Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2014 Oct;4(10):a014266. doi: 10.1101/cshperspect.a014266

Therapeutic Strategies to Inhibit MYC

Michael R McKeown 1, James E Bradner 1,2,3
PMCID: PMC4200208  PMID: 25274755

Each year, an estimated 450,000 Americans are diagnosed with MYC-dependent cancer. The development of direct-acting MYC inhibitors remains a challenge; indirect modulators of MYC may have more potential.

Abstract

MYC is a master regulator of stem cell state, embryogenesis, tissue homeostasis, and aging. As in health, in disease MYC figures prominently. Decades of biological research have identified a central role for MYC in the pathophysiology of cancer, inflammation, and heart disease. The centrality of MYC to such a vast breadth of disease biology has attracted significant attention to the historic challenge of developing inhibitors of MYC. This review will discuss therapeutic strategies toward the development of inhibitors of MYC-dependent transcriptional signaling, efforts to modulate MYC stability, and the elusive goal of developing potent, direct-acting inhibitors of MYC.


MYC (v-myc myelocytomatosis viral oncogene homolog) is a nuclear DNA-binding transcription factor that orchestrates transcriptional regulatory pathways underlying cell growth, cell-cycle progression, metabolism, and survival (Meyer and Penn 2008; Conacci-Sorrell et al. 2014). MYC was first described 30 years ago in the context of viral oncogenesis (Duesberg and Vogt 1979; Hu and Vogt 1979; Sheiness and Bishop 1979), and subsequently a human homolog was identified and characterized (Dalla-Favera et al. 1982; Taub et al. 1982) Since its discovery, MYC has emerged as a master regulator of numerous biological and disease processes.

MYC is the most frequently amplified oncogene in human cancers, and MYC alteration is observed in a wide range of tissue types including breast, lung, and prostate cancer (Beroukhim et al. 2010). MYC overexpression occurs in 30% of all human cancers and frequently predicts for a poor clinical outcome, aggressive biological behavior, increased likelihood of relapse, and advanced stage of disease (Gamberi et al. 1998; Nesbit et al. 1999). An estimated 450,000 Americans each year are diagnosed with a MYC-dependent cancer. Deregulated expression of MYC is a hallmark feature of cancer (Conacci-Sorrell et al. 2014; Gabay et al. 2014), uncoupling physiologic, growth factor–dependent proliferation. Deregulated expression of MYC in cancer occurs through gene amplification (Sauter et al. 1995), chromosomal translocation (Klein 1983; Leder et al. 1983), focal enhancer amplification (Herrmann et al. 2012), germline enhancer polymorphism (Easton et al. 2007; Greenman et al. 2007; Kiemeney et al. 2008; Pomerantz et al. 2009; Wright et al. 2010), or commonly through constitutive activation of upstream signaling pathways. Each of these mechanisms serves to uncouple physiologic, growth factor–dependent proliferation.

MYC has been multiply validated as essential for tumor initiation and maintenance in numerous tumor histologies (Huang and Weiss 2013; Roussel and Robinson 2013; Gabay et al. 2014; Schmitz et al. 2014). Studies in transgenic mouse models identify that MYC inactivation leads to prompt tumor regressions, often associated with phenotypes of differentiation and cellular senescence with or without apoptosis (Felsher and Bishop 1999; Pelengaris et al. 1999, 2002; Jain et al. 2002; Flores et al. 2004; Soucek et al. 2008). In models of osteosarcoma, even brief MYC inactivation significantly improved survival rates owing to terminal differentiation of tumor cells (Jain et al. 2002).

The pleiotropic role of MYC in developmental biology and tissue homeostasis has prompted reasonable concern that targeting MYC may provoke severe, untoward toxicity. Indeed, genetic ablation of MYC in mice is embryonic lethal at an early stage of gestation (between days 9.5 and 10.5), with failed hematopoietic development (Davis et al. 1993; Hurlin 2013). Although subsequent studies have shown tolerance of tissue-specific MYC loss or down-regulation, such as in the liver and small intestine (Bettess et al. 2005; Hurlin 2013), concerns remain that pharmacologic inhibition of MYC may feature a narrow therapeutic index. For example, tolerance of MYC loss in intestinal epithelium could be conferred by compensatory up-regulation of MYCN (Bettess et al. 2005; Hurlin 2013), suggesting that selective inhibition of MYC-family isoforms may be required to firmly establish a therapeutic index.

Recently, a powerful genetic instrument developed by Evan and colleagues has provided compelling evidence that a therapeutic index does exist for targeting MYC. Structure-guided design allowed the optimization and characterization of a 93 residue, dominant-negative peptide comprising the basic helix-loop-helix leucine zipper (bHLHZ) domain (see Conacci-Sorrell et al. 2014) called Omomyc (Soucek et al. 2008; Savino et al. 2011). Four amino acid substitutions in the extended leucine zipper domain relieve repulsive charge interactions allowing for a coiled-coil-binding event to occur, presumably with endogenous MYC. Thus, Omomyc functions to competitively bind MYC in a manner preventing MYC:MAX heterodimerization, and expression of Omomyc prompts rapid growth arrest and down-regulation of MYC target genes (Soucek et al. 1998, 2002). Tetracycline-inducible expression of Omomyc has allowed a simulation of what pharmacologic modulation of MYC function may hold, in vitro and in vivo. As described in Gabay et al. (2014), constitutive induction of Omomyc prompts tonic inhibition of MYC:MAX heterodimerization associated with significant toxicity in proliferative tissue compartments in rodents (bone marrow and bowel). However, periodic induction of Omomyc has proven well tolerated. Dosing regimens have already been identified that provoke unprecedented responses in aggressive murine models of cancer, notably including the KRAS:TP53 model of non-small-cell lung carcinoma (Soucek et al. 2013). This research further validates MYC as a therapeutic target in cancer, importantly downstream from other intractable protein targets and tumor suppressors, and establishes a therapeutic index for MYC inhibition, in vivo.

Beyond cancer, MYC is also a positive effector of tissue inflammation. Activation of MYC is observed in immune cells during inflammatory cell expansion (Wang et al. 2011). MYC amplifies the transcriptional response to inflammatory transcription factor signaling in conditions such as rheumatoid arthritis (Pap et al. 2004). MYC function has also been implicated in the pathophysiology of heart failure, during tissue remodeling associated with hypertrophy and dilatation (Ahuja et al. 2010). Hence, the ability to inhibit MYC could extend beyond cancer therapy.

TOWARD DIRECT INHIBITION OF MYC

In the postgenomic era, the emergent paradigm of drug discovery is increasingly target directed. Disease biology identifies a critical effector mediating the pathophysiology of illness or the maladaptive response, and target-directed ligand discovery is undertaken. Innovation in the science of therapeutics over the last three decades has generated diverse strategies, which may be deployed to realize direct-acting inhibitors of protein targets. Still, the prioritization of targets is heavily influenced by perceptions of success and the horizons of supported research (Rask-Andersen et al. 2011; London et al. 2013; Swinney 2013).

As a class, transcription factors have proven particularly evasive in the discipline of ligand discovery (Darnell 2002; Berg 2008). Transcription factors are master regulators of cell state, controlling gene expression programs driving cell type specification and contributing to the pathophysiology of a broad range of diseases (Ptashne and Gann 1997, 2002). As a class, transcription factors are highly desirable proteins for therapeutic targeting. Yet, also as a class, transcription factors biophysically associate through extensive interfacial associations featuring large surface areas, a lack of hydrophobic invaginations, and noncontiguous contacts. These features are at odds with traditional binding models for small organic drug molecules (Fletcher and Hamilton 2007). The number of transcription factors successfully approached to date with discovery chemistry is therefore rather small. It has been our experience, developing the first direct-acting inhibitors of the NOTCH1 transactivation complex for probe and therapeutic development in T-cell acute lymphoblastic leukemia, that significant challenges exist in each phase of assay development, small-molecule optimization, biophysical characterization, mechanistic validation, and cellular permeability (Moellering et al. 2009). For these and other reasons, despite a confluence of detailed mechanistic insights and unmet medical needs, direct inhibition of MYC remains a historic challenge (Meyer and Penn 2008). Indeed, MYC has emerged as arguably the prototypical example of an “undruggable target,” a term reserved for protein targets that have proven intractable to coordinated efforts in ligand discovery.

MYC presents specific, significant obstacles to discovery chemistry. MYC lacks enzymatic activity, limiting many effective approaches to direct inhibition. Rather, MYC functions via protein–protein interactions, which remain a technical and psychological barrier to organized efforts in drug discovery (Wells and McClendon 2007). Structurally, MYC lacks globular functional domains, which might be approached with structure-based or empirical biochemical screening. MYC is comprised of a largely unstructured amino-terminal region involved in transactivation and protein stability (Conacci-Sorrell et al. 2014; Farrell and Sears 2014). Four highly conserved functional modules exist at the amino terminus, termed MYC boxes (MBI–IV). MBI possesses a degron targeted by FBW-7 (Welcker et al. 2004; Yada et al. 2004). MBII recruits a chromatin-modifying complex containing TRRAP, GCN-5, and TIP60 (McMahon et al. 1998; Hann 2014). MBIII and MBIV contribute to MYC-specific phenotypes, such as MYC-induced apoptosis, but are not clearly relevant for MYC-induced cell proliferation and survival (Herbst et al. 2005; Cowling et al. 2006). Among the MYC boxes, structural data is only available for MBI. Arrowsmith and colleagues have reported a structural model for MBI binding to the SH3 domain of the BIN-1 tumor suppressor (Andresen et al. 2012). However, this interaction does not present a compelling therapeutic rationale, and the structure is limited to 13 residues of MYC (Fig. 1A).

Figure 1.

Figure 1.

Structural studies of MYC reveal challenges of developing direct-acting inhibitors of (A) MYC Box I (MBI as red ribbon and BIN-1 as white space-fill; PDB: 1MV0) and (B) the MYC:MAX heterodimer (MYC as red ribbon and MAX as white ribbon on white DNA; PDB: 1NKP).

The carboxyl terminus of MYC encodes a 100-residue basic helix-loop-helix-leucine-zipper (bHLH-LZ) DNA-binding domain. The leucine zipper forms a coiled-coil heterodimer with a homologous region on the transcriptional repressor MAX, which together engage E-box DNA-binding sites (see Conacci-Sorrell et al. 2014). Localization of the heterodimer to promoter and enhancer regions positively regulates transcription of proliferation-associated genes through control of transcription elongation (Eilers and Eisenman 2008; Meyer and Penn 2008; Rahl et al. 2010). Heterodimerization of MYC and MAX is required for transcriptional activation and oncogenic transformation, validating this interaction for further study. A seminal structural study by Nair and Burley (Nair and Burley 2003), however, elegantly displays the extended interaction between the two proteins that offers no apparent site for positioning a small-molecule inhibitor (Fig. 1B). The structurally and functionally similar MYCN and MYCL are comparably organized, posing then identical challenges for direct-acting therapeutic development in diseases attributable to these two oncogenes.

Intrinsically disordered proteins, such as MYC, comprise >30% of eukaryotic proteins (Wright and Dyson 1999; Metallo 2010; Andresen et al. 2012). Many such proteins are found at signaling nodes, where rapid physiologic response and dynamic cell state changes demand finely tuned posttranslational regulatory mechanisms that may be facilitated by intrinsic disorder (Gsponer and Babu 2009). Disordered proteins are often encoded by transcripts featuring short poly(A) tails, which reduce mRNA stability, and feature amino-terminal degrons and internal PEST sequences allowing for precise control of protein half-life (Gsponer et al. 2008). As discussed in detail in Farrell and Sears (2014), MYC has an ephemeral half-life (20–30 min), and is tightly regulated by E3 ubiquitin ligase recruitment and proteasomal degradation through at least two complexes (SKP-2 and FBW-7) and an evolutionarily conserved PEST domain (Herbst et al. 2004; Cowling and Cole 2006). Perhaps the intrinsic disorder of MYC and additional counter-regulatory measures (short protein and mRNA half-lives, transcriptional attenuation) are then ancient evolutionary safeguards to having such a dangerous oncogene in the genome.

Despite the apparent obstacles involved in studying the function of intrinsically disordered proteins, several research groups have organized around the challenge of developing first direct-acting inhibitors of MYC. The concept underlying these studies is theoretical in nature, arguing that the inherent, intrinsic disorder of native MYC (Nair and Burley 2003; Follis et al. 2008; Hammoudeh et al. 2009) establishes an opportunity for small-molecule-mediated induction of a neomorphic-binding site, a perhaps extreme example of induced fit. Induced fit implies a conformational change in the protein target provoked by a ligand, which may be accompanied by target inhibition or functional modulation as has been characterized extensively in enzymology and protein–nucleic acid interactions (Williamson 2000; Johnson 2008). The concept of induced fit applied to intrinsically disordered proteins possesses additional considerations. The lack of secondary structure in proteins such as MYC limits competing native intramolecular interactions, which are often low affinity, yet is complicated by the need for significant binding energy to overcome entropic penalties (Metallo 2010), and the likelihood of multiple binding states. This latter consideration presents challenges in establishing modes of molecular recognition, capturing low-energy assembly states for structural characterization, or even rigorously validating inhibitory biochemistry. As such, current described inhibitors of the MYC:MAX interaction serve as early, provocative examples of prototypical compounds, but, as a group, lack properties of target specificity biophysical characterization, and show utility in cell and model organism assays of MYC biology, which would define these compounds as chemical probes of MYC function and bona fide leads for therapeutic development (Frye 2010).

To our knowledge, the first efforts to inhibit MYC:MAX heterodimerization were reported by Vogt and colleagues in 2002 (Berg et al. 2002). Using a CFP/YFP fluorescence resonance energy transfer (FRET) assay, the bHLH-LZ domains of MYC and MAX were studied for inhibition of dimerization by a library of 7000 small molecules. These studies identified a series of related, substituted isoindolines (e.g., IIA4B20) (Fig. 2), which showed weak but reproducible inhibitory activity in FRET and ELISA assays (75–210 μm). Soon thereafter, Prochownik and colleagues (Yin et al. 2003), studied a commercial library of 10,000 compounds using a Gal-4-based yeast two-hybrid assay and the minimal bHLH-LZ interacting domains. Emerging from these studies were a series of structurally diverse compounds that impaired MYC:MAX binding by electrophoretic mobility shift assay (EMSA), including compounds that have been subsequently studied in cellular models of MYC biology (10074-A4, 10074-G5, and 10058-F4) (Fig. 2). A second study by Yang and coworkers reported curcuminoid inhibitors of MYC:MAX binding, in the context of a study aiming to establish binding constants for the heterodimeric interaction on DNA (Park et al. 2004). By measuring binding of MYC:MAX heterodimers to a fluorescein-labeled oligonucleotide, Berg and colleagues screened 17,298 compounds to discover pyrazolo-pyrimidine inhibitors with measurable biochemical inhibitory activity for MYC (Fig. 2) (e.g., Mycro1; IC50 = 30 μm) and a narrow window of selectivity for MAX:MAX homodimers (IC50 = 72 μm) (Kiessling et al. 2006, 2007). Additional efforts to target the MYC: MAX heterodimerization event have been undertaken subsequently, including efforts using further FRET screens (Xu et al. 2006), cellular transformation assays (Shi et al. 2009), and further EMSA screens (Jeong et al. 2010). These and other studies have led to a list of structurally diverse compounds with reported activity against the MYC:MAX:DNA-binding event (Fig. 2) (Yap et al. 2013).

Figure 2.

Figure 2.

Small-molecule disruptors of MYC:MAX heterodimerization.

As described above, this research comprises more than a decade of study to identify the first qualified chemical probes of MYC function, for mechanistic use in cellular biology. Medicinal chemistry efforts directed at improving the limited potency of these compounds are described in the published and patent literature (WO2010083404 A2). However, many of the active species belong to structural classes annotated as nonselective ligands in pharmaceutical screening libraries, so-called pan-assay interference compounds (PAINS) (Baell and Holloway 2010; Whitty 2011), establishing important first directions for follow-up chemistry. Perhaps consistent with this behavior, incisive and important studies have observed multiple binding events of bioactive compounds along the MYC protein by circular dichroism (Hammoudeh et al. 2009), nuclear magnetic resonance (NMR) (Hammoudeh et al. 2009), and most recently by mass spectrometry (Harvey et al. 2012). The relatively low potency, demonstrable selectivity, and durable biostability of lead compounds have prevented use in MYC model systems in vivo (Guo et al. 2009; Clausen et al. 2010), but insights are emerging from this work and structural modeling. Still needed are direct-acting MYC inhibitors with properties commensurate with a bona fide chemical probe, such as defined by Frye (2010). There exists, therefore, an opportunity to revisit this biochemistry with new screening methodologies, new types of expansive chemical libraries, and new measurements to characterize the mode of molecular recognition of candidate inhibitors.

Beyond the MYC:MAX heterodimerization interface, functional studies highlight MBII as a putative target for chemical probe development. The requirement for MBII in MYC-specific transactivation and cellular transformation highlights the emerging validation of this protein–protein interaction. Unfortunately, limited biochemical and no structural information is available for MBII. Focused biophysical and structural studies of MBII may pave the way to miniaturized assays capable of interrogating MBII with discovery chemistry.

INHIBITION OF MYC-DEPENDENT TRANSCRIPTIONAL SIGNALING

MYC-dependent transcription requires the assembly and function of critical transcriptional and chromatin-modifying enzyme complexes (Fig. 3) (Dang et al. 2006; Eilers and Eisenman 2008; also see Hann 2014). Communication of transcriptional impulses is initiated by promoter- and enhancer-bound MYC, which serves to organize histone acetyltransferases (such as TIP60, GCN5, and CBP/P300) and the mediator complex. These events promote the preparation and stabilization of accessible euchromatin, as well as bridging interactions with initiated RNA Pol II. Recently, the Young laboratory described a pivotal role of MYC also in genome-wide transcriptional elongation mediated by proximal promoter pause release (Rahl et al. 2010). MYC importantly functions to promote CDK-9-dependent phosphorylation of RNA Pol II, triggering the release of paused polymerase to execute definitive target gene transcription (see Levens 2013; Rahl and Young 2014; Sabò and Amati 2014).

Figure 3.

Figure 3.

Chromatin-dependent MYC transcriptional signaling. MYC and MAX bind at E-box motifs (boxes) in enhancers and promoters. Protein complex formation causes looping to engage promoters adjacent to MYC target genes (arrow to gene body), inducing RNA Pol II (RNA POL-2) loading to initiation site. Recruitment of TRRAP and histone acetyltransferases (HAT) leads to covalent modification of nucleosomes (clustered gray spheres) with lysine side-chain acetylation (Kac; small black circles). Acetylated, open chromatin is bound by bromodomain-containing proteins, including HATs (CBP, EP300, and GCN5) and BET-family coactivators (BET; yellow). BET bromodomains recruit the P-TEFb elongation complex (cyclin T and CDK-9; cyan), phosphorylates (small green circles) the carboxy-terminal domain of RNA POL-2, causing pause release and leading to elongation.

These insights support a model of targeting MYC via coactivator proteins critical to MYC-specific initiation and elongation. The pronounced effect of MYC on local chromatin acetylation (via HAT recruitment) (McMahon et al. 2000; Knoepfler et al. 2006), and the emerging role for MYC in elongation (via recruitment of CDK-9), prompted our consideration that the bromodomain and extraterminal domain (BET) family of coactivator proteins (BRD-2, BRD-3, BRD-4, and BRDT) figure prominently in MYC-mediated transcriptional pause release. BET bromodomains bind to polyacetylated histone tails via molecular recognition of ε-acetyl lysine (Kac) by an antiparallel bundle of four α-helices that forms a hydrophobic pocket with a conserved asparagine and an organized network of deep water molecules (Mujtaba et al. 2004, 2007; Filippakopoulos et al. 2012; Prinjha et al. 2012). Prior research established a role for BET bromodomains in cell-cycle progression and mitotic memory (Dey et al. 2003). Biochemical studies in virology by Verdin and coworkers importantly established that BET bromodomains possess a high-affinity binding motif for the CDK-9-containing positive transcription elongation factor complex (P-TEFb) at the distal carboxyl terminus (Bisgrove et al. 2007). We thus hypothesized that BET bromodomains may function to recruit CDK-9 to sites of hyperacetylated chromatin in cancer cells, thereby facilitating MYC-specific transcription elongation.

Having developed and characterized the first bromodomain inhibitor (Filippakopoulos et al. 2010), targeting the BET family of bromodomains, we performed a chemical genetic study of BET inhibition on MYC transactivation in vitro and in vivo (Delmore et al. 2011). Mechanistic studies performed in translational models of multiple myeloma (a MYC-dependent plasma cell malignancy) revealed that chemical displacement or genetic silencing of BET bromodomains results in the selective, coordinate down-regulation of the MYC transcriptional program. Concurrent, collaborative studies of BET inhibition in acute myeloid leukemia with Vakoc and Lowe comparably showed pronounced MYC pathway inhibition associated with marked down-regulation of transcription of the MYC gene itself (Zuber et al. 2011). In both studies, abrogation of chromatin-dependent MYC gene transcription via BET bromodomain inhibition elicited meaningful therapeutic responses in murine models of these aggressive hematological malignancies. Targeting MYC function via BET bromodomain inhibition has now been validated by studies in Burkitt lymphoma (Mertz et al. 2011), B- and T-cell acute lymphoblastic leukemia (Ott et al. 2012), non-small-cell lung carcinoma (Shimamura et al. 2013), and diffuse large B-cell lymphoma (Chapuy et al. 2013). In model systems of neuroblastoma, MYCN transcription and function was found to be comparably sensitive to BET inhibition. This hypothesis is presently being assessed in clinical trials of BET bromodomain inhibitors in MYC-dependent solid and hematopoietic tumors.

Another noteworthy approach to targeting MYC transcription derives from a unique and well-characterized feature of the MYC locus, namely, a predisposition for formation of G-quadruplex DNA (Siddiqui-Jain et al. 2002; Mathad et al. 2011). Small-molecule stabilizers of G-quadruplex DNA have been developed as tool compounds, which establish an impediment to RNA Pol II transcription of MYC. Further optimization is required before these compounds can be clinical deployed, but MYC-specific biological consequences have been observed with these compounds in experimental systems (Han et al. 1999a,b, 2000, 2001; Han and Hurley 2000; Hurley et al. 2006).

Beyond BET bromodomains, additional MYC-associated coactivator proteins may also then be considered for therapeutic development. As shown in Figure 4 (and as described in Conacci-Sorrell et al. 2014; Hann 2014) the MYC interactome features known critical mediators of transcriptional output, among them protein targets amenable to small-molecule discovery. Through MBII, MYC recruits the TRRAP complex that includes the histone acetyltransferases TIP60 and GCN5. The successful development of P300 histone acetyltransferase inhibitors by Cole and colleagues (Zheng et al. 2005; Bowers et al. 2010), suggests that discovery chemistry directed at these validated effectors of MYC-dependent chromatin remodeling and transcription is within reach. As P300 is a known binding partner of MYC via interactions in the transactivation domain (Faiola et al. 2005), the C646 inhibitor of P300 could contribute to the further understanding of this enzyme in MYC stability and function. Downstream from enhancer-bound factors such as these, MYC-mediated recruitment of mediator and TFIIH suggests consideration for the development of selective inhibitors of transcriptional kinases, such as CDK-7, CDK-8, and CDK-9, as MYC-directed tools and prototype therapeutics. Small-molecule inhibitors of transcriptional kinases are being intensively pursued in academia and industry, and a partial list of emerging compounds is presented in Table 1.

Figure 4.

Figure 4.

The MYC protein interactome (STRING).

Table 1.

Small molecules linked to MYC-pathway inhibition

Compound name Class Target References
Flavopiridol CDK inhibitor Cdk-9 Chen et al. 2005; Rahl et al. 2010
Purvalanol A CDK inhibitor Cdk-1 Goga et al. 2007
SU9516 CDK inhibitor Cdk-2, Cdk-9 Gao et al. 2006
PHA 767491 HCI CDK inhibitor Cdc-7 and Cdk-9 Montagnoli et al. 2008; Natoni et al. 2011
SNS-032 CDK inhibitor Cdk-2, Cdk-7, and Cdk-9 Walsby et al. 2011
JQ1 BET bromodomain inhibitor Brd-4, Brd-3, Brd-2 Filippakopoulos et al. 2010; Delmore et al. 2011
SGI-1776 PIM kinase inhibitor Pim-1 Zippo et al. 2007, 2009
EPZ004777 Dot1 L inhibitor Dot1 L Daigle et al. 2011
C464 p300/CBP ACTfrase inhibitor p300 McMahon et al. 1998
SAHA HDAC inhibitor
Triptolide TFIIH/XPB XPB Titov et al. 2011
Nutlin-3a p53-MDM2 inhibitor p53-MDM-2 Felsher et al. 2000
SB220025 MAPK inhibitor p38 Zhu et al. 2008
LY294002 PI3K inhibitor PI3K Zhu et al. 2008; Liu et al. 2011; Muellner et al. 2011
VAV-939 Wnt inhibitor Tankyrase 1,2 He et al. 1998
LY-411575 γ-Secratase inhibitor Notch 1 Moellering et al. 2009
VX-680 Aurora kinase inhibitor Aurora kinases Yang et al. 2011
NSC71948 BAG1 inhibitor BAG-1 Zhang et al. 2011
ABT263 BCL2 inhibitor BCL-2 Zhang et al. 2011
VER-155008 HSP70 inhibitor Hsp70/Hsc70 Zhang et al. 2011
KW-2478 HSP90 inhibitor Hsp90 Nakashima et al. 2010
SB 218078 Chk1 inhibitor Hck-1 Jiang et al. 2011
Leflunomide DHODH inhibitor DHODH White et al. 2011

Insights from developmental and disease biology suggest additional opportunities to target MYC function at the level of nuclear chromatin. Functioning alongside MYC-dependent transcription are parallel transcriptional networks that may be approached for synthetic lethality. Eisenman and colleagues discovered and characterized a functional interaction between Drosophila Myc and the Trithorax group protein Little imaginal discs (Lid) (Secombe et al. 2007), a homolog of the human proto-oncogene JARID1. They hypothesize a role for JARID1 in MYC-dependent neoplasia (Secombe and Eisenman 2007; de Rooij et al. 2013), supported by recent evidence that the NUP98-JARID1A oncogene in acute myeloid leukemia (AML) functions, in part, through an essential interaction between a plant homeodomain (PHD) finger encoded by JARID1A and histone 3 lysine 4, providing functional validation of JARID1A in a MYC-dependent tumor (Wang et al. 2009; Li et al. 2010).

Beyond chromatin-dependent MYC transcriptional signaling, other chromatin-associated factors such as the PIM-1 kinase may contribute to high global transcriptional output at MYC target genes (Breuer et al. 1989; van Lohuizen et al. 1989). PIM-1 phosphorylates histone H3 and, interestingly and similar to MYC, PIM-1 regulates transcriptional elongation (Zippo et al. 2007, 2009). PIM-1 inhibitors are in development as antitumor agents, including the ADP-competitive inhibitor SGI-1776. Additional small molecules targeting MYC-associated transcriptional regulators such as HDAC1, HDAC2 (Peart et al. 2005; Frumm et al. 2013), TFIIH (Titov et al. 2011), P53-MDM2 (Felsher et al. 2000), and DOT1L (Daigle et al. 2011) are referenced in Table 1.

TARGETING PATHWAYS UPSTREAM OF MYC

As an end-effector of numerous upstream signal transduction pathways, it is highly likely that effective targeting of dominant growth factor–independent signaling networks will mute upstream activation of MYC. Often, mitogenic signaling pathways activate intermediary effector transcription factors that integrate upstream surface-to-nuclear signaling with the common transcriptional language of MYC-dependent cell proliferation. A classic example is KRAS activation, which is among the most prevalent alterations in human cancer. KRAS activation in cancer drives MYC activation. Both genetic (Omomyc) (Soucek et al. 2013) and chemical (BET inhibition with JQ1) (Shimamura et al. 2013) perturbation of MYC function arrest proliferation downstream from KRAS and validate MYC as a target in KRAS-dependent tumors.

Indeed, many other upstream signaling pathways lead to deregulated MYC function, including the MAPK, PI3K, Wnt/β-catenin, and Notch pathways (see Eilers and Eisenman 2008; Meyer and Penn 2008; Conacci-Sorrell et al. 2014). The Wnt/β-catenin pathway regulates MYC expression through signaling to the Tcf-4 transcription factor that binds tissue-specific enhancers upstream of the MYC gene (Wright et al. 2010). Stabilization of NOTCH1 by juxtamembrane or PEST domain alteration in T-ALL leads to deregulation of MYC expression and function. Targeting the Notch pathway with upstream inhibition of γ-secretase (Weng et al. 2006), or direct inhibition using stabilized α-helical peptides (Moellering et al. 2009), leads to prompt down-regulation of MYC associated with antiproliferative consequences. Exemplary, additional inhibitors of upstream signal transduction pathways, such as MAPK and PI3K are presented in Table 1. As targeted agents are brought forward for clinical trials, preclinical studies assessing combinations of upstream and downstream modulators of MYC function should be pursued.

MODULATING MYC STABILITY

MYC has a naturally short half-life (Andresen et al. 2012). Regulation and preservation of the short transcript and protein half-life is essential to long-term survival without cancer. MYC protein homeostasis is considered in detail in Farrell and Sears (2014) and elsewhere by Sears and colleagues (2000). Here, we describe only briefly the opportunity to target MYC protein stability using therapeutic approaches. The transactivating domain is subject to phosphorylation, acetylation, and ubiquitylation, which serve to regulate stability and binding interactions (Cowling and Cole 2006; Andresen et al. 2012). Alterations in MYC and associated protein homeostatic pathway effectors are observed in human tumors, leading to the inappropriate posttranslational processing and stability of the MYC oncoprotein.

The facile development of kinase inhibitors warrants consideration of approaches to target MYC degradation, as MYC degradation is heavily influenced by serine and threonine phosphorylation. Biochemical and functional studies have established that phosphorylation of serine 62 (pSer62) by ERK pathway kinase activity stabilizes MYC and promotes proliferation. In contrast, phosphorylation of threonine 58 (pThr58) by GSK-3β destabilizes MYC. Notably, mutations of Thr58 are observed in Burkitt lymphoma resulting in constitutive MYC stabilization. Thus, therapeutic approaches to MEK-ERK pathway inhibition, or efforts to inhibit GSK-3β such as inhibition of PI3K-AKT signaling, may favorably modulate MYC stability for therapeutic benefit. This may be particularly true in tumors with up-regulated MEK-ERK signaling (KRAS or EGFR activated) or phosphatase and tensin (PTEN) loss. Table 1 lists exemplary compounds, which may be used as chemical probes of pathway function.

Opposing MYC phosphorylation at serine 62 is protein phosphatase 2 (PP2A), which functions as a heterotrimeric enzyme complex to indirectly and directly modulate MYC protein stability. The Sears group has shown that pSer62 is a direct target of PP2A, supporting a model in which PP2A loss (a common tumor suppressor loss in cancer) promotes MYC stability. Presently, pharmaceutical compounds targeting PP2A do not exist. However, in collaborative research with the laboratories of Thomas Look, Jon Aster, and Hanno Steen, we recently discovered PP2A as the antiproliferative target of perphenazine (and other phenothiazines), by ligand-affinity chromatography (Gutierrez et al. 2014). As small molecules targeting and activating tumor suppressors are rarely described, we are interested now to optimize these compounds for enhanced PP2A binding and activation, to destabilize MYC and modulate other critical PP2A target proteins. Recently, Eilers and colleagues reported a novel approach to destabilize MYCN via inhibition of Aurora A kinases (Brockmann et al. 2013). MYCN is targeted for degradation by the FBXW7 ubiquitin ligase. MYCN protein stability is promoted by an interaction with the Aurora A mitotic kinase. Although the kinase activity of AURKA is not required for MYCN protein stability, two ATP-competitive azepine inhibitors of AURKA (MLN-80545 and MLN-8237) disrupted binding to MYCN. In mechanistic and translational experiments performed in MYCN-dependent neuroblastoma models, AURKA inhibition prompted MYCN degradation and an antiproliferative effect.

A final opportunity for therapeutic targeting of MYC protein stability is presented by MYC-associated histone acetyltransferases. Beyond their established role in promoting chromatin remodeling, bromodomain recruitment, and gene expression, acetylation of MYC by CBP/P300, GCN5, and TIP60 promotes MYC stability and transcriptional activity (Cowling and Cole 2006). As above, inhibition of these acetyltransferase enzymes may display favorable activity in cancer model systems, in support of further clinical development.

MYC AND SYNTHETIC LETHALITY

MYC-addicted cancer cells may harbor unique dependencies that a normal cell may not require. Such synthetic lethal interactions with MYC may prompt the development of context-specific inhibitors. Two kinase targets have been identified that are required for cell survival in the context of enforced MYC expression: Aurora kinase B (Yang et al. 2010) and Cdk-1 (Goga et al. 2007). Inhibition of these kinases with cell-permeable small molecules has shown compelling evidence of a leveraged antiprolifeartive effect in MYC-dependent cancer models, albeit with a narrow therapeutic index. Emerging research approaches using highly parallel reverse genetic screens (RNA interference; RNAi), has reinvigorated efforts to identify MYC-specific tumor dependencies. Recently, two laboratories performed RNAi screening in a MYC-dependent context leading to the suggestion of CSNK1e kinase (Toyoshima et al. 2012) and the SAE1/2 SUMO-activating enzyme (Kessler et al. 2012) as putative targets for the development of synthetic lethal small molecules. Rational approaches to synthetic lethality have included consideration of metabolic adaptation to MYC-mediated tumorigenesis. Observing therapy-associated cellular senescence in treated Eµ-MYC cells, Schmitt and colleagues targeted glucose uptake and autophagy with impressive antiproliferative results (Dorr et al. 2013). Together, these studies establish a new path to MYC-specific therapeutic development, revealed by powerful new technologies and a detailed mechanistic understanding of MYC biology at the interface of transcription, translation, and metabolism.

CONCLUDING REMARKS

The centrality of MYC to the pathogenesis of cancer, illustrated in granular detail in the accompanying cited literature, demands the creativity and commitment of the allied fields of ligand discovery. In this review, we provide a rationale for targeting MYC that balances high-risk, high-reward approaches of direct MYC:MAX modulation with near-term opportunities to approach MYC transcriptional signaling via chromatin-dependent processes and pathways of protein homeostasis. Still, direct inhibition of MYC remains an elusive goal in cancer medicine—an inadequacy of current therapeutic science perhaps at the heart of our inability to eradicate this disease.

ACKNOWLEDGMENTS

The authors thank Jason J. Marineau, Peter B. Rahl, Richard A. Young, Chi Dang, and Robert N. Eisenman for influential discussions regarding therapeutic strategies toward inhibiting MYC. In addition, we thank Jaime M. Reyes for help with graphic art design in Figure 3.

REFERENCES

*Reference is also in this collection.

  1. Ahuja P, Zhao P, Angelis E, Ruan H, Korge P, Olson A, Wang Y, Jin ES, Jeffrey FM, Portman M, et al. 2010. Myc controls transcriptional regulation of cardiac metabolism and mitochondrial biogenesis in response to pathological stress in mice. J Clin Invest 120: 1494–1505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Andresen C, Helander S, Lemak A, Fares C, Csizmok V, Carlsson J, Penn LZ, Forman-Kay JD, Arrowsmith CH, Lundstrom P, et al. 2012. Transient structure and dynamics in the disordered c-Myc transactivation domain affect Bin1 binding. Nucleic Acids Res 40: 6353–6366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baell JB, Holloway GA. 2010. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J Med Chem 53: 2719–2740 [DOI] [PubMed] [Google Scholar]
  4. Berg T. 2008. Inhibition of transcription factors with small organic molecules. Curr Opin Chem Biol 12: 464–471 [DOI] [PubMed] [Google Scholar]
  5. Berg T, Cohen SB, Desharnais J, Sonderegger C, Maslyar DJ, Goldberg J, Boger DL, Vogt PK. 2002. Small-molecule antagonists of Myc/Max dimerization inhibit Myc-induced transformation of chicken embryo fibroblasts. Proc Natl Acad Sci 99: 3830–3835 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, et al. 2010. The landscape of somatic copy-number alteration across human cancers. Nature 463: 899–905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bettess MD, Dubois N, Murphy MJ, Dubey C, Roger C, Robine S, Trumpp A. 2005. c-Myc is required for the formation of intestinal crypts but dispensable for homeostasis of the adult intestinal epithelium. Mol Cell Biol 25: 7868–7878 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bisgrove DA, Mahmoudi T, Henklein P, Verdin E. 2007. Conserved P-TEFb-interacting domain of BRD4 inhibits HIV transcription. Proc Natl Acad Sci 104: 13690–13695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bowers EM, Yan G, Mukherjee C, Orry A, Wang L, Holbert MA, Crump NT, Hazzalin CA, Liszczak G, Yuan H, et al. 2010. Virtual ligand screening of the p300/CBP histone acetyltransferase: Identification of a selective small molecule inhibitor. Chem Biol 17: 471–482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Breuer M, Slebos R, Verbeek S, van Lohuizen M, Wientjens E, Berns A. 1989. Very high frequency of lymphoma induction by a chemical carcinogen in pim-1 transgenic mice. Nature 340: 61–63 [DOI] [PubMed] [Google Scholar]
  11. Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, et al. 2013. Small molecule inhibitors of aurora-A induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell 24: 75–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chapuy B, McKeown MR, Lin CY, Monti S, Roemer MG, Qi J, Rahl PB, Sun HH, Yeda KT, Doench JG, et al. 2013. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell 24: 777–790 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Clausen DM, Guo J, Parise RA, Beumer JH, Egorin MJ, Lazo JS, Prochownik EV, Eiseman JL. 2010. In vitro cytotoxicity and in vivo efficacy, pharmacokinetics, and metabolism of 10074-G5, a novel small-molecule inhibitor of c-Myc/Max dimerization. J Pharmacol Exp Ther 335: 715–727 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. *.Conacci-Sorrell M, McFerrin L, Eisenman RN. 2014. An overview of MYC and its interactome. Cold Spring Harb Perspect Med 4: a014357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cowling VH, Cole MD. 2006. Mechanism of transcriptional activation by the Myc oncoproteins. Semin Cancer Biol 16: 242–252 [DOI] [PubMed] [Google Scholar]
  16. Cowling VH, Chandriani S, Whitfield ML, Cole MD. 2006. A conserved Myc protein domain, MBIV, regulates DNA binding, apoptosis, transformation, and G2 arrest. Mol Cell Biol 26: 4226–4239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Daigle SR, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J, Johnston LD, Scott MP, Smith JJ, Xiao Y, et al. 2011. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20: 53–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Dalla-Favera R, Bregni M, Erikson J, Patterson D, Gallo RC, Croce CM. 1982. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc Natl Acad Sci 79: 7824–7827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Dang CV, O’Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F. 2006. The c-Myc target gene network. Semin Cancer Biol 16: 253–264 [DOI] [PubMed] [Google Scholar]
  20. Darnell JE Jr. 2002. Transcription factors as targets for cancer therapy. Nat Rev Cancer 2: 740–749 [DOI] [PubMed] [Google Scholar]
  21. Davis AC, Wims M, Spotts GD, Hann SR, Bradley A. 1993. A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice. Genes Dev 7: 671–682 [DOI] [PubMed] [Google Scholar]
  22. 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. de Rooij JD, Hollink IH, Arentsen-Peters ST, van Galen JF, Berna Beverloo H, Baruchel A, Trka J, Reinhardt D, Sonneveld E, Zimmermann M, et al. 2013. NUP98/JARID1A is a novel recurrent abnormality in pediatric acute megakaryoblastic leukemia with a distinct HOX gene expression pattern. Leukemia 27: 2280–2288 [DOI] [PubMed] [Google Scholar]
  24. Dey A, Chitsaz F, Abbasi A, Misteli T, Ozato K. 2003. The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis. Proc Natl Acad Sci 100: 8758–8763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Dorr JR, Yu Y, Milanovic M, Beuster G, Zasada C, Dabritz JH, Lisec J, Lenze D, Gerhardt A, Schleicher K, et al. 2013. Synthetic lethal metabolic targeting of cellular senescence in cancer therapy. Nature 501: 421–425 [DOI] [PubMed] [Google Scholar]
  26. Duesberg PH, Vogt PK. 1979. Avian acute leukemia viruses MC29 and MH2 share specific RNA sequences: Evidence for a second class of transforming genes. Proc Natl Acad Sci 76: 1633–1637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Easton DF, Pooley KA, Dunning AM, Pharoah PD, Thompson D, Ballinger DG, Struewing JP, Morrison J, Field H, Luben R, et al. 2007. Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447: 1087–1093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Eilers M, Eisenman RN. 2008. Myc’s broad reach. Genes Dev 22: 2755–2766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Faiola F, Liu X, Lo S, Pan S, Zhang K, Lymar E, Farina A, Martinez E. 2005. Dual regulation of c-Myc by p300 via acetylation-dependent control of Myc protein turnover and coactivation of Myc-induced transcription. Mol Cell Biol 25: 10220–10234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. *.Farrell AS, Sears RC. 2014. MYC degradation. Cold Spring Harb Perspect Med 4: a014365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Felsher DW, Bishop JM. 1999. Reversible tumorigenesis by MYC in hematopoietic lineages. Mol Cell 4: 199–207 [DOI] [PubMed] [Google Scholar]
  32. Felsher DW, Zetterberg A, Zhu J, Tlsty T, Bishop JM. 2000. Overexpression of MYC causes p53-dependent G2 arrest of normal fibroblasts. Proc Natl Acad Sci 97: 10544–10548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Filippakopoulos P, Picaud S, Mangos M, Keates T, Lambert JP, Barsyte-Lovejoy D, Felletar I, Volkmer R, Muller S, Pawson T, et al. 2012. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 149: 214–231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Fletcher S, Hamilton AD. 2007. Protein–protein interaction inhibitors: Small molecules from screening techniques. Curr Top Med Chem 7: 922–927 [DOI] [PubMed] [Google Scholar]
  36. Flores I, Murphy DJ, Swigart LB, Knies U, Evan GI. 2004. Defining the temporal requirements for Myc in the progression and maintenance of skin neoplasia. Oncogene 23: 5923–5930 [DOI] [PubMed] [Google Scholar]
  37. Follis AV, Hammoudeh DI, Wang H, Prochownik EV, Metallo SJ. 2008. Structural rationale for the coupled binding and unfolding of the c-Myc oncoprotein by small molecules. Chem Biol 15: 1149–1155 [DOI] [PubMed] [Google Scholar]
  38. Frumm SM, Fan ZP, Ross KN, Duvall JR, Gupta S, VerPlank L, Suh BC, Holson E, Wagner FF, Smith WB, et al. 2013. Selective HDAC1/HDAC2 inhibitors induce neuroblastoma differentiation. Chem Biol 20: 713–725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Frye SV. 2010. The art of the chemical probe. Nat Chem Biol 6: 159–161 [DOI] [PubMed] [Google Scholar]
  40. *.Gabay M, Li Y, Felsher DW. 2014. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect Med 10.1101/cshperspect.a014241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Gamberi G, Benassi MS, Bohling T, Ragazzini P, Molendini L, Sollazzo MR, Pompetti F, Merli M, Magagnoli G, Balladelli A, et al. 1998. c-myc and c-fos in human osteosarcoma: Prognostic value of mRNA and protein expression. Oncology 55: 556–563 [DOI] [PubMed] [Google Scholar]
  42. Goga A, Yang D, Tward AD, Morgan DO, Bishop JM. 2007. Inhibition of CDK1 as a potential therapy for tumors over-expressing MYC. Nat Med 13: 820–827 [DOI] [PubMed] [Google Scholar]
  43. Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, et al. 2007. Patterns of somatic mutation in human cancer genomes. Nature 446: 153–158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Gsponer J, Babu MM. 2009. The rules of disorder or why disorder rules. Prog Biophys Mol Biol 99: 94–103 [DOI] [PubMed] [Google Scholar]
  45. Gsponer J, Futschik ME, Teichmann SA, Babu MM. 2008. Tight regulation of unstructured proteins: From transcript synthesis to protein degradation. Science 322: 1365–1368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Guo J, Parise RA, Joseph E, Egorin MJ, Lazo JS, Prochownik EV, Eiseman JL. 2009. Efficacy, pharmacokinetics, tissue distribution, and metabolism of the Myc-Max disruptor, 10058-F4 [Z,E]-5-[4-ethylbenzylidine]-2-thioxothiazolidin-4-one, in mice. Cancer Chemother Pharmacol 63: 615–625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gutierrez A, Pan L, Groen RW, Baleydier F, Kentsis A, Marineau J, Grebliunaite R, Kozakewich E, Reed C, Pflumio F, et al. 2014. Phenothiazines induce PP2A-mediated apoptosis in T cell acute lymphoblastic leukemia. J Clin Invest 124: 644–655 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Hammoudeh DI, Follis AV, Prochownik EV, Metallo SJ. 2009. Multiple independent binding sites for small-molecule inhibitors on the oncoprotein c-Myc. J Am Chem Soc 131: 7390–7401 [DOI] [PubMed] [Google Scholar]
  49. Han H, Hurley LH. 2000. G-quadruplex DNA: A potential target for anti-cancer drug design. Trends Pharmacol Sci 21: 136–142 [DOI] [PubMed] [Google Scholar]
  50. Han H, Cliff CL, Hurley LH. 1999a. Accelerated assembly of G-quadruplex structures by a small molecule. Biochemistry 38: 6981–6986 [DOI] [PubMed] [Google Scholar]
  51. Han H, Hurley LH, Salazar M. 1999b. A DNA polymerase stop assay for G-quadruplex-interactive compounds. Nucleic Acids Res 27: 537–542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Han H, Bennett RJ, Hurley LH. 2000. Inhibition of unwinding of G-quadruplex structures by Sgs1 helicase in the presence of N,N′-bis[2-(1-piperidino)ethyl]-3,4,9,10-perylenetetracarboxylic diimide, a G-quadruplex-interactive ligand. Biochemistry 39: 9311–9316 [DOI] [PubMed] [Google Scholar]
  53. Han H, Langley DR, Rangan A, Hurley LH. 2001. Selective interactions of cationic porphyrins with G-quadruplex structures. J Am Chem Soc 123: 8902–8913 [DOI] [PubMed] [Google Scholar]
  54. *.Hann SR. 2014. MYC cofactors: Molecular switches controlling diverse biological outcomes. Cold Spring Harb Perspect Med 10.1101/cshperspect.a014399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Harvey SR, Porrini M, Stachl C, MacMillan D, Zinzalla G, Barran PE. 2012. Small-molecule inhibition of c-MYC: MAX leucine zipper formation is revealed by ion mobility mass spectrometry. J Am Chem Soc 134: 19384–19392 [DOI] [PubMed] [Google Scholar]
  56. Herbst A, Salghetti SE, Kim SY, Tansey WP. 2004. Multiple cell-type-specific elements regulate Myc protein stability. Oncogene 23: 3863–3871 [DOI] [PubMed] [Google Scholar]
  57. Herbst A, Hemann MT, Tworkowski KA, Salghetti SE, Lowe SW, Tansey WP. 2005. A conserved element in Myc that negatively regulates its proapoptotic activity. EMBO Rep 6: 177–183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Herrmann H, Blatt K, Shi J, Gleixner KV, Cerny-Reiterer S, Mullauer L, Vakoc CR, Sperr WR, Horny HP, Bradner JE, et al. 2012. Small-molecule inhibition of BRD4 as a new potent approach to eliminate leukemic stem- and progenitor cells in acute myeloid leukemia AML. Oncotarget 3: 1588–1599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hu SS, Vogt PK. 1979. Avian oncovirus MH2 is defective in Gag, Pol, and Env. Virology 92: 278–284 [DOI] [PubMed] [Google Scholar]
  60. *.Huang M, Weiss WA. 2013. Neuroblastoma and MYNC. Cold Spring Harb Perspect Med 3: a014415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hurley LH, Von Hoff DD, Siddiqui-Jain A, Yang D. 2006. Drug targeting of the c-MYC promoter to repress gene expression via a G-quadruplex silencer element. Semin Oncol 33: 498–512 [DOI] [PubMed] [Google Scholar]
  62. *.Hurlin PJ. 2013. Control of vertebrate development by MYC. Cold Spring Harb Perspect Med 3: a014332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Jain M, Arvanitis C, Chu K, Dewey W, Leonhardt E, Trinh M, Sundberg CD, Bishop JM, Felsher DW. 2002. Sustained loss of a neoplastic phenotype by brief inactivation of MYC. Science 297: 102–104 [DOI] [PubMed] [Google Scholar]
  64. Jeong KC, Ahn KO, Yang CH. 2010. Small-molecule inhibitors of c-Myc transcriptional factor suppress proliferation and induce apoptosis of promyelocytic leukemia cell via cell cycle arrest. Mol Biosyst 6: 1503–1509 [DOI] [PubMed] [Google Scholar]
  65. Johnson KA. 2008. Role of induced fit in enzyme specificity: A molecular forward/reverse switch. J Biol Chem 283: 26297–26301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kessler JD, Kahle KT, Sun T, Meerbrey KL, Schlabach MR, Schmitt EM, Skinner SO, Xu Q, Li MZ, Hartman ZC, et al. 2012. A SUMOylation-dependent transcriptional subprogram is required for Myc-driven tumorigenesis. Science 335: 348–353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kiemeney LA, Thorlacius S, Sulem P, Geller F, Aben KK, Stacey SN, Gudmundsson J, Jakobsdottir M, Bergthorsson JT, Sigurdsson A, et al. 2008. Sequence variant on 8q24 confers susceptibility to urinary bladder cancer. Nat Genet 40: 1307–1312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kiessling A, Sperl B, Hollis A, Eick D, Berg T. 2006. Selective inhibition of c-Myc/Max dimerization and DNA binding by small molecules. Chem Biol 13: 745–751 [DOI] [PubMed] [Google Scholar]
  69. Kiessling A, Wiesinger R, Sperl B, Berg T. 2007. Selective inhibition of c-Myc/Max dimerization by a pyrazolo [1,5-a]pyrimidine. ChemMedChem 2: 627–630 [DOI] [PubMed] [Google Scholar]
  70. Klein G. 1983. Specific chromosomal translocations and the genesis of B-cell-derived tumors in mice and men. Cell 32: 311–315 [DOI] [PubMed] [Google Scholar]
  71. Knoepfler PS, Zhang XY, Cheng PF, Gafken PR, McMahon SB, Eisenman RN. 2006. Myc influences global chromatin structure. EMBO J 25: 2723–2734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Leder P, Battey J, Lenoir G, Moulding C, Murphy W, Potter H, Stewart T, Taub R. 1983. Translocations among antibody genes in human cancer. Science 222: 765–771 [DOI] [PubMed] [Google Scholar]
  73. *.Levens D. 2013. Cellular MYCro economics: Balancing MYC function with MYC expression. Cold Spring Harb Perspect Med 3: a014233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Li L, Greer C, Eisenman RN, Secombe J. 2010. Essential functions of the histone demethylase lid. PLoS Genet 6: e1001221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. London N, Raveh B, Schueler-Furman O. 2013. Druggable protein–protein interactions—From hot spots to hot segments. Curr Opin Chem Biol 17: 952–959 [DOI] [PubMed] [Google Scholar]
  76. Mathad RI, Hatzakis E, Dai J, Yang D. 2011. c-MYC promoter G-quadruplex formed at the 5′-end of NHE III1 element: Insights into biological relevance and parallel-stranded G-quadruplex stability. Nucleic Acids Res 39: 9023–9033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. McMahon SB, Van Buskirk HA, Dugan KA, Copeland TD, Cole MD. 1998. The novel ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F oncoproteins. Cell 94: 363–374 [DOI] [PubMed] [Google Scholar]
  78. McMahon SB, Wood MA, Cole MD. 2000. The essential cofactor TRRAP recruits the histone acetyltransferase hGCN5 to c-Myc. Mol Cell Biol 20: 556–562 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S, Mele DA, Bergeron L, Sims RJ III. 2011. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci 108: 16669–16674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Metallo SJ. 2010. Intrinsically disordered proteins are potential drug targets. Curr Opin Chem Biol 14: 481–488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Meyer N, Penn LZ. 2008. Reflecting on 25 years with MYC. Nat Rev Cancer 8: 976–990 [DOI] [PubMed] [Google Scholar]
  82. Moellering RE, Cornejo M, Davis TN, Del Bianco C, Aster JC, Blacklow SC, Kung AL, Gilliland DG, Verdine GL, Bradner JE. 2009. Direct inhibition of the NOTCH transcription factor complex. Nature 462: 182–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Mujtaba S, He Y, Zeng L, Yan S, Plotnikova O, Sachchidanand, Sanchez R, Zeleznik-Le NJ, Ronai Z, Zhou MM. 2004. Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol Cell 13: 251–263 [DOI] [PubMed] [Google Scholar]
  84. Mujtaba S, Zeng L, Zhou MM. 2007. Structure and acetyl-lysine recognition of the bromodomain. Oncogene 26: 5521–5527 [DOI] [PubMed] [Google Scholar]
  85. Nair SK, Burley SK. 2003. X-ray structures of Myc-Max and Mad-Max recognizing DNA. Molecular bases of regulation by proto-oncogenic transcription factors. Cell 112: 193–205 [DOI] [PubMed] [Google Scholar]
  86. Nesbit CE, Tersak JM, Prochownik EV. 1999. MYC oncogenes and human neoplastic disease. Oncogene 18: 3004–3016 [DOI] [PubMed] [Google Scholar]
  87. Ott CJ, Kopp N, Bird L, Paranal RM, Qi J, Bowman T, Rodig SJ, Kung AL, Bradner JE, Weinstock DM. 2012. BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood 120: 2843–2852 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Pap T, Nawrath M, Heinrich J, Bosse M, Baier A, Hummel KM, Petrow P, Kuchen S, Michel BA, Gay RE, et al. 2004. Cooperation of Ras- and c-Myc-dependent pathways in regulating the growth and invasiveness of synovial fibroblasts in rheumatoid arthritis. Arthritis Rheum 50: 2794–2802 [DOI] [PubMed] [Google Scholar]
  89. Park S, Chung S, Kim KM, Jung KC, Park C, Hahm ER, Yang CH. 2004. Determination of binding constant of transcription factor myc-max/max-max and E-box DNA: The effect of inhibitors on the binding. Biochim Biophys Acta 1670: 217–228 [DOI] [PubMed] [Google Scholar]
  90. Peart MJ, Smyth GK, van Laar RK, Bowtell DD, Richon VM, Marks PA, Holloway AJ, Johnstone RW. 2005. Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. Proc Natl Acad Sci 102: 3697–3702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Pelengaris S, Littlewood T, Khan M, Elia G, Evan G. 1999. Reversible activation of c-Myc in skin: Induction of a complex neoplastic phenotype by a single oncogenic lesion. Mol Cell 3: 565–577 [DOI] [PubMed] [Google Scholar]
  92. Pelengaris S, Khan M, Evan GI. 2002. Suppression of Myc-induced apoptosis in β cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell 109: 321–334 [DOI] [PubMed] [Google Scholar]
  93. Pomerantz MM, Ahmadiyeh N, Jia L, Herman P, Verzi MP, Doddapaneni H, Beckwith CA, Chan JA, Hills A, Davis M, et al. 2009. The 8q24 cancer risk variant rs6983267 shows long-range interaction with MYC in colorectal cancer. Nat Genet 41: 882–884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Prinjha RK, Witherington J, Lee K. 2012. Place your BETs: The therapeutic potential of bromodomains. Trends Pharmacol Sci 33: 146–153 [DOI] [PubMed] [Google Scholar]
  95. Ptashne M, Gann A. 1997. Transcriptional activation by recruitment. Nature 386: 569–577 [DOI] [PubMed] [Google Scholar]
  96. Ptashne M, Gann A. 2002. Genes & signals. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY [Google Scholar]
  97. *.Rahl PB, Young RA. 2014. MYC and transcription elongation. Cold Spring Harb Perspect Med 4: a020990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Rahl PB, Lin CY, Seila AC, Flynn RA, McCuine S, Burge CB, Sharp PA, Young RA. 2010. c-Myc regulates transcriptional pause release. Cell 141: 432–445 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Rask-Andersen M, Almen MS, Schioth HB. 2011. Trends in the exploitation of novel drug targets. Nat Rev Drug Discov 10: 579–590 [DOI] [PubMed] [Google Scholar]
  100. *.Roussel MF, Robinson GW. 2013. Role of MYC in medulloblastoma. Cold Spring Harb Perspect Med 3: a014308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. *.Sabò A, Amati B. 2014. Genome recognition by MYC. Cold Spring Harb Perspect Med 4: a014191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Sauter G, Carroll P, Moch H, Kallioniemi A, Kerschmann R, Narayan P, Mihatsch MJ, Waldman FM. 1995. c-Myc copy number gains in bladder cancer detected by fluorescence in situ hybridization. Am J Pathol 146: 1131–1139 [PMC free article] [PubMed] [Google Scholar]
  103. Savino M, Annibali D, Carucci N, Favuzzi E, Cole MD, Evan GI, Soucek L, Nasi S. 2011. The action mechanism of the Myc inhibitor termed Omomyc may give clues on how to target Myc for cancer therapy. PLoS ONE 6: e22284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. *.Schmitz R, Ceribelli M, Pittaluga S, Wright G, Staudt LM. 2014. Oncogenic mechanisms in Burkitt lymphoma. Cold Spring Harb Perspect Med 4: a014282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. 2000. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev 14: 2501–2514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Secombe J, Eisenman RN. 2007. The function and regulation of the JARID1 family of histone H3 lysine 4 demethylases: The Myc connection. Cell Cycle 6: 1324–1328 [DOI] [PubMed] [Google Scholar]
  107. Secombe J, Li L, Carlos L, Eisenman RN. 2007. The Trithorax group protein Lid is a trimethyl histone H3K4 demethylase required for dMyc-induced cell growth. Genes Dev 21: 537–551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Sheiness D, Bishop JM. 1979. DNA and RNA from uninfected vertebrate cells contain nucleotide sequences related to the putative transforming gene of avian myelocytomatosis virus. J Virol 31: 514–521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Shi J, Stover JS, Whitby LR, Vogt PK, Boger DL. 2009. Small molecule inhibitors of Myc/Max dimerization and Myc-induced cell transformation. Bioorg Med Chem Lett 19: 6038–6041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Shimamura T, Chen Z, Soucheray M, Carretero J, Kikuchi E, Tchaicha JH, Gao Y, Cheng KA, Cohoon TJ, Qi J, et al. 2013. Efficacy of BET bromodomain inhibition in Kras-mutant non-small cell lung cancer. Clin Cancer Res 19: 6183–6192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Siddiqui-Jain A, Grand CL, Bearss DJ, Hurley LH. 2002. Direct evidence for a G-quadruplex in a promoter region and its targeting with a small molecule to repress c-MYC transcription. Proc Natl Acad Sci 99: 11593–11598 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Soucek L, Helmer-Citterich M, Sacco A, Jucker R, Cesareni G, Nasi S. 1998. Design and properties of a Myc derivative that efficiently homodimerizes. Oncogene 17: 2463–2472 [DOI] [PubMed] [Google Scholar]
  113. Soucek L, Jucker R, Panacchia L, Ricordy R, Tato F, Nasi S. 2002. Omomyc, a potential Myc dominant negative, enhances Myc-induced apoptosis. Cancer Res 62: 3507–3510 [PubMed] [Google Scholar]
  114. Soucek L, Whitfield J, Martins CP, Finch AJ, Murphy DJ, Sodir NM, Karnezis AN, Swigart LB, Nasi S, Evan GI. 2008. Modelling Myc inhibition as a cancer therapy. Nature 455: 679–683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Soucek L, Whitfield JR, Sodir NM, Masso-Valles D, Serrano E, Karnezis AN, Swigart LB, Evan GI. 2013. Inhibition of Myc family proteins eradicates KRas-driven lung cancer in mice. Genes Dev 27: 504–513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Swinney DC. 2013. Phenotypic vs. target-based drug discovery for first-in-class medicines. Clin Pharmacol Ther 93: 299–301 [DOI] [PubMed] [Google Scholar]
  117. Taub R, Kirsch I, Morton C, Lenoir G, Swan D, Tronick S, Aaronson S, Leder P. 1982. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc Natl Acad Sci 79: 7837–7841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Titov DV, Gilman B, He QL, Bhat S, Low WK, Dang Y, Smeaton M, Demain AL, Miller PS, Kugel JF, et al. 2011. XPB, a subunit of TFIIH, is a target of the natural product triptolide. Nat Chem Biol 7: 182–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Toyoshima M, Howie HL, Imakura M, Walsh RM, Annis JE, Chang AN, Frazier J, Chau BN, Loboda A, Linsley PS, et al. 2012. Functional genomics identifies therapeutic targets for MYC-driven cancer. Proc Natl Acad Sci 109: 9545–9550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. van Lohuizen M, Verbeek S, Krimpenfort P, Domen J, Saris C, Radaszkiewicz T, Berns A. 1989. Predisposition to lymphomagenesis in pim-1 transgenic mice: Cooperation with c-myc and N-myc in murine leukemia virus-induced tumors. Cell 56: 673–682 [DOI] [PubMed] [Google Scholar]
  121. Wang GG, Song J, Wang Z, Dormann HL, Casadio F, Li H, Luo JL, Patel DJ, Allis CD. 2009. Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger. Nature 459: 847–851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick LL, Fitzgerald P, Chi H, Munger J, et al. 2011. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35: 871–882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Welcker M, Orian A, Jin J, Grim JE, Harper JW, Eisenman RN, Clurman BE. 2004. The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc Natl Acad Sci 101: 9085–9090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Wells JA, McClendon CL. 2007. Reaching for high-hanging fruit in drug discovery at protein-protein interfaces. Nature 450: 1001–1009 [DOI] [PubMed] [Google Scholar]
  125. Weng AP, Millholland JM, Yashiro-Ohtani Y, Arcangeli ML, Lau A, Wai C, Del Bianco C, Rodriguez CG, Sai H, Tobias J, et al. 2006. c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. Genes Dev 20: 2096–2109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Whitty A. 2011. Growing PAINS in academic drug discovery. Future Med Chem 3: 797–801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Williamson JR. 2000. Induced fit in RNA-protein recognition. Nat Struct Biol 7: 834–837 [DOI] [PubMed] [Google Scholar]
  128. Wright PE, Dyson HJ. 1999. Intrinsically unstructured proteins: Re-assessing the protein structure-function paradigm. J Mol Biol 293: 321–331 [DOI] [PubMed] [Google Scholar]
  129. Wright JB, Brown SJ, Cole MD. 2010. Upregulation of c-MYC in cis through a large chromatin loop linked to a cancer risk-associated single-nucleotide polymorphism in colorectal cancer cells. Mol Cell Biol 30: 1411–1420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Xu Y, Shi J, Yamamoto N, Moss JA, Vogt PK, Janda KD. 2006. A credit-card library approach for disrupting protein–protein interactions. Bioorg Med Chem 14: 2660–2673 [DOI] [PubMed] [Google Scholar]
  131. Yada M, Hatakeyama S, Kamura T, Nishiyama M, Tsunematsu R, Imaki H, Ishida N, Okumura F, Nakayama K, Nakayama KI. 2004. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. EMBO J 23: 2116–2125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Yang D, Liu H, Goga A, Kim S, Yuneva M, Bishop JM. 2010. Therapeutic potential of a synthetic lethal interaction between the MYC proto-oncogene and inhibition of aurora-B kinase. Proc Natl Acad Sci 107: 13836–13841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Yap JL, Wang H, Hu A, Chauhan J, Jung KY, Gharavi RB, Prochownik EV, Fletcher S. 2013. Pharmacophore identification of c-Myc inhibitor 10074-G5. Bioorg Med Chem Lett 23: 370–374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Yin X, Giap C, Lazo JS, Prochownik EV. 2003. Low molecular weight inhibitors of Myc–Max interaction and function. Oncogene 22: 6151–6159 [DOI] [PubMed] [Google Scholar]
  135. Zheng Y, Balasubramanyam K, Cebrat M, Buck D, Guidez F, Zelent A, Alani RM, Cole PA. 2005. Synthesis and evaluation of a potent and selective cell-permeable p300 histone acetyltransferase inhibitor. J Am Chem Soc 127: 17182–17183 [DOI] [PubMed] [Google Scholar]
  136. Zippo A, De Robertis A, Serafini R, Oliviero S. 2007. PIM1-dependent phosphorylation of histone H3 at serine 10 is required for MYC-dependent transcriptional activation and oncogenic transformation. Nat Cell Biol 9: 932–944 [DOI] [PubMed] [Google Scholar]
  137. Zippo A, Serafini R, Rocchigiani M, Pennacchini S, Krepelova A, Oliviero S. 2009. Histone crosstalk between H3S10ph and H4K16ac generates a histone code that mediates transcription elongation. Cell 138: 1122–1136 [DOI] [PubMed] [Google Scholar]
  138. Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, Sison EA, Magoon D, Qi J, Blatt K, Wunderlich M, et al. 2011. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478: 524–528 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES