáñez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, Horn D, Kayserili H, Opitz JM, Laxova R et al. (2015) Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161:1012-1025.).

Phase separation

Phase separation has been recently discovered to be an important part of enhancer function. Phase separation is the formation of membraneless organelles inside the cell when groups of molecules interact with each other. Phase separation plays an important part in enhancer function and gene regulation (Sabari et al., 2018Sabari BR, Dall’Agnese A, Boija A, Klein IA, Coffey EL, Shrinivas K, Abraham BJ, Hannett NM, Zamudio AV, Manteiga JC et al. (2018) Coactivator condensation at super-enhancers links phase separation and gene control. Science 361:eaar3958.; Nair et al., 2019Nair SJ, Yang L, Meluzzi D, Oh S, Yang F, Friedman MJ, Wang S, Suter T, Alshareedah I, Gamliel A et al. (2019) Phase separation of ligand-activated enhancers licenses cooperative chromosomal enhancer assembly. Nat Struct Mol Biol 26:193-203.; Zhang et al., 2021Zhang J, Yue W, Zhou Y, Liao M, Chen X and Hua J (2021) Super enhancers-functional cores under the 3D genome. Cell Prolif 54:e12970.; Lee et al., 2022Lee R, Kang MK, Kim YJ, Yang B, Shim H, Kim S, Kim K, Yang CM, Min BG, Jung WJ et al. (2022) CTCF-mediated chromatin looping provides a topological framework for the formation of phase-separated transcriptional condensates. Nucleic Acids Res 50:207-226.). On the other hand, it is also shown to be essential in decommissioning of enhancers (Jia et al., 2021Jia P, Li X, Wang X, Yao L, Xu Y, Hu Y, Xu W, He Z, Zhao Q, Deng Y et al. (2021) ZMYND8 mediated liquid condensates spatiotemporally decommission the latent super-enhancers during macrophage polarization. Nat Commun 12:6535.). It is also discovered to be an important mechanism of aberrant chromatin looping and cancer development (Ahn et al., 2021Ahn JH, Davis ES, Daugird TA, Zhao S, Quiroga IY, Uryu H, Li J, Storey AJ, Tsai YH, Keeley DP et al. (2021) Phase separation drives aberrant chromatin looping and cancer development. Nature 595:591-595.; Owen et al., 2021Owen I, Yee D, Wyne H, Perdikari TM, Johnson V, Smyth J, Kortum R, Fawzi NL and Shewmaker F (2021) The oncogenic transcription factor FUS-CHOP can undergo nuclear liquid-liquid phase separation. J Cell Sci 134:jcs258578.; Kabra and Bushweller, 2022Kabra A and Bushweller J (2022) The Intrinsically Disordered Proteins MLLT3 (AF9) and MLLT1 (ENL) - Multimodal transcriptional switches with roles in normal hematopoisis. MLL fusion leukemia and kidney cancer. J Mol Biol 434:167117.; Suzuki and Onimaru, 2022Suzuki HI and Onimaru K (2022) Biomolecular condensates in cancer biology. Cancer Sci 113:382-391.).

Function of enhancer in cancer

Enhancer reprogramming in cancer

There is extensive enhancer reprogramming resulting in the expression of essential players in cancer invasive progression in various types of cancer (Roe et al., 2017Roe JS, Hwang CI, Somerville TDD, Milazzo JP, Lee EJ, Da Silva B, Maiorino L, Tiriac H, Young CM, Miyabayashi K et al. (2017) Enhancer repogramming promotes anceratic cancer metastasis. Cell 170:875-888.e820.; Teng et al., 2020Teng S, Li YE, Yang M, Qi R, Huang Y, Wang Q, Zhang Y, Chen S, Li S, Lin K et al. (2020) Tissue-specific transcription reprogramming promotes liver metastasis of colorectal cancer. Cell Res 30:34-49.; Yi et al., 2020Yi M, Tan Y, Wang L, Cai J, Li X, Zeng Z, Xiong W, Li G, Li X, Tan P et al. (2020) TP63 links chromatin remodeling and enhancer reprogramming to epidermal differentiation and squamous cell carcinoma development. Cell Mol Life Sci 77:4325-4346.; Zhou et al., 2020Zhou J, Wang D, Tang D and Huang W (2020) Abnormal activations of super-enhancers enhance the carcinogenicity in lung adenocarcinoma. Cancer Manag Res 12:8509-8518.; Ye et al., 2021Ye B, Fan D, Xiong W, Li M, Yuan J, Jiang Q, Zhao Y, Lin J, Liu J, Lv Y et al. (2021) Oncogenic enhancers drive esophageal squamous cell carcinogenesis and metastasis. Nat Commun 12:4457.; Huang et al., 2022Huang P, Zhang B, Zhao J and Li MD (2022) Integrating the epigenome and transcriptome of hepatocellular carcinoma to identify systematic enhancer abberations and establish an abberant enhancers-related prognostic signature. Front Cell Dev Biol 10:827657.). Some enhancers gained activity and drive the expression of oncogenes, while others lose their enhancer activity, which may result in the repression of tumor suppressor genes.

cis-acting factors that drive oncogenic enhancer reprogramming

Both cis-elements and trans-acting factors can induce enhancer reprogramming in cancer progression. Cis-acting alterations that drive oncogenic enhancer activity include single-nucleotide polymorphisms (SNPs), small insertions or deletions (INDELs), and enhancer hijacking. SNPs and INDELs represent hereditary cancer predisposition, whereas enhancer hijacking is done through somatic chromosomal rearrangements. SNPs and INDELS result in the gain or loss of enhancer function by creating new or disrupting existing TF binding sites (Figure 1). Enhancer hijacking is the utilization of otherwise harmless enhancers to drive oncogene expression. Large chromosomal rearrangements, including deletions, translocations, inversions, and copy number changes, are responsible for enhancer hijacking (Figure 2).

Figure 1
SNP and INDELs can disrupt TF binding motifs in existing enhancers or create new binding motifs for new enhancers, which results in oncogenic gene expression program.

Figure 2
Enhancer hijacking resulting from chromosomal rearrangements can also lead to oncogenic gene expression.

Large amounts of SNPs linked to diseases have been found to be in noncoding regions and the majority of these SNPs are located in enhancer regions (Hindorff et al., 2009Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS and Manolio TA (2009) Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A 106:9362-9367.; Maurano et al., 2012Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, Reynolds AP, Sandstrom R, Qu H, Brody J et al. (2012) Systematic localization of common disease-associated variation in regulatory DNA. Science 337:1190-1195.; Hnisz et al., 2013Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-André V, Sigova AA, Hoke HA and Young RA (2013) Super-enhancers in the control of cell identity and disease. Cell 155:934-947.; Weinhold et al., 2014Weinhold N, Jacobsen A, Schultz N, Sander C and Lee W (2014) Genome-wide analysis of noncoding regulatory mutations in cancer. Nat Genet 46:1160-1165.; Nasser et al., 2021Nasser J, Bergman DT, Fulco CP, Guckelberger P, Doughty BR, Patwardhan TA, Jones TR, Nguyen TH, Ulirsch JC, Lekschas F et al. (2021) Genome-wide enhancer maps link risk variants to disease genes. Nature 593:238-243.). The SNP rs2168101 within the SE of the neuroblastoma oncogene LMO1 influences neuroblastoma susceptibility through differential GATA TF binding and regulation of LMO1 expression (Oldridge et al., 2015Oldridge DA, Wood AC, Weichert-Leahey N, Crimmins I, Sussman R, Winter C, McDaniel LD, Diamond M, Hart LS, Zhu S et al. (2015) Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature 528:418-421.). INDELs acquired upstream of the TAL1 oncogene introduce de novo binding motifs for the TF MYB, which creates a SE and drives TAL1 overexpression in primary patient T-cell acute lymphoblastic leukemia (T-ALL) (Mansour et al., 2014Mansour MR, Abraham BJ, Anders L, Berezovskaya A, Gutierrez A, Durbin AD, Etchin J, Lawton L, Sallan SE, Silverman LB et al. (2014) Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346:1373-1377.).

Chromosomal translocation causing the repositioning of a single enhancer could result in aberrant expression of oncogene EVI1 and acute myeloid leukemia (Gröschel et al., 2014Gröschel S, Sanders MA, Hoogenboezem R, de Wit E, Bouwman BAM, Erpelinck C, van der Velden VHJ, Havermans M, Avellino R, van Lom K et al. (2014) A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157:369-381.). Structural variants that juxtapose GFI1 (Growth Factor Independent 1) family oncogenes proximal to active enhancers are discovered to instigate oncogenic activities in medulloblastoma (Northcott et al., 2014Northcott PA, Lee C, Zichner T, Stütz AM, Erkek S, Kawauchi D, Shih DJ, Hovestadt V, Zapatka M, Sturm D et al. (2014) Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511:428-434.). Duplication of an enhancer region near the androgen receptor (AR) locus has been found in advanced prostate cancer that causes therapeutic resistance (Takeda et al., 2018Takeda DY, Spisák S, Seo JH, Bell C, O’Connor E, Korthauer K, Ribli D, Csabai I, Solymosi N, Szállási Z et al. (2018) A somatically acquired enhancer of androgen receptor is a noncoding driver of advamced prostate cancer. Cell 174:422-432.e413.). The duplication causes enhanced AR expression, which undermines the effectiveness of clinical treatment targeting the AR signaling pathway.

trans-acting factors that drive oncogenic enhancer reprogramming

Besides cis-elements that define the intrinsic ability of an enhancer region to attract TF binding, another important factor is the chromatin landscape, which determines whether the DNA of a robust enhancer is accessible for TF to bind to initiate gene expression. This important aspect of oncogenic enhancer reprogramming involves epigenetic modifications of the enhancers. A myriad of trans-acting factors play essential roles in enhancer epigenetic modification, such as chromatin remodelers, epigenetic modifiers, and pioneer TFs.

Chromatin remodelers maintain or change chromatin landscape and DNA accessibility by moving or ejecting nucleosomes. Mutations in the SWI/SNF family of chromatin remodeler account for about 20% of all human cancers (St Pierre and Kadoch, 2017St Pierre R and Kadoch C (2017) Mammalian SWI/SNF complexes in cancer: emerging therapeutic opportunities. Curr Opin Genet Dev 42:56-67.). The ARID1A subunit targets SWI/SNF complex to enhancers and loss of ARID1A impairs the enhancer-mediated transcriptional program of colonic epithelium and drives colon cancer in mice (Mathur et al., 2017Mathur R, Alver BH, San Roman AK, Wilson BG, Wang X, Agoston AT, Park PJ, Shivdasani RA and Roberts CW (2017) ARID1A loss impairs enhancer-mediated gene regulation and drives colon cancer in mice. Nat Genet 49:296-302.). Besides chromatin remodelers that move nucleosomes around, the type of histone variant used in nucleosomes is also an important regulator of enhancer activity in cancer cells. In breast cancer cell lines, H2A.Z occupancy is linked to enhancer activation (Brunelle et al., 2015Brunelle M, Nordell Markovits A, Rodrigue S, Lupien M, Jacques P and Gévry N (2015) The histone variant H2A.Z is an important regulator of enhancer activity. Nucleic Acids Res 43:9742-9756.). Other epigenetic modifiers, such as DNA methyltransferases (Lu et al., 2016Lu R, Wang P, Parton T, Zhou Y, Chrysovergis K, Rockowitz S, Chen WY, Abdel-Wahab O, Wade PA, Zheng D et al. (2016) Epigenetic perturbations by Arg882-mutated DNMT3A potentiate aberrant stem cell gene-expression program and acute leukemia development. Cancer Cell 30:92-107.; Yang et al., 2016Yang L, Rodriguez B, Mayle A, Park HJ, Lin X, Luo M, Jeong M, Curry CV, Kim SB, Ruau D et al. (2016) DNMT3A loss drives enhancer hypomehtylation in FLT3-ITD associated leukemias. Cancer Cell 29:922-934.), histone methyltransferases, and demethylases (Sze and Shilatifard, 2016Sze CC and Shilatifard A (2016) MLL3/MLL4/COMPASS family on epigenetic regulation of enhancer function and cancer. Cold Spring Harb Perspect Med 6:a026427.; Andricovich et al., 2018Andricovich J, Perkail S, Kai Y, Casasanta N, Peng W and Tzatsos A (2018) Loss of KDM6A Activates Super-Enhancers to Induce Gender-Specific Squamous-like Pancreatic Cancer and Confers Sensitivity to BET Inhibitors. Cancer Cell 33:512-526.e518.; Tran et al., 2020Tran N, Broun A and Ge K (2020) Lysine Demethylase KDM6A in differentiation, development, and cancer. Mol Cell Biol 40:e00341-20.) are also implicated in oncogenic enhancer function through modulating local active/repressive DNA and histone modifications.

Pioneer TFs drive chromatin remodeling and opening in enhancer regions and facilitate gene activation. It is observed that metastatic transition in pancreatic ductal adenocarcinoma is accompanied by large-scale enhancer reprogramming. The pioneer TF FOXA1 is a driver of enhancer activation in this process, which leads to a retrograde developmental transition to embryonic foregut endoderm and a more metastatic nature in vivo (Roe et al., 2017Roe JS, Hwang CI, Somerville TDD, Milazzo JP, Lee EJ, Da Silva B, Maiorino L, Tiriac H, Young CM, Miyabayashi K et al. (2017) Enhancer repogramming promotes anceratic cancer metastasis. Cell 170:875-888.e820.).

It is important to note that components of the enhancer-promoter loop, whose formation is an essential step in transcription initiation, are also essential trans-acting factors in oncogenic enhancer reprogramming. These structural components include CTCF (Fiorito et al., 2016Fiorito E, Sharma Y, Gilfillan S, Wang S, Singh SK, Satheesh SV, Katika MR, Urbanucci A, Thiede B, Mills IG et al. (2016) CTCF modulates Estrogen Receptor function through specific chromatin and nuclear matrix interactions. Nucleic Acids Res 44:10588-10602.), cohesin (Rao et al., 2017Rao SSP, Huang SC, Glenn St Hilaire B, Engreitz JM, Perez EM, Kieffer-Kwon KR, Sanborn AL, Johnstone SE, Bascom GD, Bochkov ID et al. (2017) Cohesin loss elminates all loop domains. Cell 171:305-320.e324.), BRD4 (Lovén et al., 2013Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.), and Mediator (Lovén et al., 2013Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.). BRD4 turns out to be a promising therapeutic target for cancer treatment, which will be discussed in more detail later in this review.

SE & cancer

Disease-associated SNPs are most frequently found in noncoding regions of the genome and the majority of those noncoding SNPs are located inside enhancers (Hnisz et al., 2013Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-André V, Sigova AA, Hoke HA and Young RA (2013) Super-enhancers in the control of cell identity and disease. Cell 155:934-947.). SEs have been implicated in various types of cancer such as adenoid cystic carcinoma (Drier et al., 2016Drier Y, Cotton MJ, Williamson KE, Gillespie SM, Ryan RJ, Kluk MJ, Carey CD, Rodig SJ, Sholl LM, Afrogheh AH et al. (2016) An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nat Genet 48:265-272.), basal-like breast cancer (Chen et al., 2019Chen CH, Yang N, Zhang Y, Ding J, Zhang W, Liu R, Liu W and Chen C (2019) Inhibition of super enhancer downregulates the expression of KLF5 in basal-like breast cancers. Int J Biol Sci 15:1733-1742.), colon cancer (Göndör, 2020Göndör A (2020) WNT-mediated gene gating: A novel principle connecting oncogenic super-enhancers with the nuclear pore to drive pathological expression of MYC. Mol Cell Oncol 7:1710992.), colorectal cancer (Li et al., 2021Li QL, Lin X, Yu YL, Chen L, Hu QX, Chen M, Cao N, Zhao C, Wang CY, Huang CW et al. (2021) Genome-wide profiling in colorectal cancer identifies PHF19 and TBC1D16 as oncogenic super enhancers. Nat Commun 12:6407.; Yu et al., 2021Yu D, Yang X, Lin J, Cao Z, Lu C, Yang Z, Zheng M, Pan R and Cai W (2021) Super-enhancer induced Il-20RA promotes proliferation/metatasis and immune evasion on colorectal cancer. Front Oncol 11:724655.), endometrial carcinoma (Zhang et al., 2016Zhang X, Choi PS, Francis JM, Imielinski M, Watanabe H, Cherniack AD and Meyerson M (2016) Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat Genet 48:176-182.), follicular lymphoma (Heckman et al., 2002Heckman CA, Mehew JW and Boxer LM (2002) NF-kappaB activates Bcl-2 expression in t(14;18) lymphoma cells. Oncogene 21:3898-3908.), leukemia (Gröschel et al., 2014Gröschel S, Sanders MA, Hoogenboezem R, de Wit E, Bouwman BAM, Erpelinck C, van der Velden VHJ, Havermans M, Avellino R, van Lom K et al. (2014) A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157:369-381.; Mansour et al., 2014Mansour MR, Abraham BJ, Anders L, Berezovskaya A, Gutierrez A, Durbin AD, Etchin J, Lawton L, Sallan SE, Silverman LB et al. (2014) Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346:1373-1377.), lung adenocarcinoma (Zhang et al., 2016Zhang X, Choi PS, Francis JM, Imielinski M, Watanabe H, Cherniack AD and Meyerson M (2016) Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat Genet 48:176-182.), multiple myeloma (Delmore et al., 2011Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J et al. (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146:904-917.; Alvarez-Benayas et al., 2021Alvarez-Benayas J, Trasanidis N, Katsarou A, Ponnusamy K, Chaidos A, May PC, Xiao X, Bua M, Atta M, Roberts IAG et al. (2021) Chromatin-based, in cis and in trans regulatory rewiring underpins distinct oncogenic transcriptomes in multiple myeloma. Nat Commun 12:5450.; Jia et al., 2022Jia Y, Zhou J, Tan TK, Chung TH, Chen Y, Chooi JY, Sanda T, Fullwood MJ, Xiong S, Toh SHM et al. (2022) Super enhancer-mediated upregulation of HJURP promotes growth and survival of t(4;14)-positive multiple myeloma. Cancer Res 82:406-418.), nasopharyngeal carcinoma (Ke et al., 2017Ke L, Zhou H, Wang C, Xiong G, Xiang Y, Ling Y, Khabir A, Tsao GS, Zeng Y, Zeng M et al. (2017) Nasopharyngeal carcinoma super-enhancer-driven ETV6 correlates with prognosis. Proc Natl Acad Sci U S A 114:9683-9688.; Cai et al., 2020Cai J, Chen S, Yi M, Tan Y, Peng Q, Ban Y, Yang J, Li X, Zeng Z, Xiong W et al. (2020) ΔNp63α is a super enhancer-enriched master factor controlling the basal-to-luminal differentiation transcriptional program and gene regulatory networks in nasopharyngeal carcinoma. Carcinogenesis 41:1282-1293.), neuroblastoma (Oldridge et al., 2015Oldridge DA, Wood AC, Weichert-Leahey N, Crimmins I, Sussman R, Winter C, McDaniel LD, Diamond M, Hart LS, Zhu S et al. (2015) Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature 528:418-421.), oesophageal squamous cell carcinoma (Jiang et al., 2017Jiang YY, Lin DC, Mayakonda A, Hazawa M, Ding LW, Chien WW, Xu L, Chen Y, Xiao JF, Senapedis W et al. (2017) Targeting super-enhancer-associated oncogenes in oesophageal squamous cell carcinoma. Gut 66:1358-1368.), pancreatic cancer (Kim et al., 2021Kim HR, Yim J, Yoo HB, Lee SE, Oh S, Jung S, Hwang CI, Shin DM, Kim T, Yoo KH et al. (2021) EVI1 activates tumor-promoting transcriptional enhancers in pancreatic cancer. NAR Cancer 3:zcab023.), pleomorphic adenoma (Afshari et al., 2020Afshari MK, Fehr A, Nevado PT, Andersson MK and Stenman G (2020) Activation of PLAG1 and HMGA2 by gene fusions involving the transcriptional regulator gene NFIB. Genes Chromosomes Cancer 59:652-660.), prostate cancer (Takeda et al., 2018Takeda DY, Spisák S, Seo JH, Bell C, O’Connor E, Korthauer K, Ribli D, Csabai I, Solymosi N, Szállási Z et al. (2018) A somatically acquired enhancer of androgen receptor is a noncoding driver of advamced prostate cancer. Cell 174:422-432.e413.; Xiao et al., 2022Xiao L, Parolia A, Qiao Y, Bawa P, Eyunni S, Mannan R, Carson SE, Chang Y, Wang X, Zhang Y et al. (2022) Targeting SWI/SNF ATPases in enhancer-addicted prostate cancer. Nature 601:434-439.), primary effusion lymphoma (Wang et al., 2020Wang C, Zhang L, Ke L, Ding W, Jiang S, Li D, Narita Y, Hou I, Liang J, Li S et al. (2020) Primary effusion lymphoma enhancer connectome links super-enhancers to dependency factors. Nat Commun 11:6318.), and rhabdomyosarcoma (Gryder et al., 2020Gryder BE, Wachtel M, Chang K, El Demerdash O, Aboreden NG, Mohammed W, Ewert W, Pomella S, Rota R, Wei JS et al. (2020) Miswired Enhancer Logic Drives a Cancer of the Muscle Lineage. iScience 23:101103.). Known SEs, their target genes, and relative SE formation mechanisms are summarized in Table 1. SEs are associated with key oncogene expressions in many cancer cells. SEs are found near oncogenes in cancer cells, whereas in their healthy counterparts, these SEs are absent (Tang et al., 2020Tang F, Yang Z, Tan Y and Li Y (2020) Super-enhancer function and its application in cancer targeted therapy. NPJ Precis Oncol 4:2.). Many events could lead to SE formation during tumor pathogenesis, including DNA amplification (Zhang et al., 2016Zhang X, Choi PS, Francis JM, Imielinski M, Watanabe H, Cherniack AD and Meyerson M (2016) Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat Genet 48:176-182.) and translocation (Drier et al., 2016Drier Y, Cotton MJ, Williamson KE, Gillespie SM, Ryan RJ, Kluk MJ, Carey CD, Rodig SJ, Sholl LM, Afrogheh AH et al. (2016) An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nat Genet 48:265-272.).

Table 1-
Known SEs and their target genes in various cancers.

Enhancer and therapy resistance

Therapy resistance is a major issue in anticancer treatment, and the underlying molecular mechanisms are not completely understood. It is recently discovered that enhancer is also an essential factor in cancer therapy resistance (Bao et al., 2019Bao J, Li M, Liang S, Yang Y, Wu J, Zou Q, Fang S, Chen S and Guo L (2019) Integrated high-throughput analysis identifies super enhancers associated with chemoresistance in SCLC. BMC Med Genomics 12:67.; Canella et al., 2022Canella A, Van Belle S, Brouns T, Nigita G, Carlon MS, Christ F and Debyser Z (2022) LEDGF/p75-mediated chemoresistance of mixed-lineage leukemia involves cell survival pathways and super enhancer activators. Cancer Gene Ther 29:133-140.). BRD4 downregulation is implicated in SE activity and might constitute a novel mechanism for chemoresistance in mixed-lineage leukemia (Canella et al., 2022Canella A, Van Belle S, Brouns T, Nigita G, Carlon MS, Christ F and Debyser Z (2022) LEDGF/p75-mediated chemoresistance of mixed-lineage leukemia involves cell survival pathways and super enhancer activators. Cancer Gene Ther 29:133-140.). Global enhancer reprogramming changes breast cancer transcriptional programs profoundly to promote cellular plasticity and therapy resistance (Bi et al., 2020Bi M, Zhang Z, Jiang YZ, Xue P, Wang H, Lai Z, Fu X, De Angelis C, Gong Y, Gao Z et al. (2020) Enhancer reprogramming driven by high-order assemblies of transcription factors promotes phenotypic plasticity and breast cancer endocrine resistance. Nat Cell Biol 22:701-715.). It was shown that oncogenic TFs GATA3 and AP1 regulate enhancers that are lost and gained respectively during treatment resistance acquisition. GATA3 is responsible for luminal lineage-specific gene expression, whereas AP1 regulates cancer invasion-related gene programs. The high-order enhancer machinery mediated by differential TF-TF and TF-enhancer interactions is a mechanism of enhancer reprogramming and therapy resistance (Bi et al., 2020Bi M, Zhang Z, Jiang YZ, Xue P, Wang H, Lai Z, Fu X, De Angelis C, Gong Y, Gao Z et al. (2020) Enhancer reprogramming driven by high-order assemblies of transcription factors promotes phenotypic plasticity and breast cancer endocrine resistance. Nat Cell Biol 22:701-715.).

Application of enhancer reprogramming in cancer treatment

Targeting mediators of super-enhancer function

Since it has been observed in cancer cells that enhancers are driving oncogenic transcriptional programs, enhancers have become a potential pharmacological target for interventions of cancer.

BETi

Recently there has been a lot of research effort to explore possibilities to treat cancer with the inhibition of bromodomain and extraterminal (BET) proteins (Whyte et al., 2013Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH, Rahl PB, Lee TI and Young RA (2013) Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153:307-319.). There are four human BET proteins: BRD2, BRD3, BRD4, and testes-specific BRDT, out of which the most studied is BRD4. BRD4 contains two bromodomains, which can bind acetylated lysine on histone tails and transcription factors (Yang, 2004Yang XJ (2004) Lysine acetylation and the bromodomain: A new partnership for signaling. Bioessays 26:1076-1087.), and a C-terminal motif which can interact with positive transcription elongation factor b (PTEF-b). By binding to acetylated histones, acetylated transcription factors, and PTEF-b, BRD4 serves as a scaffold for transcription machinery to come together. The interaction between BRD4 and PTEF-b permits transcription initiation and elongation (Itzen et al., 2014Itzen F, Greifenberg AK, Bösken CA and Geyer M (2014) Brd4 activates P-TEFb for RNA polymerase II CTD phosphorylation. Nucleic Acids Res 42:7577-7590.). BRD4 is widely distributed along the genome and drives the transcription of many cell-lineage-determining genes in somatic cells and oncogenes in cancer (Lovén et al., 2013Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.; Donati et al., 2018Donati B, Lorenzini E and Ciarrocchi A (2018) BRD4 and Cancer: Going beyond transcriptional regulation. Mol Cancer 17:164.). BRD4 is found at essentially all active promoters and a significant fraction of active enhancers in both normal and transformed cell types (Anand et al., 2013Anand P, Brown JD, Lin CY, Qi J, Zhang R, Artero PC, Alaiti MA, Bullard J, Alazem K, Margulies KB et al. (2013) BET bromodomains mediate transcriptional pause release in heart failure. Cell 154:569-582.).

BET inhibitors (BETi) disrupt BET protein binding to acetylated lysine residues of chromatin and suppress the transcription of various genes, including oncogenes and oncogenic transcription factors. BETi is emerging as one of the most promising drugs to treat various types of cancer. There are several classes of BETi depending on whether they bind the BD of BET proteins noncovalently, bivalently, or if they also target BET proteins for degradation (Kulikowski et al., 2021Kulikowski E, Rakai BD and Wong NCW (2021) Inhibitors of bromodomain and extra‐terminal proteins for treating multiple human diseases. Med Res Rev 41:223-245.). Noncovalent BETi has the largest number of currently available BETi, they can bind bromodomains of BET proteins noncovalently and compete with acetylated peptides, thus displacing BET proteins from acetylated chromatin (Filippakopoulos et al., 2010Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I et al. (2010) Selective inhibition of BET bromodomains. Nature 468:1067-1073.; Nicodeme et al., 2010Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H et al. (2010) Suppression of inflammation by a synthetic histone mimic. Nature 468:1119-1123.). JQ1, IBET-762, IBET-151, OTX015, and ZEN-3694 all belong to this type, and they have shown antitumor activity in both cancer cell lines and murine cancer models (Dawson et al., 2011Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf C, Savitski MM et al. (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478:529-533.; Delmore et al., 2011Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J et al. (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146:904-917.; Boi et al., 2015Boi M, Gaudio E, Bonetti P, Kwee I, Bernasconi E, Tarantelli C, Rinaldi A, Testoni M, Cascione L, Ponzoni M et al. (2015) The BET Bromodomain Inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs. Clin Cancer Res 21:1628-1638.; Baldan et al., 2019Baldan F, Allegri L, Lazarevic M, Catia M, Milosevic M, Damante G and Milasin J (2019) Biological and molecular effects of bromodomain and extra-terminal (BET) inhibitors JQ1, IBET-151, and IBET-762 in OSCC cells. J Oral Pathol Med 48:214-221.).

Although it is still not clear how BET confers cancer-specific susceptibility, BETi is effective in reducing the transcription of several oncogenes (Delmore et al., 2011Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J et al. (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146:904-917.; Lovén et al., 2013Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.; Fowler et al., 2014Fowler T, Ghatak P, Price DH, Conaway R, Conaway J, Chiang CM, Bradner JE, Shilatifard A and Roy AL (2014) Regulation of MYC expression and differential JQ1 sensitivity in cancer cells. PLoS One 9:e87003.) and is potentially effective in treating various types of cancers including pancreatic ductal adenocarcinoma, leukemia, ovarian cancer (Yokoyama et al., 2016Yokoyama Y, Zhu H, Lee JH, Kossenkov AV, Wu SY, Wickramasinghe JM, Yin X, Palozola KC, Gardini A, Showe LC et al. (2016) BET inhibitors suppress ALDH activity by targeting ALDH1A1 super enhancer in ovarian cancer. Cancer Res 76:6320-6330.) and mature B-cell lymphoma (Dawson et al., 2011Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf C, Savitski MM et al. (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478:529-533.; Sahai et al., 2014Sahai V, Kumar K, Knab LM, Chow CR, Raza SS, Bentrem DJ, Ebine K and Munshi HG (2014) BET bromodomain inhibitors block growth of pancreatic cancer cells in three-dimensional collagen. Mol Cancer Ther 13:1907-1917.; Boi et al., 2015Boi M, Gaudio E, Bonetti P, Kwee I, Bernasconi E, Tarantelli C, Rinaldi A, Testoni M, Cascione L, Ponzoni M et al. (2015) The BET Bromodomain Inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs. Clin Cancer Res 21:1628-1638.; Mazur et al., 2015Mazur PK, Herner A, Mello SS, Wirth M, Hausmann S, Sánchez-Rivera FJ, Lofgren SM, Kuschma T, Hahn SA, Vangala D et al. (2015) Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat Med 21:1163-1171.; Garcia et al., 2016Garcia PL, Miller AL, Kreitzburg KM, Council LN, Gamblin TL, Christein JD, Heslin MJ, Arnoletti JP, Richardson JH, Chen D et al. (2016) The BET bromodomain inhibitor JQ1 suppresses growth of pancreatic ductal adenocarcinoma in patient-derived xenograft models. Oncogene 35:833-845.). It is worth mentioning that besides cancer, BETi has also shown promising therapeutic benefits in cardiovascular (atherosclerosis (Tsujikawa et al., 2019Tsujikawa LM, Fu L, Das S, Halliday C, Rakai BD, Stotz SC, Sarsons CD, Gilham D, Daze E, Wasiak S et al. (2019) Apabetalone (RVX-208) reduces vascular inflammation in vitro and in CVD patients by a BET-dependent epigenetic mechanism. Clin Epigenetics 11:102.) and heart failure (Anand et al., 2013Anand P, Brown JD, Lin CY, Qi J, Zhang R, Artero PC, Alaiti MA, Bullard J, Alazem K, Margulies KB et al. (2013) BET bromodomains mediate transcriptional pause release in heart failure. Cell 154:569-582.; Duan et al., 2017Duan Q, McMahon S, Anand P, Shah H, Thomas S, Salunga HT, Huang Y, Zhang R, Sahadevan A, Lemieux ME et al. (2017) BET bromodomain inhibition suppresses innate inflammatory and profibrotic transcriptional networks in heart failure. Sci Transl Med 9:eaah5084.)), autoimmune (juvenile idiopathic arthritis (Klein, 2018Klein K (2018) Bromodomain protein inhibition: A novel therapeutic strategy in rheumatic diseases. RMD Open 4:e000744.)) and metabolic diseases (obesity (Goupille et al., 2016Goupille O, Penglong T, Kadri Z, Granger-Locatelli M, Fucharoen S, Maouche-Chrétien L, Prost S, Leboulch P and Chrétien S (2016) Inhibition of the acetyl lysine-binding pocket of bromodomain and extraterminal domain proteins interferes with adipogenesis. Biochem Biophys Res Commun 472:624-630.; Duan et al., 2020Duan Q, Wu P, Liu Z, Xia F, Zhu L, Zheng Z, Yang T and Qi J (2020) BET bromodomain inhibition suppresses adipogenesis in mice. Endocrine 67:264-267.)).

The molecular mechanisms of how BETis exert their anti-cancer function are not completely understood. Theoretically, inhibition of BRD4 would not only interfere with its oncogene targets but also other housekeeping genes essential for maintaining cell identity. It was hypothesized that BETi impacts the transcription of SE-associated genes more effectively than that of typical enhancers bound by BRD4 (Lovén et al., 2013Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.). This “off-target” effect might be exacerbated with higher doses, which highlights the importance of discovering effective biomarkers to help visualize drug maximum activity and supervising dose control.

Treatment-associated toxicity, drug resistance, and lack of predictive biomarkers have limited BETi’s progression in clinical trials (Sarnik et al., 2021Sarnik J, Popławski T and Tokarz P (2021) BET proteins as attractive targets for cancer therapeutics. Int J Mol Sci 22:11102.). Future studies defining the mechanism of BETi activity, finding predictive biomarkers to predict sensitivity to BETi, and identifying potent combinational drugs would help prevent toxicities and facilitate its clinical success as anti-cancer agents (Shorstova et al., 2021Shorstova T, Foulkes WD and Witcher M (2021) Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 124:1478-1490.).

CDK7 inhibitors

Cyclin-dependent kinase 7 (CDK7) drives cell cycle progression through the phosphorylation of cell cycle CDKs. CDK7 also phosphorylates RNA polymerase II which permits transcription at active genes. CDK7 is upregulated in various types of cancers including estrogen receptor-positive breast cancer (Patel et al., 2016Patel H, Abduljabbar R, Lai CF, Periyasamy M, Harrod A, Gemma C, Steel JH, Patel N, Busonero C, Jerjees D et al. (2016) Expression of CDK7, Cyclin H, and MAT1 is elevated in breast cancer and is prognostic in estrogen-receptor-positive breast cancers. Clin Cancer Res 22:5929-5938.), gastric cancer (Wang et al., 2016Wang Q, Li M, Zhang X, Huang H, Huang J, Ke J, Ding H, Xiao J, Shan X, Liu Q et al. (2016) Upregulation of CDK7 in gastric cancer cell promotes tumor cell proliferation and predicts poor prognosis. Exp Mol Pathol 100:514-521.), triple-negative breast cancer(Li et al., 2017Li B, Ni Chonghaile T, Fan Y, Madden SF, Klinger R, O’Connor AE, Walsh L, O’Hurley G, Mallya Udupi G, Joseph J et al. (2017) Therapeutic rationale to target highly expressed CDK7 conferring poor outcomes in triple-negative breast cancer. Cancer Res 77:3834-3845.), ovarian cancer(Zhang et al., 2017Zhang Z, Peng H, Wang X, Yin X, Ma P, Jing Y, Cai MC, Liu J, Zhang M, Zhang S et al. (2017) Preclinical efficacy and molecular mechanism of targeting CDK7-dependent transcriptional addiction in ovarian cancer. Mol Cancer Ther 16:1739-1750.) and oral squamous cell carcinoma(Jiang et al., 2019Jiang L, Huang R, Wu Y, Diao P, Zhang W, Li J, Li Z, Wang Y, Cheng J and Yang J (2019) Overexpression of CDK7 is associated with unfavourable prognosis in oral squamous cell carcinoma. Pathology 51:74-80.). CDK7 inhibitors are emerging as promising cancer therapeutic targets. Their anti-tumor effect is mediated through both cell cycle arrest and inhibition of oncogenic transcriptional programs. Examples of CDK7 inhibitors include covalent inhibitors such as THZ1 (Kwiatkowski et al., 2014Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB, Dastur A, Amzallag A, Ramaswamy S, Tesar B et al. (2014) Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 511:616-620.), THZ2 (Zhang et al., 2020Zhang J, Liu W, Zou C, Zhao Z, Lai Y, Shi Z, Xie X, Huang G, Wang Y, Zhang X et al. (2020) Targeting super enhancer-associated oncogenes in osteosarcoma with THZ2, a covalent CDK7 inhibitor. Clin Cancer Res 26:2681-2692.), and SY-1365 (Hu et al., 2019Hu S, Marineau JJ, Rajagopal N, Hamman KB, Choi YJ, Schmidt DR, Ke N, Johannessen L, Bradley MJ, Orlando DA et al. (2019) Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res 79:3479-3491.) and noncovalent inhibitors such as BS-181 (Ali et al., 2009Ali S, Heathcote DA, Kroll SH, Jogalekar AS, Scheiper B, Patel H, Brackow J, Siwicka A, Fuchter MJ, Periyasamy M et al. (2009) The development of a selective cyclin-dependent kinase inhibitor that shows antitumor activity. Cancer Res 69:6208-6215.) and LDC4297 (Kelso et al., 2014Kelso TW, Baumgart K, Eickhoff J, Albert T, Antrecht C, Lemcke S, Klebl B and Meisterernst M (2014) Cyclin-dependent kinase 7 controls mRNA synthesis by affecting stability of preinitiation complexes, leading to altered gene expression, cell cycle progression, and survival of tumor cells. Mol Cell Biol 34:3675-3688.).

CDK7 inhibition is most effective in suppressing SE-linked oncogenic transcription compared with other genes without SE association (Chipumuro et al., 2014Chipumuro E, Marco E, Christensen CL, Kwiatkowski N, Zhang T, Hatheway CM, Abraham BJ, Sharma B, Yeung C, Altabef A et al. (2014) CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159:1126-1139.; Eliades et al., 2018Eliades P, Abraham BJ, Ji Z, Miller DM, Christensen CL, Kwiatkowski N, Kumar R, Njauw CN, Taylor M, Miao B et al. (2018) High MITF expression is associated with super-enhancers an suppressed CFK7 inhibition in melanoma. J Invest Dermatol 138:1582-1590.; Cao et al., 2019Cao X, Dang L, Zheng X, Lu Y, Lu Y, Ji R, Zhang T, Ruan X, Zhi J, Hou X et al. (2019) Targeting super-enhancer-driven oncogenic transcription by CDK7 inhibition in anaplastic thyroid carcinoma. Thyroid 29:809-823.). SE is loaded with PolII, co-activators, Mediator complex, and transcription factors. And it is shown that SE-associated genes are particularly sensitive to small perturbations in CDK7 function and PolII-mediated transcription (Kwiatkowski et al., 2014Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB, Dastur A, Amzallag A, Ramaswamy S, Tesar B et al. (2014) Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 511:616-620.). Treatments with covalent inhibitors inhibit downstream phophorylation of Pol II (Hu et al., 2019Hu S, Marineau JJ, Rajagopal N, Hamman KB, Choi YJ, Schmidt DR, Ke N, Johannessen L, Bradley MJ, Orlando DA et al. (2019) Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res 79:3479-3491.). CDK7 inhibitors also exert their anti-cancer function by reducing levels of SE-associated oncogenic TFs (Hu et al., 2019Hu S, Marineau JJ, Rajagopal N, Hamman KB, Choi YJ, Schmidt DR, Ke N, Johannessen L, Bradley MJ, Orlando DA et al. (2019) Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res 79:3479-3491.). CDK7 inhibition leads to reduced recruitment of oncogenic TFs and the repression of associated oncogene expression (Yuan et al., 2022Yuan J, Li X and Yu S (2022) CDK7-dependent transcriptional addiction in bone and soft tissue sarcomas: Present and future. Biochim Biophys Acta Rev Cancer 1877:188680.).

Due to its essential role in cell cycle progression, inhibition of CDK7 causes cell cycle arrest (Ali et al., 2009Ali S, Heathcote DA, Kroll SH, Jogalekar AS, Scheiper B, Patel H, Brackow J, Siwicka A, Fuchter MJ, Periyasamy M et al. (2009) The development of a selective cyclin-dependent kinase inhibitor that shows antitumor activity. Cancer Res 69:6208-6215.; Chipumuro et al., 2014Chipumuro E, Marco E, Christensen CL, Kwiatkowski N, Zhang T, Hatheway CM, Abraham BJ, Sharma B, Yeung C, Altabef A et al. (2014) CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159:1126-1139.; Kelso et al., 2014Kelso TW, Baumgart K, Eickhoff J, Albert T, Antrecht C, Lemcke S, Klebl B and Meisterernst M (2014) Cyclin-dependent kinase 7 controls mRNA synthesis by affecting stability of preinitiation complexes, leading to altered gene expression, cell cycle progression, and survival of tumor cells. Mol Cell Biol 34:3675-3688.; Choi et al., 2019Choi YJ, Kim DH, Yoon DH, Suh C, Choi CM, Lee JC, Hong JY and Rho JK (2019) Efficacy of the novel CDK7 inhibitor QS1189 in mantle cell lymphoma. Sci Rep 9:7193.; Olson et al., 2019Olson CM, Liang Y, Leggett A, Park WD, Li L, Mills CE, Elsarrag SZ, Ficarro SB, Zhang T, Düster R et al. (2019) Development of a selectiv CDK7 covalent inhibitor reveals predominant cell-cycle phenotype. Cell Chem Biol 26:792-803.e710.). The extent and timing of cell cycle arrest vary among different cancer types: LDC4297 causes G1 arrest in A549 lung cancer cells, but in HCT116 colon cancer cells only causes G2/M delay after extended incubation(Kelso et al., 2014Kelso TW, Baumgart K, Eickhoff J, Albert T, Antrecht C, Lemcke S, Klebl B and Meisterernst M (2014) Cyclin-dependent kinase 7 controls mRNA synthesis by affecting stability of preinitiation complexes, leading to altered gene expression, cell cycle progression, and survival of tumor cells. Mol Cell Biol 34:3675-3688.).

Genome editing to target enhancer

Another way to modify SE function in cancer is based on CRISPR/Cas9 gene editing system. The mutated form of transcription factor RUNX1 is associated with poorer outcomes in acute myeloid leukemia (AML). It is shown that CRISPR/Cas9 mediated knocking out of RUNX1 SE epicenter (a 24kb enhancer region inside the 170kb SE) results in repression of RUNX1 and higher apoptosis of AML cells (Mill et al., 2019Mill CP, Fiskus W, DiNardo CD, Qian Y, Raina K, Rajapakshe K, Perera D, Coarfa C, Kadia TM, Khoury JD et al. (2019) RUNX1-targeted therapy for AML expressing somatic or germline mutation in RUNX1. Blood 134:59-73.). In a subset of T-cell acute lymphoblastic leukemia (T-ALL) cases, there are indels in a conserved noncoding region that create an SE upstream of the TAL1 oncogene through introducing MYB transcription factor binding motifs. CRISPR/Cas9 experiments to cut out the mutated site resulted in the collapse of the SE and abrogation of TAL1 expression (Mansour et al., 2014Mansour MR, Abraham BJ, Anders L, Berezovskaya A, Gutierrez A, Durbin AD, Etchin J, Lawton L, Sallan SE, Silverman LB et al. (2014) Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346:1373-1377.).

A few clinical trials have been completed or are ongoing that leverage NHEJ-mediated genetic disruption of BCL11A enhancer. Another way to modify enhancers for therapeutic purposes without introducing double-strand breaks is to base edit. CRISPR/Cas9 system and a cytidine deaminase enzyme could be fused together to mediate cytidine to uridine conversion and subsequently C to T substitution at the target site (Komor et al., 2016Komor AC, Kim YB, Packer MS, Zuris JA and Liu DR (2016) Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420-424.; Gehrke et al., 2018Gehrke JM, Cervantes O, Clement MK, Wu Y, Zeng J, Bauer DE, Pinello L and Joung JK (2018) An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nature Biotechnol 36:977-982.). A single therapeutic base edit of the BCL11A enhancer in patient-derived human hematopoietic stem and progenitor cells (HSPCs) prevents sickling and globin chain imbalance in their erythroid progeny (Zeng et al., 2020Zeng J, Wu Y, Ren C, Bonanno J, Shen AH, Shea D, Gehrke JM, Clement K, Luk K, Yao Q et al. (2020) Therapeutic base editing of human hematopoietic stem cells. Nat Med 26:535-541.).

Future directions

The function of enhancers in tumorigenesis has been the target of intensive research efforts for some years. It is foreseeable that more types of cancer would be found to be related to enhancer reprogramming. Identification of major enhancers, including SEs associated with different types of cancer and subgroups, would pave the way for finding more potential targets for treatment. Targeting both cis and trans factors in enhancer function has been utilized in cancer therapy through genome editing and anti-cancer agents (BETi and CDK7i), although the molecular mechanisms are not completely understood. There are issues associated with these agents’ progression in clinical trials. Defining mechanisms of activity and finding suitable biomarkers would aid their successful translation in cancer therapy. It is shown that enhancers also play important roles in cancer therapy resistance recently, and research on the molecular mechanism of enhancer function would enable more strategies to resolve therapy resistance.

Acknowledgements

This work was supported by Specialized Research Fund of Department of Education in Shaanxi Province (No. 21JK0896).

References

  • Afshari MK, Fehr A, Nevado PT, Andersson MK and Stenman G (2020) Activation of PLAG1 and HMGA2 by gene fusions involving the transcriptional regulator gene NFIB. Genes Chromosomes Cancer 59:652-660.
  • Ahn JH, Davis ES, Daugird TA, Zhao S, Quiroga IY, Uryu H, Li J, Storey AJ, Tsai YH, Keeley DP et al (2021) Phase separation drives aberrant chromatin looping and cancer development. Nature 595:591-595.
  • Ali S, Heathcote DA, Kroll SH, Jogalekar AS, Scheiper B, Patel H, Brackow J, Siwicka A, Fuchter MJ, Periyasamy M et al (2009) The development of a selective cyclin-dependent kinase inhibitor that shows antitumor activity. Cancer Res 69:6208-6215.
  • Alvarez-Benayas J, Trasanidis N, Katsarou A, Ponnusamy K, Chaidos A, May PC, Xiao X, Bua M, Atta M, Roberts IAG et al (2021) Chromatin-based, in cis and in trans regulatory rewiring underpins distinct oncogenic transcriptomes in multiple myeloma. Nat Commun 12:5450.
  • Anand P, Brown JD, Lin CY, Qi J, Zhang R, Artero PC, Alaiti MA, Bullard J, Alazem K, Margulies KB et al (2013) BET bromodomains mediate transcriptional pause release in heart failure. Cell 154:569-582.
  • Andersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, Boyd M, Chen Y, Zhao X, Schmidl C, Suzuki T et al (2014a) An atlas of active enhancers across human cell types and tissues. Nature 507:455-461.
  • Andersson R, Refsing Andersen P, Valen E, Core LJ, Bornholdt J, Boyd M, Heick Jensen T and Sandelin A (2014b) Nuclear stability and transcriptional directionality separate functionally distinct RNA species. Nat Commun 5:5336.
  • Andersson R and Sandelin A (2020) Determinants of enhancer and promoter activities of regulatory elements. Nat Rev Genet 21:71-87.
  • Andricovich J, Perkail S, Kai Y, Casasanta N, Peng W and Tzatsos A (2018) Loss of KDM6A Activates Super-Enhancers to Induce Gender-Specific Squamous-like Pancreatic Cancer and Confers Sensitivity to BET Inhibitors. Cancer Cell 33:512-526.e518.
  • Baldan F, Allegri L, Lazarevic M, Catia M, Milosevic M, Damante G and Milasin J (2019) Biological and molecular effects of bromodomain and extra-terminal (BET) inhibitors JQ1, IBET-151, and IBET-762 in OSCC cells. J Oral Pathol Med 48:214-221.
  • Bao J, Li M, Liang S, Yang Y, Wu J, Zou Q, Fang S, Chen S and Guo L (2019) Integrated high-throughput analysis identifies super enhancers associated with chemoresistance in SCLC. BMC Med Genomics 12:67.
  • Bi M, Zhang Z, Jiang YZ, Xue P, Wang H, Lai Z, Fu X, De Angelis C, Gong Y, Gao Z et al (2020) Enhancer reprogramming driven by high-order assemblies of transcription factors promotes phenotypic plasticity and breast cancer endocrine resistance. Nat Cell Biol 22:701-715.
  • Boi M, Gaudio E, Bonetti P, Kwee I, Bernasconi E, Tarantelli C, Rinaldi A, Testoni M, Cascione L, Ponzoni M et al (2015) The BET Bromodomain Inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs. Clin Cancer Res 21:1628-1638.
  • Bose DA, Donahue G, Reinberg D, Shiekhattar R, Bonasio R and Berger SL (2017) RNA binding to CBP stimulates histone acetylation and transcription. Cell 168:135-149.e122.
  • Bradner JE, Hnisz D and Young RA (2017) Transcriptional addiction in cancer. Cell 168:629-643.
  • Brunelle M, Nordell Markovits A, Rodrigue S, Lupien M, Jacques P and Gévry N (2015) The histone variant H2A.Z is an important regulator of enhancer activity. Nucleic Acids Res 43:9742-9756.
  • Cai J, Chen S, Yi M, Tan Y, Peng Q, Ban Y, Yang J, Li X, Zeng Z, Xiong W et al (2020) ΔNp63α is a super enhancer-enriched master factor controlling the basal-to-luminal differentiation transcriptional program and gene regulatory networks in nasopharyngeal carcinoma. Carcinogenesis 41:1282-1293.
  • Canella A, Van Belle S, Brouns T, Nigita G, Carlon MS, Christ F and Debyser Z (2022) LEDGF/p75-mediated chemoresistance of mixed-lineage leukemia involves cell survival pathways and super enhancer activators. Cancer Gene Ther 29:133-140.
  • Cao Q, Anyansi C, Hu X, Xu L, Xiong L, Tang W, Mok MTS, Cheng C, Fan X, Gerstein M et al (2017) Reconstruction of enhancer-target networks in 935 samples of human primary cells, tissues and cell lines. Nat Genet 49:1428-1436.
  • Cao X, Dang L, Zheng X, Lu Y, Lu Y, Ji R, Zhang T, Ruan X, Zhi J, Hou X et al (2019) Targeting super-enhancer-driven oncogenic transcription by CDK7 inhibition in anaplastic thyroid carcinoma. Thyroid 29:809-823.
  • Chen CH, Yang N, Zhang Y, Ding J, Zhang W, Liu R, Liu W and Chen C (2019) Inhibition of super enhancer downregulates the expression of KLF5 in basal-like breast cancers. Int J Biol Sci 15:1733-1742.
  • Chipumuro E, Marco E, Christensen CL, Kwiatkowski N, Zhang T, Hatheway CM, Abraham BJ, Sharma B, Yeung C, Altabef A et al (2014) CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159:1126-1139.
  • Choi YJ, Kim DH, Yoon DH, Suh C, Choi CM, Lee JC, Hong JY and Rho JK (2019) Efficacy of the novel CDK7 inhibitor QS1189 in mantle cell lymphoma. Sci Rep 9:7193.
  • Collins T, Read MA, Neish AS, Whitley MZ, Thanos D and Maniatis T (1995) Transcriptional regulation of endothelial cell adhesion molecules: NF-kappa B and cytokine-inducible enhancers. FASEB J 9:899-909.
  • Core LJ, Martins AL, Danko CG, Waters CT, Siepel A and Lis JT (2014) Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers. Nat Genet 46:1311-1320.
  • Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf C, Savitski MM et al (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478:529-533.
  • Delabesse E, Ogilvy S, Chapman MA, Piltz SG, Gottgens B and Green AR (2005) Transcriptional regulation of the SCL locus: Identification of an enhancer that targets the primitive erythroid lineage in vivo. Mol Cell Biol 25:5215-5225.
  • Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J et al (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146:904-917.
  • Donati B, Lorenzini E and Ciarrocchi A (2018) BRD4 and Cancer: Going beyond transcriptional regulation. Mol Cancer 17:164.
  • Drier Y, Cotton MJ, Williamson KE, Gillespie SM, Ryan RJ, Kluk MJ, Carey CD, Rodig SJ, Sholl LM, Afrogheh AH et al (2016) An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nat Genet 48:265-272.
  • Duan Q, McMahon S, Anand P, Shah H, Thomas S, Salunga HT, Huang Y, Zhang R, Sahadevan A, Lemieux ME et al (2017) BET bromodomain inhibition suppresses innate inflammatory and profibrotic transcriptional networks in heart failure. Sci Transl Med 9:eaah5084.
  • Duan Q, Wu P, Liu Z, Xia F, Zhu L, Zheng Z, Yang T and Qi J (2020) BET bromodomain inhibition suppresses adipogenesis in mice. Endocrine 67:264-267.
  • Eliades P, Abraham BJ, Ji Z, Miller DM, Christensen CL, Kwiatkowski N, Kumar R, Njauw CN, Taylor M, Miao B et al (2018) High MITF expression is associated with super-enhancers an suppressed CFK7 inhibition in melanoma. J Invest Dermatol 138:1582-1590.
  • Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I et al (2010) Selective inhibition of BET bromodomains. Nature 468:1067-1073.
  • Fiorito E, Sharma Y, Gilfillan S, Wang S, Singh SK, Satheesh SV, Katika MR, Urbanucci A, Thiede B, Mills IG et al (2016) CTCF modulates Estrogen Receptor function through specific chromatin and nuclear matrix interactions. Nucleic Acids Res 44:10588-10602.
  • Fowler T, Ghatak P, Price DH, Conaway R, Conaway J, Chiang CM, Bradner JE, Shilatifard A and Roy AL (2014) Regulation of MYC expression and differential JQ1 sensitivity in cancer cells. PLoS One 9:e87003.
  • Fudenberg G, Imakaev M, Lu C, Goloborodko A, Abdennur N and Mirny LA (2016) Formation of chromosomal domains by loop Extrusion. Cell Rep 15:2038-2049.
  • Furlong EEM and Levine M (2018) Developmental enhancers and chromosome topology. Science 361:1341-1345.
  • Garcia PL, Miller AL, Kreitzburg KM, Council LN, Gamblin TL, Christein JD, Heslin MJ, Arnoletti JP, Richardson JH, Chen D et al (2016) The BET bromodomain inhibitor JQ1 suppresses growth of pancreatic ductal adenocarcinoma in patient-derived xenograft models. Oncogene 35:833-845.
  • Gehrke JM, Cervantes O, Clement MK, Wu Y, Zeng J, Bauer DE, Pinello L and Joung JK (2018) An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nature Biotechnol 36:977-982.
  • Gibbons MD, Fang Y, Spicola AP, Linzer N, Jones SM, Johnson BR, Li L, Xie M and Bungert J (2022) Enhancer-mediated formation of nuclear transcription initiation domains. Int J Mol Sci 23:9290.
  • Göndör A (2020) WNT-mediated gene gating: A novel principle connecting oncogenic super-enhancers with the nuclear pore to drive pathological expression of MYC. Mol Cell Oncol 7:1710992.
  • Göttgens B, Broccardo C, Sanchez MJ, Deveaux S, Murphy G, Göthert JR, Kotsopoulou E, Kinston S, Delaney L, Piltz S et al (2004) The scl +18/19 stem cell enhancer is not required for hematopoiesis: identification of a 5’ bifunctional hematopoietic-endothelial enhancer bound by Fli-1 and Elf-1. Mol Cell Biol 24:1870-1883.
  • Goupille O, Penglong T, Kadri Z, Granger-Locatelli M, Fucharoen S, Maouche-Chrétien L, Prost S, Leboulch P and Chrétien S (2016) Inhibition of the acetyl lysine-binding pocket of bromodomain and extraterminal domain proteins interferes with adipogenesis. Biochem Biophys Res Commun 472:624-630.
  • Gröschel S, Sanders MA, Hoogenboezem R, de Wit E, Bouwman BAM, Erpelinck C, van der Velden VHJ, Havermans M, Avellino R, van Lom K et al (2014) A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157:369-381.
  • Gryder BE, Wachtel M, Chang K, El Demerdash O, Aboreden NG, Mohammed W, Ewert W, Pomella S, Rota R, Wei JS et al (2020) Miswired Enhancer Logic Drives a Cancer of the Muscle Lineage. iScience 23:101103.
  • Heckman CA, Mehew JW and Boxer LM (2002) NF-kappaB activates Bcl-2 expression in t(14;18) lymphoma cells. Oncogene 21:3898-3908.
  • Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre C, Singh H and Glass CK (2010) Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38:576-589.
  • Heinz S, Romanoski CE, Benner C and Glass CK (2015) The selection and function of cell type-specific enhancers. Nat Rev Mol Cell Biol 16:144-154.
  • Henriques T, Scruggs BS, Inouye MO, Muse GW, Williams LH, Burkholder AB, Lavender CA, Fargo DC and Adelman K (2018) Widespread transcriptional pausing and elongation control at enhancers. Genes Dev 32:26-41.
  • Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS and Manolio TA (2009) Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A 106:9362-9367.
  • Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-André V, Sigova AA, Hoke HA and Young RA (2013) Super-enhancers in the control of cell identity and disease. Cell 155:934-947.
  • Hoffman JA, Trotter KW, Day CR, Ward JM, Inoue K, Rodriguez J and Archer TK (2022) Multimodal regulatory elements within a hormone-specific super enhancer control a heterogeneous transcriptional response. Mol Cell 82:803-815.e805.
  • Hu S, Marineau JJ, Rajagopal N, Hamman KB, Choi YJ, Schmidt DR, Ke N, Johannessen L, Bradley MJ, Orlando DA et al (2019) Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res 79:3479-3491.
  • Huang P, Zhang B, Zhao J and Li MD (2022) Integrating the epigenome and transcriptome of hepatocellular carcinoma to identify systematic enhancer abberations and establish an abberant enhancers-related prognostic signature. Front Cell Dev Biol 10:827657.
  • Itzen F, Greifenberg AK, Bösken CA and Geyer M (2014) Brd4 activates P-TEFb for RNA polymerase II CTD phosphorylation. Nucleic Acids Res 42:7577-7590.
  • Jia P, Li X, Wang X, Yao L, Xu Y, Hu Y, Xu W, He Z, Zhao Q, Deng Y et al (2021) ZMYND8 mediated liquid condensates spatiotemporally decommission the latent super-enhancers during macrophage polarization. Nat Commun 12:6535.
  • Jia Y, Zhou J, Tan TK, Chung TH, Chen Y, Chooi JY, Sanda T, Fullwood MJ, Xiong S, Toh SHM et al (2022) Super enhancer-mediated upregulation of HJURP promotes growth and survival of t(4;14)-positive multiple myeloma. Cancer Res 82:406-418.
  • Jiang L, Huang R, Wu Y, Diao P, Zhang W, Li J, Li Z, Wang Y, Cheng J and Yang J (2019) Overexpression of CDK7 is associated with unfavourable prognosis in oral squamous cell carcinoma. Pathology 51:74-80.
  • Jiang YY, Lin DC, Mayakonda A, Hazawa M, Ding LW, Chien WW, Xu L, Chen Y, Xiao JF, Senapedis W et al (2017) Targeting super-enhancer-associated oncogenes in oesophageal squamous cell carcinoma. Gut 66:1358-1368.
  • Kabra A and Bushweller J (2022) The Intrinsically Disordered Proteins MLLT3 (AF9) and MLLT1 (ENL) - Multimodal transcriptional switches with roles in normal hematopoisis. MLL fusion leukemia and kidney cancer. J Mol Biol 434:167117.
  • Kaikkonen MU, Spann NJ, Heinz S, Romanoski CE, Allison KA, Stender JD, Chun HB, Tough DF, Prinjha RK, Benner C et al (2013) Remodeling of the enhancer landscape during macrophage activation is coupled to enhancer transcription. Mol Cell 51:310-325.
  • Karin M, Haslinger A, Heguy A, Dietlin T and Cooke T (1987) Metal-responsive elements act as positive modulators of human metallothionein-IIA enhancer activity. Mol Cell Biol 7:606-613.
  • Ke L, Zhou H, Wang C, Xiong G, Xiang Y, Ling Y, Khabir A, Tsao GS, Zeng Y, Zeng M et al (2017) Nasopharyngeal carcinoma super-enhancer-driven ETV6 correlates with prognosis. Proc Natl Acad Sci U S A 114:9683-9688.
  • Kelso TW, Baumgart K, Eickhoff J, Albert T, Antrecht C, Lemcke S, Klebl B and Meisterernst M (2014) Cyclin-dependent kinase 7 controls mRNA synthesis by affecting stability of preinitiation complexes, leading to altered gene expression, cell cycle progression, and survival of tumor cells. Mol Cell Biol 34:3675-3688.
  • Khan A and Zhang X (2019) Integrative modeling reveals key chromatin and sequence signatures predicting super-enhancers. Sci Rep 9:2877.
  • Kim HR, Yim J, Yoo HB, Lee SE, Oh S, Jung S, Hwang CI, Shin DM, Kim T, Yoo KH et al (2021) EVI1 activates tumor-promoting transcriptional enhancers in pancreatic cancer. NAR Cancer 3:zcab023.
  • Kim TK, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara-Haley K, Kuersten S et al (2010) Widespread transcription at neuronal activity-regulated enhancers. Nature 465:182-187.
  • Klein K (2018) Bromodomain protein inhibition: A novel therapeutic strategy in rheumatic diseases. RMD Open 4:e000744.
  • Koch F, Fenouil R, Gut M, Cauchy P, Albert TK, Zacarias-Cabeza J, Spicuglia S, de la Chapelle AL, Heidemann M, Hintermair C et al (2011) Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat Struct Mol Biol 18:956-963.
  • Komor AC, Kim YB, Packer MS, Zuris JA and Liu DR (2016) Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420-424.
  • Kulikowski E, Rakai BD and Wong NCW (2021) Inhibitors of bromodomain and extra‐terminal proteins for treating multiple human diseases. Med Res Rev 41:223-245.
  • Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, Heravi-Moussavi A, Kheradpour P, Zhang Z, Wang J, Ziller MJ et al (2015) Integrative analysis of 111 reference human epigenomes. Nature 518:317-330.
  • Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB, Dastur A, Amzallag A, Ramaswamy S, Tesar B et al (2014) Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 511:616-620.
  • Lee R, Kang MK, Kim YJ, Yang B, Shim H, Kim S, Kim K, Yang CM, Min BG, Jung WJ et al (2022) CTCF-mediated chromatin looping provides a topological framework for the formation of phase-separated transcriptional condensates. Nucleic Acids Res 50:207-226.
  • Li B, Ni Chonghaile T, Fan Y, Madden SF, Klinger R, O’Connor AE, Walsh L, O’Hurley G, Mallya Udupi G, Joseph J et al (2017) Therapeutic rationale to target highly expressed CDK7 conferring poor outcomes in triple-negative breast cancer. Cancer Res 77:3834-3845.
  • Li QL, Lin X, Yu YL, Chen L, Hu QX, Chen M, Cao N, Zhao C, Wang CY, Huang CW et al (2021) Genome-wide profiling in colorectal cancer identifies PHF19 and TBC1D16 as oncogenic super enhancers. Nat Commun 12:6407.
  • Li W, Notani D, Ma Q, Tanasa B, Nunez E, Chen AY, Merkurjev D, Zhang J, Ohgi K, Song X et al (2013) Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498:516-520.
  • Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI and Young RA (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153:320-334.
  • Lu R, Wang P, Parton T, Zhou Y, Chrysovergis K, Rockowitz S, Chen WY, Abdel-Wahab O, Wade PA, Zheng D et al (2016) Epigenetic perturbations by Arg882-mutated DNMT3A potentiate aberrant stem cell gene-expression program and acute leukemia development. Cancer Cell 30:92-107.
  • Lubas M, Andersen PR, Schein A, Dziembowski A, Kudla G and Jensen TH (2015) The human nuclear exosome targeting complex is loaded onto newly synthesized RNA to direct early ribonucleolysis. Cell Rep 10:178-192.
  • Lupiáñez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, Horn D, Kayserili H, Opitz JM, Laxova R et al (2015) Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161:1012-1025.
  • Mansour MR, Abraham BJ, Anders L, Berezovskaya A, Gutierrez A, Durbin AD, Etchin J, Lawton L, Sallan SE, Silverman LB et al (2014) Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346:1373-1377.
  • Mathur R, Alver BH, San Roman AK, Wilson BG, Wang X, Agoston AT, Park PJ, Shivdasani RA and Roberts CW (2017) ARID1A loss impairs enhancer-mediated gene regulation and drives colon cancer in mice. Nat Genet 49:296-302.
  • Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, Reynolds AP, Sandstrom R, Qu H, Brody J et al (2012) Systematic localization of common disease-associated variation in regulatory DNA. Science 337:1190-1195.
  • Mazur PK, Herner A, Mello SS, Wirth M, Hausmann S, Sánchez-Rivera FJ, Lofgren SM, Kuschma T, Hahn SA, Vangala D et al (2015) Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat Med 21:1163-1171.
  • Melgar MF, Collins FS and Sethupathy P (2011) Discovery of active enhancers through bidirectional expression of short transcripts. Genome Biol 12:R113.
  • Mikhaylichenko O, Bondarenko V, Harnett D, Schor IE, Males M, Viales RR and Furlong EEM (2018) The degree of enhancer or promoter activity is reflected by the levels and directionality of eRNA transcription. Genes Dev 32:42-57.
  • Mill CP, Fiskus W, DiNardo CD, Qian Y, Raina K, Rajapakshe K, Perera D, Coarfa C, Kadia TM, Khoury JD et al (2019) RUNX1-targeted therapy for AML expressing somatic or germline mutation in RUNX1. Blood 134:59-73.
  • Mousavi K, Zare H, Dell’orso S, Grontved L, Gutierrez-Cruz G, Derfoul A, Hager GL and Sartorelli V (2013) eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol Cell 51:606-617.
  • Nair SJ, Yang L, Meluzzi D, Oh S, Yang F, Friedman MJ, Wang S, Suter T, Alshareedah I, Gamliel A et al (2019) Phase separation of ligand-activated enhancers licenses cooperative chromosomal enhancer assembly. Nat Struct Mol Biol 26:193-203.
  • Nasser J, Bergman DT, Fulco CP, Guckelberger P, Doughty BR, Patwardhan TA, Jones TR, Nguyen TH, Ulirsch JC, Lekschas F et al (2021) Genome-wide enhancer maps link risk variants to disease genes. Nature 593:238-243.
  • Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H et al (2010) Suppression of inflammation by a synthetic histone mimic. Nature 468:1119-1123.
  • Northcott PA, Lee C, Zichner T, Stütz AM, Erkek S, Kawauchi D, Shih DJ, Hovestadt V, Zapatka M, Sturm D et al (2014) Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511:428-434.
  • Oldridge DA, Wood AC, Weichert-Leahey N, Crimmins I, Sussman R, Winter C, McDaniel LD, Diamond M, Hart LS, Zhu S et al (2015) Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature 528:418-421.
  • Olson CM, Liang Y, Leggett A, Park WD, Li L, Mills CE, Elsarrag SZ, Ficarro SB, Zhang T, Düster R et al (2019) Development of a selectiv CDK7 covalent inhibitor reveals predominant cell-cycle phenotype. Cell Chem Biol 26:792-803.e710.
  • Owen I, Yee D, Wyne H, Perdikari TM, Johnson V, Smyth J, Kortum R, Fawzi NL and Shewmaker F (2021) The oncogenic transcription factor FUS-CHOP can undergo nuclear liquid-liquid phase separation. J Cell Sci 134:jcs258578.
  • Park YK, Lee JE, Yan Z, McKernan K, O’Haren T, Wang W, Peng W and Ge K (2021) Interplay of BAF and MLL4 promotes cell type-specific enhancer activation. Nat Commun 12:1630.
  • Patel H, Abduljabbar R, Lai CF, Periyasamy M, Harrod A, Gemma C, Steel JH, Patel N, Busonero C, Jerjees D et al (2016) Expression of CDK7, Cyclin H, and MAT1 is elevated in breast cancer and is prognostic in estrogen-receptor-positive breast cancers. Clin Cancer Res 22:5929-5938.
  • Rada-Iglesias A, Bajpai R, Swigut T, Brugmann SA, Flynn RA and Wysocka J (2011) A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470:279-283.
  • Rao SSP, Huang SC, Glenn St Hilaire B, Engreitz JM, Perez EM, Kieffer-Kwon KR, Sanborn AL, Johnstone SE, Bascom GD, Bochkov ID et al (2017) Cohesin loss elminates all loop domains. Cell 171:305-320.e324.
  • Robson MI, Ringel AR and Mundlos S (2019) Regulatory landscaping: How enhancer-promoter communication is sculpted in 3D. Mol Cell 74:1110-1122.
  • Roe JS, Hwang CI, Somerville TDD, Milazzo JP, Lee EJ, Da Silva B, Maiorino L, Tiriac H, Young CM, Miyabayashi K et al (2017) Enhancer repogramming promotes anceratic cancer metastasis. Cell 170:875-888.e820.
  • Sabari BR, Dall’Agnese A, Boija A, Klein IA, Coffey EL, Shrinivas K, Abraham BJ, Hannett NM, Zamudio AV, Manteiga JC et al (2018) Coactivator condensation at super-enhancers links phase separation and gene control. Science 361:eaar3958.
  • Sahai V, Kumar K, Knab LM, Chow CR, Raza SS, Bentrem DJ, Ebine K and Munshi HG (2014) BET bromodomain inhibitors block growth of pancreatic cancer cells in three-dimensional collagen. Mol Cancer Ther 13:1907-1917.
  • Sánchez M, Göttgens B, Sinclair AM, Stanley M, Begley CG, Hunter S and Green AR (1999) An SCL 3’ enhancer targets developing endothelium together with embryonic and adult haematopoietic progenitors. Development 126:3891-3904.
  • Sarnik J, Popławski T and Tokarz P (2021) BET proteins as attractive targets for cancer therapeutics. Int J Mol Sci 22:11102.
  • Shlyueva D, Stelzer C, Gerlach D, Yáñez-Cuna JO, Rath M, Boryń Ł M, Arnold CD and Stark A (2014) Hormone-responsive enhancer-activity maps reveal predictive motifs, indirect repression, and targeting of closed chromatin. Mol Cell 54:180-192.
  • Shorstova T, Foulkes WD and Witcher M (2021) Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 124:1478-1490.
  • Sigova AA, Abraham BJ, Ji X, Molinie B, Hannett NM, Guo YE, Jangi M, Giallourakis CC, Sharp PA and Young RA (2015) Transcription factor trapping by RNA in gene regulatory elements. Science 350:978-981.
  • St Pierre R and Kadoch C (2017) Mammalian SWI/SNF complexes in cancer: emerging therapeutic opportunities. Curr Opin Genet Dev 42:56-67.
  • Suzuki HI and Onimaru K (2022) Biomolecular condensates in cancer biology. Cancer Sci 113:382-391.
  • Symmons O, Uslu VV, Tsujimura T, Ruf S, Nassari S, Schwarzer W, Ettwiller L and Spitz F (2014) Functional and topological characteristics of mammalian regulatory domains. Genome Res 24:390-400.
  • Sze CC and Shilatifard A (2016) MLL3/MLL4/COMPASS family on epigenetic regulation of enhancer function and cancer. Cold Spring Harb Perspect Med 6:a026427.
  • Takeda DY, Spisák S, Seo JH, Bell C, O’Connor E, Korthauer K, Ribli D, Csabai I, Solymosi N, Szállási Z et al (2018) A somatically acquired enhancer of androgen receptor is a noncoding driver of advamced prostate cancer. Cell 174:422-432.e413.
  • Tan Y, Yao L, Gamliel A, Nair SJ, Taylor H, Ohgi K, Aggarwal AK and Rosenfeld MG (2023) Signal-induced enhancer activation requires Ku70 to read topoisomerase1-DNA covalent complexes. Nat Struct Mol Biol 30:148-158.
  • Tang F, Yang Z, Tan Y and Li Y (2020) Super-enhancer function and its application in cancer targeted therapy. NPJ Precis Oncol 4:2.
  • Teng S, Li YE, Yang M, Qi R, Huang Y, Wang Q, Zhang Y, Chen S, Li S, Lin K et al (2020) Tissue-specific transcription reprogramming promotes liver metastasis of colorectal cancer. Cell Res 30:34-49.
  • Thanos D (1996) Mechanisms of transcriptional synergism of eukaryotic genes. The interferon-beta paradigm. Hypertension 27:1025-1029.
  • Thanos D, Du W and Maniatis T (1993) The high mobility group protein HMG I(Y) is an essential structural component of a virus-inducible enhancer complex. Cold Spring Harb Symp Quant Biol 58:73-81.
  • Tran N, Broun A and Ge K (2020) Lysine Demethylase KDM6A in differentiation, development, and cancer. Mol Cell Biol 40:e00341-20.
  • Tsujikawa LM, Fu L, Das S, Halliday C, Rakai BD, Stotz SC, Sarsons CD, Gilham D, Daze E, Wasiak S et al (2019) Apabetalone (RVX-208) reduces vascular inflammation in vitro and in CVD patients by a BET-dependent epigenetic mechanism. Clin Epigenetics 11:102.
  • Tu YH, Juan HF and Huang HC (2021) Identification of cell states using super-enhancer RNA. BMC Genomics 22:787.
  • van Steensel B and Furlong EEM (2019) The role of transcription in shaping the spatial organization of the genome. Nat Rev Mol Cell Biol 20:327-337.
  • Wang C, Zhang L, Ke L, Ding W, Jiang S, Li D, Narita Y, Hou I, Liang J, Li S et al (2020) Primary effusion lymphoma enhancer connectome links super-enhancers to dependency factors. Nat Commun 11:6318.
  • Wang Q, Li M, Zhang X, Huang H, Huang J, Ke J, Ding H, Xiao J, Shan X, Liu Q et al (2016) Upregulation of CDK7 in gastric cancer cell promotes tumor cell proliferation and predicts poor prognosis. Exp Mol Pathol 100:514-521.
  • Weinhold N, Jacobsen A, Schultz N, Sander C and Lee W (2014) Genome-wide analysis of noncoding regulatory mutations in cancer. Nat Genet 46:1160-1165.
  • Westin G and Schaffner W (1988) A zinc-responsive factor interacts with a metal-regulated enhancer element (MRE) of the mouse metallothionein-I gene. EMBO J 7:3763-3770.
  • Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH, Rahl PB, Lee TI and Young RA (2013) Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153:307-319.
  • Xiao L, Parolia A, Qiao Y, Bawa P, Eyunni S, Mannan R, Carson SE, Chang Y, Wang X, Zhang Y et al (2022) Targeting SWI/SNF ATPases in enhancer-addicted prostate cancer. Nature 601:434-439.
  • Xiong L, Kang R, Ding R, Kang W, Zhang Y, Liu W, Huang Q, Meng J and Guo Z (2018) Genome-wide identification and characterization of enhancers across 10 human tissues. Int J Biol Sci 14:1321-1332.
  • Yang L, Rodriguez B, Mayle A, Park HJ, Lin X, Luo M, Jeong M, Curry CV, Kim SB, Ruau D et al (2016) DNMT3A loss drives enhancer hypomehtylation in FLT3-ITD associated leukemias. Cancer Cell 29:922-934.
  • Yang XJ (2004) Lysine acetylation and the bromodomain: A new partnership for signaling. Bioessays 26:1076-1087.
  • Ye B, Fan D, Xiong W, Li M, Yuan J, Jiang Q, Zhao Y, Lin J, Liu J, Lv Y et al (2021) Oncogenic enhancers drive esophageal squamous cell carcinogenesis and metastasis. Nat Commun 12:4457.
  • Yi M, Tan Y, Wang L, Cai J, Li X, Zeng Z, Xiong W, Li G, Li X, Tan P et al (2020) TP63 links chromatin remodeling and enhancer reprogramming to epidermal differentiation and squamous cell carcinoma development. Cell Mol Life Sci 77:4325-4346.
  • Yokoyama Y, Zhu H, Lee JH, Kossenkov AV, Wu SY, Wickramasinghe JM, Yin X, Palozola KC, Gardini A, Showe LC et al (2016) BET inhibitors suppress ALDH activity by targeting ALDH1A1 super enhancer in ovarian cancer. Cancer Res 76:6320-6330.
  • Yoshino S and Suzuki HI (2022) The molecular understanding of super-enhancer dysregulation in cancer. Nagoya J Med Sci 84:216-229.
  • Yu D, Yang X, Lin J, Cao Z, Lu C, Yang Z, Zheng M, Pan R and Cai W (2021) Super-enhancer induced Il-20RA promotes proliferation/metatasis and immune evasion on colorectal cancer. Front Oncol 11:724655.
  • Yuan J, Li X and Yu S (2022) CDK7-dependent transcriptional addiction in bone and soft tissue sarcomas: Present and future. Biochim Biophys Acta Rev Cancer 1877:188680.
  • Zeng J, Wu Y, Ren C, Bonanno J, Shen AH, Shea D, Gehrke JM, Clement K, Luk K, Yao Q et al (2020) Therapeutic base editing of human hematopoietic stem cells. Nat Med 26:535-541.
  • Zhang J, Liu W, Zou C, Zhao Z, Lai Y, Shi Z, Xie X, Huang G, Wang Y, Zhang X et al (2020) Targeting super enhancer-associated oncogenes in osteosarcoma with THZ2, a covalent CDK7 inhibitor. Clin Cancer Res 26:2681-2692.
  • Zhang J, Yue W, Zhou Y, Liao M, Chen X and Hua J (2021) Super enhancers-functional cores under the 3D genome. Cell Prolif 54:e12970.
  • Zhang X, Choi PS, Francis JM, Imielinski M, Watanabe H, Cherniack AD and Meyerson M (2016) Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat Genet 48:176-182.
  • Zhang Z, Peng H, Wang X, Yin X, Ma P, Jing Y, Cai MC, Liu J, Zhang M, Zhang S et al (2017) Preclinical efficacy and molecular mechanism of targeting CDK7-dependent transcriptional addiction in ovarian cancer. Mol Cancer Ther 16:1739-1750.
  • Zhou J, Wang D, Tang D and Huang W (2020) Abnormal activations of super-enhancers enhance the carcinogenicity in lung adenocarcinoma. Cancer Manag Res 12:8509-8518.

Associate Editor:

Carlos Menck

Publication Dates

  • Publication in this collection
    04 Aug 2023
  • Date of issue
    2023

History

  • Received
    31 Oct 2022
  • Accepted
    19 June 2023
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br