Despite extensive progress in developing anticancer therapies, therapy resistance remains a major challenge that promotes disease relapse. The changes that lead to therapy resistance can be intrinsically present or may be initiated during treatment. Genetic and epigenetic heterogeneity in tumors make it more challenging to deal with therapy resistance. Recent advances in genome-wide analyses have revealed that the deregulation of distal gene regulatory elements, such as enhancers, appears in several pathophysiological conditions, including cancer. Beyond the conventional function of enhancers in recruiting transcription factors to gene promoters, enhancer elements are also transcribed into noncoding RNAs known as enhancer RNAs (eRNA). Accumulating evidence suggests that uncontrolled enhancer activity with aberrant eRNA expression promotes oncogenesis. Interestingly, tissue-specific, transcribed eRNAs from active enhancers can serve as potential therapeutic targets or biomarkers in several cancer types. This review provides a comprehensive overview of the mechanisms of enhancer transcription and eRNAs as well as their potential roles in cancer and drug resistance.

Cancer is one of the leading causes of death worldwide; it is a heterogeneous disease controlled by genetic and epigenetic alterations and deregulated transcription (1, 2). Advances in DNA-based technologies have led to the development of precision-based therapeutic strategies, including surgery, targeted therapy, radiation therapy, chemotherapy, hormonal therapy, and immunotherapy (3–5). Yet, most cancers evolve and become refractory to therapy, resulting in death because the continuous selection of fit cells leads to resistance against therapeutic strategies (6–8).

In the past two decades, with the completion of the Human Genome Project and advances in high throughput sequencing, it has become quite feasible to identify regulatory and functional elements and determine their function, especially abnormal biological processes such as cancer. Sequencing has provided a blueprint for gene expression and has led to new study areas comprising the assessment of multiple action mechanisms involved in gene regulation, providing researchers with therapeutic and diagnostic targets for many heritable diseases. Transcription is a critical regulatory step controlled by promoters, which are nearby cis-regulatory elements, and other distal regulatory elements, such as enhancers and silencing elements (9). Understanding the intricate details of transcriptional regulation at enhancers in cancer is the key to developing next-generation therapeutic strategies to treat this ever-evolving disease.

Enhancers and enhancer transcription

Enhancers are short clusters of regulatory DNA elements, spanning 500–2,000 bp, that are transcribed and contain transcription factor (TF) recognition sequences. They are present either near to or distant from the target gene promoters and regulate expression across space and time (10–12). The first enhancer was described as a distal cis-regulatory DNA element of the gene activation of the simian virus 40 (SV40; ref. 13). Active enhancers have been found in undifferentiated and pluripotent embryonic stem cells, which drive gene expression to maintain pluripotency (14). However, those involved in lineage commitment remain inactive (15). During cellular differentiation, enhancers become active and control lineage specification (14, 15). Emerging studies have shown that most oncogenes are governed by actively transcribing enhancers and/or clusters of enhancers (sometimes termed super-enhancers), previously known as locus control regions (16–18). The binding of TFs to enhancers leads to the recruitment of chromatin-modifying machinery and chromatin remodeling complexes. Enhancers are typically marked by histone modifications, such as histone H3 lysine 27 acetylation (H3K27ac) and H3 lysine 4 monomethylation (H3K4me1; Fig. 1A), which result in enhancer transcription to generate enhancer RNAs (eRNA) via RNA polymerase II (RNA Pol II; Fig. 1A; refs. 19–25). The discovery of eRNAs has revealed an untapped wealth of biological information. We have just begun to understand their potential in designing strategies to combat many diseases, including cancer.

Figure 1.

The mechanism of eRNA action. A, Increased histone modifications H3K27ac and H3K4me1 promote eRNA transcription from the enhancer of target genes. B, eRNAs interact with TFs and target them to bind to their cognate site. C, Enhancer–promoter interaction. eRNA interacts with the RAD21 and SMC3 subunits of the cohesion complex to mediate the E–P interaction via the mediator complex, promoting chromatin looping. D, Interaction with the transcriptional machinery. eRNAs remove NELF and recruit P-TEFb to release the pause of RNA Pol II, driving processive transcription and nascent mRNA transcription.

Figure 1.

The mechanism of eRNA action. A, Increased histone modifications H3K27ac and H3K4me1 promote eRNA transcription from the enhancer of target genes. B, eRNAs interact with TFs and target them to bind to their cognate site. C, Enhancer–promoter interaction. eRNA interacts with the RAD21 and SMC3 subunits of the cohesion complex to mediate the E–P interaction via the mediator complex, promoting chromatin looping. D, Interaction with the transcriptional machinery. eRNAs remove NELF and recruit P-TEFb to release the pause of RNA Pol II, driving processive transcription and nascent mRNA transcription.

Close modal

eRNAs were first identified in neurons and macrophages (26, 27). eRNAs are bidirectionally transcribed from enhancers (24, 26) via RNA Pol II and are usually noncoding (10, 22, 26, 28–32). In contrast to mRNAs, the majority of these eRNAs are unspliced and have nonpolyadenylated tails (11). Furthermore, eRNAs are more unstable, more abundant, and shorter, and they are retained in the nucleus, making them vulnerable to exosome-mediated decay (11, 33–35). The lack of polyadenylation (polyA) is one of the primary causes of their reduced stability (11, 33, 34, 36). Mechanistically, the proximity of the polyA signal to the transcription start site (TSS) prevents the assembly of polyadenylation machinery at RNA Pol II's C-terminal domain (CTD), which results in the recruitment of exosomes (37). It is evident from recent studies that eRNAs have secondary structures similar to other ncRNAs and undergo posttranscriptional modifications, such as cytosine methylation via Nsun7, that stabilize the eRNA (38). eRNAs bind to TFs and are densely located at the target gene's TSS to induce enhancer–promoter (E–P) interactions (Fig. 1B and C; ref. 39). These characteristics make them poorly detectable through conventional polyA+ RNA-seq (10, 11, 22, 24, 26).

The identification and quantification of eRNAs in specific target cell lines and tissues are feasible via techniques such as ribosomal RNA (rRNA)-depleted total RNA-seq or cap analysis of gene expression (CAGE; refs. 11, 40, 41). Other studies have identified eRNAs using Global Run-On Sequencing (GRO-seq), Precision Nuclear Run-On Sequencing (PRO-seq), or Transient Transcriptome Sequencing (TT-seq; refs. 42–48). The CAGE by the FANTOM consortium revealed 43,011 active enhancers that transcribe eRNA (49). The genomic regions marked with H3K27ac and H3K4me1 reveal the active sites of eRNA transcription (50). BruUV-seq, which involves the transcription blocking of DNA lesions using UV light coupled with bromouridine labeling and nascent RNA-seq, is also an efficient method to detect eRNAs (51). Other techniques, such as FISH, use fluorescein-tagged RNA probes to detect eRNAs and their steady-state levels (52). These methods reveal an unprecedented view of the active transcription landscape, enabling the discovery of eRNAs.

This review will discuss the relevance of enhancer transcription and eRNAs in cancer, including therapy-resistant cancers. Lastly, we will propose plausible new therapeutic strategies to combat resistant cancers.

eRNAs are the primary products of active enhancers, which promote gene transcription and facilitate the interaction of TFs (Fig. 1B) and coactivators via an E–P loop (Fig. 1C) and prevent Pol II release from pausing by interacting with NELF (Fig. 1D; refs. 39, 42, 53–56). Recent studies have suggested that the deletion or disruption of DNA-encoding ncRNAs may not reveal their molecular functions (57, 58); hence, ncRNAs such as eRNAs should be targeted using classical approaches such as shRNA- or siRNA-mediated silencing. Possible precautions should be taken to ensure minimal or no interference in TF or RNA Pol II functions (34, 39, 42, 59, 60). Generally, nonpolyadenylated eRNAs act in cis; however, recent reports have indicated the existence of trans-acting, stable polyadenylated eRNAs (10, 42, 53, 59).

eRNAs as positive regulators of gene transcription

eRNAs alter the epigenetic landscape and chromatin architecture at the enhancer site (61); for example, the SERPINB2 eRNA upregulates its mRNA transcription (62). Conversely, the number of transcripts generated from enhancers located in the intragenic regions can inversely correlate with the host's gene expression levels (63). Nonetheless, recent reports have suggested that eRNAs have an enormous impact on transcription regulation, with eRNAs inducing neighboring mRNA transcription (11, 26, 40, 42, 61, 64). RNA Pol II is not ubiquitously bound to the enhancers; however, its recruitment and removal are crucial for the initiation and termination of eRNA, which regulate the length of transcripts (65). Knockdown studies have revealed that the downregulation of eRNAs leads to fewer target mRNAs (62, 66). The specificity of eRNAs to their target mRNA synthesis is contextually dependent on the chromatin landscape, TFs, and E–P interactions (67).

E–P interactions

Historically, active enhancers have been shown to regulate distal gene promoters via chromatin looping, and recent GRO-seq-based studies have indicated that eRNAs are actively transcribed from these enhancers, yet their biological function remains elusive (47, 68). In the initial study, researchers performed loss of function and chromosomal conformation capture assays, which showed an eRNA–E–P interaction in the presence of estrogen in breast cancer cells (42). It was found that eRNAs regulate the interaction between active enhancers and proximal promoters through association with the cohesion complex via the RAD21 and SMC3 subunits (Fig. 1C and D). Later, it was affirmed that the eRNA–RAD21 interaction is required for the stability of the E–P loop, as RNase H1-mediated digestion prevents RAD21 recruitment to the chromatin (69). Furthermore, such chromatin looping and eRNA interactions were found to regulate gene expression at the β-globin gene locus (70). Similarly, using 4C-seq and ChIP-seq assays, it was found that Evf2 eRNA, transcribed from the ultra-conserved enhancer, modulates cohesion positioning in interneurons, thereby regulating enhancer interactions in long-range cis (>30 Mb) and trans (71). In addition, in muscle, DRR eRNA modulates cohesin recruitment in distal promoters, facilitating a long-range E–P interaction and regulating muscle differentiation (72).

eRNAs have also been shown to interact with heterogeneous nuclear ribonucleoprotein U (hnRNPU) and the mediator complex to regulate the E–P interaction (66, 73). Heparanase (HPSE) eRNA regulates the transcription of the HPSE oncogene through an interaction with hnRNPU and p300, which is mediated by EGR1. This interaction facilitates the increased recruitment of p300 to the super-enhancer, may promote chromatin looping, and activates the promoter (66). eRNAs have also been found to associate with the mediator coactivator complex at E–P looping sites. The loss of eRNA removes the mediator complex from the target promoters, and the looping is disrupted (53, 74). For instance, eRNA interacts with mediator complex subunit 1 (MED1), facilitating the E–P interaction via chromatin looping in prostate cancer cells (53). In T-cell acute lymphoblastic leukemia cells, ARIEL eRNA connects to mediator complex MED12 (75), and in colon cancer cells, CTCF (the CCCTC binding factor) eRNAs facilitate the E–P interaction via chromatin looping (76). Likewise, PRC2, which trimethylates H3K27, interacts with the mediator complex, assisting in poised enhancers' looping by binding to RNA (77, 78). However, the PRC2-dependent mechanism of the eRNA-driven transition of poised to active enhancers requires further investigation.

Furthermore, the integrator, which is a multi-subunit complex associated with the CTD of RNA Pol II, regulates eRNA processing, which, in turn, modulates the eRNA-dependent E–P interaction (35, 79). Contrastingly, several studies have shown that the E–P interaction is maintained even after the inhibition of eRNA production or enhancer transcription (54, 80). Collectively, the exact mechanism of the eRNA-mediated E–P interaction remains to be explored and may be distinct for different eRNAs.

Interactions with DNA, R-loop formation, and the RNA exosome

It is evident from recent studies that eRNAs hybridize with templated DNA, leaving nontemplated DNA single-stranded, which is known as the R-loop (81). These R-loops are associated with DNA damage and genomic instability and are implicated in cancer (82, 83).

Studies have suggested that R-loops are associated with an RNA processing complex known as the RNA exosome (34). RNA exosomes have nine core subunits that bind to RNA, while the two non-core subunits are endowed with ribonuclease activity (84). The catalytic subunit DIS3 regulates CTCF binding element eRNAs (cbeRNA; cbeRNAs and DIS3-sensitive RNAs have a substantial overlap), which has been shown to play a crucial role in the maintenance of immunoglobulin heavy chain integrity and somatic hypermutation in B cells (85). The genetic depletion of exosome component 3 (Exosc3) and exosome component 10 (Exosc10) of the RNA exosome complex leads to the upregulation of eRNAs that results in R-loop formation and genomic instability at the enhancers of mouse embryonic stem cells and B cells (34). Significantly, tumor aneuploidy correlates with global enhancer activation and eRNA transcription, indicating genomic instability, DNA damage, and cancer progression (86). Further studies are required to elucidate the relationship between eRNA transcription and R-loop formation at the enhancers of cancer cells.

eRNAs have emerged as excellent markers of active enhancers and genes (Table 1). Aberrant changes in eRNA levels are associated with deregulated enhancer transcription and gene expression in cancer (32, 41, 86, 87). For example, in breast cancer, the estrogen-dependent binding of estrogen receptor alpha (ERα) to oncogenic enhancers leads to the production of eRNAs (80, 88). Similarly, in prostate cancer, androgen receptor (AR)-induced KLK3 eRNA, a regulator of KLK3 oncogene expression eRNA, acts as a scaffold for the AR-protein complex (53). Conversely, studies have indicated that tumor suppressors, such as TP53, induce eRNAs, which play a critical role in cell-cycle regulation in several cancer cells (60). In acute myeloid leukemia (AML), chromosomal rearrangements such as inv (3)/t(3;3) leads to aberrant transcriptional activity at GATA2 gene distal enhancer, which elevates EVI1 expression. Mechanistically, inv (3)/t(3;3) loops with the EVI1 promoter to control EVI1 expression (89). In addition, in pancreatic cancer, the enhancer landscape's reprogramming, which leads to genome-wide alterations in enhancer activity, promotes oncogenesis with the worst prognosis (90–92). These observations have revealed the role of enhancer transcription in regulating oncogenesis (93), and may generate eRNAs, which can be potential therapeutic targets, although a direct connection between pancreatic cancer and eRNA expression is yet to be established.

Table 1.

A list of the eRNAs implicated in oncogenesis.

Cancer typeeRNAsTranscriptional directionReferences
Breast cancer NRIPe, TFF1e, PGRe, KCNK5e, FOXC1e, GREB1e, CA12e, SMAD7e, P2RY2e, SIAH2e, NET1e, P21e, PRKAG2e, SUFUe, TOB1e Bidirectional 21, 42, 94 
Castration-resistant prostate cancer PSAe, ARHGEF26e, SLC16A7e, KLK15e, TLE1e, HTR3Ae, NCAM2e, FTOe, MARC1e, LUZP2e Bidirectional 98, 117 
Colorectal cancer CCAT1e Bidirectional 99 
Basal cell carcinoma ACTRT1e Bidirectional 95 
AML MYC (E1–E5) Not determined 113 
ALL ARIELe, LUNAR1e Unidirectional 75, 114, 122 
Cancer typeeRNAsTranscriptional directionReferences
Breast cancer NRIPe, TFF1e, PGRe, KCNK5e, FOXC1e, GREB1e, CA12e, SMAD7e, P2RY2e, SIAH2e, NET1e, P21e, PRKAG2e, SUFUe, TOB1e Bidirectional 21, 42, 94 
Castration-resistant prostate cancer PSAe, ARHGEF26e, SLC16A7e, KLK15e, TLE1e, HTR3Ae, NCAM2e, FTOe, MARC1e, LUZP2e Bidirectional 98, 117 
Colorectal cancer CCAT1e Bidirectional 99 
Basal cell carcinoma ACTRT1e Bidirectional 95 
AML MYC (E1–E5) Not determined 113 
ALL ARIELe, LUNAR1e Unidirectional 75, 114, 122 

eRNAs as tumor suppressors

eRNAs function as tumor suppressors by regulating tumor suppressor gene expression (32), such as tumor suppressor p53. The transcription of several eRNAs upon Nutlin-3a treatment in breast cancer cells is stimulated by p53 (60). Intriguingly, the loss of these eRNAs perturbs p53-dependent gene transcription (60). In another instance, LED lncRNA (the lncRNA activator of enhancer domains) regulates enhancer activity and eRNA transcription to control the expression of the p53-dependent genes CDKN1A, p21, PRKAG2, TOB1, and SUFU (94). Interestingly, LED expression is regulated by DNA hypermethylation; 5-azacytidine treatment promotes LED expression, which, in turn, facilitates p53's tumor-suppressive role. Therefore, eRNAs are crucial for maintaining tumor-suppressive functions.

eRNAs as positive regulators of oncogenes

eRNAs are master regulators of oncogenes and show potential oncogenic functions in various human cancers (32, 93). In ER+ breast cancers, 17β-estradiol (E2) treatment leads to ERα-bound enhancers' activation, inducing eRNA transcription and E2-dependent gene regulation (42). E2 treatment activates eRNA transcription and promotes the expression of genes, including GREB1, NRIP1, P2RY2, SMAD7, PGR, KCNK5, SIAH2, CA12, TFF1, and FOXC1 (42). Interestingly, the interaction between NRIP1 and TFF1 loci is lost upon the depletion of NRIP1e, indicating eRNA-mediated E–P looping. Mutation in the enhancer element of the ACTRT1 gene, which encodes actin-related protein T1 (ARP-T1), leads to aberrant eRNA transcription (95). Several reports have suggested that mutations or misregulation in enhancer regions lead to aberrant eRNA production (95) and deregulated signaling pathways (47, 61, 95–97). Therefore, the ectopic expression of ACTRT1 reduces cell proliferation and activates the Hedgehog signaling pathway aberrantly (95).

In prostate cancer, AR binding to target the PSA gene enhancer produces PSA eRNA (PSAe) and activates its expression in castration-resistant prostate cancer (CRPC) cells (98). PSAe regulates androgen-dependent gene expression both in cis and trans and promotes tumor progression (98). PSAe is structurally similar to the HIV-1 TAR-L motif and interacts with P-TEFb and cyclin T1 to regulate transcription. However, during trans-activation, PSAe modulates AR-dependent gene expression via a feed forward loop.

In other malignancies, such as CpG island methylator phenotype colon cancer, cMYC oncogene expression is driven by super-enhancer–derived colon cancer–associated transcript 1 (CCAT1; ref. 99). Furthermore, the interrogation of the cancer genome atlas has shown a positive correlation between CCAT1 and cMYC expression in several tumor types (99), indicating a functional relationship between them. Inhibition of BRD4 by JQ1 or its knockdown leads to reduced cMYC expression, which arrests tumor growth (99). CCAT1L, a longer form of CCAT1, mediates a long-distance interaction between the CCAT1-producing enhancer and the cMYC promoter (76).

In the past few years, genome-wide studies have shown aberrant enhancer activity in the deregulation of many signaling pathways in cancer (12, 100, 101). For example, in breast cancer cells, the hormone-driven YAP/TEAD of the Hippo pathway modulates eRNA transcription (102); other pathways, such as inflammatory signaling in p53-mutated cancers, drive eRNA transcription to regulate tumor progression (103). Specifically, inflammatory gene enhancers such as CCL2 are activated by mutant p53 and NFκB (103, 104). Collectively, these studies have suggested that eRNAs play a role in promoting tumor initiation and maintenance.

Resistant cancer is a well-defined phenomenon that occurs when cancer cells become tolerant of an administered drug dose. Resistance to therapy occurs due to various factors, including genetic and/or epigenetic alterations, the increased efflux of drugs, and several other molecular processes (Fig. 2A). These alterations collude to develop a patient's resistance to classical or novel chemotherapeutic drugs and immunotherapy-based approaches, leading to the disease's relapse (3, 5–8). Traditionally, many chemotherapeutic drugs induce DNA damage in cancer cells; however, off-target effects could potentially lead to cytotoxicity in normal cells. After repeated exposure, the drug's efficacy recedes, leading to a relapse of the disease in most patients. For instance, 40%–55% of triple-negative breast cancer patients develop resistance to chemotherapy and radiation therapy (105, 106). Similarly, 50%–70% of ovarian cancers relapse within a year post-surgery and -chemotherapy (107). Approximately 40% of non–small cell lung cancers relapse due to acquired resistance, resulting in death (108). Furthermore, 20% of lymphoblastic leukemia and pediatric cancers develop resistance and relapse (109). Hence, continuous efforts are being made to understand and identify the mechanisms of resistance in cancer.

Figure 2.

Mechanism of drug resistance, eRNA-dependent therapeutic strategy for cancer treatment. A, The epigenetic mechanism of drug resistance. B, A schematic model of silencing eRNA production by CRISPR interference (CRISPRi), small-molecule–based targeted drugs, and inhibiting eRNA–protein interactions, thus suppressing the function of eRNA to deactivate its target oncogenes.

Figure 2.

Mechanism of drug resistance, eRNA-dependent therapeutic strategy for cancer treatment. A, The epigenetic mechanism of drug resistance. B, A schematic model of silencing eRNA production by CRISPR interference (CRISPRi), small-molecule–based targeted drugs, and inhibiting eRNA–protein interactions, thus suppressing the function of eRNA to deactivate its target oncogenes.

Close modal

In the past decade, the deregulation of enhancer transcription, affecting ncRNA expression (eRNAs, lncRNAs, etc.), has been implicated in several cancers and its therapies (Fig. 2A; refs. 93, 110–114); however, the role of eRNAs in drug resistance is not well known. Though the role of enhancers in therapy-resistant cancers is emerging, such as, in combination with cisplatin, BET inhibitors sensitize cisplatin-resistant ovarian cancer to cisplatin by inhibiting the ALDH1A1 super-enhancer (115). Enhancer overactivation is also implicated in hormone-refractory breast cancers; a recent report has suggested that TFs such as GATA3 and AP1 reprogram the enhancer landscape to promote enhancer overactivation, leading to phenotypic plasticity in endocrine-resistant breast cancer (116).

Given the role of enhancers in therapy-resistant cancers and eRNAs being shown as functional units of enhancer activity, the eRNAs involvement in resistant cancers can't be ruled out. Neuroepithelial cell transforming gene 1 eRNA (NET1e) is abundantly expressed in breast cancer, and, notably, its overexpression results in resistance to the drugs BEZ235 and Obatoclax in MCF7 cells (21). In enzalutamide CRPC cells, AR-eRNAs are differentially expressed (117); the study has shown that prolonged treatment with enzalutamide leads to resistance and enhances the expression of AR-eRNAs and oncogenic mRNAs such as FTO, LUZP2, MARC1, and NCAM2, thereby promoting more resistant phenotypes (117).

eRNA transcription from active enhancers is implicated in both drug-resistant solid tumors and therapy-resistant leukemias. In daunorubicin-resistant AML, in response to stress, enhancers are activated to induce ABCB1 expression and promote resistance (118). Daunorubicin treatment to fresh blast cells from human AML samples leads to the activation of ABCB1 expression via the binding of ATF4-like TFs to stress-responsive enhancers. It has been reported that U0126 and ISRIB inhibit ABCB1 expression, thereby sensitizing cancer cells to drug treatment; however, the mechanism of inhibition is still unknown (118). These inhibitors may also affect enhancer activity and eRNA transcription by suppressing ABCB1 expression. Thus, by uncovering eRNA mechanisms in therapy-resistant cancers, researchers can identify potential therapeutic targets in resistant cancers.

eRNAs as emerging targets for anticancer therapy

Emerging studies have shown aberrant enhancer activity and eRNA expression in tumor samples when compared to normal tissues (21, 86). Enhancer overactivation is a unique phenomenon in oncogenesis, and targeting eRNAs could be a potential new anticancer therapeutic strategy (21, 119–121). The tissue-specific expression of eRNAs across different cancer types may make them tissue-specific therapeutic targets. These eRNAs can be targeted by shRNAs, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) Cas9, CRISPR interference (CRISPRi)-dCas9, antisense oligos [ASO or locked nucleic acids (LNA)], or enhancer activity suppression using BET inhibitors, which can control target gene expression and tumor growth (Fig. 2B; refs. 21, 53, 66, 122–124). NET1e is overexpressed in breast cancer cell lines, such as MCF7, with low or no expression seen in normal breast cell lines, such as MCF10A, or other cancer cell lines, such as HeLa or HepG2 (21). Targeting NET1e with LNAs inhibits cell proliferation (21), and this eRNA could serve as a potential drug target; however, it needs to be tested in preclinical models. In another study, the HPSE eRNA has been shown to have an oncogenic function in vivo (66); a detailed mechanism of action will reveal its therapeutic potential.

Interestingly, regarding other potential targets, a strong correlation between enhancer 9 expression (chr9:5580709–5581016) and programmed death-ligand 1 (PD-L1) has been found in a plethora of cancers (86). PD-L1 reportedly plays a significant role in cancer immunotherapy; therefore, using the expression of eRNA as a biomarker can help in designing immunotherapy-based approaches since the deletion of this enhancer impairs PD-L1 expression (86). These studies have suggested that eRNAs significantly contribute to the development of novel therapeutic targets that are highly specific and unique due to their tight regulation and tissue specificity.

Tumor heterogeneity poses a considerable challenge in understanding tumor progression and creates obstacles in identifying the best therapeutic strategy; failure to determine a potential strategy could lead to drug resistance. Identifying the driver oncogene, which promotes drug resistance at any stage of tumorigenesis, is possible with high throughput genomics, proteomics, and metabolomics analyses. These technologies have helped the scientific community to advance the design of combinatorial or personalized therapy.

Therapy with a single drug often fails to affect heterogeneous polyclonal cells and increases the possibility of relapse. Therefore, combinatorial treatment shows promising results and minimizes the likelihood of a gain of resistance. However, predicting therapeutic outcomes can be difficult, as resistant tumors are genetically different across patients. As tumor cells often develop alternative mechanisms for survival, finding an alternative therapeutic strategy will not be the final step.

In this regard, studies that focus on understanding the mechanisms that drive enhancer function and the role of eRNAs in drug resistance have emerged as a potential avenue for investigating the identification of potential targets. However, there is still much to study before we can completely understand and exploit enhancer transcription's therapeutic potential in cancer (11, 21, 32, 40, 86, 93, 112). Given the instability and dynamic nature of enhancer transcripts, the primary challenge is to validate their functional outcomes in vivo to explore their clinical utility. The chromatin regions and proteome associated with eRNAs must also be investigated and can be identified via Chromatin Isolation by RNA Purification (ChIRP), RNA antisense purification (RAP), or identification of direct RNA interacting proteins (iDRiP; refs. 125–127), which will potentially help to inhibit critical interactions by targeting protein partners. However, structural analyses would help in designing small-molecule modulators that can target eRNA–protein interactions with specificity.

Another potential avenue is to identify the role that RNA modifications play, including 5-cytosine methylation (m5C), N6-adenosine methylation (m6A), hydroxy-methyl cytosine (5hmC), and methyl-1 adenosine (m1A), which have been reported in several regulatory RNAs (128–131). The existence of such modifications in the context of eRNAs has not yet been explored. In addition, the role of eRNAs in higher-order chromatin organization, as mediated by RNA–DNA, RNA–RNA, or RNA–protein interactions, should be investigated in future studies (132, 133). Addressing these outstanding questions will not only broaden our knowledge of the enhanceosome but will significantly help in uncovering new eRNA-based cancer therapies, specifically for therapy-resistant cancers.

No disclosures were reported.

The authors thank Prof. Jessica K. Tyler for her critical comments on the manuscript. They also thank Ms. Melina J. Sedano and Ms. Alana L. Harrison for their helpful comments. The authors deeply regret being unable to cite many of the references due to space limitations. Figure illustrations were created with BioRender. Figures are adapted from “Regulation of Transcription in Eukaryotic Cells”, by BioRender.com (2021); Retrieved from https://app.biorender.com/biorender-templates. This work was supported in part by research grants received from Basic and Applied Research in Biophysics & Material Science (RSI 4002) from the Department of Atomic Energy; Science & Engineering Research Board (SERB) (CRG/2018/000985), Department of Science & Technology; SwarnaJayanti Fellowship (DST/SJF/LSA-02/2017–18), Department of Science and Technology; Department of Biotechnology (BT/PR28920/MED/122/176/2018); S. Ramachandran National Bioscience Award for Career Development 2019 (BT/HRD-NBA-NWB/38/2019–20), Department of Biotechnology, Govt. of India to CD; S.S. Gadad is a CPRIT Scholar in Cancer Research; first-time faculty recruitment award from Cancer Prevention and Research Institute of Texas (CPRIT; RR170020 to S.S. Gadad); Science & Engineering Research Board (SERB; EMR/2016/006233), Department of Science & Technology (DST), Govt. of India (to S. Roy). S. Adhikary thanks the Council of Scientific and Industrial Research (CSIR), Govt. of India, for fellowship support.

1.
Suzuki
A
,
Makinoshima
H
,
Wakaguri
H
,
Esumi
H
,
Sugano
S
,
Kohno
T
, et al
Aberrant transcriptional regulations in cancers: genome, transcriptome and epigenome analysis of lung adenocarcinoma cell lines
.
Nucleic Acids Res
2014
;
42
:
13557
72
.
2.
Bradner
JE
,
Hnisz
D
,
Young
RA
. 
Transcriptional addiction in cancer
.
Cell
2017
;
168
:
629
43
.
3.
Baskar
R
,
Lee
KA
,
Yeo
R
,
Yeoh
KW
. 
Cancer and radiation therapy: current advances and future directions
.
Int J Med Sci
2012
;
9
:
193
99
.
4.
Huang
B
,
Warner
M
,
Gustafsson
. 
Estrogen receptors in breast carcinogenesis and endocrine therapy
.
Mol Cell Endocrinol
2015
;
3
:
240
44
.
5.
Khalil
DN
,
Smith
EL
,
Brentjens
RJ
,
Wolchok
JD
. 
The future of cancer treatment: immunomodulation, CARs and combination immunotherapy
.
Nat Rev Clin Oncol
2016
;
13
:
273
90
.
6.
Holohan
C
,
Van Schaeybroeck
S
,
Longley
DB
,
Johnston
PG
. 
Cancer drug resistance: an evolving paradigm
.
Nat Rev Cancer
2013
;
13
:
714
26
.
7.
Borst
P
. 
Cancer drug pan-resistance: pumps, cancer stem cells, quiescence, epithelial to mesenchymal transition, blocked cell death pathways, persisters or what?
Open Biol
2012
;
2
:
120066
.
8.
Alfarouk
KO
,
Stock
CM
,
Taylor
S
,
Walsh
M
,
Muddathir
AK
,
Verduzco
D
, et al
Resistance to cancer chemotherapy: failure in drug response from ADME to P-gp
.
Cancer Cell Int
2015
;
15
:
71
.
9.
Heintzman
ND
,
Ren
B
. 
Finding distal regulatory elements in the human genome
.
Curr Opin Genet Dev
2009
;
19
:
541
49
.
10.
Natoli
G
,
Andrau
J-C
. 
Noncoding transcription at enhancers: general principles and functional models
.
Annu Rev Genet
2012
;
46
:
1
19
.
11.
Andersson
R
,
Gebhard
C
,
Miguel-Escalada
I
,
Hoof
I
,
Bornholdt
J
,
Boyd
M
, et al
An atlas of active enhancers across human cell types and tissues
.
Nature
2014
;
507
:
455
61
.
12.
Li
W
,
Notani
D
,
Rosenfeld
MG
. 
Enhancers as non-coding RNA transcription units: recent insights and future perspectives
.
Nat Rev Genet
2016
;
17
:
207
23
.
13.
Banerji
J
,
Rusconi
S
,
Schaffner
W
. 
Expression of a β-globin gene is enhanced by remote SV40 DNA sequences
.
Cell
1981
;
27
:
299
308
.
14.
Whyte
WA
,
Orlando
DA
,
Hnisz
D
,
Abraham
BJ
,
Lin
CY
,
Kagey
MH
, et al
Master transcription factors and mediator establish super-enhancers at key cell identity genes
.
Cell
2013
;
153
:
307
19
.
15.
Rada-Iglesias
A
,
Bajpai
R
,
Swigut
T
,
Brugmann
SA
,
Flynn
RA
,
Wysocka
J
. 
A unique chromatin signature uncovers early developmental enhancers in humans
.
Nature
2011
;
470
:
279
85
.
16.
Fraser
P
,
Grosveld
F
. 
Locus control regions, chromatin activation and transcription
.
Curr Opin Cell Biol
1998
;
10
:
361
65
.
17.
Li
Q
,
Peterson
KR
,
Fang
X
,
Stamatoyannopoulos
G
. 
Locus control regions
.
Blood
2002
;
100
:
3077
86
.
18.
Sengupta
S
,
George
RE
. 
Super-enhancer-driven transcriptional dependencies in cancer
.
Trends Cancer
2017
;
3
:
269
81
.
19.
Creyghton
MP
,
Cheng
AW
,
Welstead
GG
,
Kooistra
T
,
Carey
BW
,
Steine
EJ
, et al
Histone H3K27ac separates active from poised enhancers and predicts developmental state
.
Proc Natl Acad Sci U S A
2010
;
107
:
21931
36
.
20.
Sur
I
,
Taipale
J
. 
The role of enhancers in cancer
.
Nat Rev Cancer
2016
;
16
:
483
93
.
21.
Zhang
Z
,
Lee
JH
,
Ruan
H
,
Ye
Y
,
Krakowiak
J
,
Hu
Q
, et al
Transcriptional landscape and clinical utility of enhancer RNAs for eRNA-targeted therapy in cancer
.
Nat Commun
2019
;
10
:
4562
.
22.
Lam
MTY
,
Li
W
,
Rosenfeld
MG
,
Glass
CK
. 
Enhancer RNAs and regulated transcriptional programs
.
Trends Biochem Sci
2014
;
39
:
170
82
.
23.
Koch
F
,
Fenouil
R
,
Gut
M
,
Cauchy
P
,
Albert
TK
,
Zacarias-Cabeza
J
, et al
Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters
.
Nat Struct Mol Biol
2011
;
18
:
956
63
.
24.
de Santa
F
,
Barozzi
I
,
Mietton
F
,
Ghisletti
S
,
Polletti
S
,
Tusi
BK
, et al
A large fraction of extragenic RNA Pol II transcription sites overlap enhancers
.
PLoS Biol
2010
;
8
:
e1000384
.
25.
Heinz
S
,
Benner
C
,
Spann
N
,
Bertolino
E
,
Lin
YC
,
Laslo
P
, et al
Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities
.
Mol Cell
2010
;
38
:
576
89
.
26.
Kim
TK
,
Hemberg
M
,
Gray
JM
,
Costa
AM
,
Bear
DM
,
Wu
J
, et al
Widespread transcription at neuronal activity-regulated enhancers
.
Nature
2010
;
465
:
182
87
.
27.
Ghisletti
S
,
Barozzi
I
,
Mietton
F
,
Polletti
S
,
De Santa
F
,
Venturini
E
, et al
Identification and characterization of enhancers controlling the inflammatory gene expression program in macrophages
.
Immunity
2010
;
32
:
317
28
.
28.
Plank
JL
,
Dean
A
. 
Enhancer function: mechanistic and genome-wide insights come together
.
Mol Cell
2014
;
55
:
5
14
.
29.
Sedano
MJ
,
Harrison
AL
,
Zilaie
M
,
Das
C
,
Choudhari
R
,
Ramos
E
, et al
Emerging roles of estrogen-regulated enhancer and long non-coding RNAs
.
Int J Mol Sci
2020
;
21
:
3711
.
30.
Murakami
S
,
Gadad
SS
,
Kraus
WL
. 
A PreSTIGEous use of LncRNAs to predict enhancers
.
Cell Cycle
2015
;
14
:
1619
20
.
31.
Hou
TY
,
Kraus
WL
. 
Spirits in the material world: enhancer RNAs in transcriptional regulation
.
Trends Biochem Sci
2021
;
46
:
138
53
.
32.
Lee
JH
,
Xiong
F
,
Li
W
. 
Enhancer RNAs in cancer: regulation, mechanisms and therapeutic potential
.
RNA Biol
2020
;
7
:
1550
59
.
33.
Andersson
R
,
Refsing Andersen
P
,
Valen
E
,
Core
LJ
,
Bornholdt
J
,
Boyd
M
, et al
Nuclear stability and transcriptional directionality separate functionally distinct RNA species
.
Nat Commun
2014
;
5
:
5336
.
34.
Pefanis
E
,
Wang
J
,
Rothschild
G
,
Lim
J
,
Kazadi
D
,
Sun
J
, et al
RNA exosome-regulated long non-coding RNA transcription controls super-enhancer activity
.
Cell
2015
;
161
:
774
89
.
35.
Lai
F
,
Gardini
A
,
Zhang
A
,
Shiekhattar
R
. 
Integrator mediates the biogenesis of enhancer RNAs
.
Nature
2015
;
525
:
399
403
.
36.
Imamura
K
,
Takaya
A
,
Ishida
Y
,
Fukuoka
Y
,
Taya
T
,
Nakaki
R
, et al
Diminished nuclear RNA decay upon Salmonella infection upregulates antibacterial noncoding RNAs
.
EMBO J
2018
;
37
:
e97723
.
37.
Ntini
E
,
Järvelin
AI
,
Bornholdt
J
,
Chen
Y
,
Boyd
M
,
Jørgensen
M
, et al
Polyadenylation site-induced decay of upstream transcripts enforces promoter directionality
.
Nat Struct Mol Biol
2013
;
20
:
923
8
.
38.
Aguilo
F
,
Li
SiDe
,
Balasubramaniyan
N
,
Sancho
A
,
Benko
S
,
Zhang
F
, et al
Deposition of 5-methylcytosine on enhancer RNAs enables the coactivator function of PGC-1α
.
Cell Rep
2016
;
14
:
479
92
.
39.
Lam
MTY
,
Cho
H
,
Lesch
HP
,
Gosselin
D
,
Heinz
S
,
Tanaka-Oishi
Y
, et al
Rev-Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription
.
Nature
2013
;
498
:
511
5
.
40.
Arner
E
,
Daub
CO
,
Vitting-Seerup
K
,
Andersson
R
,
Lilje
B
,
Drabløs
F
, et al
Transcribed enhancers lead waves of coordinated transcription in transitioning mammalian cells
.
Science
2015
;
347
:
1010
4
.
41.
Murakawa
Y
,
Yoshihara
M
,
Kawaji
H
,
Nishikawa
M
,
Zayed
H
,
Suzuki
H
, et al
Enhanced identification of transcriptional enhancers provides mechanistic insights into diseases
.
Trends Genet
2016
;
32
:
76
88
.
42.
Li
W
,
Notani
D
,
Ma
Q
,
Tanasa
B
,
Nunez
E
,
Chen
AY
, et al
Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation
.
Nature
2013
;
498
:
516
20
.
43.
Hah
N
,
Danko
CG
,
Core
L
,
Waterfall
JJ
,
Siepel
A
,
Lis
JT
, et al
A rapid, extensive, and transient transcriptional response to estrogen signaling in breast cancer cells
.
Cell
2011
;
145
:
622
34
.
44.
Core
LJ
,
Waterfall
JJ
,
Lis
JT
. 
Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters
.
Science
2008
;
322
:
1845
48
.
45.
Schwalb
B
,
Michel
M
,
Zacher
B
,
Hauf
KF
,
Demel
C
,
Tresch
A
, et al
TT-seq maps the human transient transcriptome
.
Science
2016
;
352
:
1225
28
.
46.
Core
LJ
,
Martins
AL
,
Danko
CG
,
Waters
CT
,
Siepel
A
,
Lis
JT
. 
Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers
.
Nat Genet
2014
;
46
:
1311
20
.
47.
Wang
D
,
Garcia-Bassets
I
,
Benner
C
,
Li
W
,
Su
X
,
Zhou
Y
, et al
Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA
.
Nature
2011
;
474
:
390
94
.
48.
Kwak
H
,
Fuda
NJ
,
Core
LJ
,
Lis
JT
. 
Precise maps of RNA polymerase reveal how promoters direct initiation and pausing
.
Science
2013
;
339
:
950
53
.
49.
Forrest
ARR
,
Kawaji
H
,
Rehli
M
,
Baillie
JK
,
De Hoon
MJL
,
Haberle
V
, et al
A promoter-level mammalian expression atlas
.
Nature
2014
;
507
:
462
70
.
50.
Blinka
S
,
Reimer
MH
,
Pulakanti
K
,
Pinello
L
,
Yuan
GC
,
Rao
S
. 
Identification of transcribed enhancers by genome-wide chromatin immunoprecipitation sequencing
.
Methods Mol Biol
2017
;
1468
:
91
109
.
51.
Magnuson
B
,
Veloso
A
,
Kirkconnell
KS
,
De Andrade Lima
LC
,
Paulsen
MT
,
Ljungman
EA
, et al
Identifying transcription start sites and active enhancer elements using BruUV-seq
.
Sci Rep
2015
;
5
:
17978
.
52.
Shibayama
Y
,
Fanucchi
S
,
Mhlanga
MM
. 
Visualization of enhancer-derived noncoding RNA
.
Methods Mol Biol
2017
;
1468
:
19
32
.
53.
Hsieh
CL
,
Fei
T
,
Chen
Y
,
Li
T
,
Gao
Y
,
Wang
X
, et al
Enhancer RNAs participate in androgen receptor-driven looping that selectively enhances gene activation
.
Proc Natl Acad Sci U S A
2014
;
111
:
7319
24
.
54.
Schaukowitch
K
,
Joo
JY
,
Liu
X
,
Watts
JK
,
Martinez
C
,
Kim
TK
. 
Enhancer RNA facilitates NELF release from immediate early genes
.
Mol Cell
2014
;
56
:
29
42
.
55.
Sigova
AA
,
Abraham
BJ
,
Ji
X
,
Molinie
B
,
Hannett
NM
,
Guo
YE
, et al
Transcription factor trapping by RNA in gene regulatory elements
.
Science
2015
;
350
:
978
81
.
56.
Adelman
K
,
Lis
JT
. 
Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans
.
Nat Rev Genet
2012
;
13
:
720
31
.
57.
Shechner
DM
,
Hacisuleyman
E
,
Younger
ST
,
Rinn
JL
. 
Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display
.
Nat Methods
2015
;
12
:
664
70
.
58.
Bassett
AR
,
Akhtar
A
,
Barlow
DP
,
Bird
AP
,
Brockdorff
N
,
Duboule
D
, et al
Considerations when investigating lncRNA function in vivo
.
Elife
2014
;
3
:
1
14
.
59.
Mousavi
K
,
Zare
H
,
Dell'Orso
S
,
Grontved
L
,
Gutierrez-Cruz
G
,
Derfoul
A
, et al
ERNAs promote transcription by establishing chromatin accessibility at defined genomic loci
.
Mol Cell
2013
;
51
:
606
17
.
60.
Melo
CA
,
Drost
J
,
Wijchers
PJ
,
van de Werken
H
,
de Wit
E
,
Vrielink
JAFO
, et al
ERNAs are required for p53-dependent enhancer activity and gene transcription
.
Mol Cell
2013
;
49
:
524
35
.
61.
Kaikkonen
MU
,
Spann
NJ
,
Heinz
S
,
Romanoski
CE
,
Allison
KA
,
Stender
JD
, et al
Remodeling of the enhancer landscape during macrophage activation is coupled to enhancer transcription
.
Mol Cell
2013
;
51
:
310
25
.
62.
Shii
L
,
Song
L
,
Maurer
K
,
Zhang
Z
,
Sullivan
KE
. 
SERPINB2 is regulated by dynamic interactions with pause-release proteins and enhancer RNAs
.
Mol Immunol
2017
;
88
:
20
31
.
63.
Cinghu
S
,
Yang
P
,
Kosak
JP
,
Conway
AE
,
Kumar
D
,
Oldfield
AJ
, et al
Intragenic enhancers attenuate host gene expression
.
Mol Cell
2017
;
68
:
104
17
.
64.
Dorighi
KM
,
Swigut
T
,
Henriques
T
,
Bhanu
NV
,
Scruggs
BS
,
Nady
N
, et al
Mll3 and Mll4 facilitate enhancer RNA synthesis and transcription from promoters independently of H3K4 monomethylation
.
Mol Cell
2017
;
66
:
568
76
.
65.
Tan
Y
,
Jin
C
,
Ma
W
,
Hu
Y
,
Tanasa
B
,
Oh
S
, et al
Dismissal of RNA polymerase II underlies a large ligand-induced enhancer decommissioning program
.
Mol Cell
2018
;
71
:
526
39
.
66.
Jiao
W
,
Chen
Y
,
Song
H
,
Li
D
,
Mei
H
,
Yang
F
, et al
HPSE enhancer RNA promotes cancer progression through driving chromatin looping and regulating hnRNPU/p300/EGR1/HPSE axis
.
Oncogene
2018
;
37
:
2728
45
.
67.
van Arensbergen
J
,
van Steensel
B
,
Bussemaker
HJ
. 
In search of the determinants of enhancer-promoter interaction specificity
.
Trends Cell Biol
2014
;
24
:
695
702
.
68.
Sanyal
A
,
Lajoie
BR
,
Jain
G
,
Dekker
J
. 
The long-range interaction landscape of gene promoters
.
Nature
2012
;
489
:
109
13
.
69.
Pezone
A
,
Zuchegna
C
,
Tramontano
A
,
Romano
A
,
Russo
G
,
de Rosa
M
, et al
RNA stabilizes transcription-dependent chromatin loops induced by nuclear hormones
.
Sci Rep
2019
;
9
:
3925
.
70.
Kim
YW
,
Lee
S
,
Yun
J
,
Kim
AR
. 
Chromatin looping and eRNA transcription precede the transcriptional activation of gene in the β-globin locus
.
Biosci Rep
2015
;
35
:
e00179
.
71.
Cajigas
I
,
Chakraborty
A
,
Swyter
KR
,
Luo
H
,
Bastidas
M
,
Nigro
M
, et al
The Evf2 ultraconserved enhancer lncRNA functionally and spatially organizes megabase distant genes in the developing forebrain
.
Mol Cell
2018
;
71
:
956
72
.
72.
Tsai
PF
,
Dell'Orso
S
,
Rodriguez
J
,
Vivanco
KO
,
Ko
KD
,
Jiang
K
, et al
A muscle-specific enhancer RNA mediates cohesin recruitment and regulates transcription In trans
.
Mol Cell
2018
;
71
:
129
41
.
73.
Pnueli
L
,
Rudnizky
S
,
Yosefzon
Y
,
Melamed
P
. 
RNA transcribed from a distal enhancer is required for activating the chromatin at the promoter of the gonadotropin α-subunit gene
.
Proc Natl Acad Sci U S A
2015
;
112
:
4369
74
.
74.
Lai
F
,
Orom
UA
,
Cesaroni
M
,
Beringer
M
,
Taatjes
DJ
,
Blobel
GA
, et al
Activating RNAs associate with mediator to enhance chromatin architecture and transcription
.
Nature
2013
;
494
:
497
501
.
75.
Chiang
MY
. 
Finding ARIEL under the sea of T-ALL circuits
.
Blood
2019
;
134
:
219
21
.
76.
Xiang
JF
,
Yin
QF
,
Chen
T
,
Zhang
Y
,
Zhang
XO
,
Wu
Z
, et al
Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus
.
Cell Res
2014
;
24
:
513
31
.
77.
Kaneko
S
,
Son
J
,
Shen
SS
,
Reinberg
D
,
Bonasio
R
. 
PRC2 binds active promoters and contacts nascent RNAs in embryonic stem cells
.
Nat Struct Mol Biol
2013
;
20
:
1258
64
.
78.
Cruz-Molina
S
,
Respuela
P
,
Tebartz
C
,
Kolovos
P
,
Nikolic
M
,
Fueyo
R
, et al
PRC2 facilitates the regulatory topology required for poised enhancer function during pluripotent stem cell differentiation
.
Cell Stem Cell
2017
;
20
:
689
705
.
79.
Barbieri
E
,
Trizzino
M
,
Welsh
SA
,
Owens
TA
,
Calabretta
B
,
Carroll
M
, et al
Targeted enhancer activation by a subunit of the integrator complex
.
Mol Cell
2018
;
71
:
103
16
.
80.
Hah
N
,
Murakami
S
,
Nagari
A
,
Danko
CG
,
Lee Kraus
W
. 
Enhancer transcripts mark active estrogen receptor binding sites
.
Genome Res
2013
;
23
:
1210
23
.
81.
Sanz
LA
,
Hartono
SR
,
Lim
YW
,
Steyaert
S
,
Rajpurkar
A
,
Ginno
PA
, et al
Prevalent, dynamic, and conserved R-loop structures associate with specific epigenomic signatures in mammals
.
Mol Cell
2016
;
63
:
167
78
.
82.
Bhatia
V
,
Barroso
SI
,
García-Rubio
ML
,
Tumini
E
,
Herrera-Moyano
E
,
Aguilera
A
. 
BRCA2 prevents R-loop accumulation and associates with TREX-2 mRNA export factor PCID2
.
Nature
2014
;
511
:
362
65
.
83.
Gan
W
,
Guan
Z
,
Liu
J
,
Gui
T
,
Shen
K
,
Manley
JL
, et al
R-loop-mediated genomic instability is caused by impairment of replication fork progression
.
Genes Dev
2011
;
25
:
2041
56
.
84.
Wasmuth
EV
,
Lima
CD
. 
Structure and activities of the eukaryotic RNA exosome
.
Enzymes
2012
;
31
:
53
75
.
85.
Laffleur
B
,
Lim
J
,
Zhang
W
,
Chen
Y
,
Pefanis
E
,
Bizarro
J
, et al
Noncoding RNA processing by DIS3 regulates chromosomal architecture and somatic hypermutation in B cells
.
Nat Genet
2021
;
53
:
230
42
.
86.
Chen
H
,
Li
C
,
Peng
X
,
Zhou
Z
,
Weinstein
JN
Cancer Genome Atlas Research Network
et al 
A pan-cancer analysis of enhancer expression in nearly 9000 patient samples
.
Cell
2018
;
173
:
386
99
.
87.
Schmitt
AD
,
Hu
M
,
Ren
B
. 
Genome-wide mapping and analysis of chromosome architecture
.
Nat Rev Mol Cell Biol
2016
;
17
:
743
55
.
88.
Li
W
,
Hu
Y
,
Oh
S
,
Ma
Q
,
Merkurjev
D
,
Song
X
, et al
Condensin I and II complexes license full estrogen receptor α-dependent enhancer activation
.
Mol Cell
2015
;
59
:
188
202
.
89.
Yamaoka
A
,
Suzuki
M
,
Katayama
S
,
Orihara
D
,
Engel
JD
,
Yamamoto
M
. 
EVI1 and GATA2 misexpression induced by inv(3)(q21q26) contribute to megakaryocyte-lineage skewing and leukemogenesis
.
Blood Adv
2020
;
4
:
1722
36
.
90.
Roe
JS
,
Hwang
CI
,
Somerville
TDD
,
Milazzo
JP
,
Lee
EJ
,
Da Silva
B
, et al
Enhancer reprogramming promotes pancreatic cancer metastasis
.
Cell
2017
;
170
:
875
88
.
91.
Somerville
TDD
,
Xu
Y
,
Miyabayashi
K
,
Tiriac
H
,
Cleary
CR
,
Maia-Silva
D
, et al
TP63-mediated enhancer reprogramming drives the squamous subtype of pancreatic ductal adenocarcinoma
.
Cell Rep
2018
;
25
:
1741
55
.
92.
Hamdan
FH
,
Johnsen
SA
. 
DeltaNp63-dependent super enhancers define molecular identity in pancreatic cancer by an interconnected transcription factor network
.
Proc Natl Acad Sci U S A
2018
;
115
:
e12343
52
.
93.
Léveillé
N
,
Melo
CA
,
Agami
R
. 
Enhancer-associated RNAs as therapeutic targets
.
Expert Opin Biol Ther
2015
;
15
:
723
34
.
94.
Léveillé
N
,
Melo
CA
,
Rooijers
K
,
Díaz-Lagares
A
,
Melo
SA
,
Korkmaz
G
, et al
Genome-wide profiling of p53-regulated enhancer RNAs uncovers a subset of enhancers controlled by a lncRNA
.
Nat Commun
2015
;
6
:
6520
.
95.
Bal
E
,
Park
HS
,
Belaid-Choucair
Z
,
Kayserili
H
,
Naville
M
,
Madrange
M
, et al
Mutations in ACTRT1 and its enhancer RNA elements lead to aberrant activation of Hedgehog signaling in inherited and sporadic basal cell carcinomas
.
Nat Med
2017
;
23
:
1226
33
.
96.
He
B
,
Gao
P
,
Ding
YY
,
Chen
CH
,
Chen
G
,
Chen
C
, et al
Diverse noncoding mutations contribute to deregulation of cis-regulatory landscape in pediatric cancers
.
Sci Adv
2020
;
6
:
3064
.
97.
Hnisz
D
,
Schuijers
J
,
Lin
CY
,
Weintraub
AS
,
Abraham
BJ
,
Lee
TI
, et al
Convergence of developmental and oncogenic signaling pathways at transcriptional super-enhancers
.
Mol Cell
2015
;
58
:
362
70
.
98.
Zhao
Y
,
Wang
L
,
Ren
S
,
Wang
L
,
Blackburn
PR
,
McNulty
MS
, et al
Activation of P-TEFb by androgen receptor-regulated enhancer RNAs in castration-resistant prostate cancer
.
Cell Rep
2016
;
15
:
599
610
.
99.
McCleland
ML
,
Mesh
K
,
Lorenzana
E
,
Chopra
VS
,
Segal
E
,
Watanabe
C
, et al
CCAT1 is an enhancer-templated RNA that predicts BET sensitivity in colorectal cancer
.
J Clin Invest
2016
;
126
:
639
52
.
100.
Galli
GG
,
Carrara
M
,
Yuan
WC
,
Valdes-Quezada
C
,
Gurung
B
,
Pepe-Mooney
B
, et al
YAP drives growth by controlling transcriptional pause release from dynamic enhancers
.
Mol Cell
2015
;
60
:
328
37
.
101.
Wang
H
,
Zang
C
,
Taing
L
,
Arnett
KL
,
Wong
YJ
,
Pear
WS
, et al
NOTCH1-RBPJ complexes drive target gene expression through dynamic interactions with superenhancers
.
Proc Natl Acad Sci U S A
2014
;
111
:
705
10
.
102.
Zhu
C
,
Li
L
,
Zhang
Z
,
Bi
M
,
Wang
H
,
Su
W
, et al
A non-canonical role of YAP/TEAD is required for activation of estrogen-regulated enhancers in breast cancer
.
Mol Cell
2019
;
75
:
791
806
.
103.
Rahnamoun
H
,
Lu
H
,
Duttke
SH
,
Benner
C
,
Glass
CK
,
Lauberth
SM
. 
Mutant p53 shapes the enhancer landscape of cancer cells in response to chronic immune signaling
.
Nat Commun
2017
;
8
:
754
.
104.
Rahnamoun
H
,
Lee
J
,
Sun
Z
,
Lu
H
,
Ramsey
KM
,
Komives
EA
, et al
RNAs interact with BRD4 to promote enhanced chromatin engagement and transcription activation
.
Nat Struct Mol Biol
2018
;
25
:
687
97
.
105.
Rivera
E
,
Gomez
H
. 
Chemotherapy resistance in metastatic breast cancer: the evolving role of ixabepilone
.
Breast Cancer Res
2010
;
2
:
S2
.
106.
Wind
NS
,
Holen
I
. 
Multidrug resistance in breast cancer: from in vitro models to clinical studies
.
Int J Breast Cancer
2011
;
967419
.
107.
Castells
M
,
Thibault
B
,
Delord
JP
,
Couderc
B
. 
Implication of tumor microenvironment in chemoresistance: tumor-associated stromal cells protect tumor cells from cell death
.
Int J Mol Sci
2012
;
13
:
9545
71
.
108.
Uramoto
H
,
Tanaka
F
. 
Recurrence after surgery in patients with NSCLC
.
Transl Lung Cancer Res
2014
;
3
:
242
49
.
109.
O'Connor
D
,
Sibson
K
,
Caswell
M
,
Connor
P
,
Cummins
M
,
Mitchell
C
, et al
Early UK experience in the use of clofarabine in the treatment of relapsed and refractory paediatric acute lymphoblastic leukaemia
.
Br J Haematol
2011
;
154
:
482
85
.
110.
Schmitt
AM
,
Chang
HY
. 
Long noncoding RNAs in cancer pathways
.
Cancer Cell
2016
;
29
:
452
63
.
111.
Morlando
M
,
Fatica
A
. 
Alteration of epigenetic regulation by long noncoding RNAs in cancer
.
Int J Mol Sci
2018
;
19
:
570
.
112.
Hamdan
FH
,
Johnsen
SA
. 
Perturbing enhancer activity in cancer therapy
.
Cancers
2019
;
11
:
634
.
113.
Call
SG
,
Duren
RP
,
Panigrahi
AK
,
Nguyen
L
,
Freire
PR
,
Grimm
SL
, et al
Targeting oncogenic super enhancers in MYC-dependent AML using a small molecule activator of NR4A nuclear receptors
.
Sci Rep
2020
;
10
:
2851
.
114.
Trimarchi
T
,
Bilal
E
,
Ntziachristos
P
,
Fabbri
G
,
Dalla-Favera
R
,
Tsirigos
A
, et al
Genome-wide mapping and characterization of notch-regulated long noncoding RNAs in acute leukemia
.
Cell
2014
;
158
:
593
606
.
115.
Yokoyama
Y
,
Zhu
H
,
Lee
JH
,
Kossenkov
AV
,
Wu
SY
,
Wickramasinghe
JM
, et al
BET inhibitors suppress ALDH activity by targeting ALDH1A1 super-enhancer in ovarian cancer
.
Cancer Res
2016
;
76
:
6320
30
.
116.
Bi
M
,
Zhang
Z
,
Jiang
YZ
,
Xue
P
,
Wang
H
,
Lai
Z
, et al
Enhancer reprogramming driven by high-order assemblies of transcription factors promotes phenotypic plasticity and breast cancer endocrine resistance
.
Nat Cell Biol
2020
;
22
:
701
15
.
117.
Zhao
J
,
Zhao
Y
,
Wang
L
,
Zhang
J
,
Karnes
RJ
,
Kohli
M
, et al
Alterations of androgen receptor-regulated enhancer RNAs (eRNAs) contribute to enzalutamide resistance in castration-resistant prostate cancer
.
Oncotarget
2016
;
7
:
38551
65
.
118.
Williams
MS
,
Amaral
FMR
,
Simeoni
F
,
Somervaille
TCP
. 
A stress-responsive enhancer induces dynamic drug resistance in acute myeloid leukemia
.
J Clin Invest
2020
;
130
:
1217
32
.
119.
Zhang
X
,
Choi
PS
,
Francis
JM
,
Imielinski
M
,
Watanabe
H
,
Cherniack
AD
, et al
Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers
.
Nat Genet
2016
;
48
:
176
82
.
120.
Bahr
C
,
Von Paleske
L
,
Uslu
VV
,
Remeseiro
S
,
Takayama
N
,
Ng
SW
, et al
A Myc enhancer cluster regulates normal and leukaemic haematopoietic stem cell hierarchies
.
Nature
2018
;
553
:
515
20
.
121.
Mansour
MR
,
Abraham
BJ
,
Anders
L
,
Berezovskaya
A
,
Gutierrez
A
,
Durbin
AD
, et al
An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element
.
Science
2014
;
346
:
1373
7
.
122.
Tan
SH
,
Leong
WZ
,
Ngoc
PCT
,
Tan
TK
,
Bertulfo
FC
,
Lim
MC
, et al
The enhancer RNA ARIEL activates the oncogenic transcriptional program in T-cell acute lymphoblastic leukemia
.
Blood
2019
;
134
:
239
51
.
123.
Ding
M
,
Zhan
H
,
Liao
X
,
Li
A
,
Zhong
Y
,
Gao
Q
, et al
Enhancer RNA – P2RY2e induced by estrogen promotes malignant behaviors of bladder cancer
.
Int J Biol Sci
2018
;
14
:
1268
76
.
124.
Liang
J
,
Zhou
H
,
Gerdt
C
,
Tan
M
,
Colson
T
,
Kaye
KM
, et al
Epstein-Barr virus super-enhancer eRNAs are essential for MYC oncogene expression and lymphoblast proliferation
.
Proc Natl Acad Sci U S A
2016
;
113
:
14121
26
.
125.
Chu
C
,
Zhang
QC
,
Da Rocha
ST
,
Flynn
RA
,
Bharadwaj
M
,
Calabrese
JM
, et al
Systematic discovery of Xist RNA binding proteins
.
Cell
2015
;
161
:
404
16
.
126.
McHugh
CA
,
Chen
CK
,
Chow
A
,
Surka
CF
,
Tran
C
,
McDonel
P
, et al
The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3
.
Nature
2015
;
521
:
232
36
.
127.
Minajigi
A
,
Froberg
JE
,
Wei
C
,
Sunwoo
H
,
Kesner
B
,
Colognori
D
, et al
A comprehensive Xist interactome reveals cohesin repulsion and an RNA-directed chromosome conformation
.
Science
2015
;
349
:
aab2276
.
128.
Batista
PJ
,
Molinie
B
,
Wang
J
,
Qu
K
,
Zhang
J
,
Li
L
, et al
M6A RNA modification controls cell fate transition in mammalian embryonic stem cells
.
Cell Stem Cell
2014
;
15
:
707
19
.
129.
Liu
N
,
Dai
Q
,
Zheng
G
,
He
C
,
Parisien
M
,
Pan
T
. 
N6 -methyladenosine-dependent RNA structural switches regulate RNA-protein interactions
.
Nature
2015
;
518
:
560
64
.
130.
Liu
RJ
,
Long
T
,
Li
J
,
Li
H
,
Wang
ED
. 
Structural basis for substrate binding and catalytic mechanism of a human RNA:m5C methyltransferase NSun6
.
Nucleic Acids Res
2017
;
45
:
6684
97
.
131.
Roovers
M
,
Wouters
J
,
Bujnicki
JM
,
Tricot
C
,
Stalon
V
,
Grosjean
H
, et al
A primordial RNA modification enzyme: the case of tRNA (m1A) methyltransferase
.
Nucleic Acids Res
2004
;
32
:
465
76
.
132.
Li
X
,
Fu
XD
. 
Chromatin-associated RNAs as facilitators of functional genomic interactions
.
Nat Rev Genet
2019
;
20
:
503
19
.
133.
Xiao
R
,
Chen
JY
,
Liang
Z
,
Luo
D
,
Chen
G
,
Lu
ZJ
, et al
Pervasive chromatin-RNA binding protein interactions enable RNA-based regulation of transcription
.
Cell
2019
;
178
:
107
21
.