Abstract
Chromatin serves to organize and compact the genome but also functions as a signaling hub for the dynamic regulation of transcriptional programs that control cell type specification. The historical discovery that several pro-differentiation anti-cancer agents target chromatin regulatory enzymes buoyed early interest in developing drugs that modulate chromatin structure and function. Chromatin-based drug discovery has since flourished alongside major advances in discovery chemistry and target selection, producing a rich collection of chemical probes, drugs, and drug candidates targeting chromatin regulatory processes. The substantial growth and maturity of this field over the last several decades provides an opportunity to reflect on the successes and failures associated with translating chromatin regulatory targets into anti-cancer drugs. Taking a target-centric perspective, we discuss the motivation for pursuing specific chromatin regulatory proteins and review the chemistries that enabled small molecule discovery and development. In so doing, we hope to evaluate the strength of these targets, the agents that prosecute them, and the prospects for future efforts in this field.
1. Introduction
Chromatin is a DNA-histone complex that compacts the genome and controls the accessibility of DNA to regulatory factors that act in trans. It is therefore both the template for transcribing DNA into RNA and a signaling hub for gene regulation. The full appreciation that chromatin influences transcription required several decades of research, dating to initial observations that changes in chromatin structure coincide with changes in gene activity.1 Over 30 years later, when histone acetyltransferase (HAT) and histone deacetylase (HDAC) enzymes were first isolated, cloned, and sequenced, they were found to share the identity of known transcriptional activators and repressors, providing the first direct link between chromatin modifiers and transcription.2–7 The disciplines of synthetic chemistry, pharmacology, and chemical biology were featured prominently in this seminal work, wherein affinity purification was used to identify the cellular targets of the natural product, trapoxin, an agent known to induce hyperacetylation. The modern field of chemical biology is nicely reflected in this early study and through its foundational impact on the fields of chromatin and chemical biology, it has shaped, considerably, the present state of chromatin-directed drug discovery. Nearing another 30 years since this report, we will consider how chromatin regulation has matured as a target for small-molecule drugs, focusing on the development of anti-cancer therapeutics and the evolution of target selection. We will highlight the foundational discoveries that made specific target classes available to discovery chemistry and provide an assessment on the current outlook for translating these targets and agents into therapeutics. Our treatment of this subject will be mostly limited to compounds targeting chromatin regulatory factors wherein their potential anti-cancer effects are thought to be primarily and directly related to gene transcription. For example, we will not discuss the DNA repair protein PARP1, even though it is strongly associated with transcriptional biology, as the anti-cancer effects of PARP inhibitors in the clinic can be confidently ascribed to the modulation DNA repair [28676700]. Similarly, the inhibition of metabolic proteins that indirectly affect chromatin regulators, like IDH1/2 [cite], will not be discussed.
1.1. Chromatin biology background
Chromatin is a nucleoprotein polymer consisting of DNA wrapped around histones to form repeating nucleosome structures. Generally, a nucleosome is formed by ~1.6 turns of DNA (~147 base pairs) around a core histone octamer, forming a left-handed superhelix.8 This octamer is produced by two copies each of the histone proteins H2A, H2B, H3, and H4, but alternative assemblies that feature histone variants or other stoichiometries also exist.9 Linker histone H1 binds to DNA between and adjacent to nucleosomes, assisting with local and higher-order chromatin compaction [Cite]. Nucleosomes are post-translationally modified in abundance, most frequently on the unstructured amino (N)-terminal tails of histone subunits but also on their globular carboxy (C)-terminal cores (Figure 1a). These modifications can influence transcriptionally signaling by altering the accessibility of DNA sequences through chromatin compaction or by binding directly to regulatory factors that act in trans.10–13
Figure 1.

Chromatin regulation and transcriptional signaling. (a) Chromatin regulators and chromatin-dependent transcriptional signaling. (b) Control of enhancer accessibility and activation.
Transcription begins with the recruitment of RNA polymerase (Pol) II and other general transcriptional machinery to gene promoters (located just upstream of the transcription start site), followed by an initial transcription of ~20–100 bp before Pol II reaches a regulated state of stable pausing.14,15 Release from this promoter-proximal pause is controlled by P-TEFb (positive elongation factor b), a heterodimeric complex of CDK9 (cyclin dependent kinase 9) and cyclin T, which phosphorylates RNA Pol II and other negative regulatory factors, such as DSIF and NELF, allowing for productive transcriptional elongation (Figure 1a).16 During elongation, Pol II requires accessory factors to traverse through tightly bound nucleosome structures and maintain histone post translational modifications until termination sites are reached and the transcription cycle is completed.17–19
The production of specialized cell types through organismal development is controlled by the spatiotemporally regulated transcription of specific subsets of genes encoded in the genome.20 Gene-selective transcriptional processes are largely controlled by enhancer sequences located in cis.21 These cis-regulatory elements are densely populated with transcription factor (TF)-binding motifs and are most frequently located distal to the gene promoter, often encoded 10 to 100 kb away or further. Enhancer selection is thought to be controlled by a class of pioneering DNA-binding transcription factors (pioneer TFs) that possess a specialized ability to bind their motifs even when embedded in nucleosomes (Figure 1b).22,23 After binding these occluded motifs within permissive regions of chromatin, pioneering TFs then function to enforce local regions of increased DNA accessibility (Figure 1b). Most TFs can only bind their cognate motifs in the context of free DNA, so this pioneering function allows for additional TFs to bind at enhancers and create an active regulatory element (Figure 1b). Despite being distally encoded, these enhancer elements become physically juxtaposed with the promoter of their target genes in three-dimensional space. This too is highly regulated by chromatin, as the signaling from enhancer-bound TFs to promoter-bound RNA Pol II involves the concerted activity of transcription co-regulators, many of which act directly by binding to chromatin or modifying its structure and function.
Two studies reported in 1996 played an outsized role in establishing that histone modifications could impact transcription.2,3 Working to clone and sequence genes encoding histone acetyltransferases (HATs), Professor Allis and colleagues discovered that a HAT from Tetrahymena, a unicellular eukaryote, was highly conserved with the yeast transcriptional co-activator, Gcn5.2 Contemporaneously, Professor Schreiber and colleagues reported the molecular target of the natural product, trapoxin, which was correctly presumed to be a direct-acting inhibitor of histone deacetylases (HDACs) based on structural similarity between its aliphatic epoxyketone and an acetylated lysine side chain.3 Once isolated through an affinity matrix purification, it was discovered that mammalian HDACs targeted by trapoxin were conserved with the yeast transcriptional co-repressor, Rpd3p. These two studies provided the first direct links between chromatin modifiers and transcriptional regulation, greatly accelerating interest in the modern field of chromatin biology.
Today, we know many distinct classes of chromatin modifiers that dynamically regulate the state of post-translational modifications (PTMs) on histones and other chromatin-bound proteins (Figure 1a).24 Acetylation is joined by methylation, phosphorylation, ubiquitination, ADP ribosylation, and many other modifications of varying abundance. Site-specific histone modifications are often associated with discrete genomic features and transcriptional regulatory states. For example, nucleosomes within the promoter regions of actively transcribed genes are enriched with the trimethylation of lysine 4 of histone H3 (H3K4me3) [cite], active enhancers are enriched with H3K27ac [cite], and repressed enhancers are marked by H3K27me3 [cite]. Histone modifications can serve many different purposes but can often be understood to function through two main mechanisms: (i) altering chromatin structure to make DNA more or less accessible and (ii) serving as binding sites that are selectively recognized by chromatin reader domains (Figure 1a). In many cases, these mechanisms are mutually dependent, as PTMs can have intrinsic effects on protein-DNA interactions but can also serve to recruit regulatory factors that alter chromatin compaction and accessibility by acting in trans.18,25
Chromatin reader domains are found in scaffolding proteins that recruit other regulatory factors to local regions of the genome and can also function as modules that allosterically regulate enzymatic activities encoded in the same protein or protein complex.26 They are evolved to selectively bind chemical modifications on specific side chains of a protein (often histone tails), enabling combinatorial control in the recruitment of chromatin readers, depending on which modifications are present locally in the genome. For example, lysine acetylation is recognized by bromodomains, DPF (double PHD finger) domains, and YEATS (Yaf9, ENL, AF9, Taf14, Sas5) domains.27–30 Lysine methylation is bound by chromodomains, WD40 domains, Tudor domains, and PWWP domains, among others.31–39 Like chromatin modifiers, chromatin reader domains have often been the subject of targeted drug discovery efforts, resulting in many high-quality chemical probes and several investigational new drugs.40
1.2. Genetic dependencies and common essential genes
Unlike many of the targets we will discuss in this review, pharmacological tools have been known for HDACs for decades.41 Chemical tools for HDACs are now widely available, representing an extraordinary range of selectivity and potency profiles, and have contributed enormously to the biological study of chromatin and transcription.42 Given their long history, extensive study, and chemical diversity, HDACs provide a particularly instructive example for the considerations that are relevant to targeting chromatin regulatory processes with small molecules. Much of the attention given to HDAC inhibitors can be attributed to the early observation that HDAC inhibition provokes terminal differentiation of cancer cells in tissue culture.43–47 This work ultimately enabled the development of 4 FDA-approved drugs targeting HDACs for cancer therapy. However, HDAC inhibitors have failed in many clinical trials due to dose-limiting toxicity, which is equally instructive when considering the selection of targets for drug discovery. Outside of a few notable indications, the therapeutic utility of HDAC inhibitors has been limited by on-target toxicity, some of which can be attributed to the broad-spectrum inhibition of many HDAC isoforms by these agents. Another issue with HDAC inhibitors is that several HDAC isoforms are encoded by common essential genes – genes that are required for the survival of most or all cell types. Pharmacological inhibition is often not equivalent to genetic knockout, and there are important examples of common essential proteins that can be targeted with an acceptable therapeutic index, as recently highlighted [cite]. For example, the essential genes HMGCR and PTGS2 are the targets of statins and aspirin, respectively [cite]. However, in many instances, the intrinsic therapeutic window for common essential targets is narrowed by the effects of inhibiting these proteins in normal tissues.48
The prevailing thought is that more effective therapeutics can be brought forth by targeting cancer-specific dependencies – genes that are selectively required for the survival of tumor cells versus normal tissues as a result of defined genetic alterations or other changes in cell state.49 Thanks to consortia-led genetic screening efforts, which have used pooled CRISPR/Cas9 loss-of-function screens to catalog genetic dependencies in thousands of cancer cell lines (see the cancer dependency map, or DepMap, at depmap.org50–52), it is now trivial to determine if a potential drug target is encoded by a common essential gene and would therefore likely be required by most normal tissues as well. DepMap uses the algorithm Chronos to transform CRISPR screening datasets into “gene effect” scores that report the impact of gene disruption on cell fitness [cite]. It affixes the median gene effect score of essential genes to −1 and non-essential genes to 0 such that the magnitude of effect can be rapidly interpreted. Since gene effect scores are calculated across thousands of cancer cell lines the distribution of scores can be used to quickly distinguish between common essential genes (those with a distribution centered on –1) and selective dependencies (those with a distribution centered near 0 with a fraction of cell lines with scores approaching –1). For selective dependencies, these score can be integrated with genetic and phenotypic features to identify the underlying cause of the dependency, further probe the biological pathway, and/or nominate biomarkers of drug response.
In chromatin biology, the promise of targeting context-specific cancer dependencies, as opposed to common essential genes, can be seen in the Food and Drug Administration (FDA) approval of EZH2 inhibitors for the treatment of EZH2-mutant lymphomas and SWI/SNF-mutant sarcomas, as EZH2 is not a common essential gene but is a selective dependency in these tumors (Figure 2a,b).53 Likewise, the menin inhibitor revumenib is now approved for the treatment of acute leukemia caused by chromosomal rearrangements involving the MLL gene. Menin, which is not a common essential gene, is selectively required in these and other genetically defined acute leukemias.54–60 In contrast to the favorable therapeutic window afforded by these targets, common essential gene products can be difficult to drug due to their importance in normal tissues. For example, BET bromodomain inhibitors and CDK9 inhibitors, which target the products of common essential genes (Figure 2c), have shown less promise in clinical trials. We will discuss these fundamental differences in target biology below, beginning our review, part of the thematic Special Issue on Drugging the Undruggable, with a discussion of HDACs, highlighting many of the strengths and challenges associated with targeting chromatin regulatory processes.
Figure 2.

Targeting context-specific cancer dependencies versus common essential genes. (a) Chemical structures of FDA-approved drugs targeting the chromatin regulatory enzymes, EZH2 and menin-MLL. (b) Histogram of DepMap gene effect scores for EZH2 and MEN1 (encodes menin protein) across 1150 cancer cell lines, as determined using pooled CRISPR/Cas9-based gene essentiality screens. Gene effect scores, which were downloaded from the DepMap CRISPR Public 24Q2 dataset hosted at DepMap.org, were calculated using Chronos, an algorithm that affixes the median score of essential genes to −1 and non-essential genes to 0. (c) Gene effect scores for the common essential genes BRD4 and CDK9.
2. Targeting essential proteins
2.1. Histone deacetylases
Zinc-dependent HDACs are categorized into four phylogenetic groups: Class I (HDAC1/2/3/8), Class IIa (HDAC4/5/7/9), Class IIb (HDAC6/10), and Class IV (HDAC11) HDACs [cite]. Class I HDACs are nuclear localized and principally associated with chromatin regulation, whereas class IIa/b enzymes can localize to the cytoplasm where they show diverse functions [cite]. HDAC inhibitors were used to discover the first HDAC enzymes and they have remained essential tools for studying HDAC biology [cite].3,61 In the 1990s, the study of natural product HDAC inhibitors and the serendipitous development of synthetic HDAC inhibitors followed fascinatingly parallel paths that were eventually linked conceptually by their shared pro-differentiation effects. The first synthetic HDAC inhibitor, SAHA (suberoylanilide hydroxamic acid), originated from efforts to improve on the pro-differentiation activity of dimethyl sulfoxide (DMSO), first described to induce cellular differentiation in the 1970s.62 The unlikely path of medicinal chemistry that led from DMSO to SAHA was elegantly recounted in a past perspective by Marks and Breslow.41 In it, they highlighted the serendipity that contributed to the discovery of SAHA,62–66 which relied on the iterative synthesis of polar compounds meant to mimic the properties of DMSO (Figure 3a). While both DMSO and SAHA inhibit HDAC enzymes, some intermediate compounds that led to the discovery of SAHA, like hexamethylene bisacetamide (HMBA), are not HDAC inhibitors. Moreover, only after the initial discovery of SAHA as an optimized pro-differentiation agent was it later realized that SAHA functions through the inhibition of HDACs. This realization was triggered by the structural similarity between SAHA and trichostatin A (TSA), a natural product that also harbors a hydroxamic acid and was known to inhibit HDACs (Figure 3a).46 SAHA is now approved for the treatment of cutaneous T-cell lymphoma (CTCL) under the generic name, vorinostat.67
Figure 3.

Common features of HDAC inhibitors. (a) Chemical structure of prototypical hydroxamic acid HDAC inhibitors, indicating cap, linker, and chelating functionalities on SAHA. (b-d) Chemical structures of prototypical macrocyclic (b), benzamide (c), and 4’-arylbenzamide HDAC inhibitors (d).
2.1.1. Overview of HDAC inhibitors
While the mechanism of HDAC inhibition by DMSO is not well understood, SAHA and TSA function by inserting a hydroxamic acid moiety deep into the HDAC active site where it chelates zinc (Figure 3a). These compounds share a cap-linker-chelate arrangement that combines a Zn-binding group, a surface recognition “cap”, and a linker that traverses through the HDAC active-site channel to join the two, which mimics the insertion of lysine side chains that HDACs have evolved to bind (Figure 3a). The majority of HDAC inhibitors share these features and fall into 3 major structural classes – hydroxamic acids, macrocyclic tetrapeptides, and ortho-aminoanilides (also known as benzamides) (Figure 3b–d). For example, the macrocycle, FK228, features an intramolecular disulfide that is reduced in the cellular environment to reveal a sulfhydryl that interacts with zinc in the HDAC active site.68,69 The sulfydryl is connected to the cyclictetrapeptide “capping” structure through a hydrocarbon “linker”, conceptually analogous to the attachment of the hydroxamate of SAHA to its anilide capping group. Benzamides, like the prototypical CI-994 and MS-275, bind to zinc through their ortho amine groups, a structure that enabled later agents, the 4’-arylbenzamides, to exploit a deeper internal cavity located beyond the active site zinc (Figure 3c,d).70–72 There are a few notable exceptions to the cap-linker-chelate logic, including HDAC inhibitors without a metal chelating group, which were discovered through efforts to systematically evaluate the structure-activity relationships (SAR) governing isoform-specific HDAC inhibition by macrocyclic tetrapeptide inhibitors,73–75 as well as pharmacophores that enable covalent engagement.76–79
HDACs stand out from other classes of chromatin regulatory proteins for the length of time that small-molecule inhibitors have been available to the research community. The convergence of several serendipitous HDAC inhibitor discoveries not only provided an early indication of the chemical tractability of these enzymes but also created an immediate wealth of structurally distinct chemical tools for biological research. In retrospect, the early discovery of HDAC inhibitors can likely be attributed to the intrinsic ligandability of the HDAC active site and the large effects that HDAC inhibition has on diverse cellular processes. These qualities of HDAC enzymes would, in principle, increase the chances that a pharmacopeia of bioactive small molecules might contain an HDAC inhibitor. Like SAHA, the ortho-aminoanilide class of HDAC inhibitors were also identified serendipitously. The first two compounds of this class, CI-994 and MS-275, were discovered contemporaneously in the 1990s as antiproliferative agents and then later recognized to inhibit HDACs. CI-994 was first used as an anticonvulsant before its antiproliferative activity was recognized in clinical observations and later studied in rodent models of leukemia.80–82 MS-275 was developed out of a synthetic effort to discover agents that overcome cancer models of multidrug resistance.83 Both were later discovered to inhibit HDACs, providing an orthogonal chelating group to target HDACs.
2.1.2. Common essential HDAC isoforms
In the years following the discovery of SAHA and its approval for CTCL, we have witnessed a broad effort to discover small-molecule modulators of chromatin regulatory proteins. Often, the motivation for developing these agents has been the expectation that they would make effective anti-cancer agents. Several striking examples of clinical successes have resulted from this work – for example, the approval of EZH2 and menin inhibitors as anti-cancer drugs. However, the field is now mature enough to offer several instructive counter examples that failed to yield the clinical advances expected from preclinical experimentation. HDAC inhibitors highlight this duality well. Vorinostat, for example, has been tested in hundreds of Phase 1 and Phase 2 clinical trials but only approved for CTCL.48 This can, in part, be attributed to its broad-spectrum inhibition of Zn-dependent HDACs, of which there are 11 encoded in humans.48 However, more selective HDAC inhibitors, such as trapoxin, have been known for as long as SAHA and have still failed to produce more widespread anti-cancer drug approvals, suggesting that improving isoform selectivity is not sufficient to eliminate on-target toxicity, the predominant limitation of these HDAC inhibitors. Consistent with the classification of HDACs by primary sequence similarity,84 isoform selectivity correlates strongly with the HDAC classes.85 However, many class I-selective compounds show less potent inhibition of Class I HDAC8 than Class IIb HDAC6,85 which regulates tubulin acetylation.86 FK228,68,69 a natural product prodrug inhibitor of Class I HDACs is the only class-selective HDAC inhibitor to receive FDA approval – for CTCL and briefly for peripheral T-cell lymphoma (PTCL) before it was withdrawn for failing to meet specified endpoints in a confirmatory Phase 3 trial.87–89 These approvals are joined by two additional broad-spectrum hydroxamate HDAC inhibitors, belinostat (approved for PTCL) and panobinostat (approved for relapsed and refractory multiple myeloma),90,91 demonstrating that isoform-selective HDAC inhibitors have failed to yield major therapeutic breakthroughs beyond what has also been achieved with broad-spectrum HDAC inhibitors.
Focusing on Class I-selective HDAC inhibitors, much of the difficulty in developing these agents into therapeutics can be attributed to the general essentiality of their targets. Among Class I HDACs, HDAC3 is encoded by a common essential gene (Figure 4a). It is ubiquitously expressed across tissues, indispensable in mice for viability, and required for the fitness of all cancer cell lines.48,61,92–96 The latter is considered a strong indication that many untransformed cell types and normal tissues will also depend on the gene for survival. These targets present significant challenges for drug development as on-target toxicity to normal cells can create adverse effects that limit drug efficacy.48 4’-aryl ortho-aminoanilides were previously reported to achieve selectivity for HDAC1/2 over HDAC3 by occupying a unique internal cavity in HDAC1/2 located beyond the resident zinc.71 This structural feature was originally reported to prevent HDAC3 inhibition and preserve HDAC1/2 inhibition but was recently demonstrated to be an artifact of differences in assay design between HDAC1/2 and HDAC3. Most isoform selectivity profiling has used purified protein preparations, but newly developed Cora-Fluor TR-FRET probes97 have enabled the development of ligand displacement assays (FITC-labelled SAHA) that are compatible with crude lysates.98 These assays demonstrated that 4’-aryl benzamide inhibitors, like Cpd-60 (also known as Merck60) bind with high affinity to free, but not complex-bound, HDAC1/2/3. In cells, where HDAC1/2/3 are predominantly complex-bound, these compounds show no selectivity between HDAC1/2/3. HDAC1 and HDAC2 are subunits of several large multiprotein complexes, including CoREST, NuRD, SIN3A, and MiDAC. HDAC3 is a component of the NCoR complex. Biochemical assays using isolated recombinant protein have been possible to develop for HDAC1/2, but HDAC3 assays have required the addition of NCoR. In the past, comparison of in vitro assays performed on isolated HDAC1/2 versus NCoR-bound HDAC3 incorrectly suggested that 4’-aryl benzamides, like Cpd-60 (also known as Merck 60), are selective for HDAC1/2 over HDAC3. Furthermore, the kinetics of these HDAC inhibitors are often slow and frequently show state-dependent differences in activity, relying, for example, on protein partners and inositol phosphates.99–102 Carefully tracking enzyme inhibition using continuous enzyme kinetics has similarly found reports of selectivity within HDAC1/2/3 – namely, the selective inhibition of HDAC3 over HDAC1/2 by RGFP966 – to be artifacts of standard end-point assays.102,103
Figure 4.

Context-dependent inhibition of essential HDAC proteins. (a) Gene effect scores for the common essential gene, HDAC3. (b) Chemical structure of corin, a dual HDAC and LSD1 inhibitor designed to preferentially inhibit CoREST-bound HDAC enzymes. (c) Chemical structure of the molecular glue UM171, which selectively degrades CoREST by inducing proximity between KBTBD4 and HDAC1/2-CoREST.
Given the results of these studies, which challenge previous reports of selectivity within the HDAC1/2/3 subfamily, it is prudent to consider that many studies reporting the effects of HDAC1/2 inhibition on cancer cells are likely confounded by the inhibition of HDAC3, a common essential protein. Nevertheless, the combined activities of HDAC1 and HDAC2 are also broadly essential,93,104–115 suggesting that even if HDAC1/2-selective inhibitors are possible to develop, they would likely be broadly toxic as well. HDAC1 and HDAC2 are highly homologous paralogs that incorporate into the same protein complexes (CoREST, NuRD, SIN3A, and MiDAC).42 Loss of one gene or protein is compensated by an increase in the other through post-translational changes in stability.106,107,110 Combined loss of both paralogs is not viable for mouse development and conditional deletion experiments have shown that dual knockout is incompatible with the survival of many diverse cell types. In cancer cells, loss of one paralog is frequently tolerated, whereas loss of both is not. This redundancy creates a selective dependency on one paralog when the other is compromised by cancer-specific genetic deletions.93 In Neuroblastoma, for example, HDAC1 is frequently compromised by deletions of chromosome 1p, creating a reliance on HDAC2 in these cells. Conversely, HDAC2 is deleted in multiple myeloma cells causing a requirement for HDAC1. This genetic relationship, where HDAC2 is only indispensable for cell fitness in the context of HDAC1 deletion (or vice versa), is defined as synthetic lethality – a term borrowed from classical genetics to describe 2 genetic alterations that are tolerated individually but lethal in combination.116 If HDAC1 or HDAC2 could be selectively targeted by small molecules, these genetic deletions could represent vulnerabilities for therapeutic development that would avoid the toxicity of combined HDAC1/2 inhibition to normal cells and potentially widen the therapeutic window of HDAC inhibition. However, pharmacological discrimination between these two paralogs is not currently possible and will likely rely on the development of new pharmacological approaches that go beyond active-site inhibition. For example, several studies have suggested the possibility of selectively degrading certain HDAC isoforms with small molecules that recruit E3 ubiquitin ligase machinery.117–124 Later, we will discuss several other examples of synthetic lethal relationships between paralogs and emerging pharmacological approaches to exploit them, notably including similar methods that induce paralog-selective targeted protein degradation.
2.1.3. State-specific HDAC inhibitors
In retrospect, the growing appreciation that HDAC1/2/3 are indiscriminately important for cell fitness can likely rationalize much of the past difficulty in translating HDAC inhibitors into effective therapeutics. One interesting approach is to further improve specificity and reduce general toxicity by selectively targeting HDAC1/2 in specific multiprotein complexes. Rodin Therapeutics has disclosed benzamide inhibitors with selectivity for CoREST-bound HDAC1/2.125 Here, selectivity was evaluated on substrate assays performed on immunoprecipiated complexes. Recent CoraFluor assays could be used to revisit these compounds, as dissociation of HDAC1/2 from the immunoprecipitated complex might create a pool of free HDAC1/2 that alters the apparent selectivity profile.98 If it is too difficult to create orthosteric inhibitors with improved selectivity within Class I isoforms or across protein complexes, bidentate inhibitors have been suggested as a compelling path forward.126,127 The CoREST inhibitor, Corin, is synthesized by tethering the Class I HDAC inhibitor, entinostat, to an inhibitor of the CoREST demethylase subunit, LSD1 (Figure 4b).126 This bivalent compound showed improved activity against melanoma cell lines and decreased toxicity to normal melanocytes, potentially owing to its preferential inhibition of the intact CoREST complex versus other HDAC complexes.126 Corin is also highly active against preclinical models of diffuse intrinsic pontine glioma, where a CRISPR screen found HDAC inhibition sensitizes these cancer cells to LSD1 inhibition.128 The molecular glue UM171 causes a specific degradation of the CoREST complex by inducing interactions between HDAC1/2 and the ubiquitin ligase complex substrate receptor, KBTBD4 (Figure 4c),129–133 suggesting that small-molecule degraders may also provide a method to antagonize specific HDAC1/2 complexes. Indeed, HDAC2 degradation in HDAC1-deficient cancer cells selectively destabilizes the NuRD complex compared to other HDAC1/2 complexes.93
2.2. DNA methyltransferases
2.2.1. Nucleoside DNMT inhibitors
The nucleoside analog, azacitidine (5-azacytidine), was approved for the treatment of MDS in 2004.134 Today, azacitidine and the related nucleoside analog, decitabine (5-aza-2′-deoxycytidine) (Figure 5a), are used in several additional indications, most notably in combination with the BCL2 inhibitor, venetoclax, for AML. These hypomethylating agents are DNA methyltransferase (DNMT) inhibitors that work through a fascinating mechanism. DNMTs catalyze the methylation of cytosine at the C5 position by first forming a covalent intermediate between an active site cysteine and C6.135 The activated C5 position is then methylated by S-adenosylmethionine (SAM) followed by β-elimination to release the enzyme. Azacitidine was first discovered and described as an anti-cancer agent in 1964 and later found to cause the selective hypomethylation of DNA strands into which azacitidine is incorporated.135,136 When these nucleoside analogs are incorporated into DNA, their 5-aza modifications prevent the release of DNMTs from DNA by β-elimination (Figure 5b), causing global DNA hypomethylation through the trapping of covalent DNMT-nucleic acid intermediates.137 Reducing the levels of DNMT protein genetically causes a decrease in cellular sensitivity to azacytidine, demonstrating that the trapped species of DNMT represents a gain-of-function toxicity to cells that is independent of DNMT degradation and DNA hypomethylation.137 Unlike decitabine, which is only incorporated into DNA, azacytidine is also incorporated into RNA, which causes additional cellular effects that are likely related to its improved efficacy in preclinical models.138–141
Figure 5.

Progress in DNMT drug discovery. (a) Chemical structures of FDA-approved cytidine nucleoside analogs that inhibit DNMTs. (b) Mechanism of DNMT inhibition by nucleoside analog inhibitors. (c) Chemical structures of DNMT1-selective non-nucleoside inhibitors. (d) Co-crystal structure (PDB 6X9J) of GSK3830052 (blue) bound to the interface of DNA (beige) and DNMT1 (white). (e) Gene effect scores for the common essential gene DNMT1.
Azacitidine and decitabine inhibit all three enzymes responsible for DNA methylation – DNMT1, DNMT3A, and DNMT3B. DNMT3A/B are the de novo methyltransferases responsible for methylating new sites of DNA, whereas DNMT1 is responsible for propagating methylation marks onto hemi-methylated DNA after replication.142–155 The therapeutic window of azacitidine and decitabine is likely limited, in part, by nucleoside-specific off-target effects at the high doses that are required to overcome the poor pharmacokinetic properties related to their nucleoside structures. Unfortunately, guadecitabine, a dinucleotide analog of decitabine and deoxyguanosine developed for improved stability,156–158 failed in Phase 3 clinical trials for MDS and AML. Therefore, much effort has been invested in identifying non-nucleoside DNMT inhibitors that would reduce off-target nucleoside toxicities.
2.2.2. Non-nucleoside DNMT inhibitors
Early examples of non-nucleoside inhibitors include the phthalimide, RG-108, which was discovered through a virtual screen for DNMT1 inhibitors;159 the quinoline, SGI-1027,160 which was discovered among a collection of molecules that can bind the minor groove of DNA; and nanaomycin, a quinone antibiotic, which was also identified through virtual screening.161 RG-108, which was originally characterized on bacterial DNMT, was later found to possess minimal activity against mammalian DNMTs.162,163 The manuscript first reporting SGI-1027 was recently retracted, but other groups have validated the activity of SGI-1027 and its analog, MC3343, in purified DNMT assays.162,163 Still, this class of quinolines are relatively weak inhibitors of DNMTs and were recently reported to have minimal effects on global DNA methylation levels in AML cells.163 Nanaomycin is reported to inhibit DNMT3A selectively, sparing DNMT1 in biochemical assays, and showing decreased DNA methylation in cells.161,164 However, its effects have not been thoroughly characterized in cells, it has not been tested in vivo, and its anthracycline core presents concerns for tolerability.
A series of dicyanopyridine compounds represent the most substantial advance in the development of non-nucleoside DNMT inhibitors (Figure 5c).163 These were discovered in a high-throughput screening campaign to identify enzymatic inhibitors of DNMT1.163 This screen identified GW623415X, a 6-amino-3,5-dicyano-4-ethylpyridine scaffold with low micromolar potency against DNMT1. A dimethylamino analog, GSK3484862, showed a nearly 10-fold improvement in activity and no inhibition of other DNMTs up to a concentration of 50 μM, while its enantiomer was inactive up to a concentration of 10 μM. This compound and related analogs, GSK3685032 and GSK3830052, inhibit DNMT1 reversibly and show impressive selectivity against DNMT3A/B. GSK3685032 is more than 2,500-fold selective for DNMT1 over DNMT3A/B. They induce hypomethylation in cells causing anti-proliferative responses in AML models in vitro and in vivo. Extensive characterization of their on-target effects, including genetic manipulations to rule out confounding off-target effects, established this series of compounds as the first potent and specific non-nucleoside DNMT1 inhibitors. Their binding to DNMT1 is context-dependent, showing dramatically improved affinity in the presence of hemimethylated DNA, and co-crystal structures revealed that the compound binds across the protein-DNA interface with the 3,5-dicyanopyridine portion intercalating into DNA (Figure 5d). This intercalation is dependent on the presence of DNMT1, as the compound does not bind to DNA nonspecifically. Not only is this intercalation important for compound binding, but it also displaces the active-site loop of DNMT1 from intercalating into DNA and forming a closed, active conformation of the enzyme. This fascinating mechanism of context-dependent DNA intercalation establishes that DNMT enzymes can be inhibited by non-nucleoside small molecules and may provide future advantages for drug development. However, DNMT1 is a common essential gene (Figure 5e), and it remains unclear what challenges non-nucleoside inhibitors might encounter in human clinical investigation related to on-target toxicity.
2.3. BRD4
2.3.1. Discovery of BET bromodomain inhibitors
BET (bromodomains and extraterminal domains) family proteins (BRD2, BRD3, BRD4, and BRDT) are chromatin readers that bind to acetylated lysine side chains through a tandem pair of N-terminal bromodomains. In 2010, two papers reported the discovery of JQ1 and I-BET (Figure 6a), triazolodiazepines that bind competitively with chromatin to the acetyllysine-binding pocket of BET bromodomains.165,166 These reports provided the first demonstration that chromatin reader domains are pharmacologically tractable targets. The origins of both JQ1 and I-BET can be traced back to phenotypic screens for compounds with anti-inflammatory activity. Therefore, like HDAC and DNMT inhibitors, the biological activity of these compounds was known before their cellular targets were described. BET bromodomain inhibitors were quickly shown to possess extraordinary anti-cancer activity in preclinical models of diverse cancer types, leading to many clinical development programs and clinical trials. On-target toxicity has complicated therapeutic translation but BET bromodomain inhibitors remain under active clinical investigation.
Figure 6.

Prototypical BET bromodomain inhibitors. (a) Chemical structures of JQ1 and I-BET, the first reported BET bromodomain inhibitors. The (R)-JQ1 is an inactive stereoisomer that does not bind BET bromodomains. (b) Chemical structures of prototypical BET bromodomain inhibitors with alternative acetyl-lysine mimetic groups
The thienodiazepine BET bromodomain inhibitor, JQ1, was developed by Professors Bradner and Knapp and studied in midline carcinomas that are driven by oncogenic rearrangements of the BET protein, BRD4.165 JQ1 was synthesized after patent filings from Mitsubishi Pharmaceuticals disclosed a connection between thienodiazepines (originally identified from anti-inflammatory phenotypic screens) and BET bromodomain proteins.165 JQ1 is decorated with solvent-exposed t-butylester and 4-chlorophenyl pendant groups, in addition to a triazolyl moiety that inserts deep into the bromodomain pocket and mimics acetyllysine interactions with a key conserved asparagine. Biochemical profiling showed that this structure binds with 50 to 90 nM affinity to both bromodomain 1 (BD1) and BD2 on all BET isoforms (BRD2/3/4/T) and no significant binding is observed to non-BET bromodomains.
Further establishing the tractability of this class, Professor Tarakhovsky and colleagues at GSK contemporaneously reported the BET bromodomain inhibitor, I-BET.166 This compound was discovered by GlaxoSmithKline through a phenotypic screen for ApoA1 reporter gene activation and studied in models of inflammatory gene signaling.166,167 Here, a benzodiazepine compound, GW841819X, was discovered by phenotypic screening and an analog with similar activity, I-BET, was then found to target BET proteins through ligand affinity chromatography. Sharing the triazolodiazepine core and 4-chloro pendant group with JQ1, it featured a fused benzene instead of a thiophene and an aminoethyl amide instead of a t-butylester. It binds competitively with chromatin to the acetyllysine-binding pocket in the same mode as JQ1, contacting the conserved asparagine and exhibiting similar 50 to 60 nM affinity.
As shown in these index studies, both JQ1 and I-BET inhibit BET bromodomain binding to chromatin, which causes cell cycle arrest and differentiation in BRD4-NUT-driven NUT midline carcinoma (NMC) and anti-inflammatory effects in activated macrophages.165,166 The anti-inflammatory effects of I-BET were attributed to the suppression of pro-inflammatory gene regulatory signaling. By chromatin immunoprecipitation and DNA sequencing (ChIP-seq) studies, I-BET could be shown to evict BET protein from chromatin, leading to selective downregulation of stimulus-responsive inflammatory genes. At these genes, transcriptional elongation by RNA Pol II was suppressed, consistent with prior studies connecting BRD4 to the activity of the elongation factor, P-TEFb.168,169 These class-selective effects would later be shown many times over in diverse model systems with a major focus on the potential to induce anti-cancer effects.170 In NMCs, JQ1 was shown to elicit a potent pro-differentiation effect and G1 cell growth arrest in tissue culture, ultimately prolonging survival of tumor-bearing mice.165
2.3.2. Clinical evaluation of BET bromodomain inhibitors
Interactions between BRD4 and P-TEFb quickly inspired the study of BET bromodomain inhibitors in MYC-driven tumors, as previous studies had suggested an important role for P-TEFb in maintaining MYC expression.168,169,171 BET bromodomain inhibition was accordingly found to possess anti-proliferative effects against several MYC-dependent hematological malignancies.172–175 In the first report, BRD4 was identified in a pooled genetic knockdown screen that aimed to identify essential genes in acute myeloid leukemia (AML) cells from among ~250 chromatin regulatory proteins.172 Both genetic knockdown of BRD4 and BET bromodomain inhibition by JQ1 showed anti-leukemia activity in vivo as a result of induced myeloid differentiation and growth arrest,172 not unlike the pro-differentiation effects observed in NMC.165 Transcriptional downregulation of MYC, together with eviction of BRD4 from the MYC enhancer were also observed in multiple lineage leukemia (MLL) where oncogenesis is driven by MLL fusion oncoproteins, in multiple myeloma (MM) cells where elevated MYC expression is driven by chromosomal translocations that connect the IgH enhancer to MYC promoter regulation, and in a variety of other leukemia and lymphoma cell lines.172–178 Here, MYC was shown to be among the most quickly and severely downregulated genes in response to BET bromodomain inhibition.
BET bromodomain inhibition has since been studied in preclinical models of countless cancers, resulting in many clinical trials in oncology. Results of the initial trials evaluating the thienodiazepine OTX015 (birabresib) in leukemia, lymphoma, multiple myeloma, lung, prostate, and NMC have now been reported.179–182 In general, thrombocytopenia and adverse gastrointestinal events lead to dose-limiting on-target toxicities. Drug holidays have been explored to address these toxicities, especially thrombocyteopenia, which is reversible upon drug cessation, but BET inhibition is now widely considered to be poorly tolerated. BET bromodomain inhibitors have advanced to a Phase 3 clinical trial in one instance, which evaluated CPI-0610 (pelabresib) in combination with the JAK inhibitor ruxolitinib for myelofibrosis (NCT04603495).183 Preclinical studies have previously demonstrated that combined JAK and BET bromodomain inhibition is effective in animal models of myeloproliferative neoplasms (MPN), interestingly related to the anti-inflammatory effects of BET bromodomain inhibition in suppressing NF-kB transcriptional networks.184 This recalls the original discovery of BET bromodomain inhibitors as anti-inflammatory agents and past characterizations of BET-dependent NF-kB signaling.166,185 Conference presentations have reported that the Phase 3 trial of pelabresib and ruxolitinib met its primary endpoint, providing renewed hope for BET bromodomain inhibitors, but these agents have otherwise not shown promising therapeutic profiles in the clinic.
2.3.3. Chemical probe studies of BRD4
BET bromodomain inhibitors have made for exceptionally useful chemical probes of transcription and chromatin biology, which has assisted in illuminating the complications associated with translating BET bromodomain inhibitors into the clinic. When using pharmacological tools to study complex biological systems, assigning a phenotype to the on-target or off-target effects of a compound can be exceedingly difficult.186 Several reviews and perspectives have previously crystalized the experimental strategies to definitively link biological phenotypes to on-target pharmacological activities.186–190 First is to identify a drug-resistant mutant allele that blocks drug binding and suppresses the biological effects of the compound, second is to compare physiochemically matched active and inactive enantiomers, when possible, and third is to use multiple structurally distinct compounds that share the same intended target, as they are likely to have different off-target effects. Evaluating these three criteria for BET bromodomain inhibitors, resistance mutations that disrupt drug binding to BRD4 while also preserving BRD4 function have not yet been discovered.191–194 However, the study of BET bromodomain proteins has benefited from the existence of stereochemistry-selective inhibitors (i.e. the existence of enantiomeric inactive controls) and several classes of chemical scaffolds that inhibit the same sites on BET proteins.40 Indeed, an attractive feature of JQ1 that has contributed to its widespread use as a chemical probe for BET bromodomain proteins is the availability of an inactive enantiomeric control. The t-butyl ester substituent on JQ1 creates a chiral center on the diazepine core, resulting in a physiochemically matched inactive control (Figure 6a).165 The active enantiomer, (S)-JQ1, and inactive control, (R)-JQ1, have been used extensively to help assign cellular phenotypes as being caused by the inhibition of BET bromodomains and not a cellular off-target. In addition to the diazepine BET bromodomain inhibitors, chemical probes with several distinct pharmacophores have been reported, as previously reviewed,170 notably including early examples like the dimethylisoxazole (e.g. I-BET151) and dihydroquinazolinone (e.g. PFI-1) inhibitors (Figure 6b).173,195–197
The rigorous use of structurally distinct chemical probes and paired enantiomeric controls has greatly assisted the study of BET proteins in cancer. Indeed, the preferential downregulation of MYC has been observed repeatedly in diverse cancer types,176–178,191,198–201 including in a pre-registered replication study,202 and can be extended to the MYC paralog MYCN.203 However, this preferential downregulation of MYC is accompanied by a more general downregulation of lineage-specifying TFs, which has been rationalized by the disproportionate abundance of BRD4 at enhancers controlling the expression of these genes.177,185 In addition to regulating the expression of lineage-driving TFs, BRD4 also binds to acetylated TFs, allowing for BRD4-dependent signaling between enhancer-bound acetylated TFs and RNA Pol II.204,205 Altogether, the effects of BET bromodomain inhibition converge on the preferential suppression of transcriptional programs that are important for cell state and identity (Figure 7a), which is congruent with their pro-differentiation effects across cancer models.
Figure 7.

BRD4 is a global regulator of transcriptional activation. (a) BET bromodomain inhibition causes gene-selective inhibition of transcription. (b) Schematic depiction of PROTACs, heterobifunctional compounds that induce targeted protein degradation. (c) Chemical structure of the BET PROTAC, dBET6. (d) BET degradation causes global inhibition of transcriptional activity. (e) Selective degradation of BRD4 using the auxin-inducible degron (AID) system, which relies on auxin-induced proximity between a genetically fused AID tag and an ectopically expressed E3 substrate receptor, Tir1. BRD4 degradation causes global inhibition of transcriptional activity.
Despite these class-selective transcriptional consequences of BET bromodomain inhibition, it is now clear that BRD4 functions more broadly as an essential regulator of transcriptional elongation at most, if not all, actively transcribed genes.206,207 This insight was made possible by the discovery of chemical tools that induce targeted protein degradation of BRD4 rather than inhibiting its isolated bromodomain functions. For over 2 decades, the concept of inducing targeted protein degradation has been pursued through the synthesis of PROTACs (proteolysis targeting chimeras) – bifunctional compounds that independently bind to an E3 ubiquitin ligase and a target protein of interest.208 These compounds can induce ubiquitin transfer onto non-native substrates through chemically enforced proximity to the E3 (Figure 7b). After the drug thalidomide was discovered to bind cereblon (CRBN),209 a cullin-RING E3 ligase complex substrate receptor, Bradner and colleagues developed the first CRBN-based PROTACs, selecting BET proteins to demonstrate proof-of-concept.210 Attaching thalidomide to JQ1 through a polymethylene linker resulted in a bifunctional small molecule, dBET1, which elicits potent and selective degradation of BET proteins in living cells and animals.210 An optimized molecule, dBET6 (Figure 7c), which featured a longer polymethylene linker was subsequently reported and used to investigate BET-dependent transcriptional regulation.206
Using kinetically resolved and unbiased genomic measurements of transcriptional activity following dBET6 treatment, BET degradation was found to universally halt the transcription of all genes by preventing the transition of RNA Pol II from promoter-proximal pausing to productive transcriptional elongation (Figure 7d).206 A later study, which used a genetically encoded system to enable selective degradation of BRD4 (sparing BRD2/3 from degradation), found that the global inhibition of transcriptional elongation by BET degraders could be specifically attributed to the activity of BRD4 (Figure 7e).207 Several direct-acting PROTACs have now also been reported that selectively degrade BRD4 over other BET proteins.211,212 A notable early example, AT1, was developed from JQ1 and a small-molecule ligand for the von Hippel-Lindau E3 substrate receptor (VHL) using structure-guided optimization.212 Small-molecule ligands for VHL that mimic a key hydroxyproline degron were developed by iterative optimization of peptidomimetic ligands.213–216 This afforded VHL-based small-molecule PROTACs capable of degrading diverse targets, including BRD4, which was reported contemporaneously with the first CRBN-based BRD4 degraders.211,217 It is now well-established that multi-specific ligands can be fashioned into heterobifunctional PROTACs to degrade a subset of its targets.211,212,218–224 These compounds rely on the determinants of ternary complex formation rather than ligand affinity to achieve improved selectivity, as shown by BRD4-selective PROTACs that spare other BET proteins from degradation.211,212,220
The discovery that BRD4 is universally required for transcription likely explains the finding that BRD4 is a common essential gene.48 It might also explain why so many diverse preclinical models of cancer showed striking responses to BET bromodomain inhibition whereas clinical trials have not been nearly as promising. As discussed by Professor Sellers and colleagues, targeting common essential genes as an anti-cancer strategy presents major difficulties with on-target toxicity, limiting the therapeutic window in human patients.48 Due to the likelihood that a common essential gene is important for an indispensable cell type, tissue, or function, systemic exposure to drugs targeting essential genes can create unacceptable toxicities. The global suppression of transcription by BRD4 degradation likely explains why the BET degraders that were once disclosed by industry groups have not been pursued as clinical programs.225 However, it is important to consider that BET bromodomain inhibitors specifically target the interaction between bromodomains and acetyllysine and do not necessarily disrupt all associated functions of BRD4. Indeed, BRD4 degradation elicits a much more severe effect on transcription and cell fitness than BET bromodomain inhibition,206 the former being much closer in nature, and in consequence, to the genetic loss-of-function screens that led to the annotation of BRD4 as a common essential gene. BET bromodomain inhibition, in contrast, elicits a class-selective suppression of a subset of genes.177,206,207 Why BET bromodomain inhibition differs from BET degradation might be rationalized by the observation that bromodomain inhibition preferentially evicts BRD4 from enhancer regions where it is most densely localized.177 Therefore, BET bromodomain inhibition preferentially impacts the genes targeted by these enhancers without eliciting the same global transcriptional suppression as BET degradation. Despite these differences, BET bromodomain inhibitors have still not led to the therapeutic breakthroughs that were originally signaled by early preclinical studies, further highlighting the challenges of targeting the protein products of common essential genes.48
Chemically induced degradation has demonstrated that BRD4 functions as a global transcriptional regulator by controlling the kinase activity of CDK9,206,207 the kinase component of P-TEFb. RNA Pol II is precisely phosphorylated at specific side chains within a heptapeptide repeat (the YSPTSPS sequence repeated 52 times in mammalian cells) on its C-terminal domain.226 CDK9 is responsible for phosphorylating RNA Pol II on Ser2 of this repeat, which—along with the CDK9-mediated phosphorylation of DSIF and NELF— is responsible for releasing RNA Pol II from promoter-proximal transcriptional pausing.16 BRD4 was previously thought to regulate transcription by recruiting CDK9 to chromatin,168,169 but acute BRD4 degradation has shown that it is required for the activity, not recruitment, of CDK9.206,207 Ultimately, CDK9 inhibition and BET degradation provoke substantively identical transcriptional responses.206
In general, cyclin dependent kinases that regulate transcription have proven to be fruitful targets for discovery chemistry (Figure 8a). Chemical probes and drug candidates targeting CDK7, CDK8, CDK9, CDK12, and CDK13 are also now well represented,227–235 as was recently reviewed.236,237 Inhibition of CDK9 by the ATP analog, DRB (5,6-dichloro-1-b-D-ribofuranosylbenzimidazole), was critical to the discovery of factors controlling pause release and transcriptional elongation.238–251 The more cellularly active CDK inhibitor, flavopiridol, has been similarly useful for studying P-TEFb in cells, revealing a global requirement for P-TEFb across all transcribed genes (Figure 8b).252–256 Most recently, compounds with improved selectivity for CDK9 over other CDKs, such as the bipyridine inhibitors, iCDK9 and NVP-2, or the pyrazolopyrimidine, KB-0742, have enabled unambiguous assignment of these effects to CDK9 (Figure 8c).206,221,257,258 Following on the clinical failures of non-selective CDK9 inhibitors, one CDK9-selective inhibitor, AZD4573, was developed for clinical use against hematological malignancies.259 Here, the logic was to target the short-lived transcript of the antiapoptotic gene, MCL1, by using selective CDK9 inhibition to transiently inhibit transcriptional activity globally, such that transcripts with short half-lives would be preferentially depleted. This strategy was developed with the goal of overcoming the challenging safety profiles seen with previous CDK9 inhibitors.259 However, after clinical trials reached as far as Phase 2, AstraZeneca suspended clinical development of AZD4573, citing strategic portfolio prioritization. Kronos, the developers of KB-0742, have terminated clinical investigation of their selective CDK9 inhibitor due to a lack of safety and futility (NCT04718675). Similar to BET bromodomain degraders, these developments present concerns that the global effects of CDK9 inhibition on transcription will prohibit therapeutic dosing in humans.
Figure 8.

CDK9 inhibitors and transcription elongation. (a) Early chemical tools to study transcription elongation and CDK9 function. (b) Inhibition of CDK9 inhibits transcriptional activity globally. (c) Chemical structures of selective CDK9 inhibitors.
2.3.4. Domain-selective BET bromodomain inhibitors
Each BET protein contains two bromodomains located in tandem near the N-terminus. Most BET bromodomain inhibitors (e.g. JQ1, I-BET, pelabresib) inhibit both bromodomain 1 (BD1) and BD2. In recent years, several groups in academia and industry have sought to mitigate the toxicity associated with BET bromodomain inhibition by pursuing selectivity between BD1 and BD2, which has resulted in compounds that selectively inhibit BD1 of BRD2/3/4 or BD2 of BRD2/3/4 (termed BD1-selective BET bromodomain inhibitors and BD2-selective BET bromodomain inhibitors, respectively).260–286 BD1 and BD2 have been reported to mediate different biological activities, with BD1 generally suggested to be the primary mediator of binding to acetylated histones and BD2 thought to bind acetylated lysines on other proteins that are present on chromatin.287–292 Each BD1 domain is more closely related to the BD1 domains from other proteins than they are to the BD2 domain in the same protein, which has enabled the development of compounds that inhibit all BET proteins but discriminate between BD1 and BD2. This was first indicated as a possibility by resveratrol analogs in 2013,261 but more firmly established by a series of papers publishing high-quality BD1-selective and BD2-selective chemical probes in 2020.268–272 ABBV-744, a BD2-selective chemical probe, contains a pyrrolopyridone core and was developed through structure-guided optimization to exploit side chain differences present on loops at the periphery of the ligand-binding site (Figure 9a).268,269 In BRD4, for example, Asp144 and Ile146 in BD1 are replaced by His437 and Val439 in BD2 (Figure 9b). The team at AbbVie that developed ABBV-744 recognized modest selectivity (7-fold) for BD2 in one compound from a pyrrolopyridone series that had previously led to the development of the dual BD1/2 BET inhibitor, ABBV-075 (mivebresib) (Figure 9b). This compound features a primary amide group on the C2 position of the pyrrolopyridone core, which is unsubstituted in ABBV-075 but positioned in close proximity to the Asp144/His437 and Ile146/Val439 side chains of BRD4 BD1/BD2. Expansion of the amide to an ethyl amide further improved selectivity to 17-fold for BD2, rationalized in co-crystal structures by the inward projection of BD2-His437 into the periphery of the binding pocket, compared to BD1-Asp144 facing outward (Figure 9b). Optimization of the 2,4-difluorophenoxy group, which is positioned proximally to Ile146/Val439, led to >100-fold selectivity. Further optimization of pharmacokinetic properties led to ABBV-744, which maintained single-digit nanomolar potency on BD2 (Ki = 1.6 nM) and over 300-fold selectivity against BD1 (520 nM). Overall, this series of ligands showed far improved shape complementarity for BD2 than BD1, justifying their impressive selectivity.
Figure 9.

BD1-selective and BD2-selective BET bromodomain inhibitors. (a) Discovery of ABBV-774, a BD2-selective BET bromodomain inhibitor. (b) Co-crystal structures of BRD4 BD1 (PDB 6VIW) and BRD4 BD2 (PDB 6VIX) with a BD2-selective inhibitor from the ABBV-744 series. (c) Discovery of BD2-selective acetamide BET bromodomain inhibitor series. (d) Chemical structure of iBET-BD2, an optimized BD2-selective BET bromodomain inhibitor. (e) Discovery of iBET-BD1, a BD1-selective BET bromodomain inhibitor. (f) Effects of dual BD1/2 BET bromodomain inhibition versus BD2-selective BET bromodomain inhibition.
Contemporaneously, GSK reported both BD1-selective and BD2-selective BET bromodomain inhibitors, termed iBET-BD1 (GSK778) and iBET-BD2 (GSK046).270–272 In comparison to the discovery of ABBV-744, which traced back to a pre-existing ligand series, GSK discovered the starting point for iBET-BD2 from a high-throughput screen of over 2 million compounds. Using a TR-FRET-based screen, two BRD4 constructs were employed, each containing both tandem bromodomains, but with either BD1 or BD2 inactivated by Tyr to Ala mutations (Figure 9c). An acetamide screening hit showed ~10-fold selectivity for BD2 over BD1 across all BET family members (Figure 9c). Co-crystal structures with BRD4 BD1 and BRD2 BD2 revealed the importance of His433 in BRD2 BD2 (analogous to His437 in BRD4 BD2), which showed conformational flexibility of rotamers alternately facing inward or outward. In contrast, Asp144 of BRD4 BD1 was conformationally restrained and pointed inward. Further structure-informed improvements led to iBET-BD2, which is moderately less potent than ABBV-744, but similarly selective (KD,BD2 = 30 nM, >300-fold selective) (Figure 9d). iBET-BD1 was developed from a previously disclosed BD1/2 inhibitor – the dimethylisoxazole I-BET151 – using structure-guided efforts to capture interactions with Asp144 through a local water network that is not present in BD2, ultimately achieving 19 nM affinity for BD1 and >100-fold selectivity against BD2 (Figure 9e). While the BD1 selectivity of iBET-BD1 was less impressive compared to iBET-BD2, further selectivity for BD1 is possible. A structurally distinct compound with >1000-fold selectivity for BD1 over BD2, GSK789, was developed in a separate effort following observations that the ATAD2 bromodomain inhibitor, GSK8814, showed >50-fold selectivity for BRD4 BD1 over BD2.293
Comparison of the BD2-selective inhibitor, ABBV-744, to the dual BD1/2 inhibitor, ABBV-075, revealed a striking difference in living systems (Figure 9f).268 While ABBV-075 was shown to be broadly toxic across cell lines of diverse cancer types, ABBV-744 was selectively cytotoxic, preferentially impacting AML and prostate cancer cell lines. This selectivity was observed alongside a muted impact on global transcriptional control compared to dual inhibition and translated to a more tolerated profile in vivo that was associated with reduced thrombocytopenia and GI toxicity in mice. Promisingly, ABBV-744 has progressed into early-stage clinical trials, including a study in combination with ruxolitinib or navitoclax for myelofibrosis (NCT04454658), the indication where BET bromodomain inhibitors have performed best in the clinic.
In another study, iBET-BD2 was reported to be ineffective as an anti-cancer agent both in vitro and in vivo, whereas iBET-BD1 phenocopied dual inhibition. These data further establish the importance of BD1 to basal BRD4 function, but it is unclear why iBET-BD2 failed to elicit similar effects as ABBV-744 in preclinical cancer models. One possibility is that this reflects the weaker potency of iBET-BD2 compared to ABBV-744. The exceptionally quick maturation of BD1- and BD2-selective chemical probes and drug candidates will allow for unambiguous testing of domain-selective BET inhibitors in preclinical studies and clinical trials.
2.3.5. Repurposing BET bromodomain inhibitors to induce transcription
Beyond their utility as chemical probes of BET family proteins, JQ1 and other prototypical BET bromodomain inhibitors have also emerged as exceptionally useful tools for the development of heterobifunctional chemical inducers of proximity (CIPs). Several attributes have made JQ1 particularly attractive for proof-of-concept studies that seek to establish new forms of chemically induced proximity. First, it is readily available from commercial suppliers and is relatively inexpensive. Second, the tert-butylester group of JQ1 is solvent exposed when bound to bromodomains and therefore provides a conveniently accessible protected carboxylic acid that can be used to attach linkers without obstructing bromodomain engagement.294 Finally, the inactive enantiomer of JQ1 provides a valuable negative control to determine whether the cellular effects of a JQ1-based heterobifunctional CIP require on-target engagement of BET bromodomains. These qualities likely contributed to JQ1 being used in each of the first 3 proof-of-concept studies showing that CRBN ligands can be used to construct heterobifunctional PROTACs210,211,225 and it has continued to be overrepresented in the development of new methods for inducing targeted protein degradation.120,211,212,295–315 Beyond targeted protein degradation, JQ1 has also helped to establish several other forms of chemically induced proximity, notably including chemically induced phosphorylation (PHICS),316 O-GlcNAcylation (OGTAC),317 nuclear import (NICE),318 and regulated induced proximity targeting chimeras (RIPTACs).319
A particularly inventive use of JQ1 is embodied in recent reports of chemical inducers of proximity that use JQ1 to recruit the powerful transcriptional co-activator functions of BRD4 to silenced genes, conditionally reactivating their expression (Figure 10).318,320,321 The primary challenge in developing chemical inducers of proximity that selectively regulate gene transcription is the identification of small molecules that can localize to specific gene regulatory elements. This was first addressed using genetically encoded systems – for example, using chemically induced proximity to recruit a tagged transcriptional regulatory proteins to an engineered locus.322–329 Later methods evoked pyrrole- and imidazole-based polyamides, which bind directly to DNA,330 to create heterobifunctional compounds that circumvent the need for locus engineering.320,331–335 While the limited specificity of polyamides has presented challenges for this approach, it has been particularly effective in models of Friedreich’s Ataxia,320,336 where the pathophysiological silencing of FXN transcription is caused by the expansion of repressive GAA microsatellite repeats near the FXN promoter.337,338 A heterobifunctional molecule attaching JQ1 to a GAA-targeted polyamide resulted in a remarkably selective activator of FXN transcription, called synthetic transcription elongation factor 1 (Syn-TEF1) (Figure 10a).320 By localizing to the GAA repeats near FXN, SynTEF1 can recruit BRD4 to the FXN promoter, which activates paused RNA Pol II for productive transcriptional elongation. Domain-selective BET bromodomain inhibitors have also been used in place of JQ1 to synthesize new FXN-activating SynTEFs, revealing a dramatic preference for BD2-based recruitment over BD1, which was suggested to reflect differences in the evolved functions of these domains.336
Figure 10.

Chemical inducers of proximity that control the assembly of transcriptional regulatory complexes. (a) SynTEF1 binds GAA repeats near the silenced FXN promoter through its polyamide DNA-binding moiety (blue) and recruits BRD4 to activate FXN transcription. (b) CEM87 binds FKBP12-tagged dCas9 through its FK506 moiety (blue), localizing to dCas9-bound regions of the genome and recruiting BRD4 to activate gene expression in a locus-specific manner. (c) TCIP1 binds to the transcriptional repressor BCL6 (blue) and recruits BRD4 to active BCL6-repressed gene targets. (d) CDK-TCIP1 recruits CDK9 (orange) to BCL6 (blue) through its kinase inhibitor moiety (orange), producing a locally high effective molarity of CDK9 near BCL6-bound promoters to active expression of BCL6-repressed gene targets.
Tag-specific heterobifunctional small molecules have been used to enable flexible targeting of BRD4 to specific genomic locations bound by Cas9. This method requires expression of a nuclease “dead” Cas9 mutant (dCas9) fused to FKBP12 and a locus-specific small guide RNA (sgRNA) that navigates dCas9 to a desired region of the genome. A heterobifunctional fusion of JQ1 and FK506 can then be used to recruit BRD4 to dCas9-bound locations of the genome through its FKBP12 tag, allowing for locus-specific and ligand-dependent activation of gene expression (Figure 10b). JQ1 has even been conjugated directly onto dCas9 using split intein chemistry, allowing for the programmable localization of JQ1 on the genome by cellular delivery of semisynthetic dCas9-JQ1 conjugates.339 While the use of dCas9 to direct small molecules to specific genomic locations has enabled elegant proof-of-concept studies in cells, the challenge of delivering the large dCas9 construct in vivo present a substantial hurdle for clinical translation. More recent designs have used recombinant zinc finger (ZF) DNA-binding domains in place of dCas9 such that delivery is possible by adeno-associated virus (AAV), yet these have only been applied in tissue culture to date [cite].340,341
The repressive transcription factor BCL6, has been coopted to enable locus-specific delivery of JQ1 without the need for any genetic engineering or protein delivery.321 In this elegant approach, BCL6 inhibitors were repurposed to synthesize heterobifunctional transcriptional chemical inducers of proximity (TCIP) compounds that can recruit BRD4 to BCL6, resulting in small molecules that induce remarkably potent anti-lymphoma activity by activating genes that are ordinarily repressed by BCL6.321 The BCL6 transcription factor is an essential driver of diffuse large B-cell lymphoma (DLBCL) and functions by interacting with transcriptional co-repressor complexes to suppress BCL6 target genes, as previously reviewed.342 Small molecules that bind to the BCL6 BTB domain and inhibit its interactions with co-repressors have previously been discovered using a high-throughput fluorescence polarization screen of nearly 2 million compounds, which measured the displacement of a co-repressor-derived peptide from the BTB domain.343 From an initial 4-amino-5-chloropyrimidine screening hit, which showed 20 μM affinity for the BTB domain, structure-guided optimization provided dramatic improvements in binding affinity, affording two notable chemical probes, BI-3802 and BI-3812. Both compounds inhibit the interaction between the BTB domain and co-repressor peptide with single-digit nanomolar potency and possess only slight differences in their overall chemical structures. However, different substituents on a terminal piperidine group afford BI-3802 the ability to induce potent and selective BCL6 degradation.343 Later studies determined that BCL6 degradation results from chemically induced polymerization of BCL6, a proteoform that is recognized and ubiquitinated by the SIAH1 E3 ubiquitin ligase.344
Compared to BCL6 inhibition by BI-3812, BCL6 degradation by BI-3802 produces more substantial de-repression of BCL6 target genes and causes anti-proliferative responses in DLBCL cell lines that do not respond to the inhibitor. However, even stronger activation of BCL6 target genes can be elicited by compounds that recruit BRD4 to BCL6. TCIP1, a chimera of BI-3812 and JQ1, recruits BRD4 to BCL6, activating the expression of BCL6 target genes beyond the simple de-repression that is achieved by BCL6 degraders.321 This heterobifunctional compound induces cooperative BRD4-BCL6 interactions, recruiting BRD4 to BCL6 target genes without affecting the normal bulk distribution of BRD4 elsewhere in the genome (Figure 10c). The recruitment of BRD4 causes an immediate increase in transcriptional elongation by RNA Pol II at BCL6 target genes, including important pro-apoptotic genes, ultimately causing the death of DLBCL cell lines. These important proof-of-concept studies could provide a renewed purpose for the large pharmacopeia of BET bromodomain inhibitors.
Like JQ1, CDK9 inhibitors have also been used to construct BCL6 TCIPs.345 CDK-TCIP1, a chimera of BI-3812 and the CDK inhibitor SNS-032 can induce BCL6-CDK9 proximity and rapidly activate BCL6 target genes by inducing CDK9 to phosphorylate RNA Pol II at BCL6-bound promoters (Figure 10d). CDK-TCIP2, which harbors a more rigid and structured spirocyclic linker, showed more potent activity and improved pharmacokinetic behavior, allowing its use in animal models where it potently inhibits the proliferation of B cells in the germinal center. An interesting distinction from BCL6-BRD4 TCIPs is that BCL6-CDK9 TCIPs are constructed from active-site inhibitors of CDK9 enzymatic activity. Recruitment of mutant CDK9 isoforms lacking catalytic activity failed to activate BCL6 target genes, indicating that the catalytic activity of CDK9, and not its scaffolding functions, is strictly required for TCIP activity. This discovery indicates that an inhibited form of CDK9 can be recruited to BCL6 and then released to form a locally high effective molarity of catalytically competent CDK9 molecules that then mediate locus-specific transcriptional activation. This is consistent with a previous study that successfully repurposed HDAC inhibitors to recruit HDACs to a specific genomic locus for chemically induced transcriptional silencing.328 Similar to BET bromodomain inhibitors, TCIP pharmacology may provide a renewed interest in CDK9 inhibitors, which have otherwise proven challenging to translate into the clinic.
Expansion of TCIP pharmacology beyond BCL6 will require further advances in TF ligand discovery. However, with few exceptions, such as nuclear hormone receptors,346 TFs typically lack the types of naturally evolved ligand-binding domains that are preferred for chemical probe and drug discovery.347,348 Chemoproteomic-based methods for ligand discovery, which have matured at a remarkable pace in recent years,349–351 show great promise for the future of TF-directed drug discovery.352–356 Cysteine-reactive ligands are now reported for MYC, β-catenin, SOX10, NF-κB1, and FOXA1.352,353,355,356 These early-stage compounds have produced a great diversity of functional effects, for example, inducing degradation of β-catenin, stabilizing SOX10 dimerization, and altering the sequence specificity of FOXA1-DNA interactions. Most relevant to the chromatin-specific focus of this review, covalent ligands for the pioneer factor FOXA1 were recently discovered by chemoproteomic-based ligand discovery.352 The tryptoline acrylamide, WX-02–23, stereoselectively engages a cysteine (C258) within the FOXA1 DNA-binding domain, improving its affinity for weak motifs without affecting strong ones, thus relaxing the basal sequence preferences of FOXA1. In cells, this results in many thousands of gained and lost FOXA1 binding sites, with these sites showing increases and decreases in chromatin accessibility, respectively.352 This effect on chromatin accessibility is consistent with the description of FOXA1 as a pioneer TF,357,358 and, interestingly, the mechanism by which WX-02–23 affects FOXA1 after engaging C258 resembles a recurrent cancer-associated mutation (R261G) located nearby.352,359–361 This cellularly active small-molecule modulator of FOXA1 highlights the potential for chemoproteomic-based ligand discovery to address historically challenging targets, like transcription factors.
2.4. Context-specific inhibition of pan-essential gene products
While the products of common essential genes typically make for challenging drug targets, the complex genetic alterations observed in human cancers can present opportunities to selectively modulate an essential protein in tumor cells versus a normal genetic background. This principle has led to the clinical development of PRMT5 (protein arginine methyltransferase) inhibitors that target cancers harboring homozygous deletions of methylthioadenosine phosphorylase (MTAP). Homozygous deletions within the chromosome 9p21 region, which inactivate the tumor suppressor gene CDKN2A, are among the most commonly observed genetic alterations in cancer.362 MTAP, which is in close genomic proximity to CDKN2A, is often co-deleted, causing a dramatic sensitization to knockdown of PRMT5, WDR77 and RIOK1 – genes encoding subunits of the PRMT5 methylosome.363–365 In the methionine salvage pathway, the conversion of methylthioadenosine (MTA) to adenine and methionine is catalyzed by MTAP (Figure 11a). Tumor cells harboring MTAP deletions therefore accumulate high concentrations of MTA, which selectively inhibits PRMT5 by binding competitively with S-adenosylmethionine (SAM), the cofactor required for methylation by PRMT5 (Figure 11b).363–365 This partial inhibition of PRMT5 by MTA in MTAP-deficient tumors has enabled the development of compounds that target the cell-essential functions of PRMT5 selectively in tumor cells.366–369
Figure 11.

Development of MTA-cooperative PRMT5 inhibitors to selectively target the common essential protein PRMT5 in MTAP-deleted tumors. (a) Metabolism of MTA by MTAP. (b) MTAP deletion elevates cellular MTA concentrations, partially inhibiting PRMT5-mediated deposition of symmetric dimethyl arginine (SDMA) modifications. (c) PRMT5 inhibitors that do not bind cooperatively with MTA inhibit PRMT5 in “normal” cells and MTAP-deleted tumor cells, diminishing their therapeutic window. (d) MTA-cooperative inhibitors selectively inhibit PRMT5 in MTAP-deleted tumor cells. (e) Chemical structures of MTA-cooperative PRMT5 inhibitors.
Knockout of Prmt5 causes embryonic lethality in mice and DepMap demonstrates PRMT5 is encoded by a common essential gene (Figure 11c).370 It catalyzes the post-translational modification of arginine to symmetric dimethylarginine (SDMA), primarily regulating histone proteins, RNA splicing factors, and ribosomal proteins.371–379 While RNAi knockdown of PRMT5 is tolerated by wild-type cells, the hypomorphic state of PRMT5 caused by the accumulation of MTA makes MTAP-deleted cells sensitive to further PRMT5 loss-of-function,363–365 but a complete knockout impairs the viability of both wild-type and MTAP-deleted cancer cells.364 This proved to be a critically important distinction in developing PRMT5 agents that can exploit MTAP synthetic lethality. EPZ015666, a selective PRMT5 methyltransferase inhibitor predated the report of PRMT5-MTAP synthetic lethality.380,381 Originally developed to target PRMT5 in mantle cell lymphoma and discovered by high-throughput screening, EPZ015666 binds cooperatively with SAM and competitively with PRMT5 substrates.380,381 This mechanism of inhibition is indifferent to MTA concentrations, causing antiproliferative effects indiscriminate to MTAP status (Figure 11d).363–365 This inspired the need to develop inhibitors that could bind cooperatively with MTA to the PRMT5-MTA complex an effectively exploit the synthetic lethal relationship.363–365
Several MTA-cooperative PRMT5 inhibitors have now been reported, including MRTX1719, TNG908, AZ-PRMT5i-1, and AMG 193 (Figure 11e,f).366–369,382 MRTX1719, the first reported MTA-cooperative inhibitor, was developed using structure-based optimization of 4-(aminomethyl)phthalazin-1(2H)-one, a hit discovered by surface plasmon resonance (SPR)-based fragment screening.366,367 The fragment showed 5-fold preference in affinity for PRMT5-MTA over PRMT5-SAM (KD = 10 μM versus 51 μM) and was co-crystalized with both complexes to understand the structural basis of its selectivity. It was further optimized using consecutive rounds of structure-based design, with analogs being tested in biochemical enzyme assays with and without MTA present to optimize for MTA cooperativity. MRTX1719 inhibits PRMT5-mediated SDMA marks with a potency of 8 nM in MTAP-deleted cells and inhibits growth with a similar potency of 12 nM. In cells with wild-type MTAP, inhibition of SDMA is shifted to 653 nM and cellular growth inhibition drops to 890 nM. This is consistent with SPR experiments showing that MRTX1719 has a 67-fold greater preference for binding PRMT5-MTA over PRMT5-SAM (KD= 0.140 pM versus 9.4 pM). An atropisomer of MRTX1719 was nearly 200-fold less active in MTAP-deleted cells, further confirming its on-target activity. In mouse models, this leads to potent anti-tumor activity against MTAP-deleted, but not wild-type xenografts, validating their sensitization to MTA-cooperative PRMT5 inhibition in vivo.366,367 Clinical reports from patients treated with MRTX1719 in a Phase 1/2 study have also confirmed SDMA inhibition in MTAP-deleted tumors and several objective responses (RECIST 1.1 criteria) have been observed with tumor size decreasing over time, providing an impressive clinical proof-of-concept (NCT05245500).367 Another clinical agent, AMG 193 was optimized from an aminoquinoline scaffold discovered in a DNA-encoded library (DEL) screen that was run in the presence or absence of MTA.369 Selectivity for MTAP-deleted cancer cells was improved from 3-fold to 40-fold through structure-based optimization, with AMG 193 showing remarkable selectivity for MTAP-deleted cells and a strong correlation to PRMT5 knockdown across nearly 1000 cancer cell lines. Like MRTX1719, AMG 193 shows selectivity for MTAP-deleted xenografts and early clinical reports show activity against MTAP-deleted cancers (NCT05094336).369 These initial clinical reports ease concerns about a prior study that suggested the synthetic lethality between MTAP and PRMT5 would be muted in vivo by stroma-mediated metabolism of excess MTA.383
3. Targeting genetically restricted vulnerabilities
To overcome the challenges related to on-target toxicity that can be presented by targeting common essential genes, there have been broad efforts to identify genes that are required for the survival of cancer cells harboring specific genetic alterations. In principle, drugs targeting these genes can leverage synthetic lethality to selectively target cancer cells and spare normal tissues and physiological functions. These genetically restricted cancer vulnerabilities can take many forms and are richly associated with chromatin biology. For example, we discuss dependencies that arise from the unnatural behavior of chimeric oncoproteins, like MLL fusions, or hypermorphic mutant enzymes, like EZH2. In a more specialized case—paralog synthetic lethality—genetic loss of function affecting one of two closely related paralogs can create a highly selective vulnerability on the other. This most often occurs when the overlapping function of these paralogs is required, in general, for cell survival. Thus, when one paralog is missing in cancer cells but not normal cells, the other paralog can present a highly attractive drug target. While chromatin biology harbors many examples of this synthetic lethality, paralogs also tend to have highly similar structures and ligand-binding pocket. This presents substantial challenges for discovery chemistry, as a chemical agent must selectively target the remaining paralog to effectively leverage the synthetic lethal relationship. As we will discuss, this challenge remains a major area of ongoing effort and innovation for chromatin drug discovery.
3.1. MLL-fusion dependencies
MLL (mixed lineage leukemia), also known as KMT2A, is a histone methyltransferase critical for the transcriptional regulation of hematopoiesis and cellular differentiation.384 MLL controls the expression of developmental programs, most notably by activating the transcription of HOX cluster genes.385,386 The MLL gene is frequently rearranged by chromosome 11q23 translocations, leading to the production of more than 80 different MLL fusion oncoproteins that drive the development of acute leukemia.387–391 These fusion proteins combine the N-terminal chromatin-binding domains of MLL with the C-terminal region of a partner protein, most often a transcriptional coactivator.392–394 The resulting oncoprotein localizes to MLL target sites on chromatin where it aberrantly activates transcription via the C-terminal fusion partner (Figure 12a).395 In this way, MLL fusions dysregulate the developmental control of MLL target genes, promoting the ectopic expression of leukemic proto-oncogenes, such as HOXA9 and MEIS1.396,397 In preclinical models, leukemias driven by this mechanism are preferentially vulnerable to the disruption of many transcriptional co-activators and chromatin regulators that support the function of MLL and MLL-fusion oncoproteins.398 This has motivated the discovery of chemical tools and clinical development programs for several chromatin regulatory proteins such as ENL, DOT1L, WDR5, and menin, which are discussed throughout this chapter. Notably, the success of targeting menin with a small molecule menin-MLL inhibitor, recently approved by the FDA for the treatment of MLL-fusion leukemia, provides strong validation for this therapeutic approach in a clinical setting.
Figure 12.

Oncogenic MLL fusions create cancer-specific vulnerabilities. (a) MLL fusion proteins form pathogenic transcriptional regulator complexes and vulnerabilities to inhibition of specific transcriptional regulators, such as DOT1L. (b) DOT1L is a selective dependency in MLL-rearranged leukemia. Boxplot of CRISPR gene effect scores for DOT1L across 1150 cancer cell lines (DepMap) grouped by the presence of an MLL-rearrangement (N = 12). Boxes represent the median and IQR 25–75%. Whiskers extend 5–95%.
3.1.1. DOT1L
Fusion proteins resulting from MLL chromosomal translocations retain the N-terminus of MLL fused with the C-terminus of another protein. While dozens of MLL fusion partners have been described, the most prevalent are proteins involved in transcriptional activation, including AF4, AF9, ENL, and AF10. Many of these fusion partners have been shown to interact with the histone methyltransferase, DOT1L, either directly or via higher-order multiprotein complexes.395,399–408 DOT1L (disruptor of telomeric silencing 1-like) was originally discovered in yeast as a regulator of telomere silencing and has since been identified in mammals as the only known histone H3 lysine 79 (H3K79) methyltransferase.409–412 H3K79me2/3 modifications are present in the body of actively transcribed genes and elevated levels of H3K79me2/3 are positively correlated with transcriptional output.413–415 DOT1L activity is required during development for proper erythropoiesis and has important roles in adult hematopoiesis.416,417 However, MLL-fusion leukemia are hypersensitive to DOT1L loss-of-function, with MLL-fusion oncoproteins shown to colocalize with a disproportionately high amount of H3K79me2 marks at MLL-specific target genes (Figure 12b).401,405,418–420 The interaction between MLL-fusion proteins and DOT1L triggers the recruitment of DOT1L to ectopic sites, resulting in the hypermethylation of H3K79 and the dysregulated expression of genes such as HOXA9 and MEIS1, which are crucial for initiating and sustaining leukemogenic proliferation. Genetic knock-out and mutagenesis experiments have shown that DOT1L, and more specifically its histone methyltransferase (HMT) activity, are indispensable for the growth of MLL-fusion leukemia.419,421–426
Unlike other lysine methyltransferase proteins, DOT1L does not contain a canonical SET (su(var)3–9, enhancer-of-zeste, and trithorax) methyltransferase domain. Instead, it contains a class 1 methyltransferase domain that is structurally similar to arginine methyltransferases and DNA methyltransferases.427,428 Initial efforts to discover DOT1L methyltransferase inhibitors used libraries of nucleoside analogs based on the native methyl donor, SAM (Figure 13a).420,429–432 The SAM cofactor binds a narrow channel on DOT1L with the methionine group of SAM projecting into the center of the protein (Figure 13a). A loop at the entrance to the tunnel is unique to DOT1L and likely contributes to its specificity for H3K79.427 All histone methyltransferases use SAM as a methyl group donor; however, likely due to its distinct non-SET domain, DOT1L inhibitors show excellent selectivity against other methyltransferases, as witnessed by the first prototypical DOT1L inhibitor, EPZ004777.420,432 EPZ004777 retains the adenosyl core of SAM but replaces the sulfur atom with a tertiary amine that connects to an isopropyl group and a tert-butyl phenyl urea (Figure 13b). EPZ004777 binds competitively to the SAM-binding pocket and causes the opening of a previously inaccessible hydrophobic pocket (Figure 13b). EPZ004777 exhibits a similar binding pose as SAM, maintaining interactions with key amino acid residues within the active site, as well as a novel interaction with the carbonyl oxygen of Gly163.432 Treatment with EPZ004777 in vitro causes a global decrease in H3K79me2 levels, but several days are required to observe this effect.420 There is no cellular demethylase known for the removal of H3K79 methylation, so the global depletion of this mark appears to rely on histone H3 turnover, resulting in a delay in the onset of effects from DOT1L inhibition.433–435 EPZ004777 treatment produces a dose and time dependent decrease in HOXA9 and MEIS1 transcripts, key drivers of MLL-fusion leukemia, and selectively inhibits the proliferation of MLL-rearranged leukemia cells, inducing differentiation and apoptosis.420 Evaluating the anti-leukemia activity of EPZ004777 in mice required administration via subcutaneous pumps, where it reduced H3K79me2 levels and significantly increased the survival of mice bearing xenografts of the MV4;11 cell line, a model of MLL-AF4-rearranged AML. This compound validated the therapeutic strategy for targeting DOT1L in MLL-rearranged leukemia, but it had insufficient pharmacokinetic properties for clinical development.420
Figure 13.

DOT1L methyltransferase inhibitors (a) DOT1L cofactor, SAM (blue), in complex with DOT1L (PDB: 3QOW). (b) EPZ004777 bound to DOT1L (PDB: 4EKI). (c) EPZ-5676 bound to DOT1L (PDB: 4HRA). The DOT1L loop at the entrance to the tunnel is shown in pink in each structure.
Many groups underwent efforts to optimize the pharmacokinetic properties of adenosyl DOT1L inhibitors.430–432,436 The addition of a halogen atom (bromine) to S-adenosyl homocysteine (SAH) resulted in an almost 8-fold greater affinity for DOT1L,431 and application of this finding to EPZ004777 yielded the brominated chemical probe, SGC0946.437 The cryptic hydrophobic pocket revealed by EPZ004777 suggested that DOT1L could accommodate larger hydrophobic substituents.432,437 Structure-guided optimization of EPZ004777 led to the development of the first clinical candidate for DOT1L, EPZ-5676 (Figure 13c). EPZ-5767 (pinometostat), replaced the tert-butyl phenylurea from EPZ004777 with a tert-butyl benzimidazole, connecting it with a cyclobutyl group, rather than a polymethylene chain, introducing more rigidity to the molecule.436 X-ray crystallography of pinometostat and DOT1L revealed that, similar to EPZ004777, EPZ5676 induces a conformational change that widens the cryptic hydrophobic binding pocket and increases favorable interactions with the compound (Figure 13c).432,436 Pinometostat shows greater HMT inhibition, potently decreasing expression of HOXA9 and MEIS1 and robustly inhibiting the proliferation of MLL rearranged leukemia cells. Due to its more suitable pharmacokinetic properties, it was administered by continuous IV infusion to MV4;11 xenograft mice, where it decreased tumor volume, oncogenic gene expression, and H3K79 methylation.436,438,439 In the clinic, pinometostat was well tolerated at levels sufficient to inhibit DOT1L HMT activity, but therapeutic responses were minimal.440 In contrast to the anti-tumor effects seen in preclinical models, clinical responses were far more varied, with 2 of 51 patients achieving complete remission. In pediatric studies, pinometostat treatment caused a brief reduction in peripheral or bone marrow blasts but did not provoke sustained responses.441,442 This showed that the effects of disrupting DOT1L methyltransferase activity are likely not sufficient for single-agent anti-cancer activity.443 Promising in vitro results have pointed toward the potential for combination therapies, but no such studies have been initiated in the clinic.444,445
DOT1L inhibitors have been explored as possible therapies for other cancer types, including glioblastoma, breast cancer, NPM1-mutant leukemia, and others.446–455, Given the unfavorable pharmacokinetics of adenosine nucleoside inhibitors, many research groups have sought to develop compounds with novel chemical scaffolds to address the rapid degradation of previous inhibitors and improve bioavailability.456–458 A variety of screening methods have led to the discovery of non-nucleoside DOT1L inhibitors.459–461 Using SAH production to identify DOT1L inhibitors in a high-throughput chemical screen, a team at Novartis identified a non-nucleoside hit that was optimized to sub-nanomolar potency through structure-based design, ultimately yielding compounds with suitable pharmacokinetic properties for use in vivo (Figure 14a).462 Interestingly, these compounds bind competitively with SAM despite not entering the SAM-binding pocket. They form contacts with Phe243 and Pro130, which are positioned on opposite sides of the entrance to the SAM-binding pocket (Figure 14b).462 Two optimized compounds (Compounds 10 and 11) have been shown to inhibit H3K79me2 deposition, cell proliferation, and target gene expression in DOT1L sensitive cell lines, consistent with EPZ5676 treatment. In MLL-AF9 and MLL-AF6 patient derived xenograft (PDX) mouse models, oral delivery of either compound reduces leukemia burden in bone marrow, spleen and the peripheral blood.463
Figure 14.

Chemical structures of bioavailable non-nucleoside DOT1L inhibitors. (a) Chemical structure of high throughput screening (HTS) hit compound and optimized compound 10. (b) Crystal structure of Compound 10 bound to DOT1L (PDB: 6TEL) showing interactions with key amino acids in green.
The use of such non-nucleoside inhibitors could help advance preclinical assessment of DOT1L target biology in vivo; however, none are currently suitable for clinical investigation due to limited efficacy.462–464 In addition to these methyltransferase inhibitors, there have also been efforts to directly perturb DOT1L through alternative mechanisms, notably by inhibiting its interactions with MLL fusion proteins.403,465–470 DOT1L interacts with the ANC-1 homology domain (AHD) of the common MLL fusion partners, AF9 and ENL, which is required for SEC recruitment and proliferation in MLL-r leukemias. Peptidomimetics based on the DOT1L peptide that binds the AHD of ENL/AF9 validated that disrupting DOT1L protein-protein interactions is sufficient to inhibit proliferation in dependent leukemias.403,465 Small-molecule inhibitors have been designed for the AHD of ENL/AF9 and inhibit proliferation in the low micromolar range, validating the tractability and utility of targeting DOT1L protein-protein interactions via the AHD domain.400,401,428,468,469,471 Despite the structural diversity of DOT1L agents, clinical development appears to have stalled with the termination of EPZ5676 clinical trials. DOT1L has all the makings of a profoundly compelling anti-leukemia target. It is a highly selective dependency in MLL-rearranged leukemia and DOT1L inhibitors show promising anti-cancer effects in preclinical studies. It is therefore surprising that pinometostat did not show more success in the clinic. The fact that it elicited positive pharmacodynamic responses but not therapeutic responses, together with the lack of development for non-nucleoside inhibitors, indicates that DOT1L dependency may simply not be strong enough to elicit sufficient therapeutic responses in patients.
3.1.2. Menin
MEN1, which encodes for the protein menin, was initially discovered through efforts to map the gene on chromosome 11q13 that is responsible for the autosomal dominant familial cancer syndrome, multiple endocrine neoplasia-type 1 (MEN1).472 Germline loss-of-function mutations in this gene lead to tumorigenesis in multiple endocrine organs, suggesting its role as a tumor suppressor.472 Later, it was discovered to form stable interactions with the N-terminal region of the methyltransferase, MLL (Figure 15a).473–475 This N-terminal region is retained in MLL-fusion oncoproteins and menin is required for the pathogenic function of MLL-fusion oncoproteins. Genetic loss of menin or deletion of the menin-MLL interaction site results in decreased gene expression of HOX genes and causes cell differentiation.474 Acute leukemia driven by MLL-fusion oncoproteins are dependent upon menin to maintain stem-like gene expression and evade differentiation, but menin is dispensable in many healthy tissues.474,476 These studies revealed the importance of menin in MLL-rearranged leukemias and motivated the menin-MLL interaction as a possible target for small-molecule inhibition.
Figure 15.

Small molecule inhibitors of menin-MLL. (a) Depiction of minimal MLL peptide interacting with menin (PDB: 3U88). (b) structure of early lead compound MI-2 and its interactions with menin (PDB: 4GQ3). Key amino acid residues are highlighted in green with the novel interactions compared to MLL in maroon. (c) Crystal structure of menin interacting with VTP-50469 (PDB: 6PKC). A novel distal interaction with tryptophan 341 is shown in purple.
The first small-molecule inhibitor of the menin-MLL protein-protein interaction, reported in 2012, was discovered using a fluorescence polarization (FP) assay to screen for compounds that disrupt the interaction of menin with a synthetic and fluorescently-labeled MLL peptide.54 Optimization of a thienopyrimidine screening hit (MI-1) resulted in the discovery of MI-2 and MI-3, which bind to menin with sub-micromolar affinity (Figure 15b). These compounds recapitulate genetic knockout of MEN1 in leukemia cell lines, causing a decrease in HOX and MEIS1 gene expression, as well as inducing G1/G0 cell cycle arrest in MLL-rearranged (MLLr) leukemia cells. A high-resolution crystal structure of MI-2 bound to menin demonstrated that these inhibitors bind the same cavity as the MLL peptide and engage in many of the same interactions (Figure 15b).477 The nitrogen atoms in the thienopyrimidine ring of MI-2 form hydrogen bonds with menin amino acid residues Tyr276 and Asn282 and a gem-dimethylthiazole moiety fits into a cavity formed by Tyr319, Tyr323 and Met322, mimicking interactions that are crucial for MLL binding. This structure revealed the relatively shallow nature of the menin binding pocket, highlighting the impressive ability of these compounds to inhibit a protein-protein interaction (PPI). Structure-guided optimization resulted in the discovery of MI-2–2, which replaced a propyl substituent on the thienopyrimidine core with a trifluoroethyl group and improved potency by approximately 10-fold compared to MI-2.477
Further improvements primarily focused on retaining the thienopyrimidine core477–480 and optimizing pendant groups on the other end of the molecule to improve affinity and enable in vivo study, which was not possible with MI-2–2 due to modest activity and poor metabolic stability.479,481 Efforts to develop the MI-2–2 scaffold for in vivo assessment led to MI-136, which contains a cyanoindole-ring connected to the thienopyrimidine core through a piperidine linker. A further optimized ligand, MI-503, binds menin with single-digit nanomolar affinity and is able to inhibit leukemic gene expression in MLL-rearranged cell lines. It shows suitable pharmacokinetic properties and when tested in MV4–11 xenograft mice,479,481 improves overall survival, and decreases HOX/MEIS1 gene expression. It causes differentiation of leukemia blasts without substantially impairing normal hematopoiesis, providing the first in vivo demonstration that menin inhibition could provide a well-tolerated therapeutic approach in MLL rearranged. Additional improvements of this scaffold have led to orally bioavailable thienopyrimidine menin inhibitors (MI-3454), which induce regression of established tumors, and ziftomenib (KO-539), which is in clinical trials to evaluate its use as a single agent or combinatorial therapeutic in patients with relapsed or refractory AML.56,479,481–485
VTP-50469 was designed from the piperazinyl pyrimidine fragment of MI-2 using an iterative structure-based approach.57 VTP-50469 lacks the fused thiophene moiety found in the thienopyrimidine structures but maintains the key pyrimidine interactions with Tyr276. The cyclohexyl sulfonamide group extends beyond the canonical binding site to form a distant hydrogen bond with Trp341 (Figure 15c). Replacing the piperazine with a diamino spirocyclic moiety kept key cation interactions with Tyr319 and Tyr323 and caused a ~30-fold increase in potency (Ki = 104 pM). VTP-50469 (also SNDX-50469) showed greater cell growth inhibition than previous menin inhibitors in vitro, suppressing the growth of MLL-rearranged PDX models so substantially that a subset of mice remained disease-free and on treatment for over a year.57
VTP-50469 and closely related analogs have consistently shown profound activity as a single agent and combination therapeutic in mouse models of AML.58,483,486 Most notably, this includes not only MLL-r leukemia, but also AML harboring nucleophosmin (NPM1) mutations. NPM1 is among the most frequently mutated genes in AML (>25%), encoding a mutant protein that is mis-localized from the nucleolus to the cytoplasm (NPM1c).487,488 Expression of genes within both the HOXA and HOXB cluster is characteristic of NPM1c-positive AML and is a direct and pathogenic consequence of the mis-localization of the protein.489–491 The activity of menin was first studied in NPM1-mutant AML based on the hypothesis that MLL would regulate HOX expression in diverse leukemia, not only MLL-r.451 Indeed, MI-503 was shown to be highly active against the NPM1c AML cell line, OCI/AML-3, in vitro and to cause a significant, albeit modest, extension of lifespan for mice bearing OCI/AML-3 xenografts. Later studies, using MI-3454 and VTP-50469 to treat PDX models of NPM1c AML showed remarkable activity, with treatment causing disease regression rather than simply slowing its progression.56,58 VTP-50469 treatment was tolerated for over 100 days with overall survival being prolonged by hundreds of days after withdrawal of treatment, highlighting the exceptionally impressive activity of menin inhibition in these aggressive PDX models. It is perhaps instructive to compare the preclinical effects of menin-MLL inhibitors with DOT1L inhibitors, which never showed the same degree of profound and durable responses in mouse models.
The impressive preclinical effects of menin-MLL inhibitors resulted in clinical trials to evaluate their effects in both MLL-r and NPM1c acute leukemia. Revumenib (SNDX-5613), a close analog of VTP-50469, was recently reported to be safe and efficacious in a Phase 1/2 clinical trial evaluating its use in relapsed or refractory disease (NCT04065399, AUGMENT-101) (Figure 16a).59,60 Complete remission or complete remission with partial hematological recovery was observed in 30% of 60 patients.60 The overall response rate to revumenib treatment was 59% in MLL-r leukemia and 36% in NPM1c leukemia, with patients showing on-target decreases in leukemogenic MLL target genes, such as HOXA9/MEIS1, and an increase in the differentiation marker, CD11b.60 No responses were observed in the 8 patients who lacked MLL or NPM1 mutations, supporting the conclusion that the efficacy of revumenib is due to on-target inhibition of menin-MLL. This was definitively established by sequencing of patient samples, which revealed acquired menin mutations as a primary cause of therapeutic resistance.59 Recently, the FDA approved revumenib for relapsed and refractory acute leukemia with MLL rearrangements, marking a major breakthrough for both patients and the field.
Figure 16.

FDA-approved menin inhibitor, revumenib. (a) Chemical structure of revumenib. (b) Crystal structure showing revumenib bound to menin (PDB: 7UJ4). Amino acids involved in acquired resistance (M322 and T344) are shown in green. (c) Schemic depiction of revumenib (hexagon) mechanism of action and resistance mechanisms. Resistance mutations preventing binding of menin to revumenib but allow menin-MLL reassociation and MLL target gene expression.
In four patients who initially responded to revumenib but then relapsed or progressed while on treatment, each harbored at least one mutation in MEN1.59 This motivated a broader search for MEN1 mutations, examining 38 patients before and after nearly two months of treatment. While no mutations were detected before treatment, 12 of 38 patients carried one or more MEN1 mutations after treatment. These mutations, such as M322I and T344M, are closely clustered to a distal region of the ligand-binding pocket that contacts the cyclohexyl sulfonamide moiety, opposite the pyrimidine core (Figure 16b). They selectively disrupt the binding of revumenib to menin without substantially affecting the menin-MLL interaction (Figure 16c). Further study in preclinical models demonstrated that these mutations were sufficient to confer evasive resistance to revumenib and similar mutations within the MLL binding pocket could be identified by unbiased forward genetic approaches, both in tissue culture and in mouse models of acute leukemia. Altogether, these clinical results (i) validated the on-target activity of revumenib, (ii) confirmed the long-standing preclinical hypothesis that menin is a viable target for safe and effective leukemia therapeutics, and (iii) highlighted several opportunities to optimize this class of therapeutics. Interesting, while M327I affects revumenib and MI3454 equivalently, T349M more severely disrupted revumenib binding compared to MI3454, suggesting different scaffolds could possess non-overlapping liabilities to mutations in menin, which might allow for sequential therapy with distinct menin inhibitors to overcome resistance. The concentration of mutations within a region of menin that is dispensable for MLL binding suggests the need to avoid drug contacts with these side chains. However, therapeutic combinations may also minimize the opportunities for resistance to emerge through MEN1 mutations and clinical trials are currently underway to assess revumenib as part of combination regimens.485,492–494
In addition to the thienopyrimidine class of menin inhibitors, several other scaffolds have been reported.478,480,495 Structural analysis of the menin-MLL peptide complex lead to the development of the macrocyclic peptidomimetic inhibitor, MCP-1.495 MCP-1 binds with high-affinity to menin but requires further optimization due to poor cell permeability and its large molecular weight. A hydroxy- and amino-methylpiperidine class, which binds the same MLL pocket, was identified from a high-throughput screen shortly after the initial report of thienopyrimidine inhibitors.480 Since prolonged menin inhibition is required for optimal anti-leukemia effects, the aminomethylpiperidine compound, MIV-6, was utilized to explore the effects of irreversible covalent inhibition (Figure 17a). Structure-based optimization of this scaffold was followed by installation of an electrophilic acrylamide to capture covalent interaction with Cys329. This resulted in M-525, a potent covalent menin inhibitor that shows long-lasting anti-tumor activity in vivo (Figure 17b).496 Extensive efforts to improve this scaffold have resulted in compounds with greater potency against menin and dramatically improved pharmacokinetic properties, including an orally active covalent inhibitor, M-1121.497,498 A covalent inhibitor, BMF-219, is currently in clinical trials for the treatment of patients with relapsed and refractory AML.499
Figure 17.

Development of covalent inhibitors for MENIN. a) Chemical structure of MIV-6 and crystal structure of MIV-6 bound to MENIN (PDB:4OG8). b) Chemical structure of covalent inhibitor, M-525, bound to MENIN (PDB:6B41). Key amino acid resides for the MENIN-MLL interaction are shown in green and the covalently bound cysteine 329 is shown in purple.
The past decade of research has validated both the dependency and pharmacological tractability of the menin-MLL interaction in MLL- and NPM1-mutant cancers. Promising clinical trial results in leukemia patients have shown that these compounds are safe and efficacious as single agents and could be promising in combinatorial therapies.500 The continued exploration and development of more effective menin inhibitors alone, or in combination with other clinically relevant anticancer therapies, aims to address resistance mechanisms observed in vitro, that potentially await clinical validation.501 Interestingly, over the last few years, menin inhibitors have shown promising results in other indications, demonstrating anti-cancer effects across a wide breadth of cancer cell types.55,502–505
3.1.3. WDR5
MLL fusion proteins have been shown to activate oncogenic gene expression in cooperation with the wild-type MLL protein.506,507 MLL fusion proteins typically lack the C-terminal region of MLL, which contains both the SET domain and an arginine-containing sequence that is recognized by a binding site on WDR5. This sequence, called the WIN motif, is conserved across other SET-domain-containing proteins and the conserved arginine residue is required for recognition by WDR5.508,509 The extent of MLL methyltransferase activity depends on the stability of the MLL-WRAD complex, which consists of MLL, WDR5, RbBP5, ASH2L, and two copies of DPY-30.509 Mutations in the MLL WIN motif or the WDR5 WIN binding site (WIN site) can disrupt the MLL-WDR5 interaction, resulting in a loss of MLL methyltransferase activity.509 Since MLL is an activator of oncogenic HOX gene expression, targeting MLL catalytic activity through its interaction with WDR5 could offer a novel therapeutic strategy.
WDR5 is a member of the WD40 repeat domain-containing family and is a core scaffolding component of MLL/SET methyltransferase complexes. WD40 repeat (WDR) domains are comprised of a conserved 40 amino acid sequence ending with tryptophan and aspartic acid residues. Most WDR proteins, like WDR5, contain 7 repeats and forms a seven-bladed propeller conformation (Figure 18a). There are two binding sites on WDR5 through which its protein-protein interactions occur, the WBM (WDR5 Binding Motif) site and the WIN (WDR5 Interaction) site (Figure 18b).510 The WIN site is an arginine binding cavity on the surface of WDR5 that interacts with the MLL WIN motif as well as functions to read H3K4me marks.38,511,512 Genetic loss of WDR5 causes global decreases in H3K4 methylation and, ultimately, leads to inactivation of many essential genes.38,511 MLL chromatin localization, however, is unchanged after WDR5 loss, suggesting that WDR5 regulates the methyltransferase activity of MLL and not its localization.38
Figure 18.

WDR5 WIN site ligandability. (a-b) WDR5 structure showing the WIN site (pink) and the WBM site (blue) located on opposing faces of the seven-bladed barrel structure (PDB: 3EG6). (c) Development of WDR5 WIN site inhibitors based on the MLL WIN peptide sequence. The key arginine motif is highlighted in orange.
The arginine residue on the MLL WIN motif (R3765) is required for binding to the central cavity of WDR5 and inspired the development of peptidomimetic inhibitors containing a common arginine residue.513–516 Motivated by the promising effects of peptidomimetics, multiple groups sought small-molecule inhibitors of the MLL-WDR5 interaction.517–521 One group used a fluorescent polarization (FP)-based high-throughput screen to identify a single hit compound, the benzamide WDR5–0101.522 A follow-up screen with structurally similar benzamide compounds identified WDR5–47 (Figure 19a), which was structurally determined to occupy the central arginine binding cavity on WDR5. It binds competitively with and displaces the binding of WIN motif containing proteins, such as MLL.523 These discoveries inspired further compound optimization that, ultimately, identified numerous selective small-molecule ligands for WDR5, like OICR-9429 (Figure 19a).523–527 OICR-9429 effectively inhibits the methyltransferase activity of MLL in vitro and decreases the proliferation of MLL-rearranged cells.
Figure 19.

Small-molecule inhibitors of the WDR5 WIN domain. (a) Development of the WIN site inhibitor OICR-9429. (b) Chemical structure of bioavailable WIN site inhibitors DDO-2093 and DDO-2213. (c) Gene effect scores for the common essential gene, WDR5 (DepMap).
Additional work on this scaffold led to the discovery of compounds with suitable pharmacokinetic properties for in vivo use.528,529 These compounds retained the methylpiperazine and central phenyl groups of OICR-9429 but included modifications to the C5 position of the phenyl ring (Figure 19b). One compound, DDO-2213, was discovered using a scaffold hopping approach, resulting in the replacement of the benzamide with a pyrimidine ring.528 DDO-2213 binds to WDR5 with high affinity and selectively reduces the MLL methyltransferase activity over other HMT proteins.528 Another optimized compound, DDO-2093, retained the core scaffold of OICR-9429 but replaced the phenyl morpholine ring of OICR-9429 with a phenyltriazole moiety (Figure 19b). Both compounds have been shown to possess suitable properties for assessment in mouse xenograft models of leukemia, each causing a decrease in tumor mass, tumor weight, and HOXA1/MEIS1 expression without obvious toxicities. Promisingly, DDO-2213 has suitable pharmacokinetic properties to be administered orally. These compounds are the first inhibitors of the WIN site to show proof-of-concept in vivo. However, WDR5 is a common essential protein (Figure 19c), and it remains unclear whether a safe therapeutic window can be achieved with WDR5 inhibition.
The WDR5 inhibitor OICR-9429 has been used to fashion WDR5 PROTACs.530–533 The resulting degraders showed greater efficacy in cells than OICR-9429 and were highly selective for WDR5.530–532 One CRBN-based PROTAC showed degradation of WDR5 and off-target degradation of the CRBN neo-substrates, IKZF1 and IKZF3. This degrader elicits greater inhibition of WRAD complex target gene expression, attributed to decreased MLL occupancy on chromatin, and more effectively suppresses the growth of MV4;11 and EOL-1 AML cell lines compared to a WDR5-selective degrader.533 Interestingly, despite WDR5 being considered common essential, the WDR5 and IKZF1/3 degrader lacks general toxicity, suggesting that WDR5 may still present a viable target for MLLr leukemias.533
WIN site inhibitors and PROTACs are joined by WBM site inhibitors, where the oncogenic transcription factor, MYC, binds to WDR5.534 Multiple groups have shown that disruption of the WBM site prevents MYC from associating with a subset of its target genes,535,536 providing support for targeting MYC through its interaction with WDR5. A high-throughput screen for small-molecule inhibitors of this interaction revealed a biarylsulfonamide hit that binds the WBM site.537 Structure-guided optimization afforded compounds that inhibit the WDR5 interaction with MYC in cells, along with structurally related negative control compounds that do not bind WDR5.537 Hits from an NMR-based fragment screen combined with these biarylsulfonamide inhibitors allowed for structure-guided scaffold “merging” to design new hits with improved drug-like properties.538 The compound with the highest affinity, compound 12 (KD = 0.10 nM), causes a fourfold decrease in WDR5-MYC association in cells and decreases MYC binding to WDR5-dependent chromatin targets.538
Both WIN site inhibition and WBM disruption appears to cause a reduction of WDR5 on select chromatin targets, specifically those encoding ribosomal proteins.517 WDR5 functions at these targets to recruit MYC through its WBM. Therefore, it is possible that disrupting either the WIN site or the WBM causes the same result – reduced MYC recruitment to genes that encode ribosomal biogenesis proteins.534 While its importance in MLL-rearranged leukemia is supported by both chemical and genetic studies,539 a role for WDR5 in ribosomal biogenesis may be consistent with its annotation as a common essential protein. Site-specific WDR5 inhibitors, which affect a subset of WDR5 functions,540,541 may therefore allow for selective targeting of non-essential chromatin related functions of WDR5 while avoiding toxicity predicted by genetic loss of function. Contrary to earlier reports, the effects of WIN inhibition are caused by disruption of WDR5 binding to chromatin and cannot be attributed directly to changes in histone methylation, which only occur subsequently,517,541 indicating a less privileged link to MLL fusion tumors than previously hypothesized.
CRISPR/Cas9-based loss-of-function genetic modifier screens have shown that the cellular response to WIN inhibitors relies on decreased ribosomal gene transcription and activation of p53 response pathways, with p53-deficient cancer cell lines showing a lack of sensitivity to WIN inhibition.542 These studies afford more comprehensive understanding of the mechanisms underlying the sensitivity to WDR5 WIN inhibitors, which may help to identify biomarkers that predict responsiveness to WDR5 antagonists in the diverse set of cancers that have been shown sensitive to WDR5 loss-of-function.543–548 Overall, WIN inhibitors appear to target an oncogenic role of WDR5 that partially avoids its common essential functions, implying that there may be opportunities to develop selective anti-cancer therapeutics targeting WDR5.
3.1.4. ENL
ENL was first identified as a potential acute leukemia target in two studies reported together in 2017.549,550 In one, a genome-scale CRISPR/Cas9-based loss-of-function screen identified ENL as an essential gene in leukemia driven by MLL fusion oncoproteins.549 In another study, the prevalence of ENL and AF9 in MLL fusion oncogenes motivated a reverse genetic approach to study the function of wild-type ENL and AF9 alleles in MLL-rearranged leukemias.550 This revealed ENL, but not AF9, as being required for the fitness of MLL-rearranged acute leukemia cells. Both reports demonstrated normal hematopoietic stem and progenitor cells are insensitive to ENL loss-of-function, suggesting ENL-targeted therapeutics might possess limited on-target toxicity to normal tissues, a finding that is further supported by its profile in the DepMap dataset (Figure 20a).
Figure 20.

Exploiting ENL dependencies in MLL-rearranged leukemias. (a) Gene effect scores for ENL in cell lines grouped by MLL-rearrangement (DepMap). (b) H3K27ac (blue) occupying the YEATS domain of ENL (PDB:5J9S). Tyrosine 78 (Y78) and Phenylalanine 59 (F59) on loops 6 and 4, respectively, are shown in green. (c) JalView sequence alignment of all 4 human YEATS domains from ENL, AF9, YEATS2 and YEATS4. Darker red corresponding to higher conservation. Conserved secondary structure is depicted below alignment L corresponds to loops and arrows correspond to beta sheets. (d) Chemical structure of ENL/AF9 YEATS domain inhibitor, SGC-iMLLT1 (top), and the X-ray crystal structure showing SGC-iMLLT1 (blue) bound to ENL (bottom) (PDB: 6HT1). At top, SGC-iMLLT is colored by its orientation of binding to the ENL YEATS domain relative to acetyllysine. Green depicts binding to the entrance of the tunnel into which acetyllysine binds. Pink shows the central amide that mimics the binding of the acetyllysine amide and blue depicts binding to the exit of tunnel. (e) Chemical structures of other YEATS domain inhibitors, colored the same as SGC-iMLLT1 including coloring for their E3 ligase. (f) Chemical structures of ENL PROTACs and a molecular glue degrader.
ENL functions as a chromatin reader protein through its conserved YEATS domain, first reported in 2014 to bind acetylated lysine side chains (Figure 20b).28 Four YEATS domains (named for the founding members Yaf9, ENL, AF9, Taf14, and Sas5)551 are encoded in the human genome (ENL, AF9, GAS41/YEATS4 and YEATS2) and each is part of chromatin regulatory complexes.552,553 This conserved domain (Figure 20c) is organized into an immunoglobulin fold and forms an open-ended channel between loops 4 and 6 to accommodate acyllysine side chains (Figure 20b).28,554–557 Early structural and mutagenesis studies of the YEATS domain identified key amino acids involved in binding acetyllysine, which makes π-π-π stacking interactions with Tyr78 and Phe59.549,550,554,558 Electrostatic interactions play key roles in acyllysine recognition, including hydrogen bond interactions with the backbone amide of Tyr78 and the Ser58 side chain,559 CH-π interactions with Phe28 and the tip of the acetyllysine, and amide-π interactions with Tyr78 and Phe59 (Figure 20b).28,560,561
Based on the structural analyses of ENL/AF9 interactions with acetyllysine, point mutations have been discovered that decrease acetyllysine affinity in vitro and prevent ENL and AF9 localization to chromatin in cells.28,549,550 In acute leukemia cells that depend on ENL for survival, these mutant alleles were unable to complement the loss of wild-type ENL, suggesting that interactions between ENL YEATS and acetyllysine are essential for its pro-leukemic function.549,550 Based on this discovery, there have been widespread efforts to identify small molecules that competitively inhibit the ENL YEATS domain. The first reported inhibitor, SGC-iMLLT, originated from the Structural Genomics Consortium (SGC) in a screen of 40,000 small molecules that measured the displacement of a synthetic histone peptide from the ENL YEATS domain.559 Optimized from a benzimidazole hit, SGC-iMLLT1 contains a central amide bond that mimics the acetyllysine substrate by engaging in key hydrogen bond interactions with Ser58, Tyr78, and Ala79 (Figure 20d). The flanking indazole and benzamidazole moieties make contacts with the entrance and exit of the binding tunnel, respectively (the entrance and exit are defined relative to the insertion of acetyllysine). Binding of the native acetyllysine substrate and SGC-iMLLT1 is facilitated by conformational flexibility of Tyr78, which is a more constricted tryptophan residue in YEATS2/4, potentially contributing to its selectivity for ENL/AF9 (Figure 20d). Many ENL/AF9 YEATS domain inhibitors have since been reported, which has revealed recurrent structural themes of YEATS domain inhibitors (Figure 20e). These contain, for example, central amide or urea groups flanked by piperazine, imidazopyridine, and isoxazole groups that contact the exit tunnel and indazole, cyclobutyl, and indane groups that contact its entrance.559,562–571
The imidazopyridine inhibitor, SR-0813, was optimized from a FRET-based HTS hit using a sulfur(VI) fluoride exchange (SuFEx) based high-throughput medicinal chemistry approach (Figure 20e).562 SR-0813 is potent and selective for ENL/AF9 YEATS domains, (ENL YEATS KD = 30 nM) with minimal effects on YEATS2 at high concentrations (10 – 50 μM) and virtually no binding to YEATS4 by SPR. SR-0813 inhibits the proliferation of ENL-dependent, MLL-rearranged leukemias by evicting ENL from chromatin through competitive binding with acetyllysine, resulting in a concomitant downregulation of ENL target gene expression. Promisingly, SR-0813 treatment showed no detectable anti-proliferative effects in cell lines insensitive to ENL loss, indicating that the observed effects in ENL-dependent cell lines are likely due to its on-target activity. This research recapitulated the effects of ENL degradation and validated that the pharmacological inhibition of the ENL YEATS domain represents an effective way to exploit ENL dependencies in MLL rearranged leukemias.562 Despite the in vitro potency of SR-0813, however, its poor metabolic stability precluded it from further assessment in vivo.
Modification of SGC-iMLLT produced the first inhibitor capable of probing ENL/AF9 YEATS domain in vivo. This compound, TDI-11055, replaced the benzimidazole of SGC-iMLLT with a pyrrolopyridine group to yield much improved pharmacokinetic properties, including acceptable oral bioavailability (Figure 20e).565,567 TDI-11055 selectively binds the ENL/AF9 YEATS domains (KD = 119 nM), prevents the association of ENL with chromatin, and reduces the expression of ENL target genes in MLL-rearranged cell lines. Encouragingly, TDI-11055 is well tolerated in vivo and produces anti-cancer effects in xenograft models of MLL-rearranged leukemias. Furthermore, TDI-11055 was used to uncover a dependency on ENL in NPM1-mutant leukemia and it showed strong anti-cancer effects in animal models of the disease.567 This aspect of ENL target biology is notably similar to menin, which is a targetable dependency in both MLL-rearranged and NPM1-mutant leukemia. A CRISPR/Cas9-based ENL mutagenesis strategy was able to identify an in-frame deletion that confers resistance to TDI-11055, validating its anti-leukemic effects as being on-target. Interestingly, this deletion overlaps a hotspot region of recurrent gain-of-function insertion-deletion mutations observed in Favorable Histology Wilms tumors that cause increased biomolecular condensate formation.572–578 Another reported orally bioavailable inhibitor, 563SR-C-107, is based on an amido-triazolopyridine screening hit,568 with the addition of the pyrrolopyridine group from TDI-111055 to improve pharmacokinetic behavior..564,565,567
SR-0813, TDI-11055, SR-C-107, and other chemical probes for the ENL/AF9 YEATS domains elicit anti-proliferative effects that are highly selective for ENL-dependent leukemias, and their suppression of gene transcription is similarly selective for ENL target genes.562,564,567 However, their anti-proliferative effects are delayed, requiring several cell divisions and more than a week of drug exposure to cause an effect in vitro. The strengths of their effects are also weaker than compounds inhibiting similar targets, like menin, with high doses required to inhibit ENL-dependent leukemia growth in vitro and in vivo (both TDI-11055 and SR-C-107 are used at 200 mg/kg doses in mice). While these compounds leave significant room to improve affinity, it is likely that YEATS domain inhibition elicits a sub-maximal effect compared to degradation or genetic knockout.549,550,563,579,580
YEATS domain ligands have been used to design several ENL/AF9 PROTACs. A precursor to SR-0813 was used to discover the first ENL/AF9 degrader, the CRBN-based PROTAC, SR-1114.562 However, SR-1114 also induces degradation of the transcription factor IKZF1, a common neosubstrate target of CRBN-based degraders, limiting its deployment as a chemical probe. Compound 1, which used SGC-iMLLT to construct a more potent ENL degrader (3-fold over SR-1114) also elicits anti-leukemic effects against ENL-dependent cell lines, but its selectivity for ENL has not been evaluated at proteome scale.580 MS41, which was designed by linking the ENL/AF9 YEATS ligand, PFI-6,566 to a VHL ligand, is a potent (DC50 = 3.5 nM) and selective degrader of ENL.579 In vitro, it selectively inhibits the growth of ENL-dependent leukemia cell lines and in vivo, it suppresses the growth of an MV4;11 xenograft model of acute leukemia without causing toxicity to normal hematopoiesis. Joining these heterobifunctional PROTACs is dHTC1, a molecular glue degrader based on the SR-0813 scaffold (Figure 20f).563 dHTC1 was developed using a new, prospective approach for molecular glue discovery,563 which relies on sulfur(VI) fluoride exchange (SuFEx)-based transformations for parallel ligand diversification.562,581–583 This approach allows for the discovery of rare chemical modifications that can transform pre-existing ligands into molecular glues, affording dHTC1, a potent and selective ENL degrader.563 It binds to CRBN with high affinity only after pre-forming a bimolecular complex with ENL, inducing ENL degradation and eliciting anti-leukemic effects in vitro and in vivo.563 While further comparative analysis of YEATS domain inhibitors and ENL degraders is needed, and additional improvement in YEATS domain affinity are likely possible, these early-stage degraders strongly suggest that degradation would be the preferred method to perturb ENL therapeutically. These degraders are excellent tool compounds for assessing ENL degradation as potential therapeutics, but no ENL-targeted agents have advanced into clinical investigation.
YEATS domain inhibitors have also been reported for YEATS4, which was originally identified as an overexpressed oncoprotein in glioma, for which it was named GAS41 (glioma amplified sequence 41).584 Like other YEATS-domain-containing proteins, YEATS4 regulates transcription by recognizing post translational modifications on histone tails and recruiting multiprotein complexes to influence gene expression.585 YEATS4 amplifications have been found in many indications including sarcomas, non-small cell lung cancers (NSCLC),586 colorectal, and gastric cancers.587–591 Genetic knock out of YEATS4 has been shown to impair proliferation in NCSLC with YEATS4 amplifications.592 The prevalence of YEATS4 amplifications in various cancer types and its role in transcriptional regulation suggest that it could be tractable target for small-molecule inhibitors, but it is also annotated as a common essential gene by DepMap (Figure 21a).
Figure 21.

YEATS domain of YEATS4 is chemically tractable. (a) Histogram of gene effect scores for common essential gene YEATS4 (DepMap). (b) YEATS4 YEATS domain inhibitors. Chemical structures are colored based on their orientation of binding to the acetyllysine channel, as described in Figure 19 (blueD= exit, pink = central amide, and green = entrance).
The first reported YEATS4 YEATS domain inhibitor was developed from fragment library screening hit, a thiophene amide scaffold that was later optimized to contain a benzothiazolyl substituent (compound 11).593 Based on findings that YEATS4 binds to histones more strongly as a dimer, a bivalent analog of compound 11, connected by a short polymethylene linker, was developed (Compound 9) (Figure 21b).593,594 This compound decreases the expression of YEATS4 target genes and showed moderate antiproliferative effects in cancers with YEATS4 amplifications. A second reported scaffold, a pyrazolopyridine series of potent and selective YEATS4 inhibitors with suitable pharmacokinetic properties for use in vivo, was developed from a virtual screening hit.595 Interestingly, although YEATS4 is a common essential protein, these compounds show minimal cellular toxicity, which suggests that the YEATS domain of YEATS4 may not be required for its cell-essential functions.592
3.1.5. ASH1L
ASH1L (absent, small, or homeotic-like 1) is a large, 333 kDa protein that contains a SET domain, bromodomain, PHD, and BAH domain, as well as a long, unstructured N-terminal region. The SAM-dependent SET domain catalyzes H3K36 mono- and di-methylation of histone 3 lysine 36 (H3K36) while the other domains bind to posttranslational modifications.596 It is expressed in hematopoietic stem and progenitor cells (HSPCs), where is regulates HOX gene expression and supports self-renewal.597 This activity is reminiscent of MLL, with the two proteins sharing a conserved functional relationship.598–600 Indeed, ASH1L regulates an essential writer-reader-eraser axis in MLL-rearranged leukemias, where it catalyzes the deposition of H3K36me2 marks that are recognized by the MLL interactor, LEDGF, and colocalizes with MLL to activate a leukemogenic transcriptional program.601 Loss of ASH1L mitigates leukemogenesis in MLL-transformed cell lines and xenografts, altogether supporting a model in which ASH1L is critical in MLL-rearranged leukemias.601 Furthermore, in a mouse model harboring an in-frame deletion of the Ash1l SET domain, bone marrow progenitors are resistant to transformation by MLL-fusion oncogenes but not by other control oncogenes, leading to the pursuit of ASH1L methyltransferase inhibitors.602
The first ASH1L inhibitors originated from an NMR-based screen of approximately 1,600 compounds. The chemical shifts from a thioamide hit mapped to a region of the protein near an autoinhibitory loop, which was confirmed via point mutation. Medicinal chemistry optimization of this compound yielded a compound, AS-5, that binds to a buried site and is conformationally restrained by the concomitant binding of the endogenous SAM cofactor (Figure 22). This compound features an indole scaffold with hydroxyethyl and aminomethyl substituents that engage in stabilizing hydrogen-bonding interactions within the pocket. These were replaced with 1-trifuloromethylsulfonylpiperidine and methyl azetidine moieties, respectively, to yield the lead compound AS-99, which binds in a similar fashion. The thioamide engages in a chalcogen-bonding interaction with the backbone carbonyl of H2193 and replacement of the sulfur with oxygen abolishes activity, providing an excellent negative control compound (Figure 22). AS-99 binds the SET domain with a KD of 0.89 μM and inhibits ASH1L with an IC50 of 0.79 μM. It is selective against 20 other HMTs including NSD1, NSD2 and NSD3, as well as SETD2, likely due to the lack of structural conservation in their autoinhibitory loops. AS-99 is antiproliferative in cell lines harboring MLL fusions and recapitulates the pro-differentiation phenotype seen with genetic ASH1L knockdown and SET domain deletion. A decrease in H3K36me2 abundance is seen after drug treatment, which produces a dose-dependent decrease in the transcription of gene signatures associated with MLL fusions. The compound also decreases leukemic burden in mice. Altogether, this report provides a compelling proof-of-concept for selectively inhibiting ASH1L, further demonstrating its potential as a therapeutic target.602
Figure 22.

Summary of medicinal chemistry results from hit (AS-5) to lead (AS-99) inhibitor of ASH1L., as well as negative chemical control (AS-nc).
3.2. PRC2
Polycomb repressive complex 2 (PRC2) is a multi-subunit chromatin regulatory complex best known for its role in transcriptional repression. Mechanistically, PRC2 functions through its enzymatic subunit, enhancer of zeste homologue 2 (EZH2), which catalyzes the methylation of H3K27, a canonically repressive histone PTMs (notably, PRC2 is the exclusive writer of H3K27me3). EZH2 exists in the PRC2 complex alongside suppressor of zeste 12 homolog (SUZ12) and embryonic ectoderm development (EED), a scaffolding and chromatin reader protein, respectively, which are indispensable for the histone methyltransferase activity of PRC2 (Figure 23a).326,603,604 As first discovered in Drosophila and later shown to be conserved in humans, PRC2 activity is counteracted by that of SWItch/Sucrose Non-Fermentable (SWI/SNF).605,606 While SWI/SNF remodels chromatin to a more open and active regulatory state through ATP-dependent remodeling of nucleosomes—allowing transcription factors and transcriptional machinery to access DNA within cis-regulatory elements—PRC2 mediates gene-silencing through its methyltransferase activity. This oppositional relationship is also directly competitive. Genetically engineered systems that allow for rapamycin-dependent recruitment of the BAF complex to a specific genomic locus have shown that the most immediate effect of BAF recruitment, even preceding chromatin decondensation, is the eviction of PRC2, as evidenced by anti-Ezh2 ChIP and proxied by anti-H3K27me3 ChIP.326
Figure 23.

PRC complexes mediate repressive transcriptional markers and play a role in malignant transformation. (a) PRC1 and PRC2 complexes. (b) PRC2 exists in precise regulatory balance with SWI/SNF. Genetic aberrations that disrupt this balance play pivotal roles in oncogenesis.
Genetic alterations that disrupt the balance between BAF and PRC2 can lead to tumor formation (Figure 23b). Hyperactivation of PRC2 in cancer can be caused by mutations607–609 and amplifications610–615 of EZH2, as well as by the inactivation of certain BAF subunits.616–621 Activating point mutations at tyrosine 641 (Y641) (which include Y641F, Y641N, Y641S, and Y641H) are present on a single allele of EZH2 in a large percentage of non-Hodgkin lymphomas—a diverse group of cancers that arise from malignant proliferation of B-, T-, or NK-lymphocytes outside the classic Hodgkin’s lymphoma subtype—creating an oncogenic hypermorph that works cooperatively with the wild-type protein to drive a chromatin architecture with elevated levels of H3K27me3 (Figure 23b).607,622
Wild-type EZH2 is most active as a mono-methyltransferase, with substantially reduced activity for catalyzing the second and third methylation. In contrast, mutated EZH2 has highest activity for the final methylation and little activity for catalyzing mono-methlyation. Hyperaccumulation of trimethylated substrates occurs in cells expressing both the wild-type and mutant enzymes, which cooperate to catalyze the early and late methylations.607,622 EZH2 overexpression can also result from loss-of-function mutations within the SWI/SNF complex. For example, malignant rhabdoid tumors (MRT)—a rare and highly aggressive pediatric cancer most commonly originating in the kidney or CNS—are nearly uniform in harboring biallelic inactivation of SMARCB1,623 which leads to EZH2 overexpression and hypersensitivity to EZH2 loss-of-function.624 These genetically restricted sensitivities to EZH2 loss-of-function motivated the development of PRC2 inhibitors. Because EZH2 is the enzymatic subunit, early drug discovery efforts focused heavily on orthosteric inhibitors of its SET catalytic domain. While this has yielded clinically approved agents, to be discussed in the following subsections, resistance mutations in EZH2 highlight an important limitation of targeting only the catalytic site. This has led to growing interest in other PRC2 subunits—particularly EED, whose interaction with EZH2 is required for enzymatic activity—as alternative therapeutic entry points. For example, the allosteric EED inhibitor EED226, also to be discussed, can suppress PRC2 activity even in the setting of EZH2 mutations, establishing a compelling rationale for exploring non-catalytic PRC2 subunits as drug targets.
3.2.1. EZH2
The molecule 3-deazaneplanocin A (DZNep), an S-adenosylhomocysteine hydrolase inhibitor, elicits downstream by-product inhibition of SAM dependent methyltransferases, a large superfamily of enzymes, consisting of over 200 enzymes in humans alone to which EZH2 belongs.625–627 While the discovery of this molecule contributed to initial investigations of protein function, it was found to be insufficiently specific, resulting in changes to global chromatin architecture that could not be reliably sourced to EZH2 inhibition. This called for direct-acting and selective small molecules that inhibit EZH2/PRC2 activity.
Groups at Epizyme, GSK, and Novartis ushered in the first of these molecules with the disclosure of SAM-competitive EZH2 inhibitors in 2012.628–630 Epizyme first reported EPZ005687, which was discovered through the screening of 175,000 compounds against recombinant PRC2, followed by an additional 5,000 compounds representing 25 standout structural clusters (Figure 24a). The resulting hit was a pyridone-containing compound, which was further optimized to improve solubility and potency, yielding EPZ005687.628 This first report highlighted what would continue to be a common hurdle in PRC2 inhibitor discovery – that it is difficult to generate both apo- and co-crystal structures of ligand bound to full PRC2 complex, making structure-based drug design and optimization difficult (although recent efforts toward structural elucidation may be changing this for future work).631–635 Nevertheless, the reported compound demonstrated competitive antagonism with the native SAM substrate, while remaining noncompetitive with oligonucleosomes. EPZ005687 selectively inhibits EZH2 (Ki = 24 nM) over EZH1 and other methyltransferases (50 and >500 - fold selectivity, respectively) resulting in the decrease of H3K27me3 marks in diverse cell types while sparing other post-translational modifications. Consistent with genetic studies, the antiproliferative activity of EPZ005687 is selective for EZH2-mutant cancer cell lines over EZH2-wildtype cancers.628
Figure 24.

Disruption of PRC2 by small-molecule inhibitors of EZH2 methyltransferase. (a) Prototypical chemical probes targeting EZH2. (b) Clinically evaluated small-molecule inhibitors of EZH1/EZH2.
Contemporaneously, GSK disclosed a similar pyridone EZH2 inhibitor, which had resulted from the screening of an internal compound collection.629 Like EPZ005687, the key pharmacophore consists of a pyridone, 4,6-disubstitution pattern, a linking amide, and a branched alkyl group at the 1-azaindazole. Medicinal chemistry optimization led to GSK126, an EZH2-selective, SAM-competitive, and in vivo-active inhibitor that elicits a transcriptional activation profile correlated to antiproliferative activity in EZH2-mutant cancers over wild-type cancers (Figure 24a).629,636 EI1, another pyridone EZH2 methyltransferase inhibitor, was also disclosed in 2012 by Novartis (Figure 24a). EI1 invokes cell cycle arrest and apoptosis in EZH2-mutant DLBCL cancer cells and reactivates expression of PRC2 target genes.630 A series of other compounds, UNC1999, CPI-169 and CPI-360, were disclosed shortly after and used to validate similar biological activity, namely, an antiproliferative phenotype in EZH2-mutant DLBCL cells that correlates with a global reduction of H3K27me3 marks.630,637,638
The first, and as-yet only, EZH2 inhibitor to be approved by the FDA is EPZ-6438, or tazemetostat (Figure 24b).624,639 Tazemetostat featured remarkable improvements in potency, selectivity, and pharmacokinetic properties compared to the previous tool compound, EPZ005687. In lymphoma cell lines bearing EZH2 point mutations, treatment with tazemetostat leads to G1 arrest and induction of apoptosis, with cell death correlating to a commensurate reduction in H3K27me3 abundance. PRC2/EZH2 target genes are upregulated upon compound treatment, indicating a beneficial transcriptional response elicited by a change in the chromatin landscape. In mice and rats, the compound demonstrates low clearance with steady volumes of distribution, oral bioavailability, and rapid absorption, among other beneficial drug-like properties. In mouse xenograft studies, doses as low as 34.2 mg/kg are sufficient to inhibit tumor growth while higher doses, above 114 mg/kg, are sufficient to eradicate xenograft tumors, with no tumor regrowth after drug cessation. These promising results led to the clinical exploration of tazemetostat and its eventual FDA approval for follicular lymphoma in 2020.640,641 In a phase ll clinical trial (NCT01897571) of 42 patients with EZH2 mutant follicular lymphoma, there was an overall response rate of 69% (12% complete and 57% partial responses); these numbers were lower in patients with EZH2 wild-type follicular lymphoma, concordant with preclinical mechanistic results.642
Epizyme further optimized the tazemetostat scaffold, seeking to mitigate oxidative metabolic liabilities associated with the pyridone-benzamide core. Modification of the pyran of tazemetostat, addition of a methoxyethyl group to the cyclohexaylamine, and replacement of the second benzene ring with an acetylene linker produced EPZ011989.643 While EPZ011989 shows excellent oral bioavailability and improved metabolic stability, it did not supplant the development of tazemetostat. Others have explored the degree of selectivity between EZH2 and its paralog, EZH1. Tazemeteostat and EPZ011989 are 35-fold and 15-fold selective for EZH2 over EZH1, respectively. Compounds that inhibit EZH1 more effectively than tazemetostat, such as the dual inhibitor valemetostat, may be able to address some cancers where EZH1 can compensate for EZH2, although dual inhibition carries potential limitations, such as increased toxicity (Figure 24b).644 While valemetostat does not carry the FDA approval that tazemetostat does, it is being evaluated in trials for peripheral T-cell lymphoma (PTCL) and adult T-cell leukemia/lymphoma (ATLL).645
Tazemetostat is also highly efficacious against MRT. The biallelic inactivation of the SWI/SNF subunit, SMARCB1, which is pathognomonic for MRT, sensitizes cells to inhibition of EZH2.606 In a 2013 report, EZH2 inhibition was shown to inhibit the growth of SMARCB1-deficient MRT cells in vitro and cause tumor regression in xenograft models of MRT in vivo.624 Based on positive clinical studies,640 tazemetostat has now been approved for the treatment of SMARCB1-mutant sarcoma, but response rates seen in sarcoma are well below those seen in EZH2-mutant lymphoma.
3.2.2. EED
EZH2 functions within the PRC2 complex together with EED and SUZ12, and disrupting these interactions can also block PRC2 activity. A hydrocarbon-stapled peptide called SAH-EZH2 was designed to mimic a short helical region of EZH2 that normally binds EED. By occupying this interface, SAH-EZH2 prevents EZH2 from associating with EED, leading to EZH2 destabilization. In leukemia models, this approach halted cell growth and promoted differentiation, illustrating that PRC2 can be inhibited by breaking apart the complex rather than directly targeting its catalytic site.646,647
EZH2 catalytic activity can also be modulated through inhibition of EED, which allosterically activates EZH2 by binding H3K27me3 and other methylated lysines.604,647–649 EED is a WD40-repeat protein, folding into a seven-bladed β-propeller structure that recognizes trimethyllysine within an aromatic cage.604,650 In 2017, groups at Novartis and Abbvie reported the potent and selective allosteric PRC2 inhibitors, EED226 and A-395, which function by disrupting the interaction of EED with methylated lysine (Figure 25a).651,652 The EED226 scaffold was identified in a high throughput screen using homogenous time-resolved fluorescence (HTRF) to detect the methylation of H3K27 by recombinant PRC2,651,653,654 a screen that had previously yielded SAM-competitive EZH2 inhibitors.630 Characterization of non-competitive inhibitors led to the discovery of a triazolopyridine scaffold that binds competitively with H3K27me3 to the WD40 domain of EED.651,654 Treatment of G401 cells with EED226 leads to an inhibition of H3K27 methylation, which is concomitant with a transcriptional response that mirrors orthosteric EZH2 inhibition.651 EED226 is effective in vivo in DLBCL xenograft models and represents an opportunity to overcome resistance to orthosteric EZH2 inhibitors. In preclinical models, forward genetic approaches have identified secondary EZH2 mutations causing acquired resistance to SAM-competitive EZH2 inhibitors.655,656 In cellular models, EED226 was able to overcome this resistance, suggesting that combination or sequential therapy with EZH2 and EED inhibitors could prevent or overcome resistance.651
Figure 25.

EED inhibitors and other methods to disrupt PRC2. (a) First-in-class EED chemical probes. (b) Other representative PRC2 inhibitors. (c) Summary of improvement of EED degrader UNC6852 to UNC7700.
A-395, a structurally divergent EED inhibitor that also binds the H3K27me3 recognition pocket of EED, was reported contemporaneously with EED226 (Figure 25a). It effectively reduces H3K27 methylation, inhibits cancer cell proliferation, and demonstrates potent anti-tumor efficacy in preclinical xenograft models.652 Importantly, A-395 retained activity in cell lines resistant to EZH2 inhibitors, suggesting its potential to overcome therapeutic resistance. Its PK properties, however, limit its clinical application, although it, alongside EED226, represented a valuable chemical tool for probing PRC2 biology.
In 2022, Novartis disclosed significant improvements to the tool compound EED226, fashioning a more drug-like molecule MAK683 (Figure 25b). EED226 suffers from undesirable solubility and permeability, and the furan moiety is prone to oxidation and reactive metabolite formation, making it a poor clinical candidate. To this end, Novartis focused on three main moieties: (1) the furan, which resides in a deep pocket that prefers electron rich aromatic systems; (2) the triazolopyrimidine core, which resides in the aromatic cage that prefers electron deficient systems; and (3) the solvent-exposed sulfone, which is inherently permissive to change and a desirable location for property modulation. Studies illustrated that CYP3A4 and CYP11A1 act on EED226 to form reactive species, particularly through modification of the furan.657,658 Replacement of the furan with a dihydrobenzo furan yielded significant improvements and fluorination of the ring helped to improve off-target liabilities. Additional modifications to shield the compound from heme ligation and decrease its metabolic reactivity produced MAK683, which is effective against Karpas422 xenograft models and demonstrates favorable cross-species ADME properties, including little to no CYP inhibition.657 It was eventually progressed into clinical evaluation for the treatment of DLBCL, but a full report of the trial results has not been disclosed, and further development was halted before Phase 2.659
Non-enzymatic scaffolding functions of EZH2 have been reported to be important for SWI/SNF-mutant tumors, providing a rationale to develop targeted protein degradation approaches for EZH2.53,660,661 Small-molecule inhibitors of EZH2 and EED have enabled the development of PROTACs and other targeted degradation approaches to inactivate PRC2 function. The first example, MS-1943, which degrades EZH2 by hydrophobic tagging, was synthesized by appending an adamantane group to the EZH2 ligand C24 (Figure 25b).662 This molecule degrades EZH2, causing collateral degradation of SUZ12 but not EED, as EED is a core scaffold component that can exist stably independently and is not dependent on EZH2. The degrader suppresses the H3K27me3 mark, albeit less effectively than the parent compound C24. MS-1943 is, however, more cytotoxic in EZH2 dependent cell lines than C24, due to chronic stimulation of the unfolded protein response caused by compound-induced endoplasmic reticulum (ER) stress. This may be an off-target activity of the adamantane tag, as this effect has not been reported for PRC2-targeted PROTACs. Such PROTACs include UNC6852, a bivalent degrader designed by connecting a EED226 to a ligand for the E3 ubiquitin ligase VHL (Figure 25c).663 UNC6852 degrades EED and causes the collateral degradation of the PRC2 components EZH2 and SUZ12, which depend on EED for proper folding and stabilization. Disruption of the PRC2 complex results in reduced H3K27me3 levels in EZH2-Y641N-mutant cells and is antiproliferative in DLBCL. Other PROTACs have been built on both EED ligands and EZH2 inhibitors.664,665 UNC7700, an analog of UNC6852 that implements a cis-cyclobutane linker in place of the more flexible polymethylene linker, shows greatly improved potency of degradation (Figure 25c).666
A recent study also suggests that covalent engagement of EZH2 is sufficient to result in the depletion of EZH2 protein levels.667 IHMT-337, which covalently binds EZH2 at Cys663 was discovered from a collection of 200 compounds designed to irreversibly inhibit EZH2 (Figure 25b). EZH2 is subsequently degraded via the CHIP-mediated ubiquitination pathway. Interestingly, this molecule was useful in elucidating a non-enzymatic, PRC2-independent function of EZH2 as a transcriptional cofactor for CDK4. The compound is not only effective in EZH2-mutant DLBCL cells, but also in triple negative breast cancer (TNBC), which are not dependent on EZH2 enzymatic activity. Several other degraders of EZH2 and EED have been reported, combining various PRC2-ligands with various E3-ligase ligands to fashion heterobifunctional molecules capable of depleting protein levels in vitro and in vivo.663–665,668,669
While allosteric approaches to target PRC2 show preclinical activity against EZH2-mutant lymphoma similar to orthosteric inhibition, their ultimate success in the clinic remains uncertain. They might enable major therapeutic advances if able to overcome mutations causing acquired resistance to SAM-competitive EZH2 inhibitors. Indeed, there is preliminary evidence that acquired EZH2 mutations identified to promote resistance in cell culture are also observed in patients treated with tazemetostat.670 Ongoing clinical trials will determine if EED inhibitors, such as ORIC-944, which is being tested in prostate cancer (NCT05413421), can provide therapeutic benefit as single agents or combination therapeutics.671
3.2.3. PRC1
PRC1 is a multiprotein complex that functions together with PRC2 to enable gene silencing. It possesses a core E3 ubiquitin ligase, RING1A/RING1B, a chromobox (CBX) reader protein, and one of 6 polycomb group ring finger (PCGF) paralog proteins (Figure 26a).672 Of the 6 PCGF proteins, PCGF4, also called BMI1, is a member of the canonical PRC1 complex. CBX proteins recognize repressive H3K27me3 marks deposited by PRC2 and the PRC1 complex mono-ubiquitinates H2AK118 and H2AK119 via RING1A/B.673 Together, PRC1 and PRC2 silence gene transcription to promote stemness during development and cancer.674 Interestingly, continual BMI1 expression has been shown to be selectively required for the proliferation of leukemia stem cells and it has therefore has been proposed as an attractive target for small molecule inhibition.675 It has also been suggested as a target in multiple other indications beyond AML.676–678 E3 ligase activity of RING1B/A is essential for the maintenance of leukemia stem cells and leukemogenesis, which has inspired several attempts to inhibit H2A ubiquitination (H2Aub) levels using small molecules.679–684
Figure 26.

PRC1 inhibition. (a) Chromatin regulation by PRC1. (b) chemical structures of RING1B inhibitors RB-2 and RB-3. (c) Chemical structures of RING1B/BMI1 PROTACs constructed with ligands binding EED (purple) and the E3 ligase CRBN (yellow) or VHL (grey).
Initial efforts to discover small molecule inhibitors of RING1B and BMI1 focused on inhibiting the ligase activity of PRC1. Small molecule chemical screens that used H2Aub signal as the output led to the discovery of two compounds, GW-519 and PRT4165.685,686 Both compounds inhibit H2Aub and decreased proliferation, but their mechanisms of action have not been elucidated. Screening for compounds that decreased BMI1 expression has led to the discovery of PTC-209 which decreases BMI1 expression in colorectal cancer.680 The mechanism of PTC-209 remains unknown, but its proposed effect is on BMI1 translation leading to the observed reduction in BMI1 protein expression. While targeting the PRC1 through disruption of BMI1 scaffolding function remains an attractive strategy, the mechanism of PTC-209 has not been the focus of active investigation. Instead, recent efforts have shifted toward the development of next-generation compounds with improved pharmacological properties and less convoluted mechanisms of action.687,688
The first direct-acting and mechanistically characterized inhibitor to be reported was discovered in a fragment screen using 1H-15N heteronuclear single quantum coherence (HSQC) experiments and a 15N-labeled RING1B-BMI1 fusion protein.679 A benzene- and thiophene-containing fragment hit was found to bind the RING1B-BMI1 fusion protein in the mM range. Subsequent medicinal chemistry, including replacement of the thiophene ring with a pyrrole to increase solubility, yielded RB-2, which binds with 11.5 μM affinity to the RING domain of RING1B (Figure 26b). Replacement of the RB-2 ethyl with an isopropyl and the chloro-phenyl with a chloro-indole led to increased hydrophobic interactions with RING1B and improved affinity to 2.8 μM. The resulting compound, RB-3, inhibits ubiquitination in vitro and in cells and causes a decrease in PRC1 localization to its target genes.679 Further optimization has been employed on this scaffold,689 but improvements in potency and in vivo activity are crucial to more fully explore RING1B as a potential therapeutic target.
PRC1 can also be targeted by inducing collateral degradation of BMI1 and RING1B through EED-based PROTACs.690 This discovery was motivated by the previous observation that an EED PROTACs can induce the degradation of additional PRC2 subunits.663,664 EED has been shown to incorporate into PRC1 with RING1B and BMI1,691,692 suggesting that EED PROTACS may also be able to induce the degradation of PRC1 components.690 MS147, a heterobifunctional construction of ligands for EED and VHL, preferentially degrades RING1B and BMI1 over PRC2 components, including EED, causing a decrease in H2AK119ub without altering H3K27me3.690 Interestingly, MS147 shares a similar VHL and EED ligand to the previously reported EED/PRC2 degrader, PROTAC 2 (Figure 26c).664 The main difference between these compounds is linker length, which contributes to MS147 inducing degradation of both RING1B and BMI1, but only degrading PRC2 subunits at high concentrations. Similar results were observed with MS181, a CRBN-based EED degrader that induces PRC1 subunit degradation (Figure 26c).693 However, proteome-scale measures of selectivity have not been reported for MS147 and MS181.
3.3. SWI/SNF
The mammalian SWI/SNF (switch/sucrose non-fermentable) complex, also known as the BAF (BRG-/BRM-associated factor) complex, is an ATP-dependent chromatin remodeler complex and transcriptional co-activator.694,695 A diverse collection of protein products from 29 genes are incorporated into three distinct assemblies of the BAF complex: canonical BAF (cBAF), polybromo-associated BAF complex (PBAF), and the recently described non-canonical BAF (also reported as GLTSCR1 BAF, or GBAF, hereafter referred to as ncBAF) (Figure 27).696–699 While all three complexes share a core set of subunits, each distinct complex specifically incorporates certain subunits not shared by the others. ARID1A/ARID1B and DPF2 are incorporated into cBAF; PBRM1, ARID2, and BRD7 are incorporated into PBAF; and GLTSCR1/GLTSCR1L and BRD9 are incorporated into ncBAF (Figure 27). Both SMARCA2 or SMARCA4, the paralogous ATPase subunits of the complex, can be incorporated into each of the three complexes. They couple the hydrolysis of ATP to the disruption of histone-DNA contacts to open chromatin structures within cis-regulatory elements of the genome and allow transcriptional activation.700 Chromatin immunoprecipitation using antibodies specific to each complex (e.g., anti-SMARCA2/SMARCA4 for pan-SWI/SNF recognition, anti-BRD9/GLTSCR1 for ncBAF specific recognition, anti-DPF2 for cBAF specific recognition, and anti-BRD7 for PBAF specific recognition) have shown that ncBAF, cBAF and PBAF are differentially localized to promoters, distal sites, and gene bodies, respectively (Figure 23). Furthermore, like SMARCA2/4, many BAF subunits can be alternately fulfilled by close paralogs, leading to important functional diversifications of BAF complexes in specific cell types.701 The hyper-diversity of these complexes has been a rich source of biological investigation and underlies a number of cancer-specific vulnerabilities, whereby an imbalance in BAF activity makes other chromatin regulatory proteins indispensable for cell fitness.
Figure 27.

Graphical representation of BAF complexes, with subunits uniquely incorporated into each complex labeled. Complexes are centered over their asymmetric genomic destinations.
The SWI/SNF complexes are large, multi-subunit transcriptional regulators that have specialized functions across organismal development and are recurrently mutated in diverse human malignancies.702 Synovial sarcoma (SS) and malignant rhabdoid tumor (MRT) are among the clearest examples of SWI/SNF-driven tumors, where nearly all cases are driven by a specific SWI/SNF alteration. In SS, the t(X;18)(p11.2;q11.2) translocation is found in virtually all tumors and considered pathognomonic for disease.703–706 The result is an onco-fusion protein SS18-SSX, which is defined by the replacement of eight C-terminal amino acids of SS18 with the 78 C-terminal amino acids of one SSX protein paralog.704–706 This fusion product is the sole genetic anomaly in a cancer characterized by an otherwise remarkably low mutational burden, and is causal of disease.707 SS18 is a subunit of the SWI/SNF complex and, when fused to SSX, its incorporation leads to eviction of SMARCB1 (also known as BAF47/SNF5/INI1) from the complex (Figure 28a).708,709 This causes SWI/SNF to be redistributed on the genome, dysregulating its opposition to PRC-mediated gene repression and activating a synovial sarcoma gene signature.710,711
Figure 28.

BRD9 is an induced sensitivity in SWI/SNF-mutant tumors. (a) Schematic of SMARCB1 mutations in synovial sarcoma and malignant rhabdoid tumor. (b) Representative small-molecules chemical probes that inhibit the BRD9 bromodomain. (c) Development of BRD9 PROTACs based on BI9564.
3.3.1. BRD9 synthetic lethality
In MRT, SMARCB1 is biallelically inactivated in up to 95% of all tumors (Figure 28a).712,713 This alteration evokes a synthetic lethal dependency on BRD9 and the ncBAF complex, which was discovered in an unbiased genetic screen and shown further with chemical degraders of BRD9.701 It has also been validated by multiple independent groups.714 A similar dependency on BRD9 is seen in synovial sarcoma, where the degradation of BRD9 disrupts SS18-SSX complexes, downregulates oncogenic genes, and inhibits tumor growth.715 BRD9 is essential for the incorporation of GLTSCR1, a defining subunit of the non-canonical BAF (ncBAF) complex. GLTSCR1 functions as the signature scaffolding subunit that distinguishes ncBAF from canonical and PBAF complexes, enabling ncBAF to localize to specific chromatin sites and regulate a distinct set of genes. Without GLTSCR1, ncBAF cannot assemble into a functional complex. In the setting of SMARCB1 loss, BRD9 incorporation is increased, driving ncBAF formation and creating an acquired vulnerability to BRD9 inhibition (Figure 28a).
Given the discovery of BRD9 as a genetically defined dependency in SWI/SNF-mutant tumors, the protein has become an attractive target for drug discovery. However, BRD9 chemical probes preceded the identification of BRD9 as a potential drug target and were critical to the discovery and validation of BRD9 as a cancer-specific dependency. Like many other bromodomain proteins, BRD9 bromodomain inhibitors were widely pursued after the first generation of BET bromodomain inhibitors established the ligandability of this class. LP99, disclosed in 2015, is a sub-micromolar, selective inhibitor of BRD7/9 bromodomains, discovered using complexity-building cascade chemistry to assist with structure-guided optimization of a quinolone fragment hit (Figure 28b).716 Other early compounds include triazolophthalazine717 and 9H-purine718 scaffolds that compete with acetyllysine to bind the bromodomain of BRD9. However, these early compounds did not possess sufficient activity and selectivity to faithfully study BRD9 biology.
The thienopyridone, I-BRD9, was disclosed in 2016 and reported to possess >700-fold selectivity against the BET bromodomain family and 200-fold selectivity against its paralog, BRD7, representing a major advance for BRD9 tool development (Figure 28b).719 This selectivity is driven by a charged amidine favoring the less hydrophobic environment of BRD9 relative to other bromodomains. A structurally divergent pyridone series with suitable pharmacokinetic and pharmacodynamic (PK/PD) properties to interrogate BRD9 biology in vivo was reported shortly after I-BRD9.720 These compounds, exemplified by the chemical probes, BI-7273 and BI-9564 (Figure 28b,c), originated from a fragment library screen and virtual ligand screening, which converged on pyrimidinone and dimethylpyridinone cores as starting points for structure-based medicinal chemistry. Elaboration of the fragments yielded BI-7273, which binds with 15 nM affinity to BRD9 and does not bind BET bromodomains up to 100 μM (Figure 28b). This improved selectivity, compared to LP99 and I-BRD9, proved exceptionally useful for studying BRD9 biology. Based on prior studies demonstrating a role for the SWI/SNF ATPase, SMARCA4, in acute leukemia,721 BI-7273 was evaluated in acute leukemia cells and compared to genetic BRD9 loss-of-function perturbations.722 Both chemical and genetic perturbations demonstrated cell-type-selective anti-leukemic activity, which could be positively attributed to BRD9 through genetic domain-swap experiments that made drug-resistant alleles of BRD9. BI-9564, a more selective inhibitor from this series, also inhibits cancer cell proliferation in vitro, demonstrates an acceptable pharmacokinetic profile, and is efficacious in a murine model of AML (Figure 28c).720 These chemical probes are joined by several more examples of structurally diverse BRD9 bromodomain ligands.723–726
In contrast to acute leukemia cells, which are sensitive to BRD9 bromodomain inhibition, multiple independent reports have demonstrated that SS and MRT are dependent on BRD9 for survival but insensitive to BRD9 bromodomain inhibition, suggesting an essential scaffolding role rather than a dependence on its acetyl-lysine-binding bromodomain.701,714,715 This suggested that chemically induced degradation of BRD9 would be required to effectively exploit BRD9 vulnerabilities in SWI/SNF-mutant tumors. A series of CRBN-based BRD9 PROTACs had previously been constructed from I-BRD9 or BI-7273, including dBRD9, a BI-7273-pomalidomide heterobifunctional that induces potent and selective BRD9 degradation (Figure 26c).218 Mass spectrometry (MS)-based proteomics has shown that dBRD9 induces the degradation of BRD9 exclusively. No other proteins were found to be degraded by dBRD9, including BRD7, the paralogous target of BI-7273. This was a particularly interesting discovery, as it was one of the first examples showing that a multi-targeted ligand can be used to construct a more selective degrader, now a well-established feature of PROTAC pharmacology.222
The PROTAC, dBRD9, and an optimized analog, dBRD9-A, were subsequently used to demonstrate that synovial sarcoma and malignant rhabdoid tumors are hypersensitive to BRD9 degradation (Figure 28c).701,715 The BRD9-specific BAF complex, ncBAF, is a critical dependency in SS and MRT. This was discovered using CRISPR-Cas9-based gene essentiality screens, in which a pooled library of single guide RNAs is introduced into a population of cells, where each guide directs Cas9 to disrupt a specific gene. By tracking which guides become depleted or enriched over time, one can identify genes that are essential for cell survival or confer selective advantages under defined conditions. This experiment demonstrated that both SS and MRT cell lines are selectively sensitive to loss of BAF subunits that are specific to ncBAF.701 Independently, a domain-focused CRISPR screen, which can be used to identify domain-specific dependencies within protein targets,727 demonstrated that the bromodomain of BRD9 is important to the fitness of SS cells but dispensable for other sarcomas.715 These results were corroborated with rescue experiments showing that wild-type, but not bromodomain-inactive, BRD9 can support SS fitness. Interestingly, an additional domain, containing amino acids 311–345, was identified as being necessary for association into the ncBAF complex.715 These amino acids are within the DUF3512 domain (amino acids 274–505), which was similarly reported by another group to be necessary and sufficient for BRD9 incorporation into ncBAF.701 This was hypothesized to explain the observation that BRD9 degradation is more effective at inhibiting the proliferation of SS cells than BRD7/9 bromodomain inhibition.715 Ultimately, these findings motivated the clinical development of BRD9 PROTACs, including CFT8634 (C4 Therapeutics, NCT05355753) and FHD-609 (Foghorn Therapeutics, NCT04965753), for the treatment of SS and MRT. However, both trials were terminated due to FHD-609 causing dose-limiting cardiotoxicity and CFT8634 showing a lack of therapeutic efficacy despite efficient BRD9 degradation, according to company press releases.
3.3.2. SMARCA2/4 paralog synthetic lethality
SWI/SNF complexes can incorporate one of two paralogous ATPase subunits, SMARCA2 (SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin, Subfamily A, Member 2; also known as BRM) and SMARCA4 (also known as BRG1), which share 75% sequence identity, 93% conserved ATPase domain, and functional redundancy (Figure 29a). These subunits are mutually exclusive with each other and highly homologous, possessing catalytic ATPase domains and acetyl-lysine reader bromodomains.728 Recurrent homozygous mutations of SMARCA4 are found in 10–35% of NSCLC,619,729–732 15% of Burkett Lymphoma,617,733 5–10% of childhood medulloblastoma,734–736 as well as in pancreatic adenocarcinoma,737 melanoma,738 and other cancers, while SMARCA2 is often epigenetically silenced in cancer.620 The structural and functional redundancy of SMARCA2 and SMARCA4 create a potentially targetable synthetic lethality whereby SMARCA2 is essential for the survival of SMARCA4-deficient tumors (Figure 29a). We have also included DepMap data for these genes (Figure 29b).
Figure 29.

SMARCA2 is a synthetic lethality in SMARCA4-deficient cancers. (a) Schematic of SMARCA2 synthetic lethality. (b) Histogram of gene effect scores for the genes SMARCA2 and SMARCA4 (DepMap). (c) Chemical probe for the SMARCA2/4 bromodomain. (d) Chemical probes that inhibit SMARCA2/4 ATPase activity. (e) Degraders of SMARCA2/4. (f) Development of optimized PROTACs, such as SMD-1087, with SMI-1074, a recently reported SMARCA2/4 bromodomain ligand.
This synthetic lethal relationship was first discovered in a hypothesis-driven study. Here, RNAi-based silencing of SMARCA2 expression was shown to suppress the growth of SMARCA4-deficient tumors through the p21/CDKN1A axis, whereby p53-induced expression of the cyclin-dependent kinase inhibitor p21 enforces cell-cycle arrest, indicative of a senescent phenotype.739 However, SMARCA2 knockdown had no effect on non-transformed fibroblasts that express both SMARCA2 and SMARCA4, highlighting a clear synthetic lethal relationship. This senescent phenotype is rescued through ectopic expression of wild-type SMARCA4 but not mutant SMARCA4 alleles that inactivate the ATPase domain, nominating its catalytic domain as a potential target for drug discovery. However, as noted by the authors of this study, some cancers show genetic alterations affecting both SMARCA2 and SMARCA4, potentially pointing to a limitation of exploiting SMARCA2/4 synthetic lethality.739
Shortly after this publication, two concurrent manuscripts reported the discovery of SMARCA2/4 synthetic lethality through unbiased, large-scale genetic screens.621,740 In the first, a collection of pooled RNAi essentiality screens performed across hundreds of cell lines (Project Achilles), identified SMARCA2 as the most differentially essential gene in cancers with inactivating mutations in SMARCA4. After validating the context-specific essentiality of SMARCA2, the authors demonstrated that SMARCA4 mutation leads to the upregulation of SMARCA2 and increases its incorporation into the SWI/SNF complex. Contemporaneously, another large-scale RNAi-based screen for essential genes, which used a chromatin-focused library to screen dozens of cancer cell lines, similarly identified SMARCA2 as a strong and selective dependency in SMARCA4-deficient cancer.621 The integrity of the SWI/SNF complex was found to be maintained upon SMARCA2 knockdown in SMARCA4-deficient cancers, suggesting that the antiproliferative effect of SMARCA2 loss-of-function is caused by the redundancy of SMARCA2/4 ATPase activity and not due to destabilization of the entire complex (Figure 29a).
These genetic findings galvanized interest in the pursuit of small molecule ligands that target SMARCA2/4, both to further probe the biology of this system and to potentially exploit the synthetic lethal relationship for therapeutic discovery. Bromodomain ligands were the first small molecules to be reported for SMARCA2/4. The earliest example, PFI-3, was collaboratively reported by the Structural Genomics Consortium and Pfizer and binds SMARCA2/4 bromodomains with an affinity of 89 nM (Figure 29b).616,741,742 Interesting, the selectivity of PFI-3 against other bromodomain families is attributed to a unique binding mode in which the phenolic head group of the molecule displaces a series of water molecules that are traditionally conserved when liganded by other bromodomain inhibitors. While the compound can elicit a dose-dependent displacement of the isolated SMARCA2 bromodomain from chromatin, it does not show antiproliferative activity in SMARCA4-deficient cancer cells. Subsequent genetic rescue experiments using mutant alleles of SMARCA2 demonstrated that its ATPase activity is required for cell survival but its bromodomain is dispensable.616 This demonstrated that the ATPase domain is the relevant target for drug discovery in oncology, which was further supported by the discovery that PFI-3 is unable to displace full-length SMARCA2 (as opposed to the isolated bromodomain) from chromatin.742 Interestingly the dispensability of the bromodomain is consistent with Drosophila experiments reported in 2002, which demonstrated that the bromodomain of the fly ortholog is not essential for its localization to chromatin.743 It is important to note that even if the bromodomain had been relevant to SMARCA2 function, its close homology with the SMARCA4 bromodomain would have presented an enormous challenge for the goal of discovering ligands that selectively inhibit SMARCA2 over SMARCA4, which is ultimately needed to exploit this synthetic lethality.
The functionally relevant ATPase domain of SMARCA2/4 represented a more difficult target for ligand discovery than the bromodomain. Toward the goal of developing ATPase inhibitors of SMARCA2/4, a team at Novartis designed an ATP turnover assay using a truncated version of SMARCA2 (ATPase and SnAC domains) for high-throughput screening, which was further supported by multiple downstream biophysical assays to assess hit quality.744 A diheteroaryl-urea hit emerged from a screen of 72,000 compounds and was moved forward for optimization, leading to the discovery of tool compounds, BRM011 and BRM014 (Figure 29c). Crystal structures demonstrated hydrogen bonding interactions between the urea nitrogen and the catalytic Glu852 side chain, which traps it in an inactive conformation. Interestingly, these compounds were found to bind in an allosteric pocket near the active site, a potential way to circumvent challenges with liganding the associated orthosteric cleft. BRM014 has been used effectively as a chemical tool, for example, demonstrating the widespread importance of SMARCA2/4 for maintaining chromatin accessibility.745–747 While BRM014 showed potent activity in cell line models and in vivo, it elicited tolerability issues likely owing to the dual inhibition of both paralogs. Similar to the SMARCA2/4 bromodomains, selectively inhibiting one of the two ATPase domains will likely present a major challenge for drug discovery, due to their highly homologous structures.748,749 While a SMARCA2/4 ATPase inhibitor, FHD-286, is being evaluated in a Phase 1 clinical trials, it suffered a partial clinical hold in relapsed/refractory AML and myelodysplastic syndrome and a full suspension of its clinical exploration in metastatic uveal melanoma.750
While SMARCA2/4 bromodomain inhibitors are incapable of phenocopying genetic loss-of-function, they have found a renewed purpose in the development of PROTACs that can exploit SMARCA2/4 synthetic lethality. Here, small molecules targeting the bromodomain have been used to construct PROTACs that selectively degrade SMARCA2 over SMARCA4. While developing bromodomain inhibitors with high isoform selectivity has proven difficult due to the strong structural conservation of bromodomain binding pockets, degraders can achieve apparent selectivity through a different principle. Degraders rely not only on bromodomain binding but also on the formation of a productive ternary complex with an E3 ligase, which introduces an additional layer of specificity based on surface complementarity and complex stability. Thus, bromodomain selectivity is rarely achieved at the inhibitor level but can emerge during targeted protein degradation because the degrader exploits unique degron–ligase–target interactions. The first SMARCA2/4 PROTAC, ACBI1, which degrades both paralogs, was discovered using a rational design strategy that emphasized the formation of a cooperative ternary complex between SMARCA2/4 and VHL (Figure 29d).751 It was constructed from an aminopyridazine bromodomain ligand and the VHL ligand, VH101, and induces degradation of both SMARCA2 and SMARCA4 at sub-micromolar concentrations. It also degrades PBRM1, a high-affinity bromodomain target of the aminopyridazine ligand. These effects result in potent anti-proliferative activity, recapitulating SMARCA2/4 genetic loss-of-function and SMARCA2/4 ATPase inhibition. The SMARCA2/4 PROTAC, AU-15330, which is constructed from the same bromodomain and VHL ligands but uses a shorter linker and different linker attachments, also degrades both paralogs (Figure 29d).752 Despite degrading SMARCA2/4 and PRBM1, prostate cancer cells were reported to be preferentially sensitive to AU-15330.
A next-generation degrader, ACBI2, offers an improved selectivity window (Figure 29d). This VHL-based PROTAC of SMARCA2 was redesigned using a previously reported quinazolinone PRBM1 bromodomain ligand that was further optimized for use in PROTACs by considering rigidity and minimization of hydrogen bond donors.753 Multiple exit vectors were considered on the VHL ligand, arriving at a benzylic attachment site that was evaluated with a variety of linker compositions. Iteratively improving on moderate initial selectivity for SMARCA2 over SMARCA4 with this series of compounds, ACBI2 was found to elicit degradation of SMARCA2 at single-digit nanomolar concentrations and is 30-fold selective against SMARCA4. It also demonstrates PK/PD properties that allow for oral delivery in vivo, eliciting tumor stasis in mouse xenograft models of SMARCA4-deficient lung cancer. A series of selective SMARCA2 degraders have since been disclosed, including A947, SMD-3040 and a collection of diazabicyclooctane molecules.754–756 Comparative analyses have demonstrated the most improved selectivity with SMD-3040.755 Whereas other published PROTACs show relatively strong maximal degradation (Dmax) of SMARCA4 at higher concentrations,751,753,754 SMD-3040, which is constructed by tethering a pyrazolo-aminopyridine bromodomain ligand to a VHL ligand through a rigid spirocyclic linker, is ~100-fold selective for SMARCA2 and shows a diminished Dmax for SMARCA4 (Figure 29d).755 This compound is useful for in vivo experimentation and demonstrates effective tumor inhibition in SMARCA4-deficient xenograft mouse models with a well-tolerated toxicity profile.755 Another redesigned SMARCA2/4 ligand has contributed to the discovery of PROTACs, such as SMD-1087 and SMD-3236, with further improved profiles in vitro and in vivo, which could enable rapid clinical translation (Figure 29e).757,758 Indeed, as a class, SMARCA2-selective PROTACs are already in clinical-stage investigation, with Phase 1 trials ongoing for the reportedly SMARCA2-selective degraders, PRT3789 (NCT05639751) and PRT7732 (NCT06560645).
3.3.3. ARID1A/ARID1B synthetic lethality
The AT-rich interaction domain 1A (ARID1A) is a SWI/SNF complex subunit that has physiologically relevant roles in transcriptional regulation and chromatin remodeling.759 It also serves as a tumor suppressor gene,760,761 and, in some stage- and tissue-specific cases, as an oncogene.762 ARID1A is mutated in almost 10% of human cancers, making it the most commonly mutated SWI/SNF subunit.763 ARID1B, a mutually exclusive and functionally unique counterpart, shares approximately 60% identity with ARID1A. As first revealed by examination of the Project Achilles dataset, ARID1A-mutant tumors are hyper-dependent on ARID1B for survival,764 as it incorporates into residual SWI/SNF complexes necessary for cell survival.618,764,765 No small-molecule ligands for ARID1A/B have been reported, but these genetic data make a compelling argument for developing pharmacologic tools to study this synthetic lethality. Small-molecule tools have been utilized to explore other vulnerabilities of ARID1A-mutant cancers, such as molecules targeting DNA damage repair, cell cycle, and immune checkpoint pathways.766–768
3.4. CBP/p300
The paralogs, CBP (encoded by CREB-binding protein, CREBBP) and p300 (encoded by E1A binding protein p300, EP300), are transcriptional coactivators and lysine acetyltransferases with diverse cellular functions, most notably including redundant and non-redundant roles in transcriptional regulation. They share nine structurally conserved functional domains involved in protein-protein interactions, recognition of post-translational modifications, and catalysis of lysine acetylation.769,770 Two of these are recognized as promising targets for small-molecule therapeutics: the bromodomain and the HAT domain. The bromodomain binds to acetylated lysines and regulates CBP/p300 HAT activity, which is responsible for acetylating thousands of lysines on histone and non-histone substrates, like transcription factors and transcriptional co-regulators.771 CBP/p300 regulate enhancer-mediated transcriptional activation, highlighting their critical roles in both normal physiological and pathophysiological processes.772 These proteins play a critical role in the development and maintenance of solid and hematological tumors alike, most notably as oncogenic fusions and non-oncogene dependencies, which has motivated broad efforts in chemical probe and drug discovery.772
CBP/p300 are critical to a variety of essential cellular functions, including cell cycle regulation, DNA synthesis and repair, and transcriptional control. Heterozygous loss-of-function mutations are known to cause Rubinstein-Taybi syndrome (RTS), a congenital disease characterized by growth deficiency, mental deficit, developmental delays and abnormalities, and a characteristic dysmorphology.773 Another critical feature of RTS is the propensity to develop cancer, potentially due to somatic mutation of the remaining functional allele. In addition to RTS tumors, loss-of-function alterations to EP300 and CREBBP – including loss-of-heterozygosity, truncations, and missense mutations – have been known to occur in colorectal, breast, ovarian, lung, and gastric cancers for several decades.774–780
Inactivating mutations are also common in some hematological malignancies, including acute lymphoblastic leukemia and lymphoma.781,782 In healthy lymphoid tissues, CBP/p300 maintains the acetylation pattern required for the terminal differentiation of mature B cells—that is, the final differential step in which B cells irreversibly commit to a cellular identity of plasma cell or memory B cell.783 In normal immunophysiology, a subset of B cells will endure mutagenic germinal center reactions, in which they deliberately acquire somatic hypermutations and class-switch recombination to diversify the antibody repertoire, in response to encountering immunogenic antigens. Loss-of-function mutations of CREBBP, however, impair the reacetylation of enhancers that are necessary to facilitate germinal center exit, ultimately promoting lymphomagenesis.781 Follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL), for example, exhibit frequent inactivating structural mutations in the HAT domain of CBP and, less frequently, p300.781 These mutations were discovered by whole-exome sequencing and predicted to cause the elimination or truncation of the HAT domain in CBP, reducing its affinity for acetyl-CoA binding and preventing acetylation of its substrates.781 The lack of BCL6 acetylation by mutant CBP has been shown to result in unopposed activity of BCL6/SMRT/HDAC3 complexes at B-cell signaling enhancers, contributing to lymphomagenesis.784
Cancers with CREBBP loss-of-function alterations are hyper-dependent on p300 function, creating a synthetic lethal target for drug discovery and development. This was originally discovered in an unbiased forward genetic screen using a library of siRNAs to knock down the expression of chromatin regulatory proteins.785 It has been further reinforced in vivo using mouse models for conditional deletion of Crebbp or Ep300 in the germinal center (GC), altogether showing the combined essentiality of CBP/p300.786 The loss of both paralogs in B cells compromises GC formation, creating a novel vulnerability to EP300 deletion in DLBCL cell lines that harbor CREBBP mutations.786
Instability of the genome near the 5’ end of the CREBBP gene, alongside potential breakpoints in the second intron, predispose CREBBP to chromosomal translocations yielding oncogenic CBP fusion proteins.787,788 This phenomenon is more common with CBP than p300, and these translocations are frequently implicated in the transformation of hematological malignancies. These include CBP fusions with monocytic leukemia zing-finger (MOZ, also known as KAT6A),789,790 MOZ-related factor (MORF, or KAT6B),791 and MLL.792,793 Rarely, fusions can also be seen with p300, as evidenced by MOZ-p300 fusion oncoprotein.790 These leukemogenic fusion proteins form aberrant chromatin acetylation and transcriptional co-activation patterns through the mis-localization of their enzymatic activity on the genome.
The first pharmacological perturbations of HAT proteins were enabled by the design and synthesis of bisubstrate inhibitors that engage the binding sites for both histone substrates and the acetyl-CoA cofactor. This is best exemplified by Lys-CoA, a potent CBP/p300 HAT inhibitor with ~500 nM potency.794 While bisubstrate inhibitors were exceptionally useful for in vitro biochemistry and structural biology studies,795 their lack of drug-like physiochemical properties prohibited cell biological studies. The natural products, garcinol and anacardic acid, were later reported as CBP/p300 inhibitors,796,797 along with a synthetic small-molecule, C646,798 but these tools were later found to be complicated by non-specific off-target effects.799 C646, which found widespread use as a p300 inhibitor, shows off-target reactivity with thiols in cells and was recently reported to function through XPO1, with p300 being perturbed indirectly by C646-induced degradation of XPO1.800,801 In more recent years, these tools have been succeeded by highly potent and selective chemical probes for both the bromodomains and HAT domains of CBP/p300, providing a structurally and functionally diverse collection of chemical tools to study CBP/p300.
3.4.1. CBP/p300 bromodomain inhibitors
CBP/p300 bromodomain inhibitors were among the first non-BET bromodomain inhibitors to be reported after the discovery of JQ1 and I-BET. Preceded by the discovery of fragments and macrocyclic peptide with relatively weak potency,802–804 the first potent and selective chemical probe for CBP/p300 bromodomains, SGC-CBP30, was reported by the Structural Genomics Consortium in 2014.805 This discovery was enabled by optimizing a 3,5-dimethylisoxazole BET bromodomain inhibitor that had previously been shown to possess off-target activity against CBP.806 A collection of 83 analogs – synthesized by parallel Suzuki couplings of commercially available pinacol esters and heteroaryl bromides – were screened by differential scanning fluorimetry (DSF) for selective engagement of CBP over BRD4.807 Further modifications of a hit from this screen, together with structure-guided design, afforded the chemical probe SGC-CBP30 (Compound 59 in the index manuscript), which binds with 20–30 nM affinity to CBP/p300, but is only 40-fold selective against BRD4 (Figure 30a).807 808 Based on this scaffold, additional structure-based drug design afforded the chemical probe PF-CBP1, an inhibitor with slightly reduced potency for CBP/p300 (IC50 = 130 nM), but dramatically improved selectivity against BRD4 (IC50 = 18 μM) (Figure 30a).809 The same isoxazolylbenzimidazole scaffold is shared by CCS1477, the first CBP/p300 bromodomain inhibitor to advance into human clinical investigation (NCT03568656 and NCT04068597) (Figure 30a).810 It is highly potent against CBP/p300, showing 1.3 nM affinity for p300, and is more than 100-fold selective against BRD4 (222 nM affinity). Clinical trials for CCS1477 are focused on prostate cancer, where CBP/p300 function as co-activators for androgen receptor (AR). CCS1477 is orally bioavailable and preclinical experiments have shown it can delay tumor growth in patient-derived xenograft models of prostate cancer.810 which is consistent with prior reports showing that prostate cancer models are hypersensitive to CBP/p300 bromodomain and HAT domains inhibitors.811,812
Figure 30.

Small-molecules targeting CBP/p300. (a) Benzimidazole-based bromodomain inhibitors. (b) Piperidine-based bromodomain inhibitors. (c) Benzodiazepinone-based bromodomain inhibitor. (d) Benzoxazepine based bromodomain inhibitor. (e) Structurally distinct CBP/p300 HAT inhibitors. (f) CBP/p300 PROTACs, which share piperidine-based bromodomain ligands to target CBP/p300 for degradation.
A pyrazolopiperidine core has also afforded CBP/p300 bromodomain inhibitors with potent activity in vivo and excellent selectivity against BET bromodomains.806 This scaffold was first discovered as a screening hit from a thermal shift assay for CBP.813 Structure-based drug design motivated three areas for hit optimization: an N-acetylpiperidine group was preferred over alternative acetyllysine mimics, a pyrazole substituent was found to make favorable interactions with the WPF shelf, and a tetrahydrofuran moiety occupies the ZA loop. An analog that used an N-methylpyrazole moiety to contact the WPF shelf showed good potency and physicochemical properties, yet suffered from metabolic instability. This was improved by an analog that moved the N-methylpyrazole from the 3 position to the 4 position of a phenyl ring that was also fluorinated at 2 position to decrease glucoronidation. The resulting chemical probe, GNE-272, shows low clearance, good oral bioavailability, and excellent selectivity for CBP/p300 over other off targets (Figure 30b). In a leukemia xenograft model (using the MOLM-16 AML cell line), it was shown to decrease MYC expression and repress leukemia burden at doses that pharmacokinetic studies predicted would inhibit CBP/p300 but not BET bromodomains.813
Additional improvements on GNE-272 were subsequently reported with contemporaneous disclosures of GNE-049, GNE-781, and GNE-207 (Figure 30b).811,814,815 Concerns that the aniline in GNE-272 could be a possible toxicophore motivated the direct attachment of the phenyl group to the pyrazolopiperidine core, which maintained activity.816 Successive replacements of the phenyl group with indole and isoquinolone groups, together with exchanging the tetrahydrofuran for a tetrahydropyran, delivered progressive improvements in potency. Further substitution of the isoquinolone with an amidopyridine yielded GNE-207, a highly potent inhibitor of the CBP bromodomain (IC50 = 1 nM) with >2000-fold selectivity over BRD4 (Figure 30b). While this compound demonstrated acceptable PK for in vivo study,816 more substantial improvements were obtained with the contemporaneous report of GNE-781 (Figure 30b).817 Here, it was found that constraining the aniline nitrogen of GNE-272 into a tetrahydroquinoline group prevented the molecule from adopting a planar conformation when bound to BRD4, introducing a steric clash and improving selectivity. Introduction of a difluoromethyl substituent to position 7 of the tetrahydroquinoline further improved CBP potency and selectivity over BRD4 and replacement of the tetrahydrofuran with a tetrahydropyran (as in GNE-207) improved metabolic stability. A final replacement of the acetamide moiety, which mimics the binding of acetyllysine to the conserved bromodomain asparagine, with a methylurea group addressed adverse effects on the central nervous system, ultimately yielding a compound with subnanomolar potency in biochemical assays, <10 nM potency in cells, >5,000-fold selectivity over BET bromodomains, and suitable oral bioavailability. This compound, GNE-781, is highly selective for CBP/p300 over a large panel of other bromodomains, kinases, and cytochromes p450s, providing an excellent chemical probe for CBP/p300 both in vitro and in vivo, while other compounds were prioritized for clinical development (Figure 30b).817
Additional scaffolds to probe CBP/p300 are well represented by CPI-637 and I-CBP112. The benzodiazepinone CPI-637 is a potent and selective chemical probe for CBP/p300 that arose from the optimization of a fragment screening hit for CBP (Figure 30c).818 A co-crystal structure demonstrated that CBP Asn1168, the conserved asparagine that contacts acetyllysine side chains in all bromodomains, makes a critical hydrogen-bonding interaction with the benzodiazepinone carbonyl group. It also showed electron density only for the R enantiomer, leading to the discovery that this scaffold stereoselectively inhibits CBP/p300. Structure-based drug design ultimately yielded CPI-637, a potent and stereoselective CBP/p300 bromodomain inhibitor (IC50 = 30 nM for CBP) with favorable selectivity against BRD4 bromodomains (IC50 = 11 μM).818 The benzodiazipinone scaffold of I-CBP112 was discovered by screening benzoxazepine molecules similar to the benzodiazepine class of BET bromodomain inhibitors (Figure 30d).808 Compared to more recent inhibitors, I-CBP112 only shows moderate affinity for CBP/p300 (KD = 151 nM) and modest selectivity against BRD4 bromodomains (KD = 5.6 μM).
3.4.2. CBP/p300 HAT inhibitors
A-485, the first potent, selective, and cellularly active chemical probe for a HAT domain—that of CBP/p300—was reported in 2017 (Figure 30e).812 Compounds that resulted from earlier efforts to inhibit HAT domains were typically limited by non-specific assay interference and off-target activities.799 This major breakthrough in HAT pharmacology was enabled by a virtual screen of 800,000 commercially available compounds.819 Hypothesizing that p300 may undergo conformational changes known to take place when other proteins bind acetyl-CoA, open conformations of the enzyme were computationally predicted and used for virtual ligand screening (VLS), which deviated from prior VLS efforts that had used published X-ray crystal structures. Among 1,300 VLS hits that were experimentally tested in a radioactivity-based p300 HAT enzyme assay, a hydantoin compound was prioritized for its low micromolar potency, lack of potentially reactive groups, and synthetic tractability. Iterative medicinal chemistry optimization arrived at a spiro-indane hydantoin core substituted with a methylurea moiety and then a spiro oxazolidinedione core with improved permeability, clearance, and oral bioavailability. Interestingly, the stereogenic spiro center showed opposite enantioselectivity in the spirooxazolidinedione series compared to the spirohydantoin (Figure 30e). Additional modifications to improve PK properties resulted in A-485, a potent, selective, and in vivo active chemical probe that inhibits the acetylation of CBP/p300 substrates (e.g. H3K27Ac and H3K18ac) without affecting other acetylation sites (e.g. H3K9ac).812,819 A co-crystal structure of A-485 bound to the HAT domain of p300 showed direct engagement of the acetyl-CoA binding pocket and biochemical analyses determined that it binds competitively with this cofactor.812,819 Notable interactions are made with a loop (L1) that is present in CBP/p300 but absent in other HATs, which was hypothesized to explain both its selectivity against other acetyltransferases and the lack of activity for the inactive control compound, A-486, a urea regioisomer of A485.812
Additional analogs of the oxazolidinedione and hydantoin scaffolds have been reported since the initial discovery of A-485.820–822 This includes the highly optimized spirohydantoin, B026, a CBP/p300 HAT inhibitor 30-fold more potent than A-485 (Figure 30e).823 B026 and other A-485 analogs have independently converged on replacing the methylurea with a methylpyrazole group, removing hydrogen bond donors to improve cellular permeability and oral bioavailability.820,821 A structural analysis of an optimized hydantoin analog has provided a possible explanation for the switch of stereochemical preference between the spirohydantoin and spirooxazolidinedione scaffolds, with the (S) spirocenter in the hydantoin series allowing for an additional hydrogen bonding interaction between the hydantoin core and p300. Structural analyses also allowed for the design of a covalent CBP/p300 inhibitor by attaching a phenylacrylamide to the urea of A485, which is located just 7 Å from Cys1450 of p300.824
A structurally distinct 2-aminopyridine chemical probe, CPI-1612, complements the spirohydantoin and spiroxazolidinedione scaffolds for CBP/p300.825 CPI-1612 originated from a high-throughput screen of 200,000 small molecules, which used a FRET-based assay to monitor p300-mediated acetylation of a histone peptide substrate.826 From ~2,500 hits, a total of 18 compounds passed a series of counter-screen assays, including both acetyl-CoA-competitive and histone peptide-competitive inhibitors.826 Medicinal chemistry optimization of an acetyl-CoA-competitive indole compound led to a redesigned amidopyridine core and, ultimately, the discovery of CPI-1612.825 Interestingly, these optimization efforts identified the same methylpyrazole group that is found in the optimized spirohydantoin compounds. Despite their unique chemical scaffolds, both classes of compounds position the methylpyrazole group in the same site on p300 and with the same orientation.821,825 CPI-1612 shows impressive sub-nanomolar potency against p300 and reduces H3K27Ac and tumor growth in vivo with just twice daily oral dosing of 0.5 mg/kg.825
3.4.3. CBP/p300 PROTACs
Both bromodomain and HAT domain inhibitors have been used to design CBP/p300 PROTACs. The first example, dCBP1, is a highly potent and selective CRBN-based PROTAC designed from the bromodomain ligand, GNE-781.827 Using an in silico docking analysis to nominate potential linker attachment sites for GNE-781 and A-485, the tetrahydropyran of GNE-781 was prioritized for PROTAC construction. dCBP1 was synthesized by substituting the tetrahydropyran of GNE-781 for a conjugatable piperidine and attaching 5’-aminothalidomide through a polyethylene glycol (PRG)-4 linker (Figure 29f). It induces potent, selective, and rapid degradation of both CBP and p300 in cell culture.827 The rapid disruption of CBP/p300-mediated acetylation and enhancer-dependent transcription by dCBP1 offers a well-characterized chemical probe for cellular study that complements bromodomain and HAT domain inhibitors.
Following the discovery of dCBP1, more potent degraders have been reported, including compounds that can induce degradation at low picomolar concentrations. JET-209, a CRBN-based PROTAC, is ~10-fold more potent than dCBP1 (Figure 30f).828 It is constructed from GNE-207 with a short and rigid piperidine linker using an alternative linker attachment site compared to dCBP1. It is highly active in vivo, degrading CBP/p300 within 3 hours of a 1 mg/kg dose.828 The GNE-049 scaffold was used in the successful pursuit of orally bioavailable degraders, as exemplified by the contemporaneously reported PROTACs, CBPD-409 and CBPD-268 (Figure 30f).829,830 CBPD-268 features a redesigned CRBN ligand, TX-16, which had previously afforded potent and orally available nuclear hormone receptor PROTACs.831,832 To minimize hydrogen bond donors, TX-16 was attached directly to a GNE-049 analog that replaced the tetrahydropyran for a cyclohexane, resulting in a compound, CBPD-268, with low picomolar potency and improved pharmacokinetic properties (Figure 30f).833 Impressively, it is capable of inducing tumor regression in a xenograft model of prostate cancer with 1 mg/kg oral dosing given just 3 times per week.
These and other bromodomain-based CBP/p300 degraders, such as the CCS1477-based PROTAC, QC-182,834 induce degradation of both paralogs equivalently.835 Interestingly, early progress in the development of paralog-selective degraders, which will be needed if the synthetic lethality observed between CBP and p300 is to be exploited therapeutically, has come in the form of HAT-based PROTACs. The first example, JQAD1, is a CRBN-based PROTAC constructed from the CBP/p300 HAT inhibitor, A485.836 While the degradation induced by JQAD1 is relatively slow for both CBP and p300, there is a kinetic window in which p300 degradation is observed without CBP degradation.836 In vivo, this window enables the selective degradation of p300 over CBP, establishing an important proof-of-concept for paralog-selective degradation.836 The HAT inhibitor CPI-1612 has also been used to develop CBP/p300 PROTACs,837 with a recently reported VHL-based compound, MC1, selectively degrading p300 over CBP at 1 μM.838 MC1 is more broadly active across cell lines compared to JQAD1 and appears to show improved kinetics of degradation. These studies highlight the possibility of progressing p300-selective PROTACs as drug candidates for CREBBP-mutant cancers.
3.4.4. Clinical translation of CBP/p300 antagonists
While the synthetic lethality between CBP and p300 provides a potentially powerful mechanism to selectively target several cancer types, indiscriminate inhibition of both CBP and p300 presents the possibility for on-target toxicity to normal tissues. Nevertheless, some cancer subtypes show hypersensitivity to CBP/p300 inhibition, suggesting that pharmacological inhibition may have nuanced effects across different lineages. Profiling of the orthosteric CBP/p300 HAT inhibitor, A-485, across diverse cancer cell lines has shown it is preferentially cytotoxic to multiple myeloma and prostate cancer cell lines.839 Even within a single lineage, sensitivity to A-485 varies widely, a phenomenon that cannot be attributed to differences in the extent of CBP/p300 inhibition.812,840,841 Therefore, it can be inferred that there are intrinsic differences in the cellular requirement for CBP/p300 activity, both within and across cancer cell lineages. This might reasonably be expected to hold true across normal tissues as well, which could make dual inhibition or degradation better tolerated than perhaps initially expected. Multiple myeloma, which is among the most acutely sensitive lineages to dual inhibitors and degraders, are also the most sensitive to genetic deletion of CREBBP/EP300,812,840,842 supporting the idea that CBP/p300 activity might be a targetable dependency with a reasonable therapeutic window for some cancer lineages.
If dual degraders or orthosteric HAT inhibitors prove to be too toxic, bromodomain inhibitors may prove a better tolerated approach to exploit lineage-restricted CBP/p300 dependencies. Unlike BET bromodomain inhibitors, which evict their targets from chromatin by competitively blocking their association with acetyllysine side chains, CBP/p300 bromodomain inhibitors do not inhibit chromatin localization.843 Both genetic and chemical studies show that the bromodomain-acetyllysine interaction is not required for CBP/p300 localization to chromatin.843 Instead, the bromodomain is part of the catalytic core, with CBP/p300 bromdomain inhibitors inhibiting catalytic activity rather than chromatin localization.843,844 Interestingly, however, the inhibition of CBP/p300 catalytic activity by bromodomain inhibitors is substrate-selective.771 Whereas CBP/p300 HAT inhibition provokes the loss of acetylation across thousands of lysines, bromodomain inhibition affects a much smaller subset of CBP/p300 substrates.771 This is very clearly observed by H3K27ac, which is depleted by both HAT and bromodomain inhibitors, and H3K18ac, which is only depleted by HAT inhibitors.843
Mechanistically, this milder inhibition of CBP/p300 function by bromodomain inhibitors might lead to improved tolerability. The lineage-specific dependency on CBP/p300 in prostate cancer has also been observed with CBP/p300 bromodomain inhibitors, motivating ongoing clinical trials for CCS1477 in castration-resistant prostate cancer (CRPC).811,845 Unfortunately, CCS1477 and the structurally distinct CBP/p300 bromodomain inhibitor, GNE-781, both induce dose-limiting thrombocytopenia in mice, presenting a possible on-target toxicity concern for CBP/p300 inhibition that has led to the implementation of intermittent dosing in humans.846,847
In the coming years, the results of clinical trials for CBP/p300 bromodomain inhibitors will answer many questions about CBP/p300 tolerability and lineage-restricted dependencies. The development of CBP/p300 HAT inhibitors has not yet matured to the point of clinical approval, which may, in part, be due to the toxicity of such compounds, but any new trials would be similarly informative. Much excitement would remain for paralog-selective degraders if this pharmacology can continue to progress. Other strategies to target CBP/p300 continue to be reported, such as the recent description of a photorelease strategy that aims to improve tolerability to this class of drugs.848 CBP/p300 bromodomain inhibitors have also found use in bifunctional molecules that recruit CBP/p300 for inducing targeted lysine acetylation in cells,329,849–851 suggesting new strategies to target or harness CBP/p300 for anti-cancer therapeutics may yet be awaited.
3.5. GCN5/PCAF
The proteins KAT2A and KAT2B, also known as GCN5 and PCAF, respectively, are two closely related lysine acetyltransferases that play critical roles in the regulation of gene expression. PCAF (P300/CBP-associated factor) and GCN5 (general control non-derepressible 5) are members of the GCN5-related N-acetyltransferase (GNAT) family and are key components of multi-protein transcriptional co-regulatory complexes, importantly the Spt-Ada-Gcn5-acetyltransferase (SAGA) complex. Several recent studies have reported SAGA complex subunits as potentially targetable cancer dependencies. An analysis of DepMap recently revealed that the core subunits TADA2B, SUPT20H, TAA1, and TAF5L are critical neuroblastoma-specific dependencies.852 This was orthogonally validated with CRISPR/Cas9- and degron-based loss-of-function perturbations and led to the further discovery that the combined activity of GCN5/PCAF is essential for MYCN-amplified neuroblastoma.852 Two contemporaneous reports found the SAGA subunit, SGF29, to be a leukemia-specific vulnerability.853,854 In particular, domain-focused CRISPR-Cas9 screening experiments in these reports found its chromatin reader Tudor domain to be necessary for SGF29-dependent leukemia survival, nominating a potential new target for drug discovery.853,854
Fragments that bind the bromodomain of PCAF and block acetyllysine recognition have been known since 2005 but generally featured limited affinity.855–858 In 2008, a phenotypic screening hit, CPTH2, was reported to inhibit the yeast Gcn5p,859 but it has since been described as a promiscuous electrophile with likely limited value as a chemical probe.799 Discovery of the first potent and cell-active PCAF/GCN5 chemical probes arrived with multiple reports published in 2016. The PCAF/GCN5 bromodomain inhibitor, GSK4027, originated from a screen of 30,000 molecules with known and putative acetyllysine mimetic groups using a fluorescence polarization (FP) assay that measured the displacement of a promiscuous bromodomain ligand (Figure 31a).860 A prioritized pyridazinone hit was used for structure-based optimization, which focused on improving PCAF/GCN5 potency while also improving selectivity against BRD4, ultimately leading to the discovery of GSK4027 and an enantiomeric negative control.860 The chemical probe, L-45 (also known as L-Moses), was reported contemporaneously and features a structurally distinct triazolopthalazine scaffold (Figure 31b). Using promiscuous bromodomain inhibitors as starting points, structure-based drug design, virtual screening, and iterative medicinal chemistry optimization delivered the chemical probe L-45 and an inactive enantiomer.861 Both GSK4027 (KD,PCAF = 1.4 nM) and L-45 (KD,PCAF = 126 nM) exhibit excellent selectivity against BRD4 bromodomains, with GSK4027 being more potent.861 Several additional compounds have been reported for GCN5/PCAF,862,863 notably including compounds related to GSK4027 that replace the pyridazinone warhead with a pyrrolopyrimidinone system.864 This new scaffold was discovered by virtual screening and optimized with 2-position substitutions to improve its potency (Figure 31c).864
Figure 31.

PCAF/GCN5 drug discovery. (a) Summary of medicinal chemistry optimization leading to GSK4027. (b) Summary of medicinal chemistry campaign leading to L-45. (c) Summary of hit to lead [(R,R)-36n] medicinal chemistry optimization of PCAF/GCN5 chemical probe. (d) Degrader of PCAF/GCN5.
In an immunological context, PCAF and GCN5 are reported to play a role in the production of inflammatory cytokines, as evidenced by the reduced levels of interleukin-6 and tumor necrosis factor in LPS challenged mice that have had PCAF genetically ablated.865,866 865,866Bromodomain inhibition of PCAF/GCN5 by GSK4027 are unable to phenocopy the immunomodulatory effect of PCAF knockout, which inspired the development of PROTAC degraders.867 Examination of the crystal structure of GSK4027 bound to PCAF illuminated an appropriate exit vector to which an E3 ligase ligand could be appended, resulting in the CRBN-based PROTAC, GSK699, and an inactive enantiomer, GSK702 (Figure 31d).867 GSK699 was able to deplete PCAF and GCN5 protein levels with 1.5–3 nM potency and induce a marked reduction of inflammatory markers. In addition to serving as a prominent proof of concept for inducing PCAF and GCN5 degradation, this study convincingly established the need to degrade GCN5/PCAF rather than inhibit their bromodomains. While GCN5/PCAF bromodomain inhibitors were not studied in the recent report of SAGA dependencies in neuroblastoma, treatment with GSK699 was found to inhibit the growth of neuroblastoma cells in vitro and in vivo.852 In support of this being an on-target effect, the inactive enantiomer of GSK699 could not suppress neuroblastoma growth, establishing SAGA dependencies as potential new targets for drug development.
3.6. MYST HAT dependencies
The first orthosteric HAT inhibitor to enter human clinical investigation targets the paralogous acetyltransferases, KAT6A (also known as MOZ) and KAT6B (MORF).868 KAT6A/B contain a MYST family (named for MOZ, Ybf2, Sas2, Tip60) HAT domain consisting of an acetyl-CoA-binding motif and a zinc finger fold. They function as transcriptional co-regulators by catalyzing lysine acetylation, with an emerging consensus on their importance for H3K23 acetylation.869–872 KAT6 can impact diverse physiological processes, including development,873–878 cellular senescence,879–881 and hematopoiesis.882–886 In cancer, KAT6A was originally discovered as a partner of CBP-fusion oncogenes and has since been described in several other oncogenic fusions.789,791,887–900 In addition to its presence in fusion oncogenes, multiple studies have nominated wild-type KAT6A as a genetic dependency of hematological malignancies, namely lymphoma and AML.900,901 However, KAT6A acetyltransferase activity is also essential for the maintenance of normal hematopoietic stem cells and its roles in normal and malignant hematopoiesis are closely linked.883,884,886 For example, its importance in lymphoma is closely related to its effect on the proliferation of normal B-cell progenitors901 and, similarly, the discovery of KAT6A dependency in AML originated in a forward genetic screen for chromatin regulators that block terminal differentiation.900
The first MYST acetyltransferase inhibitors were discovered in a high-throughput screen using a biochemical assay that measures the acetylation of a synthetic histone peptide.902 Aided by efficient biophysical assays for hit triage, a naphthylsulfonohydrazide screening hit with low micromolar potency, CTx-0123143, was identified as the only true-positive hit from a screen of 243,000 compounds.902 Using this scaffold for extensive medicinal chemistry optimization resulted in the discovery of WM-8014 and WM-1119, two potent and selective KAT6A/B acetyltransferase inhibitors (Figure 32a,b).881,903,904 These acetyl-CoA-competitive inhibitors directly engage the MYST catalytic pocket, positioning the acylsulfonylhdrazide to mimic the contacts of the acetyl-CoA diphosphate group.904 Both WM-8014 and WM-1119 bind KAT6A/B with high affinity (KD < 10 nM), but WM-1119 features improvements in selectivity (250-fold selective over KAT7) and pharmacokinetic properties.881 These improvements enabled limited studies of KAT6A/B inhibition in vivo, with WM-1119 provoking an anti-lymphoma effect in mice with 3–4 times daily dosing (50 mg/kg per dose ).881
Figure 32.

Small molecules targeting MYST family lysine acetyltransferases. (a-c) Chemical probes that inhibit KAT6A and KAT6B acetyltransferase activity. (d) Small molecule inhibitor of KAT7/HBO1 acetyltransferase activity.
The same high-throughput screening cascade that was used to discover the acylsulfonylhdrazide series was later used to discover a benzisoxazole sulfonamide scaffold with improved pharmacological properties.870 An initial screen of 250,000 compounds led to the discovery of 3 biophysically validated hits, including the benzisoxazole CTx-533. Medicinal chemistry was supported by biochemical, biophysical, and cellular assays to measure potency and selectivity for KAT6A, ultimately resulting in the discovery of PF-9363 (Figure 32c).870 A co-crystal structure with KAT6A demonstrated that the sulfonamide and isoxazole form the functional acetyl-CoA diphosphate mimic, similar to the acylsulfonylhydrazide moiety in WM-1119. PF-9363 is exceptionally potent, inhibiting KAT6A and KAT6B with potencies of 270 pM and 2.4 nM, respectively, and it possesses excellent drug-like physicochemical properties for study in animal model systems.870 Among the other 3 MYST acetyltransferases encoded in the human genome, PF-9363 is >250-fold less potent at inhibiting KAT7 and >1,000-fold less potent for KAT5/8, altogether affording an excellent chemical probe to study KAT6A/B and, at higher concentrations, KAT7.870
In AML, KAT6A dependency was first proposed to be mediated through the chromatin reader protein ENL, another AML dependency.900 Specifically, it was suggested that ENL binds to chromatin by recognizing H3K9ac marks deposited by KAT6A. While PF-9363 was later used to demonstrate that KAT6A/B are not likely involved in the acetylation of H3K9 (at least not globally),870 a functional connection between ENL and KAT6A is supported by a physical interaction whereby acetylated lysine side chains from KAT6A bind into the YEATS domains of AF9 and ENL.905 More recent study has shown that KAT6A is a dependency across several models of acute leukemia, but combining KAT6A/B with KAT7 inhibition provides a stronger anti-leukemic effect.906,907 Prior to this report, KAT7 acetyltransferase activity had been discovered as a critical requirement for the maintenance of leukemia stem cells populations,908 which has been attributed to a functional and physical interaction between KAT7 and menin-MLL complexes.908–910 Consistent with this link, both genetic knockout and pharmacological inhibition of KAT6A have been found to sensitize MLL-fusion leukemia to menin-MLL inhibition in vitro.911 Indeed, combining PF-9363 with the menin-MLL inhibitor, VTP-50469, elicits profound anti-leukemia effects in vivo, overcoming primary and acquired resistance to menin-MLL inhibitors.906,907 A related effect has been observed in gastrointestinal stromal tumors (GIST), where a correlated dependency on KAT6A and MLL has recently been described.912 Interestingly, combined treatment with WM-1119 and the menin-MLL inhibitor, VTP-50469, inhibits GIST growth in vivo, despite both compounds failing to show single-agent activity in this disease.912
In the future, KAT6/7 acetyltransferase inhibitors may have considerable potential as acute leukemia therapeutics, especially in combination with the now FDA-approved class of menin-MLL inhibitors. Currently, however, both clinical-stage KAT6A/B inhibitors, PF-07248144 and MEN-2312, are being evaluated in breast, prostate, and lung cancer (NCT04606446 and NCT06638307). Based on prior observations that KAT6A is frequently amplified in breast cancer and that KAT6A knockdown reduces estrogen receptor (ER) levels,913,914 PF-9363 was first studied in ER-positive breast cancers with KAT6A overexpression.870 In breast cancer cell lines, anti-proliferative responses to PF-9363 are most closely associated with KAT6A overexpression and ER-positive luminal subtypes, which is consistent with the observed ability of PF-9363 to suppress ER-dependent transcriptional programs.870 PF-9363 elicits on-target anti-tumor activity in mouse xenograft models of breast cancer, including some models that have shown drug-induced tumor regressions.870 In a first clinical report from the Phase 1 clinical trial, NCT04606446, the KAT6A/B inhibitor, PF-07248144, was found to be sufficiently well tolerated to be given at doses that cause a pharmacodynamic response (loss of H3K23ac) in peripheral blood mononuclear cells and tumors.868 This preliminary report showed PF-07248144 possesses antitumor activity both as monotherapy and in combination with the selective estrogen receptor degrader, fulvestrant, establishing KAT6A/B inhibition as a compelling therapeutic strategy for metastatic breast cancer.
3.7. NSD2 oncogene dependency
H3K36 dimethylation (H3K36me2) is catalyzed by the nuclear SET domain (NSD) family of histone methyltransferases – NSD1, NSD2, and NSD3 – and is associated with active gene transcription, mRNA processing, and DNA damage repair.915 NSD2 is also known as MMSET (multiple myeloma SET) and WHSC1 (Wolf Hirschhorn syndrome candidate 1), names that originate from the association of NSD2 alterations with these diseases. In multiple myeloma (MM), recurrent NSD2 mutations or increases in NSD2 expression create an increase in H3K36 methylation,916,917 with consequences on cellular adhesion,918 proliferation,919 and transformation.920 NSD2 also plays consequential roles in a variety of other cancer types, including lung cancer,921 prostate cancer,922 and pediatric malignancies,923 among others.924–929 In such cases, NSD2 can either be overexpressed, commonly due to chromosomal translocations, or feature point mutations. For example, oncogenic t(4;14) translocations, which are observed in 15–20% of MM patients, place the NSD2 gene under control of the Ig heavy chain locus, dramatically increasing its tissue-specific expression.930 E1099K, D1125N and T1150A mutations enhance its interaction with the nucleosome by destabilizing the autoinhibitory loop, keeping it in an open hypermorphic state and allowing for hypermethylation.931–935 E1099K and T1150A mutations are oncogenic and poor prognostic indicators for B-ALL and are associated with relapse in patients with mantle cell lymphoma.936–939 These findings have nominated NSD2 as a potential therapeutic target, motivating the development of chemical tools and drug candidates.
3.7.1. NSD SET domain inhibitors
NSD2 harbors a SET methyltransferase domain and multiple protein folds that mediate chromatin interactions, including two PWWP (proline-tryptophan-tryptophan-proline) domains, multiple PHD (plant homeodomain) fingers, and an HMG (high mobility group) box (Figure 33a). While potent inhibitors have been achieved through the development of peptidomimetic inhibitors like PTD2, a pentapeptide that replaces the substrate lysine with norleucine, these compounds are not suitable for cellular studies.940 Many efforts to develop small-molecule NSD2 inhibitors have focused on repurposing quinazoline inhibitors of the G9a methyltransferase, like BIX-01294 (Figure 33b). These quinazoline inhibitors, which bind to the histone substrate-binding site of G9a,941–946 can also possess weak off-target activity against NSD2 in vitro.947,948 After initial difficulties with structure-guided optimization, extensive structure-activity relationship studies have recently discovered analogs that potently inhibit NSD2 and are selective against G9a and several other non-NSD methyltransferases.949,950 While still early, preliminary biological studies with these compounds suggest they can preferentially inhibit the growth of cell lines with oncogenic NSD2 alterations and suppress tumor growth in mouse xenograft models.949
Figure 33.

NSD2 drug discovery. (a) Summary of NSD2 domain structure for drug discovery. (b) Small-molecule inhibitors of the NSD SET domain. (c) Discovery and optimization of NSD2-PWWP1 ligands and structure of UNC6852 bound to NSD2-PWWP1 (PDB: 6XCG). (d) Small-molecule degraders of NSD2 based on UNC6834 and a summary of their mechanism of action.
The NSD2 SET domain has shown signs of ligandability with other scaffolds as well, but none have yielded potent and selective inhibitors.951–953 For example, the natural product sinefungin, which is an analog of the SAM cofactor used by the NSD2 SET domain, is known to inhibit NSD2 but is weakly potent and has yet to afford highly active analogs.948,954 A recently developed high-throughput screening assay for NSD2 may afford additional scaffolds, but the hits identified in the first reported screen possess notable liabilities.951 The assay for this screen used native nucleosomes purified from HeLa cells as the NSD2 substrate, since NSD2 is not catalytically active on synthetic histone peptides,955 ultimately enabling high-throughput screens.951 However, in a preliminary screen of approximately 16,000 compounds, most confirmed hits show notable liabilities. For example, the screening hit, DA3003–1, a previously reported Cdc25 phosphatase inhibitor, was validated to bind the SET domain and inhibit NSD2 activity, but it was also found to inhibit dozens of other methyltransferases.951 Some improvements in selectivity have been reported for structural analogs of DA3003–1,956 but DA3003–1 is known to participate in redox cycling and was originally proposed to engage NSD2 through nonspecific interactions.951 While NSD2 SET has proven a challenging target for discovery chemistry, further improvements are likely possible. Indeed, a Phase 1 clinical trial is currently underway to evaluate KTX-1001, described as a selective NSD2 inhibitor, for the treatment of multiple myeloma (NCT05651932).957 Recent work described the discovery of potent, selective NSD2 catalytic inhibitors related to KTX-1001, IACS-17596 and IACS-17817) that deplete H3k36me2, reprogram chromatin and suppress oncogenic transcriptional programs in KRAS-driven pancreatic and lung cancer models.958 These compounds act through a SAM-competitve mechanism coupled with catalytic channel obstruction, leading to durable tumor regression and prolonged survival in preclinical systems.958 Together, these findings highlight the therapeutic potential of directly targeting NSD2’s enzymatic activity, complementing ongoing clinical evaluation of KTX-1001 in multiple myeloma.
Selectivity within the NSD family has recently been accomplished with the discovery of covalent NSD1 inhibitors.959 NSD1 is involved in recurrent leukemogenic chromosomal translocations that fuse it with NUP98, making it an attractive target for drug discovery.960–962 A series of covalent NSD1 inhibitors were recently developed from a benzothiazole scaffold identified in an NMR-based fragment screen of 1,600 compounds.959 The initial benzothiazole fragment hit, BT1, was fashioned with a thiocyanate handle after NMR studies showed compound-induced perturbations to the chemical shift of Cys2062.959 A co-crystal structure with the resulting compound, BT3, showed formation of a disulfide bond and illustrated the emergence of a channel-like pocket that is absent in the apo structure.959 Replacing the thiocyanate with a methyl aziridine yielded BT5, which irreversibly engages NSD1 SET.959 The compound binds NSD1-SET in cells, inhibits HMT activity, and selectively inhibits the growth of murine bone marrow progenitor cells transformed with the oncogenic NSD1-NUP98 fusion over other oncogenes.
3.7.2. PWWP domains and the NSD family
The NSD2 PWWP1 domain, and PWWP domains more generally, represent tractable targets for ligand discovery. The PWWP1 domain of NSD2 binds to H3K36me2-modified nucleosomes through a conserved aromatic cage, making cation-π interactions with the methyl lysine ammonium.963–965 BI-9321, a chemical probe for NSD3, was the first small-molecule ligand to be reported for a PWWP domain.966 NSD3 has been proposed to mediate the binding of the chromatin remodeler protein, CHD8, to BRD4, providing a function that is indispensable for acute leukemia survival.967 This function is specific to the short isoform of NSD3 which includes its first, but not second, PWWP domain. Fragment-based ligand discovery for the NSD3 PWWP1 domain previously identified a methylimidazole core that makes CH-π interactions with the conserved PWWP aromatic cage that binds methyl lysine.966 Structure-based design afforded a ligand, BI-9321, that possesses mid-nanomolar affinity for NSD3 and is selective against 14 other PWWP domains tested when assayed by DSF.966 High concentrations of the ligand were required to show anti-leukemia activity in tissue culture, suggesting that additional improvements in ligand affinity would be required to more fully validate NSD3 as a target for drug discovery.966 However, BI-9321 has been used to construct NSD3 PROTACs, such as MS9715, and additional scaffolds have recently been reported to support targeted protein degradation.968–970 MS9715 shows weakly potent but selective degradation of NSD3 and modest anti-leukemia activity in tissue culture.
In contrast to NSD3, the PWWP domain of LEDGF (also known as PSIP1) has proven more challenging for ligand discovery. LEDGF is selectively required for transformation and oncogenic gene expression in MLL-rearranged leukemias.971 Its PWWP domain binds to nucleosomal H3K36me3 and is important for facilitating the binding of MLL to its chromatin targets.972–974 While a recently reported DNA encoded library (DEL) screen failed to identify any starting points for LEDGF PWWP ligand optimization, a chemoproteomics-based method for mapping ligand-binding sites in cells suggests that the PWWP proteins, HDGF and LEDGF, can be stereoselectively bound by small-molecule ligands.975
Small-molecule ligands of the NSD2 PWWP1 domain were first identified from a virtual screen for the PWWP domain of ZMYND11.976 While none of the hits from this virtual screen were found to bind ZMYND11, additional testing against other PWWP domains led to the discovery of a fragment-like ligand with mid-micromolar affinity for NSD2 PWWP1. A scaffold-hopping approach was then used to identify a ligand with a greater number of commercially available analogs that could be used to evaluate SAR. This led to the discovery of a tertiary N-cyclopropyl amide compound with single-digit micromolar affinity. A co-crystal structure was solved, which confirmed engagement of the methyllysine-binding site of NSD2 and showed that the isopropyl group, which was critical for activity in SAR studies, fills a narrow hydrophobic pocket at the bottom of the aromatic cage. This molecule, MR837, demonstrated that the NSD2 PWWP1 domain is a ligandable target and, furthermore, served as a crucial starting point for the optimization of potent and selective chemical probes (Figure 33c).
Structure-guided optimization of MR837, via the key intermediate, MRT866, led to the discovery of UNC6934, a potent and selective chemical probe for the NSD2 PWWP1 domain (Figure 33c).977 It binds NSD2 PWWP1 with an affinity of 91 nM and is selective against all PWWP domains tested. Fascinatingly, liberation of the cyclopropyl ring to the corresponding isopropyl was sufficient to entirely abrogate compound binding, providing a useful inactive negative control, UNC7145. UNC6934 effectively disrupts the interaction between purified H3K36me2 nucleosomes and the isolated PWWP1 domain of NSD2, but cannot do the same to full-length NSD2 protein, likely because it makes additional contacts with nucleosomal DNA outside of the methyllysine-binding pocket. Crystallographic studies proved informative, highlighting that UNC6934 binds adjacent to the basic DNA binding surface in the canonical methyllysine binding pocket of the PWWP1 domain, but does not compete with DNA. This structure again highlighted the importance of the constrained geometry of the cyclopropyl group, which inserts into the aromatic cage with exceptional shape complementarity. It also allowed for the rational design of a biotin-conjugated affinity probe, which was used to demonstrate the exquisite selectivity of UNC6934 for NSD2. In cells, inhibition of the NSD2 PWWP1 domain by UNC6934 modestly increases nucleolar accumulation of NSD2, but it does not displace NSD2 from chromatin globally, reduce H3K36me2 levels, or impair the growth of multiple myeloma cells. This highlights a broader challenge with domain-selective probes: while they can be useful for dissecting local structural functions, targeting a single domain of a large, multidomain chromatin regulator may not phenocopy inhibition or loss of the full-length protein, limiting their translational potential.
Due to PWWP1 engagement eliciting minimal phenotypic effects, UNC6934 was quickly repurposed for targeted protein degradation. However, rather than conjugating UNC6934 to the CRBN or VHL ligands that are most frequently used for PROTAC discovery, the first reported NSD2 degraders were designed by decorating UNC6934 with basic amino acid side chains.978 Inspired by a previous report,979 this approach was meant to recruit UBR-box E3 ligases that recognize N-degons that are rich in arginine, lysine, and histidine. Using the same attachment site as previously used to synthesize biotinylated analogs, simple heterobifunctional constructions of UNC6934 and arginine were found to cause modest NSD2 degradation.978 An optimized molecule, UNC8153, which features a simple polymethylene-linked primary amine, was found to elicit potent (sub-micromolar) and exclusive degradation of NSD2 (Figure 33d). However, while a free N-terminal alpha amino group is necessary for degron recognition by UBR-box domain E3 ligases,980–982 eliminating this element still afforded active degraders and knockdown of UBR proteins could not rescue NSD2 degradation.978 A later study found that the primary amine of UNC8732 is converted to an aldehyde, which forms a reversible covalent bond with the E3 substrate receptor, P22, to mediate targeted protein degradation (Figure 33d).983 These findings were reported contemporaneously with an independent discovery of FKBP12 degraders that proceed through the same mechanism. Both studies found that a previously reported XIAP degrader proceeds through this mechanism,984–986 highlighting it as a generalizable approach to convert ligands into degraders.985 (Another contemporaneous report, which used a CRISPR activation screen to uncover an alternative mechanism of ligand-induced degradation through FBXO22, further established the ability to FBXO22 to support targeted protein degradation.312)
UNC8153-induces rapid degradation of NSD2, which causes a delayed reduction in H3K36me2 levels after nearly a week of treatment, likely demonstrating that H3K36me2 modifications must be lost by dilution through cell divisions.978 While UNC8153 was shown to reduce the growth of multiple myeloma cells expressing NSD2-E1099K, these effects are modest and only observed at high concentrations of the drug.978 Furthermore, UNC8153 is completely ineffective against t(4;14)-positive multiple myeloma cells in vitro. This is potentially due to the sub-maximal degradation elicited by UNC8153, as a more effective analog, UNC8732, showed more potent anti-proliferative activity.984 UNC8732, which features a constrained linker attachment site but preserves the primary amine modification, is 10-fold more potent and elicits more complete degradation of NSD2 than to UNC8153 (Figure 31c). It also suppresses the growth of ALL cells expressing NSD2 E1099K in tissue culture, although multiple weeks of drug exposure are required to observe this effect.984
Multiple CRBN-based NSD2 PROTACs have now been reported. MS159, which is constructed from the UNC6934 scaffold, degrades NSD2 through CRBN but elicits off-target degradation of the CRBN neo-substrates, IKZF1 and IKZF3.987 Since IKZF1/3 degradation is responsible for the therapeutic effect of thalidomide analogs in multiple myeloma,988,989 the degree to which NSD2 degradation is responsible for MS159 anti-proliferative effects is unclear. The PROTAC LLC0424, which boasts a 5’ thalidomide moiety linked to a UNC6934 derivative through a conformationally rigid amido piperidine linker, induces potent and exclusive degradation of NSD2 and is active both in vitro and in vivo.990 The steadily improving activity of NSD2 degraders suggest that they may have substantial potential for clinical development alongside the undisclosed SET domain inhibitor, KTX-1001.
3.7.3. SETD2
The histone methyltransferase SETD2 is the exclusive writer of the trimethylation mark on lysine 36 of histone H3 (H3K36),991 a chromatin modification linked to transcriptional fidelity, RNA splicing, DNA repair, and genomic stability.992–994 While SETD2 has been shown to function as a classic tumor suppressor in many solid malignancies—where it is often biallelically inactivated995,996—emerging evidence supports a different and targetable role in hematologic malignancies. SETD2 mutations are commonly heterozygous in leukemias, lymphomas, and multiple myeloma, suggesting a haploinsufficiency as opposed to complete loss of function.997 This partial loss sensitizes these cells to SETD2 inhibition, creating a unique therapeutic opportunity, especially in genetically or epigenetically primed contexts. Some flagship examples include that of t(4;14) multiple myeloma, as well as in MLL-rearranged leukemias. In t(4;14) MM, NSD2 overexpression leads to elevated H3k36me2 levels, driving cells toward a reliance on SETD2 to mediate the balance between di- and tri-methylated states.998 In MLL-rearranged leukemia, SETD2 disruption alters transcriptional programs and DNA repair pathways; selective inhibition of SETD2 catalytic activity may phenocopy or exacerbate these states, leading to synthetic lethality while sparing healthy tissue.999 Furthermore, recent pan-cancer analyses demonstrate that SETD2 mutation or reduced expression is associated with widespread dysregulation of DNA methylation across diverse cancer types—including hematologic and solid tumors—suggesting that SETD2 loss may drive tumorigenesis via epigenetic reprogramming beyond histone methylation.1000 Altogether, these biological observations have motivated chemical efforts to fashion potent and selective SETD2 inhibitors to continue to study SETD2 biology.
The first selective chemical probes for SETD2 inhibition were derived from the cofactor analog sinefungin by introducing simple alkyl substituents, yielding compounds such as N-propyl sinefungin (Pr-SNF) and N-benzyl sinefungin.1001 These analogs showed sub-micromolar potency (IC₅₀ = 0.48–0.80 μM) and more than ten-fold selectivity for SETD2 over related methyltransferases, providing the first demonstration that selective inhibition of SETD2 is feasible.1001 Beyond serving as early tool compounds, they highlighted the tractability of SETD2 as a drug target and set the stage for subsequent inhibitor development by establishing important structural foundations.
Building on these insights, Epizyme undertook a structure-based drug discovery campaign, screening a methyltransferase-biased library and identifying a 2-amidoindole scaffold with activity against SETD2.1002 Medicinal chemistry optimization yielded EPZ-719, a potent, reversible SETD2 inhibitor with nanomolar biochemical potency (IC₅₀ = 8 nM) and low-nanomolar activity in cellular assays monitoring H3K36me3 levels. EPZ-719 demonstrated sufficient metabolic stability and selectivity to serve as an in vivo tool compound for target validation studies.1002 However, it lacked adequate solubility and oral bioavailability to advance into clinical development.
To address these limitations, a conformational design strategy was used, incorporating a rigid cis-(1R,3S)-diaminocyclohexyl linker to enhance metabolic stability and oral pharmacokinetics.1003 This approach yielded EZM0414, a highly selective and orally bioavailable SETD2 inhibitor with potent biochemical and cellular activity (IC₅₀ = 18 nM and 31 nM, respectively) and robust antiproliferative effects in multiple myeloma cell lines bearing t(4;14) translocations.1003 Crystallographic studies confirmed that EZM0414 binds deep within the lysine channel of SETD2, displacing autoinhibitory residues and stabilizing the enzyme in an open conformation.1003 EZM0414 exhibited favorable ADME properties and demonstrated in vivo activity in xenograft models, ultimately entering clinical evaluation for the treatment of multiple myeloma and B-cell malignancies.1003
The evolution from sinefungin-based probes to EPZ-719 and finally to the clinical candidate EZM0414 exemplifies the power of rational, structure-guided drug discovery in the epigenetic landscape. In hematologic cancers, where SETD2 functions as a haploinsufficient dependency rather than a tumor suppressor, these chemical probes provide critical tools for mechanistic dissection and therapeutic intervention.
4. State- and lineage-specific dependencies
In addition to genetically restricted vulnerabilities, some targets show cancer-specific activity that is independent of a specific genetic alteration. These include lineage-specific dependencies, in which a target or a drug-like chemical agent shows activity in specific cancer lineages, sometimes without a clear understanding of the underlying basis for its cancer-specific effect. State-specific dependencies—factors that are important only under certain conditions or cell states—can also make for compelling anti-cancer drug targets.
4.1. LSD1
LSD1 (lysine-specific histone demethylase 1, also known as KDM1A) is a flavin adenine dinucleotide (FAD)-dependent lysine demethylase and transcriptional co-regulator. Before being reported as the first known example of a histone demethylase,1004 LSD1 had initially been identified in transcriptional co-repressor complexes (e.g. CoREST and NuRD) and found to possess homology with amine oxidases.1005–1008 LSD1 regulates gene expression by removing H3K4 and H3K9 mono- and di-methylation marks from gene enhancers and also through non-enzymatic scaffolding functions.1004,1009–1015 Its FAD-dependent mechanism of catalysis, which requires a protonated amine as a substrate, makes it unable to demethylate the trimethylated H3K4 modification that is primarily present at promoters.1004 Current interest in LSD1 drug development is largely motivated by experiments showing that LSD1 is required for AML and lung cancer, in addition to other malignancies.1011,1013,1016–1025
The earliest efforts to inhibit LSD1 used mechanisms similar to inhibitors of related monoamine oxidase enzymes, which covalently modify the FAD cofactor within the enzyme active site.1026 The first, a peptidic inhibitor based on the H3K4 substrate, used a propargylamine analog of Lys4 to form a covalent adduct with FAD, making it a suicide inhibitor of LSD1 enzyme function.1027–1030 In a contemporaneous report, the FDA-approved antidepressant monoamine oxidase inhibitor, trans-2-phenylcyclopropylamine (tranylcypromine or TCP), was found to be the first small-molecule and cell-active inhibitor of LSD1.1031,1032 Tranylcypromine reacts to form a covalent linkage to FAD within a hydrophobic pocket of the LSD1 active site (Figure 34a,b).1031–1034 Many groups have discovered or designed tranylcypromine-based LSD1 inhibitors with substitutions on both the phenyl ring and the primary amino group,1016,1033,1035–1037 which has afforded several clinical-stage tranylcypromine-based LSD1 inhibitors.1031,1033,1035,1038–1040 These include GSK2879552, ORY-1001 (iadademstat), bomedemstat, INCB059872, and JBI-801 (LSD1/HDAC dual inhibitor) (Figure 34c).1016,1021,1041–1045 In addition to these irreversible inhibitors, two structurally differentiated reversible inhibitors, pulrodemstat (CC-90011) and seclidemstat (sp-2577), have also advanced to clinical investigation (Figure 34c).1046–1048
Figure 34.

LSD1 inhibitors. (a) Formation of the TCP-FAD tetracyclic adduct by LSD1 inhibitors. (b) Crystal structure of (+)-tPCPA in complex with LSD1 (PDB: 2XAH). Important interactions of TCP-FAD with LSD1 amino acids shown in green. (c) Chemical structures of additional irreversible inhibitors (TCP moiety highlighted in pink). (d) Structure of reversible LSD1 inhibitors.
ORY-1001 was discovered using computational modeling of LSD1 crystal structures to assist in the design of more than 800 tranylcypromine analogs.1021 It is a potent (IC50 = 18 nM) and selective inhibitor of LSD1, binding rapidly and irreversibly to FAD. It elicits antiproliferative effects at low nanomolar concentrations in AML cell lines, causing an increase in terminal differentiation, and is active against AML and T-ALL xenografts in vivo. As a clinical agent, ORY-1001 has shown promising signals for tolerability and efficacy in a Phase 1 report of relapsed and refractory acute myeloid leukemia (R/R AML),1045 further encouraging LSD1 as a target for drug development. Due to promising pre-clinical results, iadademstat was explored as a combinatorial therapy, recently progressing to a Phase 2 clinical trial in combination with the azacitidine.1049 In patients with newly diagnosed AML, the therapeutic combination displayed a manageable safety profile and showed an encouraging 52% of patients achieving complete remission.1049 Additional clinical trials exploring other combinatorial therapies with ORY-1001 in myeloid malignancies and solid tumors are ongoing (NCT05546580, NCT05420636, NCT06357182, and NCT06287775).
Several years before ORY-1001 was reported, structurally related compounds were discovered in a screen of 2.5 million compounds using a biochemical assay of LSD1 enzyme activity.1016 Optimization efforts led to the discovery of the selective LSD1 inhibitors, GSK-LSD1 and GSK2699537, as well as the clinical agent, GSK2879552 (Figure 34c).1016 GSK-LSD1 and GSK2879552 both contain a tranylcypromine core and a piperidine moiety, while GSK2879552 includes a benzoic acid extending from the piperidine nitrogen. In a proliferation screen, GSK2879552 inhibited the growth of AML and some small cell lung carcinoma SCLC cell lines. This translated to in vivo antitumor activity in SCLC xenograft mouse models,1016 which has motivated the clinical translation of GSK2879552 and other tranylcypromine-based inhibitors, such as INCB059872,1050 for SCLC. However, despite its preclinical promise, clinical trials using GSK2879552 were suspended due to severe adverse effects and insubstantial efficacy.1051 In mouse models, GSK2879552 was reported to cause anemia, but the most common clinical toxicity in humans was thrombocytopenia and the most serious adverse event was encephalopathy, which caused several patient deaths.1051 Fortunately, encephalopathy has not been observed in other clinical trials, suggesting it is not likely a class-wide effect of on-target LSD1 inhibition. According to clinical trial records (NCT02712905), a strategic business decision was made to terminate clinical investigation of INCB059872, leaving only ORY-1001 and bomedemstat (IMG-7289) in clinical development. IMG-7289, which has advanced to Phase 2b clinical trials to assess pharmacokinetic and safety in patients with myelofibrosis,1052 is effective in preclinical models of myeloproliferative neoplasms (MPN), castration resistant prostate cancer, and AML, among other cancers.1041,1053–1055
Despite being designed and demonstrated to inhibit the enzymatic activity of LSD1, it is now clear that the effects of LSD1 inhibitors are primarily mediated by disrupting a protein-protein interaction with GFI1.1011,1013,1014 This was first uncovered in characterization of the tranylcypromine-based inhibitor, T-3775440, which was found to inhibit LSD1 interactions with the transcriptional repressor, GFI1, while leaving the CoREST complex intact.1011 A study published shortly after this initial report found that the tranylcypromine-based inhibitor, OG86, alters gene transcription much earlier than it impacts histone lysine methylation levels, which led to the discovery that the demethylase activity of LSD1 is not required for LSD1-dependent AML growth and survival.1013 Instead, OG68 also disrupts the LSD1-GFI1 interaction, leading to loss of LSD1 from chromatin and a decrease in GFI1-mediated transcriptional repression.1013 A CRISPR-based method for high-density mutagenesis that was later used to uncover mechanisms of resistance to LSD1 inhibitors discovered that several mutations conferring drug resistance to LSD1 inhibitors inactivated enzyme function, unambiguously demonstrating that demethylase inhibition is not necessary for the phenotypic effects of LSD1 inhibitors.1014
The interaction between LSD1 and GFI1 has also been highlighted in a study showing that LSD1 inhibitors are effective against GFI1-driven medulloblastoma, both in vitro and in vivo.1056 Interestingly, it was recently discovered that UM171,1057 a small molecule that induces the degradation of LSD1/CoREST through the E3 substrate receptor KBTBD4,133,1058 mimics the effects of medulloblastoma mutations in KBTBD4 that also lead to neomorphic degradation of LSD1/CoREST.131 Mechanistically, UM171 induces CoREST degradation by binding into the active site of HDAC1/2 and stabilizing their interaction with KBTBD4, ultimately making contacts with HDAC1/2 that are analogous to those made by mutant forms of KBTBD4 in medulloblastoma, both of which lead to LSD1/CoREST degradation.129,130 This pro-tumorigenic effect of LSD1/CoREST degradation stands in contrast to LSD1 inhibitors and may provide an important phenotype to monitor in future preclinical and clinical investigation.
A reversible LSD1 inhibitor, puldrodemstat (CC-90011) has also advanced to clinical trials for AML and SCLC (Figure 34d).1046,1047,1059 CC-90011 was developed from a hit identified in a high-throughput screen of more than 300,000 compounds and has an optimized pyrimidinone core (Figure 34d).1059 It binds reversibly to LSD1, contacting the catalytic lysine and extending into the FAD-binding pocket, selectively inhibiting LSD1 activity with sub-nanomolar potency and showing anti-AML effects at single-digit nanomolar concentrations. Clinical trial results will be required to evaluate the benefits of using covalent tranylcypromine-based inhibitors versus this new class of reversible LSD1 inhibitors, but a recent comparative study of LSD1 inhibitors found that while CC-90011 and ORY-1001 are equivalently potent enzymatic inhibitors (the most potent among all clinical stage inhibitors of LSD1), CC-90011 is a weaker inhibitor of both LSD1-GFI1 interaction and AML cell growth.1060
4.2. KDM6B
The Jumonji C (JmjC) family of histone demethylase enzymes differ from those of the LSD family through their demethylase mechanisms. Unlike the LSD enzymes that utilize an amine oxidase reaction in the removal of methylation marks, the Jumonji domain family are iron-containing demethylase enzymes that use α-ketoglutarate as a cofactor to catalyze a hydroxylation reaction that can also operate on trimethyllysine substrates.1061–1068 KDM6A (UTX), and KDM6B (JMJD3) selectively demethylate H3K27me2/3,1069–1071 reversing transcriptionally repressive chromatin states mediated by PRC1 and PRC2.600 A loss of these marks is associated with stem-like gene expression during development and KDM6A/B are essential for normal transcriptional programs in development and for oncogenic programs in cancers such as T-cell acute lymphoblastic leukemia (T-ALL) and glioma.1072–1074
The first KDM6 inhibitor, GSK-J1, was developed using structure-guided optimization of a weak hit identified in a screen of 2 million small molecules (Figure 35a).1075 GSK-J1 binds competitively with a-ketoglutarate but not with the histone substrate. Its propanoic acid mimics the a-ketoglutarate cofactor binding, the tetrahydrobenzazepine fits into a narrow cleft to mimic a proline residue on the histone substrate (Pro30) and the pyridylpyrimidine interacts with the Fe(II) cofactor, which is critical for inhibition of JMJD3 (Figure 35b).1075 The polar carboxylate of GSK-J1 limits cell permeability, so GSK-J4, an ethyl ester prodrug of GSK-J1 is used for cell-based studies (Figure 35a).1075 GSK-J4 significantly reduces the proliferation of T-ALL, prostate, glioma, and other cancers.1072,1074,1076 Unlike LSD1, these effects appear to be due to the catalytic activity of JMJD3, since introduction of catalytically inactive JMJD3 could not rescue the effects of GSK-J4 inhibition.1072 More advanced compounds, including those with activity in animal model systems, are awaited for further evaluation of KDM6B as a potential cancer target.
Figure 35.

Jumonji domain inhibitors. (a) chemical structures of JMJD3 inhibitor GSK-J1 and prodrug GSK-J4. (b) Crystal structure depicting the essential interactions of GSK-J1 with the iron atom (pink) in the JMJD3 active site (PDB: 4ASK). (c) Chemical structure of pan-KDM5 family inhibitor CPI-455.
4.3. KDM5/6 and drug-tolerant persistence
The KDM5 family of lysine demethylases, part of the larger JmjC-domain-containing class, play critical roles in gene regulation and chromatin remodeling. These enzymes, which demethylate H3K4, are associated with gene repression.1077 Overexpression and amplification of KDM5 proteins, particularly KDM5A and KDM5B, have been observed in a wide variety of cancers where they regulate the expression of both tumor suppressor genes and oncogenes.1078–1084 KDM5 and KDM6 family members are also implicated in maintaining drug-tolerant cell states.1085–1089 Drug-tolerant persister (DTP) cells represent a population of cancer cells that emerge following drug treatment and are characterized by a slow cell cycle and a reversible state of drug insensitivity. Unlike drug resistance driven by genetic mutations, DTP cells evade treatment through non-genetic mechanisms, such as chromatin rewiring and transcriptional changes. Notably, these cells often regain drug sensitivity and proliferative growth after treatment is removed, making them a key target for therapeutic intervention. Gene expression profiling of DTP cells has shown that the expression of KDM5A/B is elevated in these populations and that DTPs are dependent on KDM5 enzymes, specifically their enzymatic activity for maintaining the DTP state.1088 CPI-455, a selective pan-KDM5 family inhibitor that binds competitively with α-ketoglutarate, has shown the ability to prevent the survival of DTP cells (Figure 35c).1090 While more potent KDM5 inhibitors show activity in a variety of cancers, none of these have progressed to the clinic.1091–1096
4.4. GLP/G9a
G9a is a SET domain containing methyltransferase protein which specifically catalyzes the mono- and di-methylation of H3K9 (H3K9me1/2), modifications associated with transcriptional repression.1097 Aberrant H3K9 methylation has been observed in pathogenesis where it contributes to the silencing of tumor suppressors, such as p53.1098–1103 G9a and its paralog, GLP (G9a-like protein), are primarily responsible for the addition of this modification and G9a overexpression has been observed in many cancers and is correlated with a poor prognosis.1104 Knockdown of G9a reduced H3K9me2 at silenced loci leads to the re-expression of tumor suppressor genes, inhibition of cell proliferation and invasion, and supports G9a as a therapeutic target.1105,1106
There are two main classes of G9a/GLP inhibitors: SAM-competitive and substrate-competitive inhibitors. The most successful and specific compounds are those that fall into the substrate-competitive category. The first substrate inhibitor for GLP/G9a emerged from a high-throughput screen for inhibition of enzyme activity. This compound, BIX-01294, specifically inhibited the methyltransferase activity of GLP/G9a (IC50=1.7 μM and IC50=38 μM respectively) without activity on other methyltransferases (up to 45 μM).941,942 However, BIX-01294 is cytotoxic at concentrations > 4.1 μM limiting its use. SAR of BIX-01294 lead to the discovery of several compounds and the report of G9a-inhibitor co-crystal structures.943–946,1107 The first structure of G9a-UNC0224 validated compound binding and facilitated the discovery of subsequent structure-based inhibitors with greater potency, permeability, and selectivity, many of which possessed the quinazoline scaffold of BIX-01294.944 Further optimization of the substituents off the quinazoline scaffold lead to the development of UNC0642, an in vivo probe suitable for animal studies. UNC0642 had greater in vivo activity, potency (IC50 < 2.5nM), and selectivity (>2,000 fold) over other histone methyltransferases.1108 Additionally, expanding the quinazoline ring to a benzodiazepine led to a novel chemotype of compounds with similar activity as the previous scaffold.1109
Recently, molecular glue degraders for the transcription factor WIZ (dWIZ-1/2) were disclosed.1110 WIZ was revealed as a repressor of fetal hemoglobin via association with repressive complexes and GLP/G9a to facilitate methylation and gene repression.1111,1112 Degradation of WIZ reduced H3K9me2 and increased the expression of GLP/G9a gene targets. dWIZ-2 toxicity was assessed in >300 cell lines and was revealed to be significantly less cytotoxic than G9a/GLP inhibition with UNC0642.1110 This finding presents a novel approach for targeting proteins via disrupting or degrading G9a/GLP interactors.
5. Conclusion
By focusing on the potential for chromatin regulation to support the discovery of anti-cancer therapeutics, and by reviewing chromatin targets from the perspective of the genetic evidence that might influence their chances for therapeutic success, we have attempted to make for a manageable perspective on the field as it currently stands. Several reviews of exceptional quality have been published previously on related subjects in chromatin and drug discovery,1113,1114 including one published while this work was in development that is similarly organized around genetic rationale for target selection.1115 We sought a unique perspective in highlighting the evolution of chemistries that have enabled target prosecution and therapeutic translation. Still, we must sincerely apologize for any references missed within this work, especially from among the many biological studies that follow the discovery of a new chemical agent.
Assisted tremendously by the growing sophistication of chemical biology as a discipline, the study of chromatin biology has been advanced immeasurably by the pursuit of chemical tools. The discovery of HDACs was made possible by the study HDAC inhibitors, which provided a first concrete example of transcriptional control as a chromatin-dependent regulatory process. The first therapeutic hypotheses originated from these early studies, propelling HDAC inhibitors into the clinic where they continue to find use today. Buoyed by human genetics, functional genomics, and chemical biology, far more targets are now at our disposal, which will undoubtedly animate many more years of groundbreaking discoveries in chromatin biology, cancer pharmacology, and, hopefully, clinical development.
These new targets can take valuable lessons from the historical evidence supporting targets like HDACs and DNMTs, which revealed the therapeutic potential for targeting essential genes while also highlighting the challenges associated with their therapeutic windows. Target-based drug discovery has dominated chromatin drug discovery for many years now, but resources like DepMap have only become widely available in the last 10 years. While EZH2 and menin predate the existence of large-scale cancer dependency maps, the clinical successes for these cancer-specific vulnerabilities stand out as particularly hopeful signs for the richly informed targets of the future. We expect that ongoing clinical trials for targets like KAT6/7, LSD1, CBP/p300, and SWI/SNF, among others, will prove similarly instructive, giving new opportunities for us to revisit this document and evaluate—or reevaluate—the qualities that make for successful target selection.
As our synthesis of chromatin regulation and tumor biology continues to improve, we can expect continued challenges for discovery chemistry. Attractive targets like those formed by the paralog synthetic lethality between CBP/p300 and SMARCA2/4 have already provided formidable challenges for drug discovery, but new innovations have provided inroads to these otherwise intractable targets. Targeted protein degradation, which has great potential for achieving the selectivity needed to exploit paralog synthetic lethality, has only become a widespread field of study in the last 10 years. Importantly, there was no thought of being able to address paralog synthetic lethality with target protein degradation at its earliest inception,1116 highlighting the need to continue innovating new approaches in drug discovery and chemical biology. The emerging ability to rewire chromatin regulatory machinery is among the most exciting new possibilities, as it enables gain-of-function outcomes that are not possible with conventional inhibitors. Importantly, TCIPs, synTEFs, and related proximity agents enable small molecules that target common essential proteins to be repurposed as recruitment handles that redirect an essential protein for therapeutic benefit. This could allow for unprecedented precision in chromatin modulation and a new generation of therapeutic agents with unique considerations for target selection. We await these and other advances with great excitement.
Supplementary Material
Acknowledgements.
This work was supported by the Ono Pharma Foundation Breakthrough Science Initiative Awards Program, the Baxter Foundation Young Investigator Award, and the National Cancer Institute (R01CA280720) (M.A.E.). Order of authorship for equally contributing authors was determined by alphabetical order of surname.
Biographies
Paige Barta earned her B.A. in Biochemistry and Molecular Biology from Lewis & Clark College in 2020. After graduation, she worked as a research technician in Jared Rutter’s laboratory at the University of Utah before beginning her graduate studies at The Scripps Research Institute. She is currently pursuing her Ph.D. in the Erb Group, where she couples chemical probe perturbations with transcriptional genomics and forward genetics to study transcriptional control in cancer.
Trever Carter obtained his B.S. in Biochemistry from the University of Notre Dame in 2019. After a year of working with Prof. Craig Lindsley as a research assistant, he joined the Medical Scientist Training Program at the University of California – San Diego in 2020. Currently, he is pursuing his PhD in the Erb Group at The Scripps Research Institute, where he utilizes high-throughput chemistry toward the prospective discovery of molecular glues.
Michael Erb is an Associate Professor in the Department of Chemistry at The Scripps Research Institute. Before starting his independent career as a Scripps Fellow, he received his B.A. in Biochemistry from Claremont McKenna College (2014) and his PhD from Harvard University (2017) under the mentorship of Prof. James Bradner. The Erb laboratory combines expertise in transcriptional biology and discovery chemistry to drug cancer-associated transcriptional regulatory pathways. His research has been recognized and supported by an NIH Director’s Early Independence Award and the Ono Pharma Foundation Breakthrough Science Initiative Award.
References
- (1).ALLFREY VG; FAULKNER R; MIRSKY AE ACETYLATION AND METHYLATION OF HISTONES AND THEIR POSSIBLE ROLE IN THE REGULATION OF RNA SYNTHESIS. Proc Natl Acad Sci U S A 1964, 51 (5), 786–794. 10.1073/pnas.51.5.786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (2).Brownell JE; Zhou J; Ranalli T; Kobayashi R; Edmondson DG; Roth SY; Allis CD Tetrahymena Histone Acetyltransferase A: A Homolog to Yeast Gcn5p Linking Histone Acetylation to Gene Activation. Cell 1996, 84 (6), 843–851. 10.1016/s0092-8674(00)81063-6. [DOI] [PubMed] [Google Scholar]
- (3).Taunton J; Hassig CA; Schreiber SL A Mammalian Histone Deacetylase Related to the Yeast Transcriptional Regulator Rpd3p. Science 1996, 272 (5260), 408–411. 10.1126/science.272.5260.408. [DOI] [PubMed] [Google Scholar]
- (4).Mizzen CA; Yang XJ; Kokubo T; Brownell JE; Bannister AJ; Owen-Hughes T; Workman J; Wang L; Berger SL; Kouzarides T; Nakatani Y; Allis CD The TAF(II)250 Subunit of TFIID Has Histone Acetyltransferase Activity. Cell 1996, 87 (7), 1261–1270. 10.1016/s0092-8674(00)81821-8. [DOI] [PubMed] [Google Scholar]
- (5).Yang XJ; Ogryzko VV; Nishikawa J; Howard BH; Nakatani Y A P300/CBP-Associated Factor That Competes with the Adenoviral Oncoprotein E1A. Nature 1996, 382 (6589), 319–324. 10.1038/382319a0. [DOI] [PubMed] [Google Scholar]
- (6).Ogryzko VV; Schiltz RL; Russanova V; Howard BH; Nakatani Y The Transcriptional Coactivators P300 and CBP Are Histone Acetyltransferases. Cell 1996, 87 (5), 953–959. 10.1016/s0092-8674(00)82001-2. [DOI] [PubMed] [Google Scholar]
- (7).Bannister AJ; Kouzarides T The CBP Co-Activator Is a Histone Acetyltransferase. Nature 1996, 384 (6610), 641–643. 10.1038/384641a0. [DOI] [PubMed] [Google Scholar]
- (8).Luger K; Mäder AW; Richmond RK; Sargent DF; Richmond TJ Crystal Structure of the Nucleosome Core Particle at 2.8 A Resolution. Nature 1997, 389 (6648), 251–260. 10.1038/38444. [DOI] [PubMed] [Google Scholar]
- (9).Talbert PB; Henikoff S Histone Variants on the Move: Substrates for Chromatin Dynamics. Nat Rev Mol Cell Biol 2017, 18 (2), 115–126. 10.1038/nrm.2016.148. [DOI] [PubMed] [Google Scholar]
- (10).Schreiber SL; Bernstein BE Signaling Network Model of Chromatin. Cell 2002, 111 (6), 771–778. 10.1016/s0092-8674(02)01196-0. [DOI] [PubMed] [Google Scholar]
- (11).Rea S; Eisenhaber F; O’Carroll D; Strahl BD; Sun ZW; Schmid M; Opravil S; Mechtler K; Ponting CP; Allis CD; Jenuwein T Regulation of Chromatin Structure by Site-Specific Histone H3 Methyltransferases. Nature 2000, 406 (6796), 593–599. 10.1038/35020506. [DOI] [PubMed] [Google Scholar]
- (12).Strahl BD; Allis CD The Language of Covalent Histone Modifications. Nature 2000, 403 (6765), 41–45. 10.1038/47412. [DOI] [PubMed] [Google Scholar]
- (13).Jenuwein T; Allis CD Translating the Histone Code. Science 2001, 293 (5532), 1074–1080. 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
- (14).Sainsbury S; Bernecky C; Cramer P Structural Basis of Transcription Initiation by RNA Polymerase II. Nat Rev Mol Cell Biol 2015, 16 (3), 129–143. 10.1038/nrm3952. [DOI] [PubMed] [Google Scholar]
- (15).Jonkers I; Lis JT Getting up to Speed with Transcription Elongation by RNA Polymerase II. Nat Rev Mol Cell Biol 2015, 16 (3), 167–177. 10.1038/nrm3953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Guo J; Price DH RNA Polymerase II Transcription Elongation Control. Chem Rev 2013, 113 (11), 8583–8603. 10.1021/cr400105n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Armstrong JA Negotiating the Nucleosome: Factors That Allow RNA Polymerase II to Elongate through Chromatin. Biochem Cell Biol 2007, 85 (4), 426–434. 10.1139/O07-054. [DOI] [PubMed] [Google Scholar]
- (18).Tessarz P; Kouzarides T Histone Core Modifications Regulating Nucleosome Structure and Dynamics. Nat Rev Mol Cell Biol 2014, 15 (11), 703–708. 10.1038/nrm3890. [DOI] [PubMed] [Google Scholar]
- (19).Porrua O; Libri D Transcription Termination and the Control of the Transcriptome: Why, Where and How to Stop. Nat Rev Mol Cell Biol 2015, 16 (3), 190–202. 10.1038/nrm3943. [DOI] [PubMed] [Google Scholar]
- (20).Levine M; Cattoglio C; Tjian R Looping Back to Leap Forward: Transcription Enters a New Era. Cell 2014, 157 (1), 13–25. 10.1016/j.cell.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Heinz S; Romanoski CE; Benner C; Glass CK The Selection and Function of Cell Type-Specific Enhancers. Nat Rev Mol Cell Biol 2015, 16 (3), 144–154. 10.1038/nrm3949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Zaret KS Pioneer Transcription Factors Initiating Gene Network Changes. Annu Rev Genet 2020, 54, 367–385. 10.1146/annurev-genet-030220-015007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Balsalobre A; Drouin J Pioneer Factors as Master Regulators of the Epigenome and Cell Fate. Nat Rev Mol Cell Biol 2022, 23 (7), 449–464. 10.1038/s41580-022-00464-z. [DOI] [PubMed] [Google Scholar]
- (24).Bannister AJ; Kouzarides T Regulation of Chromatin by Histone Modifications. Cell Res 2011, 21 (3), 381–395. 10.1038/cr.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (25).Verdin E; Ott M 50 Years of Protein Acetylation: From Gene Regulation to Epigenetics, Metabolism and Beyond. Nat Rev Mol Cell Biol 2015, 16 (4), 258–264. 10.1038/nrm3931. [DOI] [PubMed] [Google Scholar]
- (26).Musselman CA; Lalonde M-E; Côté J; Kutateladze TG Perceiving the Epigenetic Landscape through Histone Readers. Nat Struct Mol Biol 2012, 19 (12), 1218–1227. 10.1038/nsmb.2436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Dhalluin C; Carlson JE; Zeng L; He C; Aggarwal AK; Zhou MM Structure and Ligand of a Histone Acetyltransferase Bromodomain. Nature 1999, 399 (6735), 491–496. 10.1038/20974. [DOI] [PubMed] [Google Scholar]
- (28).Li Y; Wen H; Xi Y; Tanaka K; Wang H; Peng D; Ren Y; Jin Q; Dent SYR; Li W; Li H; Shi X AF9 YEATS Domain Links Histone Acetylation to DOT1L-Mediated H3K79 Methylation. Cell 2014, 159 (3), 558–571. 10.1016/j.cell.2014.09.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (29).Lange M; Kaynak B; Forster UB; Tönjes M; Fischer JJ; Grimm C; Schlesinger J; Just S; Dunkel I; Krueger T; Mebus S; Lehrach H; Lurz R; Gobom J; Rottbauer W; Abdelilah-Seyfried S; Sperling S Regulation of Muscle Development by DPF3, a Novel Histone Acetylation and Methylation Reader of the BAF Chromatin Remodeling Complex. Genes Dev 2008, 22 (17), 2370–2384. 10.1101/gad.471408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (30).Zeng L; Zhang Q; Li S; Plotnikov AN; Walsh MJ; Zhou M-M Mechanism and Regulation of Acetylated Histone Binding by the Tandem PHD Finger of DPF3b. Nature 2010, 466 (7303), 258–262. 10.1038/nature09139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (31).Vezzoli A; Bonadies N; Allen MD; Freund SMV; Santiveri CM; Kvinlaug BT; Huntly BJP; Göttgens B; Bycroft M Molecular Basis of Histone H3K36me3 Recognition by the PWWP Domain of Brpf1. Nat Struct Mol Biol 2010, 17 (5), 617–619. 10.1038/nsmb.1797. [DOI] [PubMed] [Google Scholar]
- (32).Wang Y; Reddy B; Thompson J; Wang H; Noma K; Yates JR; Jia S Regulation of Set9-Mediated H4K20 Methylation by a PWWP Domain Protein. Mol Cell 2009, 33 (4), 428–437. 10.1016/j.molcel.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (33).Huang Y; Fang J; Bedford MT; Zhang Y; Xu R-M Recognition of Histone H3 Lysine-4 Methylation by the Double Tudor Domain of JMJD2A. Science 2006, 312 (5774), 748–751. 10.1126/science.1125162. [DOI] [PubMed] [Google Scholar]
- (34).Flanagan JF; Mi L-Z; Chruszcz M; Cymborowski M; Clines KL; Kim Y; Minor W; Rastinejad F; Khorasanizadeh S Double Chromodomains Cooperate to Recognize the Methylated Histone H3 Tail. Nature 2005, 438 (7071), 1181–1185. 10.1038/nature04290. [DOI] [PubMed] [Google Scholar]
- (35).Jacobs SA; Khorasanizadeh S Structure of HP1 Chromodomain Bound to a Lysine 9-Methylated Histone H3 Tail. Science 2002, 295 (5562), 2080–2083. 10.1126/science.1069473. [DOI] [PubMed] [Google Scholar]
- (36).Nielsen PR; Nietlispach D; Mott HR; Callaghan J; Bannister A; Kouzarides T; Murzin AG; Murzina NV; Laue ED Structure of the HP1 Chromodomain Bound to Histone H3 Methylated at Lysine 9. Nature 2002, 416 (6876), 103–107. 10.1038/nature722. [DOI] [PubMed] [Google Scholar]
- (37).Bannister AJ; Zegerman P; Partridge JF; Miska EA; Thomas JO; Allshire RC; Kouzarides T Selective Recognition of Methylated Lysine 9 on Histone H3 by the HP1 Chromo Domain. Nature 2001, 410 (6824), 120–124. 10.1038/35065138. [DOI] [PubMed] [Google Scholar]
- (38).Wysocka J; Swigut T; Milne TA; Dou Y; Zhang X; Burlingame AL; Roeder RG; Brivanlou AH; Allis CD WDR5 Associates with Histone H3 Methylated at K4 and Is Essential for H3 K4 Methylation and Vertebrate Development. Cell 2005, 121 (6), 859–872. 10.1016/j.cell.2005.03.036. [DOI] [PubMed] [Google Scholar]
- (39).Botuyan MV; Lee J; Ward IM; Kim J-E; Thompson JR; Chen J; Mer G Structural Basis for the Methylation State-Specific Recognition of Histone H4-K20 by 53BP1 and Crb2 in DNA Repair. Cell 2006, 127 (7), 1361–1373. 10.1016/j.cell.2006.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (40).Engelberg IA; Foley CA; James LI; Frye SV Improved Methods for Targeting Epigenetic Reader Domains of Acetylated and Methylated Lysine. Curr Opin Chem Biol 2021, 63, 132–144. 10.1016/j.cbpa.2021.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (41).Marks PA; Breslow R Dimethyl Sulfoxide to Vorinostat: Development of This Histone Deacetylase Inhibitor as an Anticancer Drug. Nat Biotechnol 2007, 25 (1), 84–90. 10.1038/nbt1272. [DOI] [PubMed] [Google Scholar]
- (42).Millard CJ; Watson PJ; Fairall L; Schwabe JWR Targeting Class I Histone Deacetylases in a “Complex” Environment. Trends Pharmacol Sci 2017, 38 (4), 363–377. 10.1016/j.tips.2016.12.006. [DOI] [PubMed] [Google Scholar]
- (43).Kijima M; Yoshida M; Sugita K; Horinouchi S; Beppu T Trapoxin, an Antitumor Cyclic Tetrapeptide, Is an Irreversible Inhibitor of Mammalian Histone Deacetylase. Journal of Biological Chemistry 1993, 268 (30), 22429–22435. 10.1016/S0021-9258(18)41547-5. [DOI] [PubMed] [Google Scholar]
- (44).Itazaki H; Nagashima K; Sugita K; Yoshida H; Kawamura Y; Yasuda Y; Matsumoto K; Ishii K; Uotani N; Nakai H Isolation and Structural Elucidation of New Cyclotetrapeptides, Trapoxins A and B, Having Detransformation Activities as Antitumor Agents. J Antibiot (Tokyo) 1990, 43 (12), 1524–1532. 10.7164/antibiotics.43.1524. [DOI] [PubMed] [Google Scholar]
- (45).Yoshida H; Sugita K A Novel Tetracyclic Peptide, Trapoxin, Induces Phenotypic Change from Transformed to Normal in Sis-Oncogene-Transformed NIH3T3 Cells. Jpn J Cancer Res 1992, 83 (4), 324–328. 10.1111/j.1349-7006.1992.tb00109.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (46).Yoshida M; Kijima M; Akita M; Beppu T Potent and Specific Inhibition of Mammalian Histone Deacetylase Both in Vivo and in Vitro by Trichostatin A. Journal of Biological Chemistry 1990, 265 (28), 17174–17179. 10.1016/S0021-9258(17)44885-X. [DOI] [PubMed] [Google Scholar]
- (47).Yoshida M; Nomura S; Beppu T Effects of Trichostatins on Differentiation of Murine Erythroleukemia Cells. Cancer Res 1987, 47 (14), 3688–3691. [PubMed] [Google Scholar]
- (48).Chang L; Ruiz P; Ito T; Sellers WR Targeting Pan-Essential Genes in Cancer: Challenges and Opportunities. Cancer Cell 2021, 39 (4), 466–479. 10.1016/j.ccell.2020.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (49).Luo J; Solimini NL; Elledge SJ Principles of Cancer Therapy: Oncogene and Non-Oncogene Addiction. Cell 2009, 136 (5), 823–837. 10.1016/j.cell.2009.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (50).Meyers RM; Bryan JG; McFarland JM; Weir BA; Sizemore AE; Xu H; Dharia NV; Montgomery PG; Cowley GS; Pantel S; Goodale A; Lee Y; Ali LD; Jiang G; Lubonja R; Harrington WF; Strickland M; Wu T; Hawes DC; Zhivich VA; Wyatt MR; Kalani Z; Chang JJ; Okamoto M; Stegmaier K; Golub TR; Boehm JS; Vazquez F; Root DE; Hahn WC; Tsherniak A Computational Correction of Copy Number Effect Improves Specificity of CRISPR-Cas9 Essentiality Screens in Cancer Cells. Nat Genet 2017, 49 (12), 1779–1784. 10.1038/ng.3984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (51).Tsherniak A; Vazquez F; Montgomery PG; Weir BA; Kryukov G; Cowley GS; Gill S; Harrington WF; Pantel S; Krill-Burger JM; Meyers RM; Ali L; Goodale A; Lee Y; Jiang G; Hsiao J; Gerath WFJ; Howell S; Merkel E; Ghandi M; Garraway LA; Root DE; Golub TR; Boehm JS; Hahn WC Defining a Cancer Dependency Map. Cell 2017, 170 (3), 564–576.e16. 10.1016/j.cell.2017.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (52).Dempster JM; Pacini C; Pantel S; Behan FM; Green T; Krill-Burger J; Beaver CM; Younger ST; Zhivich V; Najgebauer H; Allen F; Gonçalves E; Shepherd R; Doench JG; Yusa K; Vazquez F; Parts L; Boehm JS; Golub TR; Hahn WC; Root DE; Garnett MJ; Tsherniak A; Iorio F Agreement between Two Large Pan-Cancer CRISPR-Cas9 Gene Dependency Data Sets. Nat Commun 2019, 10 (1), 5817. 10.1038/s41467-019-13805-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (53).Kim KH; Roberts CWM Targeting EZH2 in Cancer. Nat Med 2016, 22 (2), 128–134. 10.1038/nm.4036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (54).Grembecka J; He S; Shi A; Purohit T; Muntean AG; Sorenson RJ; Showalter HD; Murai MJ; Belcher AM; Hartley T; Hess JL; Cierpicki T Menin-MLL Inhibitors Reverse Oncogenic Activity of MLL Fusion Proteins in Leukemia. Nat Chem Biol 2012, 8 (3), 277–284. 10.1038/nchembio.773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (55).Kempinska K; Malik B; Borkin D; Klossowski S; Shukla S; Miao H; Wang J; Cierpicki T; Grembecka J Pharmacologic Inhibition of the Menin-MLL Interaction Leads to Transcriptional Repression of PEG10 and Blocks Hepatocellular Carcinoma. Mol Cancer Ther 2018, 17 (1), 26–38. 10.1158/1535-7163.MCT-17-0580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (56).Klossowski S; Miao H; Kempinska K; Wu T; Purohit T; Kim E; Linhares BM; Chen D; Jih G; Perkey E; Huang H; He M; Wen B; Wang Y; Yu K; Lee SC-W; Danet-Desnoyers G; Trotman W; Kandarpa M; Cotton A; Abdel-Wahab O; Lei H; Dou Y; Guzman M; Peterson L; Gruber T; Choi S; Sun D; Ren P; Li L-S; Liu Y; Burrows F; Maillard I; Cierpicki T; Grembecka J Menin Inhibitor MI-3454 Induces Remission in MLL1-Rearranged and NPM1-Mutated Models of Leukemia. J Clin Invest 2020, 130 (2), 981–997. 10.1172/JCI129126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (57).Krivtsov AV; Evans K; Gadrey JY; Eschle BK; Hatton C; Uckelmann HJ; Ross KN; Perner F; Olsen SN; Pritchard T; McDermott L; Jones CD; Jing D; Braytee A; Chacon D; Earley E; McKeever BM; Claremon D; Gifford AJ; Lee HJ; Teicher BA; Pimanda JE; Beck D; Perry JA; Smith MA; McGeehan GM; Lock RB; Armstrong SA A Menin-MLL Inhibitor Induces Specific Chromatin Changes and Eradicates Disease in Models of MLL-Rearranged Leukemia. Cancer Cell 2019, 36 (6), 660–673.e11. 10.1016/j.ccell.2019.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (58).Uckelmann HJ; Kim SM; Wong EM; Hatton C; Giovinazzo H; Gadrey JY; Krivtsov AV; Rücker FG; Döhner K; McGeehan GM; Levine RL; Bullinger L; Vassiliou GS; Armstrong SA Therapeutic Targeting of Preleukemia Cells in a Mouse Model of NPM1 Mutant Acute Myeloid Leukemia. Science 2020, 367 (6477), 586–590. 10.1126/science.aax5863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (59).Perner F; Stein EM; Wenge DV; Singh S; Kim J; Apazidis A; Rahnamoun H; Anand D; Marinaccio C; Hatton C; Wen Y; Stone RM; Schaller D; Mowla S; Xiao W; Gamlen HA; Stonestrom AJ; Persaud S; Ener E; Cutler JA; Doench JG; McGeehan GM; Volkamer A; Chodera JD; Nowak RP; Fischer ES; Levine RL; Armstrong SA; Cai SF MEN1 Mutations Mediate Clinical Resistance to Menin Inhibition. Nature 2023, 615 (7954), 913–919. 10.1038/s41586-023-05755-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (60).Issa GC; Aldoss I; DiPersio J; Cuglievan B; Stone R; Arellano M; Thirman MJ; Patel MR; Dickens DS; Shenoy S; Shukla N; Kantarjian H; Armstrong SA; Perner F; Perry JA; Rosen G; Bagley RG; Meyers ML; Ordentlich P; Gu Y; Kumar V; Smith S; McGeehan GM; Stein EM The Menin Inhibitor Revumenib in KMT2A-Rearranged or NPM1-Mutant Leukaemia. Nature 2023, 615 (7954), 920–924. 10.1038/s41586-023-05812-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (61).Falkenberg KJ; Johnstone RW Histone Deacetylases and Their Inhibitors in Cancer, Neurological Diseases and Immune Disorders. Nat Rev Drug Discov 2014, 13 (9), 673–691. 10.1038/nrd4360. [DOI] [PubMed] [Google Scholar]
- (62).Friend C; Scher W; Holland JG; Sato T Hemoglobin Synthesis in Murine Virus-Induced Leukemic Cells in Vitro: Stimulation of Erythroid Differentiation by Dimethyl Sulfoxide. Proc Natl Acad Sci U S A 1971, 68 (2), 378–382. 10.1073/pnas.68.2.378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (63).Tanaka M; Levy J; Terada M; Breslow R; Rifkind RA; Marks PA Induction of Erythroid Differentiation in Murine Virus Infected Eythroleukemia Cells by Highly Polar Compounds. Proc Natl Acad Sci U S A 1975, 72 (3), 1003–1006. 10.1073/pnas.72.3.1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (64).Richon VM; Webb Y; Merger R; Sheppard T; Jursic B; Ngo L; Civoli F; Breslow R; Rifkind RA; Marks PA Second Generation Hybrid Polar Compounds Are Potent Inducers of Transformed Cell Differentiation. Proc Natl Acad Sci U S A 1996, 93 (12), 5705–5708. 10.1073/pnas.93.12.5705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (65).Richon VM; Emiliani S; Verdin E; Webb Y; Breslow R; Rifkind RA; Marks PA A Class of Hybrid Polar Inducers of Transformed Cell Differentiation Inhibits Histone Deacetylases. Proc Natl Acad Sci U S A 1998, 95 (6), 3003–3007. 10.1073/pnas.95.6.3003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (66).Finnin MS; Donigian JR; Cohen A; Richon VM; Rifkind RA; Marks PA; Breslow R; Pavletich NP Structures of a Histone Deacetylase Homologue Bound to the TSA and SAHA Inhibitors. Nature 1999, 401 (6749), 188–193. 10.1038/43710. [DOI] [PubMed] [Google Scholar]
- (67).Olsen EA; Kim YH; Kuzel TM; Pacheco TR; Foss FM; Parker S; Frankel SR; Chen C; Ricker JL; Arduino JM; Duvic M Phase IIb Multicenter Trial of Vorinostat in Patients with Persistent, Progressive, or Treatment Refractory Cutaneous T-Cell Lymphoma. J Clin Oncol 2007, 25 (21), 3109–3115. 10.1200/JCO.2006.10.2434. [DOI] [PubMed] [Google Scholar]
- (68).Nakajima H; Kim YB; Terano H; Yoshida M; Horinouchi S FR901228, a Potent Antitumor Antibiotic, Is a Novel Histone Deacetylase Inhibitor. Exp Cell Res 1998, 241 (1), 126–133. 10.1006/excr.1998.4027. [DOI] [PubMed] [Google Scholar]
- (69).Furumai R; Matsuyama A; Kobashi N; Lee K-H; Nishiyama M; Nakajima H; Tanaka A; Komatsu Y; Nishino N; Yoshida M; Horinouchi S FK228 (Depsipeptide) as a Natural Prodrug That Inhibits Class I Histone Deacetylases. Cancer Res 2002, 62 (17), 4916–4921. [PubMed] [Google Scholar]
- (70).Witter DJ; Harrington P; Wilson KJ; Chenard M; Fleming JC; Haines B; Kral AM; Secrist JP; Miller TA Optimization of Biaryl Selective HDAC1&2 Inhibitors (SHI-1:2). Bioorg Med Chem Lett 2008, 18 (2), 726–731. 10.1016/j.bmcl.2007.11.047. [DOI] [PubMed] [Google Scholar]
- (71).Methot JL; Chakravarty PK; Chenard M; Close J; Cruz JC; Dahlberg WK; Fleming J; Hamblett CL; Hamill JE; Harrington P; Harsch A; Heidebrecht R; Hughes B; Jung J; Kenific CM; Kral AM; Meinke PT; Middleton RE; Ozerova N; Sloman DL; Stanton MG; Szewczak AA; Tyagarajan S; Witter DJ; Secrist JP; Miller TA Exploration of the Internal Cavity of Histone Deacetylase (HDAC) with Selective HDAC1/HDAC2 Inhibitors (SHI-1:2). Bioorg Med Chem Lett 2008, 18 (3), 973–978. 10.1016/j.bmcl.2007.12.031. [DOI] [PubMed] [Google Scholar]
- (72).Bressi JC; Jennings AJ; Skene R; Wu Y; Melkus R; De Jong R; O’Connell S; Grimshaw CE; Navre M; Gangloff AR Exploration of the HDAC2 Foot Pocket: Synthesis and SAR of Substituted N-(2-Aminophenyl)Benzamides. Bioorg Med Chem Lett 2010, 20 (10), 3142–3145. 10.1016/j.bmcl.2010.03.091. [DOI] [PubMed] [Google Scholar]
- (73).Montero A; Beierle JM; Olsen CA; Ghadiri MR Design, Synthesis, Biological Evaluation, and Structural Characterization of Potent Histone Deacetylase Inhibitors Based on Cyclic Alpha/Beta-Tetrapeptide Architectures. J Am Chem Soc 2009, 131 (8), 3033–3041. 10.1021/ja809508f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (74).Olsen CA; Ghadiri MR Discovery of Potent and Selective Histone Deacetylase Inhibitors via Focused Combinatorial Libraries of Cyclic Alpha3beta-Tetrapeptides. J Med Chem 2009, 52 (23), 7836–7846. 10.1021/jm900850t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (75).Vickers CJ; Olsen CA; Leman LJ; Ghadiri MR Discovery of HDAC Inhibitors That Lack an Active Site Zn(2+)-Binding Functional Group. ACS Med Chem Lett 2012, 3 (6), 505–508. 10.1021/ml300081u. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (76).Daniel KB; Sullivan ED; Chen Y; Chan JC; Jennings PA; Fierke CA; Cohen SM Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release. J Med Chem 2015, 58 (11), 4812–4821. 10.1021/acs.jmedchem.5b00539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (77).Keeley AB; Kopranovic A; Di Lorenzo V; Ábrányi-Balogh P; Jänsch N; Lai LN; Petri L; Orgován Z; Pölöske D; Orlova A; Németh AG; Desczyk C; Imre T; Bajusz D; Moriggl R; Meyer-Almes F-J; Keserü GM Electrophilic MiniFrags Revealed Unprecedented Binding Sites for Covalent HDAC8 Inhibitors. J Med Chem 2024, 67 (1), 572–585. 10.1021/acs.jmedchem.3c01779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (78).Raouf YS; Sedighi A; Geletu M; Frere GA; Allan RG; Nawar N; de Araujo ED; Gunning PT Discovery of YSR734: A Covalent HDAC Inhibitor with Cellular Activity in Acute Myeloid Leukemia and Duchenne Muscular Dystrophy. J Med Chem 2023, 66 (24), 16658–16679. 10.1021/acs.jmedchem.3c01236. [DOI] [PubMed] [Google Scholar]
- (79).Raouf YS; Moreno-Yruela C Slow-Binding and Covalent HDAC Inhibition: A New Paradigm? JACS Au 2024, 4 (11), 4148–4161. 10.1021/jacsau.4c00828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (80).el-Beltagi HM; Martens AC; Dahab GM; Hagenbeek A Efficacy of Acetyldinaline for Treatment of Minimal Residual Disease (MRD): Preclinical Studies in the BNML Rat Model for Human Acute Myelocytic Leukemia. Leukemia 1993, 7 (11), 1795–1800. [PubMed] [Google Scholar]
- (81).el-Beltagi HM; Martens AC; Lelieveld P; Haroun EA; Hagenbeek A Acetyldinaline: A New Oral Cytostatic Drug with Impressive Differential Activity against Leukemic Cells and Normal Stem Cells--Preclinical Studies in a Relevant Rat Model for Human Acute Myelocytic Leukemia. Cancer Res 1993, 53 (13), 3008–3014. [PubMed] [Google Scholar]
- (82).LoRusso PM; Demchik L; Foster B; Knight J; Bissery MC; Polin LM; Leopold WR; Corbett TH Preclinical Antitumor Activity of CI-994. Invest New Drugs 1996, 14 (4), 349–356. 10.1007/BF00180810. [DOI] [PubMed] [Google Scholar]
- (83).Saito A; Yamashita T; Mariko Y; Nosaka Y; Tsuchiya K; Ando T; Suzuki T; Tsuruo T; Nakanishi O A Synthetic Inhibitor of Histone Deacetylase, MS-27–275, with Marked in Vivo Antitumor Activity against Human Tumors. Proc Natl Acad Sci U S A 1999, 96 (8), 4592–4597. 10.1073/pnas.96.8.4592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (84).Gregoretti IV; Lee Y-M; Goodson HV Molecular Evolution of the Histone Deacetylase Family: Functional Implications of Phylogenetic Analysis. J Mol Biol 2004, 338 (1), 17–31. 10.1016/j.jmb.2004.02.006. [DOI] [PubMed] [Google Scholar]
- (85).Bradner JE; West N; Grachan ML; Greenberg EF; Haggarty SJ; Warnow T; Mazitschek R Chemical Phylogenetics of Histone Deacetylases. Nat Chem Biol 2010, 6 (3), 238–243. 10.1038/nchembio.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (86).Hubbert C; Guardiola A; Shao R; Kawaguchi Y; Ito A; Nixon A; Yoshida M; Wang X-F; Yao T-P HDAC6 Is a Microtubule-Associated Deacetylase. Nature 2002, 417 (6887), 455–458. 10.1038/417455a. [DOI] [PubMed] [Google Scholar]
- (87).Bachy E; Camus V; Thieblemont C; Casasnovas R-O; Ysebaert L; Damaj GL; Guidez S; Pica G-M; Kim WS; Lim ST; André M; Martin Garcia-Sancho A; Penarrubia Ponce MJ; Staber PB; Trotman J; Hüttmann A; Stefoni V; Re A; Gaulard P; Delfau-Larue M-H; De Leval L; Meignan M; Li J; Morschhauser F; Delarue R Final Analysis of the Ro-CHOP Phase III Study (Conducted by LYSA): Romidepsin Plus CHOP in Patients with Peripheral T-Cell Lymphoma. Blood 2020, 136 (Supplement 1), 32–33. 10.1182/blood-2020-134440. [DOI] [Google Scholar]
- (88).Whittaker SJ; Demierre M-F; Kim EJ; Rook AH; Lerner A; Duvic M; Scarisbrick J; Reddy S; Robak T; Becker JC; Samtsov A; McCulloch W; Kim YH Final Results from a Multicenter, International, Pivotal Study of Romidepsin in Refractory Cutaneous T-Cell Lymphoma. J Clin Oncol 2010, 28 (29), 4485–4491. 10.1200/JCO.2010.28.9066. [DOI] [PubMed] [Google Scholar]
- (89).Coiffier B; Pro B; Prince HM; Foss F; Sokol L; Greenwood M; Caballero D; Borchmann P; Morschhauser F; Wilhelm M; Pinter-Brown L; Padmanabhan S; Shustov A; Nichols J; Carroll S; Balser J; Balser B; Horwitz S Results from a Pivotal, Open-Label, Phase II Study of Romidepsin in Relapsed or Refractory Peripheral T-Cell Lymphoma after Prior Systemic Therapy. J Clin Oncol 2012, 30 (6), 631–636. 10.1200/JCO.2011.37.4223. [DOI] [PubMed] [Google Scholar]
- (90).O’Connor OA; Horwitz S; Masszi T; Van Hoof A; Brown P; Doorduijn J; Hess G; Jurczak W; Knoblauch P; Chawla S; Bhat G; Choi MR; Walewski J; Savage K; Foss F; Allen LF; Shustov A Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results of the Pivotal Phase II BELIEF (CLN-19) Study. J Clin Oncol 2015, 33 (23), 2492–2499. 10.1200/JCO.2014.59.2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (91).San-Miguel JF; Hungria VTM; Yoon S-S; Beksac M; Dimopoulos MA; Elghandour A; Jedrzejczak WW; Günther A; Nakorn TN; Siritanaratkul N; Corradini P; Chuncharunee S; Lee J-J; Schlossman RL; Shelekhova T; Yong K; Tan D; Numbenjapon T; Cavenagh JD; Hou J; LeBlanc R; Nahi H; Qiu L; Salwender H; Pulini S; Moreau P; Warzocha K; White D; Bladé J; Chen W; de la Rubia J; Gimsing P; Lonial S; Kaufman JL; Ocio EM; Veskovski L; Sohn SK; Wang M-C; Lee JH; Einsele H; Sopala M; Corrado C; Bengoudifa B-R; Binlich F; Richardson PG Panobinostat plus Bortezomib and Dexamethasone versus Placebo plus Bortezomib and Dexamethasone in Patients with Relapsed or Relapsed and Refractory Multiple Myeloma: A Multicentre, Randomised, Double-Blind Phase 3 Trial. Lancet Oncol 2014, 15 (11), 1195–1206. 10.1016/S1470-2045(14)70440-1. [DOI] [PubMed] [Google Scholar]
- (92).Yang X-J; Seto E The Rpd3/Hda1 Family of Lysine Deacetylases: From Bacteria and Yeast to Mice and Men. Nat Rev Mol Cell Biol 2008, 9 (3), 206–218. 10.1038/nrm2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (93).Zhang Y; Remillard D; Onubogu U; Karakyriakou B; Asiaban JN; Ramos AR; Bowland K; Bishop TR; Barta PA; Nance S; Durbin AD; Ott CJ; Janiszewska M; Cravatt BF; Erb MA Collateral Lethality between HDAC1 and HDAC2 Exploits Cancer-Specific NuRD Complex Vulnerabilities. Nat Struct Mol Biol 2023, 30 (8), 1160–1171. 10.1038/s41594-023-01041-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (94).Montgomery RL; Potthoff MJ; Haberland M; Qi X; Matsuzaki S; Humphries KM; Richardson JA; Bassel-Duby R; Olson EN Maintenance of Cardiac Energy Metabolism by Histone Deacetylase 3 in Mice. J Clin Invest 2008, 118 (11), 3588–3597. 10.1172/JCI35847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (95).Knutson SK; Chyla BJ; Amann JM; Bhaskara S; Huppert SS; Hiebert SW Liver-Specific Deletion of Histone Deacetylase 3 Disrupts Metabolic Transcriptional Networks. EMBO J 2008, 27 (7), 1017–1028. 10.1038/emboj.2008.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (96).Bhaskara S; Chyla BJ; Amann JM; Knutson SK; Cortez D; Sun Z-W; Hiebert SW Deletion of Histone Deacetylase 3 Reveals Critical Roles in S Phase Progression and DNA Damage Control. Mol Cell 2008, 30 (1), 61–72. 10.1016/j.molcel.2008.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (97).Payne NC; Maksoud S; Tannous BA; Mazitschek R A Direct High-Throughput Protein Quantification Strategy Facilitates Discovery and Characterization of a Celastrol-Derived BRD4 Degrader. Cell Chem Biol 2022, 29 (8), 1333–1340.e5. 10.1016/j.chembiol.2022.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (98).Payne NC; Mazitschek R Resolving the Deceptive Isoform and Complex Selectivity of HDAC1/2 Inhibitors. Cell Chem Biol 2022, 29 (7), 1140–1152.e5. 10.1016/j.chembiol.2022.03.002. [DOI] [PubMed] [Google Scholar]
- (99).Chou CJ; Herman D; Gottesfeld JM Pimelic Diphenylamide 106 Is a Slow, Tight-Binding Inhibitor of Class I Histone Deacetylases. J Biol Chem 2008, 283 (51), 35402–35409. 10.1074/jbc.M807045200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (100).Bantscheff M; Hopf C; Savitski MM; Dittmann A; Grandi P; Michon A-M; Schlegl J; Abraham Y; Becher I; Bergamini G; Boesche M; Delling M; Dümpelfeld B; Eberhard D; Huthmacher C; Mathieson T; Poeckel D; Reader V; Strunk K; Sweetman G; Kruse U; Neubauer G; Ramsden NG; Drewes G Chemoproteomics Profiling of HDAC Inhibitors Reveals Selective Targeting of HDAC Complexes. Nat Biotechnol 2011, 29 (3), 255–265. 10.1038/nbt.1759. [DOI] [PubMed] [Google Scholar]
- (101).Becher I; Dittmann A; Savitski MM; Hopf C; Drewes G; Bantscheff M Chemoproteomics Reveals Time-Dependent Binding of Histone Deacetylase Inhibitors to Endogenous Repressor Complexes. ACS Chem Biol 2014, 9 (8), 1736–1746. 10.1021/cb500235n. [DOI] [PubMed] [Google Scholar]
- (102).Moreno-Yruela C; Olsen CA Determination of Slow-Binding HDAC Inhibitor Potency and Subclass Selectivity. ACS Med Chem Lett 2022, 13 (5), 779–785. 10.1021/acsmedchemlett.1c00702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (103).Malvaez M; McQuown SC; Rogge GA; Astarabadi M; Jacques V; Carreiro S; Rusche JR; Wood MA HDAC3-Selective Inhibitor Enhances Extinction of Cocaine-Seeking Behavior in a Persistent Manner. Proc Natl Acad Sci U S A 2013, 110 (7), 2647–2652. 10.1073/pnas.1213364110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (104).Lagger G; O’Carroll D; Rembold M; Khier H; Tischler J; Weitzer G; Schuettengruber B; Hauser C; Brunmeir R; Jenuwein T; Seiser C Essential Function of Histone Deacetylase 1 in Proliferation Control and CDK Inhibitor Repression. EMBO J 2002, 21 (11), 2672–2681. 10.1093/emboj/21.11.2672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (105).Jamaladdin S; Kelly RDW; O’Regan L; Dovey OM; Hodson GE; Millard CJ; Portolano N; Fry AM; Schwabe JWR; Cowley SM Histone Deacetylase (HDAC) 1 and 2 Are Essential for Accurate Cell Division and the Pluripotency of Embryonic Stem Cells. Proc Natl Acad Sci U S A 2014, 111 (27), 9840–9845. 10.1073/pnas.1321330111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (106).Yamaguchi T; Cubizolles F; Zhang Y; Reichert N; Kohler H; Seiser C; Matthias P Histone Deacetylases 1 and 2 Act in Concert to Promote the G1-to-S Progression. Genes Dev 2010, 24 (5), 455–469. 10.1101/gad.552310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (107).Wilting RH; Yanover E; Heideman MR; Jacobs H; Horner J; van der Torre J; DePinho RA; Dannenberg J-H Overlapping Functions of Hdac1 and Hdac2 in Cell Cycle Regulation and Haematopoiesis. EMBO J 2010, 29 (15), 2586–2597. 10.1038/emboj.2010.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (108).LeBoeuf M; Terrell A; Trivedi S; Sinha S; Epstein JA; Olson EN; Morrisey EE; Millar SE Hdac1 and Hdac2 Act Redundantly to Control P63 and P53 Functions in Epidermal Progenitor Cells. Dev Cell 2010, 19 (6), 807–818. 10.1016/j.devcel.2010.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (109).Heideman MR; Wilting RH; Yanover E; Velds A; de Jong J; Kerkhoven RM; Jacobs H; Wessels LF; Dannenberg J-H Dosage-Dependent Tumor Suppression by Histone Deacetylases 1 and 2 through Regulation of c-Myc Collaborating Genes and P53 Function. Blood 2013, 121 (11), 2038–2050. 10.1182/blood-2012-08-450916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (110).Dovey OM; Foster CT; Conte N; Edwards SA; Edwards JM; Singh R; Vassiliou G; Bradley A; Cowley SM Histone Deacetylase 1 and 2 Are Essential for Normal T-Cell Development and Genomic Stability in Mice. Blood 2013, 121 (8), 1335–1344. 10.1182/blood-2012-07-441949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (111).Matthews GM; Mehdipour P; Cluse LA; Falkenberg KJ; Wang E; Roth M; Santoro F; Vidacs E; Stanley K; House CM; Rusche JR; Vakoc CR; Zuber J; Minucci S; Johnstone RW Functional-Genetic Dissection of HDAC Dependencies in Mouse Lymphoid and Myeloid Malignancies. Blood 2015, 126 (21), 2392–2403. 10.1182/blood-2015-03-632984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (112).Stubbs MC; Kim W; Bariteau M; Davis T; Vempati S; Minehart J; Witkin M; Qi J; Krivtsov AV; Bradner JE; Kung AL; Armstrong SA Selective Inhibition of HDAC1 and HDAC2 as a Potential Therapeutic Option for B-ALL. Clin Cancer Res 2015, 21 (10), 2348–2358. 10.1158/1078-0432.CCR-14-1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (113).Frumm SM; Fan ZP; Ross KN; Duvall JR; Gupta S; VerPlank L; Suh B-C; Holson E; Wagner FF; Smith WB; Paranal RM; Bassil CF; Qi J; Roti G; Kung AL; Bradner JE; Tolliday N; Stegmaier K Selective HDAC1/HDAC2 Inhibitors Induce Neuroblastoma Differentiation. Chem Biol 2013, 20 (5), 713–725. 10.1016/j.chembiol.2013.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (114).Ito T; Young MJ; Li R; Jain S; Wernitznig A; Krill-Burger JM; Lemke CT; Monducci D; Rodriguez DJ; Chang L; Dutta S; Pal D; Paolella BR; Rothberg MV; Root DE; Johannessen CM; Parida L; Getz G; Vazquez F; Doench JG; Zamanighomi M; Sellers WR Paralog Knockout Profiling Identifies DUSP4 and DUSP6 as a Digenic Dependence in MAPK Pathway-Driven Cancers. Nat Genet 2021, 53 (12), 1664–1672. 10.1038/s41588-021-00967-z. [DOI] [PubMed] [Google Scholar]
- (115).DeWeirdt PC; Sanson KR; Sangree AK; Hegde M; Hanna RE; Feeley MN; Griffith AL; Teng T; Borys SM; Strand C; Joung JK; Kleinstiver BP; Pan X; Huang A; Doench JG Optimization of AsCas12a for Combinatorial Genetic Screens in Human Cells. Nat Biotechnol 2021, 39 (1), 94–104. 10.1038/s41587-020-0600-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (116).Huang A; Garraway LA; Ashworth A; Weber B Synthetic Lethality as an Engine for Cancer Drug Target Discovery. Nat Rev Drug Discov 2020, 19 (1), 23–38. 10.1038/s41573-019-0046-z. [DOI] [PubMed] [Google Scholar]
- (117).Smalley JP; Adams GE; Millard CJ; Song Y; Norris JKS; Schwabe JWR; Cowley SM; Hodgkinson JT PROTAC-Mediated Degradation of Class I Histone Deacetylase Enzymes in Corepressor Complexes. Chem Commun (Camb) 2020, 56 (32), 4476–4479. 10.1039/d0cc01485k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (118).Xiong Y; Donovan KA; Eleuteri NA; Kirmani N; Yue H; Razov A; Krupnick NM; Nowak RP; Fischer ES Chemo-Proteomics Exploration of HDAC Degradability by Small Molecule Degraders. Cell Chem Biol 2021, 28 (10), 1514–1527.e4. 10.1016/j.chembiol.2021.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (119).Smalley JP; Baker IM; Pytel WA; Lin L-Y; Bowman KJ; Schwabe JWR; Cowley SM; Hodgkinson JT Optimization of Class I Histone Deacetylase PROTACs Reveals That HDAC1/2 Degradation Is Critical to Induce Apoptosis and Cell Arrest in Cancer Cells. J Med Chem 2022, 65 (7), 5642–5659. 10.1021/acs.jmedchem.1c02179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (120).Toriki ES; Papatzimas JW; Nishikawa K; Dovala D; Frank AO; Hesse MJ; Dankova D; Song J-G; Bruce-Smythe M; Struble H; Garcia FJ; Brittain SM; Kile AC; McGregor LM; McKenna JM; Tallarico JA; Schirle M; Nomura DK Rational Chemical Design of Molecular Glue Degraders. ACS Cent Sci 2023, 9 (5), 915–926. 10.1021/acscentsci.2c01317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (121).Cross JM; Coulson ME; Smalley JP; Pytel WA; Ismail O; Trory JS; Cowley SM; Hodgkinson JT A “click” Chemistry Approach to Novel Entinostat (MS-275) Based Class I Histone Deacetylase Proteolysis Targeting Chimeras. RSC Med Chem 2022, 13 (12), 1634–1639. 10.1039/d2md00199c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (122).Patel U; Smalley JP; Hodgkinson JT PROTAC Chemical Probes for Histone Deacetylase Enzymes. RSC Chem Biol 2023, 4 (9), 623–634. 10.1039/d3cb00105a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (123).Smalley JP; Cowley SM; Hodgkinson JT MDM2 Antagonist Idasanutlin Reduces HDAC1/2 Abundance and Corepressor Partners but Not HDAC3. ACS Med Chem Lett 2024, 15 (1), 93–98. 10.1021/acsmedchemlett.3c00449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (124).Pavan AR; Smalley JP; Patel U; Pytel WA; Dos Santos JL; Cowley SM; Schwabe JWR; Hodgkinson JT Cereblon-Recruiting Proteolysis Targeting Chimeras (PROTACs) Can Determine the Selective Degradation of HDAC1 over HDAC3. Chem Commun (Camb) 2024, 60 (94), 13879–13882. 10.1039/d4cc05138f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (125).Fuller NO; Pirone A; Lynch BA; Hewitt MC; Quinton MS; McKee TD; Ivarsson M CoREST Complex-Selective Histone Deacetylase Inhibitors Show Prosynaptic Effects and an Improved Safety Profile To Enable Treatment of Synaptopathies. ACS Chem Neurosci 2019, 10 (3), 1729–1743. 10.1021/acschemneuro.8b00620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (126).Kalin JH; Wu M; Gomez AV; Song Y; Das J; Hayward D; Adejola N; Wu M; Panova I; Chung HJ; Kim E; Roberts HJ; Roberts JM; Prusevich P; Jeliazkov JR; Roy Burman SS; Fairall L; Milano C; Eroglu A; Proby CM; Dinkova-Kostova AT; Hancock WW; Gray JJ; Bradner JE; Valente S; Mai A; Anders NM; Rudek MA; Hu Y; Ryu B; Schwabe JWR; Mattevi A; Alani RM; Cole PA Targeting the CoREST Complex with Dual Histone Deacetylase and Demethylase Inhibitors. Nat Commun 2018, 9 (1), 53. 10.1038/s41467-017-02242-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (127).Milelli A; Marchetti C; Turrini E; Catanzaro E; Mazzone R; Tomaselli D; Fimognari C; Tumiatti V; Minarini A Novel Polyamine-Based Histone Deacetylases-Lysine Demethylase 1 Dual Binding Inhibitors. Bioorg Med Chem Lett 2018, 28 (6), 1001–1004. 10.1016/j.bmcl.2018.02.034. [DOI] [PubMed] [Google Scholar]
- (128).Anastas JN; Zee BM; Kalin JH; Kim M; Guo R; Alexandrescu S; Blanco MA; Giera S; Gillespie SM; Das J; Wu M; Nocco S; Bonal DM; Nguyen Q-D; Suva ML; Bernstein BE; Alani R; Golub TR; Cole PA; Filbin MG; Shi Y Re-Programing Chromatin with a Bifunctional LSD1/HDAC Inhibitor Induces Therapeutic Differentiation in DIPG. Cancer Cell 2019, 36 (5), 528–544.e10. 10.1016/j.ccell.2019.09.005. [DOI] [PubMed] [Google Scholar]
- (129).Xie X; Zhang O; Yeo MJR; Lee C; Tao R; Harry SA; Payne NC; Nam E; Paul L; Li Y; Kwok HS; Jiang H; Mao H; Hadley JL; Lin H; Batts M; Gosavi PM; D’Angiolella V; Cole PA; Mazitschek R; Northcott PA; Zheng N; Liau BB Converging Mechanism of UM171 and KBTBD4 Neomorphic Cancer Mutations. Nature 2025, 639 (8053), 241–249. 10.1038/s41586-024-08533-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (130).Yeo MJR; Zhang O; Xie X; Nam E; Payne NC; Gosavi PM; Kwok HS; Iram I; Lee C; Li J; Chen NJ; Nguyen K; Jiang H; Wang ZA; Lee K; Mao H; Harry SA; Barakat IA; Takahashi M; Waterbury AL; Barone M; Mattevi A; Carr SA; Udeshi ND; Bar-Peled L; Cole PA; Mazitschek R; Liau BB; Zheng N UM171 Glues Asymmetric CRL3-HDAC1/2 Assembly to Degrade CoREST Corepressors. Nature 2025, 639 (8053), 232–240. 10.1038/s41586-024-08532-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (131).Chen Z; Ioris RM; Richardson S; Van Ess AN; Vendrell I; Kessler BM; Buffa FM; Busino L; Clifford SC; Bullock AN; D’Angiolella V Disease-Associated KBTBD4 Mutations in Medulloblastoma Elicit Neomorphic Ubiquitylation Activity to Promote CoREST Degradation. Cell Death Differ 2022, 29 (10), 1955–1969. 10.1038/s41418-022-00983-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (132).Žemaitis K; Ghosh S; Hansson J; Subramaniam A The Stem Cell-Supporting Small Molecule UM171 Triggers Cul3-KBTBD4-Mediated Degradation of ELM2 Domain-Harboring Proteins. J Biol Chem 2023, 299 (5), 104662. 10.1016/j.jbc.2023.104662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (133).Chagraoui J; Girard S; Spinella J-F; Simon L; Bonneil E; Mayotte N; MacRae T; Coulombe-Huntington J; Bertomeu T; Moison C; Tomellini E; Thibault P; Tyers M; Marinier A; Sauvageau G UM171 Preserves Epigenetic Marks That Are Reduced in Ex Vivo Culture of Human HSCs via Potentiation of the CLR3-KBTBD4 Complex. Cell Stem Cell 2021, 28 (1), 48–62.e6. 10.1016/j.stem.2020.12.002. [DOI] [PubMed] [Google Scholar]
- (134).Silverman LR; Demakos EP; Peterson BL; Kornblith AB; Holland JC; Odchimar-Reissig R; Stone RM; Nelson D; Powell BL; DeCastro CM; Ellerton J; Larson RA; Schiffer CA; Holland JF Randomized Controlled Trial of Azacitidine in Patients with the Myelodysplastic Syndrome: A Study of the Cancer and Leukemia Group B. J Clin Oncol 2002, 20 (10), 2429–2440. 10.1200/JCO.2002.04.117. [DOI] [PubMed] [Google Scholar]
- (135).Jones PA; Taylor SM Cellular Differentiation, Cytidine Analogs and DNA Methylation. Cell 1980, 20 (1), 85–93. 10.1016/0092-8674(80)90237-8. [DOI] [PubMed] [Google Scholar]
- (136).Sorm F; Pískala A; Cihák A; Veselý J 5-Azacytidine, a New, Highly Effective Cancerostatic. Experientia 1964, 20 (4), 202–203. 10.1007/BF02135399. [DOI] [PubMed] [Google Scholar]
- (137).Jüttermann R; Li E; Jaenisch R Toxicity of 5-Aza-2’-Deoxycytidine to Mammalian Cells Is Mediated Primarily by Covalent Trapping of DNA Methyltransferase Rather than DNA Demethylation. Proc Natl Acad Sci U S A 1994, 91 (25), 11797–11801. 10.1073/pnas.91.25.11797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (138).Li LH; Olin EJ; Buskirk HH; Reineke LM Cytotoxicity and Mode of Action of 5-Azacytidine on L1210 Leukemia. Cancer Res 1970, 30 (11), 2760–2769. [PubMed] [Google Scholar]
- (139).Hollenbach PW; Nguyen AN; Brady H; Williams M; Ning Y; Richard N; Krushel L; Aukerman SL; Heise C; MacBeth KJ A Comparison of Azacitidine and Decitabine Activities in Acute Myeloid Leukemia Cell Lines. PLoS One 2010, 5 (2), e9001. 10.1371/journal.pone.0009001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (140).Aimiuwu J; Wang H; Chen P; Xie Z; Wang J; Liu S; Klisovic R; Mims A; Blum W; Marcucci G; Chan KK RNA-Dependent Inhibition of Ribonucleotide Reductase Is a Major Pathway for 5-Azacytidine Activity in Acute Myeloid Leukemia. Blood 2012, 119 (22), 5229–5238. 10.1182/blood-2011-11-382226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (141).Diesch J; Zwick A; Garz A-K; Palau A; Buschbeck M; Götze KS A Clinical-Molecular Update on Azanucleoside-Based Therapy for the Treatment of Hematologic Cancers. Clin Epigenetics 2016, 8, 71. 10.1186/s13148-016-0237-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (142).Gruenbaum Y; Cedar H; Razin A Substrate and Sequence Specificity of a Eukaryotic DNA Methylase. Nature 1982, 295 (5850), 620–622. 10.1038/295620a0. [DOI] [PubMed] [Google Scholar]
- (143).Bestor T; Laudano A; Mattaliano R; Ingram V Cloning and Sequencing of a CDNA Encoding DNA Methyltransferase of Mouse Cells. The Carboxyl-Terminal Domain of the Mammalian Enzymes Is Related to Bacterial Restriction Methyltransferases. J Mol Biol 1988, 203 (4), 971–983. 10.1016/0022-2836(88)90122-2. [DOI] [PubMed] [Google Scholar]
- (144).Li E; Bestor TH; Jaenisch R Targeted Mutation of the DNA Methyltransferase Gene Results in Embryonic Lethality. Cell 1992, 69 (6), 915–926. 10.1016/0092-8674(92)90611-f. [DOI] [PubMed] [Google Scholar]
- (145).Lei H; Oh SP; Okano M; Jüttermann R; Goss KA; Jaenisch R; Li E De Novo DNA Cytosine Methyltransferase Activities in Mouse Embryonic Stem Cells. Development 1996, 122 (10), 3195–3205. 10.1242/dev.122.10.3195. [DOI] [PubMed] [Google Scholar]
- (146).Okano M; Xie S; Li E Cloning and Characterization of a Family of Novel Mammalian DNA (Cytosine-5) Methyltransferases. Nat Genet 1998, 19 (3), 219–220. 10.1038/890. [DOI] [PubMed] [Google Scholar]
- (147).Okano M; Bell DW; Haber DA; Li E DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for de Novo Methylation and Mammalian Development. Cell 1999, 99 (3), 247–257. 10.1016/s0092-8674(00)81656-6. [DOI] [PubMed] [Google Scholar]
- (148).Aoki A; Suetake I; Miyagawa J; Fujio T; Chijiwa T; Sasaki H; Tajima S Enzymatic Properties of de Novo-Type Mouse DNA (Cytosine-5) Methyltransferases. Nucleic Acids Res 2001, 29 (17), 3506–3512. 10.1093/nar/29.17.3506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (149).Fatemi M; Hermann A; Pradhan S; Jeltsch A The Activity of the Murine DNA Methyltransferase Dnmt1 Is Controlled by Interaction of the Catalytic Domain with the N-Terminal Part of the Enzyme Leading to an Allosteric Activation of the Enzyme after Binding to Methylated DNA. J Mol Biol 2001, 309 (5), 1189–1199. 10.1006/jmbi.2001.4709. [DOI] [PubMed] [Google Scholar]
- (150).Bostick M; Kim JK; Estève P-O; Clark A; Pradhan S; Jacobsen SE UHRF1 Plays a Role in Maintaining DNA Methylation in Mammalian Cells. Science 2007, 317 (5845), 1760–1764. 10.1126/science.1147939. [DOI] [PubMed] [Google Scholar]
- (151).Sharif J; Muto M; Takebayashi S; Suetake I; Iwamatsu A; Endo TA; Shinga J; Mizutani-Koseki Y; Toyoda T; Okamura K; Tajima S; Mitsuya K; Okano M; Koseki H The SRA Protein Np95 Mediates Epigenetic Inheritance by Recruiting Dnmt1 to Methylated DNA. Nature 2007, 450 (7171), 908–912. 10.1038/nature06397. [DOI] [PubMed] [Google Scholar]
- (152).Hashimoto H; Horton JR; Zhang X; Bostick M; Jacobsen SE; Cheng X The SRA Domain of UHRF1 Flips 5-Methylcytosine out of the DNA Helix. Nature 2008, 455 (7214), 826–829. 10.1038/nature07280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (153).Avvakumov GV; Walker JR; Xue S; Li Y; Duan S; Bronner C; Arrowsmith CH; Dhe-Paganon S Structural Basis for Recognition of Hemi-Methylated DNA by the SRA Domain of Human UHRF1. Nature 2008, 455 (7214), 822–825. 10.1038/nature07273. [DOI] [PubMed] [Google Scholar]
- (154).Arita K; Ariyoshi M; Tochio H; Nakamura Y; Shirakawa M Recognition of Hemi-Methylated DNA by the SRA Protein UHRF1 by a Base-Flipping Mechanism. Nature 2008, 455 (7214), 818–821. 10.1038/nature07249. [DOI] [PubMed] [Google Scholar]
- (155).Song J; Rechkoblit O; Bestor TH; Patel DJ Structure of DNMT1-DNA Complex Reveals a Role for Autoinhibition in Maintenance DNA Methylation. Science 2011, 331 (6020), 1036–1040. 10.1126/science.1195380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (156).Yoo CB; Jeong S; Egger G; Liang G; Phiasivongsa P; Tang C; Redkar S; Jones PA Delivery of 5-Aza-2’-Deoxycytidine to Cells Using Oligodeoxynucleotides. Cancer Res 2007, 67 (13), 6400–6408. 10.1158/0008-5472.CAN-07-0251. [DOI] [PubMed] [Google Scholar]
- (157).Chuang JC; Warner SL; Vollmer D; Vankayalapati H; Redkar S; Bearss DJ; Qiu X; Yoo CB; Jones PA S110, a 5-Aza-2’-Deoxycytidine-Containing Dinucleotide, Is an Effective DNA Methylation Inhibitor in Vivo and Can Reduce Tumor Growth. Mol Cancer Ther 2010, 9 (5), 1443–1450. 10.1158/1535-7163.MCT-09-1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (158).Issa J-PJ; Roboz G; Rizzieri D; Jabbour E; Stock W; O’Connell C; Yee K; Tibes R; Griffiths EA; Walsh K; Daver N; Chung W; Naim S; Taverna P; Oganesian A; Hao Y; Lowder JN; Azab M; Kantarjian H Safety and Tolerability of Guadecitabine (SGI-110) in Patients with Myelodysplastic Syndrome and Acute Myeloid Leukaemia: A Multicentre, Randomised, Dose-Escalation Phase 1 Study. Lancet Oncol 2015, 16 (9), 1099–1110. 10.1016/S1470-2045(15)00038-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (159).Brueckner B; Garcia Boy R; Siedlecki P; Musch T; Kliem HC; Zielenkiewicz P; Suhai S; Wiessler M; Lyko F Epigenetic Reactivation of Tumor Suppressor Genes by a Novel Small-Molecule Inhibitor of Human DNA Methyltransferases. Cancer Res 2005, 65 (14), 6305–6311. 10.1158/0008-5472.CAN-04-2957. [DOI] [PubMed] [Google Scholar]
- (160).Datta J; Ghoshal K; Denny WA; Gamage SA; Brooke DG; Phiasivongsa P; Redkar S; Jacob ST A New Class of Quinoline-Based DNA Hypomethylating Agents Reactivates Tumor Suppressor Genes by Blocking DNA Methyltransferase 1 Activity and Inducing Its Degradation. Cancer Res 2009, 69 (10), 4277–4285. 10.1158/0008-5472.CAN-08-3669. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- (161).Kuck D; Caulfield T; Lyko F; Medina-Franco JL Nanaomycin A Selectively Inhibits DNMT3B and Reactivates Silenced Tumor Suppressor Genes in Human Cancer Cells. Mol Cancer Ther 2010, 9 (11), 3015–3023. 10.1158/1535-7163.MCT-10-0609. [DOI] [PubMed] [Google Scholar]
- (162).Manara MC; Valente S; Cristalli C; Nicoletti G; Landuzzi L; Zwergel C; Mazzone R; Stazi G; Arimondo PB; Pasello M; Guerzoni C; Picci P; Nanni P; Lollini P-L; Mai A; Scotlandi K A Quinoline-Based DNA Methyltransferase Inhibitor as a Possible Adjuvant in Osteosarcoma Therapy. Mol Cancer Ther 2018, 17 (9), 1881–1892. 10.1158/1535-7163.MCT-17-0818. [DOI] [PubMed] [Google Scholar]
- (163).Pappalardi MB; Keenan K; Cockerill M; Kellner WA; Stowell A; Sherk C; Wong K; Pathuri S; Briand J; Steidel M; Chapman P; Groy A; Wiseman AK; McHugh CF; Campobasso N; Graves AP; Fairweather E; Werner T; Raoof A; Butlin RJ; Rueda L; Horton JR; Fosbenner DT; Zhang C; Handler JL; Muliaditan M; Mebrahtu M; Jaworski J-P; McNulty DE; Burt C; Eberl HC; Taylor AN; Ho T; Merrihew S; Foley SW; Rutkowska A; Li M; Romeril SP; Goldberg K; Zhang X; Kershaw CS; Bantscheff M; Jurewicz AJ; Minthorn E; Grandi P; Patel M; Benowitz AB; Mohammad HP; Gilmartin AG; Prinjha RK; Ogilvie D; Carpenter C; Heerding D; Baylin SB; Jones PA; Cheng X; King BW; Luengo JI; Jordan AM; Waddell I; Kruger RG; McCabe MT Discovery of a First-in-Class Reversible DNMT1-Selective Inhibitor with Improved Tolerability and Efficacy in Acute Myeloid Leukemia. Nat Cancer 2021, 2 (10), 1002–1017. [PMC free article] [PubMed] [Google Scholar]
- (164).Caulfield T; Medina-Franco JL Molecular Dynamics Simulations of Human DNA Methyltransferase 3B with Selective Inhibitor Nanaomycin A. J Struct Biol 2011, 176 (2), 185–191. 10.1016/j.jsb.2011.07.015. [DOI] [PubMed] [Google Scholar]
- (165).Filippakopoulos P; Qi J; Picaud S; Shen Y; Smith WB; Fedorov O; Morse EM; Keates T; Hickman TT; Felletar I; Philpott M; Munro S; McKeown MR; Wang Y; Christie AL; West N; Cameron MJ; Schwartz B; Heightman TD; La Thangue N; French CA; Wiest O; Kung AL; Knapp S; Bradner JE Selective Inhibition of BET Bromodomains. Nature 2010, 468 (7327), 1067–1073. 10.1038/nature09504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (166).Nicodeme E; Jeffrey KL; Schaefer U; Beinke S; Dewell S; Chung C-W; Chandwani R; Marazzi I; Wilson P; Coste H; White J; Kirilovsky J; Rice CM; Lora JM; Prinjha RK; Lee K; Tarakhovsky A Suppression of Inflammation by a Synthetic Histone Mimic. Nature 2010, 468 (7327), 1119–1123. 10.1038/nature09589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (167).Chung C-W; Coste H; White JH; Mirguet O; Wilde J; Gosmini RL; Delves C; Magny SM; Woodward R; Hughes SA; Boursier EV; Flynn H; Bouillot AM; Bamborough P; Brusq J-MG; Gellibert FJ; Jones EJ; Riou AM; Homes P; Martin SL; Uings IJ; Toum J; Clement CA; Boullay A-B; Grimley RL; Blandel FM; Prinjha RK; Lee K; Kirilovsky J; Nicodeme E Discovery and Characterization of Small Molecule Inhibitors of the BET Family Bromodomains. J Med Chem 2011, 54 (11), 3827–3838. 10.1021/jm200108t. [DOI] [PubMed] [Google Scholar]
- (168).Yang Z; Yik JHN; Chen R; He N; Jang MK; Ozato K; Zhou Q Recruitment of P-TEFb for Stimulation of Transcriptional Elongation by the Bromodomain Protein Brd4. Mol Cell 2005, 19 (4), 535–545. 10.1016/j.molcel.2005.06.029. [DOI] [PubMed] [Google Scholar]
- (169).Jang MK; Mochizuki K; Zhou M; Jeong H-S; Brady JN; Ozato K The Bromodomain Protein Brd4 Is a Positive Regulatory Component of P-TEFb and Stimulates RNA Polymerase II-Dependent Transcription. Mol Cell 2005, 19 (4), 523–534. 10.1016/j.molcel.2005.06.027. [DOI] [PubMed] [Google Scholar]
- (170).Filippakopoulos P; Knapp S Targeting Bromodomains: Epigenetic Readers of Lysine Acetylation. Nat Rev Drug Discov 2014, 13 (5), 337–356. 10.1038/nrd4286. [DOI] [PubMed] [Google Scholar]
- (171).Yang Z; He N; Zhou Q Brd4 Recruits P-TEFb to Chromosomes at Late Mitosis to Promote G1 Gene Expression and Cell Cycle Progression. Mol Cell Biol 2008, 28 (3), 967–976. 10.1128/MCB.01020-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (172).Zuber J; Shi J; Wang E; Rappaport AR; Herrmann H; Sison EA; Magoon D; Qi J; Blatt K; Wunderlich M; Taylor MJ; Johns C; Chicas A; Mulloy JC; Kogan SC; Brown P; Valent P; Bradner JE; Lowe SW; Vakoc CR RNAi Screen Identifies Brd4 as a Therapeutic Target in Acute Myeloid Leukaemia. Nature 2011, 478 (7370), 524–528. 10.1038/nature10334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (173).Dawson MA; Prinjha RK; Dittmann A; Giotopoulos G; Bantscheff M; Chan W-I; Robson SC; Chung C; Hopf C; Savitski MM; Huthmacher C; Gudgin E; Lugo D; Beinke S; Chapman TD; Roberts EJ; Soden PE; Auger KR; Mirguet O; Doehner K; Delwel R; Burnett AK; Jeffrey P; Drewes G; Lee K; Huntly BJP; Kouzarides T Inhibition of BET Recruitment to Chromatin as an Effective Treatment for MLL-Fusion Leukaemia. Nature 2011, 478 (7370), 529–533. 10.1038/nature10509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (174).Delmore JE; Issa GC; Lemieux ME; Rahl PB; Shi J; Jacobs HM; Kastritis E; Gilpatrick T; Paranal RM; Qi J; Chesi M; Schinzel AC; McKeown MR; Heffernan TP; Vakoc CR; Bergsagel PL; Ghobrial IM; Richardson PG; Young RA; Hahn WC; Anderson KC; Kung AL; Bradner JE; Mitsiades CS BET Bromodomain Inhibition as a Therapeutic Strategy to Target C-Myc. Cell 2011, 146 (6), 904–917. 10.1016/j.cell.2011.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (175).Mertz JA; Conery AR; Bryant BM; Sandy P; Balasubramanian S; Mele DA; Bergeron L; Sims RJ Targeting MYC Dependence in Cancer by Inhibiting BET Bromodomains. Proc Natl Acad Sci U S A 2011, 108 (40), 16669–16674. 10.1073/pnas.1108190108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (176).Ott CJ; Kopp N; Bird L; Paranal RM; Qi J; Bowman T; Rodig SJ; Kung AL; Bradner JE; Weinstock DM BET Bromodomain Inhibition Targets Both C-Myc and IL7R in High-Risk Acute Lymphoblastic Leukemia. Blood 2012, 120 (14), 2843–2852. 10.1182/blood-2012-02-413021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (177).Lovén J; Hoke HA; Lin CY; Lau A; Orlando DA; Vakoc CR; Bradner JE; Lee TI; Young RA Selective Inhibition of Tumor Oncogenes by Disruption of Super-Enhancers. Cell 2013, 153 (2), 320–334. 10.1016/j.cell.2013.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (178).Chapuy B; McKeown MR; Lin CY; Monti S; Roemer MGM; Qi J; Rahl PB; Sun HH; Yeda KT; Doench JG; Reichert E; Kung AL; Rodig SJ; Young RA; Shipp MA; Bradner JE Discovery and Characterization of Super-Enhancer-Associated Dependencies in Diffuse Large B Cell Lymphoma. Cancer Cell 2013, 24 (6), 777–790. 10.1016/j.ccr.2013.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (179).Berthon C; Raffoux E; Thomas X; Vey N; Gomez-Roca C; Yee K; Taussig DC; Rezai K; Roumier C; Herait P; Kahatt C; Quesnel B; Michallet M; Recher C; Lokiec F; Preudhomme C; Dombret H Bromodomain Inhibitor OTX015 in Patients with Acute Leukaemia: A Dose-Escalation, Phase 1 Study. Lancet Haematol 2016, 3 (4), e186–95. 10.1016/S2352-3026(15)00247-1. [DOI] [PubMed] [Google Scholar]
- (180).Amorim S; Stathis A; Gleeson M; Iyengar S; Magarotto V; Leleu X; Morschhauser F; Karlin L; Broussais F; Rezai K; Herait P; Kahatt C; Lokiec F; Salles G; Facon T; Palumbo A; Cunningham D; Zucca E; Thieblemont C Bromodomain Inhibitor OTX015 in Patients with Lymphoma or Multiple Myeloma: A Dose-Escalation, Open-Label, Pharmacokinetic, Phase 1 Study. Lancet Haematol 2016, 3 (4), e196–204. 10.1016/S2352-3026(16)00021-1. [DOI] [PubMed] [Google Scholar]
- (181).Stathis A; Zucca E; Bekradda M; Gomez-Roca C; Delord J-P; de La Motte Rouge T; Uro-Coste E; de Braud F; Pelosi G; French CA Clinical Response of Carcinomas Harboring the BRD4-NUT Oncoprotein to the Targeted Bromodomain Inhibitor OTX015/MK-8628. Cancer Discov 2016, 6 (5), 492–500. 10.1158/2159-8290.CD-15-1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (182).Lewin J; Soria J-C; Stathis A; Delord J-P; Peters S; Awada A; Aftimos PG; Bekradda M; Rezai K; Zeng Z; Hussain A; Perez S; Siu LL; Massard C Phase Ib Trial With Birabresib, a Small-Molecule Inhibitor of Bromodomain and Extraterminal Proteins, in Patients With Selected Advanced Solid Tumors. J Clin Oncol 2018, 36 (30), 3007–3014. 10.1200/JCO.2018.78.2292. [DOI] [PubMed] [Google Scholar]
- (183).Rampal RK; Grosicki S; Chraniuk D; Abruzzese E; Bose P; Gerds AT; Vannucchi AM; Palandri F; Lee S-E; Gupta V; Lucchesi A; Oh ST; Kuykendall AT; Patriarca A; Alvarez-Larran A; Mesa R; Kiladjian J-J; Talpaz M; Harris M; Kays S-K; Jegg A-M; Li Q; Brown B; Harrison CN; Mascarenhas J Pelabresib in Combination with Ruxolitinib for Janus Kinase Inhibitor Treatment-Naïve Patients with Myelofibrosis: Results of the MANIFEST-2 Randomized, Double-Blind, Phase 3 Study. Blood 2023, 142 (Supplement 1), 628–628. 10.1182/blood-2023-179141. [DOI] [Google Scholar]
- (184).Kleppe M; Koche R; Zou L; van Galen P; Hill CE; Dong L; De Groote S; Papalexi E; Hanasoge Somasundara AV; Cordner K; Keller M; Farnoud N; Medina J; McGovern E; Reyes J; Roberts J; Witkin M; Rapaport F; Teruya-Feldstein J; Qi J; Rampal R; Bernstein BE; Bradner JE; Levine RL Dual Targeting of Oncogenic Activation and Inflammatory Signaling Increases Therapeutic Efficacy in Myeloproliferative Neoplasms. Cancer Cell 2018, 33 (1), 29–43.e7. 10.1016/j.ccell.2017.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (185).Brown JD; Lin CY; Duan Q; Griffin G; Federation A; Paranal RM; Bair S; Newton G; Lichtman A; Kung A; Yang T; Wang H; Luscinskas FW; Croce K; Bradner JE; Plutzky J NF-KB Directs Dynamic Super Enhancer Formation in Inflammation and Atherogenesis. Mol Cell 2014, 56 (2), 219–231. 10.1016/j.molcel.2014.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (186).Weiss WA; Taylor SS; Shokat KM Recognizing and Exploiting Differences between RNAi and Small-Molecule Inhibitors. Nat Chem Biol 2007, 3 (12), 739–744. 10.1038/nchembio1207-739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (187).Frye SV The Art of the Chemical Probe. Nat Chem Biol 2010, 6 (3), 159–161. 10.1038/nchembio.296. [DOI] [PubMed] [Google Scholar]
- (188).Arrowsmith CH; Audia JE; Austin C; Baell J; Bennett J; Blagg J; Bountra C; Brennan PE; Brown PJ; Bunnage ME; Buser-Doepner C; Campbell RM; Carter AJ; Cohen P; Copeland RA; Cravatt B; Dahlin JL; Dhanak D; Edwards AM; Frederiksen M; Frye SV; Gray N; Grimshaw CE; Hepworth D; Howe T; Huber KVM; Jin J; Knapp S; Kotz JD; Kruger RG; Lowe D; Mader MM; Marsden B; Mueller-Fahrnow A; Müller S; O’Hagan RC; Overington JP; Owen DR; Rosenberg SH; Roth B; Ross R; Schapira M; Schreiber SL; Shoichet B; Sundström M; Superti-Furga G; Taunton J; Toledo-Sherman L; Walpole C; Walters MA; Willson TM; Workman P; Young RN; Zuercher WJ The Promise and Peril of Chemical Probes. Nat Chem Biol 2015, 11 (8), 536–541. 10.1038/nchembio.1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (189).Kaelin WG Common Pitfalls in Preclinical Cancer Target Validation. Nat Rev Cancer 2017, 17 (7), 425–440. 10.1038/nrc.2017.32. [DOI] [PubMed] [Google Scholar]
- (190).Blagg J; Workman P Choose and Use Your Chemical Probe Wisely to Explore Cancer Biology. Cancer Cell 2017, 32 (1), 9–25. 10.1016/j.ccell.2017.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (191).Shu S; Lin CY; He HH; Witwicki RM; Tabassum DP; Roberts JM; Janiszewska M; Huh SJ; Liang Y; Ryan J; Doherty E; Mohammed H; Guo H; Stover DG; Ekram MB; Brown J; D’Santos C; Krop IE; Dillon D; McKeown M; Ott C; Qi J; Ni M; Rao PK; Duarte M; Wu S-Y; Chiang C-M; Anders L; Young RA; Winer E; Letai A; Barry WT; Carroll JS; Long H; Brown M; Liu XS; Meyer CA; Bradner JE; Polyak K Response and Resistance to BET Bromodomain Inhibitors in Triple-Negative Breast Cancer. Nature 2016, 529 (7586), 413–417. 10.1038/nature16508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (192).Fong CY; Gilan O; Lam EYN; Rubin AF; Ftouni S; Tyler D; Stanley K; Sinha D; Yeh P; Morison J; Giotopoulos G; Lugo D; Jeffrey P; Lee SC-W; Carpenter C; Gregory R; Ramsay RG; Lane SW; Abdel-Wahab O; Kouzarides T; Johnstone RW; Dawson S-J; Huntly BJP; Prinjha RK; Papenfuss AT; Dawson MA BET Inhibitor Resistance Emerges from Leukaemia Stem Cells. Nature 2015, 525 (7570), 538–542. 10.1038/nature14888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (193).Rathert P; Roth M; Neumann T; Muerdter F; Roe J-S; Muhar M; Deswal S; Cerny-Reiterer S; Peter B; Jude J; Hoffmann T; Boryń ŁM; Axelsson E; Schweifer N; Tontsch-Grunt U; Dow LE; Gianni D; Pearson M; Valent P; Stark A; Kraut N; Vakoc CR; Zuber J Transcriptional Plasticity Promotes Primary and Acquired Resistance to BET Inhibition. Nature 2015, 525 (7570), 543–547. 10.1038/nature14898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (194).Ge JY; Shu S; Kwon M; Jovanović B; Murphy K; Gulvady A; Fassl A; Trinh A; Kuang Y; Heavey GA; Luoma A; Paweletz C; Thorner AR; Wucherpfennig KW; Qi J; Brown M; Sicinski P; McDonald TO; Pellman D; Michor F; Polyak K Acquired Resistance to Combined BET and CDK4/6 Inhibition in Triple-Negative Breast Cancer. Nat Commun 2020, 11 (1), 2350. 10.1038/s41467-020-16170-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (195).Hewings DS; Wang M; Philpott M; Fedorov O; Uttarkar S; Filippakopoulos P; Picaud S; Vuppusetty C; Marsden B; Knapp S; Conway SJ; Heightman TD 3,5-Dimethylisoxazoles Act as Acetyl-Lysine-Mimetic Bromodomain Ligands. J Med Chem 2011, 54 (19), 6761–6770. 10.1021/jm200640v. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (196).Fish PV; Filippakopoulos P; Bish G; Brennan PE; Bunnage ME; Cook AS; Federov O; Gerstenberger BS; Jones H; Knapp S; Marsden B; Nocka K; Owen DR; Philpott M; Picaud S; Primiano MJ; Ralph MJ; Sciammetta N; Trzupek JD Identification of a Chemical Probe for Bromo and Extra C-Terminal Bromodomain Inhibition through Optimization of a Fragment-Derived Hit. J Med Chem 2012, 55 (22), 9831–9837. 10.1021/jm3010515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (197).Picaud S; Da Costa D; Thanasopoulou A; Filippakopoulos P; Fish PV; Philpott M; Fedorov O; Brennan P; Bunnage ME; Owen DR; Bradner JE; Taniere P; O’Sullivan B; Müller S; Schwaller J; Stankovic T; Knapp S PFI-1, a Highly Selective Protein Interaction Inhibitor, Targeting BET Bromodomains. Cancer Res 2013, 73 (11), 3336–3346. 10.1158/0008-5472.CAN-12-3292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (198).Bandopadhayay P; Bergthold G; Nguyen B; Schubert S; Gholamin S; Tang Y; Bolin S; Schumacher SE; Zeid R; Masoud S; Yu F; Vue N; Gibson WJ; Paolella BR; Mitra SS; Cheshier SH; Qi J; Liu K-W; Wechsler-Reya R; Weiss WA; Swartling FJ; Kieran MW; Bradner JE; Beroukhim R; Cho Y-J BET Bromodomain Inhibition of MYC-Amplified Medulloblastoma. Clin Cancer Res 2014, 20 (4), 912–925. 10.1158/1078-0432.CCR-13-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (199).Tang Y; Gholamin S; Schubert S; Willardson MI; Lee A; Bandopadhayay P; Bergthold G; Masoud S; Nguyen B; Vue N; Balansay B; Yu F; Oh S; Woo P; Chen S; Ponnuswami A; Monje M; Atwood SX; Whitson RJ; Mitra S; Cheshier SH; Qi J; Beroukhim R; Tang JY; Wechsler-Reya R; Oro AE; Link BA; Bradner JE; Cho Y-J Epigenetic Targeting of Hedgehog Pathway Transcriptional Output through BET Bromodomain Inhibition. Nat Med 2014, 20 (7), 732–740. 10.1038/nm.3613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (200).Cheng Z; Gong Y; Ma Y; Lu K; Lu X; Pierce LA; Thompson RC; Muller S; Knapp S; Wang J Inhibition of BET Bromodomain Targets Genetically Diverse Glioblastoma. Clin Cancer Res 2013, 19 (7), 1748–1759. 10.1158/1078-0432.CCR-12-3066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (201).Garcia PL; Miller AL; Kreitzburg KM; Council LN; Gamblin TL; Christein JD; Heslin MJ; Arnoletti JP; Richardson JH; Chen D; Hanna CA; Cramer SL; Yang ES; Qi J; Bradner JE; Yoon KJ The BET Bromodomain Inhibitor JQ1 Suppresses Growth of Pancreatic Ductal Adenocarcinoma in Patient-Derived Xenograft Models. Oncogene 2016, 35 (7), 833–845. 10.1038/onc.2015.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (202).Aird F; Kandela I; Mantis C; Reproducibility Project: Cancer Biology. Replication Study: BET Bromodomain Inhibition as a Therapeutic Strategy to Target c-Myc. Elife 2017, 6, e21253. 10.7554/eLife.21253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (203).Puissant A; Frumm SM; Alexe G; Bassil CF; Qi J; Chanthery YH; Nekritz EA; Zeid R; Gustafson WC; Greninger P; Garnett MJ; McDermott U; Benes CH; Kung AL; Weiss WA; Bradner JE; Stegmaier K Targeting MYCN in Neuroblastoma by BET Bromodomain Inhibition. Cancer Discov 2013, 3 (3), 308–323. 10.1158/2159-8290.CD-12-0418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (204).Zou Z; Huang B; Wu X; Zhang H; Qi J; Bradner J; Nair S; Chen L-F Brd4 Maintains Constitutively Active NF-KB in Cancer Cells by Binding to Acetylated RelA. Oncogene 2014, 33 (18), 2395–2404. 10.1038/onc.2013.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (205).Roe J-S; Mercan F; Rivera K; Pappin DJ; Vakoc CR BET Bromodomain Inhibition Suppresses the Function of Hematopoietic Transcription Factors in Acute Myeloid Leukemia. Mol Cell 2015, 58 (6), 1028–1039. 10.1016/j.molcel.2015.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (206).Winter GE; Mayer A; Buckley DL; Erb MA; Roderick JE; Vittori S; Reyes JM; di Iulio J; Souza A; Ott CJ; Roberts JM; Zeid R; Scott TG; Paulk J; Lachance K; Olson CM; Dastjerdi S; Bauer S; Lin CY; Gray NS; Kelliher MA; Churchman LS; Bradner JE BET Bromodomain Proteins Function as Master Transcription Elongation Factors Independent of CDK9 Recruitment. Mol Cell 2017, 67 (1), 5–18.e19. 10.1016/j.molcel.2017.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (207).Muhar M; Ebert A; Neumann T; Umkehrer C; Jude J; Wieshofer C; Rescheneder P; Lipp JJ; Herzog VA; Reichholf B; Cisneros DA; Hoffmann T; Schlapansky MF; Bhat P; von Haeseler A; Köcher T; Obenauf AC; Popow J; Ameres SL; Zuber J SLAM-Seq Defines Direct Gene-Regulatory Functions of the BRD4-MYC Axis. Science 2018, 360 (6390), 800–805. 10.1126/science.aao2793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (208).Burslem GM; Crews CM Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery. Cell 2020, 181 (1), 102–114. 10.1016/j.cell.2019.11.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (209).Ito T; Ando H; Suzuki T; Ogura T; Hotta K; Imamura Y; Yamaguchi Y; Handa H Identification of a Primary Target of Thalidomide Teratogenicity. Science 2010, 327 (5971), 1345–1350. 10.1126/science.1177319. [DOI] [PubMed] [Google Scholar]
- (210).Winter GE; Buckley DL; Paulk J; Roberts JM; Souza A; Dhe-Paganon S; Bradner JE DRUG DEVELOPMENT. Phthalimide Conjugation as a Strategy for in Vivo Target Protein Degradation. Science 2015, 348 (6241), 1376–1381. 10.1126/science.aab1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (211).Zengerle M; Chan K-H; Ciulli A Selective Small Molecule Induced Degradation of the BET Bromodomain Protein BRD4. ACS Chem Biol 2015, 10 (8), 1770–1777. 10.1021/acschembio.5b00216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (212).Gadd MS; Testa A; Lucas X; Chan K-H; Chen W; Lamont DJ; Zengerle M; Ciulli A Structural Basis of PROTAC Cooperative Recognition for Selective Protein Degradation. Nat Chem Biol 2017, 13 (5), 514–521. 10.1038/nchembio.2329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (213).Van Molle I; Thomann A; Buckley DL; So EC; Lang S; Crews CM; Ciulli A Dissecting Fragment-Based Lead Discovery at the von Hippel-Lindau Protein:Hypoxia Inducible Factor 1α Protein-Protein Interface. Chem Biol 2012, 19 (10), 1300–1312. 10.1016/j.chembiol.2012.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (214).Buckley DL; Gustafson JL; Van Molle I; Roth AG; Tae HS; Gareiss PC; Jorgensen WL; Ciulli A; Crews CM Small-Molecule Inhibitors of the Interaction between the E3 Ligase VHL and HIF1α. Angew Chem Int Ed Engl 2012, 51 (46), 11463–11467. 10.1002/anie.201206231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (215).Buckley DL; Van Molle I; Gareiss PC; Tae HS; Michel J; Noblin DJ; Jorgensen WL; Ciulli A; Crews CM Targeting the von Hippel-Lindau E3 Ubiquitin Ligase Using Small Molecules to Disrupt the VHL/HIF-1α Interaction. J Am Chem Soc 2012, 134 (10), 4465–4468. 10.1021/ja209924v. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (216).Galdeano C; Gadd MS; Soares P; Scaffidi S; Van Molle I; Birced I; Hewitt S; Dias DM; Ciulli A Structure-Guided Design and Optimization of Small Molecules Targeting the Protein-Protein Interaction between the von Hippel-Lindau (VHL) E3 Ubiquitin Ligase and the Hypoxia Inducible Factor (HIF) Alpha Subunit with in Vitro Nanomolar Affinities. J Med Chem 2014, 57 (20), 8657–8663. 10.1021/jm5011258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (217).Buckley DL; Raina K; Darricarrere N; Hines J; Gustafson JL; Smith IE; Miah AH; Harling JD; Crews CM HaloPROTACS: Use of Small Molecule PROTACs to Induce Degradation of HaloTag Fusion Proteins. ACS Chem Biol 2015, 10 (8), 1831–1837. 10.1021/acschembio.5b00442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (218).Remillard D; Buckley DL; Paulk J; Brien GL; Sonnett M; Seo H-S; Dastjerdi S; Wühr M; Dhe-Paganon S; Armstrong SA; Bradner JE Degradation of the BAF Complex Factor BRD9 by Heterobifunctional Ligands. Angew Chem Int Ed Engl 2017, 56 (21), 5738–5743. 10.1002/anie.201611281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (219).Bondeson DP; Smith BE; Burslem GM; Buhimschi AD; Hines J; Jaime-Figueroa S; Wang J; Hamman BD; Ishchenko A; Crews CM Lessons in PROTAC Design from Selective Degradation with a Promiscuous Warhead. Cell Chem Biol 2018, 25 (1), 78–87.e5. 10.1016/j.chembiol.2017.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (220).Nowak RP; DeAngelo SL; Buckley D; He Z; Donovan KA; An J; Safaee N; Jedrychowski MP; Ponthier CM; Ishoey M; Zhang T; Mancias JD; Gray NS; Bradner JE; Fischer ES Plasticity in Binding Confers Selectivity in Ligand-Induced Protein Degradation. Nat Chem Biol 2018, 14 (7), 706–714. 10.1038/s41589-018-0055-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (221).Olson CM; Jiang B; Erb MA; Liang Y; Doctor ZM; Zhang Z; Zhang T; Kwiatkowski N; Boukhali M; Green JL; Haas W; Nomanbhoy T; Fischer ES; Young RA; Bradner JE; Winter GE; Gray NS Pharmacological Perturbation of CDK9 Using Selective CDK9 Inhibition or Degradation. Nat Chem Biol 2018, 14 (2), 163–170. 10.1038/nchembio.2538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (222).Gopalsamy A Selectivity through Targeted Protein Degradation (TPD). J Med Chem 2022, 65 (12), 8113–8126. 10.1021/acs.jmedchem.2c00397. [DOI] [PubMed] [Google Scholar]
- (223).Brand M; Jiang B; Bauer S; Donovan KA; Liang Y; Wang ES; Nowak RP; Yuan JC; Zhang T; Kwiatkowski N; Müller AC; Fischer ES; Gray NS; Winter GE Homolog-Selective Degradation as a Strategy to Probe the Function of CDK6 in AML. Cell Chem Biol 2019, 26 (2), 300–306.e9. 10.1016/j.chembiol.2018.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (224).Huang H-T; Dobrovolsky D; Paulk J; Yang G; Weisberg EL; Doctor ZM; Buckley DL; Cho J-H; Ko E; Jang J; Shi K; Choi HG; Griffin JD; Li Y; Treon SP; Fischer ES; Bradner JE; Tan L; Gray NS A Chemoproteomic Approach to Query the Degradable Kinome Using a Multi-Kinase Degrader. Cell Chem Biol 2018, 25 (1), 88–99.e6. 10.1016/j.chembiol.2017.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (225).Lu J; Qian Y; Altieri M; Dong H; Wang J; Raina K; Hines J; Winkler JD; Crew AP; Coleman K; Crews CM Hijacking the E3 Ubiquitin Ligase Cereblon to Efficiently Target BRD4. Chem Biol 2015, 22 (6), 755–763. 10.1016/j.chembiol.2015.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (226).Harlen KM; Churchman LS The Code and beyond: Transcription Regulation by the RNA Polymerase II Carboxy-Terminal Domain. Nat Rev Mol Cell Biol 2017, 18 (4), 263–273. 10.1038/nrm.2017.10. [DOI] [PubMed] [Google Scholar]
- (227).Kwiatkowski N; Zhang T; Rahl PB; Abraham BJ; Reddy J; Ficarro SB; Dastur A; Amzallag A; Ramaswamy S; Tesar B; Jenkins CE; Hannett NM; McMillin D; Sanda T; Sim T; Kim ND; Look T; Mitsiades CS; Weng AP; Brown JR; Benes CH; Marto JA; Young RA; Gray NS Targeting Transcription Regulation in Cancer with a Covalent CDK7 Inhibitor. Nature 2014, 511 (7511), 616–620. 10.1038/nature13393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (228).Shenvi RA; Guerrero CA; Shi J; Li C-C; Baran PS Synthesis of (+)-Cortistatin A. J Am Chem Soc 2008, 130 (23), 7241–7243. 10.1021/ja8023466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (229).Lee HM; Nieto-Oberhuber C; Shair MD Enantioselective Synthesis of (+)-Cortistatin a, a Potent and Selective Inhibitor of Endothelial Cell Proliferation. J Am Chem Soc 2008, 130 (50), 16864–16866. 10.1021/ja8071918. [DOI] [PubMed] [Google Scholar]
- (230).Nicolaou KC; Sun Y-P; Peng X-S; Polet D; Chen DY-K Total Synthesis of (+)-Cortistatin A. Angew Chem Int Ed Engl 2008, 47 (38), 7310–7313. 10.1002/anie.200803550. [DOI] [PubMed] [Google Scholar]
- (231).Cee VJ; Chen DY-K; Lee MR; Nicolaou KC Cortistatin A Is a High-Affinity Ligand of Protein Kinases ROCK, CDK8, and CDK11. Angew Chem Int Ed Engl 2009, 48 (47), 8952–8957. 10.1002/anie.200904778. [DOI] [PubMed] [Google Scholar]
- (232).Pelish HE; Liau BB; Nitulescu II; Tangpeerachaikul A; Poss ZC; Da Silva DH; Caruso BT; Arefolov A; Fadeyi O; Christie AL; Du K; Banka D; Schneider EV; Jestel A; Zou G; Si C; Ebmeier CC; Bronson RT; Krivtsov AV; Myers AG; Kohl NE; Kung AL; Armstrong SA; Lemieux ME; Taatjes DJ; Shair MD Mediator Kinase Inhibition Further Activates Super-Enhancer-Associated Genes in AML. Nature 2015, 526 (7572), 273–276. 10.1038/nature14904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (233).Dale T; Clarke PA; Esdar C; Waalboer D; Adeniji-Popoola O; Ortiz-Ruiz M-J; Mallinger A; Samant RS; Czodrowski P; Musil D; Schwarz D; Schneider K; Stubbs M; Ewan K; Fraser E; TePoele R; Court W; Box G; Valenti M; de Haven Brandon A; Gowan S; Rohdich F; Raynaud F; Schneider R; Poeschke O; Blaukat A; Workman P; Schiemann K; Eccles SA; Wienke D; Blagg J A Selective Chemical Probe for Exploring the Role of CDK8 and CDK19 in Human Disease. Nat Chem Biol 2015, 11 (12), 973–980. 10.1038/nchembio.1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (234).Zhang T; Kwiatkowski N; Olson CM; Dixon-Clarke SE; Abraham BJ; Greifenberg AK; Ficarro SB; Elkins JM; Liang Y; Hannett NM; Manz T; Hao M; Bartkowiak B; Greenleaf AL; Marto JA; Geyer M; Bullock AN; Young RA; Gray NS Covalent Targeting of Remote Cysteine Residues to Develop CDK12 and CDK13 Inhibitors. Nat Chem Biol 2016, 12 (10), 876–884. 10.1038/nchembio.2166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (235).Quereda V; Bayle S; Vena F; Frydman SM; Monastyrskyi A; Roush WR; Duckett DR Therapeutic Targeting of CDK12/CDK13 in Triple-Negative Breast Cancer. Cancer Cell 2019, 36 (5), 545–558.e7. 10.1016/j.ccell.2019.09.004. [DOI] [PubMed] [Google Scholar]
- (236).Ferguson FM; Gray NS Kinase Inhibitors: The Road Ahead. Nat Rev Drug Discov 2018, 17 (5), 353–377. 10.1038/nrd.2018.21. [DOI] [PubMed] [Google Scholar]
- (237).Vervoort SJ; Devlin JR; Kwiatkowski N; Teng M; Gray NS; Johnstone RW Targeting Transcription Cycles in Cancer. Nat Rev Cancer 2022, 22 (1), 5–24. 10.1038/s41568-021-00411-8. [DOI] [PubMed] [Google Scholar]
- (238).Sehgal PB; Darnell JE; Tamm I The Inhibition by DRB (5,6-Dichloro-1-Beta-D-Ribofuranosylbenzimidazole) of HnRNA and MRNA Production in HeLa Cells. Cell 1976, 9 (3), 473–480. 10.1016/0092-8674(76)90092-1. [DOI] [PubMed] [Google Scholar]
- (239).Fraser NW; Sehgal PB; Darnell JE DRB-Induced Premature Termination of Late Adenovirus Transcription. Nature 1978, 272 (5654), 590–593. 10.1038/272590a0. [DOI] [PubMed] [Google Scholar]
- (240).Chodosh LA; Fire A; Samuels M; Sharp PA 5,6-Dichloro-1-Beta-D-Ribofuranosylbenzimidazole Inhibits Transcription Elongation by RNA Polymerase II in Vitro. J Biol Chem 1989, 264 (4), 2250–2257. [PubMed] [Google Scholar]
- (241).Marshall NF; Price DH Purification of P-TEFb, a Transcription Factor Required for the Transition into Productive Elongation. J Biol Chem 1995, 270 (21), 12335–12338. 10.1074/jbc.270.21.12335. [DOI] [PubMed] [Google Scholar]
- (242).Marshall NF; Peng J; Xie Z; Price DH Control of RNA Polymerase II Elongation Potential by a Novel Carboxyl-Terminal Domain Kinase. J Biol Chem 1996, 271 (43), 27176–27183. 10.1074/jbc.271.43.27176. [DOI] [PubMed] [Google Scholar]
- (243).Peterlin BM; Price DH Controlling the Elongation Phase of Transcription with P-TEFb. Mol Cell 2006, 23 (3), 297–305. 10.1016/j.molcel.2006.06.014. [DOI] [PubMed] [Google Scholar]
- (244).Wada T; Takagi T; Yamaguchi Y; Watanabe D; Handa H Evidence That P-TEFb Alleviates the Negative Effect of DSIF on RNA Polymerase II-Dependent Transcription in Vitro. EMBO J 1998, 17 (24), 7395–7403. 10.1093/emboj/17.24.7395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (245).Wada T; Takagi T; Yamaguchi Y; Ferdous A; Imai T; Hirose S; Sugimoto S; Yano K; Hartzog GA; Winston F; Buratowski S; Handa H DSIF, a Novel Transcription Elongation Factor That Regulates RNA Polymerase II Processivity, Is Composed of Human Spt4 and Spt5 Homologs. Genes Dev 1998, 12 (3), 343–356. 10.1101/gad.12.3.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (246).Hartzog GA; Wada T; Handa H; Winston F Evidence That Spt4, Spt5, and Spt6 Control Transcription Elongation by RNA Polymerase II in Saccharomyces Cerevisiae. Genes Dev 1998, 12 (3), 357–369. 10.1101/gad.12.3.357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (247).Yamaguchi Y; Takagi T; Wada T; Yano K; Furuya A; Sugimoto S; Hasegawa J; Handa H NELF, a Multisubunit Complex Containing RD, Cooperates with DSIF to Repress RNA Polymerase II Elongation. Cell 1999, 97 (1), 41–51. 10.1016/s0092-8674(00)80713-8. [DOI] [PubMed] [Google Scholar]
- (248).Fujinaga K; Irwin D; Huang Y; Taube R; Kurosu T; Peterlin BM Dynamics of Human Immunodeficiency Virus Transcription: P-TEFb Phosphorylates RD and Dissociates Negative Effectors from the Transactivation Response Element. Mol Cell Biol 2004, 24 (2), 787–795. 10.1128/MCB.24.2.787-795.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (249).Kim JB; Sharp PA Positive Transcription Elongation Factor B Phosphorylates HSPT5 and RNA Polymerase II Carboxyl-Terminal Domain Independently of Cyclin-Dependent Kinase-Activating Kinase. J Biol Chem 2001, 276 (15), 12317–12323. 10.1074/jbc.M010908200. [DOI] [PubMed] [Google Scholar]
- (250).Ramanathan Y; Rajpara SM; Reza SM; Lees E; Shuman S; Mathews MB; Pe’ery T Three RNA Polymerase II Carboxyl-Terminal Domain Kinases Display Distinct Substrate Preferences. J Biol Chem 2001, 276 (14), 10913–10920. 10.1074/jbc.M010975200. [DOI] [PubMed] [Google Scholar]
- (251).Yamada T; Yamaguchi Y; Inukai N; Okamoto S; Mura T; Handa H P-TEFb-Mediated Phosphorylation of HSpt5 C-Terminal Repeats Is Critical for Processive Transcription Elongation. Mol Cell 2006, 21 (2), 227–237. 10.1016/j.molcel.2005.11.024. [DOI] [PubMed] [Google Scholar]
- (252).Chao SH; Price DH Flavopiridol Inactivates P-TEFb and Blocks Most RNA Polymerase II Transcription in Vivo. J Biol Chem 2001, 276 (34), 31793–31799. 10.1074/jbc.M102306200. [DOI] [PubMed] [Google Scholar]
- (253).Ni Z; Saunders A; Fuda NJ; Yao J; Suarez J-R; Webb WW; Lis JT P-TEFb Is Critical for the Maturation of RNA Polymerase II into Productive Elongation in Vivo. Mol Cell Biol 2008, 28 (3), 1161–1170. 10.1128/MCB.01859-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (254).Rahl PB; Lin CY; Seila AC; Flynn RA; McCuine S; Burge CB; Sharp PA; Young RA C-Myc Regulates Transcriptional Pause Release. Cell 2010, 141 (3), 432–445. 10.1016/j.cell.2010.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (255).Henriques T; Gilchrist DA; Nechaev S; Bern M; Muse GW; Burkholder A; Fargo DC; Adelman K Stable Pausing by RNA Polymerase II Provides an Opportunity to Target and Integrate Regulatory Signals. Mol Cell 2013, 52 (4), 517–528. 10.1016/j.molcel.2013.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (256).Jonkers I; Kwak H; Lis JT Genome-Wide Dynamics of Pol II Elongation and Its Interplay with Promoter Proximal Pausing, Chromatin, and Exons. Elife 2014, 3, e02407. 10.7554/eLife.02407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (257).Lu H; Xue Y; Yu GK; Arias C; Lin J; Fong S; Faure M; Weisburd B; Ji X; Mercier A; Sutton J; Luo K; Gao Z; Zhou Q Compensatory Induction of MYC Expression by Sustained CDK9 Inhibition via a BRD4-Dependent Mechanism. Elife 2015, 4, e06535. 10.7554/eLife.06535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (258).Richters A; Doyle SK; Freeman DB; Lee C; Leifer BS; Jagannathan S; Kabinger F; Koren JV; Struntz NB; Urgiles J; Stagg RA; Curtin BH; Chatterjee D; Mathea S; Mikochik PJ; Hopkins TD; Gao H; Branch JR; Xin H; Westover L; Bignan GC; Rupnow BA; Karlin KL; Olson CM; Westbrook TF; Vacca J; Wilfong CM; Trotter BW; Saffran DC; Bischofberger N; Knapp S; Russo JW; Hickson I; Bischoff JR; Gottardis MM; Balk SP; Lin CY; Pop MS; Koehler AN Modulating Androgen Receptor-Driven Transcription in Prostate Cancer with Selective CDK9 Inhibitors. Cell Chem Biol 2021, 28 (2), 134–147.e14. 10.1016/j.chembiol.2020.10.001. [DOI] [PubMed] [Google Scholar]
- (259).Cidado J; Boiko S; Proia T; Ferguson D; Criscione SW; San Martin M; Pop-Damkov P; Su N; Roamio Franklin VN; Sekhar Reddy Chilamakuri C; D’Santos CS; Shao W; Saeh JC; Koch R; Weinstock DM; Zinda M; Fawell SE; Drew L AZD4573 Is a Highly Selective CDK9 Inhibitor That Suppresses MCL-1 and Induces Apoptosis in Hematologic Cancer Cells. Clin Cancer Res 2020, 26 (4), 922–934. 10.1158/1078-0432.CCR-19-1853. [DOI] [PubMed] [Google Scholar]
- (260).Divakaran A; Harki DA; Pomerantz WCK Recent Progress and Structural Analyses of Domain-Selective BET Inhibitors. Med Res Rev 2023, 43 (4), 972–1018. 10.1002/med.21942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (261).Picaud S; Wells C; Felletar I; Brotherton D; Martin S; Savitsky P; Diez-Dacal B; Philpott M; Bountra C; Lingard H; Fedorov O; Müller S; Brennan PE; Knapp S; Filippakopoulos P RVX-208, an Inhibitor of BET Transcriptional Regulators with Selectivity for the Second Bromodomain. Proc Natl Acad Sci U S A 2013, 110 (49), 19754–19759. 10.1073/pnas.1310658110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (262).Urick AK; Hawk LML; Cassel MK; Mishra NK; Liu S; Adhikari N; Zhang W; dos Santos CO; Hall JL; Pomerantz WCK Dual Screening of BPTF and Brd4 Using Protein-Observed Fluorine NMR Uncovers New Bromodomain Probe Molecules. ACS Chem Biol 2015, 10 (10), 2246–2256. 10.1021/acschembio.5b00483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (263).Raux B; Voitovich Y; Derviaux C; Lugari A; Rebuffet E; Milhas S; Priet S; Roux T; Trinquet E; Guillemot J-C; Knapp S; Brunel J-M; Fedorov AY; Collette Y; Roche P; Betzi S; Combes S; Morelli X Exploring Selective Inhibition of the First Bromodomain of the Human Bromodomain and Extra-Terminal Domain (BET) Proteins. J Med Chem 2016, 59 (4), 1634–1641. 10.1021/acs.jmedchem.5b01708. [DOI] [PubMed] [Google Scholar]
- (264).Cheung K; Lu G; Sharma R; Vincek A; Zhang R; Plotnikov AN; Zhang F; Zhang Q; Ju Y; Hu Y; Zhao L; Han X; Meslamani J; Xu F; Jaganathan A; Shen T; Zhu H; Rusinova E; Zeng L; Zhou J; Yang J; Peng L; Ohlmeyer M; Walsh MJ; Zhang DY; Xiong H; Zhou M-M BET N-Terminal Bromodomain Inhibition Selectively Blocks Th17 Cell Differentiation and Ameliorates Colitis in Mice. Proc Natl Acad Sci U S A 2017, 114 (11), 2952–2957. 10.1073/pnas.1615601114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (265).Law RP; Atkinson SJ; Bamborough P; Chung C-W; Demont EH; Gordon LJ; Lindon M; Prinjha RK; Watson AJB; Hirst DJ Discovery of Tetrahydroquinoxalines as Bromodomain and Extra-Terminal Domain (BET) Inhibitors with Selectivity for the Second Bromodomain. J Med Chem 2018, 61 (10), 4317–4334. 10.1021/acs.jmedchem.7b01666. [DOI] [PubMed] [Google Scholar]
- (266).Shadrick WR; Slavish PJ; Chai SC; Waddell B; Connelly M; Low JA; Tallant C; Young BM; Bharatham N; Knapp S; Boyd VA; Morfouace M; Roussel MF; Chen T; Lee RE; Kiplin Guy R; Shelat AA; Potter PM Exploiting a Water Network to Achieve Enthalpy-Driven, Bromodomain-Selective BET Inhibitors. Bioorg Med Chem 2018, 26 (1), 25–36. 10.1016/j.bmc.2017.10.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (267).Jiang F; Hu Q; Zhang Z; Li H; Li H; Zhang D; Li H; Ma Y; Xu J; Chen H; Cui Y; Zhi Y; Zhang Y; Xu J; Zhu J; Lu T; Chen Y Discovery of Benzo[Cd]Indol-2(1H)-Ones and Pyrrolo[4,3,2-de]Quinolin-2(1H)-Ones as Bromodomain and Extra-Terminal Domain (BET) Inhibitors with Selectivity for the First Bromodomain with Potential High Efficiency against Acute Gouty Arthritis. J Med Chem 2019, 62 (24), 11080–11107. 10.1021/acs.jmedchem.9b01010. [DOI] [PubMed] [Google Scholar]
- (268).Faivre EJ; McDaniel KF; Albert DH; Mantena SR; Plotnik JP; Wilcox D; Zhang L; Bui MH; Sheppard GS; Wang L; Sehgal V; Lin X; Huang X; Lu X; Uziel T; Hessler P; Lam LT; Bellin RJ; Mehta G; Fidanze S; Pratt JK; Liu D; Hasvold LA; Sun C; Panchal SC; Nicolette JJ; Fossey SL; Park CH; Longenecker K; Bigelow L; Torrent M; Rosenberg SH; Kati WM; Shen Y Selective Inhibition of the BD2 Bromodomain of BET Proteins in Prostate Cancer. Nature 2020, 578 (7794), 306–310. 10.1038/s41586-020-1930-8. [DOI] [PubMed] [Google Scholar]
- (269).Sheppard GS; Wang L; Fidanze SD; Hasvold LA; Liu D; Pratt JK; Park CH; Longenecker K; Qiu W; Torrent M; Kovar PJ; Bui M; Faivre E; Huang X; Lin X; Wilcox D; Zhang L; Shen Y; Albert DH; Magoc TJ; Rajaraman G; Kati WM; McDaniel KF Discovery of N-Ethyl-4-[2-(4-Fluoro-2,6-Dimethyl-Phenoxy)-5-(1-Hydroxy-1-Methyl-Ethyl)Phenyl]-6-Methyl-7-Oxo-1H-Pyrrolo[2,3-c]Pyridine-2-Carboxamide (ABBV-744), a BET Bromodomain Inhibitor with Selectivity for the Second Bromodomain. J Med Chem 2020, 63 (10), 5585–5623. 10.1021/acs.jmedchem.0c00628. [DOI] [PubMed] [Google Scholar]
- (270).Gilan O; Rioja I; Knezevic K; Bell MJ; Yeung MM; Harker NR; Lam EYN; Chung C-W; Bamborough P; Petretich M; Urh M; Atkinson SJ; Bassil AK; Roberts EJ; Vassiliadis D; Burr ML; Preston AGS; Wellaway C; Werner T; Gray JR; Michon A-M; Gobbetti T; Kumar V; Soden PE; Haynes A; Vappiani J; Tough DF; Taylor S; Dawson S-J; Bantscheff M; Lindon M; Drewes G; Demont EH; Daniels DL; Grandi P; Prinjha RK; Dawson MA Selective Targeting of BD1 and BD2 of the BET Proteins in Cancer and Immunoinflammation. Science 2020, 368 (6489), 387–394. 10.1126/science.aaz8455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (271).Preston A; Atkinson S; Bamborough P; Chung C-W; Craggs PD; Gordon L; Grandi P; Gray JRJ; Jones EJ; Lindon M; Michon A-M; Mitchell DJ; Prinjha RK; Rianjongdee F; Rioja I; Seal J; Taylor S; Wall I; Watson RJ; Woolven J; Demont EH Design and Synthesis of a Highly Selective and In Vivo-Capable Inhibitor of the Second Bromodomain of the Bromodomain and Extra Terminal Domain Family of Proteins. J Med Chem 2020, 63 (17), 9070–9092. 10.1021/acs.jmedchem.0c00605. [DOI] [PubMed] [Google Scholar]
- (272).Wellaway CR; Bamborough P; Bernard SG; Chung C-W; Craggs PD; Cutler L; Demont EH; Evans JP; Gordon L; Karamshi B; Lewis AJ; Lindon MJ; Mitchell DJ; Rioja I; Soden PE; Taylor S; Watson RJ; Willis R; Woolven JM; Wyspiańska BS; Kerr WJ; Prinjha RK Structure-Based Design of a Bromodomain and Extraterminal Domain (BET) Inhibitor Selective for the N-Terminal Bromodomains That Retains an Anti-Inflammatory and Antiproliferative Phenotype. J Med Chem 2020, 63 (17), 9020–9044. 10.1021/acs.jmedchem.0c00566. [DOI] [PubMed] [Google Scholar]
- (273).Seal JT; Atkinson SJ; Aylott H; Bamborough P; Chung C-W; Copley RCB; Gordon L; Grandi P; Gray JRJ; Harrison LA; Hayhow TG; Lindon M; Messenger C; Michon A-M; Mitchell D; Preston A; Prinjha RK; Rioja I; Taylor S; Wall ID; Watson RJ; Woolven JM; Demont EH The Optimization of a Novel, Weak Bromo and Extra Terminal Domain (BET) Bromodomain Fragment Ligand to a Potent and Selective Second Bromodomain (BD2) Inhibitor. J Med Chem 2020, 63 (17), 9093–9126. 10.1021/acs.jmedchem.0c00796. [DOI] [PubMed] [Google Scholar]
- (274).Preston A; Atkinson SJ; Bamborough P; Chung C-W; Gordon LJ; Grandi P; Gray JRJ; Harrison LA; Lewis AJ; Lugo D; Messenger C; Michon A-M; Mitchell DJ; Prinjha RK; Rioja I; Seal J; Taylor S; Thesmar P; Wall ID; Watson RJ; Woolven JM; Demont EH GSK973 Is an Inhibitor of the Second Bromodomains (BD2s) of the Bromodomain and Extra-Terminal (BET) Family. ACS Med Chem Lett 2020, 11 (8), 1581–1587. 10.1021/acsmedchemlett.0c00247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (275).Liu Z; Chen H; Wang P; Li Y; Wold EA; Leonard PG; Joseph S; Brasier AR; Tian B; Zhou J Discovery of Orally Bioavailable Chromone Derivatives as Potent and Selective BRD4 Inhibitors: Scaffold Hopping, Optimization, and Pharmacological Evaluation. J Med Chem 2020, 63 (10), 5242–5256. 10.1021/acs.jmedchem.0c00035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (276).Cui H; Divakaran A; Pandey AK; Johnson JA; Zahid H; Hoell ZJ; Ellingson MO; Shi K; Aihara H; Harki DA; Pomerantz WCK Selective N-Terminal BET Bromodomain Inhibitors by Targeting Non-Conserved Residues and Structured Water Displacement*. Angew Chem Int Ed Engl 2021, 60 (3), 1220–1226. 10.1002/anie.202008625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (277).Harrison LA; Atkinson SJ; Bassil A; Chung C-W; Grandi P; Gray JRJ; Levernier E; Lewis A; Lugo D; Messenger C; Michon A-M; Mitchell DJ; Preston A; Prinjha RK; Rioja I; Seal JT; Taylor S; Wall ID; Watson RJ; Woolven JM; Demont EH Identification of a Series of N-Methylpyridine-2-Carboxamides as Potent and Selective Inhibitors of the Second Bromodomain (BD2) of the Bromo and Extra Terminal Domain (BET) Proteins. J Med Chem 2021, 64 (15), 10742–10771. 10.1021/acs.jmedchem.0c02155. [DOI] [PubMed] [Google Scholar]
- (278).Cui H; Carlson AS; Schleiff MA; Divakaran A; Johnson JA; Buchholz CR; Zahid H; Vail NR; Shi K; Aihara H; Harki DA; Miller GP; Topczewski JJ; Pomerantz WCK 4-Methyl-1,2,3-Triazoles as N-Acetyl-Lysine Mimics Afford Potent BET Bromodomain Inhibitors with Improved Selectivity. J Med Chem 2021, 64 (14), 10497–10511. 10.1021/acs.jmedchem.1c00933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (279).Seal JT; Atkinson SJ; Bamborough P; Bassil A; Chung C-W; Foley J; Gordon L; Grandi P; Gray JRJ; Harrison LA; Kruger RG; Matteo JJ; McCabe MT; Messenger C; Mitchell D; Phillipou A; Preston A; Prinjha RK; Rianjongdee F; Rioja I; Taylor S; Wall ID; Watson RJ; Woolven JM; Wyce A; Zhang X-P; Demont EH Fragment-Based Scaffold Hopping: Identification of Potent, Selective, and Highly Soluble Bromo and Extra Terminal Domain (BET) Second Bromodomain (BD2) Inhibitors. J Med Chem 2021, 64 (15), 10772–10805. 10.1021/acs.jmedchem.1c00365. [DOI] [PubMed] [Google Scholar]
- (280).Aylott HE; Atkinson SJ; Bamborough P; Bassil A; Chung C-W; Gordon L; Grandi P; Gray JRJ; Harrison LA; Hayhow TG; Messenger C; Mitchell D; Phillipou A; Preston A; Prinjha RK; Rianjongdee F; Rioja I; Seal JT; Wall ID; Watson RJ; Woolven JM; Demont EH Template-Hopping Approach Leads to Potent, Selective, and Highly Soluble Bromo and Extraterminal Domain (BET) Second Bromodomain (BD2) Inhibitors. J Med Chem 2021, 64 (6), 3249–3281. 10.1021/acs.jmedchem.0c02156. [DOI] [PubMed] [Google Scholar]
- (281).Rianjongdee F; Atkinson SJ; Chung C-W; Grandi P; Gray JRJ; Kaushansky LJ; Medeiros P; Messenger C; Phillipou A; Preston A; Prinjha RK; Rioja I; Satz AL; Taylor S; Wall ID; Watson RJ; Yao G; Demont EH Discovery of a Highly Selective BET BD2 Inhibitor from a DNA-Encoded Library Technology Screening Hit. J Med Chem 2021, 64 (15), 10806–10833. 10.1021/acs.jmedchem.1c00412. [DOI] [PubMed] [Google Scholar]
- (282).Li J; Zhang C; Xu H; Wang C; Dong R; Shen H; Zhuang X; Chen X; Li Q; Lu J; Zhang M; Wu X; Loomes KM; Zhou Y; Zhang Y; Liu J; Xu Y Structure-Based Discovery and Optimization of Furo[3,2-c]Pyridin-4(5H)-One Derivatives as Potent and Second Bromodomain (BD2)-Selective Bromo and Extra Terminal Domain (BET) Inhibitors. J Med Chem 2022, 65 (7), 5760–5799. 10.1021/acs.jmedchem.2c00100. [DOI] [PubMed] [Google Scholar]
- (283).Modukuri RK; Yu Z; Tan Z; Ta HM; Ucisik MN; Jin Z; Anglin JL; Sharma KL; Nyshadham P; Li F; Riehle K; Faver JC; Duong K; Nagarajan S; Simmons N; Palmer SS; Teng M; Young DW; Yi JS; Kim C; Matzuk MM Discovery of Potent BET Bromodomain 1 Stereoselective Inhibitors Using DNA-Encoded Chemical Library Selections. Proc Natl Acad Sci U S A 2022, 119 (22), e2122506119. 10.1073/pnas.2122506119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (284).Jiang W; Hou Q; Xu H; Yang K; Wang X; Zhang K; Zeng Y; Li W; Wang B; Luo G; Zhao X; Shen H; Xu Y; Wu X Discovery of Novel Phenoxyaryl Pyridones as Bromodomain and Extra-Terminal Domain (BET) Inhibitors with High Selectivity for the Second Bromodomain (BD2) to Potentially Treat Acute Myeloid Leukemia. J Med Chem 2024, 67 (2), 1513–1532. 10.1021/acs.jmedchem.3c02104. [DOI] [PubMed] [Google Scholar]
- (285).Chen X; Wu T; Du Z; Kang W; Xu R; Meng F; Liu C; Chen Y; Bao Q; Shen J; You Q; Cao D; Jiang Z; Guo X Discovery of a Brain-Permeable Bromodomain and Extra Terminal Domain (BET) Inhibitor with Selectivity for BD1 for the Treatment of Multiple Sclerosis. Eur J Med Chem 2024, 265, 116080. 10.1016/j.ejmech.2023.116080. [DOI] [PubMed] [Google Scholar]
- (286).Qiao X-P; Wang X-T; Wang S; Mu H-X; Wang Q-S; Chen S-W Discovery of Indole-2-One Derivatives as BRD4 (BD1) Selective Inhibitors. Bioorg Med Chem 2024, 106, 117752. 10.1016/j.bmc.2024.117752. [DOI] [PubMed] [Google Scholar]
- (287).Shi J; Wang Y; Zeng L; Wu Y; Deng J; Zhang Q; Lin Y; Li J; Kang T; Tao M; Rusinova E; Zhang G; Wang C; Zhu H; Yao J; Zeng Y-X; Evers BM; Zhou M-M; Zhou BP Disrupting the Interaction of BRD4 with Diacetylated Twist Suppresses Tumorigenesis in Basal-like Breast Cancer. Cancer Cell 2014, 25 (2), 210–225. 10.1016/j.ccr.2014.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (288).Baud MGJ; Lin-Shiao E; Cardote T; Tallant C; Pschibul A; Chan K-H; Zengerle M; Garcia JR; Kwan TT-L; Ferguson FM; Ciulli A Chemical Biology. A Bump-and-Hole Approach to Engineer Controlled Selectivity of BET Bromodomain Chemical Probes. Science 2014, 346 (6209), 638–641. 10.1126/science.1249830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (289).Tyler DS; Vappiani J; Cañeque T; Lam EYN; Ward A; Gilan O; Chan Y-C; Hienzsch A; Rutkowska A; Werner T; Wagner AJ; Lugo D; Gregory R; Ramirez Molina C; Garton N; Wellaway CR; Jackson S; MacPherson L; Figueiredo M; Stolzenburg S; Bell CC; House C; Dawson S-J; Hawkins ED; Drewes G; Prinjha RK; Rodriguez R; Grandi P; Dawson MA Click Chemistry Enables Preclinical Evaluation of Targeted Epigenetic Therapies. Science 2017, 356 (6345), 1397–1401. 10.1126/science.aal2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (290).Runcie AC; Zengerle M; Chan K-H; Testa A; van Beurden L; Baud MGJ; Epemolu O; Ellis LCJ; Read KD; Coulthard V; Brien A; Ciulli A Optimization of a “Bump-and-Hole” Approach to Allele-Selective BET Bromodomain Inhibition. Chem Sci 2018, 9 (9), 2452–2468. 10.1039/c7sc02536j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (291).Lamonica JM; Deng W; Kadauke S; Campbell AE; Gamsjaeger R; Wang H; Cheng Y; Billin AN; Hardison RC; Mackay JP; Blobel GA Bromodomain Protein Brd3 Associates with Acetylated GATA1 to Promote Its Chromatin Occupancy at Erythroid Target Genes. Proc Natl Acad Sci U S A 2011, 108 (22), E159–68. 10.1073/pnas.1102140108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (292).Miller TCR; Simon B; Rybin V; Grötsch H; Curtet S; Khochbin S; Carlomagno T; Müller CW A Bromodomain-DNA Interaction Facilitates Acetylation-Dependent Bivalent Nucleosome Recognition by the BET Protein BRDT. Nat Commun 2016, 7, 13855. 10.1038/ncomms13855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (293).Watson RJ; Bamborough P; Barnett H; Chung C-W; Davis R; Gordon L; Grandi P; Petretich M; Phillipou A; Prinjha RK; Rioja I; Soden P; Werner T; Demont EH GSK789: A Selective Inhibitor of the First Bromodomains (BD1) of the Bromo and Extra Terminal Domain (BET) Proteins. J Med Chem 2020, 63 (17), 9045–9069. 10.1021/acs.jmedchem.0c00614. [DOI] [PubMed] [Google Scholar]
- (294).Anders L; Guenther MG; Qi J; Fan ZP; Marineau JJ; Rahl PB; Lovén J; Sigova AA; Smith WB; Lee TI; Bradner JE; Young RA Genome-Wide Localization of Small Molecules. Nat Biotechnol 2014, 32 (1), 92–96. 10.1038/nbt.2776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (295).Zhang X; Crowley VM; Wucherpfennig TG; Dix MM; Cravatt BF Electrophilic PROTACs That Degrade Nuclear Proteins by Engaging DCAF16. Nat Chem Biol 2019, 15 (7), 737–746. 10.1038/s41589-019-0279-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (296).Pfaff P; Samarasinghe KTG; Crews CM; Carreira EM Reversible Spatiotemporal Control of Induced Protein Degradation by Bistable PhotoPROTACs. ACS Cent Sci 2019, 5 (10), 1682–1690. 10.1021/acscentsci.9b00713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (297).Tong B; Luo M; Xie Y; Spradlin JN; Tallarico JA; McKenna JM; Schirle M; Maimone TJ; Nomura DK Bardoxolone Conjugation Enables Targeted Protein Degradation of BRD4. Sci Rep 2020, 10 (1), 15543. 10.1038/s41598-020-72491-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (298).Imaide S; Riching KM; Makukhin N; Vetma V; Whitworth C; Hughes SJ; Trainor N; Mahan SD; Murphy N; Cowan AD; Chan K-H; Craigon C; Testa A; Maniaci C; Urh M; Daniels DL; Ciulli A Trivalent PROTACs Enhance Protein Degradation via Combined Avidity and Cooperativity. Nat Chem Biol 2021, 17 (11), 1157–1167. 10.1038/s41589-021-00878-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (299).Bond AG; Craigon C; Chan K-H; Testa A; Karapetsas A; Fasimoye R; Macartney T; Blow JJ; Alessi DR; Ciulli A Development of BromoTag: A “Bump-and-Hole”-PROTAC System to Induce Potent, Rapid, and Selective Degradation of Tagged Target Proteins. J Med Chem 2021, 64 (20), 15477–15502. 10.1021/acs.jmedchem.1c01532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (300).Ichikawa S; Flaxman HA; Xu W; Vallavoju N; Lloyd HC; Wang B; Shen D; Pratt MR; Woo CM The E3 Ligase Adapter Cereblon Targets the C-Terminal Cyclic Imide Degron. Nature 2022, 610 (7933), 775–782. 10.1038/s41586-022-05333-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (301).Du G; Jiang J; Henning NJ; Safaee N; Koide E; Nowak RP; Donovan KA; Yoon H; You I; Yue H; Eleuteri NA; He Z; Li Z; Huang HT; Che J; Nabet B; Zhang T; Fischer ES; Gray NS Exploring the Target Scope of KEAP1 E3 Ligase-Based PROTACs. Cell Chem Biol 2022, 29 (10), 1470–1481.e31. 10.1016/j.chembiol.2022.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (302).Sarott RC; You I; Li Y-D; Toenjes ST; Donovan KA; Seo P; Ordonez M; Byun WS; Hassan MM; Wachter F; Chouchani ET; Słabicki M; Fischer ES; Ebert BL; Hinshaw SM; Gray NS Chemical Specification of E3 Ubiquitin Ligase Engagement by Cysteine-Reactive Chemistry. J Am Chem Soc 2023, 145 (40), 21937–21944. 10.1021/jacs.3c06622. [DOI] [PubMed] [Google Scholar]
- (303).Forte N; Dovala D; Hesse MJ; McKenna JM; Tallarico JA; Schirle M; Nomura DK Targeted Protein Degradation through E2 Recruitment. ACS Chem Biol 2023, 18 (4), 897–904. 10.1021/acschembio.3c00040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (304).Shergalis AG; Marin VL; Rhee DY; Senaweera S; McCloud RL; Ronau JA; Hutchins CW; McLoughlin S; Woller KR; Warder SE; Vasudevan A; Reitsma JM CRISPR Screen Reveals BRD2/4 Molecular Glue-like Degrader via Recruitment of DCAF16. ACS Chem Biol 2023, 18 (2), 331–339. 10.1021/acschembio.2c00747. [DOI] [PubMed] [Google Scholar]
- (305).Nowak RP; Ragosta L; Huerta F; Liu H; Ficarro SB; Cruite JT; Metivier RJ; Donovan KA; Marto JA; Fischer ES; Zerfas BL; Jones LH Development of a Covalent Cereblon-Based PROTAC Employing a Fluorosulfate Warhead. RSC Chem Biol 2023, 4 (11), 906–912. 10.1039/d3cb00103b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (306).Xue G; Xie J; Hinterndorfer M; Cigler M; Dötsch L; Imrichova H; Lampe P; Cheng X; Adariani SR; Winter GE; Waldmann H Discovery of a Drug-like, Natural Product-Inspired DCAF11 Ligand Chemotype. Nat Commun 2023, 14 (1), 7908. 10.1038/s41467-023-43657-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (307).Li Y-D; Ma MW; Hassan MM; Hunkeler M; Teng M; Puvar K; Rutter JC; Lumpkin RJ; Sandoval B; Jin CY; Schmoker AM; Ficarro SB; Cheong H; Metivier RJ; Wang MY; Xu S; Byun WS; Groendyke BJ; You I; Sigua LH; Tavares I; Zou C; Tsai JM; Park PMC; Yoon H; Majewski FC; Sperling HT; Marto JA; Qi J; Nowak RP; Donovan KA; Słabicki M; Gray NS; Fischer ES; Ebert BL Template-Assisted Covalent Modification Underlies Activity of Covalent Molecular Glues. Nat Chem Biol 2024, 20 (12), 1640–1649. 10.1038/s41589-024-01668-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (308).Hsia O; Hinterndorfer M; Cowan AD; Iso K; Ishida T; Sundaramoorthy R; Nakasone MA; Imrichova H; Schätz C; Rukavina A; Husnjak K; Wegner M; Correa-Sáez A; Craigon C; Casement R; Maniaci C; Testa A; Kaulich M; Dikic I; Winter GE; Ciulli A Targeted Protein Degradation via Intramolecular Bivalent Glues. Nature 2024, 627 (8002), 204–211. 10.1038/s41586-024-07089-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (309).Lim M; Do Cong T; Orr LM; Toriki ES; Kile AC; Papatzimas JW; Lee E; Lin Y; Nomura DK DCAF16-Based Covalent Handle for the Rational Design of Monovalent Degraders. ACS Cent Sci 2024, 10 (7), 1318–1331. 10.1021/acscentsci.4c00286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (310).Hong SH; Divakaran A; Osa A; Huang OW; Wertz IE; Nomura DK Exploiting the Cullin E3 Ligase Adaptor Protein SKP1 for Targeted Protein Degradation. ACS Chem Biol 2024, 19 (2), 442–450. 10.1021/acschembio.3c00642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (311).Parker GS; Toth JI; Fish S; Blanco GA; Kampert T; Li X; Yang L; Stumpf CR; Steadman K; Jamborcic A; Chien S; Daniele E; Dearie A; Leriche G; Bailey S; Thompson PA Discovery of Monovalent Direct Degraders of BRD4 That Act Via the Recruitment of DCAF11. Mol Cancer Ther 2024, 23, 1446. 10.1158/1535-7163.MCT-24-0219. [DOI] [PubMed] [Google Scholar]
- (312).Basu AA; Zhang C; Riha IA; Magassa A; Campos MA; Caldwell AG; Ko F; Zhang X A CRISPR Activation Screen Identifies FBXO22 Supporting Targeted Protein Degradation. Nat Chem Biol 2024, 20, 1608–1616. 10.1038/s41589-024-01655-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (313).Balzarini M; Tong J; Gui W; Jayalath IM; Schell B-B; Kodadek T Recruitment to the Proteasome Is Necessary but Not Sufficient for Chemically Induced, Ubiquitin-Independent Degradation of Native Proteins. ACS Chem Biol 2024, 19 (11), 2323–2335. 10.1021/acschembio.4c00422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (314).Serebrenik YV; Mani D; Maujean T; Burslem GM; Shalem O Pooled Endogenous Protein Tagging and Recruitment for Systematic Profiling of Protein Function. Cell genomics 2024, 4 (10), 100651. 10.1016/j.xgen.2024.100651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (315).Ichikawa S; Payne NC; Xu W; Chang C-F; Vallavoju N; Frome S; Flaxman HA; Mazitschek R; Woo CM The Cyclimids: Degron-Inspired Cereblon Binders for Targeted Protein Degradation. Cell Chem Biol 2024, 31 (6), 1162–1175.e10. 10.1016/j.chembiol.2024.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (316).Siriwardena SU; Munkanatta Godage DNP; Shoba VM; Lai S; Shi M; Wu P; Chaudhary SK; Schreiber SL; Choudhary A Phosphorylation-Inducing Chimeric Small Molecules. J Am Chem Soc 2020, 142 (33), 14052–14057. 10.1021/jacs.0c05537. [DOI] [PubMed] [Google Scholar]
- (317).Ma B; Khan KS; Xu T; Xeque Amada J; Guo Z; Huang Y; Yan Y; Lam H; Cheng AS-L; Ng BW-L Targeted Protein O-GlcNAcylation Using Bifunctional Small Molecules. J Am Chem Soc 2024, 146 (14), 9779–9789. 10.1021/jacs.3c14380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (318).Gibson WJ; Sadagopan A; Shoba VM; Choudhary A; Meyerson M; Schreiber SL Bifunctional Small Molecules That Induce Nuclear Localization and Targeted Transcriptional Regulation. J Am Chem Soc 2023, 145 (48), 26028–26037. 10.1021/jacs.3c06179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (319).Raina K; Forbes CD; Stronk R; Rappi JP; Eastman KJ; Zaware N; Yu X; Li H; Bhardwaj A; Gerritz SW; Forgione M; Hundt A; King MP; Posner ZM; Correia AD; McGovern A; Puleo DE; Chenard R; Mousseau JJ; Vergara JI; Garvin E; Macaluso J; Martin M; Bassoli K; Jones K; Garcia M; Howard K; Yaggi M; Smith LM; Chen JM; Mayfield AB; De Leon CA; Hines J; Kayser-Bricker KJ; Crews CM Regulated Induced Proximity Targeting Chimeras-RIPTACs-A Heterobifunctional Small Molecule Strategy for Cancer Selective Therapies. Cell Chem Biol 2024, 31 (8), 1490–1502.e42. 10.1016/j.chembiol.2024.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (320).Erwin GS; Grieshop MP; Ali A; Qi J; Lawlor M; Kumar D; Ahmad I; McNally A; Teider N; Worringer K; Sivasankaran R; Syed DN; Eguchi A; Ashraf M; Jeffery J; Xu M; Park PMC; Mukhtar H; Srivastava AK; Faruq M; Bradner JE; Ansari AZ Synthetic Transcription Elongation Factors License Transcription across Repressive Chromatin. Science 2017, 358 (6370), 1617–1622. 10.1126/science.aan6414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (321).Gourisankar S; Krokhotin A; Ji W; Liu X; Chang C-Y; Kim SH; Li Z; Wenderski W; Simanauskaite JM; Yang H; Vogel H; Zhang T; Green MR; Gray NS; Crabtree GR Rewiring Cancer Drivers to Activate Apoptosis. Nature 2023, 620 (7973), 417–425. 10.1038/s41586-023-06348-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (322).Rivera VM; Clackson T; Natesan S; Pollock R; Amara JF; Keenan T; Magari SR; Phillips T; Courage NL; Cerasoli F; Holt DA; Gilman M A Humanized System for Pharmacologic Control of Gene Expression. Nat Med 1996, 2 (9), 1028–1032. 10.1038/nm0996-1028. [DOI] [PubMed] [Google Scholar]
- (323).Ho SN; Biggar SR; Spencer DM; Schreiber SL; Crabtree GR Dimeric Ligands Define a Role for Transcriptional Activation Domains in Reinitiation. Nature 1996, 382 (6594), 822–826. 10.1038/382822a0. [DOI] [PubMed] [Google Scholar]
- (324).Belshaw PJ; Ho SN; Crabtree GR; Schreiber SL Controlling Protein Association and Subcellular Localization with a Synthetic Ligand That Induces Heterodimerization of Proteins. Proc Natl Acad Sci U S A 1996, 93 (10), 4604–4607. 10.1073/pnas.93.10.4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (325).Hathaway NA; Bell O; Hodges C; Miller EL; Neel DS; Crabtree GR Dynamics and Memory of Heterochromatin in Living Cells. Cell 2012, 149 (7), 1447–1460. 10.1016/j.cell.2012.03.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (326).Kadoch C; Williams RT; Calarco JP; Miller EL; Weber CM; Braun SMG; Pulice JL; Chory EJ; Crabtree GR Dynamics of BAF-Polycomb Complex Opposition on Heterochromatin in Normal and Oncogenic States. Nat Genet 2017, 49 (2), 213–222. 10.1038/ng.3734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (327).Braun SMG; Kirkland JG; Chory EJ; Husmann D; Calarco JP; Crabtree GR Rapid and Reversible Epigenome Editing by Endogenous Chromatin Regulators. Nat Commun 2017, 8 (1), 560. 10.1038/s41467-017-00644-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (328).Butler KV; Chiarella AM; Jin J; Hathaway NA Targeted Gene Repression Using Novel Bifunctional Molecules to Harness Endogenous Histone Deacetylation Activity. ACS Synth Biol 2018, 7 (1), 38–45. 10.1021/acssynbio.7b00295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (329).Chiarella AM; Butler KV; Gryder BE; Lu D; Wang TA; Yu X; Pomella S; Khan J; Jin J; Hathaway NA Dose-Dependent Activation of Gene Expression Is Achieved Using CRISPR and Small Molecules That Recruit Endogenous Chromatin Machinery. Nat Biotechnol 2020, 38 (1), 50–55. 10.1038/s41587-019-0296-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (330).Dervan PB; Edelson BS Recognition of the DNA Minor Groove by Pyrrole-Imidazole Polyamides. Curr Opin Struct Biol 2003, 13 (3), 284–299. 10.1016/s0959-440x(03)00081-2. [DOI] [PubMed] [Google Scholar]
- (331).Mapp AK; Ansari AZ; Ptashne M; Dervan PB Activation of Gene Expression by Small Molecule Transcription Factors. Proc Natl Acad Sci U S A 2000, 97 (8), 3930–3935. 10.1073/pnas.97.8.3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (332).Arora PS; Ansari AZ; Best TP; Ptashne M; Dervan PB Design of Artificial Transcriptional Activators with Rigid Poly-L-Proline Linkers. J Am Chem Soc 2002, 124 (44), 13067–13071. 10.1021/ja0208355. [DOI] [PubMed] [Google Scholar]
- (333).Ansari AZ; Mapp AK; Nguyen DH; Dervan PB; Ptashne M Towards a Minimal Motif for Artificial Transcriptional Activators. Chem Biol 2001, 8 (6), 583–592. 10.1016/s1074-5521(01)00037-0. [DOI] [PubMed] [Google Scholar]
- (334).Xiao X; Yu P; Lim H-S; Sikder D; Kodadek T Design and Synthesis of a Cell-Permeable Synthetic Transcription Factor Mimic. J Comb Chem 2007, 9 (4), 592–600. 10.1021/cc070023a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (335).Xiao X; Yu P; Lim H-S; Sikder D; Kodadek T A Cell-Permeable Synthetic Transcription Factor Mimic. Angew Chem Int Ed Engl 2007, 46 (16), 2865–2868. 10.1002/anie.200604485. [DOI] [PubMed] [Google Scholar]
- (336).Mohammed A; Waddell MB; Sutkeviciute I; Danda A; Philips SJ; Lang W; Slavish PJ; Kietlinska SJ; Kaulage M; Sourav D; Ansari AZ Domain-Selective BET Ligands Yield Next-Generation Synthetic Genome Readers/Regulators with Nonidentical Cellular Functions. J Am Chem Soc 2023, 10, 1021. 10.1021/jacs.3c06297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (337).Li Y; Lu Y; Polak U; Lin K; Shen J; Farmer J; Seyer L; Bhalla AD; Rozwadowska N; Lynch DR; Butler JS; Napierala M Expanded GAA Repeats Impede Transcription Elongation through the FXN Gene and Induce Transcriptional Silencing That Is Restricted to the FXN Locus. Hum Mol Genet 2015, 24 (24), 6932–6943. 10.1093/hmg/ddv397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (338).Yandim C; Natisvili T; Festenstein R Gene Regulation and Epigenetics in Friedreich’s Ataxia. J Neurochem 2013, 126 Suppl 1, 21–42. 10.1111/jnc.12254. [DOI] [PubMed] [Google Scholar]
- (339).Liszczak GP; Brown ZZ; Kim SH; Oslund RC; David Y; Muir TW Genomic Targeting of Epigenetic Probes Using a Chemically Tailored Cas9 System. Proc Natl Acad Sci U S A 2017, 114 (4), 681–686. 10.1073/pnas.1615723114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (340).Umaña JD; Wasserman SR; Song L; Goel AA; Yu X; Jin J; Hathaway NA Chemical Epigenetic Regulation of Adeno-Associated Virus Delivered Transgenes. Hum Gene Ther 2023, 34 (17–18), 947–957. 10.1089/hum.2023.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (341).Wasserman SR; Muron S; Lee HR; Routh ML; Hepperla AJ; Scoville DM; Huber A; Umana JD; Pereira NE; Foley CA; James LI; Hathaway NA Dynamic Activation of RAAV Transgene Expression by a Small Molecule That Recruits Endogenous Transcriptional Machinery. Nucleic Acids Res 2025, 53 (8), gkaf345. 10.1093/nar/gkaf345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (342).Hatzi K; Melnick A Breaking Bad in the Germinal Center: How Deregulation of BCL6 Contributes to Lymphomagenesis. Trends Mol Med 2014, 20 (6), 343–352. 10.1016/j.molmed.2014.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (343).Kerres N; Steurer S; Schlager S; Bader G; Berger H; Caligiuri M; Dank C; Engen JR; Ettmayer P; Fischerauer B; Flotzinger G; Gerlach D; Gerstberger T; Gmaschitz T; Greb P; Han B; Heyes E; Iacob RE; Kessler D; Kölle H; Lamarre L; Lancia DR; Lucas S; Mayer M; Mayr K; Mischerikow N; Mück K; Peinsipp C; Petermann O; Reiser U; Rudolph D; Rumpel K; Salomon C; Scharn D; Schnitzer R; Schrenk A; Schweifer N; Thompson D; Traxler E; Varecka R; Voss T; Weiss-Puxbaum A; Winkler S; Zheng X; Zoephel A; Kraut N; McConnell D; Pearson M; Koegl M Chemically Induced Degradation of the Oncogenic Transcription Factor BCL6. Cell Rep 2017, 20 (12), 2860–2875. 10.1016/j.celrep.2017.08.081. [DOI] [PubMed] [Google Scholar]
- (344).Słabicki M; Yoon H; Koeppel J; Nitsch L; Roy Burman SS; Di Genua C; Donovan KA; Sperling AS; Hunkeler M; Tsai JM; Sharma R; Guirguis A; Zou C; Chudasama P; Gasser JA; Miller PG; Scholl C; Fröhling S; Nowak RP; Fischer ES; Ebert BL Small-Molecule-Induced Polymerization Triggers Degradation of BCL6. Nature 2020, 588 (7836), 164–168. 10.1038/s41586-020-2925-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (345).Sarott RC; Gourisankar S; Karim B; Nettles S; Yang H; Dwyer BG; Simanauskaite JM; Tse J; Abuzaid H; Krokhotin A; Zhang T; Hinshaw SM; Green MR; Crabtree GR; Gray NS Relocalizing Transcriptional Kinases to Activate Apoptosis. Science 2024, 386 (6717), eadl5361. 10.1126/science.adl5361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (346).Zhao L; Zhou S; Gustafsson J-Å Nuclear Receptors: Recent Drug Discovery for Cancer Therapies. Endocr Rev 2019, 40 (5), 1207–1249. 10.1210/er.2018-00222. [DOI] [PubMed] [Google Scholar]
- (347).Bushweller JH Targeting Transcription Factors in Cancer - from Undruggable to Reality. Nat Rev Cancer 2019, 19 (11), 611–624. 10.1038/s41568-019-0196-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (348).Henley MJ; Koehler AN Advances in Targeting “undruggable” Transcription Factors with Small Molecules. Nat Rev Drug Discov 2021, 20 (9), 669–688. 10.1038/s41573-021-00199-0. [DOI] [PubMed] [Google Scholar]
- (349).Boike L; Henning NJ; Nomura DK Advances in Covalent Drug Discovery. Nat Rev Drug Discov 2022, 21 (12), 881–898. 10.1038/s41573-022-00542-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (350).Niphakis MJ; Cravatt BF Ligand Discovery by Activity-Based Protein Profiling. Cell Chem Biol 2024, 31 (9), 1636–1651. 10.1016/j.chembiol.2024.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (351).Zhang X; Cravatt BF Chemical Proteomics–Guided Discovery of Covalent Ligands for Cancer Proteins. Annu Rev Cancer Biol 2024, 8 (1), 155–175. 10.1146/annurev-cancerbio-061421-041946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (352).Won SJ; Zhang Y; Reinhardt CJ; Hargis LM; MacRae NS; DeMeester KE; Njomen E; Remsberg JR; Melillo B; Cravatt BF; Erb MA Redirecting the Pioneering Function of FOXA1 with Covalent Small Molecules. Mol Cell 2024, 84 (21), 4125–4141.e10. 10.1016/j.molcel.2024.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (353).Gowans FA; Forte N; Hatcher J; Huang OW; Wang Y; Altamirano Poblano BE; Wertz IE; Nomura DK Covalent Degrader of the Oncogenic Transcription Factor β-Catenin. J Am Chem Soc 2024, 146 (24), 16856–16865. 10.1021/jacs.4c05174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (354).Ruprecht B; Wei L; Zheng L; Bodea S; Mo X; Maschberger M; Stoehr G; Hahne H; Cornella-Taracido I; Chi A Chemoproteomic Profiling to Identify Activity Changes and Functional Inhibitors of DNA-Binding Proteins. Cell Chem Biol 2022, 29 (11), 1639–1648.e4. 10.1016/j.chembiol.2022.10.008. [DOI] [PubMed] [Google Scholar]
- (355).Boike L; Cioffi AG; Majewski FC; Co J; Henning NJ; Jones MD; Liu G; McKenna JM; Tallarico JA; Schirle M; Nomura DK Discovery of a Functional Covalent Ligand Targeting an Intrinsically Disordered Cysteine within MYC. Cell Chem Biol 2021, 28 (1), 4–13.e17. 10.1016/j.chembiol.2020.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (356).Takahashi M; Chong HB; Zhang S; Yang T-Y; Lazarov MJ; Harry S; Maynard M; Hilbert B; White RD; Murrey HE; Tsou C-C; Vordermark K; Assaad J; Gohar M; Dürr BR; Richter M; Patel H; Kryukov G; Brooijmans N; Alghali ASO; Rubio K; Villanueva A; Zhang J; Ge M; Makram F; Griesshaber H; Harrison D; Koglin A-S; Ojeda S; Karakyriakou B; Healy A; Popoola G; Rachmin I; Khandelwal N; Neil JR; Tien P-C; Chen N; Hosp T; van den Ouweland S; Hara T; Bussema L; Dong R; Shi L; Rasmussen MQ; Domingues AC; Lawless A; Fang J; Yoda S; Nguyen LP; Reeves SM; Wakefield FN; Acker A; Clark SE; Dubash T; Kastanos J; Oh E; Fisher DE; Maheswaran S; Haber DA; Boland GM; Sade-Feldman M; Jenkins RW; Hata AN; Bardeesy NM; Suvà ML; Martin BR; Liau BB; Ott CJ; Rivera MN; Lawrence MS; Bar-Peled L DrugMap: A Quantitative Pan-Cancer Analysis of Cysteine Ligandability. Cell 2024, 187 (10), 2536–2556.e30. 10.1016/j.cell.2024.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (357).Cirillo LA; McPherson CE; Bossard P; Stevens K; Cherian S; Shim EY; Clark KL; Burley SK; Zaret KS Binding of the Winged-Helix Transcription Factor HNF3 to a Linker Histone Site on the Nucleosome. EMBO J 1998, 17 (1), 244–254. 10.1093/emboj/17.1.244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (358).Cirillo LA; Lin FR; Cuesta I; Friedman D; Jarnik M; Zaret KS Opening of Compacted Chromatin by Early Developmental Transcription Factors HNF3 (FoxA) and GATA-4. Mol Cell 2002, 9 (2), 279–289. 10.1016/s1097-2765(02)00459-8. [DOI] [PubMed] [Google Scholar]
- (359).Adams EJ; Karthaus WR; Hoover E; Liu D; Gruet A; Zhang Z; Cho H; DiLoreto R; Chhangawala S; Liu Y; Watson PA; Davicioni E; Sboner A; Barbieri CE; Bose R; Leslie CS; Sawyers CL FOXA1 Mutations Alter Pioneering Activity, Differentiation and Prostate Cancer Phenotypes. Nature 2019, 571 (7765), 408–412. 10.1038/s41586-019-1318-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (360).Parolia A; Cieslik M; Chu S-C; Xiao L; Ouchi T; Zhang Y; Wang X; Vats P; Cao X; Pitchiaya S; Su F; Wang R; Feng FY; Wu Y-M; Lonigro RJ; Robinson DR; Chinnaiyan AM Distinct Structural Classes of Activating FOXA1 Alterations in Advanced Prostate Cancer. Nature 2019, 571 (7765), 413–418. 10.1038/s41586-019-1347-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (361).Arruabarrena-Aristorena A; Maag JLV; Kittane S; Cai Y; Karthaus WR; Ladewig E; Park J; Kannan S; Ferrando L; Cocco E; Ho SY; Tan DS; Sallaku M; Wu F; Acevedo B; Selenica P; Ross DS; Witkin M; Sawyers CL; Reis-Filho JS; Verma CS; Jauch R; Koche R; Baselga J; Razavi P; Toska E; Scaltriti M FOXA1 Mutations Reveal Distinct Chromatin Profiles and Influence Therapeutic Response in Breast Cancer. Cancer Cell 2020, 38 (4), 534–550.e9. 10.1016/j.ccell.2020.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (362).Beroukhim R; Mermel CH; Porter D; Wei G; Raychaudhuri S; Donovan J; Barretina J; Boehm JS; Dobson J; Urashima M; Mc Henry KT; Pinchback RM; Ligon AH; Cho Y-J; Haery L; Greulich H; Reich M; Winckler W; Lawrence MS; Weir BA; Tanaka KE; Chiang DY; Bass AJ; Loo A; Hoffman C; Prensner J; Liefeld T; Gao Q; Yecies D; Signoretti S; Maher E; Kaye FJ; Sasaki H; Tepper JE; Fletcher JA; Tabernero J; Baselga J; Tsao M-S; Demichelis F; Rubin MA; Janne PA; Daly MJ; Nucera C; Levine RL; Ebert BL; Gabriel S; Rustgi AK; Antonescu CR; Ladanyi M; Letai A; Garraway LA; Loda M; Beer DG; True LD; Okamoto A; Pomeroy SL; Singer S; Golub TR; Lander ES; Getz G; Sellers WR; Meyerson M The Landscape of Somatic Copy-Number Alteration across Human Cancers. Nature 2010, 463 (7283), 899–905. 10.1038/nature08822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (363).Kryukov GV; Wilson FH; Ruth JR; Paulk J; Tsherniak A; Marlow SE; Vazquez F; Weir BA; Fitzgerald ME; Tanaka M; Bielski CM; Scott JM; Dennis C; Cowley GS; Boehm JS; Root DE; Golub TR; Clish CB; Bradner JE; Hahn WC; Garraway LA MTAP Deletion Confers Enhanced Dependency on the PRMT5 Arginine Methyltransferase in Cancer Cells. Science 2016, 351 (6278), 1214–1218. 10.1126/science.aad5214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (364).Mavrakis KJ; McDonald ER; Schlabach MR; Billy E; Hoffman GR; deWeck A; Ruddy DA; Venkatesan K; Yu J; McAllister G; Stump M; deBeaumont R; Ho S; Yue Y; Liu Y; Yan-Neale Y; Yang G; Lin F; Yin H; Gao H; Kipp DR; Zhao S; McNamara JT; Sprague ER; Zheng B; Lin Y; Cho YS; Gu J; Crawford K; Ciccone D; Vitari AC; Lai A; Capka V; Hurov K; Porter JA; Tallarico J; Mickanin C; Lees E; Pagliarini R; Keen N; Schmelzle T; Hofmann F; Stegmeier F; Sellers WR Disordered Methionine Metabolism in MTAP/CDKN2A-Deleted Cancers Leads to Dependence on PRMT5. Science 2016, 351 (6278), 1208–1213. 10.1126/science.aad5944. [DOI] [PubMed] [Google Scholar]
- (365).Marjon K; Cameron MJ; Quang P; Clasquin MF; Mandley E; Kunii K; McVay M; Choe S; Kernytsky A; Gross S; Konteatis Z; Murtie J; Blake ML; Travins J; Dorsch M; Biller SA; Marks KM MTAP Deletions in Cancer Create Vulnerability to Targeting of the MAT2A/PRMT5/RIOK1 Axis. Cell Rep 2016, 15 (3), 574–587. 10.1016/j.celrep.2016.03.043. [DOI] [PubMed] [Google Scholar]
- (366).Smith CR; Aranda R; Bobinski TP; Briere DM; Burns AC; Christensen JG; Clarine J; Engstrom LD; Gunn RJ; Ivetac A; Jean-Baptiste R; Ketcham JM; Kobayashi M; Kuehler J; Kulyk S; Lawson JD; Moya K; Olson P; Rahbaek L; Thomas NC; Wang X; Waters LM; Marx MA Fragment-Based Discovery of MRTX1719, a Synthetic Lethal Inhibitor of the PRMT5•MTA Complex for the Treatment of MTAP-Deleted Cancers. J Med Chem 2022, 65 (3), 1749–1766. 10.1021/acs.jmedchem.1c01900. [DOI] [PubMed] [Google Scholar]
- (367).Engstrom LD; Aranda R; Waters L; Moya K; Bowcut V; Vegar L; Trinh D; Hebbert A; Smith CR; Kulyk S; Lawson JD; He L; Hover LD; Fernandez-Banet J; Hallin J; Vanderpool D; Briere DM; Blaj A; Marx MA; Rodon J; Offin M; Arbour KC; Johnson ML; Kwiatkowski DJ; Jänne PA; Haddox CL; Papadopoulos KP; Henry JT; Leventakos K; Christensen JG; Shazer R; Olson P MRTX1719 Is an MTA-Cooperative PRMT5 Inhibitor That Exhibits Synthetic Lethality in Preclinical Models and Patients with MTAP-Deleted Cancer. Cancer Discov 2023, 13 (11), 2412–2431. 10.1158/2159-8290.CD-23-0669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (368).Cottrell KM; Briggs KJ; Whittington DA; Jahic H; Ali JA; Davis CB; Gong S; Gotur D; Gu L; McCarren P; Tonini MR; Tsai A; Wilker EW; Yuan H; Zhang M; Zhang W; Huang A; Maxwell JP Discovery of TNG908: A Selective, Brain Penetrant, MTA-Cooperative PRMT5 Inhibitor That Is Synthetically Lethal with MTAP-Deleted Cancers. J Med Chem 2024, 67 (8), 6064–6080. 10.1021/acs.jmedchem.4c00133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (369).Belmontes B; Slemmons KK; Su C; Liu S; Policheni AN; Moriguchi J; Tan H; Xie F; Aiello DA; Yang Y; Lazaro R; Aeffner F; Rees MG; Ronan MM; Roth JA; Vestergaard M; Cowland S; Andersson J; Sarvary I; Chen Q; Sharma P; Lopez P; Tamayo N; Pettus LH; Ghimire-Rijal S; Mukund S; Allen JR; DeVoss J; Coxon A; Rodon J; Ghiringhelli F; Penel N; Prenen H; Glad S; Chuang C-H; Keyvanjah K; Townsley DM; Butler JR; Bourbeau MP; Caenepeel S; Hughes PE AMG 193, a Clinical Stage MTA-Cooperative PRMT5 Inhibitor, Drives Antitumor Activity Preclinically and in Patients With MTAP-Deleted Cancers. Cancer Discov 2025, 15, 139–161. 10.1158/2159-8290.CD-24-0887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (370).Tee W-W; Pardo M; Theunissen TW; Yu L; Choudhary JS; Hajkova P; Surani MA Prmt5 Is Essential for Early Mouse Development and Acts in the Cytoplasm to Maintain ES Cell Pluripotency. Genes Dev 2010, 24 (24), 2772–2777. 10.1101/gad.606110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (371).Meister G; Bühler D; Pillai R; Lottspeich F; Fischer U A Multiprotein Complex Mediates the ATP-Dependent Assembly of Spliceosomal U SnRNPs. Nat Cell Biol 2001, 3 (11), 945–949. 10.1038/ncb1101-945. [DOI] [PubMed] [Google Scholar]
- (372).Friesen WJ; Paushkin S; Wyce A; Massenet S; Pesiridis GS; Van Duyne G; Rappsilber J; Mann M; Dreyfuss G The Methylosome, a 20S Complex Containing JBP1 and PICln, Produces Dimethylarginine-Modified Sm Proteins. Mol Cell Biol 2001, 21 (24), 8289–8300. 10.1128/MCB.21.24.8289-8300.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (373).Meister G; Eggert C; Bühler D; Brahms H; Kambach C; Fischer U Methylation of Sm Proteins by a Complex Containing PRMT5 and the Putative U SnRNP Assembly Factor PICln. Curr Biol 2001, 11 (24), 1990–1994. 10.1016/s0960-9822(01)00592-9. [DOI] [PubMed] [Google Scholar]
- (374).Brahms H; Meheus L; de Brabandere V; Fischer U; Lührmann R Symmetrical Dimethylation of Arginine Residues in Spliceosomal Sm Protein B/B’ and the Sm-like Protein LSm4, and Their Interaction with the SMN Protein. RNA 2001, 7 (11), 1531–1542. 10.1017/s135583820101442x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (375).Boisvert F-M; Cote J; Boulanger M-C; Cleroux P; Bachand F; Autexier C; Richard S Symmetrical Dimethylarginine Methylation Is Required for the Localization of SMN in Cajal Bodies and Pre-MRNA Splicing. J Cell Biol 2002, 159 (6), 957–969. 10.1083/jcb.200207028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (376).Pal S; Vishwanath SN; Erdjument-Bromage H; Tempst P; Sif S Human SWI/SNF-Associated PRMT5 Methylates Histone H3 Arginine 8 and Negatively Regulates Expression of ST7 and NM23 Tumor Suppressor Genes. Mol Cell Biol 2004, 24 (21), 9630–9645. 10.1128/MCB.24.21.9630-9645.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (377).Dacwag CS; Ohkawa Y; Pal S; Sif S; Imbalzano AN The Protein Arginine Methyltransferase Prmt5 Is Required for Myogenesis Because It Facilitates ATP-Dependent Chromatin Remodeling. Mol Cell Biol 2007, 27 (1), 384–394. 10.1128/MCB.01528-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (378).Lacroix M; El Messaoudi S; Rodier G; Le Cam A; Sardet C; Fabbrizio E The Histone-Binding Protein COPR5 Is Required for Nuclear Functions of the Protein Arginine Methyltransferase PRMT5. EMBO Rep 2008, 9 (5), 452–458. 10.1038/embor.2008.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (379).Mulvaney KM; Blomquist C; Acharya N; Li R; Ranaghan MJ; O’Keefe M; Rodriguez DJ; Young MJ; Kesar D; Pal D; Stokes M; Nelson AJ; Jain SS; Yang A; Mullin-Bernstein Z; Columbus J; Bozal FK; Skepner A; Raymond D; LaRussa S; McKinney DC; Freyzon Y; Baidi Y; Porter D; Aguirre AJ; Ianari A; McMillan B; Sellers WR Molecular Basis for Substrate Recruitment to the PRMT5 Methylosome. Mol Cell 2021, 81 (17), 3481–3495.e7. 10.1016/j.molcel.2021.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (380).Chan-Penebre E; Kuplast KG; Majer CR; Boriack-Sjodin PA; Wigle TJ; Johnston LD; Rioux N; Munchhof MJ; Jin L; Jacques SL; West KA; Lingaraj T; Stickland K; Ribich SA; Raimondi A; Scott MP; Waters NJ; Pollock RM; Smith JJ; Barbash O; Pappalardi M; Ho TF; Nurse K; Oza KP; Gallagher KT; Kruger R; Moyer MP; Copeland RA; Chesworth R; Duncan KW A Selective Inhibitor of PRMT5 with in Vivo and in Vitro Potency in MCL Models. Nat Chem Biol 2015, 11 (6), 432–437. 10.1038/nchembio.1810. [DOI] [PubMed] [Google Scholar]
- (381).Duncan KW; Rioux N; Boriack-Sjodin PA; Munchhof MJ; Reiter LA; Majer CR; Jin L; Johnston LD; Chan-Penebre E; Kuplast KG; Porter Scott M; Pollock RM; Waters NJ; Smith JJ; Moyer MP; Copeland RA; Chesworth R Structure and Property Guided Design in the Identification of PRMT5 Tool Compound EPZ015666. ACS Med Chem Lett 2016, 7 (2), 162–166. 10.1021/acsmedchemlett.5b00380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (382).Smith JM; Barlaam B; Beattie D; Bradshaw L; Chan HM; Chiarparin E; Collingwood O; Cooke SL; Cronin A; Cumming I; Dean E; Debreczeni JÉ; Del Barco Barrantes I; Diene C; Gianni D; Guerot C; Guo X; Guven S; Hayhow TG; Hong T; Kemmitt PD; Lamont GM; Lamont S; Lynch JT; McWilliams L; Moore S; Raubo P; Robb GR; Robinson J; Scott JS; Srinivasan B; Steward O; Stubbs CJ; Syson K; Tan L; Turner O; Underwood E; Urosevic J; Vazquez-Chantada M; Whittaker AL; Wilson DM; Winter-Holt JJ Discovery and In Vivo Efficacy of AZ-PRMT5i-1, a Novel PRMT5 Inhibitor with High MTA Cooperativity. J Med Chem 2024, 67 (16), 13604–13638. 10.1021/acs.jmedchem.4c00097. [DOI] [PubMed] [Google Scholar]
- (383).Barekatain Y; Ackroyd JJ; Yan VC; Khadka S; Wang L; Chen K-C; Poral AH; Tran T; Georgiou DK; Arthur K; Lin Y-H; Satani N; Ballato ES; Behr EI; deCarvalho AC; Verhaak RGW; de Groot J; Huse JT; Asara JM; Kalluri R; Muller FL Homozygous MTAP Deletion in Primary Human Glioblastoma Is Not Associated with Elevation of Methylthioadenosine. Nat Commun 2021, 12 (1), 4228. 10.1038/s41467-021-24240-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (384).Artinger EL; Mishra BP; Zaffuto KM; Li BE; Chung EKY; Moore AW; Chen Y; Cheng C; Ernst P An MLL-Dependent Network Sustains Hematopoiesis. Proc Natl Acad Sci U S A 2013, 110 (29), 12000–12005. 10.1073/pnas.1301278110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (385).Milne TA; Briggs SD; Brock HW; Martin ME; Gibbs D; Allis CD; Hess JL MLL Targets SET Domain Methyltransferase Activity to Hox Gene Promoters. Mol Cell 2002, 10 (5), 1107–1117. 10.1016/s1097-2765(02)00741-4. [DOI] [PubMed] [Google Scholar]
- (386).Ernst P; Fisher JK; Avery W; Wade S; Foy D; Korsmeyer SJ Definitive Hematopoiesis Requires the Mixed-Lineage Leukemia Gene. Dev Cell 2004, 6 (3), 437–443. 10.1016/s1534-5807(04)00061-9. [DOI] [PubMed] [Google Scholar]
- (387).Meyer C; Hofmann J; Burmeister T; Gröger D; Park TS; Emerenciano M; Pombo de Oliveira M; Renneville A; Villarese P; Macintyre E; Cavé H; Clappier E; Mass-Malo K; Zuna J; Trka J; De Braekeleer E; De Braekeleer M; Oh SH; Tsaur G; Fechina L; van der Velden VHJ; van Dongen JJM; Delabesse E; Binato R; Silva MLM; Kustanovich A; Aleinikova O; Harris MH; Lund-Aho T; Juvonen V; Heidenreich O; Vormoor J; Choi WWL; Jarosova M; Kolenova A; Bueno C; Menendez P; Wehner S; Eckert C; Talmant P; Tondeur S; Lippert E; Launay E; Henry C; Ballerini P; Lapillone H; Callanan MB; Cayuela JM; Herbaux C; Cazzaniga G; Kakadiya PM; Bohlander S; Ahlmann M; Choi JR; Gameiro P; Lee DS; Krauter J; Cornillet-Lefebvre P; Te Kronnie G; Schäfer BW; Kubetzko S; Alonso CN; zur Stadt U; Sutton R; Venn NC; Izraeli S; Trakhtenbrot L; Madsen HO; Archer P; Hancock J; Cerveira N; Teixeira MR; Lo Nigro L; Möricke A; Stanulla M; Schrappe M; Sedék L; Szczepański T; Zwaan CM; Coenen EA; van den Heuvel-Eibrink MM; Strehl S; Dworzak M; Panzer-Grümayer R; Dingermann T; Klingebiel T; Marschalek R The MLL Recombinome of Acute Leukemias in 2013. Leukemia 2013, 27 (11), 2165–2176. 10.1038/leu.2013.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (388).Gu Y; Nakamura T; Alder H; Prasad R; Canaani O; Cimino G; Croce CM; Canaani E The t(4;11) Chromosome Translocation of Human Acute Leukemias Fuses the ALL-1 Gene, Related to Drosophila Trithorax, to the AF-4 Gene. Cell 1992, 71 (4), 701–708. 10.1016/0092-8674(92)90603-a. [DOI] [PubMed] [Google Scholar]
- (389).McCabe NR; Burnett RC; Gill HJ; Thirman MJ; Mbangkollo D; Kipiniak M; van Melle E; Ziemin-van der Poel S; Rowley JD; Diaz MO Cloning of CDNAs of the MLL Gene That Detect DNA Rearrangements and Altered RNA Transcripts in Human Leukemic Cells with 11q23 Translocations. Proc Natl Acad Sci U S A 1992, 89 (24), 11794–11798. 10.1073/pnas.89.24.11794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (390).Tkachuk DC; Kohler S; Cleary ML Involvement of a Homolog of Drosophila Trithorax by 11q23 Chromosomal Translocations in Acute Leukemias. Cell 1992, 71 (4), 691–700. 10.1016/0092-8674(92)90602-9. [DOI] [PubMed] [Google Scholar]
- (391).Meyer C; Burmeister T; Gröger D; Tsaur G; Fechina L; Renneville A; Sutton R; Venn NC; Emerenciano M; Pombo-de-Oliveira MS; Barbieri Blunck C; Almeida Lopes B; Zuna J; Trka J; Ballerini P; Lapillonne H; De Braekeleer M; Cazzaniga G; Corral Abascal L; van der Velden VHJ; Delabesse E; Park TS; Oh SH; Silva MLM; Lund-Aho T; Juvonen V; Moore AS; Heidenreich O; Vormoor J; Zerkalenkova E; Olshanskaya Y; Bueno C; Menendez P; Teigler-Schlegel A; Zur Stadt U; Lentes J; Göhring G; Kustanovich A; Aleinikova O; Schäfer BW; Kubetzko S; Madsen HO; Gruhn B; Duarte X; Gameiro P; Lippert E; Bidet A; Cayuela JM; Clappier E; Alonso CN; Zwaan CM; van den Heuvel-Eibrink MM; Izraeli S; Trakhtenbrot L; Archer P; Hancock J; Möricke A; Alten J; Schrappe M; Stanulla M; Strehl S; Attarbaschi A; Dworzak M; Haas OA; Panzer-Grümayer R; Sedék L; Szczepański T; Caye A; Suarez L; Cavé H; Marschalek R The MLL Recombinome of Acute Leukemias in 2017. Leukemia 2018, 32 (2), 273–284. 10.1038/leu.2017.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (392).Monroe SC; Jo SY; Sanders DS; Basrur V; Elenitoba-Johnson KS; Slany RK; Hess JL MLL-AF9 and MLL-ENL Alter the Dynamic Association of Transcriptional Regulators with Genes Critical for Leukemia. Exp Hematol 2011, 39 (1), 77–86.e1–5. 10.1016/j.exphem.2010.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (393).Mueller D; García-Cuéllar M-P; Bach C; Buhl S; Maethner E; Slany RK Misguided Transcriptional Elongation Causes Mixed Lineage Leukemia. PLoS Biol 2009, 7 (11), e1000249. 10.1371/journal.pbio.1000249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (394).Slany RK MLL Fusion Proteins and Transcriptional Control. Biochim Biophys Acta Gene Regul Mech 2020, 1863 (3), 194503. 10.1016/j.bbagrm.2020.194503. [DOI] [PubMed] [Google Scholar]
- (395).Mueller D; Bach C; Zeisig D; Garcia-Cuellar M-P; Monroe S; Sreekumar A; Zhou R; Nesvizhskii A; Chinnaiyan A; Hess JL; Slany RK A Role for the MLL Fusion Partner ENL in Transcriptional Elongation and Chromatin Modification. Blood 2007, 110 (13), 4445–4454. 10.1182/blood-2007-05-090514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (396).Chowdhury T; Brady HJM Insights from Clinical Studies into the Role of the MLL Gene in Infant and Childhood Leukemia. Blood Cells Mol Dis 2008, 40 (2), 192–199. 10.1016/j.bcmd.2007.07.005. [DOI] [PubMed] [Google Scholar]
- (397).Muntean AG; Hess JL The Pathogenesis of Mixed-Lineage Leukemia. Annu Rev Pathol 2012, 7, 283–301. 10.1146/annurev-pathol-011811-132434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (398).Sun Y; Chen B-R; Deshpande A Epigenetic Regulators in the Development, Maintenance, and Therapeutic Targeting of Acute Myeloid Leukemia. Front Oncol 2018, 8, 41. 10.3389/fonc.2018.00041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (399).Bitoun E; Oliver PL; Davies KE The Mixed-Lineage Leukemia Fusion Partner AF4 Stimulates RNA Polymerase II Transcriptional Elongation and Mediates Coordinated Chromatin Remodeling. Hum Mol Genet 2007, 16 (1), 92–106. 10.1093/hmg/ddl444. [DOI] [PubMed] [Google Scholar]
- (400).Mohan M; Herz H-M; Takahashi Y-H; Lin C; Lai KC; Zhang Y; Washburn MP; Florens L; Shilatifard A Linking H3K79 Trimethylation to Wnt Signaling through a Novel Dot1-Containing Complex (DotCom). Genes Dev 2010, 24 (6), 574–589. 10.1101/gad.1898410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (401).Okada Y; Feng Q; Lin Y; Jiang Q; Li Y; Coffield VM; Su L; Xu G; Zhang Y HDOT1L Links Histone Methylation to Leukemogenesis. Cell 2005, 121 (2), 167–178. 10.1016/j.cell.2005.02.020. [DOI] [PubMed] [Google Scholar]
- (402).Park G; Gong Z; Chen J; Kim J-E Characterization of the DOT1L Network: Implications of Diverse Roles for DOT1L. Protein J 2010, 29 (3), 213–223. 10.1007/s10930-010-9242-8. [DOI] [PubMed] [Google Scholar]
- (403).Shen C; Jo SY; Liao C; Hess JL; Nikolovska-Coleska Z Targeting Recruitment of Disruptor of Telomeric Silencing 1-like (DOT1L): Characterizing the Interactions between DOT1L and Mixed Lineage Leukemia (MLL) Fusion Proteins. J Biol Chem 2013, 288 (42), 30585–30596. 10.1074/jbc.M113.457135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (404).Zhang W; Xia X; Reisenauer MR; Hemenway CS; Kone BC Dot1a-AF9 Complex Mediates Histone H3 Lys-79 Hypermethylation and Repression of ENaCalpha in an Aldosterone-Sensitive Manner. J Biol Chem 2006, 281 (26), 18059–18068. 10.1074/jbc.M601903200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (405).Deshpande AJ; Deshpande A; Sinha AU; Chen L; Chang J; Cihan A; Fazio M; Chen C-W; Zhu N; Koche R; Dzhekieva L; Ibáñez G; Dias S; Banka D; Krivtsov A; Luo M; Roeder RG; Bradner JE; Bernt KM; Armstrong SA AF10 Regulates Progressive H3K79 Methylation and HOX Gene Expression in Diverse AML Subtypes. Cancer Cell 2014, 26 (6), 896–908. 10.1016/j.ccell.2014.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (406).Biswas D; Milne TA; Basrur V; Kim J; Elenitoba-Johnson KSJ; Allis CD; Roeder RG Function of Leukemogenic Mixed Lineage Leukemia 1 (MLL) Fusion Proteins through Distinct Partner Protein Complexes. Proc Natl Acad Sci U S A 2011, 108 (38), 15751–15756. 10.1073/pnas.1111498108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (407).Deshpande AJ; Rouhi A; Lin Y; Stadler C; Greif PA; Arseni N; Opatz S; Quintanilla-Fend L; Holzmann K; Hiddemann W; Döhner K; Döhner H; Xu G; Armstrong SA; Bohlander SK; Buske C The Clathrin-Binding Domain of CALM and the OM-LZ Domain of AF10 Are Sufficient to Induce Acute Myeloid Leukemia in Mice. Leukemia 2011, 25 (11), 1718–1727. 10.1038/leu.2011.153. [DOI] [PubMed] [Google Scholar]
- (408).Deshpande AJ; Bradner J; Armstrong SA Chromatin Modifications as Therapeutic Targets in MLL-Rearranged Leukemia. Trends Immunol 2012, 33 (11), 563–570. 10.1016/j.it.2012.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (409).Singer MS; Kahana A; Wolf AJ; Meisinger LL; Peterson SE; Goggin C; Mahowald M; Gottschling DE Identification of High-Copy Disruptors of Telomeric Silencing in Saccharomyces Cerevisiae. Genetics 1998, 150 (2), 613–632. 10.1093/genetics/150.2.613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (410).Ng HH; Feng Q; Wang H; Erdjument-Bromage H; Tempst P; Zhang Y; Struhl K Lysine Methylation within the Globular Domain of Histone H3 by Dot1 Is Important for Telomeric Silencing and Sir Protein Association. Genes Dev 2002, 16 (12), 1518–1527. 10.1101/gad.1001502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (411).van Leeuwen F; Gafken PR; Gottschling DE Dot1p Modulates Silencing in Yeast by Methylation of the Nucleosome Core. Cell 2002, 109 (6), 745–756. 10.1016/s0092-8674(02)00759-6. [DOI] [PubMed] [Google Scholar]
- (412).Lacoste N; Utley RT; Hunter JM; Poirier GG; Côte J Disruptor of Telomeric Silencing-1 Is a Chromatin-Specific Histone H3 Methyltransferase. J Biol Chem 2002, 277 (34), 30421–30424. 10.1074/jbc.C200366200. [DOI] [PubMed] [Google Scholar]
- (413).Steger DJ; Lefterova MI; Ying L; Stonestrom AJ; Schupp M; Zhuo D; Vakoc AL; Kim J-E; Chen J; Lazar MA; Blobel GA; Vakoc CR DOT1L/KMT4 Recruitment and H3K79 Methylation Are Ubiquitously Coupled with Gene Transcription in Mammalian Cells. Mol Cell Biol 2008, 28 (8), 2825–2839. 10.1128/MCB.02076-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (414).Schübeler D; MacAlpine DM; Scalzo D; Wirbelauer C; Kooperberg C; van Leeuwen F; Gottschling DE; O’Neill LP; Turner BM; Delrow J; Bell SP; Groudine M The Histone Modification Pattern of Active Genes Revealed through Genome-Wide Chromatin Analysis of a Higher Eukaryote. Genes Dev 2004, 18 (11), 1263–1271. 10.1101/gad.1198204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (415).Godfrey L; Crump NT; Thorne R; Lau I-J; Repapi E; Dimou D; Smith AL; Harman JR; Telenius JM; Oudelaar AM; Downes DJ; Vyas P; Hughes JR; Milne TA DOT1L Inhibition Reveals a Distinct Subset of Enhancers Dependent on H3K79 Methylation. Nat Commun 2019, 10 (1), 2803. 10.1038/s41467-019-10844-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (416).Malcom CA; Ratri A; Piasecka-Srader J; Borosha S; Chakravarthi VP; Alvarez NS; Vivian JL; Fields TA; Karim Rumi MA; Fields PE Primitive Erythropoiesis in the Mouse Is Independent of DOT1L Methyltransferase Activity. Front Cell Dev Biol 2021, 9, 813503. 10.3389/fcell.2021.813503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (417).Arnold O; Barbosa K; Deshpande AJ; Zhu N The Role of DOT1L in Normal and Malignant Hematopoiesis. Front Cell Dev Biol 2022, 10, 917125. 10.3389/fcell.2022.917125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (418).Krivtsov AV; Feng Z; Lemieux ME; Faber J; Vempati S; Sinha AU; Xia X; Jesneck J; Bracken AP; Silverman LB; Kutok JL; Kung AL; Armstrong SA H3K79 Methylation Profiles Define Murine and Human MLL-AF4 Leukemias. Cancer Cell 2008, 14 (5), 355–368. 10.1016/j.ccr.2008.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (419).Bernt KM; Zhu N; Sinha AU; Vempati S; Faber J; Krivtsov AV; Feng Z; Punt N; Daigle A; Bullinger L; Pollock RM; Richon VM; Kung AL; Armstrong SA MLL-Rearranged Leukemia Is Dependent on Aberrant H3K79 Methylation by DOT1L. Cancer Cell 2011, 20 (1), 66–78. 10.1016/j.ccr.2011.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (420).Daigle SR; Olhava EJ; Therkelsen CA; Majer CR; Sneeringer CJ; Song J; Johnston LD; Scott MP; Smith JJ; Xiao Y; Jin L; Kuntz KW; Chesworth R; Moyer MP; Bernt KM; Tseng J-C; Kung AL; Armstrong SA; Copeland RA; Richon VM; Pollock RM Selective Killing of Mixed Lineage Leukemia Cells by a Potent Small-Molecule DOT1L Inhibitor. Cancer Cell 2011, 20 (1), 53–65. 10.1016/j.ccr.2011.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (421).Chang M-J; Wu H; Achille NJ; Reisenauer MR; Chou C-W; Zeleznik-Le NJ; Hemenway CS; Zhang W Histone H3 Lysine 79 Methyltransferase Dot1 Is Required for Immortalization by MLL Oncogenes. Cancer Res 2010, 70 (24), 10234–10242. 10.1158/0008-5472.CAN-10-3294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (422).Jo SY; Granowicz EM; Maillard I; Thomas D; Hess JL Requirement for Dot1l in Murine Postnatal Hematopoiesis and Leukemogenesis by MLL Translocation. Blood 2011, 117 (18), 4759–4768. 10.1182/blood-2010-12-327668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (423).Chen L; Deshpande AJ; Banka D; Bernt KM; Dias S; Buske C; Olhava EJ; Daigle SR; Richon VM; Pollock RM; Armstrong SA Abrogation of MLL-AF10 and CALM-AF10-Mediated Transformation through Genetic Inactivation or Pharmacological Inhibition of the H3K79 Methyltransferase Dot1l. Leukemia 2013, 27 (4), 813–822. 10.1038/leu.2012.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (424).Deshpande AJ; Chen L; Fazio M; Sinha AU; Bernt KM; Banka D; Dias S; Chang J; Olhava EJ; Daigle SR; Richon VM; Pollock RM; Armstrong SA Leukemic Transformation by the MLL-AF6 Fusion Oncogene Requires the H3K79 Methyltransferase Dot1l. Blood 2013, 121 (13), 2533–2541. 10.1182/blood-2012-11-465120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (425).Nguyen AT; Taranova O; He J; Zhang Y DOT1L, the H3K79 Methyltransferase, Is Required for MLL-AF9-Mediated Leukemogenesis. Blood 2011, 117 (25), 6912–6922. 10.1182/blood-2011-02-334359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (426).Kühn MWM; Hadler MJ; Daigle SR; Koche RP; Krivtsov AV; Olhava EJ; Caligiuri MA; Huang G; Bradner JE; Pollock RM; Armstrong SA MLL Partial Tandem Duplication Leukemia Cells Are Sensitive to Small Molecule DOT1L Inhibition. Haematologica 2015, 100 (5), e190–3. 10.3324/haematol.2014.115337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (427).Min J; Feng Q; Li Z; Zhang Y; Xu R-M Structure of the Catalytic Domain of Human DOT1L, a Non-SET Domain Nucleosomal Histone Methyltransferase. Cell 2003, 112 (5), 711–723. 10.1016/s0092-8674(03)00114-4. [DOI] [PubMed] [Google Scholar]
- (428).Feng Q; Wang H; Ng HH; Erdjument-Bromage H; Tempst P; Struhl K; Zhang Y Methylation of H3-Lysine 79 Is Mediated by a New Family of HMTases without a SET Domain. Curr Biol 2002, 12 (12), 1052–1058. 10.1016/s0960-9822(02)00901-6. [DOI] [PubMed] [Google Scholar]
- (429).Yao Y; Chen P; Diao J; Cheng G; Deng L; Anglin JL; Prasad BVV; Song Y Selective Inhibitors of Histone Methyltransferase DOT1L: Design, Synthesis, and Crystallographic Studies. J Am Chem Soc 2011, 133 (42), 16746–16749. 10.1021/ja206312b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (430).Anglin JL; Deng L; Yao Y; Cai G; Liu Z; Jiang H; Cheng G; Chen P; Dong S; Song Y Synthesis and Structure-Activity Relationship Investigation of Adenosine-Containing Inhibitors of Histone Methyltransferase DOT1L. J Med Chem 2012, 55 (18), 8066–8074. 10.1021/jm300917h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (431).Yu W; Smil D; Li F; Tempel W; Fedorov O; Nguyen KT; Bolshan Y; Al-Awar R; Knapp S; Arrowsmith CH; Vedadi M; Brown PJ; Schapira M Bromo-Deaza-SAH: A Potent and Selective DOT1L Inhibitor. Bioorg Med Chem 2013, 21 (7), 1787–1794. 10.1016/j.bmc.2013.01.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (432).Basavapathruni A; Jin L; Daigle SR; Majer CRA; Therkelsen CA; Wigle TJ; Kuntz KW; Chesworth R; Pollock RM; Scott MP; Moyer MP; Richon VM; Copeland RA; Olhava EJ Conformational Adaptation Drives Potent, Selective and Durable Inhibition of the Human Protein Methyltransferase DOT1L. Chem Biol Drug Des 2012, 80 (6), 971–980. 10.1111/cbdd.12050. [DOI] [PubMed] [Google Scholar]
- (433).Sweet SMM; Li M; Thomas PM; Durbin KR; Kelleher NL Kinetics of Re-Establishing H3K79 Methylation Marks in Global Human Chromatin. J Biol Chem 2010, 285 (43), 32778–32786. 10.1074/jbc.M110.145094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (434).Zee BM; Levin RS; Xu B; LeRoy G; Wingreen NS; Garcia BA In Vivo Residue-Specific Histone Methylation Dynamics. J Biol Chem 2010, 285 (5), 3341–3350. 10.1074/jbc.M109.063784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (435).Chory EJ; Calarco JP; Hathaway NA; Bell O; Neel DS; Crabtree GR Nucleosome Turnover Regulates Histone Methylation Patterns over the Genome. Mol Cell 2019, 73 (1), 61–72.e3. 10.1016/j.molcel.2018.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (436).Daigle SR; Olhava EJ; Therkelsen CA; Basavapathruni A; Jin L; Boriack-Sjodin PA; Allain CJ; Klaus CR; Raimondi A; Scott MP; Waters NJ; Chesworth R; Moyer MP; Copeland RA; Richon VM; Pollock RM Potent Inhibition of DOT1L as Treatment of MLL-Fusion Leukemia. Blood 2013, 122 (6), 1017–1025. 10.1182/blood-2013-04-497644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (437).Yu W; Chory EJ; Wernimont AK; Tempel W; Scopton A; Federation A; Marineau JJ; Qi J; Barsyte-Lovejoy D; Yi J; Marcellus R; Iacob RE; Engen JR; Griffin C; Aman A; Wienholds E; Li F; Pineda J; Estiu G; Shatseva T; Hajian T; Al-Awar R; Dick JE; Vedadi M; Brown PJ; Arrowsmith CH; Bradner JE; Schapira M Catalytic Site Remodelling of the DOT1L Methyltransferase by Selective Inhibitors. Nat Commun 2012, 3, 1288. 10.1038/ncomms2304. [DOI] [PubMed] [Google Scholar]
- (438).Basavapathruni A; Olhava EJ; Daigle SR; Therkelsen CA; Jin L; Boriack-Sjodin PA; Allain CJ; Klaus CR; Raimondi A; Scott MP; Dovletoglou A; Richon VM; Pollock RM; Copeland RA; Moyer MP; Chesworth R; Pearson PG; Waters NJ Nonclinical Pharmacokinetics and Metabolism of EPZ-5676, a Novel DOT1L Histone Methyltransferase Inhibitor. Biopharm Drug Dispos 2014, 35 (4), 237–252. 10.1002/bdd.1889. [DOI] [PubMed] [Google Scholar]
- (439).Chen C-W; Armstrong SA Targeting DOT1L and HOX Gene Expression in MLL-Rearranged Leukemia and Beyond. Exp Hematol 2015, 43 (8), 673–684. 10.1016/j.exphem.2015.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (440).Waters NJ Preclinical Pharmacokinetics and Pharmacodynamics of Pinometostat (EPZ-5676), a First-in-Class, Small Molecule S-Adenosyl Methionine Competitive Inhibitor of DOT1L. Eur J Drug Metab Pharmacokinet 2017, 42 (6), 891–901. 10.1007/s13318-017-0404-3. [DOI] [PubMed] [Google Scholar]
- (441).Shukla N; Wetmore C; O’Brien MM; Silverman LB; Brown P; Cooper TM; Thomson B; Blakemore SJ; Daigle S; Suttle B; Waters NJ; Krivstov AV; Armstrong SA; Ho PT; Gore L Final Report of Phase 1 Study of the DOT1L Inhibitor, Pinometostat (EPZ-5676), in Children with Relapsed or Refractory MLL-r Acute Leukemia. Blood 2016, 128 (22), 2780–2780. 10.1182/blood.V128.22.2780.2780. [DOI] [Google Scholar]
- (442).Stein EM; Garcia-Manero G; Rizzieri DA; Tibes R; Berdeja JG; Savona MR; Jongen-Lavrenic M; Altman JK; Thomson B; Blakemore SJ; Daigle SR; Waters NJ; Suttle AB; Clawson A; Pollock R; Krivtsov A; Armstrong SA; DiMartino J; Hedrick E; Löwenberg B; Tallman MS The DOT1L Inhibitor Pinometostat Reduces H3K79 Methylation and Has Modest Clinical Activity in Adult Acute Leukemia. Blood 2018, 131 (24), 2661–2669. 10.1182/blood-2017-12-818948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (443).Stein EM; Tallman MS Mixed Lineage Rearranged Leukaemia: Pathogenesis and Targeting DOT1L. Curr Opin Hematol 2015, 22 (2), 92–96. 10.1097/MOH.0000000000000123. [DOI] [PubMed] [Google Scholar]
- (444).Gilan O; Lam EYN; Becher I; Lugo D; Cannizzaro E; Joberty G; Ward A; Wiese M; Fong CY; Ftouni S; Tyler D; Stanley K; MacPherson L; Weng C-F; Chan Y-C; Ghisi M; Smil D; Carpenter C; Brown P; Garton N; Blewitt ME; Bannister AJ; Kouzarides T; Huntly BJP; Johnstone RW; Drewes G; Dawson S-J; Arrowsmith CH; Grandi P; Prinjha RK; Dawson MA Functional Interdependence of BRD4 and DOT1L in MLL Leukemia. Nat Struct Mol Biol 2016, 23 (7), 673–681. 10.1038/nsmb.3249. [DOI] [PubMed] [Google Scholar]
- (445).Klaus CR; Iwanowicz D; Johnston D; Campbell CA; Smith JJ; Moyer MP; Copeland RA; Olhava EJ; Scott MP; Pollock RM; Daigle SR; Raimondi A DOT1L Inhibitor EPZ-5676 Displays Synergistic Antiproliferative Activity in Combination with Standard of Care Drugs and Hypomethylating Agents in MLL-Rearranged Leukemia Cells. J Pharmacol Exp Ther 2014, 350 (3), 646–656. 10.1124/jpet.114.214577. [DOI] [PubMed] [Google Scholar]
- (446).MacLeod G; Bozek DA; Rajakulendran N; Monteiro V; Ahmadi M; Steinhart Z; Kushida MM; Yu H; Coutinho FJ; Cavalli FMG; Restall I; Hao X; Hart T; Luchman HA; Weiss S; Dirks PB; Angers S Genome-Wide CRISPR-Cas9 Screens Expose Genetic Vulnerabilities and Mechanisms of Temozolomide Sensitivity in Glioblastoma Stem Cells. Cell Rep 2019, 27 (3), 971–986.e9. 10.1016/j.celrep.2019.03.047. [DOI] [PubMed] [Google Scholar]
- (447).Zhang L; Deng L; Chen F; Yao Y; Wu B; Wei L; Mo Q; Song Y Inhibition of Histone H3K79 Methylation Selectively Inhibits Proliferation, Self-Renewal and Metastatic Potential of Breast Cancer. Oncotarget 2014, 5 (21), 10665–10677. 10.18632/oncotarget.2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (448).Cho M-H; Park J-H; Choi H-J; Park M-K; Won H-Y; Park Y-J; Lee CH; Oh S-H; Song Y-S; Kim HS; Oh Y-H; Lee J-Y; Kong G DOT1L Cooperates with the C-Myc-P300 Complex to Epigenetically Derepress CDH1 Transcription Factors in Breast Cancer Progression. Nat Commun 2015, 6, 7821. 10.1038/ncomms8821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (449).Kurani H; Razavipour SF; Harikumar KB; Dunworth M; Ewald AJ; Nasir A; Pearson G; Van Booven D; Zhou Z; Azzam D; Wahlestedt C; Slingerland J DOT1L Is a Novel Cancer Stem Cell Target for Triple-Negative Breast Cancer. Clin Cancer Res 2022, 28 (9), 1948–1965. 10.1158/1078-0432.CCR-21-1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (450).Nassa G; Salvati A; Tarallo R; Gigantino V; Alexandrova E; Memoli D; Sellitto A; Rizzo F; Malanga D; Mirante T; Morelli E; Nees M; Åkerfelt M; Kangaspeska S; Nyman TA; Milanesi L; Giurato G; Weisz A Inhibition of Histone Methyltransferase DOT1L Silences ERα Gene and Blocks Proliferation of Antiestrogen-Resistant Breast Cancer Cells. Sci Adv 2019, 5 (2), eaav5590. 10.1126/sciadv.aav5590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (451).Kühn MWM; Song E; Feng Z; Sinha A; Chen C-W; Deshpande AJ; Cusan M; Farnoud N; Mupo A; Grove C; Koche R; Bradner JE; de Stanchina E; Vassiliou GS; Hoshii T; Armstrong SA Targeting Chromatin Regulators Inhibits Leukemogenic Gene Expression in NPM1 Mutant Leukemia. Cancer Discov 2016, 6 (10), 1166–1181. 10.1158/2159-8290.CD-16-0237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (452).Zhang J; Yang T; Han M; Wang X; Yang W; Guo N; Ren Y; Cui W; Li S; Zhao Y; Zhai X; Jia L; Yang J; Wu C; Wang L Gain-of-Function Mutations in the Catalytic Domain of DOT1L Promote Lung Cancer Malignant Phenotypes via the MAPK/ERK Signaling Pathway. Sci Adv 2023, 9 (22), eadc9273. 10.1126/sciadv.adc9273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (453).Liu D; Zhang X-X; Li M-C; Cao C-H; Wan D-Y; Xi B-X; Tan J-H; Wang J; Yang Z-Y; Feng X-X; Ye F; Chen G; Wu P; Xi L; Wang H; Zhou J-F; Feng Z-H; Ma D; Gao Q-L C/EBPβ Enhances Platinum Resistance of Ovarian Cancer Cells by Reprogramming H3K79 Methylation. Nat Commun 2018, 9 (1), 1739. 10.1038/s41467-018-03590-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (454).Wong M; Tee AEL; Milazzo G; Bell JL; Poulos RC; Atmadibrata B; Sun Y; Jing D; Ho N; Ling D; Liu PY; Zhang XD; Hüttelmaier S; Wong JWH; Wang J; Polly P; Perini G; Scarlett CJ; Liu T The Histone Methyltransferase DOT1L Promotes Neuroblastoma by Regulating Gene Transcription. Cancer Res 2017, 77 (9), 2522–2533. 10.1158/0008-5472.CAN-16-1663. [DOI] [PubMed] [Google Scholar]
- (455).Kryczek I; Lin Y; Nagarsheth N; Peng D; Zhao L; Zhao E; Vatan L; Szeliga W; Dou Y; Owens S; Zgodzinski W; Majewski M; Wallner G; Fang J; Huang E; Zou W IL-22(+)CD4(+) T Cells Promote Colorectal Cancer Stemness via STAT3 Transcription Factor Activation and Induction of the Methyltransferase DOT1L. Immunity 2014, 40 (5), 772–784. 10.1016/j.immuni.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (456).Yi Y; Ge S Targeting the Histone H3 Lysine 79 Methyltransferase DOT1L in MLL-Rearranged Leukemias. J Hematol Oncol 2022, 15 (1), 35. 10.1186/s13045-022-01251-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (457).Chen C; Zhu H; Stauffer F; Caravatti G; Vollmer S; Machauer R; Holzer P; Möbitz H; Scheufler C; Klumpp M; Tiedt R; Beyer KS; Calkins K; Guthy D; Kiffe M; Zhang J; Gaul C Discovery of Novel Dot1L Inhibitors through a Structure-Based Fragmentation Approach. ACS Med Chem Lett 2016, 7 (8), 735–740. 10.1021/acsmedchemlett.6b00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (458).Cao M; Li T; Chen Y; Zhai X Nucleoside and Non-Nucleoside DOT1L Inhibitors: Dawn of MLLrearranged Leukemia. Mini Rev Med Chem 2021, 21 (11), 1337–1350. 10.2174/1389557521666210111144357. [DOI] [PubMed] [Google Scholar]
- (459).Gibbons GS; Chakraborty A; Grigsby SM; Umeano AC; Liao C; Moukha-Chafiq O; Pathak V; Mathew B; Lee Y-T; Dou Y; Schürer SC; Reynolds RC; Snowden TS; Nikolovska-Coleska Z Identification of DOT1L Inhibitors by Structure-Based Virtual Screening Adapted from a Nucleoside-Focused Library. Eur J Med Chem 2020, 189, 112023. 10.1016/j.ejmech.2019.112023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (460).Chen S; Li L; Chen Y; Hu J; Liu J; Liu Y-C; Liu R; Zhang Y; Meng F; Zhu K; Lu J; Zheng M; Chen K; Zhang J; Jiang H; Yao Z; Luo C Identification of Novel Disruptor of Telomeric Silencing 1-like (DOT1L) Inhibitors through Structure-Based Virtual Screening and Biological Assays. J Chem Inf Model 2016, 56 (3), 527–534. 10.1021/acs.jcim.5b00738. [DOI] [PubMed] [Google Scholar]
- (461).Zhang L; Chen Y; Liu N; Li L; Xiao S; Li X; Chen K; Luo C; Chen S; Chen H Design, Synthesis and Anti Leukemia Cells Proliferation Activities of Pyrimidylaminoquinoline Derivatives as DOT1L Inhibitors. Bioorg Chem 2018, 80, 649–654. 10.1016/j.bioorg.2018.07.022. [DOI] [PubMed] [Google Scholar]
- (462).Stauffer F; Weiss A; Scheufler C; Möbitz H; Ragot C; Beyer KS; Calkins K; Guthy D; Kiffe M; Van Eerdenbrugh B; Tiedt R; Gaul C New Potent DOT1L Inhibitors for in Vivo Evaluation in Mouse. ACS Med Chem Lett 2019, 10 (12), 1655–1660. 10.1021/acsmedchemlett.9b00452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (463).Perner F; Gadrey JY; Xiong Y; Hatton C; Eschle BK; Weiss A; Stauffer F; Gaul C; Tiedt R; Perry JA; Armstrong SA; Krivtsov AV Novel Inhibitors of the Histone Methyltransferase DOT1L Show Potent Antileukemic Activity in Patient-Derived Xenografts. Blood 2020, 136 (17), 1983–1988. 10.1182/blood.2020006113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (464).Sabatino M; Rotili D; Patsilinakos A; Forgione M; Tomaselli D; Alby F; Arimondo PB; Mai A; Ragno R Disruptor of Telomeric Silencing 1-like (DOT1L): Disclosing a New Class of Non-Nucleoside Inhibitors by Means of Ligand-Based and Structure-Based Approaches. J Comput Aided Mol Des 2018, 32 (3), 435–458. 10.1007/s10822-018-0096-z. [DOI] [PubMed] [Google Scholar]
- (465).Du L; Grigsby SM; Yao A; Chang Y; Johnson G; Sun H; Nikolovska-Coleska Z Peptidomimetics for Targeting Protein-Protein Interactions between DOT1L and MLL Oncofusion Proteins AF9 and ENL. ACS Med Chem Lett 2018, 9 (9), 895–900. 10.1021/acsmedchemlett.8b00175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (466).Grigsby SM; Friedman A; Chase J; Waas B; Ropa J; Serio J; Shen C; Muntean AG; Maillard I; Nikolovska-Coleska Z Elucidating the Importance of DOT1L Recruitment in MLL-AF9 Leukemia and Hematopoiesis. Cancers (Basel) 2021, 13 (4), 642. 10.3390/cancers13040642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (467).Yang Y; Ahmad E; Premkumar V; Liu A; Ashikur Rahman SM; Nikolovska-Coleska Z Structural Studies of Intrinsically Disordered MLL-Fusion Protein AF9 in Complex with Peptidomimetic Inhibitors. Protein Sci 2024, 33 (6), e5019. 10.1002/pro.5019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (468).Wu F; Nie S; Yao Y; Huo T; Li X; Wu X; Zhao J; Lin Y-L; Zhang Y; Mo Q; Song Y Small-Molecule Inhibitor of AF9/ENL-DOT1L/AF4/AFF4 Interactions Suppresses Malignant Gene Expression and Tumor Growth. Theranostics 2021, 11 (17), 8172–8184. 10.7150/thno.56737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (469).Li X; Wu X; Nie S; Zhao J; Yao Y; Wu F; Mishra CB; Ashraf-Uz-Zaman M; Moku BK; Song Y Discovery, Structure-Activity Relationship and In Vitro Anticancer Activity of Small-Molecule Inhibitors of the Protein-Protein Interactions between AF9/ENL and AF4 or DOT1L. Cancers (Basel) 2023, 15 (21), 5283. 10.3390/cancers15215283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (470).Zhou Z; Kang S; Huang Z; Zhou Z; Chen S Structural Characteristics of Coiled-Coil Regions in AF10-DOT1L and AF10-Inhibitory Peptide Complex. J Leukoc Biol 2021, 110 (6), 1091–1099. 10.1002/JLB.1MA0421-010R. [DOI] [PubMed] [Google Scholar]
- (471).Yokoyama A; Lin M; Naresh A; Kitabayashi I; Cleary ML A Higher-Order Complex Containing AF4 and ENL Family Proteins with P-TEFb Facilitates Oncogenic and Physiologic MLL-Dependent Transcription. Cancer Cell 2010, 17 (2), 198–212. 10.1016/j.ccr.2009.12.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (472).Chandrasekharappa SC; Guru SC; Manickam P; Olufemi SE; Collins FS; Emmert-Buck MR; Debelenko LV; Zhuang Z; Lubensky IA; Liotta LA; Crabtree JS; Wang Y; Roe BA; Weisemann J; Boguski MS; Agarwal SK; Kester MB; Kim YS; Heppner C; Dong Q; Spiegel AM; Burns AL; Marx SJ Positional Cloning of the Gene for Multiple Endocrine Neoplasia-Type 1. Science 1997, 276 (5311), 404–407. 10.1126/science.276.5311.404. [DOI] [PubMed] [Google Scholar]
- (473).Yokoyama A; Wang Z; Wysocka J; Sanyal M; Aufiero DJ; Kitabayashi I; Herr W; Cleary ML Leukemia Proto-Oncoprotein MLL Forms a SET1-like Histone Methyltransferase Complex with Menin to Regulate Hox Gene Expression. Mol Cell Biol 2004, 24 (13), 5639–5649. 10.1128/MCB.24.13.5639-5649.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (474).Yokoyama A; Somervaille TCP; Smith KS; Rozenblatt-Rosen O; Meyerson M; Cleary ML The Menin Tumor Suppressor Protein Is an Essential Oncogenic Cofactor for MLL-Associated Leukemogenesis. Cell 2005, 123 (2), 207–218. 10.1016/j.cell.2005.09.025. [DOI] [PubMed] [Google Scholar]
- (475).Caslini C; Yang Z; El-Osta M; Milne TA; Slany RK; Hess JL Interaction of MLL Amino Terminal Sequences with Menin Is Required for Transformation. Cancer Res 2007, 67 (15), 7275–7283. 10.1158/0008-5472.CAN-06-2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (476).Li BE; Gan T; Meyerson M; Rabbitts TH; Ernst P Distinct Pathways Regulated by Menin and by MLL1 in Hematopoietic Stem Cells and Developing B Cells. Blood 2013, 122 (12), 2039–2046. 10.1182/blood-2013-03-486647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (477).Shi A; Murai MJ; He S; Lund G; Hartley T; Purohit T; Reddy G; Chruszcz M; Grembecka J; Cierpicki T Structural Insights into Inhibition of the Bivalent Menin-MLL Interaction by Small Molecules in Leukemia. Blood 2012, 120 (23), 4461–4469. 10.1182/blood-2012-05-429274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (478).Xu Y; Yue L; Wang Y; Xing J; Chen Z; Shi Z; Liu R; Liu Y-C; Luo X; Jiang H; Chen K; Luo C; Zheng M Discovery of Novel Inhibitors Targeting the Menin-Mixed Lineage Leukemia Interface Using Pharmacophore- and Docking-Based Virtual Screening. J Chem Inf Model 2016, 56 (9), 1847–1855. 10.1021/acs.jcim.6b00185. [DOI] [PubMed] [Google Scholar]
- (479).Borkin D; He S; Miao H; Kempinska K; Pollock J; Chase J; Purohit T; Malik B; Zhao T; Wang J; Wen B; Zong H; Jones M; Danet-Desnoyers G; Guzman ML; Talpaz M; Bixby DL; Sun D; Hess JL; Muntean AG; Maillard I; Cierpicki T; Grembecka J Pharmacologic Inhibition of the Menin-MLL Interaction Blocks Progression of MLL Leukemia in Vivo. Cancer Cell 2015, 27 (4), 589–602. 10.1016/j.ccell.2015.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (480).He S; Senter TJ; Pollock J; Han C; Upadhyay SK; Purohit T; Gogliotti RD; Lindsley CW; Cierpicki T; Stauffer SR; Grembecka J High-Affinity Small-Molecule Inhibitors of the Menin-Mixed Lineage Leukemia (MLL) Interaction Closely Mimic a Natural Protein-Protein Interaction. J Med Chem 2014, 57 (4), 1543–1556. 10.1021/jm401868d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (481).Borkin D; Pollock J; Kempinska K; Purohit T; Li X; Wen B; Zhao T; Miao H; Shukla S; He M; Sun D; Cierpicki T; Grembecka J Property Focused Structure-Based Optimization of Small Molecule Inhibitors of the Protein-Protein Interaction between Menin and Mixed Lineage Leukemia (MLL). J Med Chem 2016, 59 (3), 892–913. 10.1021/acs.jmedchem.5b01305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (482).Wang ES; Issa GC; Erba HP; Altman JK; Montesinos P; DeBotton S; Walter RB; Pettit K; Savona MR; Shah MV; Kremyanskaya M; Baer MR; Foran JM; Schiller G; Adès L; Heiblig M; Berthon C; Peterlin P; Rodríguez-Arbolí E; Salamero O; Patnaik MM; Papayannidis C; Grembecka J; Cierpicki T; Clegg B; Ray J; Linhares BM; Nie K; Mitra A; Ahsan JM; Tabachri M; Soifer HS; Corum D; Leoni M; Dale S; Fathi AT Ziftomenib in Relapsed or Refractory Acute Myeloid Leukaemia (KOMET-001): A Multicentre, Open-Label, Multi-Cohort, Phase 1 Trial. Lancet Oncol 2024, 25 (10), 1310–1324. 10.1016/S1470-2045(24)00386-3. [DOI] [PubMed] [Google Scholar]
- (483).Fiskus W; Daver N; Boettcher S; Mill CP; Sasaki K; Birdwell CE; Davis JA; Das K; Takahashi K; Kadia TM; DiNardo CD; Burrows F; Loghavi S; Khoury JD; Ebert BL; Bhalla KN Activity of Menin Inhibitor Ziftomenib (KO-539) as Monotherapy or in Combinations against AML Cells with MLL1 Rearrangement or Mutant NPM1. Leukemia 2022, 36 (11), 2729–2733. 10.1038/s41375-022-01707-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (484).Erba HP; Fathi AT; Issa GC; Altman JK; Montesinos P; Patnaik MM; Foran JM; De Botton S; Baer MR; Schiller GJ; Walter RB; Kremyanskaya M; Pettit KM; Strickland SA; Tomkinson B; Tabachri M; Leoni M; Dale S; Wang ES Update on a Phase 1/2 First-in-Human Study of the Menin-KMT2A (MLL) Inhibitor Ziftomenib (KO-539) in Patients with Relapsed or Refractory Acute Myeloid Leukemia. Blood 2022, 140 (Supplement 1), 153–156. 10.1182/blood-2022-167412. [DOI] [Google Scholar]
- (485).Rausch J; Dzama MM; Dolgikh N; Stiller HL; Bohl SR; Lahrmann C; Kunz K; Kessler L; Echchannaoui H; Chen C-W; Kindler T; Döhner K; Burrows F; Theobald M; Sasca D; Kühn MWM Menin Inhibitor Ziftomenib (KO-539) Synergizes with Drugs Targeting Chromatin Regulation or Apoptosis and Sensitizes Acute Myeloid Leukemia with MLL Rearrangement or NPM1 Mutation to Venetoclax. Haematologica 2023, 108 (10), 2837–2843. 10.3324/haematol.2022.282160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (486).Fiskus W; Boettcher S; Daver N; Mill CP; Sasaki K; Birdwell CE; Davis JA; Takahashi K; Kadia TM; DiNardo CD; Jin Q; Qi Y; Su X; McGeehan GM; Khoury JD; Ebert BL; Bhalla KN Effective Menin Inhibitor-Based Combinations against AML with MLL Rearrangement or NPM1 Mutation (NPM1c). Blood Cancer J 2022, 12 (1), 5. 10.1038/s41408-021-00603-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (487).Falini B; Mecucci C; Tiacci E; Alcalay M; Rosati R; Pasqualucci L; La Starza R; Diverio D; Colombo E; Santucci A; Bigerna B; Pacini R; Pucciarini A; Liso A; Vignetti M; Fazi P; Meani N; Pettirossi V; Saglio G; Mandelli F; Lo-Coco F; Pelicci P-G; Martelli MF; GIMEMA Acute Leukemia Working Party. Cytoplasmic Nucleophosmin in Acute Myelogenous Leukemia with a Normal Karyotype. N Engl J Med 2005, 352 (3), 254–266. 10.1056/NEJMoa041974. [DOI] [PubMed] [Google Scholar]
- (488).Cancer Genome Atlas Research Network; Ley TJ; Miller C; Ding L; Raphael BJ; Mungall AJ; Robertson AG; Hoadley K; Triche TJ; Laird PW; Baty JD; Fulton LL; Fulton R; Heath SE; Kalicki-Veizer J; Kandoth C; Klco JM; Koboldt DC; Kanchi K-L; Kulkarni S; Lamprecht TL; Larson DE; Lin L; Lu C; McLellan MD; McMichael JF; Payton J; Schmidt H; Spencer DH; Tomasson MH; Wallis JW; Wartman LD; Watson MA; Welch J; Wendl MC; Ally A; Balasundaram M; Birol I; Butterfield Y; Chiu R; Chu A; Chuah E; Chun H-J; Corbett R; Dhalla N; Guin R; He A; Hirst C; Hirst M; Holt RA; Jones S; Karsan A; Lee D; Li HI; Marra MA; Mayo M; Moore RA; Mungall K; Parker J; Pleasance E; Plettner P; Schein J; Stoll D; Swanson L; Tam A; Thiessen N; Varhol R; Wye N; Zhao Y; Gabriel S; Getz G; Sougnez C; Zou L; Leiserson MDM; Vandin F; Wu H-T; Applebaum F; Baylin SB; Akbani R; Broom BM; Chen K; Motter TC; Nguyen K; Weinstein JN; Zhang N; Ferguson ML; Adams C; Black A; Bowen J; Gastier-Foster J; Grossman T; Lichtenberg T; Wise L; Davidsen T; Demchok JA; Shaw KRM; Sheth M; Sofia HJ; Yang L; Downing JR; Eley G Genomic and Epigenomic Landscapes of Adult de Novo Acute Myeloid Leukemia. N Engl J Med 2013, 368 (22), 2059–2074. 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (489).Alcalay M; Tiacci E; Bergomas R; Bigerna B; Venturini E; Minardi SP; Meani N; Diverio D; Bernard L; Tizzoni L; Volorio S; Luzi L; Colombo E; Lo Coco F; Mecucci C; Falini B; Pelicci PG Acute Myeloid Leukemia Bearing Cytoplasmic Nucleophosmin (NPMc+ AML) Shows a Distinct Gene Expression Profile Characterized by up-Regulation of Genes Involved in Stem-Cell Maintenance. Blood 2005, 106 (3), 899–902. 10.1182/blood-2005-02-0560. [DOI] [PubMed] [Google Scholar]
- (490).Spencer DH; Young MA; Lamprecht TL; Helton NM; Fulton R; O’Laughlin M; Fronick C; Magrini V; Demeter RT; Miller CA; Klco JM; Wilson RK; Ley TJ Epigenomic Analysis of the HOX Gene Loci Reveals Mechanisms That May Control Canonical Expression Patterns in AML and Normal Hematopoietic Cells. Leukemia 2015, 29 (6), 1279–1289. 10.1038/leu.2015.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (491).Brunetti L; Gundry MC; Sorcini D; Guzman AG; Huang Y-H; Ramabadran R; Gionfriddo I; Mezzasoma F; Milano F; Nabet B; Buckley DL; Kornblau SM; Lin CY; Sportoletti P; Martelli MP; Falini B; Goodell MA Mutant NPM1 Maintains the Leukemic State through HOX Expression. Cancer Cell 2018, 34 (3), 499–512.e9. 10.1016/j.ccell.2018.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (492).Thomas X Small Molecule Menin Inhibitors: Novel Therapeutic Agents Targeting Acute Myeloid Leukemia with KMT2A Rearrangement or NPM1 Mutation. Oncol Ther 2024, 12 (1), 57–72. 10.1007/s40487-024-00262-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (493).Issa GC; Ravandi F; DiNardo CD; Jabbour E; Kantarjian HM; Andreeff M Therapeutic Implications of Menin Inhibition in Acute Leukemias. Leukemia 2021, 35 (9), 2482–2495. 10.1038/s41375-021-01309-y. [DOI] [PubMed] [Google Scholar]
- (494).Kwon MC; Querolle O; Dai X; Thuring JW; Verhulst T; Marien A; Goffin D; Cai W; Keersmaekers V; Eyassu F; Verstraeten K; El Ashkar S; Hogeling SM; Jacob F; Vinken P; Darville N; Pande V; Krosky D; Urbanietz G; Verbist B; Ferrante LA; Drenberg CD; Wilson D; Attar RM; Schuringa JJ; Daskalakis N; Packman K; Pietsch C; Elsayed Y; Philippar U Pharmacological Characterization of JNJ-75276617, a Menin-KMT2A Inhibitor, As Targeted Treatment for KMT2A-Altered and NPM1-Mutant Acute Leukemia. Blood 2022, 140 (Supplement 1), 5928–5929. 10.1182/blood-2022-168921. [DOI] [Google Scholar]
- (495).Zhou H; Liu L; Huang J; Bernard D; Karatas H; Navarro A; Lei M; Wang S Structure-Based Design of High-Affinity Macrocyclic Peptidomimetics to Block the Menin-Mixed Lineage Leukemia 1 (MLL1) Protein-Protein Interaction. J Med Chem 2013, 56 (3), 1113–1123. 10.1021/jm3015298. [DOI] [PubMed] [Google Scholar]
- (496).Xu S; Aguilar A; Xu T; Zheng K; Huang L; Stuckey J; Chinnaswamy K; Bernard D; Fernández-Salas E; Liu L; Wang M; McEachern D; Przybranowski S; Foster C; Wang S Design of the First-in-Class, Highly Potent Irreversible Inhibitor Targeting the Menin-MLL Protein-Protein Interaction. Angew Chem Int Ed Engl 2018, 57 (6), 1601–1605. 10.1002/anie.201711828. [DOI] [PubMed] [Google Scholar]
- (497).Zhang M; Aguilar A; Xu S; Huang L; Chinnaswamy K; Sleger T; Wang B; Gross S; Nicolay BN; Ronseaux S; Harvey K; Wang Y; McEachern D; Kirchhoff PD; Liu Z; Stuckey J; Tron AE; Liu T; Wang S Discovery of M-1121 as an Orally Active Covalent Inhibitor of Menin-MLL Interaction Capable of Achieving Complete and Long-Lasting Tumor Regression. J Med Chem 2021, 64 (14), 10333–10349. 10.1021/acs.jmedchem.1c00789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (498).Xu S; Aguilar A; Huang L; Xu T; Zheng K; McEachern D; Przybranowski S; Foster C; Zawacki K; Liu Z; Chinnaswamy K; Stuckey J; Wang S Discovery of M-808 as a Highly Potent, Covalent, Small-Molecule Inhibitor of the Menin-MLL Interaction with Strong In Vivo Antitumor Activity. J Med Chem 2020, 63 (9), 4997–5010. 10.1021/acs.jmedchem.0c00547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (499).Lancet J; Ravandi F; Montesinos P; Barrientos JC; Badar T; Alegre A; Bashey A; Bergua Burgues JM; Brunetti L; Curran EK; de Leeuw DCC; Derenzini E; Huls GA; Janssen JJWM; Jeyakumar D; Jongen-Lavrencic M; Kishtagari A; Mateos M-V; Morillo D; Rambaldi A; Rosenberg AS; Schiller GJ; Ahmed U; Cacovean A; Morris SW; Follit C; Munneke B; Waghmare G; Mandap C; Vimal M; Butler T; Carraway HE Covalent Menin Inhibitor Bmf-219 in Patients with Relapsed or Refractory (R/R) Acute Leukemia (AL): Preliminary Phase 1 Data from the Covalent-101 Study. Blood 2023, 142 (Supplement 1), 2916–2916. 10.1182/blood-2023-173149. [DOI] [Google Scholar]
- (500).Candoni A; Coppola GA 2024 Update on Menin Inhibitors. A New Class of Target Agents against KMT2A-Rearranged and NPM1-Mutated Acute Myeloid Leukemia. Hematol Rep 2024, 16 (2), 244–254. 10.3390/hematolrep16020024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (501).Soto-Feliciano YM; Sánchez-Rivera FJ; Perner F; Barrows DW; Kastenhuber ER; Ho Y-J; Carroll T; Xiong Y; Anand D; Soshnev AA; Gates L; Beytagh MC; Cheon D; Gu S; Liu XS; Krivtsov AV; Meneses M; de Stanchina E; Stone RM; Armstrong SA; Lowe SW; Allis CD A Molecular Switch between Mammalian MLL Complexes Dictates Response to Menin-MLL Inhibition. Cancer Discov 2023, 13 (1), 146–169. 10.1158/2159-8290.CD-22-0416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (502).Malik R; Khan AP; Asangani IA; Cieślik M; Prensner JR; Wang X; Iyer MK; Jiang X; Borkin D; Escara-Wilke J; Stender R; Wu Y-M; Niknafs YS; Jing X; Qiao Y; Palanisamy N; Kunju LP; Krishnamurthy PM; Yocum AK; Mellacheruvu D; Nesvizhskii AI; Cao X; Dhanasekaran SM; Feng FY; Grembecka J; Cierpicki T; Chinnaiyan AM Targeting the MLL Complex in Castration-Resistant Prostate Cancer. Nat Med 2015, 21 (4), 344–352. 10.1038/nm.3830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (503).Imachi H; Murao K; Dobashi H; Bhuyan MM; Cao X; Kontani K; Niki S; Murazawa C; Nakajima H; Kohno N; Yamashita H; Iwase H; Hayashi S; Ishida T; Yamauchi A Menin, a Product of the MENI Gene, Binds to Estrogen Receptor to Enhance Its Activity in Breast Cancer Cells: Possibility of a Novel Predictive Factor for Tamoxifen Resistance. Breast Cancer Res Treat 2010, 122 (2), 395–407. 10.1007/s10549-009-0581-0. [DOI] [PubMed] [Google Scholar]
- (504).Svoboda LK; Teh SSK; Sud S; Kerk S; Zebolsky A; Treichel S; Thomas D; Halbrook CJ; Lee H-J; Kremer D; Zhang L; Klossowski S; Bankhead AR; Magnuson B; Ljungman M; Cierpicki T; Grembecka J; Lyssiotis CA; Lawlor ER Menin Regulates the Serine Biosynthetic Pathway in Ewing Sarcoma. J Pathol 2018, 245 (3), 324–336. 10.1002/path.5085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (505).Kurmasheva RT; Bandyopadhyay A; Favours E; Del Pozo V; Ghilu S; Phelps DA; McGeehan GM; Erickson SW; Smith MA; Houghton PJ Evaluation of VTP-50469, a Menin-MLL1 Inhibitor, against Ewing Sarcoma Xenograft Models by the Pediatric Preclinical Testing Consortium. Pediatr Blood Cancer 2020, 67 (7), e28284. 10.1002/pbc.28284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (506).Thiel AT; Blessington P; Zou T; Feather D; Wu X; Yan J; Zhang H; Liu Z; Ernst P; Koretzky GA; Hua X MLL-AF9-Induced Leukemogenesis Requires Coexpression of the Wild-Type Mll Allele. Cancer Cell 2010, 17 (2), 148–159. 10.1016/j.ccr.2009.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (507).Milne TA; Martin ME; Brock HW; Slany RK; Hess JL Leukemogenic MLL Fusion Proteins Bind across a Broad Region of the Hox A9 Locus, Promoting Transcription and Multiple Histone Modifications. Cancer Res 2005, 65 (24), 11367–11374. 10.1158/0008-5472.CAN-05-1041. [DOI] [PubMed] [Google Scholar]
- (508).Patel A; Vought VE; Dharmarajan V; Cosgrove MS A Conserved Arginine-Containing Motif Crucial for the Assembly and Enzymatic Activity of the Mixed Lineage Leukemia Protein-1 Core Complex. J Biol Chem 2008, 283 (47), 32162–32175. 10.1074/jbc.M806317200. [DOI] [PubMed] [Google Scholar]
- (509).Dou Y; Milne TA; Ruthenburg AJ; Lee S; Lee JW; Verdine GL; Allis CD; Roeder RG Regulation of MLL1 H3K4 Methyltransferase Activity by Its Core Components. Nat Struct Mol Biol 2006, 13 (8), 713–719. 10.1038/nsmb1128. [DOI] [PubMed] [Google Scholar]
- (510).Odho Z; Southall SM; Wilson JR Characterization of a Novel WDR5-Binding Site That Recruits RbBP5 through a Conserved Motif to Enhance Methylation of Histone H3 Lysine 4 by Mixed Lineage Leukemia Protein-1. J Biol Chem 2010, 285 (43), 32967–32976. 10.1074/jbc.M110.159921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (511).Roguev A; Schaft D; Shevchenko A; Pijnappel WW; Wilm M; Aasland R; Stewart AF The Saccharomyces Cerevisiae Set1 Complex Includes an Ash2 Homologue and Methylates Histone 3 Lysine 4. EMBO J 2001, 20 (24), 7137–7148. 10.1093/emboj/20.24.7137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (512).Song J-J; Kingston RE WDR5 Interacts with Mixed Lineage Leukemia (MLL) Protein via the Histone H3-Binding Pocket. J Biol Chem 2008, 283 (50), 35258–35264. 10.1074/jbc.M806900200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (513).Karatas H; Townsend EC; Bernard D; Dou Y; Wang S Analysis of the Binding of Mixed Lineage Leukemia 1 (MLL1) and Histone 3 Peptides to WD Repeat Domain 5 (WDR5) for the Design of Inhibitors of the MLL1-WDR5 Interaction. J Med Chem 2010, 53 (14), 5179–5185. 10.1021/jm100139b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (514).Karatas H; Townsend EC; Cao F; Chen Y; Bernard D; Liu L; Lei M; Dou Y; Wang S High-Affinity, Small-Molecule Peptidomimetic Inhibitors of MLL1/WDR5 Protein-Protein Interaction. J Am Chem Soc 2013, 135 (2), 669–682. 10.1021/ja306028q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (515).Cao F; Townsend EC; Karatas H; Xu J; Li L; Lee S; Liu L; Chen Y; Ouillette P; Zhu J; Hess JL; Atadja P; Lei M; Qin ZS; Malek S; Wang S; Dou Y Targeting MLL1 H3K4 Methyltransferase Activity in Mixed-Lineage Leukemia. Mol Cell 2014, 53 (2), 247–261. 10.1016/j.molcel.2013.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (516).Karatas H; Li Y; Liu L; Ji J; Lee S; Chen Y; Yang J; Huang L; Bernard D; Xu J; Townsend EC; Cao F; Ran X; Li X; Wen B; Sun D; Stuckey JA; Lei M; Dou Y; Wang S Discovery of a Highly Potent, Cell-Permeable Macrocyclic Peptidomimetic (MM-589) Targeting the WD Repeat Domain 5 Protein (WDR5)-Mixed Lineage Leukemia (MLL) Protein-Protein Interaction. J Med Chem 2017, 60 (12), 4818–4839. 10.1021/acs.jmedchem.6b01796. [DOI] [PubMed] [Google Scholar]
- (517).Aho ER; Wang J; Gogliotti RD; Howard GC; Phan J; Acharya P; Macdonald JD; Cheng K; Lorey SL; Lu B; Wenzel S; Foshage AM; Alvarado J; Wang F; Shaw JG; Zhao B; Weissmiller AM; Thomas LR; Vakoc CR; Hall MD; Hiebert SW; Liu Q; Stauffer SR; Fesik SW; Tansey WP Displacement of WDR5 from Chromatin by a WIN Site Inhibitor with Picomolar Affinity. Cell Rep 2019, 26 (11), 2916–2928.e13. 10.1016/j.celrep.2019.02.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (518).Tian J; Teuscher KB; Aho ER; Alvarado JR; Mills JJ; Meyers KM; Gogliotti RD; Han C; Macdonald JD; Sai J; Shaw JG; Sensintaffar JL; Zhao B; Rietz TA; Thomas LR; Payne WG; Moore WJ; Stott GM; Kondo J; Inoue M; Coffey RJ; Tansey WP; Stauffer SR; Lee T; Fesik SW Discovery and Structure-Based Optimization of Potent and Selective WD Repeat Domain 5 (WDR5) Inhibitors Containing a Dihydroisoquinolinone Bicyclic Core. J Med Chem 2020, 63 (2), 656–675. 10.1021/acs.jmedchem.9b01608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (519).Wang F; Jeon KO; Salovich JM; Macdonald JD; Alvarado J; Gogliotti RD; Phan J; Olejniczak ET; Sun Q; Wang S; Camper D; Yuh JP; Shaw JG; Sai J; Rossanese OW; Tansey WP; Stauffer SR; Fesik SW Discovery of Potent 2-Aryl-6,7-Dihydro-5 H-Pyrrolo[1,2- a]Imidazoles as WDR5-WIN-Site Inhibitors Using Fragment-Based Methods and Structure-Based Design. J Med Chem 2018, 61 (13), 5623–5642. 10.1021/acs.jmedchem.8b00375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (520).Teuscher KB; Chowdhury S; Meyers KM; Tian J; Sai J; Van Meveren M; South TM; Sensintaffar JL; Rietz TA; Goswami S; Wang J; Grieb BC; Lorey SL; Howard GC; Liu Q; Moore WJ; Stott GM; Tansey WP; Lee T; Fesik SW Structure-Based Discovery of Potent WD Repeat Domain 5 Inhibitors That Demonstrate Efficacy and Safety in Preclinical Animal Models. Proc Natl Acad Sci U S A 2023, 120 (1), 16783–16806. 10.1073/pnas.2211297120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (521).Teuscher KB; Mills JJ; Tian J; Han C; Meyers KM; Sai J; South TM; Crow MM; Van Meveren M; Sensintaffar JL; Zhao B; Amporndanai K; Moore WJ; Stott GM; Tansey WP; Lee T; Fesik SW Structure-Based Discovery of Potent, Orally Bioavailable Benzoxazepinone-Based WD Repeat Domain 5 Inhibitors. J Med Chem 2023, 66 (24), 16783–16806. 10.1021/acs.jmedchem.3c01529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (522).Senisterra G; Wu H; Allali-Hassani A; Wasney GA; Barsyte-Lovejoy D; Dombrovski L; Dong A; Nguyen KT; Smil D; Bolshan Y; Hajian T; He H; Seitova A; Chau I; Li F; Poda G; Couture J-F; Brown PJ; Al-Awar R; Schapira M; Arrowsmith CH; Vedadi M Small-Molecule Inhibition of MLL Activity by Disruption of Its Interaction with WDR5. Biochem J 2013, 449 (1), 151–159. 10.1042/BJ20121280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (523).Bolshan Y; Getlik M; Kuznetsova E; Wasney GA; Hajian T; Poda G; Nguyen KT; Wu H; Dombrovski L; Dong A; Senisterra G; Schapira M; Arrowsmith CH; Brown PJ; Al-Awar R; Vedadi M; Smil D Synthesis, Optimization, and Evaluation of Novel Small Molecules as Antagonists of WDR5-MLL Interaction. ACS Med Chem Lett 2013, 4 (3), 353–357. 10.1021/ml300467n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (524).Getlik M; Smil D; Zepeda-Velázquez C; Bolshan Y; Poda G; Wu H; Dong A; Kuznetsova E; Marcellus R; Senisterra G; Dombrovski L; Hajian T; Kiyota T; Schapira M; Arrowsmith CH; Brown PJ; Vedadi M; Al-Awar R Structure-Based Optimization of a Small Molecule Antagonist of the Interaction Between WD Repeat-Containing Protein 5 (WDR5) and Mixed-Lineage Leukemia 1 (MLL1). J Med Chem 2016, 59 (6), 2478–2496. 10.1021/acs.jmedchem.5b01630. [DOI] [PubMed] [Google Scholar]
- (525).Grebien F; Vedadi M; Getlik M; Giambruno R; Grover A; Avellino R; Skucha A; Vittori S; Kuznetsova E; Smil D; Barsyte-Lovejoy D; Li F; Poda G; Schapira M; Wu H; Dong A; Senisterra G; Stukalov A; Huber KVM; Schönegger A; Marcellus R; Bilban M; Bock C; Brown PJ; Zuber J; Bennett KL; Al-Awar R; Delwel R; Nerlov C; Arrowsmith CH; Superti-Furga G Pharmacological Targeting of the Wdr5-MLL Interaction in C/EBPα N-Terminal Leukemia. Nat Chem Biol 2015, 11 (8), 571–578. 10.1038/nchembio.1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (526).Li D-D; Chen W-L; Xu X-L; Jiang F; Wang L; Xie Y-Y; Zhang X-J; Guo X-K; You Q-D; Sun H-P Structure-Based Design and Synthesis of Small Molecular Inhibitors Disturbing the Interaction of MLL1-WDR5. Eur J Med Chem 2016, 118, 1–8. 10.1016/j.ejmech.2016.04.032. [DOI] [PubMed] [Google Scholar]
- (527).Li D-D; Chen W-L; Wang Z-H; Xie Y-Y; Xu X-L; Jiang Z-Y; Zhang X-J; You Q-D; Guo X-K High-Affinity Small Molecular Blockers of Mixed Lineage Leukemia 1 (MLL1)-WDR5 Interaction Inhibit MLL1 Complex H3K4 Methyltransferase Activity. Eur J Med Chem 2016, 124, 480–489. 10.1016/j.ejmech.2016.08.036. [DOI] [PubMed] [Google Scholar]
- (528).Chen W; Chen X; Li D; Zhou J; Jiang Z; You Q; Guo X Discovery of DDO-2213 as a Potent and Orally Bioavailable Inhibitor of the WDR5-Mixed Lineage Leukemia 1 Protein-Protein Interaction for the Treatment of MLL Fusion Leukemia. J Med Chem 2021, 64 (12), 8221–8245. 10.1021/acs.jmedchem.1c00091. [DOI] [PubMed] [Google Scholar]
- (529).Chen W; Chen X; Li D; Wang X; Long G; Jiang Z; You Q; Guo X Discovery of a Potent MLL1 and WDR5 Protein-Protein Interaction Inhibitor with in Vivo Antitumor Activity. Eur J Med Chem 2021, 223, 113677. 10.1016/j.ejmech.2021.113677. [DOI] [PubMed] [Google Scholar]
- (530).Dölle A; Adhikari B; Krämer A; Weckesser J; Berner N; Berger L-M; Diebold M; Szewczyk MM; Barsyte-Lovejoy D; Arrowsmith CH; Gebel J; Löhr F; Dötsch V; Eilers M; Heinzlmeir S; Kuster B; Sotriffer C; Wolf E; Knapp S Design, Synthesis, and Evaluation of WD-Repeat-Containing Protein 5 (WDR5) Degraders. J Med Chem 2021, 64 (15), 10682–10710. 10.1021/acs.jmedchem.1c00146. [DOI] [PubMed] [Google Scholar]
- (531).Yu X; Li D; Kottur J; Shen Y; Kim HS; Park K-S; Tsai Y-H; Gong W; Wang J; Suzuki K; Parker J; Herring L; Kaniskan HÜ; Cai L; Jain R; Liu J; Aggarwal AK; Wang GG; Jin J A Selective WDR5 Degrader Inhibits Acute Myeloid Leukemia in Patient-Derived Mouse Models. Sci Transl Med 2021, 13 (613), eabj1578. 10.1126/scitranslmed.abj1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (532).Yu X; Li D; Kottur J; Kim HS; Herring LE; Yu Y; Xie L; Hu X; Chen X; Cai L; Liu J; Aggarwal AK; Wang GG; Jin J Discovery of Potent and Selective WDR5 Proteolysis Targeting Chimeras as Potential Therapeutics for Pancreatic Cancer. J Med Chem 2023, 66 (23), 16168–16186. 10.1021/acs.jmedchem.3c01521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (533).Li D; Yu X; Kottur J; Gong W; Zhang Z; Storey AJ; Tsai Y-H; Uryu H; Shen Y; Byrum SD; Edmondson RD; Mackintosh SG; Cai L; Liu Z; Aggarwal AK; Tackett AJ; Liu J; Jin J; Wang GG Discovery of a Dual WDR5 and Ikaros PROTAC Degrader as an Anti-Cancer Therapeutic. Oncogene 2022, 41 (24), 3328–3340. 10.1038/s41388-022-02340-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (534).Thomas LR; Adams CM; Fesik SW; Eischen CM; Tansey WP Targeting MYC through WDR5. Mol Cell Oncol 2020, 7 (2), 1709388. 10.1080/23723556.2019.1709388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (535).Thomas LR; Wang Q; Grieb BC; Phan J; Foshage AM; Sun Q; Olejniczak ET; Clark T; Dey S; Lorey S; Alicie B; Howard GC; Cawthon B; Ess KC; Eischen CM; Zhao Z; Fesik SW; Tansey WP Interaction with WDR5 Promotes Target Gene Recognition and Tumorigenesis by MYC. Mol Cell 2015, 58 (3), 440–452. 10.1016/j.molcel.2015.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (536).Thomas LR; Adams CM; Wang J; Weissmiller AM; Creighton J; Lorey SL; Liu Q; Fesik SW; Eischen CM; Tansey WP Interaction of the Oncoprotein Transcription Factor MYC with Its Chromatin Cofactor WDR5 Is Essential for Tumor Maintenance. Proc Natl Acad Sci U S A 2019, 116 (50), 25260–25268. 10.1073/pnas.1910391116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (537).Macdonald JD; Chacón Simon S; Han C; Wang F; Shaw JG; Howes JE; Sai J; Yuh JP; Camper D; Alicie BM; Alvarado J; Nikhar S; Payne W; Aho ER; Bauer JA; Zhao B; Phan J; Thomas LR; Rossanese OW; Tansey WP; Waterson AG; Stauffer SR; Fesik SW Discovery and Optimization of Salicylic Acid-Derived Sulfonamide Inhibitors of the WD Repeat-Containing Protein 5-MYC Protein-Protein Interaction. J Med Chem 2019, 62 (24), 11232–11259. 10.1021/acs.jmedchem.9b01411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (538).Chacón Simon S; Wang F; Thomas LR; Phan J; Zhao B; Olejniczak ET; Macdonald JD; Shaw JG; Schlund C; Payne W; Creighton J; Stauffer SR; Waterson AG; Tansey WP; Fesik SW Discovery of WD Repeat-Containing Protein 5 (WDR5)-MYC Inhibitors Using Fragment-Based Methods and Structure-Based Design. J Med Chem 2020, 63 (8), 4315–4333. 10.1021/acs.jmedchem.0c00224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (539).duPuy Guarnaccia A; Tansey WP Moonlighting with WDR5: A Cellular Multitasker. J Clin Med 2018, 7 (2), 21. 10.3390/jcm7020021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (540).Siladi AJ; Wang J; Florian AC; Thomas LR; Creighton JH; Matlock BK; Flaherty DK; Lorey SL; Howard GC; Fesik SW; Weissmiller AM; Liu Q; Tansey WP WIN Site Inhibition Disrupts a Subset of WDR5 Function. Sci Rep 2022, 12 (1), 1848. 10.1038/s41598-022-05947-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (541).Bryan AF; Wang J; Howard GC; Guarnaccia AD; Woodley CM; Aho ER; Rellinger EJ; Matlock BK; Flaherty DK; Lorey SL; Chung DH; Fesik SW; Liu Q; Weissmiller AM; Tansey WP WDR5 Is a Conserved Regulator of Protein Synthesis Gene Expression. Nucleic Acids Res 2020, 48 (6), 2924–2941. 10.1093/nar/gkaa051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (542).Howard GC; Wang J; Rose KL; Jones C; Patel P; Tsui T; Florian AC; Vlach L; Lorey SL; Grieb BC; Smith BN; Slota MJ; Reynolds EM; Goswami S; Savona MR; Mason FM; Lee T; Fesik S; Liu Q; Tansey WP Ribosome Subunit Attrition and Activation of the P53-MDM4 Axis Dominate the Response of MLL-Rearranged Cancer Cells to WDR5 WIN Site Inhibition. Elife 2024, 12, RP90683. 10.7554/eLife.90683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (543).Zhu J; Sammons MA; Donahue G; Dou Z; Vedadi M; Getlik M; Barsyte-Lovejoy D; Al-awar R; Katona BW; Shilatifard A; Huang J; Hua X; Arrowsmith CH; Berger SL Gain-of-Function P53 Mutants Co-Opt Chromatin Pathways to Drive Cancer Growth. Nature 2015, 525 (7568), 206–211. 10.1038/nature15251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (544).Neilsen BK; Chakraborty B; McCall JL; Frodyma DE; Sleightholm RL; Fisher KW; Lewis RE WDR5 Supports Colon Cancer Cells by Promoting Methylation of H3K4 and Suppressing DNA Damage. BMC Cancer 2018, 18 (1), 673. 10.1186/s12885-018-4580-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (545).Punzi S; Balestrieri C; D’Alesio C; Bossi D; Dellino GI; Gatti E; Pruneri G; Criscitiello C; Lovati G; Meliksetyan M; Carugo A; Curigliano G; Natoli G; Pelicci PG; Lanfrancone L WDR5 Inhibition Halts Metastasis Dissemination by Repressing the Mesenchymal Phenotype of Breast Cancer Cells. Breast Cancer Res 2019, 21 (1), 123. 10.1186/s13058-019-1216-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (546).Chen X; Xu J; Wang X; Long G; You Q; Guo X Targeting WD Repeat-Containing Protein 5 (WDR5): A Medicinal Chemistry Perspective. J Med Chem 2021, 64 (15), 10537–10556. 10.1021/acs.jmedchem.1c00037. [DOI] [PubMed] [Google Scholar]
- (547).Lu C; Liu Z; Klement JD; Yang D; Merting AD; Poschel D; Albers T; Waller JL; Shi H; Liu K WDR5-H3K4me3 Epigenetic Axis Regulates OPN Expression to Compensate PD-L1 Function to Promote Pancreatic Cancer Immune Escape. J Immunother Cancer 2021, 9 (7), e002624. 10.1136/jitc-2021-002624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (548).Zhang J; Zhou Q; Xie K; Cheng L; Peng S; Xie R; Liu L; Zhang Y; Dong W; Han J; Huang M; Chen Y; Lin T; Huang J; Chen X Targeting WD Repeat Domain 5 Enhances Chemosensitivity and Inhibits Proliferation and Programmed Death-Ligand 1 Expression in Bladder Cancer. J Exp Clin Cancer Res 2021, 40 (1), 203. 10.1186/s13046-021-01989-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (549).Erb MA; Scott TG; Li BE; Xie H; Paulk J; Seo H-S; Souza A; Roberts JM; Dastjerdi S; Buckley DL; Sanjana NE; Shalem O; Nabet B; Zeid R; Offei-Addo NK; Dhe-Paganon S; Zhang F; Orkin SH; Winter GE; Bradner JE Transcription Control by the ENL YEATS Domain in Acute Leukaemia. Nature 2017, 543 (7644), 270–274. 10.1038/nature21688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (550).Wan L; Wen H; Li Y; Lyu J; Xi Y; Hoshii T; Joseph JK; Wang X; Loh Y-HE; Erb MA; Souza AL; Bradner JE; Shen L; Li W; Li H; Allis CD; Armstrong SA; Shi X ENL Links Histone Acetylation to Oncogenic Gene Expression in Acute Myeloid Leukaemia. Nature 2017, 543 (7644), 265–269. 10.1038/nature21687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (551).Le Masson I; Yu DY; Jensen K; Chevalier A; Courbeyrette R; Boulard Y; Smith MM; Mann C Yaf9, a Novel NuA4 Histone Acetyltransferase Subunit, Is Required for the Cellular Response to Spindle Stress in Yeast. Mol Cell Biol 2003, 23 (17), 6086–6102. 10.1128/MCB.23.17.6086-6102.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (552).Wang AY; Schulze JM; Skordalakes E; Gin JW; Berger JM; Rine J; Kobor MS Asf1-like Structure of the Conserved Yaf9 YEATS Domain and Role in H2A.Z Deposition and Acetylation. Proc Natl Acad Sci U S A 2009, 106 (51), 21573–21578. 10.1073/pnas.0906539106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (553).Schulze JM; Wang AY; Kobor MS YEATS Domain Proteins: A Diverse Family with Many Links to Chromatin Modification and TranscriptionThis Paper Is One of a Selection of Papers Published in This Special Issue, Entitled CSBMCB’s 51st Annual Meeting – Epigenetics and Chromatin Dynamics, and Has Undergone the Journal’s Usual Peer Review Process. Biochemistry and Cell Biology 2009, 87 (1), 65–75. 10.1139/O08-111. [DOI] [PubMed] [Google Scholar]
- (554).Li Y; Sabari BR; Panchenko T; Wen H; Zhao D; Guan H; Wan L; Huang H; Tang Z; Zhao Y; Roeder RG; Shi X; Allis CD; Li H Molecular Coupling of Histone Crotonylation and Active Transcription by AF9 YEATS Domain. Mol Cell 2016, 62 (2), 181–193. 10.1016/j.molcel.2016.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (555).Andrews FH; Shinsky SA; Shanle EK; Bridgers JB; Gest A; Tsun IK; Krajewski K; Shi X; Strahl BD; Kutateladze TG The Taf14 YEATS Domain Is a Reader of Histone Crotonylation. Nat Chem Biol 2016, 12 (6), 396–398. 10.1038/nchembio.2065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (556).Zhang Q; Zeng L; Zhao C; Ju Y; Konuma T; Zhou M-M Structural Insights into Histone Crotonyl-Lysine Recognition by the AF9 YEATS Domain. Structure 2016, 24 (9), 1606–1612. 10.1016/j.str.2016.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (557).Zhao D; Guan H; Zhao S; Mi W; Wen H; Li Y; Zhao Y; Allis CD; Shi X; Li H YEATS2 Is a Selective Histone Crotonylation Reader. Cell Res 2016, 26 (5), 629–632. 10.1038/cr.2016.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (558).Mi W; Guan H; Lyu J; Zhao D; Xi Y; Jiang S; Andrews FH; Wang X; Gagea M; Wen H; Tora L; Dent SYR; Kutateladze TG; Li W; Li H; Shi X YEATS2 Links Histone Acetylation to Tumorigenesis of Non-Small Cell Lung Cancer. Nat Commun 2017, 8 (1), 1088. 10.1038/s41467-017-01173-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (559).Moustakim M; Christott T; Monteiro OP; Bennett J; Giroud C; Ward J; Rogers CM; Smith P; Panagakou I; Díaz-Sáez L; Felce SL; Gamble V; Gileadi C; Halidi N; Heidenreich D; Chaikuad A; Knapp S; Huber KVM; Farnie G; Heer J; Manevski N; Poda G; Al-Awar R; Dixon DJ; Brennan PE; Fedorov O Discovery of an MLLT1/3 YEATS Domain Chemical Probe. Angew Chem Int Ed Engl 2018, 57 (50), 16302–16307. 10.1002/anie.201810617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (560).Krone MW; Travis CR; Lee GY; Eckvahl HJ; Houk KN; Waters ML More Than π-π-π Stacking: Contribution of Amide-π and CH-π Interactions to Crotonyllysine Binding by the AF9 YEATS Domain. J Am Chem Soc 2020, 142 (40), 17048–17056. 10.1021/jacs.0c06568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (561).Travis CR; Francis DY; Williams DC; Waters ML Evaluation of Acyllysine Isostere Interactions with the Aromatic Pocket of the AF9 YEATS Domain. Protein Sci 2023, 32 (1), e4533. 10.1002/pro.4533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (562).Garnar-Wortzel L; Bishop TR; Kitamura S; Milosevich N; Asiaban JN; Zhang X; Zheng Q; Chen E; Ramos AR; Ackerman CJ; Hampton EN; Chatterjee AK; Young TS; Hull MV; Sharpless KB; Cravatt BF; Wolan DW; Erb MA Chemical Inhibition of ENL/AF9 YEATS Domains in Acute Leukemia. ACS Cent Sci 2021, 7 (5), 815–830. 10.1021/acscentsci.0c01550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (563).Shaum JB; Steen EA; Muñoz i Ordoño M; Wenge DV; Cheong H; Hunkeler M; Bilotta EM; Rutter Z; Barta PA; Milosevich N; Hargis LM; Janowski J; Bishop TR; Carter TR; da Camara B; Hinterndorfer M; Dada L; He W-J; Offensperger F; Furihata H; Schweber SR; Hatton C; Wen Y; Cravatt BF; Engle KM; Melillo B; Kitamura S; Ciulli A; Armstrong SA; Fischer ES; Winter GE; Erb MA High-Throughput Diversification of Protein-Ligand Surfaces to Discover Chemical Inducers of Proximity. 2024. 10.1101/2024.09.30.615685. [DOI] [Google Scholar]
- (564).Guo XS; Atla S; Nyalata S; Alugubelli YR; Chen P-HC; Xu S; Liu WR Prioritization of Eleven-Nineteen-Leukemia Inhibitors as Orally Available Drug Candidates for Acute Myeloid Leukemia. J Med Chem 2024, 67 (22), 20100–20117. 10.1021/acs.jmedchem.4c01337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (565).Michino M; Khan TA; Miller MW; Fukase Y; Vendome J; Adura C; Glickman JF; Liu Y; Wan L; Allis CD; Stamford AW; Meinke PT; Renzetti LM; Kargman S; Liverton NJ; Huggins DJ Lead Optimization of Small Molecule ENL YEATS Inhibitors to Enable In Vivo Studies: Discovery of TDI-11055. ACS Med Chem Lett 2024, 15 (4), 524–532. 10.1021/acsmedchemlett.4c00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (566).Raux B; Buchan KA; Bennett J; Christott T; Dowling MS; Farnie G; Fedorov O; Gamble V; Gileadi C; Giroud C; Huber KVM; Korczynska M; Limberakis C; Narayanan A; Owen DR; Sáez LD; Stock IA; Londregan AT Discovery of PFI-6, a Small-Molecule Chemical Probe for the YEATS Domain of MLLT1 and MLLT3. Bioorg Med Chem Lett 2024, 98, 129546. 10.1016/j.bmcl.2023.129546. [DOI] [PubMed] [Google Scholar]
- (567).Liu Y; Li Q; Alikarami F; Barrett DR; Mahdavi L; Li H; Tang S; Khan TA; Michino M; Hill C; Song L; Yang L; Li Y; Pokharel SP; Stamford AW; Liverton N; Renzetti LM; Taylor S; Watt GF; Ladduwahetty T; Kargman S; Meinke PT; Foley MA; Shi J; Li H; Carroll M; Chen C-W; Gardini A; Maillard I; Huggins DJ; Bernt KM; Wan L Small-Molecule Inhibition of the Acyl-Lysine Reader ENL as a Strategy against Acute Myeloid Leukemia. Cancer Discov 2022, 12 (11), 2684–2709. 10.1158/2159-8290.CD-21-1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (568).Ma XR; Xu L; Xu S; Klein BJ; Wang H; Das S; Li K; Yang KS; Sohail S; Chapman A; Kutateladze TG; Shi X; Liu WR; Wen H Discovery of Selective Small-Molecule Inhibitors for the ENL YEATS Domain. J Med Chem 2021, 64 (15), 10997–11013. 10.1021/acs.jmedchem.1c00367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (569).Asiaban JN; Milosevich N; Chen E; Bishop TR; Wang J; Zhang Y; Ackerman CJ; Hampton EN; Young TS; Hull MV; Cravatt BF; Erb MA Cell-Based Ligand Discovery for the ENL YEATS Domain. ACS Chem Biol 2020, 15 (4), 895–903. 10.1021/acschembio.0c00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (570).Ni X; Heidenreich D; Christott T; Bennett J; Moustakim M; Brennan PE; Fedorov O; Knapp S; Chaikuad A Structural Insights into Interaction Mechanisms of Alternative Piperazine-Urea YEATS Domain Binders in MLLT1. ACS Med Chem Lett 2019, 10 (12), 1661–1666. 10.1021/acsmedchemlett.9b00460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (571).Christott T; Bennett J; Coxon C; Monteiro O; Giroud C; Beke V; Felce SL; Gamble V; Gileadi C; Poda G; Al-Awar R; Farnie G; Fedorov O Discovery of a Selective Inhibitor for the YEATS Domains of ENL/AF9. SLAS Discov 2019, 24 (2), 133–141. 10.1177/2472555218809904. [DOI] [PubMed] [Google Scholar]
- (572).Walz AL; Ooms A; Gadd S; Gerhard DS; Smith MA; Guidry Auvil JM; Meerzaman D; Chen Q-R; Hsu CH; Yan C; Nguyen C; Hu Y; Bowlby R; Brooks D; Ma Y; Mungall AJ; Moore RA; Schein J; Marra MA; Huff V; Dome JS; Chi Y-Y; Mullighan CG; Ma J; Wheeler DA; Hampton OA; Jafari N; Ross N; Gastier-Foster JM; Perlman EJ Recurrent DGCR8, DROSHA, and SIX Homeodomain Mutations in Favorable Histology Wilms Tumors. Cancer Cell 2015, 27 (2), 286–297. 10.1016/j.ccell.2015.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (573).Perlman EJ; Gadd S; Arold ST; Radhakrishnan A; Gerhard DS; Jennings L; Huff V; Guidry Auvil JM; Davidsen TM; Dome JS; Meerzaman D; Hsu CH; Nguyen C; Anderson J; Ma Y; Mungall AJ; Moore RA; Marra MA; Mullighan CG; Ma J; Wheeler DA; Hampton OA; Gastier-Foster JM; Ross N; Smith MA MLLT1 YEATS Domain Mutations in Clinically Distinctive Favourable Histology Wilms Tumours. Nat Commun 2015, 6, 10013. 10.1038/ncomms10013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (574).Gadd S; Huff V; Walz AL; Ooms AHAG; Armstrong AE; Gerhard DS; Smith MA; Auvil JMG; Meerzaman D; Chen Q-R; Hsu CH; Yan C; Nguyen C; Hu Y; Hermida LC; Davidsen T; Gesuwan P; Ma Y; Zong Z; Mungall AJ; Moore RA; Marra MA; Dome JS; Mullighan CG; Ma J; Wheeler DA; Hampton OA; Ross N; Gastier-Foster JM; Arold ST; Perlman EJ A Children’s Oncology Group and TARGET Initiative Exploring the Genetic Landscape of Wilms Tumor. Nat Genet 2017, 49 (10), 1487–1494. 10.1038/ng.3940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (575).Wan L; Chong S; Xuan F; Liang A; Cui X; Gates L; Carroll TS; Li Y; Feng L; Chen G; Wang S-P; Ortiz MV; Daley SK; Wang X; Xuan H; Kentsis A; Muir TW; Roeder RG; Li H; Li W; Tjian R; Wen H; Allis CD Impaired Cell Fate through Gain-of-Function Mutations in a Chromatin Reader. Nature 2020, 577 (7788), 121–126. 10.1038/s41586-019-1842-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (576).Hnisz D; Shrinivas K; Young RA; Chakraborty AK; Sharp PA A Phase Separation Model for Transcriptional Control. Cell 2017, 169 (1), 13–23. 10.1016/j.cell.2017.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (577).Liu Y; Li Q; Song L; Gong C; Tang S; Budinich KA; Vanderbeck A; Mathias KM; Wertheim GB; Nguyen SC; Outen R; Joyce EF; Maillard I; Wan L Condensate-Promoting ENL Mutation Drives Tumorigenesis In Vivo Through Dynamic Regulation of Histone Modifications and Gene Expression. Cancer Discov 2024, 14 (8), 1522–1546. 10.1158/2159-8290.CD-23-0876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (578).Guo C; Che Z; Yue J; Xie P; Hao S; Xie W; Luo Z; Lin C ENL Initiates Multivalent Phase Separation of the Super Elongation Complex (SEC) in Controlling Rapid Transcriptional Activation. Sci Adv 2020, 6 (14), eaay4858. 10.1126/sciadv.aay4858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (579).Xue Z; Qin L; Xuan H; Luo K; Huang M; Xie L; Su Y; Xu L; Harsh J; Dale B; Shi X; Chen X; Kaniskan HÜ; Jin J; Wen H A Potent and Selective ENL Degrader Suppresses Oncogenic Gene Expression and Leukemia Progression. Sci Adv 2024, 10 (35), eado1432. 10.1126/sciadv.ado1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (580).Li X; Yao Y; Wu F; Song Y A Proteolysis-Targeting Chimera Molecule Selectively Degrades ENL and Inhibits Malignant Gene Expression and Tumor Growth. J Hematol Oncol 2022, 15 (1), 41. 10.1186/s13045-022-01258-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (581).Carter TR; Milosevich N; Dada L; Shaum JB; Barry Sharpless K; Kitamura S; Erb MA SuFEx-Based Chemical Diversification for the Systematic Discovery of CRBN Molecular Glues. Bioorg Med Chem 2024, 104, 117699. 10.1016/j.bmc.2024.117699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (582).Kitamura S; Zheng Q; Woehl JL; Solania A; Chen E; Dillon N; Hull MV; Kotaniguchi M; Cappiello JR; Kitamura S; Nizet V; Sharpless KB; Wolan DW Sulfur(VI) Fluoride Exchange (SuFEx)-Enabled High-Throughput Medicinal Chemistry. J Am Chem Soc 2020, 142 (25), 10899–10904. 10.1021/jacs.9b13652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (583).Liu F; Wang H; Li S; Bare GAL; Chen X; Wang C; Moses JE; Wu P; Sharpless KB Biocompatible SuFEx Click Chemistry: Thionyl Tetrafluoride (SOF4 )-Derived Connective Hubs for Bioconjugation to DNA and Proteins. Angew Chem Int Ed Engl 2019, 58 (24), 8029–8033. 10.1002/anie.201902489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (584).Fischer U; Meltzer P; Meese E Twelve Amplified and Expressed Genes Localized in a Single Domain in Glioma. Hum Genet 1996, 98 (5), 625–628. 10.1007/s004390050271. [DOI] [PubMed] [Google Scholar]
- (585).Hsu C-C; Zhao D; Shi J; Peng D; Guan H; Li Y; Huang Y; Wen H; Li W; Li H; Shi X Gas41 Links Histone Acetylation to H2A.Z Deposition and Maintenance of Embryonic Stem Cell Identity. Cell Discov 2018, 4, 28. 10.1038/s41421-018-0027-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (586).Pikor LA; Lockwood WW; Thu KL; Vucic EA; Chari R; Gazdar AF; Lam S; Lam WL YEATS4 Is a Novel Oncogene Amplified in Non-Small Cell Lung Cancer That Regulates the P53 Pathway. Cancer Res 2013, 73 (24), 7301–7312. 10.1158/0008-5472.CAN-13-1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (587).Xian Q; Song Y; Gui C; Zhou Y Mechanistic Insights into Genomic Structure and Functions of a Novel Oncogene YEATS4. Front Cell Dev Biol 2023, 11, 1192139. 10.3389/fcell.2023.1192139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (588).Ji K; Li L; Liu H; Shen Y; Jiang J; Zhang M; Teng H; Yan X; Zhang Y; Cai Y; Zhou H Unveiling the Role of GAS41 in Cancer Progression. Cancer Cell Int 2023, 23 (1), 245. 10.1186/s12935-023-03098-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (589).Kiuchi J; Komatsu S; Imamura T; Nishibeppu K; Shoda K; Arita T; Kosuga T; Konishi H; Shiozaki A; Kubota T; Okamoto K; Fujiwara H; Ichikawa D; Tsuda H; Otsuji E Overexpression of YEATS4 Contributes to Malignant Outcomes in Gastric Carcinoma. Am J Cancer Res 2018, 8 (12), 2436–2452. [PMC free article] [PubMed] [Google Scholar]
- (590).Kim YR; Park MS; Eum KH; Kim J; Lee JW; Bae T; Lee DH; Choi JW Transcriptome Analysis Indicates TFEB1 and YEATS4 as Regulatory Transcription Factors for Drug Resistance of Ovarian Cancer. Oncotarget 2015, 6 (31), 31030–31038. 10.18632/oncotarget.5208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (591).Li Y; Li L; Wu J; Qin J; Dai X; Jin T; Xu J YEATS4 Is Associated with Poor Prognosis and Promotes Epithelial-to-Mesenchymal Transition and Metastasis by Regulating ZEB1 Expression in Breast Cancer. Am J Cancer Res 2021, 11 (2), 416–440. [PMC free article] [PubMed] [Google Scholar]
- (592).Zimmermann K; Ahrens K; Matthes S; Buerstedde J-M; Strätling WH; Phi-van L Targeted Disruption of the GAS41 Gene Encoding a Putative Transcription Factor Indicates That GAS41 Is Essential for Cell Viability. J Biol Chem 2002, 277 (21), 18626–18631. 10.1074/jbc.M200572200. [DOI] [PubMed] [Google Scholar]
- (593).Listunov D; Linhares BM; Kim E; Winkler A; Simes ML; Weaver S; Cho HJ; Rizo A; Zolov S; Keshamouni VG; Grembecka J; Cierpicki T Development of Potent Dimeric Inhibitors of GAS41 YEATS Domain. Cell Chem Biol 2021, 28 (12), 1716–1727.e6. 10.1016/j.chembiol.2021.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (594).Cho HJ; Li H; Linhares BM; Kim E; Ndoj J; Miao H; Grembecka J; Cierpicki T GAS41 Recognizes Diacetylated Histone H3 through a Bivalent Binding Mode. ACS Chem Biol 2018, 13 (9), 2739–2746. 10.1021/acschembio.8b00674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (595).Londregan AT; Aitmakhanova K; Bennett J; Byrnes LJ; Canterbury DP; Cheng X; Christott T; Clemens J; Coffey SB; Dias JM; Dowling MS; Farnie G; Fedorov O; Fennell KF; Gamble V; Gileadi C; Giroud C; Harris MR; Hollingshead BD; Huber K; Korczynska M; Lapham K; Loria PM; Narayanan A; Owen DR; Raux B; Sahasrabudhe PV; Ruggeri RB; Sáez LD; Stock IA; Thuma BA; Tsai A; Varghese AE Discovery of High-Affinity Small-Molecule Binders of the Epigenetic Reader YEATS4. J Med Chem 2023, 66 (1), 460–472. 10.1021/acs.jmedchem.2c01421. [DOI] [PubMed] [Google Scholar]
- (596).Gregory GD; Vakoc CR; Rozovskaia T; Zheng X; Patel S; Nakamura T; Canaani E; Blobel GA Mammalian ASH1L Is a Histone Methyltransferase That Occupies the Transcribed Region of Active Genes. Mol Cell Biol 2007, 27 (24), 8466–8479. 10.1128/MCB.00993-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (597).Jones M; Chase J; Brinkmeier M; Xu J; Weinberg DN; Schira J; Friedman A; Malek S; Grembecka J; Cierpicki T; Dou Y; Camper SA; Maillard I Ash1l Controls Quiescence and Self-Renewal Potential in Hematopoietic Stem Cells. J Clin Invest 2015, 125 (5), 2007–2020. 10.1172/JCI78124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (598).Rogawski DS; Grembecka J; Cierpicki T H3K36 Methyltransferases as Cancer Drug Targets: Rationale and Perspectives for Inhibitor Development. Future Med Chem 2016, 8 (13), 1589–1607. 10.4155/fmc-2016-0071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (599).Wagner EJ; Carpenter PB Understanding the Language of Lys36 Methylation at Histone H3. Nat Rev Mol Cell Biol 2012, 13 (2), 115–126. 10.1038/nrm3274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (600).Schuettengruber B; Chourrout D; Vervoort M; Leblanc B; Cavalli G Genome Regulation by Polycomb and Trithorax Proteins. Cell 2007, 128 (4), 735–745. 10.1016/j.cell.2007.02.009. [DOI] [PubMed] [Google Scholar]
- (601).Zhu L; Li Q; Wong SHK; Huang M; Klein BJ; Shen J; Ikenouye L; Onishi M; Schneidawind D; Buechele C; Hansen L; Duque-Afonso J; Zhu F; Martin GM; Gozani O; Majeti R; Kutateladze TG; Cleary ML ASH1L Links Histone H3 Lysine 36 Dimethylation to MLL Leukemia. Cancer Discov 2016, 6 (7), 770–783. 10.1158/2159-8290.CD-16-0058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (602).Rogawski DS; Deng J; Li H; Miao H; Borkin D; Purohit T; Song J; Chase J; Li S; Ndoj J; Klossowski S; Kim E; Mao F; Zhou B; Ropa J; Krotoska MZ; Jin Z; Ernst P; Feng X; Huang G; Nishioka K; Kelly S; He M; Wen B; Sun D; Muntean A; Dou Y; Maillard I; Cierpicki T; Grembecka J Discovery of First-in-Class Inhibitors of ASH1L Histone Methyltransferase with Anti-Leukemic Activity. Nat Commun 2021, 12 (1), 2792. 10.1038/s41467-021-23152-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (603).Montgomery ND; Yee D; Chen A; Kalantry S; Chamberlain SJ; Otte AP; Magnuson T The Murine Polycomb Group Protein Eed Is Required for Global Histone H3 Lysine-27 Methylation. Curr Biol 2005, 15 (10), 942–947. 10.1016/j.cub.2005.04.051. [DOI] [PubMed] [Google Scholar]
- (604).Margueron R; Justin N; Ohno K; Sharpe ML; Son J; Drury WJ; Voigt P; Martin SR; Taylor WR; De Marco V; Pirrotta V; Reinberg D; Gamblin SJ Role of the Polycomb Protein EED in the Propagation of Repressive Histone Marks. Nature 2009, 461 (7265), 762–767. 10.1038/nature08398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (605).Kennison JA; Tamkun JW Dosage-Dependent Modifiers of Polycomb and Antennapedia Mutations in Drosophila. Proc Natl Acad Sci U S A 1988, 85 (21), 8136–8140. 10.1073/pnas.85.21.8136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (606).Wilson BG; Wang X; Shen X; McKenna ES; Lemieux ME; Cho Y-J; Koellhoffer EC; Pomeroy SL; Orkin SH; Roberts CWM Epigenetic Antagonism between Polycomb and SWI/SNF Complexes during Oncogenic Transformation. Cancer Cell 2010, 18 (4), 316–328. 10.1016/j.ccr.2010.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (607).Morin RD; Johnson NA; Severson TM; Mungall AJ; An J; Goya R; Paul JE; Boyle M; Woolcock BW; Kuchenbauer F; Yap D; Humphries RK; Griffith OL; Shah S; Zhu H; Kimbara M; Shashkin P; Charlot JF; Tcherpakov M; Corbett R; Tam A; Varhol R; Smailus D; Moksa M; Zhao Y; Delaney A; Qian H; Birol I; Schein J; Moore R; Holt R; Horsman DE; Connors JM; Jones S; Aparicio S; Hirst M; Gascoyne RD; Marra MA Somatic Mutations Altering EZH2 (Tyr641) in Follicular and Diffuse Large B-Cell Lymphomas of Germinal-Center Origin. Nat Genet 2010, 42 (2), 181–185. 10.1038/ng.518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (608).Lohr JG; Stojanov P; Lawrence MS; Auclair D; Chapuy B; Sougnez C; Cruz-Gordillo P; Knoechel B; Asmann YW; Slager SL; Novak AJ; Dogan A; Ansell SM; Link BK; Zou L; Gould J; Saksena G; Stransky N; Rangel-Escareño C; Fernandez-Lopez JC; Hidalgo-Miranda A; Melendez-Zajgla J; Hernández-Lemus E; Schwarz-Cruz y Celis A; Imaz-Rosshandler I; Ojesina AI; Jung J; Pedamallu CS; Lander ES; Habermann TM; Cerhan JR; Shipp MA; Getz G; Golub TR Discovery and Prioritization of Somatic Mutations in Diffuse Large B-Cell Lymphoma (DLBCL) by Whole-Exome Sequencing. Proc Natl Acad Sci U S A 2012, 109 (10), 3879–3884. 10.1073/pnas.1121343109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (609).Morin RD; Mendez-Lago M; Mungall AJ; Goya R; Mungall KL; Corbett RD; Johnson NA; Severson TM; Chiu R; Field M; Jackman S; Krzywinski M; Scott DW; Trinh DL; Tamura-Wells J; Li S; Firme MR; Rogic S; Griffith M; Chan S; Yakovenko O; Meyer IM; Zhao EY; Smailus D; Moksa M; Chittaranjan S; Rimsza L; Brooks-Wilson A; Spinelli JJ; Ben-Neriah S; Meissner B; Woolcock B; Boyle M; McDonald H; Tam A; Zhao Y; Delaney A; Zeng T; Tse K; Butterfield Y; Birol I; Holt R; Schein J; Horsman DE; Moore R; Jones SJM; Connors JM; Hirst M; Gascoyne RD; Marra MA Frequent Mutation of Histone-Modifying Genes in Non-Hodgkin Lymphoma. Nature 2011, 476 (7360), 298–303. 10.1038/nature10351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (610).Kleer CG; Cao Q; Varambally S; Shen R; Ota I; Tomlins SA; Ghosh D; Sewalt RGAB; Otte AP; Hayes DF; Sabel MS; Livant D; Weiss SJ; Rubin MA; Chinnaiyan AM EZH2 Is a Marker of Aggressive Breast Cancer and Promotes Neoplastic Transformation of Breast Epithelial Cells. Proc Natl Acad Sci U S A 2003, 100 (20), 11606–11611. 10.1073/pnas.1933744100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (611).Varambally S; Dhanasekaran SM; Zhou M; Barrette TR; Kumar-Sinha C; Sanda MG; Ghosh D; Pienta KJ; Sewalt RGAB; Otte AP; Rubin MA; Chinnaiyan AM The Polycomb Group Protein EZH2 Is Involved in Progression of Prostate Cancer. Nature 2002, 419 (6907), 624–629. 10.1038/nature01075. [DOI] [PubMed] [Google Scholar]
- (612).Kirmizis A; Bartley SM; Kuzmichev A; Margueron R; Reinberg D; Green R; Farnham PJ Silencing of Human Polycomb Target Genes Is Associated with Methylation of Histone H3 Lys 27. Genes Dev 2004, 18 (13), 1592–1605. 10.1101/gad.1200204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (613).Bracken AP; Pasini D; Capra M; Prosperini E; Colli E; Helin K EZH2 Is Downstream of the PRB-E2F Pathway, Essential for Proliferation and Amplified in Cancer. EMBO J 2003, 22 (20), 5323–5335. 10.1093/emboj/cdg542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (614).Simon JA; Lange CA Roles of the EZH2 Histone Methyltransferase in Cancer Epigenetics. Mutat Res 2008, 647 (1–2), 21–29. 10.1016/j.mrfmmm.2008.07.010. [DOI] [PubMed] [Google Scholar]
- (615).Velichutina I; Shaknovich R; Geng H; Johnson NA; Gascoyne RD; Melnick AM; Elemento O EZH2-Mediated Epigenetic Silencing in Germinal Center B Cells Contributes to Proliferation and Lymphomagenesis. Blood 2010, 116 (24), 5247–5255. 10.1182/blood-2010-04-280149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (616).Vangamudi B; Paul TA; Shah PK; Kost-Alimova M; Nottebaum L; Shi X; Zhan Y; Leo E; Mahadeshwar HS; Protopopov A; Futreal A; Tieu TN; Peoples M; Heffernan TP; Marszalek JR; Toniatti C; Petrocchi A; Verhelle D; Owen DR; Draetta G; Jones P; Palmer WS; Sharma S; Andersen JN 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 Res 2015, 75 (18), 3865–3878. 10.1158/0008-5472.CAN-14-3798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (617).Deng Q; Lakra P; Gou P; Yang H; Meydan C; Teater M; Chin C; Zhang W; Dinh T; Hussein U; Li X; Rojas E; Liu W; Reville PK; Kizhakeyil A; Barisic D; Parsons S; Wilson A; Henderson J; Scull B; Gurumurthy C; Vega F; Chadburn A; Cuglievan B; El-Mallawany NK; Allen C; Mason C; Melnick A; Green MR SMARCA4 Is a Haploinsufficient B Cell Lymphoma Tumor Suppressor That Fine-Tunes Centrocyte Cell Fate Decisions. Cancer Cell 2024, 42 (4), 605–622.e11. 10.1016/j.ccell.2024.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (618).Kelso TWR; Porter DK; Amaral ML; Shokhirev MN; Benner C; Hargreaves DC Chromatin Accessibility Underlies Synthetic Lethality of SWI/SNF Subunits in ARID1A-Mutant Cancers. Elife 2017, 6, e30506. 10.7554/eLife.30506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (619).Medina PP; Romero OA; Kohno T; Montuenga LM; Pio R; Yokota J; Sanchez-Cespedes M Frequent BRG1/SMARCA4-Inactivating Mutations in Human Lung Cancer Cell Lines. Hum Mutat 2008, 29 (5), 617–622. 10.1002/humu.20730. [DOI] [PubMed] [Google Scholar]
- (620).Wong AK; Shanahan F; Chen Y; Lian L; Ha P; Hendricks K; Ghaffari S; Iliev D; Penn B; Woodland AM; Smith R; Salada G; Carillo A; Laity K; Gupte J; Swedlund B; Tavtigian SV; Teng DH; Lees E BRG1, a Component of the SWI-SNF Complex, Is Mutated in Multiple Human Tumor Cell Lines. Cancer Res 2000, 60 (21), 6171–6177. [PubMed] [Google Scholar]
- (621).Hoffman GR; Rahal R; Buxton F; Xiang K; McAllister G; Frias E; Bagdasarian L; Huber J; Lindeman A; Chen D; Romero R; Ramadan N; Phadke T; Haas K; Jaskelioff M; Wilson BG; Meyer MJ; Saenz-Vash V; Zhai H; Myer VE; Porter JA; Keen N; McLaughlin ME; Mickanin C; Roberts CWM; Stegmeier F; Jagani Z Functional Epigenetics Approach Identifies BRM/SMARCA2 as a Critical Synthetic Lethal Target in BRG1-Deficient Cancers. Proc Natl Acad Sci U S A 2014, 111 (8), 3128–3133. 10.1073/pnas.1316793111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (622).Yap DB; Chu J; Berg T; Schapira M; Cheng S-WG; Moradian A; Morin RD; Mungall AJ; Meissner B; Boyle M; Marquez VE; Marra MA; Gascoyne RD; Humphries RK; Arrowsmith CH; Morin GB; Aparicio SAJR Somatic Mutations at EZH2 Y641 Act Dominantly through a Mechanism of Selectively Altered PRC2 Catalytic Activity, to Increase H3K27 Trimethylation. Blood 2011, 117 (8), 2451–2459. 10.1182/blood-2010-11-321208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (623).Tang M; Verhaak RG A Molecular Take on Malignant Rhabdoid Tumors. Trends Cancer 2016, 2 (5), 217–218. 10.1016/j.trecan.2016.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (624).Knutson SK; Warholic NM; Wigle TJ; Klaus CR; Allain CJ; Raimondi A; Porter Scott M; Chesworth R; Moyer MP; Copeland RA; Richon VM; Pollock RM; Kuntz KW; Keilhack H Durable Tumor Regression in Genetically Altered Malignant Rhabdoid Tumors by Inhibition of Methyltransferase EZH2. Proc Natl Acad Sci U S A 2013, 110 (19), 7922–7927. 10.1073/pnas.1303800110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (625).Glazer RI; Hartman KD; Knode MC; Richard MM; Chiang PK; Tseng CK; Marquez VE 3-Deazaneplanocin: A New and Potent Inhibitor of S-Adenosylhomocysteine Hydrolase and Its Effects on Human Promyelocytic Leukemia Cell Line HL-60. Biochem Biophys Res Commun 1986, 135 (2), 688–694. 10.1016/0006-291x(86)90048-3. [DOI] [PubMed] [Google Scholar]
- (626).Tan J; Yang X; Zhuang L; Jiang X; Chen W; Lee PL; Karuturi RKM; Tan PBO; Liu ET; Yu Q Pharmacologic Disruption of Polycomb-Repressive Complex 2-Mediated Gene Repression Selectively Induces Apoptosis in Cancer Cells. Genes Dev 2007, 21 (9), 1050–1063. 10.1101/gad.1524107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (627).Miranda TB; Cortez CC; Yoo CB; Liang G; Abe M; Kelly TK; Marquez VE; Jones PA DZNep Is a Global Histone Methylation Inhibitor That Reactivates Developmental Genes Not Silenced by DNA Methylation. Mol Cancer Ther 2009, 8 (6), 1579–1588. 10.1158/1535-7163.MCT-09-0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (628).Knutson SK; Wigle TJ; Warholic NM; Sneeringer CJ; Allain CJ; Klaus CR; Sacks JD; Raimondi A; Majer CR; Song J; Scott MP; Jin L; Smith JJ; Olhava EJ; Chesworth R; Moyer MP; Richon VM; Copeland RA; Keilhack H; Pollock RM; Kuntz KW A Selective Inhibitor of EZH2 Blocks H3K27 Methylation and Kills Mutant Lymphoma Cells. Nat Chem Biol 2012, 8 (11), 890–896. 10.1038/nchembio.1084. [DOI] [PubMed] [Google Scholar]
- (629).McCabe MT; Ott HM; Ganji G; Korenchuk S; Thompson C; Van Aller GS; Liu Y; Graves AP; Della Pietra A; Diaz E; LaFrance LV; Mellinger M; Duquenne C; Tian X; Kruger RG; McHugh CF; Brandt M; Miller WH; Dhanak D; Verma SK; Tummino PJ; Creasy CL EZH2 Inhibition as a Therapeutic Strategy for Lymphoma with EZH2-Activating Mutations. Nature 2012, 492 (7427), 108–112. 10.1038/nature11606. [DOI] [PubMed] [Google Scholar]
- (630).Qi W; Chan H; Teng L; Li L; Chuai S; Zhang R; Zeng J; Li M; Fan H; Lin Y; Gu J; Ardayfio O; Zhang J-H; Yan X; Fang J; Mi Y; Zhang M; Zhou T; Feng G; Chen Z; Li G; Yang T; Zhao K; Liu X; Yu Z; Lu CX; Atadja P; Li E Selective Inhibition of Ezh2 by a Small Molecule Inhibitor Blocks Tumor Cells Proliferation. Proc Natl Acad Sci U S A 2012, 109 (52), 21360–21365. 10.1073/pnas.1210371110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (631).Finogenova K; Bonnet J; Poepsel S; Schäfer IB; Finkl K; Schmid K; Litz C; Strauss M; Benda C; Müller J Structural Basis for PRC2 Decoding of Active Histone Methylation Marks H3K36me2/3. Elife 2020, 9, e61964. 10.7554/eLife.61964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (632).Glancy E; Ciferri C; Bracken AP Structural Basis for PRC2 Engagement with Chromatin. Curr Opin Struct Biol 2021, 67, 135–144. 10.1016/j.sbi.2020.10.017. [DOI] [PubMed] [Google Scholar]
- (633).Song J; Gooding AR; Hemphill WO; Love BD; Robertson A; Yao L; Zon LI; North TE; Kasinath V; Cech TR Structural Basis for Inactivation of PRC2 by G-Quadruplex RNA. Science (1979) 2023, 381 (6664), 1331–1337. 10.1126/science.adh0059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (634).Cookis T; Lydecker A; Sauer P; Kasinath V; Nogales E Structural Basis for the Inhibition of PRC2 by Active Transcription Histone Posttranslational Modifications. 32 2025, No. 2, 393–404. 10.1038/s41594-024-01452-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (635).Grau D; Zhang Y; Lee C-H; Valencia-Sánchez M; Zhang J; Wang M; Holder M; Svetlov V; Tan D; Nudler E; Reinberg D; Walz T; Armache K-J Structures of Monomeric and Dimeric PRC2:EZH1 Reveal Flexible Modules Involved in Chromatin Compaction. Nat Commun 2021, 12 (1), 714. 10.1038/s41467-020-20775-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (636).Verma SK; Tian X; LaFrance LV; Duquenne C; Suarez DP; Newlander KA; Romeril SP; Burgess JL; Grant SW; Brackley JA; Graves AP; Scherzer DA; Shu A; Thompson C; Ott HM; Van Aller GS; Machutta CA; Diaz E; Jiang Y; Johnson NW; Knight SD; Kruger RG; McCabe MT; Dhanak D; Tummino PJ; Creasy CL; Miller WH Identification of Potent, Selective, Cell-Active Inhibitors of the Histone Lysine Methyltransferase EZH2. ACS Med Chem Lett 2012, 3 (12), 1091–1096. 10.1021/ml3003346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (637).Konze KD; Ma A; Li F; Barsyte-Lovejoy D; Parton T; Macnevin CJ; Liu F; Gao C; Huang X-P; Kuznetsova E; Rougie M; Jiang A; Pattenden SG; Norris JL; James LI; Roth BL; Brown PJ; Frye SV; Arrowsmith CH; Hahn KM; Wang GG; Vedadi M; Jin J An Orally Bioavailable Chemical Probe of the Lysine Methyltransferases EZH2 and EZH1. ACS Chem Biol 2013, 8 (6), 1324–1334. 10.1021/cb400133j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (638).Yang X; Li F; Konze KD; Meslamani J; Ma A; Brown PJ; Zhou M-M; Arrowsmith CH; Kaniskan HÜ; Vedadi M; Jin J Structure-Activity Relationship Studies for Enhancer of Zeste Homologue 2 (EZH2) and Enhancer of Zeste Homologue 1 (EZH1) Inhibitors. J Med Chem 2016, 59 (16), 7617–7633. 10.1021/acs.jmedchem.6b00855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (639).Knutson SK; Kawano S; Minoshima Y; Warholic NM; Huang K-C; Xiao Y; Kadowaki T; Uesugi M; Kuznetsov G; Kumar N; Wigle TJ; Klaus CR; Allain CJ; Raimondi A; Waters NJ; Smith JJ; Porter-Scott M; Chesworth R; Moyer MP; Copeland RA; Richon VM; Uenaka T; Pollock RM; Kuntz KW; Yokoi A; Keilhack H Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma. Mol Cancer Ther 2014, 13 (4), 842–854. 10.1158/1535-7163.MCT-13-0773. [DOI] [PubMed] [Google Scholar]
- (640).Gounder M; Schöffski P; Jones RL; Agulnik M; Cote GM; Villalobos VM; Attia S; Chugh R; Chen TW-W; Jahan T; Loggers ET; Gupta A; Italiano A; Demetri GD; Ratan R; Davis LE; Mir O; Dileo P; Van Tine BA; Pressey JG; Lingaraj T; Rajarethinam A; Sierra L; Agarwal S; Stacchiotti S Tazemetostat in Advanced Epithelioid Sarcoma with Loss of INI1/SMARCB1: An International, Open-Label, Phase 2 Basket Study. Lancet Oncol 2020, 21 (11), 1423–1432. 10.1016/S1470-2045(20)30451-4. [DOI] [PubMed] [Google Scholar]
- (641).Italiano A; Soria J-C; Toulmonde M; Michot J-M; Lucchesi C; Varga A; Coindre J-M; Blakemore SJ; Clawson A; Suttle B; McDonald AA; Woodruff M; Ribich S; Hedrick E; Keilhack H; Thomson B; Owa T; Copeland RA; Ho PTC; Ribrag V 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 2018, 19 (5), 649–659. 10.1016/S1470-2045(18)30145-1. [DOI] [PubMed] [Google Scholar]
- (642).Morschhauser F; Tilly H; Chaidos A; McKay P; Phillips T; Assouline S; Batlevi CL; Campbell P; Ribrag V; Damaj GL; Dickinson M; Jurczak W; Kazmierczak M; Opat S; Radford J; Schmitt A; Yang J; Whalen J; Agarwal S; Adib D; Salles G Tazemetostat for Patients with Relapsed or Refractory Follicular Lymphoma: An Open-Label, Single-Arm, Multicentre, Phase 2 Trial. Lancet Oncol 2020, 21 (11), 1433–1442. 10.1016/S1470-2045(20)30441-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (643).Campbell JE; Kuntz KW; Knutson SK; Warholic NM; Keilhack H; Wigle TJ; Raimondi A; Klaus CR; Rioux N; Yokoi A; Kawano S; Minoshima Y; Choi H-W; Porter Scott M; Waters NJ; Smith JJ; Chesworth R; Moyer MP; Copeland RA EPZ011989, A Potent, Orally-Available EZH2 Inhibitor with Robust in Vivo Activity. ACS Med Chem Lett 2015, 6 (5), 491–495. 10.1021/acsmedchemlett.5b00037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (644).Yamagishi M; Hori M; Fujikawa D; Ohsugi T; Honma D; Adachi N; Katano H; Hishima T; Kobayashi S; Nakano K; Nakashima M; Iwanaga M; Utsunomiya A; Tanaka Y; Okada S; Tsukasaki K; Tobinai K; Araki K; Watanabe T; Uchimaru K Targeting Excessive EZH1 and EZH2 Activities for Abnormal Histone Methylation and Transcription Network in Malignant Lymphomas. Cell Rep 2019, 29 (8), 2321–2337.e7. 10.1016/j.celrep.2019.10.083. [DOI] [PubMed] [Google Scholar]
- (645).Izutsu K; Makita S; Nosaka K; Yoshimitsu M; Utsunomiya A; Kusumoto S; Morishima S; Tsukasaki K; Kawamata T; Ono T; Rai S; Katsuya H; Ishikawa J; Yamada H; Kato K; Tachibana M; Kakurai Y; Adachi N; Tobinai K; Yonekura K; Ishitsuka K An Open-Label, Single-Arm Phase 2 Trial of Valemetostat for Relapsed or Refractory Adult T-Cell Leukemia/Lymphoma. Blood 2023, 141 (10), 1159–1168. 10.1182/blood.2022016862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (646).Kim W; Bird GH; Neff T; Guo G; Kerenyi MA; Walensky LD; Orkin SH Targeted Disruption of the EZH2-EED Complex Inhibits EZH2-Dependent Cancer. Nat Chem Biol 2013, 9 (10), 643–650. 10.1038/nchembio.1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (647).Xu C; Bian C; Yang W; Galka M; Ouyang H; Chen C; Qiu W; Liu H; Jones AE; MacKenzie F; Pan P; Li SS-C; Wang H; Min J Binding of Different Histone Marks Differentially Regulates the Activity and Specificity of Polycomb Repressive Complex 2 (PRC2). Proc Natl Acad Sci U S A 2010, 107 (45), 19266–19271. 10.1073/pnas.1008937107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (648).Sanulli S; Justin N; Teissandier A; Ancelin K; Portoso M; Caron M; Michaud A; Lombard B; da Rocha ST; Offer J; Loew D; Servant N; Wassef M; Burlina F; Gamblin SJ; Heard E; Margueron R Jarid2 Methylation via the PRC2 Complex Regulates H3K27me3 Deposition during Cell Differentiation. Mol Cell 2015, 57 (5), 769–783. 10.1016/j.molcel.2014.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (649).Ge EJ; Jani KS; Diehl KL; Müller MM; Muir TW Nucleation and Propagation of Heterochromatin by the Histone Methyltransferase PRC2: Geometric Constraints and Impact of the Regulatory Subunit JARID2. J Am Chem Soc 2019, 141 (38), 15029–15039. 10.1021/jacs.9b02321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (650).Han Z; Xing X; Hu M; Zhang Y; Liu P; Chai J Structural Basis of EZH2 Recognition by EED. Structure 2007, 15 (10), 1306–1315. 10.1016/j.str.2007.08.007. [DOI] [PubMed] [Google Scholar]
- (651).Qi W; Zhao K; Gu J; Huang Y; Wang Y; Zhang H; Zhang M; Zhang J; Yu Z; Li L; Teng L; Chuai S; Zhang C; Zhao M; Chan H; Chen Z; Fang D; Fei Q; Feng L; Feng L; Gao Y; Ge H; Ge X; Li G; Lingel A; Lin Y; Liu Y; Luo F; Shi M; Wang L; Wang Z; Yu Y; Zeng J; Zeng C; Zhang L; Zhang Q; Zhou S; Oyang C; Atadja P; Li E An Allosteric PRC2 Inhibitor Targeting the H3K27me3 Binding Pocket of EED. Nat Chem Biol 2017, 13 (4), 381–388. 10.1038/nchembio.2304. [DOI] [PubMed] [Google Scholar]
- (652).He Y; Selvaraju S; Curtin ML; Jakob CG; Zhu H; Comess KM; Shaw B; The J; Lima-Fernandes E; Szewczyk MM; Cheng D; Klinge KL; Li H-Q; Pliushchev M; Algire MA; Maag D; Guo J; Dietrich J; Panchal SC; Petros AM; Sweis RF; Torrent M; Bigelow LJ; Senisterra G; Li F; Kennedy S; Wu Q; Osterling DJ; Lindley DJ; Gao W; Galasinski S; Barsyte-Lovejoy D; Vedadi M; Buchanan FG; Arrowsmith CH; Chiang GG; Sun C; Pappano WN The EED Protein-Protein Interaction Inhibitor A-395 Inactivates the PRC2 Complex. Nat Chem Biol 2017, 13 (4), 389–395. 10.1038/nchembio.2306. [DOI] [PubMed] [Google Scholar]
- (653).Li L; Zhang H; Zhang M; Zhao M; Feng L; Luo X; Gao Z; Huang Y; Ardayfio O; Zhang J-H; Lin Y; Fan H; Mi Y; Li G; Liu L; Feng L; Luo F; Teng L; Qi W; Ottl J; Lingel A; Bussiere DE; Yu Z; Atadja P; Lu C; Li E; Gu J; Zhao K Discovery and Molecular Basis of a Diverse Set of Polycomb Repressive Complex 2 Inhibitors Recognition by EED. PLoS One 2017, 12 (1), e0169855. 10.1371/journal.pone.0169855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (654).Huang Y; Zhang J; Yu Z; Zhang H; Wang Y; Lingel A; Qi W; Gu J; Zhao K; Shultz MD; Wang L; Fu X; Sun Y; Zhang Q; Jiang X; Zhang J; Zhang C; Li L; Zeng J; Feng L; Zhang C; Liu Y; Zhang M; Zhang L; Zhao M; Gao Z; Liu X; Fang D; Guo H; Mi Y; Gabriel T; Dillon MP; Atadja P; Oyang C Discovery of First-in-Class, Potent, and Orally Bioavailable Embryonic Ectoderm Development (EED) Inhibitor with Robust Anticancer Efficacy. J Med Chem 2017, 60 (6), 2215–2226. 10.1021/acs.jmedchem.6b01576. [DOI] [PubMed] [Google Scholar]
- (655).Gibaja V; Shen F; Harari J; Korn J; Ruddy D; Saenz-Vash V; Zhai H; Rejtar T; Paris CG; Yu Z; Lira M; King D; Qi W; Keen N; Hassan AQ; Chan HM Development of Secondary Mutations in Wild-Type and Mutant EZH2 Alleles Cooperates to Confer Resistance to EZH2 Inhibitors. Oncogene 2016, 35 (5), 558–566. 10.1038/onc.2015.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (656).Baker T; Nerle S; Pritchard J; Zhao B; Rivera VM; Garner A; Gonzalvez F Acquisition of a Single EZH2 D1 Domain Mutation Confers Acquired Resistance to EZH2-Targeted Inhibitors. Oncotarget 2015, 6 (32), 32646–32655. 10.18632/oncotarget.5066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (657).Huang Y; Sendzik M; Zhang J; Gao Z; Sun Y; Wang L; Gu J; Zhao K; Yu Z; Zhang L; Zhang Q; Blanz J; Chen Z; Dubost V; Fang D; Feng L; Fu X; Kiffe M; Li L; Luo F; Luo X; Mi Y; Mistry P; Pearson D; Piaia A; Scheufler C; Terranova R; Weiss A; Zeng J; Zhang H; Zhang J; Zhao M; Dillon MP; Jeay S; Qi W; Moggs J; Pissot-Soldermann C; Li E; Atadja P; Lingel A; Oyang C Discovery of the Clinical Candidate MAK683: An EED-Directed, Allosteric, and Selective PRC2 Inhibitor for the Treatment of Advanced Malignancies. J Med Chem 2022, 65 (7), 5317–5333. 10.1021/acs.jmedchem.1c02148. [DOI] [PubMed] [Google Scholar]
- (658).Peterson LA Reactive Metabolites in the Biotransformation of Molecules Containing a Furan Ring. Chem Res Toxicol 2013, 26 (1), 6–25. 10.1021/tx3003824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (659).Ribrag V; Michot J-M; Igleias L; Tan D; Ma B; Duca M; Wainberg ZA; Fan YY; Suenaga N; Cheng Y; Lai C; Yokota T Phase I/II Study of MAK683 in Patients with Advanced Malignancies, Including Diffuse Large B-Cell Lymphoma. Blood 2021, 138 (Supplement 1), 1422–1422. 10.1182/blood-2021-147904. [DOI] [Google Scholar]
- (660).Shi B; Liang J; Yang X; Wang Y; Zhao Y; Wu H; Sun L; Zhang Y; Chen Y; Li R; Zhang Y; Hong M; Shang Y Integration of Estrogen and Wnt Signaling Circuits by the Polycomb Group Protein EZH2 in Breast Cancer Cells. Mol Cell Biol 2007, 27 (14), 5105–5119. 10.1128/MCB.00162-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (661).Gonzalez ME; Moore HM; Li X; Toy KA; Huang W; Sabel MS; Kidwell KM; Kleer CG EZH2 Expands Breast Stem Cells through Activation of NOTCH1 Signaling. Proc Natl Acad Sci U S A 2014, 111 (8), 3098–3103. 10.1073/pnas.1308953111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (662).Ma A; Stratikopoulos E; Park K-S; Wei J; Martin TC; Yang X; Schwarz M; Leshchenko V; Rialdi A; Dale B; Lagana A; Guccione E; Parekh S; Parsons R; Jin J Discovery of a First-in-Class EZH2 Selective Degrader. Nat Chem Biol 2020, 16 (2), 214–222. 10.1038/s41589-019-0421-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (663).Potjewyd F; Turner A-MW; Beri J; Rectenwald JM; Norris-Drouin JL; Cholensky SH; Margolis DM; Pearce KH; Herring LE; James LI Degradation of Polycomb Repressive Complex 2 with an EED-Targeted Bivalent Chemical Degrader. Cell Chem Biol 2020, 27 (1), 47–56.e15. 10.1016/j.chembiol.2019.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (664).Hsu JH-R; Rasmusson T; Robinson J; Pachl F; Read J; Kawatkar S; O’ Donovan DH; Bagal S; Code E; Rawlins P; Argyrou A; Tomlinson R; Gao N; Zhu X; Chiarparin E; Jacques K; Shen M; Woods H; Bednarski E; Wilson DM; Drew L; Castaldi MP; Fawell S; Bloecher A EED-Targeted PROTACs Degrade EED, EZH2, and SUZ12 in the PRC2 Complex. Cell Chem Biol 2020, 27 (1), 41–46.e17. 10.1016/j.chembiol.2019.11.004. [DOI] [PubMed] [Google Scholar]
- (665).Liu Z; Hu X; Wang Q; Wu X; Zhang Q; Wei W; Su X; He H; Zhou S; Hu R; Ye T; Zhu Y; Wang N; Yu L Design and Synthesis of EZH2-Based PROTACs to Degrade the PRC2 Complex for Targeting the Noncatalytic Activity of EZH2. J Med Chem 2021, 64 (5), 2829–2848. 10.1021/acs.jmedchem.0c02234. [DOI] [PubMed] [Google Scholar]
- (666).Bashore FM; Foley CA; Ong HW; Rectenwald JM; Hanley RP; Norris-Drouin JL; Cholensky SH; Mills CA; Pearce KH; Herring LE; Kireev D; Frye SV; James LI PROTAC Linkerology Leads to an Optimized Bivalent Chemical Degrader of Polycomb Repressive Complex 2 (PRC2) Components. ACS Chem Biol 2023, 18 (3), 494–507. 10.1021/acschembio.2c00804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (667).Mei H; Wu H; Yang J; Zhou B; Wang A; Hu C; Qi S; Jiang Z; Zou F; Wang B; Liu F; Chen Y; Wang W; Liu J; Liu Q Discovery of IHMT-337 as a Potent Irreversible EZH2 Inhibitor Targeting CDK4 Transcription for Malignancies. Signal Transduct Target Ther 2023, 8 (1), 18. 10.1038/s41392-022-01240-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (668).Xie H; Xu W; Liang J; Liu Y; Zhuo C; Zou X; Luo W; Xiao J; Lin Y; Chen L; Li H Design, Synthesis and Evaluation of EZH2-Based PROTACs Targeting PRC2 Complex in Lymphoma. Bioorg Chem 2023, 140, 106762. 10.1016/j.bioorg.2023.106762. [DOI] [PubMed] [Google Scholar]
- (669).Velez J; Dale B; Park K-S; Kaniskan HÜ; Yu X; Jin J Discovery of a Novel, Highly Potent EZH2 PROTAC Degrader for Targeting Non-Canonical Oncogenic Functions of EZH2. Eur J Med Chem 2024, 267, 116154. 10.1016/j.ejmech.2024.116154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (670).Kazansky Y; Cameron D; Mueller HS; Demarest P; Zaffaroni N; Arrighetti N; Zuco V; Kuwahara Y; Somwar R; Ladanyi M; Qu R; de Stanchina E; Dela Cruz FS; Kung AL; Gounder MM; Kentsis A Overcoming Clinical Resistance to EZH2 Inhibition Using Rational Epigenetic Combination Therapy. Cancer Discov 2024, 14 (6), 965–981. 10.1158/2159-8290.CD-23-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (671).Daemen A; Yuen N; Wang A; Pankov A; Ulicna L; Chen C; Duong FL; Long J; Marx MA; Christensen JG; Friedman LS; Junttila MR Abstract 6586: ORIC-944, a Potent and Selective Allosteric PRC2 Inhibitor with Best-in-Class Properties, Demonstrates Combination Synergy with AR Pathway Inhibitors in Prostate Cancer Models. Cancer Res 2024, 84 (6_Supplement), 6586–6586. 10.1158/1538-7445.AM2024-6586. [DOI] [Google Scholar]
- (672).Gao Z; Zhang J; Bonasio R; Strino F; Sawai A; Parisi F; Kluger Y; Reinberg D PCGF Homologs, CBX Proteins, and RYBP Define Functionally Distinct PRC1 Family Complexes. Mol Cell 2012, 45 (3), 344–356. 10.1016/j.molcel.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (673).Wang H; Wang L; Erdjument-Bromage H; Vidal M; Tempst P; Jones RS; Zhang Y Role of Histone H2A Ubiquitination in Polycomb Silencing. Nature 2004, 431 (7010), 873–878. 10.1038/nature02985. [DOI] [PubMed] [Google Scholar]
- (674).Schuettengruber B; Bourbon H-M; Di Croce L; Cavalli G Genome Regulation by Polycomb and Trithorax: 70 Years and Counting. Cell 2017, 171 (1), 34–57. 10.1016/j.cell.2017.08.002. [DOI] [PubMed] [Google Scholar]
- (675).Lessard J; Sauvageau G Bmi-1 Determines the Proliferative Capacity of Normal and Leukaemic Stem Cells. Nature 2003, 423 (6937), 255–260. 10.1038/nature01572. [DOI] [PubMed] [Google Scholar]
- (676).Vonlanthen S; Heighway J; Altermatt HJ; Gugger M; Kappeler A; Borner MM; van Lohuizen M; Betticher DC The Bmi-1 Oncoprotein Is Differentially Expressed in Non-Small Cell Lung Cancer and Correlates with INK4A-ARF Locus Expression. Br J Cancer 2001, 84 (10), 1372–1376. 10.1054/bjoc.2001.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (677).Dimri GP; Martinez J-L; Jacobs JJL; Keblusek P; Itahana K; Van Lohuizen M; Campisi J; Wazer DE; Band V The Bmi-1 Oncogene Induces Telomerase Activity and Immortalizes Human Mammary Epithelial Cells. Cancer Res 2002, 62 (16), 4736–4745. [PubMed] [Google Scholar]
- (678).Park I; Qian D; Kiel M; Becker MW; Pihalja M; Weissman IL; Morrison SJ; Clarke MF Bmi-1 Is Required for Maintenance of Adult Self-Renewing Haematopoietic Stem Cells. Nature 2003, 423 (6937), 302–305. 10.1038/nature01587. [DOI] [PubMed] [Google Scholar]
- (679).Shukla S; Ying W; Gray F; Yao Y; Simes ML; Zhao Q; Miao H; Cho HJ; González-Alonso P; Winkler A; Lund G; Purohit T; Kim E; Zhang X; Ray JM; He S; Nikolaidis C; Ndoj J; Wang J; Jaremko Ł; Jaremko M; Ryan RJH; Guzman ML; Grembecka J; Cierpicki T Small-Molecule Inhibitors Targeting Polycomb Repressive Complex 1 RING Domain. Nat Chem Biol 2021, 17 (7), 784–793. 10.1038/s41589-021-00815-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (680).Kreso A; van Galen P; Pedley NM; Lima-Fernandes E; Frelin C; Davis T; Cao L; Baiazitov R; Du W; Sydorenko N; Moon Y-C; Gibson L; Wang Y; Leung C; Iscove NN; Arrowsmith CH; Szentgyorgyi E; Gallinger S; Dick JE; O’Brien CA Self-Renewal as a Therapeutic Target in Human Colorectal Cancer. Nat Med 2014, 20 (1), 29–36. 10.1038/nm.3418. [DOI] [PubMed] [Google Scholar]
- (681).Chen D; Wu M; Li Y; Chang I; Yuan Q; Ekimyan-Salvo M; Deng P; Yu B; Yu Y; Dong J; Szymanski JM; Ramadoss S; Li J; Wang C-Y Targeting BMI1+ Cancer Stem Cells Overcomes Chemoresistance and Inhibits Metastases in Squamous Cell Carcinoma. Cell Stem Cell 2017, 20 (5), 621–634.e6. 10.1016/j.stem.2017.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (682).Sulaiman S; Arafat K; Iratni R; Attoub S PTC-209 Anti-Cancer Effects Involved the Inhibition of STAT3 Phosphorylation. Front Pharmacol 2019, 10, 1199. 10.3389/fphar.2019.01199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (683).Bolomsky A; Schlangen K; Schreiner W; Zojer N; Ludwig H Targeting of BMI-1 with PTC-209 Shows Potent Anti-Myeloma Activity and Impairs the Tumour Microenvironment. J Hematol Oncol 2016, 9, 17. 10.1186/s13045-016-0247-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (684).Ismail IH; McDonald D; Strickfaden H; Xu Z; Hendzel MJ A Small Molecule Inhibitor of Polycomb Repressive Complex 1 Inhibits Ubiquitin Signaling at DNA Double-Strand Breaks. J Biol Chem 2013, 288 (37), 26944–26954. 10.1074/jbc.M113.461699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (685).Alchanati I; Teicher C; Cohen G; Shemesh V; Barr HM; Nakache P; Ben-Avraham D; Idelevich A; Angel I; Livnah N; Tuvia S; Reiss Y; Taglicht D; Erez O The E3 Ubiquitin-Ligase Bmi1/Ring1A Controls the Proteasomal Degradation of Top2alpha Cleavage Complex - a Potentially New Drug Target. PLoS One 2009, 4 (12), e8104. 10.1371/journal.pone.0008104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (686).Su W; Han HH; Wang Y; Zhang B; Zhou B; Cheng Y; Rumandla A; Gurrapu S; Chakraborty G; Su J; Yang G; Liang X; Wang G; Rosen N; Scher HI; Ouerfelli O; Giancotti FG The Polycomb Repressor Complex 1 Drives Double-Negative Prostate Cancer Metastasis by Coordinating Stemness and Immune Suppression. Cancer Cell 2019, 36 (2), 139–155.e10. 10.1016/j.ccell.2019.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (687).Bartucci M; Hussein MS; Huselid E; Flaherty K; Patrizii M; Laddha SV; Kui C; Bigos RA; Gilleran JA; El Ansary MMS; Awad MAM; Kimball SD; Augeri DJ; Sabaawy HE Synthesis and Characterization of Novel BMI1 Inhibitors Targeting Cellular Self-Renewal in Hepatocellular Carcinoma. Target Oncol 2017, 12 (4), 449–462. 10.1007/s11523-017-0501-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (688).Nishida Y; Maeda A; Kim MJ; Cao L; Kubota Y; Ishizawa J; AlRawi A; Kato Y; Iwama A; Fujisawa M; Matsue K; Weetall M; Dumble M; Andreeff M; Davis TW; Branstrom A; Kimura S; Kojima K The Novel BMI-1 Inhibitor PTC596 Downregulates MCL-1 and Induces P53-Independent Mitochondrial Apoptosis in Acute Myeloid Leukemia Progenitor Cells. Blood Cancer J 2017, 7 (2), e527. 10.1038/bcj.2017.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (689).Yao Y; Simes ML; Ying W; Zhao Q; Winkler A; Shukla S; Gray F; Nikolaidis C; Hewett G; Grembecka J; Cierpicki T Development of PRC1 Inhibitors Employing Fragment-Based Approach and NMR-Guided Optimization. J Med Chem 2025, 68 (2), 1382–1396. 10.1021/acs.jmedchem.4c01955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (690).Park K-S; Qin L; Kabir M; Luo K; Dale B; Zhong Y; Kim A; Wang GG; Kaniskan HÜ; Jin J Targeted Degradation of PRC1 Components, BMI1 and RING1B, via a Novel Protein Complex Degrader Strategy. Adv Sci (Weinh) 2023, 10 (10), e2205573. 10.1002/advs.202205573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (691).Stuckey JI; Dickson BM; Cheng N; Liu Y; Norris JL; Cholensky SH; Tempel W; Qin S; Huber KG; Sagum C; Black K; Li F; Huang X-P; Roth BL; Baughman BM; Senisterra G; Pattenden SG; Vedadi M; Brown PJ; Bedford MT; Min J; Arrowsmith CH; James LI; Frye SV A Cellular Chemical Probe Targeting the Chromodomains of Polycomb Repressive Complex 1. Nat Chem Biol 2016, 12 (3), 180–187. 10.1038/nchembio.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (692).Cao Q; Wang X; Zhao M; Yang R; Malik R; Qiao Y; Poliakov A; Yocum AK; Li Y; Chen W; Cao X; Jiang X; Dahiya A; Harris C; Feng FY; Kalantry S; Qin ZS; Dhanasekaran SM; Chinnaiyan AM The Central Role of EED in the Orchestration of Polycomb Group Complexes. Nat Commun 2014, 5, 3127. 10.1038/ncomms4127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (693).Kabir M; Qin L; Luo K; Xiong Y; Sidi RA; Park K-S; Jin J Discovery and Characterization of a Novel Cereblon-Recruiting PRC1 Bridged PROTAC Degrader. J Med Chem 2024, 67 (8), 6880–6892. 10.1021/acs.jmedchem.4c00538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (694).Centore RC; Sandoval GJ; Soares LMM; Kadoch C; Chan HM Mammalian SWI/SNF Chromatin Remodeling Complexes: Emerging Mechanisms and Therapeutic Strategies. Trends Genet 2020, 36 (12), 936–950. 10.1016/j.tig.2020.07.011. [DOI] [PubMed] [Google Scholar]
- (695).Mittal P; Roberts CWM The SWI/SNF Complex in Cancer - Biology, Biomarkers and Therapy. Nat Rev Clin Oncol 2020, 17 (7), 435–448. 10.1038/s41571-020-0357-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (696).Innis SM; Cabot B GBAF, a Small BAF Sub-Complex with Big Implications: A Systematic Review. Epigenetics Chromatin 2020, 13 (1), 48. 10.1186/s13072-020-00370-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (697).Mashtalir N; D’Avino AR; Michel BC; Luo J; Pan J; Otto JE; Zullow HJ; McKenzie ZM; Kubiak RL; St Pierre R; Valencia AM; Poynter SJ; Cassel SH; Ranish JA; Kadoch C Modular Organization and Assembly of SWI/SNF Family Chromatin Remodeling Complexes. Cell 2018, 175 (5), 1272–1288.e20. 10.1016/j.cell.2018.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (698).Gatchalian J; Malik S; Ho J; Lee D-S; Kelso TWR; Shokhirev MN; Dixon JR; Hargreaves DC A Non-Canonical BRD9-Containing BAF Chromatin Remodeling Complex Regulates Naive Pluripotency in Mouse Embryonic Stem Cells. Nat Commun 2018, 9 (1), 5139. 10.1038/s41467-018-07528-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (699).Alpsoy A; Dykhuizen EC Glioma Tumor Suppressor Candidate Region Gene 1 (GLTSCR1) and Its Paralog GLTSCR1-like Form SWI/SNF Chromatin Remodeling Subcomplexes. J Biol Chem 2018, 293 (11), 3892–3903. 10.1074/jbc.RA117.001065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (700).Kwon H; Imbalzano AN; Khavari PA; Kingston RE; Green MR Nucleosome Disruption and Enhancement of Activator Binding by a Human SW1/SNF Complex. Nature 1994, 370 (6489), 477–481. 10.1038/370477a0. [DOI] [PubMed] [Google Scholar]
- (701).Michel BC; D’Avino AR; Cassel SH; Mashtalir N; McKenzie ZM; McBride MJ; Valencia AM; Zhou Q; Bocker M; Soares LMM; Pan J; Remillard DI; Lareau CA; Zullow HJ; Fortoul N; Gray NS; Bradner JE; Chan HM; Kadoch C A Non-Canonical SWI/SNF Complex Is a Synthetic Lethal Target in Cancers Driven by BAF Complex Perturbation. Nat Cell Biol 2018, 20 (12), 1410–1420. 10.1038/s41556-018-0221-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (702).Kadoch C; Hargreaves DC; Hodges C; Elias L; Ho L; Ranish J; Crabtree GR Proteomic and Bioinformatic Analysis of Mammalian SWI/SNF Complexes Identifies Extensive Roles in Human Malignancy. Nat Genet 2013, 45 (6), 592–601. 10.1038/ng.2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (703).Haldar M; Randall RL; Capecchi MR Synovial Sarcoma: From Genetics to Genetic-Based Animal Modeling. Clin Orthop Relat Res 2008, 466 (9), 2156–2167. 10.1007/s11999-008-0340-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (704).Clark J; Rocques PJ; Crew AJ; Gill S; Shipley J; Chan AM; Gusterson BA; Cooper CS Identification of Novel Genes, SYT and SSX, Involved in the t(X;18)(P11.2;Q11.2) Translocation Found in Human Synovial Sarcoma. Nat Genet 1994, 7 (4), 502–508. 10.1038/ng0894-502. [DOI] [PubMed] [Google Scholar]
- (705).de Leeuw B; Balemans M; Olde Weghuis D; Geurts van Kessel A Identification of Two Alternative Fusion Genes, SYT-SSX1 and SYT-SSX2, in t(X;18)(P11.2;Q11.2)-Positive Synovial Sarcomas. Hum Mol Genet 1995, 4 (6), 1097–1099. 10.1093/hmg/4.6.1097. [DOI] [PubMed] [Google Scholar]
- (706).Skytting B; Nilsson G; Brodin B; Xie Y; Lundeberg J; Uhlén M; Larsson O A Novel Fusion Gene, SYT-SSX4, in Synovial Sarcoma. J Natl Cancer Inst 1999, 91 (11), 974–975. 10.1093/jnci/91.11.974. [DOI] [PubMed] [Google Scholar]
- (707).He M; Abro B; Kaushal M; Chen L; Chen T; Gondim M; Yan W; Neidich J; Dehner LP; Pfeifer JD Tumor Mutation Burden and Checkpoint Immunotherapy Markers in Primary and Metastatic Synovial Sarcoma. Hum Pathol 2020, 100, 15–23. 10.1016/j.humpath.2020.04.007. [DOI] [PubMed] [Google Scholar]
- (708).Kadoch C; Crabtree GR Reversible Disruption of MSWI/SNF (BAF) Complexes by the SS18-SSX Oncogenic Fusion in Synovial Sarcoma. Cell 2013, 153 (1), 71–85. 10.1016/j.cell.2013.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (709).Middeljans E; Wan X; Jansen PW; Sharma V; Stunnenberg HG; Logie C SS18 Together with Animal-Specific Factors Defines Human BAF-Type SWI/SNF Complexes. PLoS One 2012, 7 (3), e33834. 10.1371/journal.pone.0033834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (710).Banito A; Li X; Laporte AN; Roe J-S; Sanchez-Vega F; Huang C-H; Dancsok AR; Hatzi K; Chen C-C; Tschaharganeh DF; Chandwani R; Tasdemir N; Jones KB; Capecchi MR; Vakoc CR; Schultz N; Ladanyi M; Nielsen TO; Lowe SW The SS18-SSX Oncoprotein Hijacks KDM2B-PRC1.1 to Drive Synovial Sarcoma. Cancer Cell 2018, 34 (2), 346–348. 10.1016/j.ccell.2018.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (711).McBride MJ; Pulice JL; Beird HC; Ingram DR; D’Avino AR; Shern JF; Charville GW; Hornick JL; Nakayama RT; Garcia-Rivera EM; Araujo DM; Wang W-L; Tsai J-W; Yeagley M; Wagner AJ; Futreal PA; Khan J; Lazar AJ; Kadoch C The SS18-SSX Fusion Oncoprotein Hijacks BAF Complex Targeting and Function to Drive Synovial Sarcoma. Cancer Cell 2018, 33 (6), 1128–1141.e7. 10.1016/j.ccell.2018.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (712).Biegel JA; Zhou JY; Rorke LB; Stenstrom C; Wainwright LM; Fogelgren B Germ-Line and Acquired Mutations of INI1 in Atypical Teratoid and Rhabdoid Tumors. Cancer Res 1999, 59 (1), 74–79. [PubMed] [Google Scholar]
- (713).Versteege I; Sévenet N; Lange J; Rousseau-Merck MF; Ambros P; Handgretinger R; Aurias A; Delattre O Truncating Mutations of HSNF5/INI1 in Aggressive Paediatric Cancer. Nature 1998, 394 (6689), 203–206. 10.1038/28212. [DOI] [PubMed] [Google Scholar]
- (714).Wang X; Wang S; Troisi EC; Howard TP; Haswell JR; Wolf BK; Hawk WH; Ramos P; Oberlick EM; Tzvetkov EP; Ross A; Vazquez F; Hahn WC; Park PJ; Roberts CWM BRD9 Defines a SWI/SNF Sub-Complex and Constitutes a Specific Vulnerability in Malignant Rhabdoid Tumors. Nat Commun 2019, 10 (1), 1881. 10.1038/s41467-019-09891-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (715).Brien GL; Remillard D; Shi J; Hemming ML; Chabon J; Wynne K; Dillon ET; Cagney G; Van Mierlo G; Baltissen MP; Vermeulen M; Qi J; Fröhling S; Gray NS; Bradner JE; Vakoc CR; Armstrong SA Targeted Degradation of BRD9 Reverses Oncogenic Gene Expression in Synovial Sarcoma. Elife 2018, 7. 10.7554/eLife.41305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (716).Clark PGK; Vieira LCC; Tallant C; Fedorov O; Singleton DC; Rogers CM; Monteiro OP; Bennett JM; Baronio R; Müller S; Daniels DL; Méndez J; Knapp S; Brennan PE; Dixon DJ LP99: Discovery and Synthesis of the First Selective BRD7/9 Bromodomain Inhibitor. Angew Chem Int Ed Engl 2015, 54 (21), 6217–6221. 10.1002/anie.201501394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (717).Fedorov O; Lingard H; Wells C; Monteiro OP; Picaud S; Keates T; Yapp C; Philpott M; Martin SJ; Felletar I; Marsden BD; Filippakopoulos P; Müller S; Knapp S; Brennan PE [1,2,4]Triazolo[4,3-a]Phthalazines: Inhibitors of Diverse Bromodomains. J Med Chem 2014, 57 (2), 462–476. 10.1021/jm401568s. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (718).Picaud S; Strocchia M; Terracciano S; Lauro G; Mendez J; Daniels DL; Riccio R; Bifulco G; Bruno I; Filippakopoulos P 9H-Purine Scaffold Reveals Induced-Fit Pocket Plasticity of the BRD9 Bromodomain. J Med Chem 2015, 58 (6), 2718–2736. 10.1021/jm501893k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (719).Theodoulou NH; Bamborough P; Bannister AJ; Becher I; Bit RA; Che KH; Chung C; Dittmann A; Drewes G; Drewry DH; Gordon L; Grandi P; Leveridge M; Lindon M; Michon A-M; Molnar J; Robson SC; Tomkinson NCO; Kouzarides T; Prinjha RK; Humphreys PG Discovery of I-BRD9, a Selective Cell Active Chemical Probe for Bromodomain Containing Protein 9 Inhibition. J Med Chem 2016, 59 (4), 1425–1439. 10.1021/acs.jmedchem.5b00256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (720).Martin LJ; Koegl M; Bader G; Cockcroft X-L; Fedorov O; Fiegen D; Gerstberger T; Hofmann MH; Hohmann AF; Kessler D; Knapp S; Knesl P; Kornigg S; Müller S; Nar H; Rogers C; Rumpel K; Schaaf O; Steurer S; Tallant C; Vakoc CR; Zeeb M; Zoephel A; Pearson M; Boehmelt G; McConnell D Structure-Based Design of an in Vivo Active Selective BRD9 Inhibitor. J Med Chem 2016, 59 (10), 4462–4475. 10.1021/acs.jmedchem.5b01865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (721).Shi J; Whyte WA; Zepeda-Mendoza CJ; Milazzo JP; Shen C; Roe J-S; Minder JL; Mercan F; Wang E; Eckersley-Maslin MA; Campbell AE; Kawaoka S; Shareef S; Zhu Z; Kendall J; Muhar M; Haslinger C; Yu M; Roeder RG; Wigler MH; Blobel GA; Zuber J; Spector DL; Young RA; Vakoc CR Role of SWI/SNF in Acute Leukemia Maintenance and Enhancer-Mediated Myc Regulation. Genes Dev 2013, 27 (24), 2648–2662. 10.1101/gad.232710.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (722).Hohmann AF; Martin LJ; Minder JL; Roe J-S; Shi J; Steurer S; Bader G; McConnell D; Pearson M; Gerstberger T; Gottschamel T; Thompson D; Suzuki Y; Koegl M; Vakoc CR Sensitivity and Engineered Resistance of Myeloid Leukemia Cells to BRD9 Inhibition. Nat Chem Biol 2016, 12 (9), 672–679. 10.1038/nchembio.2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (723).Zheng P; Zhang J; Ma H; Yuan X; Chen P; Zhou J; Zhang H Design, Synthesis and Biological Evaluation of Imidazo[1,5-a]Pyrazin-8(7H)-One Derivatives as BRD9 Inhibitors. Bioorg Med Chem 2019, 27 (7), 1391–1404. 10.1016/j.bmc.2019.02.045. [DOI] [PubMed] [Google Scholar]
- (724).Clegg MA; Bamborough P; Chung C-W; Craggs PD; Gordon L; Grandi P; Leveridge M; Lindon M; Liwicki GM; Michon A-M; Molnar J; Rioja I; Soden PE; Theodoulou NH; Werner T; Tomkinson NCO; Prinjha RK; Humphreys PG Application of Atypical Acetyl-Lysine Methyl Mimetics in the Development of Selective Inhibitors of the Bromodomain-Containing Protein 7 (BRD7)/Bromodomain-Containing Protein 9 (BRD9) Bromodomains. J Med Chem 2020, 63 (11), 5816–5840. 10.1021/acs.jmedchem.0c00075. [DOI] [PubMed] [Google Scholar]
- (725).Hügle M; Regenass P; Warstat R; Hau M; Schmidtkunz K; Lucas X; Wohlwend D; Einsle O; Jung M; Breit B; Günther S 4-Acyl Pyrroles as Dual BET-BRD7/9 Bromodomain Inhibitors Address BETi Insensitive Human Cancer Cell Lines. J Med Chem 2020, 63 (24), 15603–15620. 10.1021/acs.jmedchem.0c00478. [DOI] [PubMed] [Google Scholar]
- (726).Gazzillo E; Pierri M; Colarusso E; Chini MG; Ferraro MG; Piccolo M; Irace C; Bruno I; Bifulco G; Terracciano S; Lauro G Exploring the Chemical Space of Functionalized [1,2,4]Triazolo[4,3-a]Quinoxaline-Based Compounds Targeting the Bromodomain of BRD9. Bioorg Chem 2023, 139, 106677. 10.1016/j.bioorg.2023.106677. [DOI] [PubMed] [Google Scholar]
- (727).Shi J; Wang E; Milazzo JP; Wang Z; Kinney JB; Vakoc CR Discovery of Cancer Drug Targets by CRISPR-Cas9 Screening of Protein Domains. Nat Biotechnol 2015, 33 (6), 661–667. 10.1038/nbt.3235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (728).Khavari PA; Peterson CL; Tamkun JW; Mendel DB; Crabtree GR BRG1 Contains a Conserved Domain of the SWI2/SNF2 Family Necessary for Normal Mitotic Growth and Transcription. Nature 1993, 366 (6451), 170–174. 10.1038/366170a0. [DOI] [PubMed] [Google Scholar]
- (729).Medina PP; Carretero J; Fraga MF; Esteller M; Sidransky D; Sanchez-Cespedes M Genetic and Epigenetic Screening for Gene Alterations of the Chromatin-Remodeling Factor, SMARCA4/BRG1, in Lung Tumors. Genes Chromosomes Cancer 2004, 41 (2), 170–177. 10.1002/gcc.20068. [DOI] [PubMed] [Google Scholar]
- (730).Blanco R; Iwakawa R; Tang M; Kohno T; Angulo B; Pio R; Montuenga LM; Minna JD; Yokota J; Sanchez-Cespedes M A Gene-Alteration Profile of Human Lung Cancer Cell Lines. Hum Mutat 2009, 30 (8), 1199–1206. 10.1002/humu.21028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (731).Rodriguez-Nieto S; Cañada A; Pros E; Pinto AI; Torres-Lanzas J; Lopez-Rios F; Sanchez-Verde L; Pisano DG; Sanchez-Cespedes M Massive Parallel DNA Pyrosequencing Analysis of the Tumor Suppressor BRG1/SMARCA4 in Lung Primary Tumors. Hum Mutat 2011, 32 (2), E1999–2017. 10.1002/humu.21415. [DOI] [PubMed] [Google Scholar]
- (732).Imielinski M; Berger AH; Hammerman PS; Hernandez B; Pugh TJ; Hodis E; Cho J; Suh J; Capelletti M; Sivachenko A; Sougnez C; Auclair D; Lawrence MS; Stojanov P; Cibulskis K; Choi K; de Waal L; Sharifnia T; Brooks A; Greulich H; Banerji S; Zander T; Seidel D; Leenders F; Ansén S; Ludwig C; Engel-Riedel W; Stoelben E; Wolf J; Goparju C; Thompson K; Winckler W; Kwiatkowski D; Johnson BE; Jänne PA; Miller VA; Pao W; Travis WD; Pass HI; Gabriel SB; Lander ES; Thomas RK; Garraway LA; Getz G; Meyerson M Mapping the Hallmarks of Lung Adenocarcinoma with Massively Parallel Sequencing. Cell 2012, 150 (6), 1107–1120. 10.1016/j.cell.2012.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (733).Love C; Sun Z; Jima D; Li G; Zhang J; Miles R; Richards KL; Dunphy CH; Choi WWL; Srivastava G; Lugar PL; Rizzieri DA; Lagoo AS; Bernal-Mizrachi L; Mann KP; Flowers CR; Naresh KN; Evens AM; Chadburn A; Gordon LI; Czader MB; Gill JI; Hsi ED; Greenough A; Moffitt AB; McKinney M; Banerjee A; Grubor V; Levy S; Dunson DB; Dave SS The Genetic Landscape of Mutations in Burkitt Lymphoma. Nat Genet 2012, 44 (12), 1321–1325. 10.1038/ng.2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (734).Pugh TJ; Weeraratne SD; Archer TC; Pomeranz Krummel DA; Auclair D; Bochicchio J; Carneiro MO; Carter SL; Cibulskis K; Erlich RL; Greulich H; Lawrence MS; Lennon NJ; McKenna A; Meldrim J; Ramos AH; Ross MG; Russ C; Shefler E; Sivachenko A; Sogoloff B; Stojanov P; Tamayo P; Mesirov JP; Amani V; Teider N; Sengupta S; Francois JP; Northcott PA; Taylor MD; Yu F; Crabtree GR; Kautzman AG; Gabriel SB; Getz G; Jäger N; Jones DTW; Lichter P; Pfister SM; Roberts TM; Meyerson M; Pomeroy SL; Cho Y-J Medulloblastoma Exome Sequencing Uncovers Subtype-Specific Somatic Mutations. Nature 2012, 488 (7409), 106–110. 10.1038/nature11329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (735).Jones DTW; Jäger N; Kool M; Zichner T; Hutter B; Sultan M; Cho Y-J; Pugh TJ; Hovestadt V; Stütz AM; Rausch T; Warnatz H-J; Ryzhova M; Bender S; Sturm D; Pleier S; Cin H; Pfaff E; Sieber L; Wittmann A; Remke M; Witt H; Hutter S; Tzaridis T; Weischenfeldt J; Raeder B; Avci M; Amstislavskiy V; Zapatka M; Weber UD; Wang Q; Lasitschka B; Bartholomae CC; Schmidt M; von Kalle C; Ast V; Lawerenz C; Eils J; Kabbe R; Benes V; van Sluis P; Koster J; Volckmann R; Shih D; Betts MJ; Russell RB; Coco S; Tonini GP; Schüller U; Hans V; Graf N; Kim Y-J; Monoranu C; Roggendorf W; Unterberg A; Herold-Mende C; Milde T; Kulozik AE; von Deimling A; Witt O; Maass E; Rössler J; Ebinger M; Schuhmann MU; Frühwald MC; Hasselblatt M; Jabado N; Rutkowski S; von Bueren AO; Williamson D; Clifford SC; McCabe MG; Collins VP; Wolf S; Wiemann S; Lehrach H; Brors B; Scheurlen W; Felsberg J; Reifenberger G; Northcott PA; Taylor MD; Meyerson M; Pomeroy SL; Yaspo M-L; Korbel JO; Korshunov A; Eils R; Pfister SM; Lichter P Dissecting the Genomic Complexity Underlying Medulloblastoma. Nature 2012, 488 (7409), 100–105. 10.1038/nature11284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (736).Parsons DW; Li M; Zhang X; Jones S; Leary RJ; Lin JC-H; Boca SM; Carter H; Samayoa J; Bettegowda C; Gallia GL; Jallo GI; Binder ZA; Nikolsky Y; Hartigan J; Smith DR; Gerhard DS; Fults DW; VandenBerg S; Berger MS; Marie SKN; Shinjo SMO; Clara C; Phillips PC; Minturn JE; Biegel JA; Judkins AR; Resnick AC; Storm PB; Curran T; He Y; Rasheed BA; Friedman HS; Keir ST; McLendon R; Northcott PA; Taylor MD; Burger PC; Riggins GJ; Karchin R; Parmigiani G; Bigner DD; Yan H; Papadopoulos N; Vogelstein B; Kinzler KW; Velculescu VE The Genetic Landscape of the Childhood Cancer Medulloblastoma. Science 2011, 331 (6016), 435–439. 10.1126/science.1198056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (737).Shain AH; Giacomini CP; Matsukuma K; Karikari CA; Bashyam MD; Hidalgo M; Maitra A; Pollack JR Convergent Structural Alterations Define SWItch/Sucrose NonFermentable (SWI/SNF) Chromatin Remodeler as a Central Tumor Suppressive Complex in Pancreatic Cancer. Proc Natl Acad Sci U S A 2012, 109 (5), E252–9. 10.1073/pnas.1114817109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (738).Hodis E; Watson IR; Kryukov GV; Arold ST; Imielinski M; Theurillat J-P; Nickerson E; Auclair D; Li L; Place C; Dicara D; Ramos AH; Lawrence MS; Cibulskis K; Sivachenko A; Voet D; Saksena G; Stransky N; Onofrio RC; Winckler W; Ardlie K; Wagle N; Wargo J; Chong K; Morton DL; Stemke-Hale K; Chen G; Noble M; Meyerson M; Ladbury JE; Davies MA; Gershenwald JE; Wagner SN; Hoon DSB; Schadendorf D; Lander ES; Gabriel SB; Getz G; Garraway LA; Chin L A Landscape of Driver Mutations in Melanoma. Cell 2012, 150 (2), 251–263. 10.1016/j.cell.2012.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (739).Oike T; Ogiwara H; Tominaga Y; Ito K; Ando O; Tsuta K; Mizukami T; Shimada Y; Isomura H; Komachi M; Furuta K; Watanabe S-I; Nakano T; Yokota J; Kohno T A Synthetic Lethality-Based Strategy to Treat Cancers Harboring a Genetic Deficiency in the Chromatin Remodeling Factor BRG1. Cancer Res 2013, 73 (17), 5508–5518. 10.1158/0008-5472.CAN-12-4593. [DOI] [PubMed] [Google Scholar]
- (740).Wilson BG; Helming KC; Wang X; Kim Y; Vazquez F; Jagani Z; Hahn WC; Roberts CWM Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation. Mol Cell Biol 2014, 34 (6), 1136–1144. 10.1128/MCB.01372-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (741).Fedorov O; Castex J; Tallant C; Owen DR; Martin S; Aldeghi M; Monteiro O; Filippakopoulos P; Picaud S; Trzupek JD; Gerstenberger BS; Bountra C; Willmann D; Wells C; Philpott M; Rogers C; Biggin PC; Brennan PE; Bunnage ME; Schüle R; Günther T; Knapp S; Müller S Selective Targeting of the BRG/PB1 Bromodomains Impairs Embryonic and Trophoblast Stem Cell Maintenance. Sci Adv 2015, 1 (10), e1500723. 10.1126/sciadv.1500723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (742).Gerstenberger BS; Trzupek JD; Tallant C; Fedorov O; Filippakopoulos P; Brennan PE; Fedele V; Martin S; Picaud S; Rogers C; Parikh M; Taylor A; Samas B; O’Mahony A; Berg E; Pallares G; Torrey AD; Treiber DK; Samardjiev IJ; Nasipak BT; Padilla-Benavides T; Wu Q; Imbalzano AN; Nickerson JA; Bunnage ME; Müller S; Knapp S; Owen DR Identification of a Chemical Probe for Family VIII Bromodomains through Optimization of a Fragment Hit. J Med Chem 2016, 59 (10), 4800–4811. 10.1021/acs.jmedchem.6b00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (743).Armstrong JA; Papoulas O; Daubresse G; Sperling AS; Lis JT; Scott MP; Tamkun JW The Drosophila BRM Complex Facilitates Global Transcription by RNA Polymerase II. EMBO J 2002, 21 (19), 5245–5254. 10.1093/emboj/cdf517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (744).Papillon JPN; Nakajima K; Adair CD; Hempel J; Jouk AO; Karki RG; Mathieu S; Möbitz H; Ntaganda R; Smith T; Visser M; Hill SE; Hurtado FK; Chenail G; Bhang H-EC; Bric A; Xiang K; Bushold G; Gilbert T; Vattay A; Dooley J; Costa EA; Park I; Li A; Farley D; Lounkine E; Yue QK; Xie X; Zhu X; Kulathila R; King D; Hu T; Vulic K; Cantwell J; Luu C; Jagani Z 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 2018, 61 (22), 10155–10172. 10.1021/acs.jmedchem.8b01318. [DOI] [PubMed] [Google Scholar]
- (745).Schick S; Grosche S; Kohl KE; Drpic D; Jaeger MG; Marella NC; Imrichova H; Lin J-MG; Hofstätter G; Schuster M; Rendeiro AF; Koren A; Petronczki M; Bock C; Müller AC; Winter GE; Kubicek S Acute BAF Perturbation Causes Immediate Changes in Chromatin Accessibility. Nat Genet 2021, 53 (3), 269–278. 10.1038/s41588-021-00777-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (746).Iurlaro M; Stadler MB; Masoni F; Jagani Z; Galli GG; Schübeler D Mammalian SWI/SNF Continuously Restores Local Accessibility to Chromatin. Nat Genet 2021, 53 (3), 279–287. 10.1038/s41588-020-00768-w. [DOI] [PubMed] [Google Scholar]
- (747).Martin BJE; Ablondi EF; Goglia C; Mimoso CA; Espinel-Cabrera PR; Adelman K Global Identification of SWI/SNF Targets Reveals Compensation by EP400. Cell 2023, 186 (24), 5290–5307.e26. 10.1016/j.cell.2023.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (748).Rago F; Rodrigues LU; Bonney M; Sprouffske K; Kurth E; Elliott G; Ambrose J; Aspesi P; Oborski J; Chen JT; McDonald ER; Mapa FA; Ruddy DA; Kauffmann A; Abrams T; Bhang H-EC; Jagani Z Exquisite Sensitivity to Dual BRG1/BRM ATPase Inhibitors Reveals Broad SWI/SNF Dependencies in Acute Myeloid Leukemia. Mol Cancer Res 2022, 20 (3), 361–372. 10.1158/1541-7786.MCR-21-0390. [DOI] [PubMed] [Google Scholar]
- (749).Jagani Z; Chenail G; Xiang K; Bushold G; Bhang H-EC; Li A; Elliott G; Zhu J; Vattay A; Gilbert T; Bric A; Kikkawa R; Dubost V; Terranova R; Cantwell J; Luu C; Silver S; Shirley M; Huet F; Maher R; Reece-Hoyes J; Ruddy D; Rakiec D; Korn J; Russ C; Ruda V; Dooley J; Costa E; Park I; Moebitz H; Nakajima K; Adair CD; Mathieu S; Ntaganda R; Smith T; Farley D; King D; Xie X; Kulathila R; Hu T; Pan X; Ma Q; Vulic K; Rago F; Clarkson S; Ge R; Sigoillot F; Pardee G; Bagdasarian L; McLaughlin M; Haas K; Weiler J; Kovats S; Jaskelioff M; Apolline-Gerard M; Beil J; Naumann U; Fortin P; Stegmeier FP; Acker MG; Williams J; Meyer M; Bradner JE; Keen N; Sellers WR; Hofmann F; Engelman JA; Stuart D; Papillon JPN In-Depth Characterization and Validation in BRG1-Mutant Lung Cancers Define Novel Catalytic Inhibitors of SWI/SNF Chromatin Remodeling. 2019. 10.1101/812628. [DOI] [Google Scholar]
- (750).DiNardo CD; Savona MR; Kishtagari A; Fathi AT; Bhalla KN; Agresta S; Reilly S; Almon C; Hentemann M; Hickman D; Corrigan D; Macaraeg M; Piel J; Horrigan K; Nabhan S; Martin P; Stein EM Preliminary Results from a Phase 1 Dose Escalation Study of FHD-286, a Novel BRG1/BRM (SMARCA4/SMARCA2) Inhibitor, Administered As an Oral Monotherapy in Patients with Advanced Hematologic Malignancies. Blood 2023, 142 (Supplement 1), 4284–4284. 10.1182/blood-2023-178090. [DOI] [Google Scholar]
- (751).Farnaby W; Koegl M; Roy MJ; Whitworth C; Diers E; Trainor N; Zollman D; Steurer S; Karolyi-Oezguer J; Riedmueller C; Gmaschitz T; Wachter J; Dank C; Galant M; Sharps B; Rumpel K; Traxler E; Gerstberger T; Schnitzer R; Petermann O; Greb P; Weinstabl H; Bader G; Zoephel A; Weiss-Puxbaum A; Ehrenhöfer-Wölfer K; Wöhrle S; Boehmelt G; Rinnenthal J; Arnhof H; Wiechens N; Wu M-Y; Owen-Hughes T; Ettmayer P; Pearson M; McConnell DB; Ciulli A BAF Complex Vulnerabilities in Cancer Demonstrated via Structure-Based PROTAC Design. Nat Chem Biol 2019, 15 (7), 672–680. 10.1038/s41589-019-0294-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (752).Xiao L; Parolia A; Qiao Y; Bawa P; Eyunni S; Mannan R; Carson SE; Chang Y; Wang X; Zhang Y; Vo JN; Kregel S; Simko SA; Delekta AD; Jaber M; Zheng H; Apel IJ; McMurry L; Su F; Wang R; Zelenka-Wang S; Sasmal S; Khare L; Mukherjee S; Abbineni C; Aithal K; Bhakta MS; Ghurye J; Cao X; Navone NM; Nesvizhskii AI; Mehra R; Vaishampayan U; Blanchette M; Wang Y; Samajdar S; Ramachandra M; Chinnaiyan AM Targeting SWI/SNF ATPases in Enhancer-Addicted Prostate Cancer. Nature 2022, 601 (7893), 434–439. 10.1038/s41586-021-04246-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (753).Kofink C; Trainor N; Mair B; Wöhrle S; Wurm M; Mischerikow N; Roy MJ; Bader G; Greb P; Garavel G; Diers E; McLennan R; Whitworth C; Vetma V; Rumpel K; Scharnweber M; Fuchs JE; Gerstberger T; Cui Y; Gremel G; Chetta P; Hopf S; Budano N; Rinnenthal J; Gmaschitz G; Mayer M; Koegl M; Ciulli A; Weinstabl H; Farnaby W A Selective and Orally Bioavailable VHL-Recruiting PROTAC Achieves SMARCA2 Degradation in Vivo. Nat Commun 2022, 13 (1), 5969. 10.1038/s41467-022-33430-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (754).Cantley J; Ye X; Rousseau E; Januario T; Hamman BD; Rose CM; Cheung TK; Hinkle T; Soto L; Quinn C; Harbin A; Bortolon E; Chen X; Haskell R; Lin E; Yu S-F; Del Rosario G; Chan E; Dunlap D; Koeppen H; Martin S; Merchant M; Grimmer M; Broccatelli F; Wang J; Pizzano J; Dragovich PS; Berlin M; Yauch RL Selective PROTAC-Mediated Degradation of SMARCA2 Is Efficacious in SMARCA4 Mutant Cancers. Nat Commun 2022, 13 (1), 6814. 10.1038/s41467-022-34562-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (755).Yang L; Tu W; Huang L; Miao B; Kaneshige A; Jiang W; Leng L; Wang M; Wen B; Sun D; Wang S Discovery of SMD-3040 as a Potent and Selective SMARCA2 PROTAC Degrader with Strong in Vivo Antitumor Activity. J Med Chem 2023, 66 (15), 10761–10781. 10.1021/acs.jmedchem.3c00953. [DOI] [PubMed] [Google Scholar]
- (756).Berlin M; Cantley J; Bookbinder M; Bortolon E; Broccatelli F; Cadelina G; Chan EW; Chen H; Chen X; Cheng Y; Cheung TK; Davenport K; DiNicola D; Gordon D; Hamman BD; Harbin A; Haskell R; He M; Hole AJ; Januario T; Kerry PS; Koenig SG; Li L; Merchant M; Pérez-Dorado I; Pizzano J; Quinn C; Rose CM; Rousseau E; Soto L; Staben LR; Sun H; Tian Q; Wang J; Wang W; Ye CS; Ye X; Zhang P; Zhou Y; Yauch R; Dragovich PS PROTACs Targeting BRM (SMARCA2) Afford Selective In Vivo Degradation over BRG1 (SMARCA4) and Are Active in BRG1 Mutant Xenograft Tumor Models. J Med Chem 2024, 67 (2), 1262–1313. 10.1021/acs.jmedchem.3c01781. [DOI] [PubMed] [Google Scholar]
- (757).Yang L; Tu W; Leng L; Huang L; Jiang W; Wang M; Wang Y; Meagher JL; Chinnaswamy K; Stuckey JA; Wang M; Wen B; Sun D; Harikrishnan L; Strickland C; Rice C; Orth P; Sui Z; Wang S Discovery of SMD-3236: A Potent, Highly Selective and Efficacious SMARCA2 Degrader for the Treatment of SMARC4-Deficient Human Cancers. J Med Chem 2025, 68 (2), 1155–1178. 10.1021/acs.jmedchem.4c01904. [DOI] [PubMed] [Google Scholar]
- (758).Leng L; Tu W; Yang L; Huang L; Wang M; Meagher JL; Chinnaswamy K; Allu SR; Rej RK; Tošović J; Harikrishnan L; Li Z; Sui Z; Stuckey JA; Wang S Discovery of High-Affinity SMARCA2/4 Bromodomain Ligands and Development of Potent and Exceptionally Selective SMARCA2 PROTAC Degraders. J Med Chem 2025, 68 (2), 1113–1133. 10.1021/acs.jmedchem.4c01903. [DOI] [PubMed] [Google Scholar]
- (759).Wilson BG; Roberts CWM SWI/SNF Nucleosome Remodellers and Cancer. Nat Rev Cancer 2011, 11 (7), 481–492. 10.1038/nrc3068. [DOI] [PubMed] [Google Scholar]
- (760).Wu JN; Roberts CWM ARID1A Mutations in Cancer: Another Epigenetic Tumor Suppressor? Cancer Discov 2013, 3 (1), 35–43. 10.1158/2159-8290.CD-12-0361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (761).Pavlidou E; Balis V Diagnostic Significance and Prognostic Role of the ARID1A Gene in Cancer Outcomes (Review). World Acad Sci J 2020, 2 (2), 49–64. 10.3892/wasj.2020.37. [DOI] [Google Scholar]
- (762).Sun X; Wang SC; Wei Y; Luo X; Jia Y; Li L; Gopal P; Zhu M; Nassour I; Chuang J-C; Maples T; Celen C; Nguyen LH; Wu L; Fu S; Li W; Hui L; Tian F; Ji Y; Zhang S; Sorouri M; Hwang TH; Letzig L; James L; Wang Z; Yopp AC; Singal AG; Zhu H Arid1a Has Context-Dependent Oncogenic and Tumor Suppressor Functions in Liver Cancer. Cancer Cell 2017, 32 (5), 574–589.e6. 10.1016/j.ccell.2017.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (763).Jiang T; Chen X; Su C; Ren S; Zhou C Pan-Cancer Analysis of ARID1A Alterations as Biomarkers for Immunotherapy Outcomes. J Cancer 2020, 11 (4), 776–780. 10.7150/jca.41296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (764).Helming KC; Wang X; Wilson BG; Vazquez F; Haswell JR; Manchester HE; Kim Y; Kryukov GV; Ghandi M; Aguirre AJ; Jagani Z; Wang Z; Garraway LA; Hahn WC; Roberts CWM ARID1B Is a Specific Vulnerability in ARID1A-Mutant Cancers. Nat Med 2014, 20 (3), 251–254. 10.1038/nm.3480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (765).Mathur R; Alver BH; San Roman AK; Wilson BG; Wang X; Agoston AT; Park PJ; Shivdasani RA; Roberts CWM ARID1A Loss Impairs Enhancer-Mediated Gene Regulation and Drives Colon Cancer in Mice. Nat Genet 2017, 49 (2), 296–302. 10.1038/ng.3744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (766).Hein KZ; Stephen B; Fu S Therapeutic Role of Synthetic Lethality in ARID1A-Deficient Malignancies. J Immunother Precis Oncol 2024, 7 (1), 41–52. 10.36401/JIPO-22-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (767).Miller RE; Brough R; Bajrami I; Williamson CT; McDade S; Campbell J; Kigozi A; Rafiq R; Pemberton H; Natrajan R; Joel J; Astley H; Mahoney C; Moore JD; Torrance C; Gordan JD; Webber JT; Levin RS; Shokat KM; Bandyopadhyay S; Lord CJ; Ashworth A Synthetic Lethal Targeting of ARID1A-Mutant Ovarian Clear Cell Tumors with Dasatinib. Mol Cancer Ther 2016, 15 (7), 1472–1484. 10.1158/1535-7163.MCT-15-0554. [DOI] [PubMed] [Google Scholar]
- (768).Mandal J; Mandal P; Wang T-L; Shih I-M Treating ARID1A Mutated Cancers by Harnessing Synthetic Lethality and DNA Damage Response. J Biomed Sci 2022, 29 (1), 71. 10.1186/s12929-022-00856-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (769).Vo N; Goodman RH CREB-Binding Protein and P300 in Transcriptional Regulation. Journal of Biological Chemistry 2001, 276 (17), 13505–13508. 10.1074/jbc.R000025200. [DOI] [PubMed] [Google Scholar]
- (770).Dancy BM; Cole PA Protein Lysine Acetylation by P300/CBP. Chem Rev 2015, 115 (6), 2419–2452. 10.1021/cr500452k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (771).Weinert BT; Narita T; Satpathy S; Srinivasan B; Hansen BK; Schölz C; Hamilton WB; Zucconi BE; Wang WW; Liu WR; Brickman JM; Kesicki EA; Lai A; Bromberg KD; Cole PA; Choudhary C Time-Resolved Analysis Reveals Rapid Dynamics and Broad Scope of the CBP/P300 Acetylome. Cell 2018, 174 (1), 231–244.e12. 10.1016/j.cell.2018.04.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (772).Gou P; Zhang W Protein Lysine Acetyltransferase CBP/P300: A Promising Target for Small Molecules in Cancer Treatment. Biomedicine & Pharmacotherapy 2024, 171, 116130. 10.1016/j.biopha.2024.116130. [DOI] [PubMed] [Google Scholar]
- (773).Roelfsema JH; White SJ; Ariyürek Y; Bartholdi D; Niedrist D; Papadia F; Bacino CA; den Dunnen JT; van Ommen G-JB; Breuning MH; Hennekam RC; Peters DJM Genetic Heterogeneity in Rubinstein-Taybi Syndrome: Mutations in Both the CBP and EP300 Genes Cause Disease. The American Journal of Human Genetics 2005, 76 (4), 572–580. 10.1086/429130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (774).Jia D; Augert A; Kim D-W; Eastwood E; Wu N; Ibrahim AH; Kim K-B; Dunn CT; Pillai SPS; Gazdar AF; Bolouri H; Park K-S; MacPherson D Crebbp Loss Drives Small Cell Lung Cancer and Increases Sensitivity to HDAC Inhibition. Cancer Discov 2018, 8 (11), 1422–1437. 10.1158/2159-8290.CD-18-0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (775).George J; Lim JS; Jang SJ; Cun Y; Ozretić L; Kong G; Leenders F; Lu X; Fernández-Cuesta L; Bosco G; Müller C; Dahmen I; Jahchan NS; Park K-S; Yang D; Karnezis AN; Vaka D; Torres A; Wang MS; Korbel JO; Menon R; Chun S-M; Kim D; Wilkerson M; Hayes N; Engelmann D; Pützer B; Bos M; Michels S; Vlasic I; Seidel D; Pinther B; Schaub P; Becker C; Altmüller J; Yokota J; Kohno T; Iwakawa R; Tsuta K; Noguchi M; Muley T; Hoffmann H; Schnabel PA; Petersen I; Chen Y; Soltermann A; Tischler V; Choi C; Kim Y-H; Massion PP; Zou Y; Jovanovic D; Kontic M; Wright GM; Russell PA; Solomon B; Koch I; Lindner M; Muscarella LA; la Torre A; Field JK; Jakopovic M; Knezevic J; Castaños-Vélez E; Roz L; Pastorino U; Brustugun O-T; Lund-Iversen M; Thunnissen E; Köhler J; Schuler M; Botling J; Sandelin M; Sanchez-Cespedes M; Salvesen HB; Achter V; Lang U; Bogus M; Schneider PM; Zander T; Ansén S; Hallek M; Wolf J; Vingron M; Yatabe Y; Travis WD; Nürnberg P; Reinhardt C; Perner S; Heukamp L; Büttner R; Haas SA; Brambilla E; Peifer M; Sage J; Thomas RK Comprehensive Genomic Profiles of Small Cell Lung Cancer. Nature 2015, 524 (7563), 47–53. 10.1038/nature14664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (776).Kishimoto M; Kohno T; Okudela K; Otsuka A; Sasaki H; Tanabe C; Sakiyama T; Hirama C; Kitabayashi I; Minna JD; Takenoshita S; Yokota J Mutations and Deletions of the CBP Gene in Human Lung Cancer. Clin Cancer Res 2005, 11 (2 Pt 1), 512–519. [PubMed] [Google Scholar]
- (777).Muraoka M; Konishi M; Kikuchi-Yanoshita R; Tanaka K; Shitara N; Chong JM; Iwama T; Miyaki M P300 Gene Alterations in Colorectal and Gastric Carcinomas. Oncogene 1996, 12 (7), 1565–1569. [PubMed] [Google Scholar]
- (778).Gayther SA; Batley SJ; Linger L; Bannister A; Thorpe K; Chin SF; Daigo Y; Russell P; Wilson A; Sowter HM; Delhanty JD; Ponder BA; Kouzarides T; Caldas C Mutations Truncating the EP300 Acetylase in Human Cancers. Nat Genet 2000, 24 (3), 300–303. 10.1038/73536. [DOI] [PubMed] [Google Scholar]
- (779).Tillinghast GW; Partee J; Albert P; Kelley JM; Burtow KH; Kelly K Analysis of Genetic Stability at the EP300 and CREBBP Loci in a Panel of Cancer Cell Lines. Genes Chromosomes Cancer 2003, 37 (2), 121–131. 10.1002/gcc.10195. [DOI] [PubMed] [Google Scholar]
- (780).Bryan EJ; Jokubaitis VJ; Chamberlain NL; Baxter SW; Dawson E; Choong DYH; Campbell IG Mutation Analysis of EP300 in Colon, Breast and Ovarian Carcinomas. Int J Cancer 2002, 102 (2), 137–141. 10.1002/ijc.10682. [DOI] [PubMed] [Google Scholar]
- (781).Pasqualucci L; Dominguez-Sola D; Chiarenza A; Fabbri G; Grunn A; Trifonov V; Kasper LH; Lerach S; Tang H; Ma J; Rossi D; Chadburn A; Murty VV; Mullighan CG; Gaidano G; Rabadan R; Brindle PK; Dalla-Favera R Inactivating Mutations of Acetyltransferase Genes in B-Cell Lymphoma. Nature 2011, 471 (7337), 189–195. 10.1038/nature09730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (782).Mullighan CG; Zhang J; Kasper LH; Lerach S; Payne-Turner D; Phillips LA; Heatley SL; Holmfeldt L; Collins-Underwood JR; Ma J; Buetow KH; Pui C-H; Baker SD; Brindle PK; Downing JR CREBBP Mutations in Relapsed Acute Lymphoblastic Leukaemia. Nature 2011, 471 (7337), 235–239. 10.1038/nature09727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (783).Xu W; Fukuyama T; Ney PA; Wang D; Rehg J; Boyd K; van Deursen JMA; Brindle PK Global Transcriptional Coactivators CREB-Binding Protein and P300 Are Highly Essential Collectively but Not Individually in Peripheral B Cells. Blood 2006, 107 (11), 4407–4416. 10.1182/blood-2005-08-3263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (784).Jiang Y; Ortega-Molina A; Geng H; Ying H-Y; Hatzi K; Parsa S; McNally D; Wang L; Doane AS; Agirre X; Teater M; Meydan C; Li Z; Poloway D; Wang S; Ennishi D; Scott DW; Stengel KR; Kranz JE; Holson E; Sharma S; Young JW; Chu C-S; Roeder RG; Shaknovich R; Hiebert SW; Gascoyne RD; Tam W; Elemento O; Wendel H-G; Melnick AM CREBBP Inactivation Promotes the Development of HDAC3-Dependent Lymphomas. Cancer Discov 2017, 7 (1), 38–53. 10.1158/2159-8290.CD-16-0975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (785).Ogiwara H; Sasaki M; Mitachi T; Oike T; Higuchi S; Tominaga Y; Kohno T Targeting P300 Addiction in CBP-Deficient Cancers Causes Synthetic Lethality by Apoptotic Cell Death Due to Abrogation of MYC Expression. Cancer Discov 2016, 6 (4), 430–445. 10.1158/2159-8290.CD-15-0754. [DOI] [PubMed] [Google Scholar]
- (786).Meyer SN; Scuoppo C; Vlasevska S; Bal E; Holmes AB; Holloman M; Garcia-Ibanez L; Nataraj S; Duval R; Vantrimpont T; Basso K; Brooks N; Dalla-Favera R; Pasqualucci L Unique and Shared Epigenetic Programs of the CREBBP and EP300 Acetyltransferases in Germinal Center B Cells Reveal Targetable Dependencies in Lymphoma. Immunity 2019, 51 (3), 535–547.e9. 10.1016/j.immuni.2019.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (787).Giles RH; Petrij F; Dauwerse HG; den Hollander AI; Lushnikova T; van Ommen GJ; Goodman RH; Deaven LL; Doggett NA; Peters DJ; Breuning MH Construction of a 1.2-Mb Contig Surrounding, and Molecular Analysis of, the Human CREB-Binding Protein (CBP/CREBBP) Gene on Chromosome 16p13.3. Genomics 1997, 42 (1), 96–114. 10.1006/geno.1997.4699. [DOI] [PubMed] [Google Scholar]
- (788).Rozman M; Camós M; Colomer D; Villamor N; Esteve J; Costa D; Carrió A; Aymerich M; Aguilar JL; Domingo A; Solé F; Gomis F; Florensa L; Montserrat E; Campo E Type I MOZ/CBP (MYST3/CREBBP) Is the Most Common Chimeric Transcript in Acute Myeloid Leukemia with t(8;16)(P11;P13) Translocation. Genes Chromosomes Cancer 2004, 40 (2), 140–145. 10.1002/gcc.20022. [DOI] [PubMed] [Google Scholar]
- (789).Borrow J; Stanton VP; Andresen JM; Becher R; Behm FG; Chaganti RS; Civin CI; Disteche C; Dubé I; Frischauf AM; Horsman D; Mitelman F; Volinia S; Watmore AE; Housman DE The Translocation t(8;16)(P11;P13) of Acute Myeloid Leukaemia Fuses a Putative Acetyltransferase to the CREB-Binding Protein. Nat Genet 1996, 14 (1), 33–41. 10.1038/ng0996-33. [DOI] [PubMed] [Google Scholar]
- (790).Kitabayashi I; Aikawa Y; Yokoyama A; Hosoda F; Nagai M; Kakazu N; Abe T; Ohki M Fusion of MOZ and P300 Histone Acetyltransferases in Acute Monocytic Leukemia with a t(8;22)(P11;Q13) Chromosome Translocation. Leukemia 2001, 15 (1), 89–94. 10.1038/sj.leu.2401983. [DOI] [PubMed] [Google Scholar]
- (791).Panagopoulos I; Fioretos T; Isaksson M; Samuelsson U; Billström R; Strömbeck B; Mitelman F; Johansson B Fusion of the MORF and CBP Genes in Acute Myeloid Leukemia with the t(10;16)(Q22;P13). Hum Mol Genet 2001, 10 (4), 395–404. 10.1093/hmg/10.4.395. [DOI] [PubMed] [Google Scholar]
- (792).Taki T; Sako M; Tsuchida M; Hayashi Y The t(11;16)(Q23;P13) Translocation in Myelodysplastic Syndrome Fuses the MLL Gene to the CBP Gene. Blood 1997, 89 (11), 3945–3950. [PubMed] [Google Scholar]
- (793).Sobulo OM; Borrow J; Tomek R; Reshmi S; Harden A; Schlegelberger B; Housman D; Doggett NA; Rowley JD; Zeleznik-Le NJ MLL Is Fused to CBP, a Histone Acetyltransferase, in Therapy-Related Acute Myeloid Leukemia with a t(11;16)(Q23;P13.3). Proc Natl Acad Sci U S A 1997, 94 (16), 8732–8737. 10.1073/pnas.94.16.8732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (794).Lau OD; Kundu TK; Soccio RE; Ait-Si-Ali S; Khalil EM; Vassilev A; Wolffe AP; Nakatani Y; Roeder RG; Cole PA HATs off: Selective Synthetic Inhibitors of the Histone Acetyltransferases P300 and PCAF. Mol Cell 2000, 5 (3), 589–595. 10.1016/S1097-2765(00)80452-9. [DOI] [PubMed] [Google Scholar]
- (795).Liu X; Wang L; Zhao K; Thompson PR; Hwang Y; Marmorstein R; Cole PA The Structural Basis of Protein Acetylation by the P300/CBP Transcriptional Coactivator. Nature 2008, 451 (7180), 846–850. 10.1038/nature06546. [DOI] [PubMed] [Google Scholar]
- (796).Balasubramanyam K; Swaminathan V; Ranganathan A; Kundu TK Small Molecule Modulators of Histone Acetyltransferase P300. J Biol Chem 2003, 278 (21), 19134–19140. 10.1074/jbc.M301580200. [DOI] [PubMed] [Google Scholar]
- (797).Balasubramanyam K; Altaf M; Varier RA; Swaminathan V; Ravindran A; Sadhale PP; Kundu TK Polyisoprenylated Benzophenone, Garcinol, a Natural Histone Acetyltransferase Inhibitor, Represses Chromatin Transcription and Alters Global Gene Expression. Journal of Biological Chemistry 2004, 279 (32), 33716–33726. 10.1074/jbc.M402839200. [DOI] [PubMed] [Google Scholar]
- (798).Bowers EM; Yan G; Mukherjee C; Orry A; Wang L; Holbert MA; Crump NT; Hazzalin CA; Liszczak G; Yuan H; Larocca C; Saldanha SA; Abagyan R; Sun Y; Meyers DJ; Marmorstein R; Mahadevan LC; Alani RM; Cole PA Virtual Ligand Screening of the P300/CBP Histone Acetyltransferase: Identification of a Selective Small Molecule Inhibitor. Chem Biol 2010, 17 (5), 471–482. 10.1016/j.chembiol.2010.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (799).Dahlin JL; Nelson KM; Strasser JM; Barsyte-Lovejoy D; Szewczyk MM; Organ S; Cuellar M; Singh G; Shrimp JH; Nguyen N; Meier JL; Arrowsmith CH; Brown PJ; Baell JB; Walters MA Assay Interference and Off-Target Liabilities of Reported Histone Acetyltransferase Inhibitors. Nat Commun 2017, 8 (1), 1527. 10.1038/s41467-017-01657-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (800).Chen YF; Rahman A; Sax JL; Atala Pleshinger MJ; Friedrich RM; Adams DJ C646 Degrades Exportin-1 to Modulate P300 Chromatin Occupancy and Function. Cell Chem Biol 2024, 31 (7), 1363–1372.e8. 10.1016/j.chembiol.2024.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (801).Shrimp JH; Sorum AW; Garlick JM; Guasch L; Nicklaus MC; Meier JL Characterizing the Covalent Targets of a Small Molecule Inhibitor of the Lysine Acetyltransferase P300. ACS Med Chem Lett 2016, 7 (2), 151–155. 10.1021/acsmedchemlett.5b00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (802).Borah JC; Mujtaba S; Karakikes I; Zeng L; Muller M; Patel J; Moshkina N; Morohashi K; Zhang W; Gerona-Navarro G; Hajjar RJ; Zhou M-M A Small Molecule Binding to the Coactivator CREB-Binding Protein Blocks Apoptosis in Cardiomyocytes. Chem Biol 2011, 18 (4), 531–541. 10.1016/j.chembiol.2010.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (803).Gerona-Navarro G; Yoel-Rodríguez; Mujtaba S; Frasca A; Patel J; Zeng L; Plotnikov AN; Osman R; Zhou M-M Rational Design of Cyclic Peptide Modulators of the Transcriptional Coactivator CBP: A New Class of P53 Inhibitors. J Am Chem Soc 2011, 133 (7), 2040–2043. 10.1021/ja107761h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (804).Sachchidanand; Resnick-Silverman L; Yan S; Mutjaba S; Liu W-J; Zeng L; Manfredi JJ; Zhou M-M Target Structure-Based Discovery of Small Molecules That Block Human P53 and CREB Binding Protein Association. Chem Biol 2006, 13 (1), 81–90. 10.1016/j.chembiol.2005.10.014. [DOI] [PubMed] [Google Scholar]
- (805).Hay DA; Fedorov O; Martin S; Singleton DC; Tallant C; Wells C; Picaud S; Philpott M; Monteiro OP; Rogers CM; Conway SJ; Rooney TPC; Tumber A; Yapp C; Filippakopoulos P; Bunnage ME; Müller S; Knapp S; Schofield CJ; Brennan PE Discovery and Optimization of Small-Molecule Ligands for the CBP/P300 Bromodomains. J Am Chem Soc 2014, 136 (26), 9308–9319. 10.1021/ja412434f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (806).Hay D; Fedorov O; Filippakopoulos P; Martin S; Philpott M; Picaud S; Hewings DS; Uttakar S; Heightman TD; Conway SJ; Knapp S; Brennan PE The Design and Synthesis of 5- and 6-Isoxazolylbenzimidazoles as Selective Inhibitors of the BET Bromodomains. Medchemcomm 2013, 4 (1), 140–144. 10.1039/C2MD20189E. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (807).Hay DA; Fedorov O; Martin S; Singleton DC; Tallant C; Wells C; Picaud S; Philpott M; Monteiro OP; Rogers CM; Conway SJ; Rooney TPC; Tumber A; Yapp C; Filippakopoulos P; Bunnage ME; Müller S; Knapp S; Schofield CJ; Brennan PE Discovery and Optimization of Small-Molecule Ligands for the CBP/P300 Bromodomains. J Am Chem Soc 2014, 136 (26), 9308–9319. 10.1021/ja412434f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (808).Picaud S; Fedorov O; Thanasopoulou A; Leonards K; Jones K; Meier J; Olzscha H; Monteiro O; Martin S; Philpott M; Tumber A; Filippakopoulos P; Yapp C; Wells C; Che KH; Bannister A; Robson S; Kumar U; Parr N; Lee K; Lugo D; Jeffrey P; Taylor S; Vecellio ML; Bountra C; Brennan PE; O’Mahony A; Velichko S; Müller S; Hay D; Daniels DL; Urh M; La Thangue NB; Kouzarides T; Prinjha R; Schwaller J; Knapp S Generation of a Selective Small Molecule Inhibitor of the CBP/P300 Bromodomain for Leukemia Therapy. Cancer Res 2015, 75 (23), 5106–5119. 10.1158/0008-5472.CAN-15-0236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (809).Denny RA; Flick AC; Coe J; Langille J; Basak A; Liu S; Stock I; Sahasrabudhe P; Bonin P; Hay DA; Brennan PE; Pletcher M; Jones LH; Chekler ELP Structure-Based Design of Highly Selective Inhibitors of the CREB Binding Protein Bromodomain. J Med Chem 2017, 60 (13), 5349–5363. 10.1021/acs.jmedchem.6b01839. [DOI] [PubMed] [Google Scholar]
- (810).Welti J; Sharp A; Brooks N; Yuan W; McNair C; Chand SN; Pal A; Figueiredo I; Riisnaes R; Gurel B; Rekowski J; Bogdan D; West W; Young B; Raja M; Prosser A; Lane J; Thomson S; Worthington J; Onions S; Shannon J; Paoletta S; Brown R; Smyth D; Harbottle GW; Gil VS; Miranda S; Crespo M; Ferreira A; Pereira R; Tunariu N; Carreira S; Neeb AJ; Ning J; Swain A; Taddei D; Schiewer MJ; Knudsen KE; Pegg N; de Bono JS Targeting the P300/CBP Axis in Lethal Prostate Cancer. Cancer Discov 2021, 11 (5), 1118–1137. 10.1158/2159-8290.CD-20-0751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (811).Jin L; Garcia J; Chan E; de la Cruz C; Segal E; Merchant M; Kharbanda S; Raisner R; Haverty PM; Modrusan Z; Ly J; Choo E; Kaufman S; Beresini MH; Romero FA; Magnuson S; Gascoigne KE Therapeutic Targeting of the CBP/P300 Bromodomain Blocks the Growth of Castration-Resistant Prostate Cancer. Cancer Res 2017, 77 (20), 5564–5575. 10.1158/0008-5472.CAN-17-0314. [DOI] [PubMed] [Google Scholar]
- (812).Lasko LM; Jakob CG; Edalji RP; Qiu W; Montgomery D; Digiammarino EL; Hansen TM; Risi RM; Frey R; Manaves V; Shaw B; Algire M; Hessler P; Lam LT; Uziel T; Faivre E; Ferguson D; Buchanan FG; Martin RL; Torrent M; Chiang GG; Karukurichi K; Langston JW; Weinert BT; Choudhary C; de Vries P; Van Drie JH; McElligott D; Kesicki E; Marmorstein R; Sun C; Cole PA; Rosenberg SH; Michaelides MR; Lai A; Bromberg KD Discovery of a Selective Catalytic P300/CBP Inhibitor That Targets Lineage-Specific Tumours. Nature 2017, 550 (7674), 128–132. 10.1038/nature24028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (813).Crawford TD; Romero FA; Lai KW; Tsui V; Taylor AM; de Leon Boenig G; Noland CL; Murray J; Ly J; Choo EF; Hunsaker TL; Chan EW; Merchant M; Kharbanda S; Gascoigne KE; Kaufman S; Beresini MH; Liao J; Liu W; Chen KX; Chen Z; Conery AR; Côté A; Jayaram H; Jiang Y; Kiefer JR; Kleinheinz T; Li Y; Maher J; Pardo E; Poy F; Spillane KL; Wang F; Wang J; Wei X; Xu Z; Xu Z; Yen I; Zawadzke L; Zhu X; Bellon S; Cummings R; Cochran AG; Albrecht BK; Magnuson S Discovery of a Potent and Selective in Vivo Probe (GNE-272) for the Bromodomains of CBP/EP300. J Med Chem 2016, 59 (23), 10549–10563. 10.1021/acs.jmedchem.6b01022. [DOI] [PubMed] [Google Scholar]
- (814).Lai KW; Romero FA; Tsui V; Beresini MH; de Leon Boenig G; Bronner SM; Chen K; Chen Z; Choo EF; Crawford TD; Cyr P; Kaufman S; Li Y; Liao J; Liu W; Ly J; Murray J; Shen W; Wai J; Wang F; Zhu C; Zhu X; Magnuson S Design and Synthesis of a Biaryl Series as Inhibitors for the Bromodomains of CBP/P300. Bioorg Med Chem Lett 2018, 28 (1), 15–23. 10.1016/j.bmcl.2017.11.025. [DOI] [PubMed] [Google Scholar]
- (815).Romero FA; Murray J; Lai KW; Tsui V; Albrecht BK; An L; Beresini MH; de Leon Boenig G; Bronner SM; Chan EW; Chen KX; Chen Z; Choo EF; Clagg K; Clark K; Crawford TD; Cyr P; de Almeida Nagata D; Gascoigne KE; Grogan JL; Hatzivassiliou G; Huang W; Hunsaker TL; Kaufman S; Koenig SG; Li R; Li Y; Liang X; Liao J; Liu W; Ly J; Maher J; Masui C; Merchant M; Ran Y; Taylor AM; Wai J; Wang F; Wei X; Yu D; Zhu B-Y; Zhu X; Magnuson S GNE-781, A Highly Advanced Potent and Selective Bromodomain Inhibitor of Cyclic Adenosine Monophosphate Response Element Binding Protein, Binding Protein (CBP). J Med Chem 2017, 60 (22), 9162–9183. 10.1021/acs.jmedchem.7b00796. [DOI] [PubMed] [Google Scholar]
- (816).Lai KW; Romero FA; Tsui V; Beresini MH; de Leon Boenig G; Bronner SM; Chen K; Chen Z; Choo EF; Crawford TD; Cyr P; Kaufman S; Li Y; Liao J; Liu W; Ly J; Murray J; Shen W; Wai J; Wang F; Zhu C; Zhu X; Magnuson S Design and Synthesis of a Biaryl Series as Inhibitors for the Bromodomains of CBP/P300. Bioorg Med Chem Lett 2018, 28 (1), 15–23. 10.1016/j.bmcl.2017.11.025. [DOI] [PubMed] [Google Scholar]
- (817).Romero FA; Murray J; Lai KW; Tsui V; Albrecht BK; An L; Beresini MH; de Leon Boenig G; Bronner SM; Chan EW; Chen KX; Chen Z; Choo EF; Clagg K; Clark K; Crawford TD; Cyr P; de Almeida Nagata D; Gascoigne KE; Grogan JL; Hatzivassiliou G; Huang W; Hunsaker TL; Kaufman S; Koenig SG; Li R; Li Y; Liang X; Liao J; Liu W; Ly J; Maher J; Masui C; Merchant M; Ran Y; Taylor AM; Wai J; Wang F; Wei X; Yu D; Zhu B-Y; Zhu X; Magnuson S GNE-781, A Highly Advanced Potent and Selective Bromodomain Inhibitor of Cyclic Adenosine Monophosphate Response Element Binding Protein, Binding Protein (CBP). J Med Chem 2017, 60 (22), 9162–9183. 10.1021/acs.jmedchem.7b00796. [DOI] [PubMed] [Google Scholar]
- (818).Taylor AM; Côté A; Hewitt MC; Pastor R; Leblanc Y; Nasveschuk CG; Romero FA; Crawford TD; Cantone N; Jayaram H; Setser J; Murray J; Beresini MH; de Leon Boenig G; Chen Z; Conery AR; Cummings RT; Dakin LA; Flynn EM; Huang OW; Kaufman S; Keller PJ; Kiefer JR; Lai T; Li Y; Liao J; Liu W; Lu H; Pardo E; Tsui V; Wang J; Wang Y; Xu Z; Yan F; Yu D; Zawadzke L; Zhu X; Zhu X; Sims RJ; Cochran AG; Bellon S; Audia JE; Magnuson S; Albrecht BK Fragment-Based Discovery of a Selective and Cell-Active Benzodiazepinone CBP/EP300 Bromodomain Inhibitor (CPI-637). ACS Med Chem Lett 2016, 7 (5), 531–536. 10.1021/acsmedchemlett.6b00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (819).Michaelides MR; Kluge A; Patane M; Van Drie JH; Wang C; Hansen TM; Risi RM; Mantei R; Hertel C; Karukurichi K; Nesterov A; McElligott D; de Vries P; Langston JW; Cole PA; Marmorstein R; Liu H; Lasko L; Bromberg KD; Lai A; Kesicki EA Discovery of Spiro Oxazolidinediones as Selective, Orally Bioavailable Inhibitors of P300/CBP Histone Acetyltransferases. ACS Med Chem Lett 2018, 9 (1), 28–33. 10.1021/acsmedchemlett.7b00395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (820).Yang Y; Zhang R; Li Z; Mei L; Wan S; Ding H; Chen Z; Xing J; Feng H; Han J; Jiang H; Zheng M; Luo C; Zhou B Discovery of Highly Potent, Selective, and Orally Efficacious P300/CBP Histone Acetyltransferases Inhibitors. J Med Chem 2020, 63 (3), 1337–1360. 10.1021/acs.jmedchem.9b01721. [DOI] [PubMed] [Google Scholar]
- (821).Ji Z; Clark RF; Bhat V; Matthew Hansen T; Lasko LM; Bromberg KD; Manaves V; Algire M; Martin R; Qiu W; Torrent M; Jakob CG; Liu H; Cole PA; Marmorstein R; Kesicki EA; Lai A; Michaelides MR Discovery of Spirohydantoins as Selective, Orally Bioavailable Inhibitors of P300/CBP Histone Acetyltransferases. Bioorg Med Chem Lett 2021, 39, 127854. 10.1016/j.bmcl.2021.127854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (822).Mastracchio A; Lai C; Digiammarino E; Ready DB; Lasko LM; Bromberg KD; McClellan WJ; Montgomery D; Manaves V; Shaw B; Algire M; Patterson MJ; Sun CC; Rosenberg S; Lai A; Michaelides MR Discovery of a Potent and Selective Covalent P300/CBP Inhibitor. ACS Med Chem Lett 2021, 12 (5), 726–731. 10.1021/acsmedchemlett.0c00654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (823).Yang Y; Zhang R; Li Z; Mei L; Wan S; Ding H; Chen Z; Xing J; Feng H; Han J; Jiang H; Zheng M; Luo C; Zhou B Discovery of Highly Potent, Selective, and Orally Efficacious P300/CBP Histone Acetyltransferases Inhibitors. J Med Chem 2020, 63 (3), 1337–1360. 10.1021/acs.jmedchem.9b01721. [DOI] [PubMed] [Google Scholar]
- (824).Mastracchio A; Lai C; Digiammarino E; Ready DB; Lasko LM; Bromberg KD; McClellan WJ; Montgomery D; Manaves V; Shaw B; Algire M; Patterson MJ; Sun CC; Rosenberg S; Lai A; Michaelides MR Discovery of a Potent and Selective Covalent P300/CBP Inhibitor. ACS Med Chem Lett 2021, 12 (5), 726–731. 10.1021/acsmedchemlett.0c00654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (825).Wilson JE; Patel G; Patel C; Brucelle F; Huhn A; Gardberg AS; Poy F; Cantone N; Bommi-Reddy A; Sims RJ; Cummings RT; Levell JR Discovery of CPI-1612: A Potent, Selective, and Orally Bioavailable EP300/CBP Histone Acetyltransferase Inhibitor. ACS Med Chem Lett 2020, 11 (6), 1324–1329. 10.1021/acsmedchemlett.0c00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (826).Huhn AJ; Gardberg AS; Poy F; Brucelle F; Vivat V; Cantone N; Patel G; Patel C; Cummings R; Sims R; Levell J; Audia JE; Bommi-Reddy A; Wilson JE Early Drug-Discovery Efforts towards the Identification of EP300/CBP Histone Acetyltransferase (HAT) Inhibitors. ChemMedChem 2020, 15 (11), 955–960. 10.1002/cmdc.202000007. [DOI] [PubMed] [Google Scholar]
- (827).Vannam R; Sayilgan J; Ojeda S; Karakyriakou B; Hu E; Kreuzer J; Morris R; Herrera Lopez XI; Rai S; Haas W; Lawrence M; Ott CJ Targeted Degradation of the Enhancer Lysine Acetyltransferases CBP and P300. Cell Chem Biol 2021, 28 (4), 503–514.e12. 10.1016/j.chembiol.2020.12.004. [DOI] [PubMed] [Google Scholar]
- (828).Thomas JE; Wang M; Jiang W; Wang M; Wang L; Wen B; Sun D; Wang S Discovery of Exceptionally Potent, Selective, and Efficacious PROTAC Degraders of CBP and P300 Proteins. J Med Chem 2023, 66 (12), 8178–8199. 10.1021/acs.jmedchem.3c00492. [DOI] [PubMed] [Google Scholar]
- (829).Chen Z; Wang M; Wu D; Zhao L; Metwally H; Jiang W; Wang Y; Bai L; McEachern D; Luo J; Wang M; Li Q; Matvekas A; Wen B; Sun D; Chinnaiyan AM; Wang S Discovery of CBPD-409 as a Highly Potent, Selective, and Orally Efficacious CBP/P300 PROTAC Degrader for the Treatment of Advanced Prostate Cancer. J Med Chem 2024, 67 (7), 5351–5372. 10.1021/acs.jmedchem.3c01789. [DOI] [PubMed] [Google Scholar]
- (830).Chen Z; Wang M; Wu D; Bai L; Xu T; Metwally H; Wang Y; McEachern D; Zhao L; Li R; Takyi-Williams J; Wang M; Wang L; Li Q; Wen B; Sun D; Wang S Discovery of CBPD-268 as an Exceptionally Potent and Orally Efficacious CBP/P300 PROTAC Degrader Capable of Achieving Tumor Regression. J Med Chem 2024, 67 (7), 5275–5304. 10.1021/acs.jmedchem.3c02124. [DOI] [PubMed] [Google Scholar]
- (831).Chen Z; Hu B; Rej RK; Wu D; Acharyya RK; Wang M; Xu T; Lu J; Metwally H; Wang Y; McEachern D; Bai L; Gersch CL; Wang M; Zhang W; Li Q; Wen B; Sun D; Rae JM; Wang S Discovery of ERD-3111 as a Potent and Orally Efficacious Estrogen Receptor PROTAC Degrader with Strong Antitumor Activity. J Med Chem 2023, 66 (17), 12559–12585. 10.1021/acs.jmedchem.3c01186. [DOI] [PubMed] [Google Scholar]
- (832).Xiang W; Zhao L; Han X; Xu T; Kregel S; Wang M; Miao B; Qin C; Wang M; McEachern D; Lu J; Bai L; Yang C-Y; Kirchhoff PD; Takyi-Williams J; Wang L; Wen B; Sun D; Ator M; Mckean R; Chinnaiyan AM; Wang S Discovery of ARD-1676 as a Highly Potent and Orally Efficacious AR PROTAC Degrader with a Broad Activity against AR Mutants for the Treatment of AR + Human Prostate Cancer. J Med Chem 2023, 66 (18), 13280–13303. 10.1021/acs.jmedchem.3c01264. [DOI] [PubMed] [Google Scholar]
- (833).Chen Z; Wang M; Wu D; Bai L; Xu T; Metwally H; Wang Y; McEachern D; Zhao L; Li R; Takyi-Williams J; Wang M; Wang L; Li Q; Wen B; Sun D; Wang S Discovery of CBPD-268 as an Exceptionally Potent and Orally Efficacious CBP/P300 PROTAC Degrader Capable of Achieving Tumor Regression. J Med Chem 2024, 67 (7), 5275–5304. 10.1021/acs.jmedchem.3c02124. [DOI] [PubMed] [Google Scholar]
- (834).Chang Q; Li J; Deng Y; Zhou R; Wang B; Wang Y; Zhang M; Huang X; Li Y Discovery of Novel PROTAC Degraders of P300/CBP as Potential Therapeutics for Hepatocellular Carcinoma. J Med Chem 2024, 67 (4), 2466–2486. 10.1021/acs.jmedchem.3c01468. [DOI] [PubMed] [Google Scholar]
- (835).Kumar Tiwari P; Reddy Doda S; Vannam R; Hudlikar M; Harrison DA; Ojeda S; Rai S; Koglin A-S; Nguyen Gilbert A; Ott CJ Exploration of Bromodomain Ligand-Linker Conjugation Sites for Efficient CBP/P300 Heterobifunctional Degrader Activity. Bioorg Med Chem Lett 2024, 102, 129676. 10.1016/j.bmcl.2024.129676. [DOI] [PubMed] [Google Scholar]
- (836).Durbin AD; Wang T; Wimalasena VK; Zimmerman MW; Li D; Dharia NV; Mariani L; Shendy NAM; Nance S; Patel AG; Shao Y; Mundada M; Maxham L; Park PMC; Sigua LH; Morita K; Conway AS; Robichaud AL; Perez-Atayde AR; Bikowitz MJ; Quinn TR; Wiest O; Easton J; Schönbrunn E; Bulyk ML; Abraham BJ; Stegmaier K; Look AT; Qi J EP300 Selectively Controls the Enhancer Landscape of MYCN-Amplified Neuroblastoma. Cancer Discov 2022, 12 (3), 730–751. 10.1158/2159-8290.CD-21-0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (837).Cheng-Sánchez I; Gosselé KA; Palaferri L; Kirillova MS; Nevado C Discovery and Characterization of Active CBP/EP300 Degraders Targeting the HAT Domain. ACS Med Chem Lett 2024, 15 (3), 355–361. 10.1021/acsmedchemlett.3c00490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (838).Chen X; Crawford MC; Xiong Y; Shaik AB; Suazo KF; Bauer LG; Penikalapati MS; Williams JH; Huber KVM; Andressen T; Swenson RE; Meier JL Paralogue-Selective Degradation of the Lysine Acetyltransferase EP300. JACS Au 2024, 4 (8), 3094–3103. 10.1021/jacsau.4c00442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (839).Lasko LM; Jakob CG; Edalji RP; Qiu W; Montgomery D; Digiammarino EL; Hansen TM; Risi RM; Frey R; Manaves V; Shaw B; Algire M; Hessler P; Lam LT; Uziel T; Faivre E; Ferguson D; Buchanan FG; Martin RL; Torrent M; Chiang GG; Karukurichi K; Langston JW; Weinert BT; Choudhary C; de Vries P; Kluge AF; Patane MA; Van Drie JH; Wang C; McElligott D; Kesicki E; Marmorstein R; Sun C; Cole PA; Rosenberg SH; Michaelides MR; Lai A; Bromberg KD Discovery of a Selective Catalytic P300/CBP Inhibitor That Targets Lineage-Specific Tumours. Nature 2017, 550 (7674), 128–132. 10.1038/nature24028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (840).Hogg SJ; Motorna O; Cluse LA; Johanson TM; Coughlan HD; Raviram R; Myers RM; Costacurta M; Todorovski I; Pijpers L; Bjelosevic S; Williams T; Huskins SN; Kearney CJ; Devlin JR; Fan Z; Jabbari JS; Martin BP; Fareh M; Kelly MJ; Dupéré-Richer D; Sandow JJ; Feran B; Knight D; Khong T; Spencer A; Harrison SJ; Gregory G; Wickramasinghe VO; Webb AI; Taberlay PC; Bromberg KD; Lai A; Papenfuss AT; Smyth GK; Allan RS; Licht JD; Landau DA; Abdel-Wahab O; Shortt J; Vervoort SJ; Johnstone RW Targeting Histone Acetylation Dynamics and Oncogenic Transcription by Catalytic P300/CBP Inhibition. Mol Cell 2021, 81 (10), 2183–2200.e13. 10.1016/j.molcel.2021.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (841).Bishop TR; Subramanian C; Bilotta EM; Garnar-Wortzel L; Ramos AR; Zhang Y; Asiaban JN; Ott CJ; Rock CO; Erb MA Acetyl-CoA Biosynthesis Drives Resistance to Histone Acetyltransferase Inhibition. Nat Chem Biol 2023. 10.1038/s41589-023-01320-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (842).Vannam R; Sayilgan J; Ojeda S; Karakyriakou B; Hu E; Kreuzer J; Morris R; Herrera Lopez XI; Rai S; Haas W; Lawrence M; Ott CJ Targeted Degradation of the Enhancer Lysine Acetyltransferases CBP and P300. Cell Chem Biol 2021, 28 (4), 503–514.e12. 10.1016/j.chembiol.2020.12.004. [DOI] [PubMed] [Google Scholar]
- (843).Raisner R; Kharbanda S; Jin L; Jeng E; Chan E; Merchant M; Haverty PM; Bainer R; Cheung T; Arnott D; Flynn EM; Romero FA; Magnuson S; Gascoigne KE Enhancer Activity Requires CBP/P300 Bromodomain-Dependent Histone H3K27 Acetylation. Cell Rep 2018, 24 (7), 1722–1729. 10.1016/j.celrep.2018.07.041. [DOI] [PubMed] [Google Scholar]
- (844).Park S; Stanfield RL; Martinez-Yamout MA; Dyson HJ; Wilson IA; Wright PE Role of the CBP Catalytic Core in Intramolecular SUMOylation and Control of Histone H3 Acetylation. Proc Natl Acad Sci U S A 2017, 114 (27), E5335–E5342. 10.1073/pnas.1703105114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (845).Welti J; Sharp A; Brooks N; Yuan W; McNair C; Chand SN; Pal A; Figueiredo I; Riisnaes R; Gurel B; Rekowski J; Bogdan D; West W; Young B; Raja M; Prosser A; Lane J; Thomson S; Worthington J; Onions S; Shannon J; Paoletta S; Brown R; Smyth D; Harbottle GW; Gil VS; Miranda S; Crespo M; Ferreira A; Pereira R; Tunariu N; Carreira S; Neeb AJ; Ning J; Swain A; Taddei D; SU2C/PCF International Prostate Cancer Dream Team; Schiewer MJ; Knudsen KE; Pegg N; de Bono JS Targeting the P300/CBP Axis in Lethal Prostate Cancer. Cancer Discov 2021, 11 (5), 1118–1137. 10.1158/2159-8290.CD-20-0751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (846).Katavolos P; Cain G; Farman C; Romero FA; Magnuson S; Ly JQ; Choo EF; Katakam AK; Andaya R; Maher J Preclinical Safety Assessment of a Highly Selective and Potent Dual Small-Molecule Inhibitor of CBP/P300 in Rats and Dogs. Toxicol Pathol 2020, 48 (3), 465–480. 10.1177/0192623319898469. [DOI] [PubMed] [Google Scholar]
- (847).Nicosia L; Spencer GJ; Brooks N; Amaral FMR; Basma NJ; Chadwick JA; Revell B; Wingelhofer B; Maiques-Diaz A; Sinclair O; Camera F; Ciceri F; Wiseman DH; Pegg N; West W; Knurowski T; Frese K; Clegg K; Campbell VL; Cavet J; Copland M; Searle E; Somervaille TCP Therapeutic Targeting of EP300/CBP by Bromodomain Inhibition in Hematologic Malignancies. Cancer Cell 2023, 41 (12), 2136–2153.e13. 10.1016/j.ccell.2023.11.001. [DOI] [PubMed] [Google Scholar]
- (848).Crawford MC; Tripu DR; Barritt SA; Jing Y; Gallimore D; Kales SC; Bhanu NV; Xiong Y; Fang Y; Butler KAT; LeClair CA; Coussens NP; Simeonov A; Garcia BA; Dibble CC; Meier JL Comparative Analysis of Drug-like EP300/CREBBP Acetyltransferase Inhibitors. ACS Chem Biol 2023, 18 (10), 2249–2258. 10.1021/acschembio.3c00293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (849).Wang WW; Chen L-Y; Wozniak JM; Jadhav AM; Anderson H; Malone TE; Parker CG Targeted Protein Acetylation in Cells Using Heterobifunctional Molecules. J Am Chem Soc 2021, 143 (40), 16700–16708. 10.1021/jacs.1c07850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (850).Kabir M; Sun N; Hu X; Martin TC; Yi J; Zhong Y; Xiong Y; Kaniskan HÜ; Gu W; Parsons R; Jin J Acetylation Targeting Chimera Enables Acetylation of the Tumor Suppressor P53. J Am Chem Soc 2023, 145 (27), 14932–14944. 10.1021/jacs.3c04640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (851).Chen L-Y; Singha Roy SJ; Jadhav AM; Wang WW; Chen P-H; Bishop T; Erb MA; Parker CG Functional Investigations of P53 Acetylation Enabled by Heterobifunctional Molecules. ACS Chem Biol 2024, 19 (9), 1918–1929. 10.1021/acschembio.4c00438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (852).Malone CF; Mabe NW; Forman AB; Alexe G; Engel KL; Chen Y-JC; Soeung M; Salhotra S; Basanthakumar A; Liu B; Dent SYR; Stegmaier K The KAT Module of the SAGA Complex Maintains the Oncogenic Gene Expression Program in MYCN-Amplified Neuroblastoma. Sci Adv 2024, 10 (22), eadm9449. 10.1126/sciadv.adm9449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (853).Chan AKN; Han L; Delaney CD; Wang X; Mukhaleva E; Li M; Yang L; Pokharel SP; Mattson N; Garcia M; Wang B; Xu X; Zhang L; Singh P; Elsayed Z; Chen R; Kuang B; Wang J; Yuan Y-C; Chen B; Chan LN; Rosen ST; Horne D; Müschen M; Chen J; Vaidehi N; Armstrong SA; Su R; Chen C-W Therapeutic Targeting Tudor Domains in Leukemia via CRISPR-Scan Assisted Drug Discovery. Sci Adv 2024, 10 (8), eadk3127. 10.1126/sciadv.adk3127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (854).Barbosa K; Deshpande A; Perales M; Xiang P; Murad R; Pramod AB; Minkina A; Robertson N; Schischlik F; Lei X; Sun Y; Brown A; Amend D; Jeremias I; Doench JG; Humphries RK; Ruppin E; Shendure J; Mali P; Adams PD; Deshpande AJ Transcriptional Control of Leukemogenesis by the Chromatin Reader SGF29. Blood 2024, 143 (8), 697–712. 10.1182/blood.2023021234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (855).Zeng L; Li J; Muller M; Yan S; Mujtaba S; Pan C; Wang Z; Zhou M-M Selective Small Molecules Blocking HIV-1 Tat and Coactivator PCAF Association. J Am Chem Soc 2005, 127 (8), 2376–2377. 10.1021/ja044885g. [DOI] [PubMed] [Google Scholar]
- (856).Navratilova I; Aristotelous T; Picaud S; Chaikuad A; Knapp S; Filappakopoulos P; Hopkins AL Discovery of New Bromodomain Scaffolds by Biosensor Fragment Screening. ACS Med Chem Lett 2016, 7 (12), 1213–1218. 10.1021/acsmedchemlett.6b00154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (857).Chaikuad A; Lang S; Brennan PE; Temperini C; Fedorov O; Hollander J; Nachane R; Abell C; Müller S; Siegal G; Knapp S Structure-Based Identification of Inhibitory Fragments Targeting the P300/CBP-Associated Factor Bromodomain. J Med Chem 2016, 59 (4), 1648–1653. 10.1021/acs.jmedchem.5b01719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (858).Hu P; Wang X; Zhang B; Zhang S; Wang Q; Wang Z Fluorescence Polarization for the Evaluation of Small-Molecule Inhibitors of PCAF BRD/Tat-AcK50 Association. ChemMedChem 2014, 9 (5), 928–931. 10.1002/cmdc.201300499. [DOI] [PubMed] [Google Scholar]
- (859).Chimenti F; Bizzarri B; Maccioni E; Secci D; Bolasco A; Chimenti P; Fioravanti R; Granese A; Carradori S; Tosi F; Ballario P; Vernarecci S; Filetici P A Novel Histone Acetyltransferase Inhibitor Modulating Gcn5 Network: Cyclopentylidene-[4-(4’-Chlorophenyl)Thiazol-2-Yl)Hydrazone. J Med Chem 2009, 52 (2), 530–536. 10.1021/jm800885d. [DOI] [PubMed] [Google Scholar]
- (860).Humphreys PG; Bamborough P; Chung C-W; Craggs PD; Gordon L; Grandi P; Hayhow TG; Hussain J; Jones KL; Lindon M; Michon A-M; Renaux JF; Suckling CJ; Tough DF; Prinjha RK Discovery of a Potent, Cell Penetrant, and Selective P300/CBP-Associated Factor (PCAF)/General Control Nonderepressible 5 (GCN5) Bromodomain Chemical Probe. J Med Chem 2017, 60 (2), 695–709. 10.1021/acs.jmedchem.6b01566. [DOI] [PubMed] [Google Scholar]
- (861).Moustakim M; Clark PGK; Trulli L; Fuentes de Arriba AL; Ehebauer MT; Chaikuad A; Murphy EJ; Mendez-Johnson J; Daniels D; Hou C-FD; Lin Y-H; Walker JR; Hui R; Yang H; Dorrell L; Rogers CM; Monteiro OP; Fedorov O; Huber KVM; Knapp S; Heer J; Dixon DJ; Brennan PE Discovery of a PCAF Bromodomain Chemical Probe. Angew Chem Int Ed Engl 2017, 56 (3), 827–831. 10.1002/anie.201610816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (862).Tao H; Wang J; Lu W; Zhang R; Xie Y; Liu Y-C; Liu R; Yue L; Chen K; Jiang H; Zhang Y; Xu X; Luo C Discovery of Trisubstituted Nicotinonitrile Derivatives as Novel Human GCN5 Inhibitors through AlphaScreen-Based High Throughput Screening. RSC Adv 2019, 9 (9), 4917–4924. 10.1039/c8ra10074h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (863).Xiong H; Han J; Wang J; Lu W; Wang C; Chen Y; Fulin Lian; Zhang N; Liu Y-C; Zhang C; Ding H; Jiang H; Lu W; Luo C; Zhou B Discovery of 1,8-Acridinedione Derivatives as Novel GCN5 Inhibitors via High Throughput Screening. Eur J Med Chem 2018, 151, 740–751. 10.1016/j.ejmech.2018.02.005. [DOI] [PubMed] [Google Scholar]
- (864).Huang L; Li H; Li L; Niu L; Seupel R; Wu C; Cheng W; Chen C; Ding B; Brennan PE; Yang S Discovery of Pyrrolo[3,2- d]Pyrimidin-4-One Derivatives as a New Class of Potent and Cell-Active Inhibitors of P300/CBP-Associated Factor Bromodomain. J Med Chem 2019, 62 (9), 4526–4542. 10.1021/acs.jmedchem.9b00096. [DOI] [PubMed] [Google Scholar]
- (865).de Jong RCM; Ewing MM; de Vries MR; Karper JC; Bastiaansen AJNM; Peters HAB; Baghana F; van den Elsen PJ; Gongora C; Jukema JW; Quax PHA The Epigenetic Factor PCAF Regulates Vascular Inflammation and Is Essential for Intimal Hyperplasia Development. PLoS One 2017, 12 (10), e0185820. 10.1371/journal.pone.0185820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (866).Bastiaansen AJNM; Ewing MM; de Boer HC; van der Pouw Kraan TC; de Vries MR; Peters EAB; Welten SMJ; Arens R; Moore SM; Faber JE; Jukema JW; Hamming JF; Nossent AY; Quax PHA Lysine Acetyltransferase PCAF Is a Key Regulator of Arteriogenesis. Arterioscler Thromb Vasc Biol 2013, 33 (8), 1902–1910. 10.1161/ATVBAHA.113.301579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (867).Bassi ZI; Fillmore MC; Miah AH; Chapman TD; Maller C; Roberts EJ; Davis LC; Lewis DE; Galwey NW; Waddington KE; Parravicini V; Macmillan-Jones AL; Gongora C; Humphreys PG; Churcher I; Prinjha RK; Tough DF Modulating PCAF/GCN5 Immune Cell Function through a PROTAC Approach. ACS Chem Biol 2018, 13 (10), 2862–2867. 10.1021/acschembio.8b00705. [DOI] [PubMed] [Google Scholar]
- (868).Mukohara T; Park YH; Sommerhalder D; Yonemori K; Hamilton E; Kim S-B; Kim JH; Iwata H; Yamashita T; Layman RM; Mita M; Clay T; Chae YS; Oakman C; Yan F; Kim GM; Im S-A; Lindeman GJ; Rugo HS; Liyanage M; Saul M; Le Corre C; Skoura A; Liu L; Li M; LoRusso PM Inhibition of Lysine Acetyltransferase KAT6 in ER+HER2- Metastatic Breast Cancer: A Phase 1 Trial. Nat Med 2024, 30 (8), 2242–2250. 10.1038/s41591-024-03060-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (869).Klein BJ; Jang SM; Lachance C; Mi W; Lyu J; Sakuraba S; Krajewski K; Wang WW; Sidoli S; Liu J; Zhang Y; Wang X; Warfield BM; Kueh AJ; Voss AK; Thomas T; Garcia BA; Liu WR; Strahl BD; Kono H; Li W; Shi X; Côté J; Kutateladze TG Histone H3K23-Specific Acetylation by MORF Is Coupled to H3K14 Acylation. Nat Commun 2019, 10 (1), 4724. 10.1038/s41467-019-12551-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (870).Sharma S; Chung C-Y; Uryu S; Petrovic J; Cao J; Rickard A; Nady N; Greasley S; Johnson E; Brodsky O; Khan S; Wang H; Wang Z; Zhang Y; Tsaparikos K; Chen L; Mazurek A; Lapek J; Kung P-P; Sutton S; Richardson PF; Greenwald EC; Yamazaki S; Jones R; Maegley KA; Bingham P; Lam H; Stupple AE; Kamal A; Chueh A; Cuzzupe A; Morrow BJ; Ren B; Carrasco-Pozo C; Tan CW; Bhuva DD; Allan E; Surgenor E; Vaillant F; Pehlivanoglu H; Falk H; Whittle JR; Newman J; Cursons J; Doherty JP; White KL; MacPherson L; Devlin M; Dennis ML; Hattarki MK; De Silva M; Camerino MA; Butler MS; Dolezal O; Pilling P; Foitzik R; Stupple PA; Lagiakos HR; Walker SR; Hediyeh-Zadeh S; Nuttall S; Spall SK; Charman SA; Connor T; Peat TS; Avery VM; Bozikis YE; Yang Y; Zhang M; Monahan BJ; Voss AK; Thomas T; Street IP; Dawson S-J; Dawson MA; Lindeman GJ; Davis MJ; Visvader JE; Paul TA Discovery of a Highly Potent, Selective, Orally Bioavailable Inhibitor of KAT6A/B Histone Acetyltransferases with Efficacy against KAT6A-High ER+ Breast Cancer. Cell Chem Biol 2023, 30 (10), 1191–1210.e20. 10.1016/j.chembiol.2023.07.005. [DOI] [PubMed] [Google Scholar]
- (871).Lv D; Jia F; Hou Y; Sang Y; Alvarez AA; Zhang W; Gao W-Q; Hu B; Cheng S-Y; Ge J; Li Y; Feng H Histone Acetyltransferase KAT6A Upregulates PI3K/AKT Signaling through TRIM24 Binding. Cancer Res 2017, 77 (22), 6190–6201. 10.1158/0008-5472.CAN-17-1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (872).Simó-Riudalbas L; Pérez-Salvia M; Setien F; Villanueva A; Moutinho C; Martínez-Cardús A; Moran S; Berdasco M; Gomez A; Vidal E; Soler M; Heyn H; Vaquero A; de la Torre C; Barceló-Batllori S; Vidal A; Roz L; Pastorino U; Szakszon K; Borck G; Moura CS; Carneiro F; Zondervan I; Savola S; Iwakawa R; Kohno T; Yokota J; Esteller M KAT6B Is a Tumor Suppressor Histone H3 Lysine 23 Acetyltransferase Undergoing Genomic Loss in Small Cell Lung Cancer. Cancer Res 2015, 75 (18), 3936–3945. 10.1158/0008-5472.CAN-14-3702. [DOI] [PubMed] [Google Scholar]
- (873).Voss AK; Collin C; Dixon MP; Thomas T Moz and Retinoic Acid Coordinately Regulate H3K9 Acetylation, Hox Gene Expression, and Segment Identity. Dev Cell 2009, 17 (5), 674–686. 10.1016/j.devcel.2009.10.006. [DOI] [PubMed] [Google Scholar]
- (874).Voss AK; Vanyai HK; Collin C; Dixon MP; McLennan TJ; Sheikh BN; Scambler P; Thomas T MOZ Regulates the Tbx1 Locus, and Moz Mutation Partially Phenocopies DiGeorge Syndrome. Dev Cell 2012, 23 (3), 652–663. 10.1016/j.devcel.2012.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (875).Vanyai HK; Garnham A; May RE; McRae HM; Collin C; Wilcox S; Smyth GK; Thomas T; Voss AK MOZ Directs the Distal-Less Homeobox Gene Expression Program during Craniofacial Development. Development 2019, 146 (14), dev175042. 10.1242/dev.175042. [DOI] [PubMed] [Google Scholar]
- (876).Miller CT; Maves L; Kimmel CB Moz Regulates Hox Expression and Pharyngeal Segmental Identity in Zebrafish. Development 2004, 131 (10), 2443–2461. 10.1242/dev.01134. [DOI] [PubMed] [Google Scholar]
- (877).Crump JG; Swartz ME; Eberhart JK; Kimmel CB Moz-Dependent Hox Expression Controls Segment-Specific Fate Maps of Skeletal Precursors in the Face. Development 2006, 133 (14), 2661–2669. 10.1242/dev.02435. [DOI] [PubMed] [Google Scholar]
- (878).Sheikh BN; Downer NL; Phipson B; Vanyai HK; Kueh AJ; McCarthy DJ; Smyth GK; Thomas T; Voss AK MOZ and BMI1 Play Opposing Roles during Hox Gene Activation in ES Cells and in Body Segment Identity Specification in Vivo. Proc Natl Acad Sci U S A 2015, 112 (17), 5437–5442. 10.1073/pnas.1422872112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (879).Sheikh BN; Phipson B; El-Saafin F; Vanyai HK; Downer NL; Bird MJ; Kueh AJ; May RE; Smyth GK; Voss AK; Thomas T MOZ (MYST3, KAT6A) Inhibits Senescence via the INK4A-ARF Pathway. Oncogene 2015, 34 (47), 5807–5820. 10.1038/onc.2015.33. [DOI] [PubMed] [Google Scholar]
- (880).Rokudai S; Laptenko O; Arnal SM; Taya Y; Kitabayashi I; Prives C MOZ Increases P53 Acetylation and Premature Senescence through Its Complex Formation with PML. Proc Natl Acad Sci U S A 2013, 110 (10), 3895–3900. 10.1073/pnas.1300490110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (881).Baell JB; Leaver DJ; Hermans SJ; Kelly GL; Brennan MS; Downer NL; Nguyen N; Wichmann J; McRae HM; Yang Y; Cleary B; Lagiakos HR; Mieruszynski S; Pacini G; Vanyai HK; Bergamasco MI; May RE; Davey BK; Morgan KJ; Sealey AJ; Wang B; Zamudio N; Wilcox S; Garnham AL; Sheikh BN; Aubrey BJ; Doggett K; Chung MC; de Silva M; Bentley J; Pilling P; Hattarki M; Dolezal O; Dennis ML; Falk H; Ren B; Charman SA; White KL; Rautela J; Newbold A; Hawkins ED; Johnstone RW; Huntington ND; Peat TS; Heath JK; Strasser A; Parker MW; Smyth GK; Street IP; Monahan BJ; Voss AK; Thomas T Inhibitors of Histone Acetyltransferases KAT6A/B Induce Senescence and Arrest Tumour Growth. Nature 2018, 560 (7717), 253–257. 10.1038/s41586-018-0387-5. [DOI] [PubMed] [Google Scholar]
- (882).Perez-Campo FM; Costa G; Lie-A-Ling M; Stifani S; Kouskoff V; Lacaud G MOZ-Mediated Repression of P16(INK) (4) (a) Is Critical for the Self-Renewal of Neural and Hematopoietic Stem Cells. Stem Cells 2014, 32 (6), 1591–1601. 10.1002/stem.1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (883).Thomas T; Corcoran LM; Gugasyan R; Dixon MP; Brodnicki T; Nutt SL; Metcalf D; Voss AK Monocytic Leukemia Zinc Finger Protein Is Essential for the Development of Long-Term Reconstituting Hematopoietic Stem Cells. Genes Dev 2006, 20 (9), 1175–1186. 10.1101/gad.1382606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (884).Katsumoto T; Aikawa Y; Iwama A; Ueda S; Ichikawa H; Ochiya T; Kitabayashi I MOZ Is Essential for Maintenance of Hematopoietic Stem Cells. Genes Dev 2006, 20 (10), 1321–1330. 10.1101/gad.1393106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (885).Sheikh BN; Yang Y; Schreuder J; Nilsson SK; Bilardi R; Carotta S; McRae HM; Metcalf D; Voss AK; Thomas T MOZ (KAT6A) Is Essential for the Maintenance of Classically Defined Adult Hematopoietic Stem Cells. Blood 2016, 128 (19), 2307–2318. 10.1182/blood-2015-10-676072. [DOI] [PubMed] [Google Scholar]
- (886).Perez-Campo FM; Borrow J; Kouskoff V; Lacaud G The Histone Acetyl Transferase Activity of Monocytic Leukemia Zinc Finger Is Critical for the Proliferation of Hematopoietic Precursors. Blood 2009, 113 (20), 4866–4874. 10.1182/blood-2008-04-152017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (887).Panagopoulos I; Andersen K; Ramslien LF; Ikonomou IM; Micci F; Heim S Therapy-Related Myeloid Leukemia With the Translocation t(8;19)(P11;Q13) Leading to a KAT6A-LEUTX Fusion Gene. Anticancer Res 2021, 41 (4), 1753–1760. 10.21873/anticanres.14940. [DOI] [PubMed] [Google Scholar]
- (888).Chinen Y; Taki T; Tsutsumi Y; Kobayashi S; Matsumoto Y; Sakamoto N; Kuroda J; Horiike S; Nishida K; Ohno H; Uike N; Taniwaki M The Leucine Twenty Homeobox (LEUTX) Gene, Which Lacks a Histone Acetyltransferase Domain, Is Fused to KAT6A in Therapy-Related Acute Myeloid Leukemia with t(8;19)(P11;Q13). Genes Chromosomes Cancer 2014, 53 (4), 299–308. 10.1002/gcc.22140. [DOI] [PubMed] [Google Scholar]
- (889).Agaimy A; Clarke BA; Kolin DL; Lee C-H; Lee J-C; McCluggage WG; Pöschke P; Stoehr R; Swanson D; Turashvili G; Beckmann MW; Hartmann A; Antonescu CR; Dickson BC Recurrent KAT6B/A::KANSL1 Fusions Characterize a Potentially Aggressive Uterine Sarcoma Morphologically Overlapping With Low-Grade Endometrial Stromal Sarcoma. Am J Surg Pathol 2022, 46 (9), 1298–1308. 10.1097/PAS.0000000000001915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (890).Pei J; Cooper H; Flieder DB; Talarchek JN; Al-Saleem T; Uzzo RG; Dulaimi E; Patchefsky AS; Testa JR; Wei S NEAT1-TFE3 and KAT6A-TFE3 Renal Cell Carcinomas, New Members of MiT Family Translocation Renal Cell Carcinoma. Mod Pathol 2019, 32 (5), 710–716. 10.1038/s41379-018-0191-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (891).Esteyries S; Perot C; Adelaide J; Imbert M; Lagarde A; Pautas C; Olschwang S; Birnbaum D; Chaffanet M; Mozziconacci M-J NCOA3, a New Fusion Partner for MOZ/MYST3 in M5 Acute Myeloid Leukemia. Leukemia 2008, 22 (3), 663–665. 10.1038/sj.leu.2404930. [DOI] [PubMed] [Google Scholar]
- (892).Liang J; Prouty L; Williams BJ; Dayton MA; Blanchard KL Acute Mixed Lineage Leukemia with an Inv(8)(P11q13) Resulting in Fusion of the Genes for MOZ and TIF2. Blood 1998, 92 (6), 2118–2122. [PubMed] [Google Scholar]
- (893).Deguchi K; Ayton PM; Carapeti M; Kutok JL; Snyder CS; Williams IR; Cross NCP; Glass CK; Cleary ML; Gilliland DG MOZ-TIF2-Induced Acute Myeloid Leukemia Requires the MOZ Nucleosome Binding Motif and TIF2-Mediated Recruitment of CBP. Cancer Cell 2003, 3 (3), 259–271. 10.1016/s1535-6108(03)00051-5. [DOI] [PubMed] [Google Scholar]
- (894).Carapeti M; Aguiar RC; Goldman JM; Cross NC A Novel Fusion between MOZ and the Nuclear Receptor Coactivator TIF2 in Acute Myeloid Leukemia. Blood 1998, 91 (9), 3127–3133. [PubMed] [Google Scholar]
- (895).Shima H; Yamagata K; Aikawa Y; Shino M; Koseki H; Shimada H; Kitabayashi I Bromodomain-PHD Finger Protein 1 Is Critical for Leukemogenesis Associated with MOZ-TIF2 Fusion. Int J Hematol 2014, 99 (1), 21–31. 10.1007/s12185-013-1466-x. [DOI] [PubMed] [Google Scholar]
- (896).Chaffanet M; Gressin L; Preudhomme C; Soenen-Cornu V; Birnbaum D; Pébusque MJ MOZ Is Fused to P300 in an Acute Monocytic Leukemia with t(8;22). Genes Chromosomes Cancer 2000, 28 (2), 138–144. . [DOI] [PubMed] [Google Scholar]
- (897).Kojima K; Kaneda K; Yoshida C; Dansako H; Fujii N; Yano T; Shinagawa K; Yasukawa M; Fujita S; Tanimoto M A Novel Fusion Variant of the MORF and CBP Genes Detected in Therapy-Related Myelodysplastic Syndrome with t(10;16)(Q22;P13). Br J Haematol 2003, 120 (2), 271–273. 10.1046/j.1365-2141.2003.04059.x. [DOI] [PubMed] [Google Scholar]
- (898).Su J; Wang X; Bai Y; Sun M; Yao Y; Duan Y The Role of MOZ/KAT6A in Hematological Malignancies and Advances in MOZ/KAT6A Inhibitors. Pharmacol Res 2021, 174, 105930. 10.1016/j.phrs.2021.105930. [DOI] [PubMed] [Google Scholar]
- (899).Katsumoto T; Ogawara Y; Yamagata K; Aikawa Y; Goitsuka R; Nakamura T; Kitabayashi I MOZ Is Critical for the Development of MOZ/MLL Fusion-Induced Leukemia through Regulation of Hoxa9/Meis1 Expression. Blood Adv 2022, 6 (19), 5527–5537. 10.1182/bloodadvances.2020003490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (900).Yan F; Li J; Milosevic J; Petroni R; Liu S; Shi Z; Yuan S; Reynaga JM; Qi Y; Rico J; Yu S; Liu Y; Rokudai S; Palmisiano N; Meyer SE; Sung PJ; Wan L; Lan F; Garcia BA; Stanger BZ; Sykes DB; Blanco MA KAT6A and ENL Form an Epigenetic Transcriptional Control Module to Drive Critical Leukemogenic Gene-Expression Programs. Cancer Discov 2022, 12 (3), 792–811. 10.1158/2159-8290.CD-20-1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (901).Sheikh BN; Lee SCW; El-Saafin F; Vanyai HK; Hu Y; Pang SHM; Grabow S; Strasser A; Nutt SL; Alexander WS; Smyth GK; Voss AK; Thomas T MOZ Regulates B-Cell Progenitors and, Consequently, Moz Haploinsufficiency Dramatically Retards MYC-Induced Lymphoma Development. Blood 2015, 125 (12), 1910–1921. 10.1182/blood-2014-08-594655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (902).Falk H; Connor T; Yang H; Loft KJ; Alcindor JL; Nikolakopoulos G; Surjadi RN; Bentley JD; Hattarki MK; Dolezal O; Murphy JM; Monahan BJ; Peat TS; Thomas T; Baell JB; Parisot JP; Street IP An Efficient High-Throughput Screening Method for MYST Family Acetyltransferases, a New Class of Epigenetic Drug Targets. J Biomol Screen 2011, 16 (10), 1196–1205. 10.1177/1087057111421631. [DOI] [PubMed] [Google Scholar]
- (903).Leaver DJ; Cleary B; Nguyen N; Priebbenow DL; Lagiakos HR; Sanchez J; Xue L; Huang F; Sun Y; Mujumdar P; Mudududdla R; Varghese S; Teguh S; Charman SA; White KL; Katneni K; Cuellar M; Strasser JM; Dahlin JL; Walters MA; Street IP; Monahan BJ; Jarman KE; Sabroux HJ; Falk H; Chung MC; Hermans SJ; Parker MW; Thomas T; Baell JB Discovery of Benzoylsulfonohydrazides as Potent Inhibitors of the Histone Acetyltransferase KAT6A. J Med Chem 2019, 62 (15), 7146–7159. 10.1021/acs.jmedchem.9b00665. [DOI] [PubMed] [Google Scholar]
- (904).Priebbenow DL; Leaver DJ; Nguyen N; Cleary B; Lagiakos HR; Sanchez J; Xue L; Huang F; Sun Y; Mujumdar P; Mudududdla R; Varghese S; Teguh S; Charman SA; White KL; Shackleford DM; Katneni K; Cuellar M; Strasser JM; Dahlin JL; Walters MA; Street IP; Monahan BJ; Jarman KE; Jousset Sabroux H; Falk H; Chung MC; Hermans SJ; Downer NL; Parker MW; Voss AK; Thomas T; Baell JB Discovery of Acylsulfonohydrazide-Derived Inhibitors of the Lysine Acetyltransferase, KAT6A, as Potent Senescence-Inducing Anti-Cancer Agents. J Med Chem 2020, 63 (9), 4655–4684. 10.1021/acs.jmedchem.9b02071. [DOI] [PubMed] [Google Scholar]
- (905).Becht DC; Selvam K; Lachance C; Côté V; Li K; Nguyen MC; Pareek A; Shi X; Wen H; Blanco MA; Côté J; Kutateladze TG A Multivalent Engagement of ENL with MOZ. Nat Struct Mol Biol 2025, 32 (4), 709–718. 10.1038/s41594-024-01455-8. [DOI] [PubMed] [Google Scholar]
- (906).Gordon SJV; Perner F; MacPherson L; Wenge DV; Bourgeois W; Fennell K; Klaus T; Petrovic J; Horvath J; Cao J; Lapek J; Uryu S; White J; Lam EYN; Mu XJ; Chan Y-C; Gillespie A; Blyth B; Camerino MA; Bozikis YE; Holze H; Knezevic K; Balic J; Stupple PA; Street IP; Monahan BJ; Sharma S; Wainwright EN; Vassiliadis D; Paul TA; Armstrong SA; Dawson MA Catalytic Inhibition of KAT6/KAT7 Enhances the Efficacy and Overcomes Primary and Acquired Resistance to Menin Inhibitors in MLL Leukaemia. Cancer Discov 2025. 10.1158/2159-8290.CD-24-1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (907).Michmerhuizen NL; Heikamp EB; Iacobucci I; Umeda M; Arthur B; Mishra V; Di Giacomo D; Hiltenbrand R; Gao Q; Radko-Juettner S; Lott J; Martucci C; Subramanyam V; Hatton C; Baviskar P; Portola P; Claquin A; Chandra B; Baggett DW; Khalighifar A; Huang H; Zhou P; Long L; Shi H; Sun Y; Papachristou EK; Chilamakuri CSR; de L. Vitorino FN; Gongora JM; Janke L; Kentsis A; D’Santos CS; Garcia BA; Kriwacki RW; Chi H; Klco JM; Armstrong SA; Mullighan CG MOZ and HBO1 Histone Acetyltransferase Complexes Are Molecular Dependencies and Therapeutic Targets in NUP98 -Rearranged Acute Myeloid Leukemia. 2024. 10.1101/2024.12.02.624182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (908).MacPherson L; Anokye J; Yeung MM; Lam EYN; Chan Y-C; Weng C-F; Yeh P; Knezevic K; Butler MS; Hoegl A; Chan K-L; Burr ML; Gearing LJ; Willson T; Liu J; Choi J; Yang Y; Bilardi RA; Falk H; Nguyen N; Stupple PA; Peat TS; Zhang M; de Silva M; Carrasco-Pozo C; Avery VM; Khoo PS; Dolezal O; Dennis ML; Nuttall S; Surjadi R; Newman J; Ren B; Leaver DJ; Sun Y; Baell JB; Dovey O; Vassiliou GS; Grebien F; Dawson S-J; Street IP; Monahan BJ; Burns CJ; Choudhary C; Blewitt ME; Voss AK; Thomas T; Dawson MA HBO1 Is Required for the Maintenance of Leukaemia Stem Cells. Nature 2020, 577 (7789), 266–270. 10.1038/s41586-019-1835-6. [DOI] [PubMed] [Google Scholar]
- (909).Au YZ; Gu M; De Braekeleer E; Gozdecka M; Aspris D; Tarumoto Y; Cooper J; Yu J; Ong SH; Chen X; Tzelepis K; Huntly BJP; Vassiliou G; Yusa K KAT7 Is a Genetic Vulnerability of Acute Myeloid Leukemias Driven by MLL Rearrangements. Leukemia 2021, 35 (4), 1012–1022. 10.1038/s41375-020-1001-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (910).Takahashi S; Kanai A; Okuda H; Miyamoto R; Komata Y; Kawamura T; Matsui H; Inaba T; Takaori-Kondo A; Yokoyama A HBO1-MLL Interaction Promotes AF4/ENL/P-TEFb-Mediated Leukemogenesis. Elife 2021, 10, e65872. 10.7554/eLife.65872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (911).Fiskus W; Mill CP; Birdwell C; Davis JA; Das K; Boettcher S; Kadia TM; DiNardo CD; Takahashi K; Loghavi S; Soth MJ; Heffernan T; McGeehan GM; Ruan X; Su X; Vakoc CR; Daver N; Bhalla KN Targeting of Epigenetic Co-Dependencies Enhances Anti-AML Efficacy of Menin Inhibitor in AML with MLL1-r or Mutant NPM1. Blood Cancer J 2023, 13 (1), 53. 10.1038/s41408-023-00826-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (912).Hemming ML; Benson MR; Loycano MA; Anderson JA; Andersen JL; Taddei ML; Krivtsov AV; Aubrey BJ; Cutler JA; Hatton C; Sicinska E; Armstrong SA MOZ and Menin-MLL Complexes Are Complementary Regulators of Chromatin Association and Transcriptional Output in Gastrointestinal Stromal Tumor. Cancer Discov 2022, 12 (7), 1804–1823. 10.1158/2159-8290.CD-21-0646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (913).Turner-Ivey B; Guest ST; Irish JC; Kappler CS; Garrett-Mayer E; Wilson RC; Ethier SP KAT6A, a Chromatin Modifier from the 8p11-P12 Amplicon Is a Candidate Oncogene in Luminal Breast Cancer. Neoplasia 2014, 16 (8), 644–655. 10.1016/j.neo.2014.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (914).Yu L; Liang Y; Cao X; Wang X; Gao H; Lin S-Y; Schiff R; Wang X-S; Li K Identification of MYST3 as a Novel Epigenetic Activator of ERα Frequently Amplified in Breast Cancer. Oncogene 2017, 36 (20), 2910–2918. 10.1038/onc.2016.433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (915).Huang C; Zhu B Roles of H3K36-Specific Histone Methyltransferases in Transcription: Antagonizing Silencing and Safeguarding Transcription Fidelity. Biophys Rep 2018, 4 (4), 170–177. 10.1007/s41048-018-0063-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (916).Azagra A; Cobaleda C NSD2 as a Promising Target in Hematological Disorders. Int J Mol Sci 2022, 23 (19), 11075. 10.3390/ijms231911075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (917).Martinez-Garcia E; Popovic R; Min D-J; Sweet SMM; Thomas PM; Zamdborg L; Heffner A; Will C; Lamy L; Staudt LM; Levens DL; Kelleher NL; Licht JD The MMSET Histone Methyl Transferase Switches Global Histone Methylation and Alters Gene Expression in t(4;14) Multiple Myeloma Cells. Blood 2011, 117 (1), 211–220. 10.1182/blood-2010-07-298349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (918).Brito JLR; Walker B; Jenner M; Dickens NJ; Brown NJM; Ross FM; Avramidou A; Irving JAE; Gonzalez D; Davies FE; Morgan GJ MMSET Deregulation Affects Cell Cycle Progression and Adhesion Regulons in t(4;14) Myeloma Plasma Cells. Haematologica 2009, 94 (1), 78–86. 10.3324/haematol.13426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (919).Huang Z; Wu H; Chuai S; Xu F; Yan F; Englund N; Wang Z; Zhang H; Fang M; Wang Y; Gu J; Zhang M; Yang T; Zhao K; Yu Y; Dai J; Yi W; Zhou S; Li Q; Wu J; Liu J; Wu X; Chan H; Lu C; Atadja P; Li E; Wang Y; Hu M NSD2 Is Recruited through Its PHD Domain to Oncogenic Gene Loci to Drive Multiple Myeloma. Cancer Res 2013, 73 (20), 6277–6288. 10.1158/0008-5472.CAN-13-1000. [DOI] [PubMed] [Google Scholar]
- (920).Lauring J; Abukhdeir AM; Konishi H; Garay JP; Gustin JP; Wang Q; Arceci RJ; Matsui W; Park BH The Multiple Myeloma Associated MMSET Gene Contributes to Cellular Adhesion, Clonogenic Growth, and Tumorigenicity. Blood 2008, 111 (2), 856–864. 10.1182/blood-2007-05-088674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (921).García-Carpizo V; Sarmentero J; Han B; Graña O; Ruiz-Llorente S; Pisano DG; Serrano M; Brooks HB; Campbell RM; Barrero MJ NSD2 Contributes to Oncogenic RAS-Driven Transcription in Lung Cancer Cells through Long-Range Epigenetic Activation. Sci Rep 2016, 6, 32952. 10.1038/srep32952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (922).Aytes A; Giacobbe A; Mitrofanova A; Ruggero K; Cyrta J; Arriaga J; Palomero L; Farran-Matas S; Rubin MA; Shen MM; Califano A; Abate-Shen C NSD2 Is a Conserved Driver of Metastatic Prostate Cancer Progression. Nat Commun 2018, 9 (1), 5201. 10.1038/s41467-018-07511-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (923).Huether R; Dong L; Chen X; Wu G; Parker M; Wei L; Ma J; Edmonson MN; Hedlund EK; Rusch MC; Shurtleff SA; Mulder HL; Boggs K; Vadordaria B; Cheng J; Yergeau D; Song G; Becksfort J; Lemmon G; Weber C; Cai Z; Dang J; Walsh M; Gedman AL; Faber Z; Easton J; Gruber T; Kriwacki RW; Partridge JF; Ding L; Wilson RK; Mardis ER; Mullighan CG; Gilbertson RJ; Baker SJ; Zambetti G; Ellison DW; Zhang J; Downing JR The Landscape of Somatic Mutations in Epigenetic Regulators across 1,000 Paediatric Cancer Genomes. Nat Commun 2014, 5, 3630. 10.1038/ncomms4630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (924).Jaffe JD; Wang Y; Chan HM; Zhang J; Huether R; Kryukov GV; Bhang HC; Taylor JE; Hu M; Englund NP; Yan F; Wang Z; Robert McDonald E; Wei L; Ma J; Easton J; Yu Z; deBeaumount R; Gibaja V; Venkatesan K; Schlegel R; Sellers WR; Keen N; Liu J; Caponigro G; Barretina J; Cooke VG; Mullighan C; Carr SA; Downing JR; Garraway LA; Stegmeier F Global Chromatin Profiling Reveals NSD2 Mutations in Pediatric Acute Lymphoblastic Leukemia. Nat Genet 2013, 45 (11), 1386–1391. 10.1038/ng.2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (925).Oyer JA; Huang X; Zheng Y; Shim J; Ezponda T; Carpenter Z; Allegretta M; Okot-Kotber CI; Patel JP; Melnick A; Levine RL; Ferrando A; Mackerell AD; Kelleher NL; Licht JD; Popovic R Point Mutation E1099K in MMSET/NSD2 Enhances Its Methyltranferase Activity and Leads to Altered Global Chromatin Methylation in Lymphoid Malignancies. Leukemia 2014, 28 (1), 198–201. 10.1038/leu.2013.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (926).Zhao L-H; Li Q; Huang Z-J; Sun M-X; Lu J-J; Zhang X-H; Li G; Wu F Identification of Histone Methyltransferase NSD2 as an Important Oncogenic Gene in Colorectal Cancer. Cell Death Dis 2021, 12 (11), 974. 10.1038/s41419-021-04267-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (927).Kojima M; Sone K; Oda K; Hamamoto R; Kaneko S; Oki S; Kukita A; Machino H; Honjoh H; Kawata Y; Kashiyama T; Asada K; Tanikawa M; Mori-Uchino M; Tsuruga T; Nagasaka K; Matsumoto Y; Wada-Hiraike O; Osuga Y; Fujii T The Histone Methyltransferase WHSC1 Is Regulated by EZH2 and Is Important for Ovarian Clear Cell Carcinoma Cell Proliferation. BMC Cancer 2019, 19 (1), 455. 10.1186/s12885-019-5638-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (928).Keats JJ; Maxwell CA; Taylor BJ; Hendzel MJ; Chesi M; Bergsagel PL; Larratt LM; Mant MJ; Reiman T; Belch AR; Pilarski LM Overexpression of Transcripts Originating from the MMSET Locus Characterizes All t(4;14)(P16;Q32)-Positive Multiple Myeloma Patients. Blood 2005, 105 (10), 4060–4069. 10.1182/blood-2004-09-3704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (929).Martinez-Garcia E; Popovic R; Min D-J; Sweet SMM; Thomas PM; Zamdborg L; Heffner A; Will C; Lamy L; Staudt LM; Levens DL; Kelleher NL; Licht JD The MMSET Histone Methyl Transferase Switches Global Histone Methylation and Alters Gene Expression in t(4;14) Multiple Myeloma Cells. Blood 2011, 117 (1), 211–220. 10.1182/blood-2010-07-298349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (930).Chesi M; Nardini E; Lim RS; Smith KD; Kuehl WM; Bergsagel PL The t(4;14) Translocation in Myeloma Dysregulates Both FGFR3 and a Novel Gene, MMSET, Resulting in IgH/MMSET Hybrid Transcripts. Blood 1998, 92 (9), 3025–3034. [PubMed] [Google Scholar]
- (931).Li W; Tian W; Yuan G; Deng P; Sengupta D; Cheng Z; Cao Y; Ren J; Qin Y; Zhou Y; Jia Y; Gozani O; Patel DJ; Wang Z Molecular Basis of Nucleosomal H3K36 Methylation by NSD Methyltransferases. Nature 2021, 590 (7846), 498–503. 10.1038/s41586-020-03069-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (932).Sato K; Kumar A; Hamada K; Okada C; Oguni A; Machiyama A; Sakuraba S; Nishizawa T; Nureki O; Kono H; Ogata K; Sengoku T Structural Basis of the Regulation of the Normal and Oncogenic Methylation of Nucleosomal Histone H3 Lys36 by NSD2. Nat Commun 2021, 12 (1), 6605. 10.1038/s41467-021-26913-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (933).Swaroop A; Oyer JA; Will CM; Huang X; Yu W; Troche C; Bulic M; Durham BH; Wen QJ; Crispino JD; MacKerell AD; Bennett RL; Kelleher NL; Licht JD An Activating Mutation of the NSD2 Histone Methyltransferase Drives Oncogenic Reprogramming in Acute Lymphocytic Leukemia. Oncogene 2019, 38 (5), 671–686. 10.1038/s41388-018-0474-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (934).Narang S; Evensen NA; Saliba J; Pierro J; Loh ML; Brown PA; Kolekar P; Mulder H; Shao Y; Easton J; Ma X; Tsirigos A; Carroll WL NSD2 E1099K Drives Relapse in Pediatric Acute Lymphoblastic Leukemia by Disrupting 3D Chromatin Organization. Genome Biol 2023, 24 (1), 64. 10.1186/s13059-023-02905-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (935).Li J; Hlavka-Zhang J; Shrimp JH; Piper C; Dupéré-Richér D; Roth JS; Jing D; Casellas Román HL; Troche C; Swaroop A; Kulis M; Oyer JA; Will CM; Shen M; Riva A; Bennett RL; Ferrando AA; Hall MD; Lock RB; Licht JD PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia. Cancer Discov 2022, 12 (1), 186–203. 10.1158/2159-8290.CD-20-1771. [DOI] [PubMed] [Google Scholar]
- (936).Zhang J; Jima D; Moffitt AB; Liu Q; Czader M; Hsi ED; Fedoriw Y; Dunphy CH; Richards KL; Gill JI; Sun Z; Love C; Scotland P; Lock E; Levy S; Hsu DS; Dunson D; Dave SS The Genomic Landscape of Mantle Cell Lymphoma Is Related to the Epigenetically Determined Chromatin State of Normal B Cells. Blood 2014, 123 (19), 2988–2996. 10.1182/blood-2013-07-517177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (937).Beà S; Valdés-Mas R; Navarro A; Salaverria I; Martín-Garcia D; Jares P; Giné E; Pinyol M; Royo C; Nadeu F; Conde L; Juan M; Clot G; Vizán P; Di Croce L; Puente DA; López-Guerra M; Moros A; Roue G; Aymerich M; Villamor N; Colomo L; Martínez A; Valera A; Martín-Subero JI; Amador V; Hernández L; Rozman M; Enjuanes A; Forcada P; Muntañola A; Hartmann EM; Calasanz MJ; Rosenwald A; Ott G; Hernández-Rivas JM; Klapper W; Siebert R; Wiestner A; Wilson WH; Colomer D; López-Guillermo A; López-Otín C; Puente XS; Campo E Landscape of Somatic Mutations and Clonal Evolution in Mantle Cell Lymphoma. Proc Natl Acad Sci U S A 2013, 110 (45), 18250–18255. 10.1073/pnas.1314608110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (938).Peng X; Peng Q; Zhong L Targeting H3K36 Methyltransferases NSDs: A Promising Strategy for Tumor Targeted Therapy. Signal Transduct Target Ther 2021, 6 (1), 220. 10.1038/s41392-021-00616-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (939).Li J; Kulis M; Riva A; Casellas Román H; Dupere-Richer D; Clerio M; Trejo ML; Menes M; Lamberto AJ; Sobh A; Kaestner CL; Bennett RL; Martin Subero II; Licht JD Activating NSD2 Mutations Drive Oncogenic Reprogramming By Disturbing Epigenetic Landscape in Mantle Cell Lymphoma. Blood 2023, 142 (Supplement 1), 2771–2771. 10.1182/blood-2023-189748. [DOI] [Google Scholar]
- (940).Morrison MJ; Boriack-Sjodin PA; Swinger KK; Wigle TJ; Sadalge D; Kuntz KW; Scott MP; Janzen WP; Chesworth R; Duncan KW; Harvey DM; Lampe JW; Mitchell LH; Copeland RA Identification of a Peptide Inhibitor for the Histone Methyltransferase WHSC1. PLoS One 2018, 13 (5), e0197082. 10.1371/journal.pone.0197082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (941).Kubicek S; O’Sullivan RJ; August EM; Hickey ER; Zhang Q; Teodoro ML; Rea S; Mechtler K; Kowalski JA; Homon CA; Kelly TA; Jenuwein T Reversal of H3K9me2 by a Small-Molecule Inhibitor for the G9a Histone Methyltransferase. Mol Cell 2007, 25 (3), 473–481. 10.1016/j.molcel.2007.01.017. [DOI] [PubMed] [Google Scholar]
- (942).Chang Y; Zhang X; Horton JR; Upadhyay AK; Spannhoff A; Liu J; Snyder JP; Bedford MT; Cheng X Structural Basis for G9a-like Protein Lysine Methyltransferase Inhibition by BIX-01294. Nat Struct Mol Biol 2009, 16 (3), 312–317. 10.1038/nsmb.1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (943).Vedadi M; Barsyte-Lovejoy D; Liu F; Rival-Gervier S; Allali-Hassani A; Labrie V; Wigle TJ; Dimaggio PA; Wasney GA; Siarheyeva A; Dong A; Tempel W; Wang S-C; Chen X; Chau I; Mangano TJ; Huang X-P; Simpson CD; Pattenden SG; Norris JL; Kireev DB; Tripathy A; Edwards A; Roth BL; Janzen WP; Garcia BA; Petronis A; Ellis J; Brown PJ; Frye SV; Arrowsmith CH; Jin J A Chemical Probe Selectively Inhibits G9a and GLP Methyltransferase Activity in Cells. Nat Chem Biol 2011, 7 (8), 566–574. 10.1038/nchembio.599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (944).Liu F; Barsyte-Lovejoy D; Allali-Hassani A; He Y; Herold JM; Chen X; Yates CM; Frye SV; Brown PJ; Huang J; Vedadi M; Arrowsmith CH; Jin J Optimization of Cellular Activity of G9a Inhibitors 7-Aminoalkoxy-Quinazolines. J Med Chem 2011, 54 (17), 6139–6150. 10.1021/jm200903z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (945).Liu F; Chen X; Allali-Hassani A; Quinn AM; Wigle TJ; Wasney GA; Dong A; Senisterra G; Chau I; Siarheyeva A; Norris JL; Kireev DB; Jadhav A; Herold JM; Janzen WP; Arrowsmith CH; Frye SV; Brown PJ; Simeonov A; Vedadi M; Jin J Protein Lysine Methyltransferase G9a Inhibitors: Design, Synthesis, and Structure Activity Relationships of 2,4-Diamino-7-Aminoalkoxy-Quinazolines. J Med Chem 2010, 53 (15), 5844–5857. 10.1021/jm100478y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (946).Liu F; Chen X; Allali-Hassani A; Quinn AM; Wasney GA; Dong A; Barsyte D; Kozieradzki I; Senisterra G; Chau I; Siarheyeva A; Kireev DB; Jadhav A; Herold JM; Frye SV; Arrowsmith CH; Brown PJ; Simeonov A; Vedadi M; Jin J Discovery of a 2,4-Diamino-7-Aminoalkoxyquinazoline as a Potent and Selective Inhibitor of Histone Lysine Methyltransferase G9a. J Med Chem 2009, 52 (24), 7950–7953. 10.1021/jm901543m. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (947).Morishita M; Mevius DEHF; Shen Y; Zhao S; di Luccio E BIX-01294 Inhibits Oncoproteins NSD1, NSD2 and NSD3. Medicinal Chemistry Research 2017, 26 (9), 2038–2047. 10.1007/s00044-017-1909-7. [DOI] [Google Scholar]
- (948).Tisi D; Chiarparin E; Tamanini E; Pathuri P; Coyle JE; Hold A; Holding FP; Amin N; Martin ACL; Rich SJ; Berdini V; Yon J; Acklam P; Burke R; Drouin L; Harmer JE; Jeganathan F; van Montfort RLM; Newbatt Y; Tortorici M; Westlake M; Wood A; Hoelder S; Heightman TD Structure of the Epigenetic Oncogene MMSET and Inhibition by N-Alkyl Sinefungin Derivatives. ACS Chem Biol 2016, 11 (11), 3093–3105. 10.1021/acschembio.6b00308. [DOI] [PubMed] [Google Scholar]
- (949).Wei J; Shi Q; Li B; Yang H; Liu L; Zhou R; Feng Z; Yang Z; Zhan J; Xiong X-F; Huang X; Wang Y Discovery of a Highly Potent and Selective Inhibitor Targeting Protein Lysine Methyltransferase NSD2. J Med Chem 2024, 67 (18), 16056–16071. 10.1021/acs.jmedchem.4c00639. [DOI] [PubMed] [Google Scholar]
- (950).Yang C; Li B; Feng Z; Li H; Yang H; Yang Z; Liu L; Shi Q; Wang H; Chen Z-Z; Huang X; Wang J; Wang Y Discovery of a Highly Potent Lysine Methyltransferases G9a/NSD2 Dual Inhibitor to Treat Solid Tumors. J Med Chem 2024, 67 (18), 16072–16087. 10.1021/acs.jmedchem.4c00640. [DOI] [PubMed] [Google Scholar]
- (951).Coussens NP; Kales SC; Henderson MJ; Lee OW; Horiuchi KY; Wang Y; Chen Q; Kuznetsova E; Wu J; Chakka S; Cheff DM; Cheng KC-C; Shinn P; Brimacombe KR; Shen M; Simeonov A; Lal-Nag M; Ma H; Jadhav A; Hall MD High-Throughput Screening with Nucleosome Substrate Identifies Small-Molecule Inhibitors of the Human Histone Lysine Methyltransferase NSD2. J Biol Chem 2018, 293 (35), 13750–13765. 10.1074/jbc.RA118.004274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (952).Shen Y; Morishita M; Lee D; Kim S; Lee T; Mevius DEHF; Roh Y; di Luccio E Identification of LEM-14 Inhibitor of the Oncoprotein NSD2. Biochem Biophys Res Commun 2019, 508 (1), 102–108. 10.1016/j.bbrc.2018.11.037. [DOI] [PubMed] [Google Scholar]
- (953).Wang S; Yang H; Su M; Lian F; Cong Z; Wei R; Zhou Y; Li X; Zheng X; Li C; Fu X; Han X; Shi Q; Li C; Zhang N; Geng M; Liu H; Li J; Huang X; Wang J 5-Aminonaphthalene Derivatives as Selective Nonnucleoside Nuclear Receptor Binding SET Domain-Protein 2 (NSD2) Inhibitors for the Treatment of Multiple Myeloma. Eur J Med Chem 2021, 222, 113592. 10.1016/j.ejmech.2021.113592. [DOI] [PubMed] [Google Scholar]
- (954).Richon VM; Johnston D; Sneeringer CJ; Jin L; Majer CR; Elliston K; Jerva LF; Scott MP; Copeland RA Chemogenetic Analysis of Human Protein Methyltransferases. Chem Biol Drug Des 2011, 78 (2), 199–210. 10.1111/j.1747-0285.2011.01135.x. [DOI] [PubMed] [Google Scholar]
- (955).Li Y; Trojer P; Xu C-F; Cheung P; Kuo A; Drury WJ; Qiao Q; Neubert TA; Xu R-M; Gozani O; Reinberg D The Target of the NSD Family of Histone Lysine Methyltransferases Depends on the Nature of the Substrate. J Biol Chem 2009, 284 (49), 34283–34295. 10.1074/jbc.M109.034462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (956).Tang H; Yu A; Xing L; Chen X; Ding H; Yang H; Song Z; Shi Q; Geng M; Huang X; Zhang A Structural Modification and Pharmacological Evaluation of Substituted Quinoline-5,8-Diones as Potent NSD2 Inhibitors. J Med Chem 2023, 66 (2), 1634–1651. 10.1021/acs.jmedchem.2c01920. [DOI] [PubMed] [Google Scholar]
- (957).Berdeja JG; Dingli D; Lonial S; Yee AJ; Chung A; Gasparetto C; Bergsagel PL; Roy V; Abdallah A-O; Thanendrarajan S; Afrough A; Rosiñol L; Trudel S; Rodriguez Otero P; Touzeau C; Bories P; Bang S; Connolly T; Kim M; Lewis CA; Linberg B-VT; Quigley MT; Landgren O; Usmani SZ; Mateos M-V; Siegel DS Trial in Progress: A Phase 1 Study of KTX-1001, an Oral, First-in-Class, Selective MMSET Inhibitor in Patients with Relapsed and Refractory Multiple Myeloma. Blood 2023, 142 (Supplement 1), 3391–3391. 10.1182/blood-2023-174550. [DOI] [Google Scholar]
- (958).Jeong J; Hausmann S; Dong H; Szczepski K; Flores NM; Garcia Gonzalez A; Shi L; Lu X; Lempiäinen J; Jakab M; Zeng L; Chasan T; Bareke E; Dong R; Carlson E; Padilla R; Husmann D; Thompson J; Shipman GA; Zahn E; Barnes CA; Khan LF; Albertorio-Sáez LM; Brill E; Kumary VUS; Marunde MR; Maryanski DN; Szany CC; Venters BJ; Windham CL; Nowakowski ME; Czaban I; Jaremko M; Keogh M-C; Le K; Soth MJ; Garcia BA; Jaremko Ł; Majewski J; Mazur PK; Gozani O NSD2 Inhibitors Rewire Chromatin to Treat Lung and Pancreatic Cancers. Nature 2025. 10.1038/s41586-025-09299-y. [DOI] [PubMed] [Google Scholar]
- (959).Huang H; Howard CA; Zari S; Cho HJ; Shukla S; Li H; Ndoj J; González-Alonso P; Nikolaidis C; Abbott J; Rogawski DS; Potopnyk MA; Kempinska K; Miao H; Purohit T; Henderson A; Mapp A; Sulis ML; Ferrando A; Grembecka J; Cierpicki T Covalent Inhibition of NSD1 Histone Methyltransferase. Nat Chem Biol 2020, 16 (12), 1403–1410. 10.1038/s41589-020-0626-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (960).Wang GG; Cai L; Pasillas MP; Kamps MP NUP98-NSD1 Links H3K36 Methylation to Hox-A Gene Activation and Leukaemogenesis. Nat Cell Biol 2007, 9 (7), 804–812. 10.1038/ncb1608. [DOI] [PubMed] [Google Scholar]
- (961).Rosati R; La Starza R; Veronese A; Aventin A; Schwienbacher C; Vallespi T; Negrini M; Martelli MF; Mecucci C NUP98 Is Fused to the NSD3 Gene in Acute Myeloid Leukemia Associated with t(8;11)(P11.2;P15). Blood 2002, 99 (10), 3857–3860. 10.1182/blood.v99.10.3857. [DOI] [PubMed] [Google Scholar]
- (962).Jaju RJ; Fidler C; Haas OA; Strickson AJ; Watkins F; Clark K; Cross NC; Cheng JF; Aplan PD; Kearney L; Boultwood J; Wainscoat JS A Novel Gene, NSD1, Is Fused to NUP98 in the t(5;11)(Q35;P15.5) in de Novo Childhood Acute Myeloid Leukemia. Blood 2001, 98 (4), 1264–1267. 10.1182/blood.v98.4.1264. [DOI] [PubMed] [Google Scholar]
- (963).Sankaran SM; Wilkinson AW; Elias JE; Gozani O A PWWP Domain of Histone-Lysine N-Methyltransferase NSD2 Binds to Dimethylated Lys-36 of Histone H3 and Regulates NSD2 Function at Chromatin. J Biol Chem 2016, 291 (16), 8465–8474. 10.1074/jbc.M116.720748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (964).Taverna SD; Li H; Ruthenburg AJ; Allis CD; Patel DJ How Chromatin-Binding Modules Interpret Histone Modifications: Lessons from Professional Pocket Pickers. Nat Struct Mol Biol 2007, 14 (11), 1025–1040. 10.1038/nsmb1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (965).Sankaran SM; Wilkinson AW; Elias JE; Gozani O A PWWP Domain of Histone-Lysine N-Methyltransferase NSD2 Binds to Dimethylated Lys-36 of Histone H3 and Regulates NSD2 Function at Chromatin. J Biol Chem 2016, 291 (16), 8465–8474. 10.1074/jbc.M116.720748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (966).Böttcher J; Dilworth D; Reiser U; Neumüller RA; Schleicher M; Petronczki M; Zeeb M; Mischerikow N; Allali-Hassani A; Szewczyk MM; Li F; Kennedy S; Vedadi M; Barsyte-Lovejoy D; Brown PJ; Huber KVM; Rogers CM; Wells CI; Fedorov O; Rumpel K; Zoephel A; Mayer M; Wunberg T; Böse D; Zahn S; Arnhof H; Berger H; Reiser C; Hörmann A; Krammer T; Corcokovic M; Sharps B; Winkler S; Häring D; Cockcroft X-L; Fuchs JE; Müllauer B; Weiss-Puxbaum A; Gerstberger T; Boehmelt G; Vakoc CR; Arrowsmith CH; Pearson M; McConnell DB Fragment-Based Discovery of a Chemical Probe for the PWWP1 Domain of NSD3. Nat Chem Biol 2019, 15 (8), 822–829. 10.1038/s41589-019-0310-x. [DOI] [PubMed] [Google Scholar]
- (967).Shen C; Ipsaro JJ; Shi J; Milazzo JP; Wang E; Roe J-S; Suzuki Y; Pappin DJ; Joshua-Tor L; Vakoc CR NSD3-Short Is an Adaptor Protein That Couples BRD4 to the CHD8 Chromatin Remodeler. Mol Cell 2015, 60 (6), 847–859. 10.1016/j.molcel.2015.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (968).Xu C; Meng F; Park K-S; Storey AJ; Gong W; Tsai Y-H; Gibson E; Byrum SD; Li D; Edmondson RD; Mackintosh SG; Vedadi M; Cai L; Tackett AJ; Kaniskan HÜ; Jin J; Wang GG A NSD3-Targeted PROTAC Suppresses NSD3 and CMyc Oncogenic Nodes in Cancer Cells. Cell Chem Biol 2022, 29 (3), 386–397.e9. 10.1016/j.chembiol.2021.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (969).Sun Y; Zhang Y; Chen X; Yu A; Du W; Huang Y; Wu F; Yu L; Li J; Wen C; Yang H; Shi Q; Geng M; Huang X; Xu S Discovery of a Potent and Selective Proteolysis Targeting Chimera (PROTAC) Degrader of NSD3 Histone Methyltransferase. Eur J Med Chem 2022, 239, 114528. 10.1016/j.ejmech.2022.114528. [DOI] [PubMed] [Google Scholar]
- (970).LegaardAndersson J; Christensen J; Kleine-Kohlbrecher D; Vacher Comet I; Fullerton Støier J; Antoku Y; Poljak V; Moretti L; Dolberg J; Jacso T; Jensby Nielsen S; Nørregaard-Madsen M; Franch T; Helin K; Cloos PAC Discovery of NSD2-Degraders from Novel and Selective DEL Hits. Chembiochem 2023, 24 (24), e202300515. 10.1002/cbic.202300515. [DOI] [PubMed] [Google Scholar]
- (971).El Ashkar S; Schwaller J; Pieters T; Goossens S; Demeulemeester J; Christ F; Van Belle S; Juge S; Boeckx N; Engelman A; Van Vlierberghe P; Debyser Z; De Rijck J LEDGF/P75 Is Dispensable for Hematopoiesis but Essential for MLL-Rearranged Leukemogenesis. Blood 2018, 131 (1), 95–107. 10.1182/blood-2017-05-786962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (972).Okuda H; Kawaguchi M; Kanai A; Matsui H; Kawamura T; Inaba T; Kitabayashi I; Yokoyama A MLL Fusion Proteins Link Transcriptional Coactivators to Previously Active CpG-Rich Promoters. Nucleic Acids Res 2014, 42 (7), 4241–4256. 10.1093/nar/gkt1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (973).van Nuland R; van Schaik FM; Simonis M; van Heesch S; Cuppen E; Boelens R; Timmers HM; van Ingen H Nucleosomal DNA Binding Drives the Recognition of H3K36-Methylated Nucleosomes by the PSIP1-PWWP Domain. Epigenetics Chromatin 2013, 6 (1), 12. 10.1186/1756-8935-6-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (974).Wang H; Farnung L; Dienemann C; Cramer P Structure of H3K36-Methylated Nucleosome-PWWP Complex Reveals Multivalent Cross-Gyre Binding. Nat Struct Mol Biol 2020, 27 (1), 8–13. 10.1038/s41594-019-0345-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (975).Wang Y; Dix MM; Bianco G; Remsberg JR; Lee H-Y; Kalocsay M; Gygi SP; Forli S; Vite G; Lawrence RM; Parker CG; Cravatt BF Expedited Mapping of the Ligandable Proteome Using Fully Functionalized Enantiomeric Probe Pairs. Nat Chem 2019, 11 (12), 1113–1123. 10.1038/s41557-019-0351-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (976).Ferreira de Freitas R; Liu Y; Szewczyk MM; Mehta N; Li F; McLeod D; Zepeda-Velázquez C; Dilworth D; Hanley RP; Gibson E; Brown PJ; Al-Awar R; James LI; Arrowsmith CH; Barsyte-Lovejoy D; Min J; Vedadi M; Schapira M; Allali-Hassani A Discovery of Small-Molecule Antagonists of the PWWP Domain of NSD2. J Med Chem 2021, 64 (3), 1584–1592. 10.1021/acs.jmedchem.0c01768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (977).Dilworth D; Hanley RP; Ferreira de Freitas R; Allali-Hassani A; Zhou M; Mehta N; Marunde MR; Ackloo S; Carvalho Machado RA; Khalili Yazdi A; Owens DDG; Vu V; Nie DY; Alqazzaz M; Marcon E; Li F; Chau I; Bolotokova A; Qin S; Lei M; Liu Y; Szewczyk MM; Dong A; Kazemzadeh S; Abramyan T; Popova IK; Hall NW; Meiners MJ; Cheek MA; Gibson E; Kireev D; Greenblatt JF; Keogh MC; Min J; Brown PJ; Vedadi M; Arrowsmith CH; Barsyte-Lovejoy D; James LI; Schapira M A Chemical Probe Targeting the PWWP Domain Alters NSD2 Nucleolar Localization. Nat Chem Biol 2022, 18 (1), 56–63. 10.1038/s41589-021-00898-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (978).Hanley RP; Nie DY; Tabor JR; Li F; Sobh A; Xu C; Barker NK; Dilworth D; Hajian T; Gibson E; Szewczyk MM; Brown PJ; Barsyte-Lovejoy D; Herring LE; Wang GG; Licht JD; Vedadi M; Arrowsmith CH; James LI Discovery of a Potent and Selective Targeted NSD2 Degrader for the Reduction of H3K36me2. J Am Chem Soc 2023, 145 (14), 8176–8188. 10.1021/jacs.3c01421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (979).Shanmugasundaram K; Shao P; Chen H; Campos B; McHardy SF; Luo T; Rao H A Modular PROTAC Design for Target Destruction Using a Degradation Signal Based on a Single Amino Acid. J Biol Chem 2019, 294 (41), 15172–15175. 10.1074/jbc.AC119.010790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (980).Ryan A; Liu J; Deiters A Targeted Protein Degradation through Fast Optogenetic Activation and Its Application to the Control of Cell Signaling. J Am Chem Soc 2021, 143 (24), 9222–9229. 10.1021/jacs.1c04324. [DOI] [PubMed] [Google Scholar]
- (981).Matta-Camacho E; Kozlov G; Li FF; Gehring K Structural Basis of Substrate Recognition and Specificity in the N-End Rule Pathway. Nat Struct Mol Biol 2010, 17 (10), 1182–1187. 10.1038/nsmb.1894. [DOI] [PubMed] [Google Scholar]
- (982).Bachmair A; Finley D; Varshavsky A In Vivo Half-Life of a Protein Is a Function of Its Amino-Terminal Residue. Science (1979) 1986, 234 (4773), 179–186. 10.1126/science.3018930. [DOI] [PubMed] [Google Scholar]
- (983).Nie DY; Tabor JR; Li J; Kutera M; St-Germain J; Hanley RP; Wolf E; Paulakonis E; Kenney TMG; Duan S; Shrestha S; Owens DDG; Pon A; Szewczyk M; Lamberto AJ; Menes M; Li F; Barsyte-Lovejoy D; Brown NG; Barsotti AM; Stamford AW; Collins JL; Wilson DJ; Raught B; Licht JD; James LI; Arrowsmith CH Recruitment of FBXO22 for Targeted Degradation of NSD2. bioRxiv 2023. 10.1101/2023.11.01.564830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (984).Nie DY; Tabor JR; Li J; Kutera M; St-Germain J; Hanley RP; Wolf E; Paulakonis E; Kenney TMG; Duan S; Shrestha S; Owens DDG; Maitland MER; Pon A; Szewczyk M; Lamberto AJ; Menes M; Li F; Penn LZ; Barsyte-Lovejoy D; Brown NG; Barsotti AM; Stamford AW; Collins JL; Wilson DJ; Raught B; Licht JD; James LI; Arrowsmith CH Recruitment of FBXO22 for Targeted Degradation of NSD2. Nat Chem Biol 2024, 20 (12), 1597–1607. 10.1038/s41589-024-01660-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (985).Kagiou C; Cisneros JA; Farnung J; Liwocha J; Offensperger F; Dong K; Yang K; Tin G; Horstmann CS; Hinterndorfer M; Paulo JA; Scholes NS; Sanchez Avila J; Fellner M; Andersch F; Hannich JT; Zuber J; Kubicek S; Gygi SP; Schulman BA; Winter GE Alkylamine-Tethered Molecules Recruit FBXO22 for Targeted Protein Degradation. Nat Commun 2024, 15 (1), 5409. 10.1038/s41467-024-49739-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (986).den Besten W; Verma K; Yamazoe S; Blaquiere N; Phung W; Izrael-Tomasevic A; Mulvihill MM; Helgason E; Prakash S; Goncharov T; Vucic D; Dueber E; Fairbrother WJ; Wertz I; Yu K; Staben ST Primary Amine Tethered Small Molecules Promote the Degradation of X-Linked Inhibitor of Apoptosis Protein. J Am Chem Soc 2021, 143 (28), 10571–10575. 10.1021/jacs.1c05269. [DOI] [PubMed] [Google Scholar]
- (987).Meng F; Xu C; Park K-S; Kaniskan HÜ; Wang GG; Jin J Discovery of a First-in-Class Degrader for Nuclear Receptor Binding SET Domain Protein 2 (NSD2) and Ikaros/Aiolos. J Med Chem 2022, 65 (15), 10611–10625. 10.1021/acs.jmedchem.2c00807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (988).Krönke J; Udeshi ND; Narla A; Grauman P; Hurst SN; McConkey M; Svinkina T; Heckl D; Comer E; Li X; Ciarlo C; Hartman E; Munshi N; Schenone M; Schreiber SL; Carr SA; Ebert BL Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells. Science (1979) 2014, 343 (6168), 301–305. 10.1126/science.1244851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (989).Lu G; Middleton RE; Sun H; Naniong M; Ott CJ; Mitsiades CS; Wong K-K; Bradner JE; Kaelin WG The Myeloma Drug Lenalidomide Promotes the Cereblon-Dependent Destruction of Ikaros Proteins. Science 2014, 343 (6168), 305–309. 10.1126/science.1244917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (990).Liu L; Parolia A; Liu Y; Hou C; He T; Qiao Y; Eyunni S; Luo J; Li C; Wang Y; Zhou F; Huang W; Ren X; Wang Z; Chinnaiyan AM; Ding K Discovery of LLC0424 as a Potent and Selective in Vivo NSD2 PROTAC Degrader. J Med Chem 2024, 67 (9), 6938–6951. 10.1021/acs.jmedchem.3c01765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (991).Edmunds JW; Mahadevan LC; Clayton AL Dynamic Histone H3 Methylation during Gene Induction: HYPB/Setd2 Mediates All H3K36 Trimethylation. EMBO J 2008, 27 (2), 406–420. 10.1038/sj.emboj.7601967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (992).Carvalho S; Vítor AC; Sridhara SC; Martins FB; Raposo AC; Desterro JMP; Ferreira J; de Almeida SF SETD2 Is Required for DNA Double-Strand Break Repair and Activation of the P53-Mediated Checkpoint. Elife 2014, 3, e02482. 10.7554/eLife.02482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (993).Aymard F; Bugler B; Schmidt CK; Guillou E; Caron P; Briois S; Iacovoni JS; Daburon V; Miller KM; Jackson SP; Legube G Transcriptionally Active Chromatin Recruits Homologous Recombination at DNA Double-Strand Breaks. Nat Struct Mol Biol 2014, 21 (4), 366–374. 10.1038/nsmb.2796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (994).de Almeida SF; Grosso AR; Koch F; Fenouil R; Carvalho S; Andrade J; Levezinho H; Gut M; Eick D; Gut I; Andrau J-C; Ferrier P; Carmo-Fonseca M Splicing Enhances Recruitment of Methyltransferase HYPB/Setd2 and Methylation of Histone H3 Lys36. Nat Struct Mol Biol 2011, 18 (9), 977–983. 10.1038/nsmb.2123. [DOI] [PubMed] [Google Scholar]
- (995).Dalgliesh GL; Furge K; Greenman C; Chen L; Bignell G; Butler A; Davies H; Edkins S; Hardy C; Latimer C; Teague J; Andrews J; Barthorpe S; Beare D; Buck G; Campbell PJ; Forbes S; Jia M; Jones D; Knott H; Kok CY; Lau KW; Leroy C; Lin M-L; McBride DJ; Maddison M; Maguire S; McLay K; Menzies A; Mironenko T; Mulderrig L; Mudie L; O’Meara S; Pleasance E; Rajasingham A; Shepherd R; Smith R; Stebbings L; Stephens P; Tang G; Tarpey PS; Turrell K; Dykema KJ; Khoo SK; Petillo D; Wondergem B; Anema J; Kahnoski RJ; Teh BT; Stratton MR; Futreal PA Systematic Sequencing of Renal Carcinoma Reveals Inactivation of Histone Modifying Genes. Nature 2010, 463 (7279), 360–363. 10.1038/nature08672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (996).Li J; Duns G; Westers H; Sijmons R; van den Berg A; Kok K SETD2: An Epigenetic Modifier with Tumor Suppressor Functionality. Oncotarget 2016, 7 (31), 50719–50734. 10.18632/oncotarget.9368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (997).Leung W; Teater M; Durmaz C; Meydan C; Chivu AG; Chadburn A; Rice EJ; Muley A; Camarillo JM; Arivalagan J; Li Z; Flowers CR; Kelleher NL; Danko CG; Imielinski M; Dave SS; Armstrong SA; Mason CE; Melnick AM SETD2 Haploinsufficiency Enhances Germinal Center-Associated AICDA Somatic Hypermutation to Drive B-Cell Lymphomagenesis. Cancer Discov 2022, 12 (7), 1782–1803. 10.1158/2159-8290.CD-21-1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (998).Kuo AJ; Cheung P; Chen K; Zee BM; Kioi M; Lauring J; Xi Y; Park BH; Shi X; Garcia BA; Li W; Gozani O NSD2 Links Dimethylation of Histone H3 at Lysine 36 to Oncogenic Programming. Mol Cell 2011, 44 (4), 609–620. 10.1016/j.molcel.2011.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (999).Bu J; Chen A; Yan X; He F; Dong Y; Zhou Y; He J; Zhan D; Lin P; Hayashi Y; Sun Y; Zhang Y; Xiao Z; Grimes HL; Wang QF; Huang G SETD2-Mediated Crosstalk between H3K36me3 and H3K79me2 in MLL-Rearranged Leukemia. Leukemia 2018, 32 (4), 890–899. 10.1038/leu.2017.339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1000).Kwok HS; Freedy AM; Siegenfeld AP; Morriss JW; Waterbury AL; Kissler SM; Liau BB Drug Addiction Unveils a Repressive Methylation Ceiling in EZH2-Mutant Lymphoma. Nat Chem Biol 2023, 19 (9), 1105–1115. 10.1038/s41589-023-01299-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1001).Zheng W; Ibáñez G; Wu H; Blum G; Zeng H; Dong A; Li F; Hajian T; Allali-Hassani A; Amaya MF; Siarheyeva A; Yu W; Brown PJ; Schapira M; Vedadi M; Min J; Luo M Sinefungin Derivatives as Inhibitors and Structure Probes of Protein Lysine Methyltransferase SETD2. J Am Chem Soc 2012, 134 (43), 18004–18014. 10.1021/ja307060p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1002).Lampe JW; Alford JS; Boriak-Sjodin PA; Brach D; Cosmopoulos K; Duncan KW; Eckley ST; Foley MA; Harvey DM; Motwani V; Munchhof MJ; Raimondi A; Riera TV; Tang C; Thomenius MJ; Totman J; Farrow NA Discovery of a First-in-Class Inhibitor of the Histone Methyltransferase SETD2 Suitable for Preclinical Studies. ACS Med Chem Lett 2021, 12 (10), 1539–1545. 10.1021/acsmedchemlett.1c00272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1003).Alford JS; Lampe JW; Brach D; Chesworth R; Cosmopoulos K; Duncan KW; Eckley ST; Kutok JL; Raimondi A; Riera TV; Shook B; Tang C; Totman J; Farrow NA Conformational-Design-Driven Discovery of EZM0414: A Selective, Potent SETD2 Inhibitor for Clinical Studies. ACS Med Chem Lett 2022, 13 (7), 1137–1143. 10.1021/acsmedchemlett.2c00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1004).Shi Y; Lan F; Matson C; Mulligan P; Whetstine JR; Cole PA; Casero RA; Shi Y Histone Demethylation Mediated by the Nuclear Amine Oxidase Homolog LSD1. Cell 2004, 119 (7), 941–953. 10.1016/j.cell.2004.12.012. [DOI] [PubMed] [Google Scholar]
- (1005).Humphrey GW; Wang Y; Russanova VR; Hirai T; Qin J; Nakatani Y; Howard BH Stable Histone Deacetylase Complexes Distinguished by the Presence of SANT Domain Proteins CoREST/Kiaa0071 and Mta-L1. J Biol Chem 2001, 276 (9), 6817–6824. 10.1074/jbc.M007372200. [DOI] [PubMed] [Google Scholar]
- (1006).You A; Tong JK; Grozinger CM; Schreiber SL CoREST Is an Integral Component of the CoREST- Human Histone Deacetylase Complex. Proc Natl Acad Sci U S A 2001, 98 (4), 1454–1458. 10.1073/pnas.98.4.1454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1007).Tong JK; Hassig CA; Schnitzler GR; Kingston RE; Schreiber SL Chromatin Deacetylation by an ATP-Dependent Nucleosome Remodelling Complex. Nature 1998, 395 (6705), 917–921. 10.1038/27699. [DOI] [PubMed] [Google Scholar]
- (1008).Shi Y; Sawada J; Sui G; Affar EB; Whetstine JR; Lan F; Ogawa H; Luke MP-S; Nakatani Y; Shi Y Coordinated Histone Modifications Mediated by a CtBP Co-Repressor Complex. Nature 2003, 422 (6933), 735–738. 10.1038/nature01550. [DOI] [PubMed] [Google Scholar]
- (1009).Metzger E; Wissmann M; Yin N; Müller JM; Schneider R; Peters AHFM; Günther T; Buettner R; Schüle R LSD1 Demethylates Repressive Histone Marks to Promote Androgen-Receptor-Dependent Transcription. Nature 2005, 437 (7057), 436–439. 10.1038/nature04020. [DOI] [PubMed] [Google Scholar]
- (1010).Whyte WA; Bilodeau S; Orlando DA; Hoke HA; Frampton GM; Foster CT; Cowley SM; Young RA Enhancer Decommissioning by LSD1 during Embryonic Stem Cell Differentiation. Nature 2012, 482 (7384), 221–225. 10.1038/nature10805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1011).Ishikawa Y; Gamo K; Yabuki M; Takagi S; Toyoshima K; Nakayama K; Nakayama A; Morimoto M; Miyashita H; Dairiki R; Hikichi Y; Tomita N; Tomita D; Imamura S; Iwatani M; Kamada Y; Matsumoto S; Hara R; Nomura T; Tsuchida K; Nakamura K A Novel LSD1 Inhibitor T-3775440 Disrupts GFI1B-Containing Complex Leading to Transdifferentiation and Impaired Growth of AML Cells. Mol Cancer Ther 2017, 16 (2), 273–284. 10.1158/1535-7163.MCT-16-0471. [DOI] [PubMed] [Google Scholar]
- (1012).Sehrawat A; Gao L; Wang Y; Bankhead A; McWeeney SK; King CJ; Schwartzman J; Urrutia J; Bisson WH; Coleman DJ; Joshi SK; Kim D-H; Sampson DA; Weinmann S; Kallakury BVS; Berry DL; Haque R; Van Den Eeden SK; Sharma S; Bearss J; Beer TM; Thomas GV; Heiser LM; Alumkal JJ LSD1 Activates a Lethal Prostate Cancer Gene Network Independently of Its Demethylase Function. Proc Natl Acad Sci U S A 2018, 115 (18), E4179–E4188. 10.1073/pnas.1719168115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1013).Maiques-Diaz A; Spencer GJ; Lynch JT; Ciceri F; Williams EL; Amaral FMR; Wiseman DH; Harris WJ; Li Y; Sahoo S; Hitchin JR; Mould DP; Fairweather EE; Waszkowycz B; Jordan AM; Smith DL; Somervaille TCP Enhancer Activation by Pharmacologic Displacement of LSD1 from GFI1 Induces Differentiation in Acute Myeloid Leukemia. Cell Rep 2018, 22 (13), 3641–3659. 10.1016/j.celrep.2018.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1014).Vinyard ME; Su C; Siegenfeld AP; Waterbury AL; Freedy AM; Gosavi PM; Park Y; Kwan EE; Senzer BD; Doench JG; Bauer DE; Pinello L; Liau BB CRISPR-Suppressor Scanning Reveals a Nonenzymatic Role of LSD1 in AML. Nat Chem Biol 2019, 15 (5), 529–539. 10.1038/s41589-019-0263-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1015).Zeng C; Chen J; Cooke EW; Subuddhi A; Roodman ET; Chen FX; Cao K Demethylase-Independent Roles of LSD1 in Regulating Enhancers and Cell Fate Transition. Nat Commun 2023, 14 (1), 4944. 10.1038/s41467-023-40606-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1016).Mohammad HP; Smitheman KN; Kamat CD; Soong D; Federowicz KE; Van Aller GS; Schneck JL; Carson JD; Liu Y; Butticello M; Bonnette WG; Gorman SA; Degenhardt Y; Bai Y; McCabe MT; Pappalardi MB; Kasparec J; Tian X; McNulty KC; Rouse M; McDevitt P; Ho T; Crouthamel M; Hart TK; Concha NO; McHugh CF; Miller WH; Dhanak D; Tummino PJ; Carpenter CL; Johnson NW; Hann CL; Kruger RG A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell 2015, 28 (1), 57–69. 10.1016/j.ccell.2015.06.002. [DOI] [PubMed] [Google Scholar]
- (1017).Schenk T; Chen WC; Göllner S; Howell L; Jin L; Hebestreit K; Klein H-U; Popescu AC; Burnett A; Mills K; Casero RA; Marton L; Woster P; Minden MD; Dugas M; Wang JCY; Dick JE; Müller-Tidow C; Petrie K; Zelent A Inhibition of the LSD1 (KDM1A) Demethylase Reactivates the All-Trans-Retinoic Acid Differentiation Pathway in Acute Myeloid Leukemia. Nat Med 2012, 18 (4), 605–611. 10.1038/nm.2661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1018).McGrath JP; Williamson KE; Balasubramanian S; Odate S; Arora S; Hatton C; Edwards TM; O’Brien T; Magnuson S; Stokoe D; Daniels DL; Bryant BM; Trojer P Pharmacological Inhibition of the Histone Lysine Demethylase KDM1A Suppresses the Growth of Multiple Acute Myeloid Leukemia Subtypes. Cancer Res 2016, 76 (7), 1975–1988. 10.1158/0008-5472.CAN-15-2333. [DOI] [PubMed] [Google Scholar]
- (1019).Takagi S; Ishikawa Y; Mizutani A; Iwasaki S; Matsumoto S; Kamada Y; Nomura T; Nakamura K LSD1 Inhibitor T-3775440 Inhibits SCLC Cell Proliferation by Disrupting LSD1 Interactions with SNAG Domain Proteins INSM1 and GFI1B. Cancer Res 2017, 77 (17), 4652–4662. 10.1158/0008-5472.CAN-16-3502. [DOI] [PubMed] [Google Scholar]
- (1020).Cusan M; Cai SF; Mohammad HP; Krivtsov A; Chramiec A; Loizou E; Witkin MD; Smitheman KN; Tenen DG; Ye M; Will B; Steidl U; Kruger RG; Levine RL; Rienhoff HY; Koche RP; Armstrong SA LSD1 Inhibition Exerts Its Antileukemic Effect by Recommissioning PU.1- and C/EBPα-Dependent Enhancers in AML. Blood 2018, 131 (15), 1730–1742. 10.1182/blood-2017-09-807024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1021).Maes T; Mascaró C; Tirapu I; Estiarte A; Ciceri F; Lunardi S; Guibourt N; Perdones A; Lufino MMP; Somervaille TCP; Wiseman DH; Duy C; Melnick A; Willekens C; Ortega A; Martinell M; Valls N; Kurz G; Fyfe M; Castro-Palomino JC; Buesa C ORY-1001, a Potent and Selective Covalent KDM1A Inhibitor, for the Treatment of Acute Leukemia. Cancer Cell 2018, 33 (3), 495–511.e12. 10.1016/j.ccell.2018.02.002. [DOI] [PubMed] [Google Scholar]
- (1022).Augert A; Eastwood E; Ibrahim AH; Wu N; Grunblatt E; Basom R; Liggitt D; Eaton KD; Martins R; Poirier JT; Rudin CM; Milletti F; Cheng W-Y; Mack F; MacPherson D Targeting NOTCH Activation in Small Cell Lung Cancer through LSD1 Inhibition. Sci Signal 2019, 12 (567), eaau2922. 10.1126/scisignal.aau2922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1023).Harris WJ; Huang X; Lynch JT; Spencer GJ; Hitchin JR; Li Y; Ciceri F; Blaser JG; Greystoke BF; Jordan AM; Miller CJ; Ogilvie DJ; Somervaille TCP The Histone Demethylase KDM1A Sustains the Oncogenic Potential of MLL-AF9 Leukemia Stem Cells. Cancer Cell 2012, 21 (4), 473–487. 10.1016/j.ccr.2012.03.014. [DOI] [PubMed] [Google Scholar]
- (1024).Bennani-Baiti IM; Machado I; Llombart-Bosch A; Kovar H Lysine-Specific Demethylase 1 (LSD1/KDM1A/AOF2/BHC110) Is Expressed and Is an Epigenetic Drug Target in Chondrosarcoma, Ewing’s Sarcoma, Osteosarcoma, and Rhabdomyosarcoma. Hum Pathol 2012, 43 (8), 1300–1307. 10.1016/j.humpath.2011.10.010. [DOI] [PubMed] [Google Scholar]
- (1025).Schulte JH; Lim S; Schramm A; Friedrichs N; Koster J; Versteeg R; Ora I; Pajtler K; Klein-Hitpass L; Kuhfittig-Kulle S; Metzger E; Schüle R; Eggert A; Buettner R; Kirfel J Lysine-Specific Demethylase 1 Is Strongly Expressed in Poorly Differentiated Neuroblastoma: Implications for Therapy. Cancer Res 2009, 69 (5), 2065–2071. 10.1158/0008-5472.CAN-08-1735. [DOI] [PubMed] [Google Scholar]
- (1026).Binda C; Li M; Hubalek F; Restelli N; Edmondson DE; Mattevi A Insights into the Mode of Inhibition of Human Mitochondrial Monoamine Oxidase B from High-Resolution Crystal Structures. Proc Natl Acad Sci U S A 2003, 100 (17), 9750–9755. 10.1073/pnas.1633804100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1027).Culhane JC; Wang D; Yen PM; Cole PA Comparative Analysis of Small Molecules and Histone Substrate Analogues as LSD1 Lysine Demethylase Inhibitors. J Am Chem Soc 2010, 132 (9), 3164–3176. 10.1021/ja909996p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1028).Culhane JC; Szewczuk LM; Liu X; Da G; Marmorstein R; Cole PA A Mechanism-Based Inactivator for Histone Demethylase LSD1. J Am Chem Soc 2006, 128 (14), 4536–4537. 10.1021/ja0602748. [DOI] [PubMed] [Google Scholar]
- (1029).Szewczuk LM; Culhane JC; Yang M; Majumdar A; Yu H; Cole PA Mechanistic Analysis of a Suicide Inactivator of Histone Demethylase LSD1. Biochemistry 2007, 46 (23), 6892–6902. 10.1021/bi700414b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1030).Yang M; Culhane JC; Szewczuk LM; Gocke CB; Brautigam CA; Tomchick DR; Machius M; Cole PA; Yu H Structural Basis of Histone Demethylation by LSD1 Revealed by Suicide Inactivation. Nat Struct Mol Biol 2007, 14 (6), 535–539. 10.1038/nsmb1255. [DOI] [PubMed] [Google Scholar]
- (1031).Lee MG; Wynder C; Schmidt DM; McCafferty DG; Shiekhattar R Histone H3 Lysine 4 Demethylation Is a Target of Nonselective Antidepressive Medications. Chem Biol 2006, 13 (6), 563–567. 10.1016/j.chembiol.2006.05.004. [DOI] [PubMed] [Google Scholar]
- (1032).Schmidt DMZ; McCafferty DG Trans-2-Phenylcyclopropylamine Is a Mechanism-Based Inactivator of the Histone Demethylase LSD1. Biochemistry 2007, 46 (14), 4408–4416. 10.1021/bi0618621. [DOI] [PubMed] [Google Scholar]
- (1033).Binda C; Valente S; Romanenghi M; Pilotto S; Cirilli R; Karytinos A; Ciossani G; Botrugno OA; Forneris F; Tardugno M; Edmondson DE; Minucci S; Mattevi A; Mai A Biochemical, Structural, and Biological Evaluation of Tranylcypromine Derivatives as Inhibitors of Histone Demethylases LSD1 and LSD2. J Am Chem Soc 2010, 132 (19), 6827–6833. 10.1021/ja101557k. [DOI] [PubMed] [Google Scholar]
- (1034).Yang M; Culhane JC; Szewczuk LM; Jalili P; Ball HL; Machius M; Cole PA; Yu H Structural Basis for the Inhibition of the LSD1 Histone Demethylase by the Antidepressant Trans-2-Phenylcyclopropylamine. Biochemistry 2007, 46 (27), 8058–8065. 10.1021/bi700664y. [DOI] [PubMed] [Google Scholar]
- (1035).Fioravanti R; Romanelli A; Mautone N; Di Bello E; Rovere A; Corinti D; Zwergel C; Valente S; Rotili D; Botrugno OA; Dessanti P; Vultaggio S; Vianello P; Cappa A; Binda C; Mattevi A; Minucci S; Mercurio C; Varasi M; Mai A Tranylcypromine-Based LSD1 Inhibitors: Structure-Activity Relationships, Antiproliferative Effects in Leukemia, and Gene Target Modulation. ChemMedChem 2020, 15 (7), 643–658. 10.1002/cmdc.201900730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1036).Schulz-Fincke J; Hau M; Barth J; Robaa D; Willmann D; Kürner A; Haas J; Greve G; Haydn T; Fulda S; Lübbert M; Lüdeke S; Berg T; Sippl W; Schüle R; Jung M Structure-Activity Studies on N-Substituted Tranylcypromine Derivatives Lead to Selective Inhibitors of Lysine Specific Demethylase 1 (LSD1) and Potent Inducers of Leukemic Cell Differentiation. Eur J Med Chem 2018, 144, 52–67. 10.1016/j.ejmech.2017.12.001. [DOI] [PubMed] [Google Scholar]
- (1037).Mimasu S; Umezawa N; Sato S; Higuchi T; Umehara T; Yokoyama S Structurally Designed Trans-2-Phenylcyclopropylamine Derivatives Potently Inhibit Histone Demethylase LSD1/KDM1. Biochemistry 2010, 49 (30), 6494–6503. 10.1021/bi100299r. [DOI] [PubMed] [Google Scholar]
- (1038).Vianello P; Botrugno OA; Cappa A; Dal Zuffo R; Dessanti P; Mai A; Marrocco B; Mattevi A; Meroni G; Minucci S; Stazi G; Thaler F; Trifiró P; Valente S; Villa M; Varasi M; Mercurio C Discovery of a Novel Inhibitor of Histone Lysine-Specific Demethylase 1A (KDM1A/LSD1) as Orally Active Antitumor Agent. J Med Chem 2016, 59 (4), 1501–1517. 10.1021/acs.jmedchem.5b01209. [DOI] [PubMed] [Google Scholar]
- (1039).Teresa Borrello M; Benelkebir H; Lee A; Hin Tam C; Shafat M; Rushworth SA; Bowles KM; Douglas L; Duriez PJ; Bailey S; Crabb SJ; Packham G; Ganesan A Synthesis of Carboxamide-Containing Tranylcypromine Analogues as LSD1 (KDM1A) Inhibitors Targeting Acute Myeloid Leukemia. ChemMedChem 2021, 16 (8), 1316–1324. 10.1002/cmdc.202000754. [DOI] [PubMed] [Google Scholar]
- (1040).Ueda R; Suzuki T; Mino K; Tsumoto H; Nakagawa H; Hasegawa M; Sasaki R; Mizukami T; Miyata N Identification of Cell-Active Lysine Specific Demethylase 1-Selective Inhibitors. J Am Chem Soc 2009, 131 (48), 17536–17537. 10.1021/ja907055q. [DOI] [PubMed] [Google Scholar]
- (1041).Jutzi JS; Kleppe M; Dias J; Staehle HF; Shank K; Teruya-Feldstein J; Gambheer SMM; Dierks C; Rienhoff HY; Levine RL; Pahl HL LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. Hemasphere 2018, 2 (3), e54. 10.1097/HS9.0000000000000054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1042).Lee SH; Stubbs M; Liu XM; Diamond M; Dostalik V; Ye M; Lo Y; Favata M; Yang G; Gallagher K; Leffet L; He C; Wu L; Margulis A; Covington M; Wynn R; Yao W; Hollis G; Huber R; Ruggeri B; Scherle P Abstract 4712: Discovery of INCB059872, a Novel FAD-Directed LSD1 Inhibitor That Is Effective in Preclinical Models of Human and Murine AML. Cancer Res 2016, 76 (14_Supplement), 4712–4712. 10.1158/1538-7445.AM2016-4712. [DOI] [Google Scholar]
- (1043).Goethert JR; Gill H; Palandri F; Ross DM; Cochrane T; Larsen SR; Halpern AB; Shortt J; Rossetti JM; Liang J; Marchetti M; Wilson AJ; Innes A; Hanna M; Stevenson WS; Vannucchi AM; Kleppe M; Natsoulis G; Harrison CN; Rienhoff HY Jr. Bomedemstat (IMG-7289), an LSD1 Inhibitor, Manages the Signs and Symptoms of Essential Thrombocythemia (ET) While Reducing the Burden of Cells Homozygous for Driver Mutations. Blood 2023, 142 (Supplement 1), 747–747. 10.1182/blood-2023-179329. [DOI] [Google Scholar]
- (1044).Zhang J; Ren X; Song Y; Yu B JBI-802: The First Orally Available LSD1/HDAC6 Dual Inhibitor to Enter Clinical Trials. Expert Opin Ther Pat 2025, 35, 493–501. 10.1080/13543776.2025.2468792. [DOI] [PubMed] [Google Scholar]
- (1045).Salamero O; Montesinos P; Willekens C; Pérez-Simón JA; Pigneux A; Récher C; Popat R; Carpio C; Molinero C; Mascaró C; Vila J; Arévalo MI; Maes T; Buesa C; Bosch F; Somervaille TCP First-in-Human Phase I Study of Iadademstat (ORY-1001): A First-in-Class Lysine-Specific Histone Demethylase 1A Inhibitor, in Relapsed or Refractory Acute Myeloid Leukemia. J Clin Oncol 2020, 38 (36), 4260–4273. 10.1200/JCO.19.03250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1046).Hollebecque A; Salvagni S; Plummer R; Niccoli P; Capdevila J; Curigliano G; Moreno V; de Braud F; de Villambrosia SG; Martin-Romano P; Baudin E; Arias M; de Alvaro J; Parra-Palau JL; Sánchez-Pérez T; Aronchik I; Filvaroff EH; Lamba M; Nikolova Z; de Bono JS Clinical Activity of CC-90011, an Oral, Potent, and Reversible LSD1 Inhibitor, in Advanced Malignancies. Cancer 2022, 128 (17), 3185–3195. 10.1002/cncr.34366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1047).Hollebecque A; Salvagni S; Plummer R; Isambert N; Niccoli P; Capdevila J; Curigliano G; Moreno V; Martin-Romano P; Baudin E; Arias M; Mora S; de Alvaro J; Di Martino J; Parra-Palau JL; Sánchez-Pérez T; Aronchik I; Filvaroff EH; Lamba M; Nikolova Z; de Bono JS Phase I Study of Lysine-Specific Demethylase 1 Inhibitor, CC-90011, in Patients with Advanced Solid Tumors and Relapsed/Refractory Non-Hodgkin Lymphoma. Clin Cancer Res 2021, 27 (2), 438–446. 10.1158/1078-0432.CCR-20-2380. [DOI] [PubMed] [Google Scholar]
- (1048).Reed DR; Chawla SP; Setty B; Mascarenhas L; Meyers PA; Metts J; Harrison DJ; Loeb D; Crompton BD; Wages DS; Stenehjem DD; Santiesteban DY; Mirza NQ; DuBois SG Phase 1 Trial of Seclidemstat (SP-2577) in Patients with Relapsed/Refractory Ewing Sarcoma. Journal of Clinical Oncology 2021, 39 (15_suppl), 11514–11514. 10.1200/JCO.2021.39.15_suppl.11514. [DOI] [Google Scholar]
- (1049).Salamero O; Molero A; Pérez-Simón JA; Arnan M; Coll R; Garcia-Avila S; Acuña-Cruz E; Cano I; Somervaille TCP; Gutierrez S; Arévalo MI; Xaus J; Buesa C; Limón A; Faller DV; Bosch F; Montesinos P Iadademstat in Combination with Azacitidine in Patients with Newly Diagnosed Acute Myeloid Leukaemia (ALICE): An Open-Label, Phase 2a Dose-Finding Study. Lancet Haematol 2024, 11 (7), e487–e498. 10.1016/S2352-3026(24)00132-7. [DOI] [PubMed] [Google Scholar]
- (1050).Johnston G; Ramsey HE; Liu Q; Wang J; Stengel KR; Sampathi S; Acharya P; Arrate M; Stubbs MC; Burn T; Savona MR; Hiebert SW Nascent Transcript and Single-Cell RNA-Seq Analysis Defines the Mechanism of Action of the LSD1 Inhibitor INCB059872 in Myeloid Leukemia. Gene 2020, 752, 144758. 10.1016/j.gene.2020.144758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1051).Bauer TM; Besse B; Martinez-Marti A; Trigo JM; Moreno V; Garrido P; Ferron-Brady G; Wu Y; Park J; Collingwood T; Kruger RG; Mohammad HP; Ballas MS; Dhar A; Govindan R Phase I, Open-Label, Dose-Escalation Study of the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy of GSK2879552 in Relapsed/Refractory SCLC. J Thorac Oncol 2019, 14 (10), 1828–1838. 10.1016/j.jtho.2019.06.021. [DOI] [PubMed] [Google Scholar]
- (1052).Pettit K; Gerds AT; Yacoub A; Watts JM; Tartaczuch M; Bradley TJ; Shortt J; Stevenson WS; Curtin NJ; Rossetti JM; Burbury K; Natsoulis G; Jones A; Talpaz M; Peppe J; Ross DM; Rienhoff HY A Phase 2a Study of the LSD1 Inhibitor Img-7289 (Bomedemstat) for the Treatment of Myelofibrosis. Blood 2019, 134 (Supplement_1), 556–556. 10.1182/blood-2019-123244. [DOI] [Google Scholar]
- (1053).Cai SF; Chu SH; Goldberg AD; Parvin S; Koche RP; Glass JL; Stein EM; Tallman MS; Sen F; Famulare CA; Cusan M; Huang C-H; Chen C-W; Zou L; Cordner KB; DelGaudio NL; Durani V; Kini M; Rex M; Tian HS; Zuber J; Baslan T; Lowe SW; Rienhoff HY; Letai A; Levine RL; Armstrong SA Leukemia Cell of Origin Influences Apoptotic Priming and Sensitivity to LSD1 Inhibition. Cancer Discov 2020, 10 (10), 1500–1513. 10.1158/2159-8290.CD-19-1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1054).Jasmine S; Mandl A; Krueger TEG; Dalrymple SL; Antony L; Dias J; Celatka CA; Tapper AE; Kleppe M; Kanayama M; Jing Y; Speranzini V; Wang YZ; Luo J; Trock BJ; Denmeade SR; Carducci MA; Mattevi A; Rienhoff HY; Isaacs JT; Brennen WN Characterization of Structural, Biochemical, Pharmacokinetic, and Pharmacodynamic Properties of the LSD1 Inhibitor Bomedemstat in Preclinical Models. Prostate 2024, 84 (10), 909–921. 10.1002/pros.24707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1055).Hiatt JB; Sandborg H; Garrison SM; Arnold HU; Liao S-Y; Norton JP; Friesen TJ; Wu F; Sutherland KD; Rienhoff HY; Martins R; Houghton AM; Srivastava S; MacPherson D Inhibition of LSD1 with Bomedemstat Sensitizes Small Cell Lung Cancer to Immune Checkpoint Blockade and T-Cell Killing. Clin Cancer Res 2022, 28 (20), 4551–4564. 10.1158/1078-0432.CCR-22-1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1056).Lee C; Rudneva VA; Erkek S; Zapatka M; Chau LQ; Tacheva-Grigorova SK; Garancher A; Rusert JM; Aksoy O; Lea R; Mohammad HP; Wang J; Weiss WA; Grimes HL; Pfister SM; Northcott PA; Wechsler-Reya RJ Lsd1 as a Therapeutic Target in Gfi1-Activated Medulloblastoma. Nat Commun 2019, 10 (1), 332. 10.1038/s41467-018-08269-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1057).Fares I; Chagraoui J; Gareau Y; Gingras S; Ruel R; Mayotte N; Csaszar E; Knapp DJHF; Miller P; Ngom M; Imren S; Roy D-C; Watts KL; Kiem H-P; Herrington R; Iscove NN; Humphries RK; Eaves CJ; Cohen S; Marinier A; Zandstra PW; Sauvageau G Cord Blood Expansion. Pyrimidoindole Derivatives Are Agonists of Human Hematopoietic Stem Cell Self-Renewal. Science 2014, 345 (6203), 1509–1512. 10.1126/science.1256337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1058).Subramaniam A; Žemaitis K; Talkhoncheh MS; Yudovich D; Bäckström A; Debnath S; Chen J; Jain MV; Galeev R; Gaetani M; Zubarev RA; Larsson J Lysine-Specific Demethylase 1A Restricts Ex Vivo Propagation of Human HSCs and Is a Target of UM171. Blood 2020, 136 (19), 2151–2161. 10.1182/blood.2020005827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1059).Kanouni T; Severin C; Cho RW; Yuen NY-Y; Xu J; Shi L; Lai C; Del Rosario JR; Stansfield RK; Lawton LN; Hosfield D; O’Connell S; Kreilein MM; Tavares-Greco P; Nie Z; Kaldor SW; Veal JM; Stafford JA; Chen YK Discovery of CC-90011: A Potent and Selective Reversible Inhibitor of Lysine Specific Demethylase 1 (LSD1). J Med Chem 2020, 63 (23), 14522–14529. 10.1021/acs.jmedchem.0c00978. [DOI] [PubMed] [Google Scholar]
- (1060).Sacilotto N; Dessanti P; Lufino MMP; Ortega A; Rodríguez-Gimeno A; Salas J; Maes T; Buesa C; Mascaró C; Soliva R Comprehensive in Vitro Characterization of the LSD1 Small Molecule Inhibitor Class in Oncology. ACS Pharmacol Transl Sci 2021, 4 (6), 1818–1834. 10.1021/acsptsci.1c00223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1061).Chen Z; Zang J; Whetstine J; Hong X; Davrazou F; Kutateladze TG; Simpson M; Mao Q; Pan C-H; Dai S; Hagman J; Hansen K; Shi Y; Zhang G Structural Insights into Histone Demethylation by JMJD2 Family Members. Cell 2006, 125 (4), 691–702. 10.1016/j.cell.2006.04.024. [DOI] [PubMed] [Google Scholar]
- (1062).Fodor BD; Kubicek S; Yonezawa M; O’Sullivan RJ; Sengupta R; Perez-Burgos L; Opravil S; Mechtler K; Schotta G; Jenuwein T Jmjd2b Antagonizes H3K9 Trimethylation at Pericentric Heterochromatin in Mammalian Cells. Genes Dev 2006, 20 (12), 1557–1562. 10.1101/gad.388206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1063).Cloos PAC; Christensen J; Agger K; Maiolica A; Rappsilber J; Antal T; Hansen KH; Helin K The Putative Oncogene GASC1 Demethylates Tri- and Dimethylated Lysine 9 on Histone H3. Nature 2006, 442 (7100), 307–311. 10.1038/nature04837. [DOI] [PubMed] [Google Scholar]
- (1064).Kooistra SM; Helin K Molecular Mechanisms and Potential Functions of Histone Demethylases. Nat Rev Mol Cell Biol 2012, 13 (5), 297–311. 10.1038/nrm3327. [DOI] [PubMed] [Google Scholar]
- (1065).Klose RJ; Yamane K; Bae Y; Zhang D; Erdjument-Bromage H; Tempst P; Wong J; Zhang Y The Transcriptional Repressor JHDM3A Demethylates Trimethyl Histone H3 Lysine 9 and Lysine 36. Nature 2006, 442 (7100), 312–316. 10.1038/nature04853. [DOI] [PubMed] [Google Scholar]
- (1066).Whetstine JR; Nottke A; Lan F; Huarte M; Smolikov S; Chen Z; Spooner E; Li E; Zhang G; Colaiacovo M; Shi Y Reversal of Histone Lysine Trimethylation by the JMJD2 Family of Histone Demethylases. Cell 2006, 125 (3), 467–481. 10.1016/j.cell.2006.03.028. [DOI] [PubMed] [Google Scholar]
- (1067).Tsukada Y; Fang J; Erdjument-Bromage H; Warren ME; Borchers CH; Tempst P; Zhang Y Histone Demethylation by a Family of JmjC Domain-Containing Proteins. Nature 2006, 439 (7078), 811–816. 10.1038/nature04433. [DOI] [PubMed] [Google Scholar]
- (1068).Yamane K; Toumazou C; Tsukada Y; Erdjument-Bromage H; Tempst P; Wong J; Zhang Y JHDM2A, a JmjC-Containing H3K9 Demethylase, Facilitates Transcription Activation by Androgen Receptor. Cell 2006, 125 (3), 483–495. 10.1016/j.cell.2006.03.027. [DOI] [PubMed] [Google Scholar]
- (1069).Xiang Y; Zhu Z; Han G; Lin H; Xu L; Chen CD JMJD3 Is a Histone H3K27 Demethylase. Cell Res 2007, 17 (10), 850–857. 10.1038/cr.2007.83. [DOI] [PubMed] [Google Scholar]
- (1070).De Santa F; Totaro MG; Prosperini E; Notarbartolo S; Testa G; Natoli G The Histone H3 Lysine-27 Demethylase Jmjd3 Links Inflammation to Inhibition of Polycomb-Mediated Gene Silencing. Cell 2007, 130 (6), 1083–1094. 10.1016/j.cell.2007.08.019. [DOI] [PubMed] [Google Scholar]
- (1071).Hong S; Cho Y-W; Yu L-R; Yu H; Veenstra TD; Ge K Identification of JmjC Domain-Containing UTX and JMJD3 as Histone H3 Lysine 27 Demethylases. Proc Natl Acad Sci U S A 2007, 104 (47), 18439–18444. 10.1073/pnas.0707292104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1072).Ntziachristos P; Tsirigos A; Welstead GG; Trimarchi T; Bakogianni S; Xu L; Loizou E; Holmfeldt L; Strikoudis A; King B; Mullenders J; Becksfort J; Nedjic J; Paietta E; Tallman MS; Rowe JM; Tonon G; Satoh T; Kruidenier L; Prinjha R; Akira S; Van Vlierberghe P; Ferrando AA; Jaenisch R; Mullighan CG; Aifantis I Contrasting Roles of Histone 3 Lysine 27 Demethylases in Acute Lymphoblastic Leukaemia. Nature 2014, 514 (7523), 513–517. 10.1038/nature13605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1073).Agger K; Cloos PAC; Christensen J; Pasini D; Rose S; Rappsilber J; Issaeva I; Canaani E; Salcini AE; Helin K UTX and JMJD3 Are Histone H3K27 Demethylases Involved in HOX Gene Regulation and Development. Nature 2007, 449 (7163), 731–734. 10.1038/nature06145. [DOI] [PubMed] [Google Scholar]
- (1074).Hashizume R; Andor N; Ihara Y; Lerner R; Gan H; Chen X; Fang D; Huang X; Tom MW; Ngo V; Solomon D; Mueller S; Paris PL; Zhang Z; Petritsch C; Gupta N; Waldman TA; James CD Pharmacologic Inhibition of Histone Demethylation as a Therapy for Pediatric Brainstem Glioma. Nat Med 2014, 20 (12), 1394–1396. 10.1038/nm.3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1075).Kruidenier L; Chung C; Cheng Z; Liddle J; Che K; Joberty G; Bantscheff M; Bountra C; Bridges A; Diallo H; Eberhard D; Hutchinson S; Jones E; Katso R; Leveridge M; Mander PK; Mosley J; Ramirez-Molina C; Rowland P; Schofield CJ; Sheppard RJ; Smith JE; Swales C; Tanner R; Thomas P; Tumber A; Drewes G; Oppermann U; Patel DJ; Lee K; Wilson DM A Selective Jumonji H3K27 Demethylase Inhibitor Modulates the Proinflammatory Macrophage Response. Nature 2012, 488 (7411), 404–408. 10.1038/nature11262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1076).Cao Z; Shi X; Tian F; Fang Y; Wu JB; Mrdenovic S; Nian X; Ji J; Xu H; Kong C; Xu Y; Chen X; Huang Y; Wei X; Yu Y; Yang B; Chung LWK; Wang F KDM6B Is an Androgen Regulated Gene and Plays Oncogenic Roles by Demethylating H3K27me3 at Cyclin D1 Promoter in Prostate Cancer. Cell Death Dis 2021, 12 (1), 2. 10.1038/s41419-020-03354-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1077).Christensen J; Agger K; Cloos PAC; Pasini D; Rose S; Sennels L; Rappsilber J; Hansen KH; Salcini AE; Helin K RBP2 Belongs to a Family of Demethylases, Specific for Tri-and Dimethylated Lysine 4 on Histone 3. Cell 2007, 128 (6), 1063–1076. 10.1016/j.cell.2007.02.003. [DOI] [PubMed] [Google Scholar]
- (1078).Lin W; Cao J; Liu J; Beshiri ML; Fujiwara Y; Francis J; Cherniack AD; Geisen C; Blair LP; Zou MR; Shen X; Kawamori D; Liu Z; Grisanzio C; Watanabe H; Minamishima YA; Zhang Q; Kulkarni RN; Signoretti S; Rodig SJ; Bronson RT; Orkin SH; Tuck DP; Benevolenskaya EV; Meyerson M; Kaelin WG; Yan Q Loss of the Retinoblastoma Binding Protein 2 (RBP2) Histone Demethylase Suppresses Tumorigenesis in Mice Lacking Rb1 or Men1. Proc Natl Acad Sci U S A 2011, 108 (33), 13379–13386. 10.1073/pnas.1110104108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1079).Catchpole S; Spencer-Dene B; Hall D; Santangelo S; Rosewell I; Guenatri M; Beatson R; Scibetta AG; Burchell JM; Taylor-Papadimitriou J PLU-1/JARID1B/KDM5B Is Required for Embryonic Survival and Contributes to Cell Proliferation in the Mammary Gland and in ER+ Breast Cancer Cells. Int J Oncol 2011, 38 (5), 1267–1277. 10.3892/ijo.2011.956. [DOI] [PubMed] [Google Scholar]
- (1080).Yamane K; Tateishi K; Klose RJ; Fang J; Fabrizio LA; Erdjument-Bromage H; Taylor-Papadimitriou J; Tempst P; Zhang Y PLU-1 Is an H3K4 Demethylase Involved in Transcriptional Repression and Breast Cancer Cell Proliferation. Mol Cell 2007, 25 (6), 801–812. 10.1016/j.molcel.2007.03.001. [DOI] [PubMed] [Google Scholar]
- (1081).Dai B; Huang H; Guan F; Zhu G; Xiao Z; Mao B; Su H; Hu Z Histone Demethylase KDM5A Inhibits Glioma Cells Migration and Invasion by down Regulating ZEB1. Biomed Pharmacother 2018, 99, 72–80. 10.1016/j.biopha.2018.01.020. [DOI] [PubMed] [Google Scholar]
- (1082).Feng T; Wang Y; Lang Y; Zhang Y KDM5A Promotes Proliferation and EMT in Ovarian Cancer and Closely Correlates with PTX Resistance. Mol Med Rep 2017, 16 (3), 3573–3580. 10.3892/mmr.2017.6960. [DOI] [PubMed] [Google Scholar]
- (1083).Zeng J; Ge Z; Wang L; Li Q; Wang N; Björkholm M; Jia J; Xu D The Histone Demethylase RBP2 Is Overexpressed in Gastric Cancer and Its Inhibition Triggers Senescence of Cancer Cells. Gastroenterology 2010, 138 (3), 981–992. 10.1053/j.gastro.2009.10.004. [DOI] [PubMed] [Google Scholar]
- (1084).Yamamoto S; Wu Z; Russnes HG; Takagi S; Peluffo G; Vaske C; Zhao X; Moen Vollan HK; Maruyama R; Ekram MB; Sun H; Kim JH; Carver K; Zucca M; Feng J; Almendro V; Bessarabova M; Rueda OM; Nikolsky Y; Caldas C; Liu XS; Polyak K JARID1B Is a Luminal Lineage-Driving Oncogene in Breast Cancer. Cancer Cell 2014, 25 (6), 762–777. 10.1016/j.ccr.2014.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1085).Banelli B; Carra E; Barbieri F; Würth R; Parodi F; Pattarozzi A; Carosio R; Forlani A; Allemanni G; Marubbi D; Florio T; Daga A; Romani M The Histone Demethylase KDM5A Is a Key Factor for the Resistance to Temozolomide in Glioblastoma. Cell Cycle 2015, 14 (21), 3418–3429. 10.1080/15384101.2015.1090063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1086).Hinohara K; Wu H-J; Vigneau S; McDonald TO; Igarashi KJ; Yamamoto KN; Madsen T; Fassl A; Egri SB; Papanastasiou M; Ding L; Peluffo G; Cohen O; Kales SC; Lal-Nag M; Rai G; Maloney DJ; Jadhav A; Simeonov A; Wagle N; Brown M; Meissner A; Sicinski P; Jaffe JD; Jeselsohn R; Gimelbrant AA; Michor F; Polyak K KDM5 Histone Demethylase Activity Links Cellular Transcriptomic Heterogeneity to Therapeutic Resistance. Cancer Cell 2018, 34 (6), 939–953.e9. 10.1016/j.ccell.2018.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1087).Roesch A; Vultur A; Bogeski I; Wang H; Zimmermann KM; Speicher D; Körbel C; Laschke MW; Gimotty PA; Philipp SE; Krause E; Pätzold S; Villanueva J; Krepler C; Fukunaga-Kalabis M; Hoth M; Bastian BC; Vogt T; Herlyn M Overcoming Intrinsic Multidrug Resistance in Melanoma by Blocking the Mitochondrial Respiratory Chain of Slow-Cycling JARID1B(High) Cells. Cancer Cell 2013, 23 (6), 811–825. 10.1016/j.ccr.2013.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1088).Sharma SV; Lee DY; Li B; Quinlan MP; Takahashi F; Maheswaran S; McDermott U; Azizian N; Zou L; Fischbach MA; Wong K-K; Brandstetter K; Wittner B; Ramaswamy S; Classon M; Settleman J A Chromatin-Mediated Reversible Drug-Tolerant State in Cancer Cell Subpopulations. Cell 2010, 141 (1), 69–80. 10.1016/j.cell.2010.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1089).Liau BB; Sievers C; Donohue LK; Gillespie SM; Flavahan WA; Miller TE; Venteicher AS; Hebert CH; Carey CD; Rodig SJ; Shareef SJ; Najm FJ; van Galen P; Wakimoto H; Cahill DP; Rich JN; Aster JC; Suvà ML; Patel AP; Bernstein BE Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. Cell Stem Cell 2017, 20 (2), 233–246.e7. 10.1016/j.stem.2016.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1090).Vinogradova M; Gehling VS; Gustafson A; Arora S; Tindell CA; Wilson C; Williamson KE; Guler GD; Gangurde P; Manieri W; Busby J; Flynn EM; Lan F; Kim H-J; Odate S; Cochran AG; Liu Y; Wongchenko M; Yang Y; Cheung TK; Maile TM; Lau T; Costa M; Hegde GV; Jackson E; Pitti R; Arnott D; Bailey C; Bellon S; Cummings RT; Albrecht BK; Harmange J-C; Kiefer JR; Trojer P; Classon M An Inhibitor of KDM5 Demethylases Reduces Survival of Drug-Tolerant Cancer Cells. Nat Chem Biol 2016, 12 (7), 531–538. 10.1038/nchembio.2085. [DOI] [PubMed] [Google Scholar]
- (1091).Yang G-J; Ko C-N; Zhong H-J; Leung C-H; Ma D-L Structure-Based Discovery of a Selective KDM5A Inhibitor That Exhibits Anti-Cancer Activity via Inducing Cell Cycle Arrest and Senescence in Breast Cancer Cell Lines. Cancers (Basel) 2019, 11 (1), 92. 10.3390/cancers11010092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1092).Zhao B; Liang Q; Ren H; Zhang X; Wu Y; Zhang K; Ma L-Y; Zheng Y-C; Liu H-M Discovery of Pyrazole Derivatives as Cellular Active Inhibitors of Histone Lysine Specific Demethylase 5B (KDM5B/JARID1B). Eur J Med Chem 2020, 192, 112161. 10.1016/j.ejmech.2020.112161. [DOI] [PubMed] [Google Scholar]
- (1093).Horton JR; Liu X; Gale M; Wu L; Shanks JR; Zhang X; Webber PJ; Bell JSK; Kales SC; Mott BT; Rai G; Jansen DJ; Henderson MJ; Urban DJ; Hall MD; Simeonov A; Maloney DJ; Johns MA; Fu H; Jadhav A; Vertino PM; Yan Q; Cheng X Structural Basis for KDM5A Histone Lysine Demethylase Inhibition by Diverse Compounds. Cell Chem Biol 2016, 23 (7), 769–781. 10.1016/j.chembiol.2016.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1094).Fu Y-D; Huang M-J; Guo J-W; You Y-Z; Liu H-M; Huang L-H; Yu B Targeting Histone Demethylase KDM5B for Cancer Treatment. Eur J Med Chem 2020, 208, 112760. 10.1016/j.ejmech.2020.112760. [DOI] [PubMed] [Google Scholar]
- (1095).Johansson C; Velupillai S; Tumber A; Szykowska A; Hookway ES; Nowak RP; Strain-Damerell C; Gileadi C; Philpott M; Burgess-Brown N; Wu N; Kopec J; Nuzzi A; Steuber H; Egner U; Badock V; Munro S; LaThangue NB; Westaway S; Brown J; Athanasou N; Prinjha R; Brennan PE; Oppermann U Structural Analysis of Human KDM5B Guides Histone Demethylase Inhibitor Development. Nat Chem Biol 2016, 12 (7), 539–545. 10.1038/nchembio.2087. [DOI] [PubMed] [Google Scholar]
- (1096).Tumber A; Nuzzi A; Hookway ES; Hatch SB; Velupillai S; Johansson C; Kawamura A; Savitsky P; Yapp C; Szykowska A; Wu N; Bountra C; Strain-Damerell C; Burgess-Brown NA; Ruda GF; Fedorov O; Munro S; England KS; Nowak RP; Schofield CJ; La Thangue NB; Pawlyn C; Davies F; Morgan G; Athanasou N; Müller S; Oppermann U; Brennan PE Potent and Selective KDM5 Inhibitor Stops Cellular Demethylation of H3K4me3 at Transcription Start Sites and Proliferation of MM1S Myeloma Cells. Cell Chem Biol 2017, 24 (3), 371–380. 10.1016/j.chembiol.2017.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1097).Liu Y; Liu K; Qin S; Xu C; Min J Epigenetic Targets and Drug Discovery: Part 1: Histone Methylation. Pharmacol Ther 2014, 143 (3), 275–294. 10.1016/j.pharmthera.2014.03.007. [DOI] [PubMed] [Google Scholar]
- (1098).Poulard C; Noureddine LM; Pruvost L; Le Romancer M Structure, Activity, and Function of the Protein Lysine Methyltransferase G9a. Life (Basel) 2021, 11 (10), 1082. 10.3390/life11101082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1099).Zhang J; Wang Y; Shen Y; He P; Ding J; Chen Y G9a Stimulates CRC Growth by Inducing P53 Lys373 Dimethylation-Dependent Activation of Plk1. Theranostics 2018, 8 (10), 2884–2895. 10.7150/thno.23824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1100).McGarvey KM; Fahrner JA; Greene E; Martens J; Jenuwein T; Baylin SB Silenced Tumor Suppressor Genes Reactivated by DNA Demethylation Do Not Return to a Fully Euchromatic Chromatin State. Cancer Res 2006, 66 (7), 3541–3549. 10.1158/0008-5472.CAN-05-2481. [DOI] [PubMed] [Google Scholar]
- (1101).Watanabe H; Soejima K; Yasuda H; Kawada I; Nakachi I; Yoda S; Naoki K; Ishizaka A Deregulation of Histone Lysine Methyltransferases Contributes to Oncogenic Transformation of Human Bronchoepithelial Cells. Cancer Cell Int 2008, 8, 15. 10.1186/1475-2867-8-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1102).Seligson DB; Horvath S; McBrian MA; Mah V; Yu H; Tze S; Wang Q; Chia D; Goodglick L; Kurdistani SK Global Levels of Histone Modifications Predict Prognosis in Different Cancers. Am J Pathol 2009, 174 (5), 1619–1628. 10.2353/ajpath.2009.080874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1103).Ellinger J; Kahl P; von der Gathen J; Rogenhofer S; Heukamp LC; Gütgemann I; Walter B; Hofstädter F; Büttner R; Müller SC; Bastian PJ; von Ruecker A Global Levels of Histone Modifications Predict Prostate Cancer Recurrence. Prostate 2010, 70 (1), 61–69. 10.1002/pros.21038. [DOI] [PubMed] [Google Scholar]
- (1104).Nachiyappan A; Gupta N; Taneja R EHMT1/EHMT2 in EMT, Cancer Stemness and Drug Resistance: Emerging Evidence and Mechanisms. FEBS J 2022, 289 (5), 1329–1351. 10.1111/febs.16334. [DOI] [PubMed] [Google Scholar]
- (1105).Dong C; Wu Y; Yao J; Wang Y; Yu Y; Rychahou PG; Evers BM; Zhou BP G9a Interacts with Snail and Is Critical for Snail-Mediated E-Cadherin Repression in Human Breast Cancer. J Clin Invest 2012, 122 (4), 1469–1486. 10.1172/JCI57349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1106).Kondo Y; Shen L; Ahmed S; Boumber Y; Sekido Y; Haddad BR; Issa J-PJ Downregulation of Histone H3 Lysine 9 Methyltransferase G9a Induces Centrosome Disruption and Chromosome Instability in Cancer Cells. PLoS One 2008, 3 (4), e2037. 10.1371/journal.pone.0002037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1107).Cao H; Li L; Yang D; Zeng L; Yewei X; Yu B; Liao G; Chen J Recent Progress in Histone Methyltransferase (G9a) Inhibitors as Anticancer Agents. Eur J Med Chem 2019, 179, 537–546. 10.1016/j.ejmech.2019.06.072. [DOI] [PubMed] [Google Scholar]
- (1108).Liu F; Barsyte-Lovejoy D; Li F; Xiong Y; Korboukh V; Huang X-P; Allali-Hassani A; Janzen WP; Roth BL; Frye SV; Arrowsmith CH; Brown PJ; Vedadi M; Jin J Discovery of an in Vivo Chemical Probe of the Lysine Methyltransferases G9a and GLP. J Med Chem 2013, 56 (21), 8931–8942. 10.1021/jm401480r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1109).Milite C; Feoli A; Horton JR; Rescigno D; Cipriano A; Pisapia V; Viviano M; Pepe G; Amendola G; Novellino E; Cosconati S; Cheng X; Castellano S; Sbardella G Discovery of a Novel Chemotype of Histone Lysine Methyltransferase EHMT1/2 (GLP/G9a) Inhibitors: Rational Design, Synthesis, Biological Evaluation, and Co-Crystal Structure. J Med Chem 2019, 62 (5), 2666–2689. 10.1021/acs.jmedchem.8b02008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1110).Ting PY; Borikar S; Kerrigan JR; Thomsen NM; Aghania E; Hinman AE; Reyes A; Pizzato N; Fodor BD; Wu F; Belew MS; Mao X; Wang J; Chitnis S; Niu W; Hachey A; Cobb JS; Savage NA; Burke A; Paulk J; Dovala D; Lin J; Clifton MC; Ornelas E; Ma X; Ware NF; Sanchez CC; Taraszka J; Terranova R; Knehr J; Altorfer M; Barnes SW; Beckwith REJ; Solomon JM; Dales NA; Patterson AW; Wagner J; Bouwmeester T; Dranoff G; Stevenson SC; Bradner JE A Molecular Glue Degrader of the WIZ Transcription Factor for Fetal Hemoglobin Induction. Science 2024, 385 (6704), 91–99. 10.1126/science.adk6129. [DOI] [PubMed] [Google Scholar]
- (1111).Renneville A; Van Galen P; Canver MC; McConkey M; Krill-Burger JM; Dorfman DM; Holson EB; Bernstein BE; Orkin SH; Bauer DE; Ebert BL EHMT1 and EHMT2 Inhibition Induces Fetal Hemoglobin Expression. Blood 2015, 126 (16), 1930–1939. 10.1182/blood-2015-06-649087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1112).Krivega I; Byrnes C; de Vasconcellos JF; Lee YT; Kaushal M; Dean A; Miller JL Inhibition of G9a Methyltransferase Stimulates Fetal Hemoglobin Production by Facilitating LCR/γ-Globin Looping. Blood 2015, 126 (5), 665–672. 10.1182/blood-2015-02-629972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1113).Ribich S; Harvey D; Copeland RA Drug Discovery and Chemical Biology of Cancer Epigenetics. Cell Chem Biol 2017, 24 (9), 1120–1147. 10.1016/j.chembiol.2017.08.020. [DOI] [PubMed] [Google Scholar]
- (1114).Shortt J; Ott CJ; Johnstone RW; Bradner JE A Chemical Probe Toolbox for Dissecting the Cancer Epigenome. Nat Rev Cancer 2017, 17 (3), 160–183. 10.1038/nrc.2016.148. [DOI] [PubMed] [Google Scholar]
- (1115).Mabe NW; Perry JA; Malone CF; Stegmaier K Pharmacological Targeting of the Cancer Epigenome. Nat Cancer 2024, 5 (6), 844–865. 10.1038/s43018-024-00777-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (1116).Sakamoto KM; Kim KB; Kumagai A; Mercurio F; Crews CM; Deshaies RJ Protacs: Chimeric Molecules That Target Proteins to the Skp1-Cullin-F Box Complex for Ubiquitination and Degradation. Proc Natl Acad Sci U S A 2001, 98 (15), 8554–8559. 10.1073/pnas.141230798. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
