Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jun 1.
Published in final edited form as: Trends Pharmacol Sci. 2019 May 8;40(6):388–402. doi: 10.1016/j.tips.2019.04.004

Pharmacological modulation of transcriptional co-regulators in cancer

Timothy R Bishop 1,2, Yuxiang Zhang 1,2, Michael A Erb 1,*
PMCID: PMC6746237  NIHMSID: NIHMS1528937  PMID: 31078321

Abstract

Upon binding of transcription factors to cis-regulatory DNA sequences, transcriptional co-regulators are required for activation or suppression of chromatin-dependent transcriptional signaling. These co-regulators are frequently implicated in oncogenesis via causal roles in dysregulated, malignant transcriptional control and have represented one of the fastest growing target classes in small-molecule drug discovery. However, challenges in targeting co-regulators include identifying evidence of a cancer-specific genetic dependency, matching the pharmacologically addressable protein fold to a functional role in disease pathology, and achieving the necessary selectivity to exploit a given genetic dependency. Here, we discuss how recent trends in cancer pharmacology have confronted these challenges, positioning co-regulators as tractable targets towards the development of emergent cancer therapies.

Keywords: cancer, transcription, chromatin, therapeutics

Transcriptional Biology and Cancer Pharmacology

Proper spatiotemporal regulation of lineage-specific transcriptional processes is essential for normal organismal development, tissue function, and homeostasis. In a malignant cell state, mutationally altered genomes are accompanied by widespread dysregulation of transcriptional programs that ultimately promote self-renewal and unrestricted proliferation – as recently reviewed [1]. Knowledge of this type has motivated academic and industrial efforts to selectively alter transcriptional regulators with small-molecule tools and drugs. Unfortunately, with the exception of nuclear hormone receptors and limited other examples, a lack of ligandable (see Glossary) sites on transcription factors has made them impractical targets for direct-acting inhibitors. Instead, efforts to target the co-regulators required for transcription factor signaling have emerged as a viable alternative strategy.

Co-regulatory activity is encoded into diverse functional classes of proteins, including, but not limited to, chromatin modifiers (writers and erasers), chromatin remodelers, chromatin readers, and other scaffolding proteins (Figure 1). Chromatin modifiers are enzymes that function to post-translationally modify histone proteins, which alters the recruitment of chromatin readers that bind to modified histone substrates and can also alter the biophysical interaction between histones and DNA [2]. Chromatin remodelers leverage ATP hydrolysis to catalytically alter nucleosome spacing and composition, affecting the accessibility of the underlying genome [3]; some also possess the ability to evict chromatin-bound complexes that possess antagonistic regulatory function [4]. Many co-regulators do not act enzymatically on a nucleosome substrate, but instead they serve as scaffolds to assemble diverse regulatory complexes at cis-regulatory elements. As with all cancer therapy, the goal of targeting co-regulators is to elicit tumor-specific cell killing in a manner that maintains the survival and physiology of normal cells in the body. Thus, the importance of high-quality target identification and validation studies – whether accomplished by genetic experimentation or by use of tool compounds – cannot be overstated (as recently highlighted [5]). Ultimately, pharmacologic development of a given target is poised to succeed when: (i) the target protein is a cancer-specific dependency, (ii) the cancer-relevant function of the protein is addressable by pharmacologic approaches, and (iii) sufficient selectivity can be achieved by a pharmacologic agent to exploit the vulnerability of interest without eliciting toxic off-target effects.

Figure 1. Mechanisms of transcriptional co-regulators.

Figure 1.

Transcriptional co-regulators mediate transcription factor signaling via multiple molecular mechanisms. Chromatin remodelers alter nucleosome positioning and composition, chromatin writers add post-translational modifications (shown as orange circles) to histones and other proteins, histone erasers remove such post-translational modifications, and chromatin readers assemble regulatory complexes by anchoring onto histones with specific states of modification.

Conceptually, a transcriptional co-regulator, like any protein, can be considered a cancer-specific dependency through two genetic mechanisms: oncogene [6] and non-oncogene [7] addiction (Box 1). For the practice of drug discovery, cancer-specific dependencies provide a genetic rationale for pharmacologically modulating a target with the prospect of achieving a high therapeutic index. In the following, we review recent advances in the pharmacology of transcriptional co-regulators, attempting to highlight each target in the context of its role as an oncogene or non-oncogene dependency in human cancers. We begin with targets regulating transcriptional elongation, discuss diverse bromodomain-containing cancer targets, and finish with a highlight of emerging targets involved in histone methylation.

Box 1. Genetics of cancer-specific dependencies.

Cancer-specific dependency refers to protein functions and regulatory pathways that are specifically required for the survival of a cancer cell but are less important to the survival and function of normal cells and tissues. Oncogene dependency, equivalently termed oncogene addiction, refers specifically to a continuous requirement by a cancer cell for the function of its oncogene [6]. Meanwhile, non-oncogene dependency is the concept of requiring the function of a protein or pathway that although not an oncogenic driver of the cancer, is still essential for its growth or survival, but comparatively less important in normal cells and tissues [7]. This concept was proposed based on the hypothesis that certain alterations present in a cancer cell may provoke a deficiency in, or additional need for, some function that does not affect physiological cell survival. Several mechanisms for this are now well-understood, including, but not limited to:

  1. Loss-of-function mutations that confer vulnerability to loss of a homolog with some redundancy in protein function.

  2. Loss-of-function mutations that confer vulnerability to loss of proteins with opposing regulatory function.

  3. Gain-of-function mutations that confer vulnerability to loss of effectors that are required for expression of the gain-of-function phenotype.

The concept of synthetic lethality has been borrowed from classical genetics to describe these classes of nononcogene dependencies in which loss of protein function – via genetic or pharmacological perturbation – is only lethal in the presence of a cancer-associated genetic alteration. Other non-oncogene dependencies may not be as easily defined by a single mutation, but by the combined action of many genetic, transcriptional, and/or post-transcriptional alterations that together, confer a cancer cell state expressing a unique vulnerability. An on-going question for translational cancer research is the best way to demonstrate that a dependency is cancer-specific, such that drugs designed to target that dependency exhibit the anticipated therapeutic index. When animal models for the disease are tractable, this can be answered by measuring effects of protein loss on normal and malignant tissues. However, when animal models are not tractable, panels of cell lines across multiple cancer lineages can be used to demonstrate lineage-restricted essentiality as a surrogate measure of cancer-specific essentiality. High-quality chemical probes for a potential target can be particularly valuable toward these ends and may also have direct relevance as starting points for drug development [139].

Targeting RNA Polymerase II Pause Release

CDK9

After transcriptional initiation, RNA Polymerase (Pol) II is subjected to a tightly regulated pausing step just downstream of transcription start sites. From this promoter-proximal pausing, Pol II awaits molecular licensing for productive transcriptional elongation via the kinase activity of positive transcription elongation factor b (P-TEFb), a heterodimeric complex of cyclin-dependent kinase 9 (CDK9) and Cyclin T. P-TEFb is inhibited by the promiscuous CDK inhibitor, flavopiridol, and thus, CDK9 pharmacology has been extensively studied in human cancers [8].

Despite positive anti-cancer efficacy, CDK inhibitors have historically been limited clinically by off-target pharmacology and dose-limiting toxicity. However, pharmacologic advances enabling selective perturbation of CDK9 have prompted a reconsideration of CDK9 inhibition as cancer therapy. Recently disclosed agents include the inhibitors, iCDK9 [9], NVP-2 [10], and MC180295 [11], and a selective degrader, THAL-SNS-032 (Figure 2, Key Figure) [12]. As selective CDK9 agents enter the clinic, it will be instructive to observe how gains in selectivity affect the therapeutic index of perturbing CDK9 in patients. Notwithstanding the notable advances in pharmacological development, the intrinsic target biology of CDK9 may yet provide challenges for drug development; namely, that CDK9 kinase activity is a universal requirement for productive gene transcription [12-15]. It follows that on-target, dose-limiting toxicity may limit the therapeutic index of CDK9 inhibition as healthy tissues may be equally affected by global loss of transcriptional activity. Careful dosing regimens may help mitigate on-target toxicities, but the anticipation of this issue has motivated efforts to inhibit transcriptional co-activators with more specialized roles across the genome.

Figure 2, Key Figure. Targeting transcriptional co-regulators in cancer.

Figure 2, Key Figure.

(A) Recent examples of pharmacological agents able to disrupt transcriptional co-regulators. Small green circles indicate acetyl-lysine, small red circles indicate H3K27me3, and the small black circle on RNA Pol II represents post-translational phosphorylation.

BET Family Proteins

Bromodomain protein 4 (BRD4) is a transcriptional co-activator known to associate with CDK9 and affect pause release of RNA Pol II. BRD4 is part of the bromodomain and extra-terminal (BET) family of proteins (BRD2, BRD3, BRD4, and BRDT), each of which possess a pair of tandem bromodomains positioned at their N-terminus. Bromodomains are protein folds evolutionarily adapted to bind to acetylated lysine side chains found within histones and on other chromatin-bound proteins and this function positions BRD4 at transcriptionally active cis-regulatory elements. Because of its links to CDK9 activity, BRD4 has emerged as a compelling target to pharmacologically disrupt pause release at select target genes, rather than across the entire genome.

BRD4 was first causally linked to cancer pathogenesis by the discovery that chimeric fusions of BRD4 to NUT (nuclear protein in testes) form oncogenes that drive a severe squamous cancer termed NUT midline carcinoma (NMC). Early efforts with JQ1 (Figure 2), a first-in-class bromodomain inhibitor, established BET bromodomain inhibition as a promising strategy to target BRD4 oncogene addiction [16], which has inspired ongoing clinical testing in NMC [17,18]. In these tumors, BET bromodomain inhibition targets BRD4-NUT oncogene dependency by displacing the fusion oncoprotein from chromatin, ultimately leading to squamous differentiation and arrested proliferation [16].

Interestingly, wild-type BRD4 has also been identified as a non-oncogene addiction in a variety of human cancers. Hematological malignancies, the first tumors shown to require wild-type BRD4, are hypersensitive to BET bromodomain inhibition [16,19-22], which has motivated a number of clinical trials that have reported preliminary signs of efficacy [23-25]. While response rates in early Phase 1 reports have not been as high as hoped based on preclinical studies, side-effects have been relatively mild – the most common being thrombocytopenia – suggesting BET bromodomain inhibition may be most compellingly used as part of combination therapies. Nevertheless, lower-than-expected response rates also highlight our need to discover predictive biomarkers of sensitivity so that drug candidates can be better matched to the patients most likely to receive benefit.

While predictive biomarkers remain elusive, the mechanisms of sensitivity to BET bromodomain inhibition in BRD4-dependent tumors have been extraordinarily well studied. In comparison to CDK9 inhibition, BET bromodomain inhibitors elicit relatively narrow transcriptional responses, preferentially affecting a class of genes driven by large enhancer clusters – often referred to as super-enhancers – that host massive amounts of chromatin-bound BRD4 [26]. This class is enriched for cell-type identity genes and in cancer, this includes many proto-oncogenes [26,27]. Thus, the mechanism of BET bromodomain inhibitor sensitivity is typically attributed to preferential downregulation of tumorigenic transcription factors, perhaps best exemplified by MYC suppression in hematological malignancies [19,20].

Recently, BET proteins have been at the center of breakthroughs in the field of targeted protein degradation. Proteolysis targeting chimeras (PROTACs) are bi-functional small molecules that recruit a liganded target protein to an E3 ubiquitin ligase complex to induce proximity-mediated poly-ubiquitination and proteolysis (Figure 3A) [28]. The recent use of small-molecule immunomodulatory drugs (IMiDs) as the E3-targeting moiety has allowed for targeted protein degradation in vivo and brought renewed interest in small-molecule degraders as therapeutics and chemical tools [29]. This was first demonstrated using dBET1, a phthalimide conjugation of JQ1 that simultaneously binds cereblon (CRBN) and BET proteins to induce CRL4CRBN- dependent degradation of the BET family [29].

Figure 3. The emerging pharmacology of small-molecule protein degraders.

Figure 3.

(A) Schematic showing the design of PROTACs. PROTACs are composed of a target protein ligand and an E3 ubiquitin ligase-binding ligand, connected by a linker region. PROTAC induce targeted protein degradation by simultaneously engaging a target protein and an E3 ubiquitin ligase complex, which results in poly-ubiquitination of the target protein and ultimately, proteasome-dependent degradation. (B) BET bromodomain inhibitors (blue ovals) inhibit the protein (BRD4) by occupying its druggable site and consequently disables its interaction with acetyl-lysine side chains (green circles) on chromatin. BRD4 PROTACs can act sub-stoichiometrically by directing multiple cycles of ubiquitination and degradation and remove the entire protein (BRD4) rather than inhibiting it.

Our work with dBET6 (Figure 2), a BET degrader optimized for cellular permeability, has revealed major differences between the cellular responses to BET degradation and BET bromodomain inhibition (Figure 3B). In contrast to the selective transcriptional effects evoked by BET bromodomain inhibition, BET degradation results in complete collapse of mRNA production by preventing the release of RNA Pol II from promoter proximal pausing genome-wide [15]. It had previously been suggested that BRD4 recruits CDK9 to chromatin and that this was the mechanism by which BRD4 affects RNA Pol II pause release [30]. Quite interestingly, while CDK9 activity is indeed eliminated by BET degradation, it does not result in loss of CDK9 occupancy on chromatin [15]. Recent application of the auxin-inducible degron system [31] to selectively degrade BRD4 has elaborated on these results, demonstrating that like BET degradation, BRD4 degradation results in transcriptional collapse without affecting CDK9 occupancy [32]. Thus, the mechanism by which BRD4 regulates CDK9 activity remains an area of open investigation.

While the BET bromodomain inhibitors and BET degraders mentioned above target the entire BET family, degradation tools have recently enabled BRD4-specific pharmacological perturbations. Remarkably, attempts to optimize BET-targeted PROTACs to only degrade BRD4 have been successful, resulting in the disclosure of multiple BRD4 degraders that do not affect BRD2 or BRD3 protein levels [33-35]. We are hopeful that PROTAC development will eventually provide chemical tools to study all BET proteins individually, each of which may have unique roles in cancer pathogenesis. For example, genetic depletion of BRD2, which can act as an oncogene to drive B-cell malignancies [36], also diminishes growth of BRD4-dependent breast cancer and chronic lymphocytic leukemia cells [37,38], but not AML cells [39]. This may reflect the fact that BRD2 and BRD4 have at least partially non-redundant function, with BRD2 functioning in three-dimensional genome organization [40,41], and highlights the need for chemical probes to disentangle BET family target biology. Still, BET degradation has already proved a very useful tool for studying BRD4-dependent transcriptional regulation in cancer cells.

Given concerns about global inhibition of mRNA synthesis causing on-target toxicity in normal tissues, BET degraders will potentially face the same set of challenges encountered by CDK9 inhibitors in the clinic. Nevertheless, disclosures of BET degrader activity in vivo have been promising [15,29,42-44], notably including a picomolar-potent molecule, QCA570, which shows anti-tumor activity in mice at 1 mg/kg dosing [44]. From a therapeutic perspective, a particularly intriguing use for pan-BET or BRD4-specific degraders would be as a means to overcome BET bromodomain inhibitor resistance. In each of three index reports, resistance to BET bromodomain inhibition was attributed to rewiring of transcriptional signaling pathways in a manner that dispensed of BET bromodomain requirement [37,39,45]. In leukemia, resistant cells are able to sustain or rapidly reactivate MYC expression upon BET bromodomain inhibition due to increases in Wingless and Int-related (WNT) signaling [39,45] – confirming the existence of BET-bromodomain-independent mechanisms of maintaining MYC expression. However, in both leukemia and breast cancer models of evasive resistance, genetic addiction to BRD4 is retained, suggesting that BET degradation, which is able to recapitulate genetic depletion of BRD4, might be able to overcome bromodomain inhibitor resistance. While this has not yet been tested, it may offer a compelling rationale for moving BET degraders into clinical trials.

ENL YEATS

ENL (eleven nineteen leukemia) is a chromatin reader and transcriptional co-activator that regulates pause release as a member of the super elongation complex (SEC) [46], a higher-order P-TEFb-containing complex [47]. It possesses an N-terminal YEATS domain, which binds to acyl-lysine side chains on histone H3, including several longer short-chain fatty acid groups (e.g. crotonyl-lysine) that bromodomains do not bind [48-50]. By means of CRISPR-Cas9 loss-of-function screening, we and other identified ENL as a non-oncogene dependency in acute leukemia [46,51]. While ENL is required for the survival of acute leukemia cells in vivo, normal hematopoietic stem and progenitors cells are insensitive to loss of ENL, suggesting ENL-targeted therapeutics might be able to elicit cancer-specific cell killing with a wide therapeutic window [46,51].

Recognizing ENL as a potential therapeutic target, we sought to identify a function within the ENL protein that would be actionable for drug discovery. Especially in multidomain proteins, it is not always obvious whether a specific domain within a potential drug target will contribute to the pathogenic function of that protein [52,53]. Thus, we and others used a mutagenesis approach to demonstrate that the YEATS domain is essential for leukemia cell survival by mediating the localization of ENL to acetylated chromatin structures [46,51]. Building on this, chemical discovery efforts have already produced small-molecule inhibitors of ENL YEATS, including SGC-iMLLT (Figure 2) [54], though further work is required to evaluate the biological activity of these molecules.

Interested in the transcriptional mechanisms underlying ENL sensitivity, we sought to study ENL by a pharmacological approach. To that end, we developed a chemically inducible protein degradation system, dTAG, to model an acute pharmacologic perturbation of ENL [46]. The rapid kinetics of this system permit evaluation of direct mechanisms of protein function, which we leveraged to study ENL-dependent transcription in leukemia [46,55]. Coupling acute degradation of ENL to integrative transcriptional genomics, we identified a class of target genes – highly enriched for undruggable leukemic driver genes, including MYC and MYB – that are selectively downregulated by loss of ENL [46]. Compared to others, these genes feature disproportionally high loads of promoter-bound ENL and upon ENL degradation, they are preferentially depleted of promoter-bound SEC before being selectively downregulated [46]. Thus, ENL selectively drives a tumorigenic gene expression program by recruiting the SEC to leukemic driver genes. Since co-regulators are often involved in rapid induction or suppression of transcriptional activity, it is often difficult to differentiate between primary mechanisms of action and secondary transcriptional effects with the use of genetic perturbations. However, modeling pharmacologic perturbations with systems like dTAG, as done with ENL, offers the ability to resolve phenotypes at earlier time points, aiding in both mechanistic basic biology and translational target validation studies [56].

These mechanistic data highlight an interesting relationship between ENL and BRD4. The phenotypic effects of ENL degradation and BET bromodomain inhibition are similar in that they both affect promoter proximal pause release and preferentially suppress expression of tumorigenic transcription factors. However, BET bromodomain inhibition affects a much larger fraction of the transcriptome, suggesting that they function distinctly. In agreement with this, combining ENL loss with BET bromodomain inhibition shows additive effects on leukemia proliferation in vivo [51]. An interesting hypothesis for their discrete regulatory roles would be that ENL and other proteins regulate P-TEFb recruitment while BRD4 controls its activity once it is placed onto chromatin.

Co-Regulators with Non-BET Bromodomains

The early success of BET bromodomain inhibition illustrated the feasibility of targeting acetyl-lysine recognition by chromatin readers and in turn, inspired a wave of efforts to target other, non-BET bromodomains. These ongoing efforts have been extraordinarily successful, ultimately drugging nearly 2 dozen different bromodomain targets [57]. Since not all of these proteins are applicable to cancer, we will focus on a handful of especially instructive examples, which highlight the complex manner by which bromodomains can contribute to protein function and affect – or not – cancer cell survival.

SWI/SNF complex – SMARCA2/4

The SWI/SNF, or BAF, complex (Box 2) is a large, multisubunit, ATP-dependent chromatin remodeler complex that functions at cis-regulatory elements to maintain accessible chromatin structures. Its ATPase function is encoded in two mutually exclusive and highly homologous proteins: SMARCA2 and SMARCA4. Mutations to SMARCA4, which frequently occur in non-small-cell lung carcinomas (NSCLC), are synthetic lethal with loss of SMARCA2, which is otherwise non-essential for cell survival [58-60]. In addition to their highly homologous ATPase domains, SMARCA2 and SMARCA4 also possess highly homologous bromodomains, which have been successfully targeted by a selective, cell-permeable small molecule, PFI-3 [61,62]. Unfortunately, this compound is unable to displace SMARCA2/4 from chromatin and does not recapitulate the anti-proliferative effects of genetic knockdown [52]. Ultimately, functional genetic experiments have revealed that the catalytic ATPase domain, but not the bromodomain, of SMARCA2 is essential in the context of SMARCA2 dependency and is thus the relevant drug target [52]. It is interesting to note that the dispensability of the SMARCA2/4 bromodomains might have been foreshadowed by work in Drosophila melanogaster defining the bromodomain of the fly homolog, BRM (Brahma), as non-essential for localization of the complex to active chromatin [63]. Nevertheless, a recent breakthrough in discovery chemistry has produced the first pharmacologic inhibitors of SMARCA2/4 ATPase activity: potent diheteroaryl ureas (Figure 2, Compound 14) that act via allostery and diminish tumor growth in vivo upon oral dosing [64]. While in-depth biological characterization has yet to be reported with this series of molecules, they are positioned to advance SMARCA2/4 pharmacology in therapeutically relevant ways. Importantly, these molecules establish the feasibility of targeting chromatin remodeler ATPase domains with drug-like small molecules and we are hopeful that their discovery will buoy increased enthusiasm for chromatin remodeler discovery chemistry. However, despite demonstrating an ability to target the relevant ATPase function, the selectivity needed to realize the promise of SMARCA2 as a synthetic lethality remains a significant hurdle to overcome, urging discovery of additional targets for pharmacological development.

Box 2. SWI/SNF complex.

The SWI/SNF complex is an evolutionarily conserved, multisubunit chromatin remodeler, first identified in yeast genetic screens and named for mating type switching (swi) and sucrose non-fermenting (snf) alleles [140-142]. In mammalian cells, SWI/SNF evolved to a ~2 MDa complex and incorporates one of two mutually exclusive ATPase subunits: SWI/SNF-related, matrix associated, actin-dependent regulator of chromatin subfamily A member 2 (SMARCA2, also Brahma or BRM) and SMARCA4 (also Brahma related gene 1 or BRG1). Also termed BAF (BRG1/BRM-associated factor), mammalian SWI/SNF complexes are able to adopt diversified functions via combinatorial assembly of different homologs across multiple gene families [143]. Moreover, multiple distinct complex formations have been characterized to date, termed BAF, PBAF, and ncBAF [65,66,144]. A remarkable 20% of human cancers feature a mutation to a gene encoding a SWI/SNF subunit [145] and many of these mutations present opportunities to exploit synthetic lethality for the development of anti-cancer therapeutics [146].

SWI/SNF complex – BRD9

BRD9, which is a member of the non-canonical SWI/SNF complex (ncBAF) [65,66], represents another target for therapeutic disruption of non-oncogene dependencies present within the SWI/SNF complex. A recent study has demonstrated that BRD9, and specifically its bromodomain, is required for the growth of acute myeloid leukemia (AML) cells through the maintenance of high MYC expression [67]. This was confirmed using the chemical tool BI-7273, a potent inhibitor of BRD7 and BRD9 [67]. Like BET bromodomain inhibitors, BRD7/9 inhibitors have also been converted into PROTACs via conjugation to E3-binding small molecules [68,69]. dBRD9 (Figure 2), a PROTAC built via IMiD conjugation of BI-7273 is a potent and selective BRD9 degrader, which remarkably, does not degrade BRD7 [68]. Already, this tool has been put to use in contemporaneous studies that discovered a synthetic lethal dependency on BRD9 in BAF-mutant tumors [65,70]. Together, these studies identified that sarcomas and rhabdoid tumors that are driven by mutations to BAF components are highly sensitive to loss of BRD9 via genetic disruption or exposure to dBRD9 [65,70].

CBP/P300

CREB binding protein (CBP) and p300 (adenovirus E1a associated 300 kDa protein) are two closely related histone acetyltransferases (HATs) that act as transcriptional co-activators by catalyzing the acylation of histone and non-histone lysine side chains (including H3K27ac, a modification found at promoters and putatively active enhancers). Owing to their synthetic lethal relationship, CBP and p300 have emerged as highly attractive cancer targets, particularly in certain lung cancers and lymphomas that frequently harbor mutations to these proteins [71-73]. In addition to their HAT domains, CBP and p300 also feature highly homologous bromodomains, the pharmacology of which has rapidly matured in recent years to include several potent and selective inhibitors [74-78]. Impressively, the successful development of CBP/p300 bromodomain inhibitors has been followed quickly by the disclosure of the first chemical probe for CBP/p300 HAT activity, an indane spirooxazolidinedione, A-485 (Figure 2) [79,80]. A-485 represents a major leap forward in HAT pharmacology, as previous agents were troubled by off-target activity, electrophilicity, and a tendency toward aggregation [81]. While none of the presently existing small-molecule inhibitors of CBP/p300 feature sufficient selectivity to exploit synthetic lethality between the two proteins, convergent chemical biology from separate groups using either A-485 or the CBP/p300 bromodomain inhibitor, GNE-272 (Figure 2), have revealed a selective dependency on CBP/p300 in androgen receptor-positive, but not androgen receptor-negative, prostate cancer growth [79,82]. Perhaps unexpectedly, these molecules are not promiscuously cytotoxic, but show context-specific anti-cancer effects, both within and among different cancer lineages and subtypes. From both a therapeutic and basic biological perspective, it will be important to identify predictive biomarkers of sensitivity and mechanisms of transcriptional response to loss of CBP/p300-mediated lysine acetylation. However, these molecules have already revealed fascinating target biology that would have been difficult to discover without pharmacologic tools. Notably, CBP/p300 bromodomain inhibition has revealed that while the bromodomain is not required for chromatin localization, it is essential for CBP/p300-mediated H3K27 acetylation and enhancer activity [83]. Moreover, CBP/p300 HAT inhibition coupled to quantitative proteomics have revealed that the repertoire of CBP/p300 substrates vastly exceeds a few histone and non-histone substrates; instead, CBP/p300 function through a so-called “acetyl-spray” mechanism, modifying thousands of sites at cis- regulatory elements [84]. Very importantly, the discovery of A-485 has definitively established that HATs can be potently and selectively inhibited by drug-like small molecules, now further confirmed by the recent disclosure of inhibitors for the structurally distinct MYST-family acetyltransferases, KAT6A and KAT6B [85]. We expect these discoveries to inspire increased consideration of HAT pharmacology for both chemical biology and cancer therapy.

The above considerations of bromodomain pharmacology offer instructive examples of the diverse outcomes that are possible when binding drug-like small molecules to a chromatin reader domain. While BET bromodomain inhibitors displace BET proteins from chromatin, CBP/p300 bromodomain inhibitors allosterically suppress HAT activity and SMARCA2/4 bromodomain ligands are entirely ineffectual. Using the co-repressor, TRIM24, as an example below, we will discuss how targeted protein degradation can rescue ineffectual ligands, creating high-quality chemical probes that phenocopy genetic depletion.

TRIM24

TRIM24 is a member of the TRIM/RBCC protein family, consisting of an N-terminal tripartite motif and a C-terminal tandem plant homeodomain finger-bromodomain, and has been implicated as a non-oncogene dependency in breast and castrate-resistant prostate cancer [86,87]. Previously, two groups independently disclosed dimethylbenzimidazolone scaffolds as dual inhibitors of TRIM24 and BRPF1 bromodomains, including IACS-9571 [88,89]. Despite double-digit nanomolar affinity, cellular pharmacology with IACS-9571 has been modest [90], inspiring PROTAC development via IMiD conjugation, which yielded a selective degrader of TRIM24, dTRIM24 [91]. Compared to IACS-9571, dTRIM24 elicits increased anti-proliferative effects in AML cells, which are preferentially sensitive to RNAi depletion of TRIM24 [91]. This increase in active pharmacology is rationalized by the discovery of an AML-specific dependency on the TRIM24 RING domain [91], which is not addressed by TRIM24 bromodomain inhibitors. Like SMARCA2/4, it seems that the bromodomain is not the relevant function of TRIM24 with respect to its role in AML, which highlights the importance of identifying domain-specific dependencies in the development of new therapeutics. However, we are hopeful that targeted protein degradation will be broadly applicable in turning ineffectual ligands into effective degraders able to address all aspects of target protein function.

Targeting Histone Methylation in Human Cancers

Polycomb repressive complexes

The polycomb repressive complexes, PRC1 and PRC2, are transcriptional co-repressors that function via chromatin compaction. Polycomb-mediated gene silencing is initiated by the histone methyltransferase (HMT) subunit of PRC2, EZH2 (enhancer of zeste homolog), which catalyzes methylation of H3K27. PRC1 recognizes H3K27me3 via chromodomain-containing substrate adapters (CBX2, CBX4, CBX6, CBX7, and CBX8) and effects subsequent gene silencing via modifications to histones and DNA. As comprehensively reviewed [92], EZH2 is implicated as an oncogene and non-oncogene dependency in a variety of cancers, owing to overexpression of EZH2, gain-of-function mutations to EZH2, and loss-of-function mutations to SWI/SNF complex subunits, which ordinarily oppose PRC2 function. This has inspired a number of clinical development programs targeting the EZH2 methyltransferase and recently, a phase 1 study of tazemetostat (Figure 2), an EZH2 HMT inhibitor, reported nearly 40% response rates in both non-Hodgkin lymphomas and SWI/SNF-mutant solid tumors [93]. Interestingly, EZH2 has also been reported to possess non-enzymatic, scaffolding function that is essential for the survival for SWI/SNF-mutant tumors [94]. In this context, genetic or chemical disruption of methyltransferase activity only partially exploits EZH2 dependency [94], presenting an attractive rationale to develop EZH2-selective PROTACs able to fully phenocopy genetic depletion.

More recently, two groups concurrently revealed first-in-class allosteric inhibitors of PRC2 that bind the WD40 domain of the core PRC2 subunit, EED [95,96]. These structurally distinct small molecules, EED226 and A-395 (Figure 2), potently inhibit methyltransferase activity in vivo and excitingly, are able to overcome mutations to EZH2 that confer resistance to EZH2 HMT inhibitors [95,96]. As yet another alternative, several groups have endeavored to inhibit the downstream effectors of PRC2-installed H3K27me3 by targeting the chromodomains of CBX family proteins in PRC1 that bind to the repressive histone mark [97,98]. These efforts have focused on peptide-based scaffolds, yielding potent tool compounds, including the chemical probe, UNC3866 (Figure 2) [98]. Whether CBX chromodomains – and chromodomains at large – will be tractable targets for small molecules remains an open question.

Methyltransferases and Hematological Malignancies

Outside of PRC2, a number of other histone methyltransferases have also been described as dependencies in cancer, particularly in hematologic malignancies. DOT1L, the methyltransferase responsible for modifying H3K79, is a well-described dependency in acute leukemia bearing fusion oncogenes involving lysine methyltransferase 2A (KMT2A) [99-101]. Pinometostat, a clinical-stage DOT1L inhibitor [102], is very well tolerated and shows initial signs of efficacy in early-phase trials of KMT2A-fusion acute leukemia [103]. Other efforts to kill KMT2A-fusion leukemia have focused on the interaction between KMT2A and its cofactor, Menin. Small-molecule inhibitors of Menin-KMT2A are effective in several leukemic subtypes [104,105] and are expected to enter clinical trials soon. ASH1-like histone lysine methytransferase (ASH1L), which methylates H3K36, has been similarly described as a dependency in KMT2A-fusion-positive leukemia [106], and intriguingly, a series of 3-(1H-indol-3-yl)benzothioamide-based inhibitors and PROTACs for ASH1L have been reported in the patent literature [107]. Additionally, the H3K36 HMT, NSD2, is known to be frequently translocated or mutated in various hematologic malignancies resulting in hyperactivity and increased sensitivity to loss of NSD2 [108]. Early efforts in assay development and discovery chemistry for the NSD2 methyltransferase have recently been reported – a likely foreshadowing of future therapeutic development [109].

Histone Demethylation and Drug-Tolerant Persistence

Histone demethylase enzymes, which function antagonistically to histone methyltransferases, are also extensively implicated in cancer pathogenesis, notably including the interesting phenomenon of drug-tolerant persistence. This is a form of non-genetic tolerance to anti-cancer drugs that emerges from rare subpopulations of cancer cells [110,111]. Converging lines of evidence implicate chromatin regulatory rewiring via activity of lysine-specific demethylases, including lysine demethylase 5 (KDM5) and KDM6 families, as important in multiple models of drug-tolerant persistence [110,112-114]. Both KDM5 and KDM6 families have proven druggable and recent efforts have extended into highly potent inhibitors with activity in vivo [115,116]. In one report, CPI-455, a selective pyrimidinone inhibitor of the KDM5 family demethylases, effectively diminished the emergence of drug-tolerant persister cells following treatment with distinct cytotoxic anti-cancer drugs in diverse cancer lineages in vitro [113]. Extension of this phenotype in vivo holds promise as a means of widely suppressing drug resistance mechanisms in the clinic.

Concluding Remarks and Future Perspectives

As witnessed by the examples above, the pharmacological science of targeting transcriptional co-regulators has matured immeasurably. Efforts in discovery chemistry are routinely able to elaborate highly selective chemical tools and drug candidates, but significant challenges and outstanding questions remain limiting in many instances (see Outstanding Questions). In highlighting exemplary molecules and their intended targets here, we hope to have provided examples that are instructive on the challenges of exploiting well-defined cancer vulnerabilities with molecularly targeted pharmacological agents.

OUTSTANDING QUESTIONS:

  • How will the first generation of drugs targeting co-regulators perform in the clinic?

  • What role will combination therapy play in bringing co-regulator pharmacology to the clinic?

  • What common, but as-yet undrugged, co-regulator protein folds might be amenable to pharmacological modulation?

  • Considering the unique advantages of targeted protein degradation, but also concerns about the size of bifunctional PROTACs, what role will targeted protein degradation play in bringing co-regulator pharmacology to the clinic?

  • What are the underlying principles governing “degradability” and can these principles be applied predictively to achieve selectivity between high-affinity targets?

The first challenge is to target a protein that is preferentially important to the survival of cancer cells compared to normal tissues. Without this intrinsic biological relationship to tumor cells, the therapeutic index of drugs that address these targets will be limited by the extent of on-target toxicity caused to essential physiological processes. Realization of this fact has motivated ongoing efforts to systematically map cancer-specific genetic dependencies [117-119]. However, for co-regulators in particular, it must be considered that screens performed in tissue culture might overlook additional drug targets that are required for growth in vivo but not in vitro. Such in vitro false negatives were previously observed for several transcription elongation factors in glioblastoma, in which these co-regulators support pro-growth transcriptional pathways specifically within the tumor microenvironment [120]. It will therefore be highly valuable, when possible, to complement large-scale screening efforts performed in tissue culture with focused screens in suitable animal models.

Even without accounting for potential false negatives, many more dependencies exist than are currently considered to be imminently druggable. However, our notion of what constitutes an actionable drug targets is continuously being reimagined. The expanding scope of protein ligandability, aided both by covalent chemical proteomics and fragment-based screening in cells [121-124], will sustain the discovery of new druggable targets. Focusing on co-regulators, lysine ligandability mapping has already resulted in the identification of a druggable site on Sin3A, which when targeted, disrupts protein-protein interactions [125]. However, reconsidered in light of PROTAC technologies, even inactive ligands have the potential to support the design of new drugs. We imagine the convergence of these technologies will catalyze a renewed definition of druggability – one that will greatly expand the pharmacology of co-regulators, particularly for those without enzymatic function or obvious ligand-binding domains.

The second major challenge highlighted here, especially important with multidomain transcriptional coregulators, is the potential for drug discovery efforts to address a domain that is not functionally involved in the dependency ascribed to an intended target. Given the difficulty in scaling genetic validation of domain-specific dependencies, researchers have adapted CRISPR-based technologies for highly parallel interrogation of individual protein domains within potential cancer targets via genetic screening [53]. Despite this breakthrough, the question remains of what to do when the relevant cancer-related function is not immediately or obviously tractable by traditional pharmacological approaches. Here, it is possible that targeted protein degradation, which circumvents the need to modulate a specific biochemical or biophysical function by eliminating the protein entirely, will be widely applicable.

Finally, we have considered the challenge of selectivity. While discovery chemistry programs are well-versed in elaborating drug-like small molecules with exquisite selectivity profiles, even the most selective compounds often target one or more closely related homologs (e.g. CBP/p300 bromodomain and HAT inhibitors target both homologs). Unfortunately, off-target activity on close homologs often limits the ability to develop drugs targeting synthetic lethality in cancer as synthetic lethal relationships are often observed between closely related homologs with redundant or semi-redundant function. It is becoming clear that PROTACs may offer assistance with this challenge as well. On multiple occasions, PROTACs have been discovered to possess improved selectivity compared to the parent ligand, sometimes affecting just a single protein [12,33-35,68,126-128].

Although we have highlighted the potential for targeted protein degradation to overcome significant challenges in co-regulator pharmacology (and drug discovery more broadly), this field still faces a number of challenges itself. Notably, the origin of improved selectivity – when it is observed – is often unclear, highlighting the need to discover generalizable principles governing “degradability” [34,35,129]. Moreover, the bulk of disclosed small-molecule PROTAC molecules have been synthesized by conjugation to one of two E3-targeting ligands. As additional E3-targeting ligands are discovered [130-134], the practical scope of degradable targets is likely to be significantly expanded, which is bound to impact the pharmacological development of transcriptional co-regulator.

Like with most, if not all, molecularly targeted cancer therapeutics, it will be essential to identify appropriate combination therapies able to increase efficacy and suppress the emergence of evasive resistance. Mounting evidence has implicated several transcriptional co-regulators as determining responses to anti-tumor immunotherapy, offering a particularly intriguing setting for combining transcriptionally targeted drug candidates. In less than two years, CDK9, SWI/SNF, and the lysine demethylase, KDM1A have all been identified as targets able to potentiate anti-cancer immunotherapy drugs [11,135-137]

The most important experiment, ongoing in many cases, is the clinical evaluation of drugs targeting coregulators in cancer. Setting aside the histone deacetylase (HDAC) inhibitors that have been approved as anticancer agents by the Food and Drug Administration (FDA) (which we have decided not to discuss here as this target class has been well-reviewed in [138]), the targets discussed in this review have reached only as far as clinical trials. Thus, the results of ongoing clinical trials are anxiously awaited to answer questions of efficacy and therapeutic index in patients. Meanwhile, sophisticated use of genetics and pharmacology will continue to grow the list of transcriptional co-regulators that represent tractable targets for therapeutic intervention.

HIGHLIGHTS:

  • Transcriptional co-activators and co-repressors (together, co-regulators) are chromatin-associated proteins that mediate signaling from DNA-bound transcription factors to the activation or suppression of gene transcription.

  • An increasing number of co-regulators are annotated as cancer-specific oncogene and non-oncogene dependencies in human malignancies.

  • Recent disclosures of drug-like small molecules have revealed many of these proteins, and the cancer-promoting activity encoded within them, to be tractable targets for pharmacological modulation.

  • Application of these small-molecule tools is opening new views into the function of co-regulators in transcriptional control and identifying underlying mechanisms by which these genes affect cancer-specific cell survival.

Acknowledgements

The authors would like to gratefully acknowledge Drs. Georg E. Winter and Rhamy Zeid for critical reading of this manuscript while in preparation. M.A.E. is supported by the National Institutes of Health through the NIH Director’s Early Independence Award (DP5-OD26380).

Glossary

Auxin-inducible degron:

auxin is a phytohormone that binds the E3 ligase substrate adapter, TIR1, enabling association and eventual degradation of AUX/IAA transcriptional repressors. Expression of TIR1 in mammalian cells, coupled with tagging of a protein of interest (POI) with an epitope from AUX/IAA, enables rapid and reversible auxin-inducible degradation of POI-AUX/IAA fusion proteins.

Cancer-specific dependency:

a vulnerability affecting cancer cell survival that is not present in normal cells and tissues.

Cis-regulatory element:

a regulatory site encoded in DNA that recruits trans-acting factors to affect the transcriptional activity of a gene in cis. The best studied cis-regulatory elements are promoters, located proximally to transcription start sites, and enhancers, which are located distal to the promoter region and act in lineage-specific patterns.

dTAG:

a ligand-inducible degradation system for rapid and reversible control of protein levels in mammalian cells. After tagging a POI with FKBP12F36V, treatment with a small molecule, dTAG-13, induces association of the POI-FKBP12F36V to CRL4CRBN, resulting in degradation of the POI.

IMiD:

thalidomide, lenalidomide, pomalidomide, and related phthalimide analogs are termed “immunomodulatory imid drugs” or IMiDs. These molecules bind to Cereblon (CRBN), the substrate receptor for the CRL4CRBN E3 ubiquitin ligase complex.

Ligandability:

the ability of a protein to support high-affinity interactions with small molecules. This term is similar to druggability, but it does not presume a functional effect of ligand-protein binding.

RING domain:

a protein fold often, but not always, possessing intrinsic E3 activity.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Bradner JE et al. (2017) Transcriptional Addiction in Cancer. Cell 168, 629–643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Zentner GE and Henikoff S (2013) Regulation of nucleosome dynamics by histone modifications. Nature Structural & Molecular Biology 20, 259–266 [DOI] [PubMed] [Google Scholar]
  • 3.Clapier CR and Cairns BR (2009) The Biology of Chromatin Remodeling Complexes. Annu. Rev. Biochem 78, 273–304 [DOI] [PubMed] [Google Scholar]
  • 4.Kadoch C et al. (2017) Dynamics of BAF-Polycomb complex opposition on heterochromatin in normal and oncogenic states. Nat. Genet 49, 213–222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kaelin WG (2017) Common pitfalls in preclinical cancer target validation. Nat Rev Cancer 17, 425–440 [DOI] [PubMed] [Google Scholar]
  • 6.Weinstein IB (2002) Cancer. Addiction to oncogenes--the Achilles heal of cancer. Science 297, 63–64 [DOI] [PubMed] [Google Scholar]
  • 7.Luo J et al. (2009) Principles of Cancer Therapy: Oncogene and Non-oncogene Addiction. Cell 136, 823–837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ferguson FM and Gray NS (2018) Kinase inhibitors: the road ahead. Nat Rev Drug Discov 17, 353–377 [DOI] [PubMed] [Google Scholar]
  • 9.Lu H et al. (2015) Compensatory induction of MYC expression by sustained CDK9 inhibition via a BRD4-dependent mechanism. Elife 4, e06535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Barsanti PA et al. Novartis AG Pyridine and pyrazine derivatives as protein kinase modulators, WO 2011/012661 A1.2010, [Google Scholar]
  • 11.Zhang H et al. (2018) Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer. Cell 175, 1244–1258. e26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Olson CM et al. (2018) Pharmacological perturbation of CDK9 using selective CDK9 inhibition or degradation. Nat Chem Biol 14, 163–170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chao SH and Price DH (2001) Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo. Journal of Biological Chemistry 276, 31793–31799 [DOI] [PubMed] [Google Scholar]
  • 14.Henriques T et al. (2013) Stable pausing by RNA polymerase II provides an opportunity to target and integrate regulatory signals. Molecular Cell 52, 517–528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Winter GE et al. (2017) BET Bromodomain Proteins Function as Master Transcription Elongation Factors Independent of CDK9 Recruitment. Molecular Cell 67, 5–18.e19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Filippakopoulos P et al. (2010) Selective inhibition of BET bromodomains. Nature 468, 1067–1073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Stathis A et al. (2016) Clinical Response of Carcinomas Harboring the BRD4-NUT Oncoprotein to the Targeted Bromodomain Inhibitor OTX015/MK-8628. Cancer Discov 6, 492–500 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lewin J et al. (2018) Phase Ib Trial With Birabresib, a Small-Molecule Inhibitor of Bromodomain and Extraterminal Proteins, in Patients With Selected Advanced Solid Tumors. J. Clin. Oncol DOI: 10.1200/JCO.2018.78.2292 [DOI] [PubMed] [Google Scholar]
  • 19.Zuber J 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]
  • 20.Delmore JE 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]
  • 21.Ott CJ et al. (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]
  • 22.Dawson MA et al. (2011) Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Berthon C et al. (2016) Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol 3, e186–95 [DOI] [PubMed] [Google Scholar]
  • 24.Amorim S et al. (2016) Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol 3, e196–204 [DOI] [PubMed] [Google Scholar]
  • 25.Stathis A and Bertoni F (2018) BET Proteins as Targets for Anticancer Treatment. Cancer Discov 8, 24–36 [DOI] [PubMed] [Google Scholar]
  • 26.Lovén J et al. (2013) Selective Inhibition of Tumor Oncogenes by Disruption of Super-Enhancers. Cell 153, 320–334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Whyte WA et al. (2013) Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cromm PM and Crews CM (2017) Targeted Protein Degradation: from Chemical Biology to Drug Discovery. Cell Chem Biol 24, 1181–1190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Winter GE et al. (2015) Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Jang MK et al. (2005) The Bromodomain Protein Brd4 Is a Positive Regulatory Component of P-TEFb and Stimulates RNA Polymerase II-Dependent Transcription. Molecular Cell 19, 523–534 [DOI] [PubMed] [Google Scholar]
  • 31.Natsume T et al. (2016) Rapid Protein Depletion in Human Cells by Auxin- Inducible Degron Tagging with Short Homology Donors. Cell Reports 15, 210–218 [DOI] [PubMed] [Google Scholar]
  • 32.Muhar M et al. (2018) SLAM-seq defines direct gene-regulatory functions of the BRD4-MYC axis. Science 360, 800–805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zengerle M et al. (2015) Selective Small Molecule Induced Degradation of the BET Bromodomain Protein BRD4. ACS Chem. Biol 10, 1770–1777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gadd MS et al. (2017) Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat Chem Biol 26, 484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nowak RP et al. (2018) Plasticity in binding confers selectivity in ligand-induced protein degradation. Nat Chem Biol 54, 9659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Greenwald RJ et al. (2004) E mu-BRD2 transgenic mice develop B-cell lymphoma and leukemia. Blood 103, 1475–1484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Shu S et al. (2016) Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature 529, 413–417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ott CJ et al. (2018) Enhancer Architecture and Essential Core Regulatory Circuitry of Chronic Lymphocytic Leukemia. Cancer Cell 34, 982–995.e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rathert P et al. (2015) Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature 525, 543–547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Cheung K et al. (2017) BET N-terminal bromodomain inhibition selectively blocks Th17 cell differentiation and ameliorates colitis in mice. Proc. Natl. Acad. Sci. U.S.A 114, 2952–2957 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hsu SC et al. (2017) The BET Protein BRD2 Cooperates with CTCF to Enforce Transcriptional and Architectural Boundaries. Molecular Cell 66, 102–116.e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Raina K et al. (2016) PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci USA 113, 7124–7129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bai L et al. (2017) Targeted Degradation of BET Proteins in Triple-Negative Breast Cancer. Cancer Research 77, 2476–2487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Qin C et al. (2018) Discovery of QCA570 as an Exceptionally Potent and Efficacious Proteolysis Targeting Chimera (PROTAC) Degrader of the Bromodomain and Extra-Terminal (BET) Proteins Capable of Inducing Complete and Durable Tumor Regression. J. Med. Chem 61,6685–6704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fong CY et al. (2015) BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Erb MA et al. (2017) Transcription control by the ENL YEATS domain in acute leukaemia. Nature 543, 270–274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Luo Z et al. (2012) The super elongation complex (SEC) family in transcriptional control. Nat. Rev. Mol. Cell Biol 13, 543–547 [DOI] [PubMed] [Google Scholar]
  • 48.Li Y et al. (2014) AF9 YEATS Domain Links Histone Acetylation to DOT1L-Mediated H3K79 Methylation. Cell 159, 558–571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Andrews FH et al. (2016) The Taf14 YEATS domain is a reader of histone crotonylation. Nat Chem Biol 12, 396–398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li Y et al. (2016) Molecular Coupling of Histone Crotonylation and Active Transcription by AF9 YEATS Domain. Molecular Cell 62, 181–193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wan L et al. (2017) ENL links histone acetylation to oncogenic gene expression in acute myeloid leukaemia. Nature 543, 265–269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vangamudi B et al. (2015) The SMARCA2/4 ATPase Domain Surpasses the Bromodomain as a Drug Target in SWI/SNF-Mutant Cancers: Insights from cDNA Rescue and PFI-3 Inhibitor Studies. Cancer Research 75, 3865–3878 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Shi J et al. (2015) Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat Biotechnol 33, 661–667 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Moustakim M et al. (2018) Discovery of an MLLT1/3 YEATS Domain Chemical Probe. Angew. Chem. Int. Ed. Engl 57, 16302–16307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Nabet B et al. (2018) The dTAG system for immediate and target-specific protein degradation. Nat Chem Biol 14, 431–441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mayor-Ruiz C and Winter GE (2019) Identification and characterization of cancer vulnerabilities via targeted protein degradation. Drug Discovery Today: Technologies DOI: 10.1016/j.ddtec.2018.12.003 [DOI] [PubMed] [Google Scholar]
  • 57.Clegg MA et al. (2019) Advancements in the Development of non-BET Bromodomain Chemical Probes. ChemMedChem 429, 457–24 [DOI] [PubMed] [Google Scholar]
  • 58.Wilson BG et al. (2014) Residual complexes containing SMARCA2 (BRM) underlie the oncogenic drive of SMARCA4 (BRG1) mutation. Molecular and Cellular Biology 34, 1136–1144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hoffman GR et al. (2014) Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers. Proc. Natl. Acad. Sci. U.S.A 111, 3128–3133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Oike T et al. (2013) A synthetic lethality-based strategy to treat cancers harboring a genetic deficiency in the chromatin remodeling factor BRG1. Cancer Research 73, 5508–5518 [DOI] [PubMed] [Google Scholar]
  • 61.Fedorov O et al. (2015) Selective targeting of the BRG/PB1 bromodomains impairs embryonic and trophoblast stem cell maintenance. Sci Adv 1, e1500723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gerstenberger BS et al. (2016) Identification of a Chemical Probe for Family VIII Bromodomains through Optimization of a Fragment Hit. J. Med. Chem 59, 4800–4811 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Armstrong JA et al. (2002) The Drosophila BRM complex facilitates global transcription by RNA polymerase II. EMBO J 21, 5245–5254 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Papillon JPN et al. (2018) Discovery of Orally Active Inhibitors of Brahma Homolog (BRM)/SMARCA2 ATPase Activity for the Treatment of Brahma Related Gene 1 (BRG1)/SMARCA4-Mutant Cancers. J. Med. Chem 61, 10155–10172 [DOI] [PubMed] [Google Scholar]
  • 65.Michel BC et al. (2018) A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation. Nat Cell Biol 154, 490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gatchalian J et al. (2018) A non-canonical BRD9-containing BAF chromatin remodeling complex regulates naive pluripotency in mouse embryonic stem cells. Nat Commun 9, 5139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hohmann AF et al. (2016) Sensitivity and engineered resistance of myeloid leukemia cells to BRD9 inhibition. Nat Chem Biol 12, 672–679 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Remillard D et al. (2017) Degradation of the BAF Complex Factor BRD9 by Heterobifunctional Ligands. Angew. Chem. Int. Ed. Engl 56, 5738–5743 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zoppi V et al. (2018) Iterative design and optimization of initially inactive Proteolysis Targeting Chimeras (PROTACs) identify VZ185 as a potent, fast and selective von Hippel-Lindau (VHL)-based dual degrader probe of BRD9 and BRD7. J. Med. Chem 62, 699–726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Brien GL et al. (2018) Targeted degradation of BRD9 reverses oncogenic gene expression in synovial sarcoma. Elife 7, 3892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pasqualucci L et al. (2011) Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 471, 189–195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wang L et al. (2011) The leukemogenicity of AML1-ETO is dependent on site-specific lysine acetylation. Science 333, 765–769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ogiwara H et al. (2016) Targeting p300 Addiction in CBP-Deficient Cancers Causes Synthetic Lethality by Apoptotic Cell Death due to Abrogation of MYC Expression. Cancer Discov 6, 430–445 [DOI] [PubMed] [Google Scholar]
  • 74.Hay DA et al. (2014) Discovery and optimization of small-molecule ligands for the CBP/p300 bromodomains. J. Am. Chem. Soc 136, 9308–9319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Picaud S et al. (2015) Generation of a Selective Small Molecule Inhibitor of the CBP/p300 Bromodomain for Leukemia Therapy. Cancer Research 75, 5106–5119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Crawford TD et al. (2016) Discovery of a Potent and Selective in Vivo Probe (GNE-272) for the Bromodomains of CBP/EP300. J. Med. Chem 59, 10549–10563 [DOI] [PubMed] [Google Scholar]
  • 77.Taylor AM et al. (2016) Fragment-Based Discovery of a Selective and Cell-Active Benzodiazepinone CBP/EP300 Bromodomain Inhibitor (CPI-637). ACS Med Chem Lett 7, 531–536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Romero FA et al. (2017) GNE-781, A Highly Advanced Potent and Selective Bromodomain Inhibitor of Cyclic Adenosine Monophosphate Response Element Binding Protein, Binding Protein (CBP). J. Med. Chem 60, 9162–9183 [DOI] [PubMed] [Google Scholar]
  • 79.Lasko LM et al. (2017) Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 550, 128–132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Michaelides MR et al. (2018) Discovery of Spiro Oxazolidinediones as Selective, Orally Bioavailable Inhibitors of p300/CBP Histone Acetyltransferases. ACS Med Chem Lett 9, 28–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dahlin JL et al. (2017) Assay interference and off-target liabilities of reported histone acetyltransferase inhibitors. Nat Commun 8, 1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jin L et al. (2017) Therapeutic Targeting of the CBP/p300 Bromodomain Blocks the Growth of Castration-Resistant Prostate Cancer. Cancer Research 77, 5564–5575 [DOI] [PubMed] [Google Scholar]
  • 83.Raisner R et al. (2018) Enhancer Activity Requires CBP/P300 Bromodomain-Dependent Histone H3K27 Acetylation. Cell Reports 24, 1722–1729 [DOI] [PubMed] [Google Scholar]
  • 84.Weinert BT et al. (2018) Time-Resolved Analysis Reveals Rapid Dynamics and Broad Scope of the CBP/p300 Acetylome. Cell 174, 231–244.e12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Baell JB et al. (2018) Inhibitors of histone acetyltransferases KAT6A/B induce senescence and arrest tumour growth. Nature 560, 253–257 [DOI] [PubMed] [Google Scholar]
  • 86.Tsai W-W et al. (2010) TRIM24 links a non-canonical histone signature to breast cancer. Nature 468, 927–932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Groner AC et al. (2016) TRIM24 Is an Oncogenic Transcriptional Activator in Prostate Cancer. Cancer Cell 29, 846–858 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bennett J et al. (2016) Discovery of a Chemical Tool Inhibitor Targeting the Bromodomains of TRIM24 and BRPF. J. Med. Chem 59, 1642–1647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Palmer WS et al. (2016) Structure-Guided Design of IACS-9571, a Selective High-Affinity Dual TRIM24-BRPF1 Bromodomain Inhibitor. J. Med. Chem 59, 1440–1454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhan Y et al. (2015) Development of novel cellular histone-binding and chromatin-displacement assays for bromodomain drug discovery. Epigenetics & Chromatin 8, 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Gechijian LN et al. (2018) Functional TRIM24 degrader via conjugation of ineffectual bromodomain and VHL ligands. Nat Chem Biol 168, 629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kim KH and Roberts CWM (2016) Targeting EZH2 in cancer. Nat Med 22, 128–134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Italiano A et al. (2018) Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study. Lancet Oncol. 19, 649–659 [DOI] [PubMed] [Google Scholar]
  • 94.Kim KH et al. (2015) SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med 21, 1491–1496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.He Y et al. (2017) The EED protein-protein interaction inhibitor A-395 inactivates the PRC2 complex. Nat Chem Biol 13, 389–395 [DOI] [PubMed] [Google Scholar]
  • 96.Qi W et al. (2017) An allosteric PRC2 inhibitor targeting the H3K27me3 binding pocket of EED. Nat Chem Biol 13, 381–388 [DOI] [PubMed] [Google Scholar]
  • 97.Milosevich N et al. (2016) Selective Inhibition of CBX6: A Methyllysine Reader Protein in the Polycomb Family. ACS Med Chem Lett 7, 139–144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Stuckey JI et al. (2016) A cellular chemical probe targeting the chromodomains of Polycomb repressive complex 1. Nat Chem Biol 12, 180–187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Okada Y et al. (2005) hDOT1L Links Histone Methylation to Leukemogenesis. Cell 121, 167–178 [DOI] [PubMed] [Google Scholar]
  • 100.Bernt KM et al. (2011) MLL-Rearranged Leukemia Is Dependent on Aberrant H3K79 Methylation by DOT1L. Cancer Cell 20, 66–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Daigle SR 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]
  • 102.Daigle SR et al. (2013) Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122,1017–1025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Stein EM et al. (2018) The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 131, blood–2017–12–818948–2669 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Borkin D et al. (2015) Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell 27, 589–602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kühn MWM et al. (2016) Targeting Chromatin Regulators Inhibits Leukemogenic Gene Expression in NPM1 Mutant Leukemia. Cancer Discov 6, 1166–1181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Zhu L et al. (2016) ASH1L Links Histone H3 Lysine 36 Dimethylation to MLL Leukemia. Cancer Discov 6, 770–783 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Grembecka J. Ash1l inhibitors and methods of treatment therewith, WO 2017/197240 A1. The Regents of the University of Michigan; [Google Scholar]
  • 108.Jaffe JD et al. (2013) Global chromatin profiling reveals NSD2 mutations in pediatric acute lymphoblastic leukemia. Nat. Genet 45, 1386–1391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Coussens NP et al. (2018) High-throughput screening with nucleosome substrate identifies small-molecule inhibitors of the human histone lysine methyltransferase NSD2. J. Biol. Chem 293, 13750–13765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Sharma SV et al. (2010) A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Spencer SL et al. (2009) Non-genetic origins of cell-to-cell variability in TRAIL-induced apoptosis. Nature 459, 428–432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Roesch A et al. (2013) Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 23, 811–825 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Vinogradova M et al. (2016) An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat Chem Biol 12, 531–538 [DOI] [PubMed] [Google Scholar]
  • 114.Liau BB et al. (2017) Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. Cell Stem Cell 20, 233–246.e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Liang J et al. (2017) From a novel HTS hit to potent, selective, and orally bioavailable KDM5 inhibitors. Bioorganic & Medicinal Chemistry Letters 27, 2974–2981 [DOI] [PubMed] [Google Scholar]
  • 116.Nie Z et al. (2018) Structure-based design and discovery of potent and selective KDM5 inhibitors. Bioorganic & Medicinal Chemistry Letters 28, 1490–1494 [DOI] [PubMed] [Google Scholar]
  • 117.Tsherniak A et al. (2017) Defining a Cancer Dependency Map. Cell 170, 564–576.e16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.McDonald ER et al. (2017) Project DRIVE: A Compendium of Cancer Dependencies and Synthetic Lethal Relationships Uncovered by Large-Scale, Deep RNAi Screening. Cell 170, 577–592.e10 [DOI] [PubMed] [Google Scholar]
  • 119.Meyers RM et al. (2017) Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells. Nat. Genet 49, 1779–1784 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Miller TE et al. (2017) Transcription elongation factors represent in vivo cancer dependencies in glioblastoma. Nature 547, 355–359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Backus KM et al. (2016) Proteome-wide covalent ligand discovery in native biological systems. Nature 534, 570–574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Parker CG et al. (2017) Ligand and Target Discovery by Fragment-Based Screening in Human Cells. Cell 168, 527–541.e29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bar-Peled L et al. (2017) Chemical Proteomics Identifies Druggable Vulnerabilities in a Genetically Defined Cancer. Cell 171, 696–709.e23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wang Y et al. (2019) Expedited mapping of the ligandable proteome using fully functionalized enantiomeric probe pairs. ChemRxiv DOI: 10.26434/chemrxiv.7764638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hacker SM et al. (2017) Global profiling of lysine reactivity and ligandability in the human proteome. Nature Chemistry 9, 1181–1190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bondeson DP et al. (2018) Lessons in PROTAC Design from Selective Degradation with a Promiscuous Warhead. Cell Chem Biol 25, 78–87.e5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Huang H-T et al. (2018) A Chemoproteomic Approach to Query the Degradable Kinome Using a Multi-kinase Degrader. Cell Chem Biol 25, 88–99.e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Brand M et al. (2019) Homolog-Selective Degradation as a Strategy to Probe the Function of CDK6 in AML. Cell Chem Biol 26, 300–306.e9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Smith BE et al. (2019) Differential PROTAC substrate specificity dictated by orientation of recruited E3 ligase. Nat Commun 10, 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Han T et al. (2017) Anticancer sulfonamides target splicing by inducing RBM39 degradation via recruitment to DCAF15. Science 356 (2017), Article eaal3755 [DOI] [PubMed] [Google Scholar]
  • 131.Uehara T et al. (2017) Selective degradation of splicing factor CAPERα by anticancer sulfonamides. Nat Chem Biol 13, 675–680 [DOI] [PubMed] [Google Scholar]
  • 132.Spradlin JN et al. (2018) Harnessing the Anti-Cancer Natural Product Nimbolide for Targeted Protein Degradation. bioRxiv DOI. 10.1101/436998 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Ward CC et al. (2018) Covalent Ligand Screening Uncovers a RNF4 E3 Ligase Recruiter for Targeted Protein Degradation Applications. bioRxiv DOI. 10.1101/439125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Zhang X et al. (2018) Electrophilic PROTACs that degrade nuclear proteins by engaging DCAF16. bioRxiv DOI: 10.1101/443804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Pan D et al. (2018) A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 359, 770–775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Miao D et al. (2018) Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 359, 801–806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Sheng W et al. (2018) LSD1 Ablation Stimulates Anti-tumor Immunity and Enables Checkpoint Blockade. Cell 174, 549–563.e19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Millard CJ et al. (2017) Targeting Class I Histone Deacetylases in a “Complex” Environment. Trends in Pharmacological Sciences 38, 363–377 [DOI] [PubMed] [Google Scholar]
  • 139.Arrowsmith CH et al. (2015) The promise and peril of chemical probes. Nat Chem Biol 11, 536–541 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Neigeborn L and Carlson M (1984) Genes affecting the regulation of SUC2 gene expression by glucose repression in Saccharomyces cerevisiae. Genetics 108, 845–858 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Stern M et al. (1984) Five SWI genes are required for expression of the HO gene in yeast. J. Mol. Biol 178, 853–868 [DOI] [PubMed] [Google Scholar]
  • 142.Peterson CL and Herskowitz I (1992) Characterization of the yeast SWI1, SWI2, and SWI3 genes, which encode a global activator of transcription. Cell 68, 573–583 [DOI] [PubMed] [Google Scholar]
  • 143.Pulice JL and Kadoch C (2016) Composition and Function of Mammalian SWI/SNF Chromatin Remodeling Complexes in Human Disease. Cold Spring Harb. Symp. Quant. Biol 81, 53–60 [DOI] [PubMed] [Google Scholar]
  • 144.Mashtalir N et al. (2018) Modular Organization and Assembly of SWI/SNF Family Chromatin Remodeling Complexes. Cell 175, 1272–1288.e20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kadoch C et al. (2013) Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet 45, 592–601 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hohmann AF and Vakoc CR (2014) A rationale to target the SWI/SNF complex for cancer therapy. Trends Genet. 30, 356–363 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES