Skip to main content
Epigenomics logoLink to Epigenomics
. 2024 Nov 27;17(1):59–74. doi: 10.1080/17501911.2024.2430169

Cancer epigenetic therapy: recent advances, challenges, and emerging opportunities

Rajita Vatapalli a,, Alex P Rossi a,b, Ho Man Chan a, Jingwen Zhang a,
PMCID: PMC11702999  PMID: 39601374

ABSTRACT

Epigenetic dysregulation is an important nexus in the development and maintenance of human cancers. This review provides an overview of how understanding epigenetic dysregulation in cancers has led to insights for novel cancer therapy development. Over the past two decades, significant strides have been made in drug discovery efforts targeting cancer epigenetic mechanisms, leading to successes in clinical development and approval of cancer epigenetic therapeutics. This article will discuss the current therapeutic rationale guiding the discovery and development of epigenetic therapeutics, key learnings from clinical experiences and new opportunities on the horizon.

KEYWORDS: Cancer epigenetics, histone modifications, DNA modifications, synthetic lethality, drug combination, drug discovery

1. Introduction

Epigenetics is the study of heritable changes in gene function that are not coded in DNA sequences [1,2]. The human body encompasses trillions of cells, and each cell contains an identical genetic blueprint encoded in DNA. The two-meter-long DNA is highly compacted into a chromatin structure in the cell nucleus. Epigenetic modifications, which involve alterations in gene expression without changes to the underlying DNA sequences, play a critical role in normal development and disease progression. Numerous modifications to DNA and histone proteins are now characterized to form an epigenetic code that regulates chromatin accessibility for transcription, replication, and DNA damage repair [3]. Dysregulation of epigenetic regulatory mechanisms leads to many human diseases, including developmental disorders, neurological diseases, autoimmune diseases, and cancers [4]. In this review, we will discuss epigenetic dysregulation in cancers, opportunities to develop precision therapeutics targeting cancer-specific epigenetic mechanisms and highlight recent progress made in the drug discovery field. Due to space limitation, we will focus our discussion on therapies undergoing active clinical investigation. We will also share our perspectives on a few potential future avenues in developing the next wave of cancer epigenetic therapies.

2. Cancer epigenetic drug discovery

Cancer epigenetics represents a promising frontier in the field of oncology drug discovery, offering new avenues for targeted therapies against cancers. There have been significant efforts to develop novel chemical inhibitors targeting protein families which regulate DNA and histone modifications. Most notable of these targets include DNA cytosine methylation, histone lysine and arginine methylation, and histone lysine acetylation and “reader” protein families that bind to specific DNA and histone modifications. The aim is to deliver anti-tumor activity and to enhance overall cancer treatment efficacy by specifically targeting and modulating these epigenetic regulators. Through a combination of innovative drug screening approaches and advanced drug design strategies (Figure 1), the quest for effective cancer epigenetic drugs holds significant promise in delivering new cancer treatment paradigms.

Figure 1.

Figure 1.

Drug discovery targeting different epigenetic target classes. (a) Proteins involved in epigenetic regulation can deposit (writers), detect (readers), and remove (erasers) the various covalent modifications on DNA and histones [5]. Direct methylation of DNA and post-translational modifications on histones are two major epigenetic mechanisms involved in regulating gene expression. The complex interplay between these factors controls genomic accessibility, which in turn regulates expression of genes involved in a variety of cellular processes. Histone acetylation and methylation networks constitute a large component of the human epigenome and have been the focus of small molecule cancer epigenetic drug discovery in the past two decades. Selected examples of inhibitors targeting writers, readers and erasers are illustrated. (b) Chromatin remodeling enzymes are responsible for the rearrangement, displacement, and replacement of nucleosomes on the chromatin. Currently, there are successful examples of small-molecule inhibitors and proteolysis-targeting chimeras (PROTACs) targeting this enzyme class. Created in BioRender. Vatapalli, R. (2024) BioRender.com/t18f526.

2.1. Reverse dysregulated epigenetic and transcriptional programs in cancers

All cancers have highly dysregulated epigenetic landscapes and transcriptional signatures [6,7]. For example, cancer cells generally exhibit focal DNA hypermethylation in gene promoters, accompanied by global DNA hypomethylation. Transcriptomic profiling of human cancers has shown heterogeneous transcriptomic features, including features reminiscent of early progenitor cell development signature, or epithelial–mesenchymal transition (EMT) gene signature or embryonic stem cell-like gene signature [8–10]. Disruption of normal gene expression contributes to tumor initiation, progression, and resistance to therapies. One of the earliest concepts of cancer epigenetic therapy aims to restore normal gene expression. While genetic alterations and mutations are largely irreversible, it was hypothesized that epigenetic changes are more readily reversible with small-molecule therapeutics [11,12].

Inhibitors of DNA methyltransferase (DNMT) and histone deacetylase (HDAC) were developed to restore epigenetic imbalance in cancers. These agents led to the initial clinical successes of cancer epigenetic therapy, with FDA approval of DNMT inhibitors azacytidine and decitabine for the treatment of myelodysplastic syndromes (MDS) and leukemia, and HDAC inhibitor vorinostat (also known as suberoylanilide hydroxamic acid (SAHA)) for the treatment of cutaneous T cell lymphoma (CTCL). The first generation of DNMT and HDAC inhibitors were developed prior to the concept of targeted drug discovery, and they are not selective against close family members and carry off-target activities. Azacytidine, a nucleoside analog, was first synthesized by Sorm and colleagues in the 1960s as a cytotoxic chemotherapeutic agent [13]. The discovery and development of DNMT inhibitors have been extensively reviewed [14,15]. Both azacytidine and decitabine are prodrugs. After uptake into the cell via nucleoside transporters, these molecules undergo a series of phosphorylation modifications, are converted to cytidine triphosphate (CTP) and deoxycytidine triphosphate (dCTP) analogs and then get incorporated into DNA and RNA. Azacytidine and decitabine can directly cause DNA damage by forming covalent adducts with DNMTs on DNA. The “trapped” DNMTs are degraded by a proteasome-mediated mechanism leading to genome-wide DNA hypomethylation [14]. Peter Jones and colleagues in 1980 made the seminal observation that azacitidine can alter the differentiated state of cultured cells and inhibit the methylation state of newly synthesized DNA [16]. DNMT inhibitors are typically used at low doses in the clinic to inhibit DNA methylation following a few rounds of cell cycle progression. At high doses, they cause acute cytotoxicity typical of chemo agents and mask the desirable effect of reprogramming the DNA methylation state. There has been continued interest in developing reversible, isoform selective DNMTi to improve therapeutic index [17]. GSK-3685032 is a novel first-in-class reversible DNMT1 selective inhibitor showing great promise in preclinical studies [17]. The highly selective DNMT inhibitors are much better chemical tools to selectively modulate DNA methylation levels in cells and to evaluate therapeutic hypotheses of inhibiting DNMT1. Thus far, no clinical study has been reported with this new generation of reversible and isoform selective DNMT1 inhibitors.

HDACs catalyze the removal of acetyl groups from lysine residues on histones [18]. These enzymes modify chromatin structure and contribute to aberrant gene expression in cancers. Therefore, targeting HDACs to restore normal cellular gene expression were actively investigated as a potential anticancer therapeutic strategy [19]. All approved HDAC inhibitors (HDACi), including vorinostat, belinostat, romidepsin, and panobinostat, have broad activities to elevate global histone acetylation by inhibiting multiple HDACs within the Zn2+ containing HDAC subfamily (HDAC1–11) [5,20]. Rich sources of natural products have greatly accelerated the discovery and development of HDAC inhibitors. Trichostatin A and trapoxin were among the first natural products verified to inhibit HDACs [21]. Cyclic depsipeptides, such as FK228 (also known as romidepsin), are a natural product originally isolated from the bacterium Chromobacterium violaceum that also potently inhibits HDACs [22]. HDACi demonstrated potent cytotoxic activity against human cancer cell lines, inducing differentiation and apoptosis of leukemic cell lines. Extensive preclinical and clinical investigations eventually led to multiple FDA approvals, including 1) vorinostat for the treatment of CTCL in 2006; 2) romidepsin for the treatment of CTCL in 2009 and peripheral T-cell lymphoma (PTCL) in 2011; 3) belinostat for the treatment of PTCL in 2014; and 4) panobinostat for the treatment of multiple myeloma (MM) in combination with other drugs in 2015. In 2019, tucidinostat (also known as Chidamide) was approved by China’s National Medical Products Administration for the treatment of breast cancer. This is the only HDAC inhibitor approved to date for a solid cancer treatment [23–25]. For details on the discovery and development of HDACi, please see reviews [26–28].

Both DNMT and HDAC inhibitors have broad mechanisms of action with pleiotropic toxicity in normal tissue, which has limited their clinical utility. Overall, these agents have a narrow therapeutic index. FDA approval is one measure of success; however, the overall clinical benefit of these agents has been fairly modest. Patients treated with pan-HDAC inhibitors have experienced severe adverse events such as fatigue or asthenia that lack a robust mitigation strategy. Ongoing efforts to develop more targeted and isoform-selective DNMT and HDAC inhibitors may overcome some of the challenges associated with the early generation of DNMT and HDAC inhibitors [29]. Beyond drug discovery efforts, better patient selection strategies are needed to maximize therapeutic benefit by identifying and enriching for responder populations. A comprehensive review of the ongoing clinical investigation on HDAC and DNMT inhibitors can be found here [30–32].

2.2. Precision targeted epigenetic therapeutics

Targeted cancer therapy has transformed cancer treatment where oncogenic driver proteins can be identified and selectively targeted by small molecule drugs [33,34]. Precision targeted cancer drugs are designed to specifically target molecular and genetic alterations in cancers. This approach helps improve tumor treatment efficacy and minimize normal tissue toxicities, potentially leading to fewer side effects. Key characteristics of precision targeted medicine include 1) identification of specific tumor biomarkers for patient selection; 2) personalized treatment matching patient with specific biomarkers with corresponding targeted therapies; and 3) therapeutic agents are highly selective against the protein drug targets of interest.

Large-scale cancer genome sequencing efforts have uncovered numerous genetic mutations of epigenetic regulators, adding new evidence that altered epigenetic regulatory mechanisms are a major hallmark of cancer [35]. Cancer genome sequencing projects have uncovered many alterations of epigenetic regulators in cancers, which fueled multiple waves of targeted drug discovery efforts to systematically target emerging enzyme classes important for epigenetic regulation, including protein methyltransferases, demethylases, acetyltransferases, DNA/RNA helicases, and numerous histone code reader family proteins [36].

Oncogenic driver mutations of epigenetic regulators are frequently observed in hematological cancers and rare pediatric cancers, including 1) hotspot gain-of-function (GoF) mutation of enhancer of zeste homolog 2 (EZH2) in lymphoma [37]; 2) chromosomal translocation of mixed lineage leukemia protein-1 (MLL1) in leukemia [38]; 3) chromosomal translocation and hotspot gain-of-function mutation of nuclear receptor binding SET domain protein 2 (NSD2) in multiple myeloma and acute lymphoblastic leukemia [39,40]; 4) chromosomal translocation of bromodomain-containing protein 3 (BRD3) and BRD4 in NUT midline carcinoma [41]; 5) chromosomal translocation of SS18 in synovial sarcoma [42]; and 6) the well-studied gain-of-function mutation in isocitrate dehydrogenase 1 (IDH1) and IDH2 in leukemia and glioma [43–45] and many more. Histones themselves are also mutated and directly linked to oncogenesis [46,47]. Missense mutations in histone H3 always occur at or near well-characterized regulatory residues that impact its post-translational modifications. For example, H3K27M mutation is found in nearly 80% of pediatric diffuse intrinsic pontine glioma (DIPG) and these mutations diminish global H3K27 trimethylation by locally trapping and inactivating the polycomb repressive complex 2 (PRC2) [48]. In most cases of DIPG, H3K27M, or H3G34V mutations are the only genetic alterations identified in the patient. H3K36M mutation is found in nearly 90% of chondroblastoma and this alteration suppresses global H3K36 methylation [49]. All histone mutations occurring in cancers have been comprehensively reviewed [50,51]. The identification of epigenetic driver oncogenes provides a strong rationale for precision-focused cancer epigenetic drug discovery. In this next section, we will discuss recent advancements in targeting epigenetic regulators as oncogenic drivers, as well as novel strategies to target loss-of-function alterations of epigenetic regulators.

2.2.1. Direct targeting of oncogenic drivers

There have been extensive efforts to develop novel therapeutics that target oncogenic driver mutations of epigenetic regulators. A number of these efforts have led to active clinical investigations (Table 1). The first success with this strategy is the FDA approval of EZH2 inhibitor tazemetostat for the treatment of follicular lymphoma (FL) in 2020 [52]. EZH2 is a member of the Polycomb-group (PcG) proteins which maintain the transcriptional repressive state by methylating histone H3 lysine 27 [53]. These proteins play a key role in regulating embryonic stem cell differentiation, renewal, cell fate decision, and the normal cellular differentiation program. Mutation of EZH2 and the PRC2 complex subunits are observed in multiple cancers, and they can be oncogenic or tumor suppressive depending on the cancer type. Oncogenic EZH2 hotspot, missense point mutations (including Y641, Y677, Y687) are concentrated in FL and diffuse large B cell lymphoma (DLBLC) of the GCB subtype [37,54–57]. Elegant biochemical investigation has demonstrated that these hotspot mutations result in neomorphic biochemical and cellular properties. While the EZH2 wild type protein is a very efficient enzyme in converting H3K27me0 to H3K27me1/2, most of the mutated EZH2 proteins showed elevated enzymatic activity that resulted in an elevated level of H3K27me3 [37,55]. Cancer cells containing EZH2 hotspot mutation display genome-wide hyper-H3K27 trimethylation. EZH2 inhibitors and EED inhibitors demonstrated profound anti-tumor activity in preclinical models, which promoted their clinical development [58–60]. Tazemetostat had an overall response rate (ORR) of 69% in EZH2 mutant FL patients, which led to its approval in FL [52]. Interestingly, tazemetostat is also efficacious in a subset of EZH2 wild type FL patients, with an ORR of 34% [61].

Table 1.

Inhibitors targeting epigenetic oncogenic drivers in clinical development (active trials only).

Target Biomarkers Inhibitors Indication Clinical trial number
EZH2 EZH2 GOF mutation Tazemetostat, GSK2816126, PF-06821497, FTX-6058, SHR2554) B-cell Non-Hodgkin lymphoma, follicular lymphoma, solid tumors NCT03456726
NCT05228158
NCT03603951
NCT03213665
NCT05467943
NCT04104776
NCT05551936
NCT04224493
  Tazemetostat, SHR2554, PF-06821497, Valmetostat,
HH-2853
PTCL, MPNST, Breast cancer, TNBC, mCRPC, R/R B-cell NHL, R/R ATL NCT05559008
NCT04355858
NCT03805399
NCT06122389
NCT04917042
NCT03460977
NCT04703192
NCT04842877
NCT04102150
NCT05633969
NCT06244485
NCT03930953
NCT04390737
EED EZH2 GOF mutation MAK-683, APG-5918, ORIC-944 DLBCL, solid tumors NCT02900651
NCT05415098.
NCT05413421
Menin MLL rearrangements/
NPM1m/FLT3m
BMF-219, SNDX-5613,
KO-539, DSP-5336
JNJ-75276617
Leukemia, R/R Leukemias,
MRD-positive AML
NCT05153330
NCT04065399
NCT04067336
NCT04811560
NCT04988555
NCT05326516
NCT05453903
NCT05521087
NCT05406817
NCT05761171
NCT04065399
NCT05360160
NCT06284486
NCT06226571
NCT05886049
NCT06222580
NCT05735184
NCT06001788
NCT04988555
KRAS mutation BMF-219 CRC, NSCLC, PDAC NCT05631574
Hox upregulation SNDX-5613 Leukemias NCT06229912
  SNDX-5613 AML, Pediatric AML, CRC, Solid tumors NCT06313437
NCT06177067
NCT05731947
NSD2 NSD2 translocation KTX-1001 Multiple myeloma NCT05651932
SMARCA2/4   FHD-286 Metastatic uveal melanoma, AML, MDS NCT04879017
NCT04891757
KAT6A   PF-07248144 metastatic ER+HER2- breast cancer, CRPC, NSCLC NCT04606446

Translocations of the MLL1 gene typify a unique group of acute leukemia that are often associated with poor prognosis. The MLL1 gene encodes a DNA binding protein that methylates histone H3K4 to positively regulate gene expression, including the Hox gene loci [62]. Leukemogenic MLL1 translocations fuse the N-terminus of MLL1 with >80 different fusion partners, and the majority of these fusions can aberrantly recruit DOT1 Like Histone Lysine Methyltransferase (DOT1L) to potentiate transcription activity [63]. DOT1L is a histone H3 lysine 79 methyltransferase [64]. DOT1L inhibitors were developed to inhibit leukemogenic transcription programs driven by MLL1-fusions [65]. DOT1L inhibitors demonstrated profound anti-tumor effects in models carrying MLL1 translocations [66,67] and in solid tumor models [66,68,69], which led to the progression of EPZ-5676 to a clinical trial [70]. Encouraging anti-tumor efficacy was observed early in the trial, though the overall anti-tumor efficacy was diminished when the trial was expanded to a larger patient population. Ultimately, DOT1L remains a target of interest but warrants further investigation.

Multiple alternate strategies were also pursued to inhibit the MLL1 fusion leukemogenic transcription, including inhibitors of Menin, WD repeat-containing protein 5 (WDR5) and MLL1 [71–74]. The most advanced of these efforts is the development of Menin inhibitors to block protein–protein interaction between MLL1 and Menin [75]. The N-terminal of the MLL1 protein contains a domain that interacts with Menin, a protein that serves as a link between MLL1 and the chromatin-binding protein lens epithelium-derived growth factor (LEDGF). The association of MLL1/Menin/LEDGF is required for the survival of leukemic cells [62,63]. Menin inhibitors potently suppress Hox and Meis1 expression and inhibit leukemic cell survival [75,76]. Multiple Menin inhibitors are now in clinical development with very promising anti-tumor efficacy observed (Table 1). While the initial proof-of-concept was demonstrated in MLL1-re-arranged leukemia, it was also found that leukemia with mutations in the nucleophosmin (NPM1) gene are also susceptible to Menin inhibition [75]. Disruption of Menin-MLL1 interaction induced growth arrest, myeloid cell differentiation and downregulation of Hox gene transcription [77]. Clinical studies with ziftomenib (KO-539) and revumenib (SNDX-5613), for which phase I clinical trial data were reported, demonstrated very encouraging efficacy in both MLL1r and NPM1mut leukemia [78]. This is now a highly active area of clinical development, and Menin inhibitors hold the promise to transform the clinical practice of MLL1r and NPM1mut leukemia.

The H3K36 histone methyltransferase NSD2 (also known as WHSC1 or MMSET) is a key oncogenic driver in multiple myeloma (MM) that carry t(4;14) translocation [39,79]. Global genomic profiling in the cancer cell line encyclopedia (CCLE) efforts also revealed hotspot, gain-of-function NSD2 mutations in pediatric acute lymphoblastic leukemia as well as in various solid tumors [40]. Cancers carrying NSD2 oncogenic mutations showed global hypermethylation of H3K36me2, with concomitant global hypomethylation of H3K27me3. Depletion of NSD2 by shRNA showed profound anti-tumor effects [79], validating NSD2 as an attractive drug target. A catalytic inhibitor selective against NSD2 (KTX-1001) has been developed and progressed to Phase 1 clinical trial (NCT05651932).

The success of EZH2 inhibitors in lymphoma and Menin inhibitors in leukemia is an important proof-of-concept that epigenetic targeted drugs are effective for cancer treatment. However, dominant oncogenic driver mutations of epigenetic regulators are mostly confined to hematological and rare cancers, therefore the concept of targeting oncogenic epigenetic regulators in solid cancers has been limited. Nonetheless, the success of targeting EZH2 and Menin is an important example of anti-tumor activity and provides important clinical data on toxicity and tolerability for this class of drugs. It is worth mentioning that EZH2 and Menin have a much broader roles in transcriptional regulation, with numerous epigenetic centered hypotheses for their contribution to disease progression in solid cancers. Building on their initial success in lymphoma and leukemia, these agents have now expanded their clinical testing in solid cancers mostly in combination with other drugs.

2.2.2. Synthetic lethality

The cancer genome sequencing project unveiled widespread genetic mutations of epigenetic regulators across all cancers. Loss-of-function and tumor suppressor mutations, including deletions, truncations, or damaging point mutations, are particularly prevalent. Chromatin remodeling proteins such as DNA and histone modifiers are mutated in every cancer type surveyed (Figure 2(a)) [84–87]. In some cases, mutations in epigenetic regulators alone are sufficient to drive tumorigenesis with high penetrance. For example, biallelic inactivation of SMARCB1 occurs in 95% of cases of malignant rhabdoid tumor (MRT) [88,89]. Unlike oncogene drivers which can be directly targeted with inhibitors, tumor suppressors which are lost in cancers cannot be directly drugged. A deep understanding of the downstream biological pathways can provide an avenue to target loss-of-function tumor suppressor pathways. For example, mutations in Tet2 and DNMT3A are known to occur early in hematopoietic progenitor cells, contributing to clonal hematopoiesis [90] and ultimately leukemia development [91–93]. With the understanding of Tet2 function in facilitating cytosine demethylation, Tet2 mutation has been associated with greater likelihood of responses to DNMTi in myeloid dysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) [94]. Similarly, mutations in DNMT3A and IDH1/2 status may predict response in MDS and AML [95]. However, in the absence of such mechanisms, alternative drug discovery strategies are needed to target tumor suppressor mutations, and in recent years, synthetic lethality has been an area of intense focus for epigenetic drug discovery.

Figure 2.

Figure 2.

Synthetic lethality approach to target cancers carrying mutations in epigenetic regulators. (a) Across all human cancers, there is a high prevalence of mutations in epigenetic genes. Mutation frequency of top 10 epigenetic regulators (green) were plotted comparing to tumor suppressor TP53 (purple) (source: cBioportal for cancer genomics pan-cancer database). (b) Synthetic lethality (SL) is defined as a circumstance in which the simultaneous mutation or inhibition of two genes is lethal, but the singular mutation or inhibition of either one is viable [80,81]. Identifying synthetic lethality relationships in cancers provides the possibility of developing therapeutics that selectively kill cancer cells harboring specific mutations, whilst sparing the wild-type normal cells. Exploiting synthetic lethality may provide a superior therapeutic index by reducing on-target toxicity. (c) SL relationships can be identified through a variety of experimental approaches. Large scale screening approaches utilizing both genetic tools (CRISPR or RNAi) and small molecule drug libraries can be explored. Such screens can be performed using large panel of cancer cell lines, or in paired isogenic knockout cell lines [82,83]. Created in BioRender. Vatapalli, R. (2024) BioRender.com/g88s208.

The concept of synthetic lethality can be illustrated by a pair of genes A and B, in which damaging mutations or deletion of gene A in cancer cells leads to epigenetic rewiring that results in a selective dependency on gene B, making gene B an attractive therapeutic target for cancers that lack gene A function [96]. Given that normal cells have both gene A and B, they can often tolerate the on-target impact targeting gene B. As such, cancer therapeutics based on the synthetic lethal concept are expected to have favorable therapeutic index by taking advantage of biological redundancy (Figure 2). With the rapid advances in large, genome-wide scale functional genomic screens using shRNA and CRISPR [82,83], together with comprehensive annotation of cancer cell lines by large consortium such as Sanger and Cancer Cell Line Encyclopedia (CCLE), these capabilities have enabled a systematic and comprehensive approach to uncover highly robust and specific synthetic lethal vulnerabilities for tumor suppressor mutations [83,96].

FDA approval of tazemetostat for patients with advanced epithelial sarcomas (ES) is the first successful example where synthetic lethality rationale is applied to target loss-of-function of an epigenetic regulator [58,89,97]. During development, gene expression states are tightly regulated during cell fate decision. Two opposing groups of epigenetic regulators, the Polycomb (PcG) protein which generally maintain gene repression and the Trithorax group (TrxG) proteins which generally activate gene expression, are vital to establish and maintain these stable and heritable gene expression states. The SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex belongs to the crucial TrxG family proteins. Mutations of the SWI/SNF complex proteins, including SMARCB1, ARID1A, SMARCA2/4, occur in ~20% of all cancers [98,99]. It was reported that SWI/SNF complex has an antagonistic relationship with EZH2 or PRC2, which belongs to the PcG group of proteins. Biallelic deletion or mutation of SMARCB1 is a defining hallmark in malignant rhabdoid tumor (MRT) and Ewing sarcoma (ES) [100]. MRT and ES are extremely aggressive cancers that respond poorly to existing therapies. Given the antagonistic relationship of SWI/SNF and PRC2, a series of very elegant preclinical studies [89] demonstrated that EZH2 and/or PRC2 inhibitors have strong anti-tumor activity in MRT and ES. In January 2020, tazemetostat received the FDA accelerated approval for the treatment of adults and children aged 16 years and older with metastatic or locally advanced ES not eligible for surgical resection [52]. The clinical activity of tazemetostat in ES is modest compared to follicular lymphoma, though this represents a major breakthrough in the development of epigenetic therapeutics to leverage the synthetic lethality concept and is the first approval of an epigenetic drug in solid cancers [96].

The Depmap consortium (https://depmap.org/portal/) has now generated CRISPR and shRNA cell line screening data which are publicly available for identification of novel synthetic lethal targets [83]. One exciting finding is the collateral vulnerability to the protein arginine methyltransferase PRMT5 in cancer models that harbor loss of methylthioadenosine phosphorylase (MTAP), which is frequently co-deleted with the tumor suppressor gene CDKN2A [101]. Genetic mutations of MTAP occur in 10–15% of all cancers. MTAP is a critical enzyme that plays a key role in polyamine metabolism and is important for both the methionine and adenine salvage pathway. It is also the only enzyme in mammalian cells known to catalyze the metabolism of methylthioadenosine (MTA). When the MTAP gene is deleted in cancer, MTA accumulates in cells and has partial inhibitory activity on PRMT5, which functions to suppress symmetric dimethylarginine (SDMA) levels. Thus, cancers with MTAP loss are susceptible to PRMT5 inhibition. First-generation PRMT5 inhibitors, including both SAM competitive and uncompetitive inhibitors, were rapidly progressed to clinical trials, though no meaningful clinical efficacy was observed due to dose-limiting hematological toxicities [102]. A major breakthrough has been the development of second generation, MTA cooperative PRMT5 inhibitors that realize the full potential of synthetic lethality in MTAP-deficient cancers. This is now an exciting area with multiple second-generation PRMT5 inhibitors, including MRTX1719, AMG193, TNG908, TNG462 and AZD3470 in clinical trials. MTA is a SAM mimetic and inhibits PRMT5 both in vitro and in cells [102]. When PRMT5 is MTA bound, it causes subtle conformational change of the Glu435 sidechain compared to the SAM-bound PRMT5 state. Remarkably, these subtle structural differences can be leveraged to develop MTA-cooperative PRMT5 inhibitors, which exploit the intracellular concentration differential of MTA and SAM in MTAP-deficient versus MTAP-wildtype cells, selectively killing cancer cells that have lost MTAP protein expression. Normal cells, such as hematopoietic cells, express MTAP protein and maintain a lower level of MTA::SAM ratio and are expected to have minimal levels of PRMT5 inhibited by the MTA cooperative inhibitor, therefore mitigating the hematological toxicities observed with the first-generation inhibitors. Preliminary data from MRTX1719 and AMG193 clinical studies demonstrated very promising safety and efficacy profile. Importantly, no dose limiting hematological toxicities were observed in patients. Altogether, this elegant drug discovery approach for the first time demonstrated that the differential cellular MTA:SAM ratio can be leveraged to create tumor-specific targeting of PRMT5 to maximize selective tumor cell killing. MTAP is widely lost in many solid cancers, including glioblastoma, mesothelioma, lung cancer, bladder cancer, gastric and esophageal cancers, pancreatic cancer, melanoma, and head and neck cancers. An MTA cooperative PRMT5 inhibitor is expected to have an exciting potential in a broad range of solid cancers, hopefully becoming the first targeted therapy for CDKN2A/MTAP negative cancers.

Another form of synthetic lethality commonly seen with epigenetic regulators is paralog synthetic lethality, which is best exemplified within the SWI/SNF chromatin remodeling complex. ARID1A/ARID1B, and SMARCA2/SMARCA4 are two pairs of paralogs which are incorporated into the SWI/SNF complex in a mutually exclusive manner [103,104]. When one of the paralogs is lost, SWI/SNF complexes utilize the remaining paralog to conduct its chromatin remodeling function, driving exclusive sensitivity to the perturbation of the remaining paralog. This paralog dependency relationship has been independently validated by several groups as well as in large-scale functional genomic studies [105–107]. Taking SMARCA2 and SMARCA4 as an example, SMARCA2 and SMARCA4 are ATPase subunits of the SWI/SNF complex. SMARCA4 loss due to mutations and deletions is frequently observed in non-small cell lung cancers and other solid tumors [108]. Oike and colleagues showed that SMARCA2 depletion by shRNA can selectively inhibit the growth of SMARCA4 deficient cells both in vitro and in vivo while sparing SMARCA4 wild-type models [106]. In addition to the ATPase domain, SMARCA2 and SMARCA4 also harbor a druggable bromodomain. Rescue experiments using SMARCA4 expression vectors with either ATPase-dead mutant or bromodomain mutation showed that the bromodomain mutant was able to completely rescue the growth phenotype upon SMARCA2 depletion, but an ATPase dead mutant was not able to. Similarly, bromodomain inhibition was insufficient to inhibit the growth and survival of SMARCA4 deficient models, indicating that the synthetic lethality phenotype is dependent on the ATPase but not the bromodomain function of the proteins [105].

The paralog dependency between SMARCA2 and SMARCA4 has attracted enormous interest to selectively target SMARCA2 for SMARCA4-deficient cancers. One of the efforts is through directly targeting the SMARCA2 ATPase function. However, the highly homologous nature between SMARCA2 and SMARCA4 ATPase domains poses a great challenge. A highly selective SMARCA2 inhibitor is desirable to achieve maximal therapeutic index. Several groups have reported an alternative approach which utilizes a SMARCA2/4 nonselective bromodomain binder and developed a SMARCA2 selective heterobifunctional degrader (also known as PROTACs) [109–111]. Both SMARCA2 selective ATPase inhibitor and selective SMARCA2 PROTACs have been reported [112,113]. Based on very promising pre-clinical data, a few of these molecules have advanced to early stages of clinical development (NCT05639751) [114].

SMARCA2 is among one of the most advanced paralog synthetic lethality-based targets for epigenetic regulators. There are many more synthetic lethal epigenetic targets that are being actively pursued by biotech and pharmaceutical companies, including ARID1A/B, EP300/CBP, DDX3X/3Y [115–117]. The concept of pursuing synthetic lethality for novel epigenetic targets is an area of active research and please refer to Table 2 for agents currently in clinical investigations.

Table 2.

Epigenetic inhibitors targeting synthetic lethal relationships in clinical development.

Epigenetic alteration Target Inhibitor Indication Rationale Clinical trial number
ARID1A mutation/LOF EZH2 Tazemetostat Ovarian cancer epigenetic antagonism between SWI/SNF and PcG (Polycomb group protein) [118] NCT05023655, NCT03348631
BAP1 loss EZH2 Tazemetostat Mesothelioma decrease in H4K20 monomethylation, increase in EZH2 expression [119] NCT02860286
SMARCA4 mutation/LOF SMARCA2 PRT-3789
PRT-7732
FHD-909
LY4050784
Solid tumors paralog dependency in SWI/SNF [106,107] NCT05639751
NCT06560645
NCT06561685
SYT-SSX EZH2 Tazemetostat Synovial sarcoma epigenetic antagonism between PcG and SWI/SNF [120] NCT02601937, NCT02601950
SMARCB1 EZH2 Tazemetostat malignant rhabdoid tumors (MRT) and atypical teratoid/rhabdoid tumors (ATRT) epigenetic antagonism between PcG and SWI/SNF [97,121] NCT03213665, NCT02601950
SMARCB1 HDAC Vorinostat MRT and ATRT restore histone acetylation in SMARCB1-null cells [122] NCT04897880
CREBBP mutation/LOF p300 CCS-1477 Lung cancer, Heme cancer paralog dependency [117] NCT04068597
MTAP PRMT5 AMG-193, TNG-908, MRTX-1719
TNG-462
AZD-3470
Multiple cancers preferential PRMT5 inhibition by MTA in MTAP deleted cancer [101,102,123] NCT05245500, NCT05094336, NCT05275478,
NCT05732831
NCT06130553
NCT06137144

2.3. Combination therapy

Building on the success of monotherapy activity of HDAC and DNMT inhibitors, combinations of HDAC or DNMT inhibitors with other therapies have been extensively tested in the clinical setting. The most notable success was the combination of DNMTi with cell death agent venetoclax for the treatment of AML. Low-intensity regimen of venetoclax combined with decitabine or azacitidine demonstrated good efficacy and a tolerable safety profile in elderly patients with AML unfit for intensive chemotherapy [124]. There are active clinical trials to explore DNMTi in combination with other agents to further extend the benefit of AML treatment, including Fms-like kinase 3 (FLT3) inhibitor gilteritinib (FLT3 inhibitor) plus azacitidine (NCT02752035) for AML with FLT3 mutation and ADI-PEG 20 (pegylated arginine deiminase) in combination with venetoclax and azacitidine triple combination for AML. Effective use of epigenetic drugs in combination setting is an area of active investigation. In the next session, we will discuss a few combination strategies and current preclinical and clinical activities exploring these opportunities.

2.3.1. Combination therapy with epigenetic agents to inhibit the oncogenic transcriptional programs

One of the most successful concepts in oncology therapy is to target lineage-dependent transcription programs, such as estrogen receptor (ER) mediated transcription in breast cancer and androgen receptor (AR) mediated transcription in prostate cancer. Epigenetic regulation plays a critical role in transcription activation and is often hijacked in the cancer setting to fuel constitutive transcriptional activities driving cancer progression. It is therefore rational to consider combining epigenetic targeted agents with other targeted drugs to suppress oncogenic transcription programs.

Several recent studies reported that EZH2 expression is upregulated in hormone-refractory metastatic prostate cancers and high EZH2 expression levels are correlated with disease stage, and poor outcome in prostate cancer patients [125,126]. In preclinical models, combining EZH2 inhibitors with enzalutamide synergistically inhibits cell proliferation and colony formation and promotes apoptosis in enzalutamide-resistant prostate cancer cells [127]. Mechanistically, EZH2 functions both as a transcription corepressor and coactivator [128]. In mouse models, EZH2 inhibition restrains tumor neuroendocrine differentiation [125,127,129]. EZH2 inhibition increases cytotoxic activity and IFN-γ production of tumor-specific CD8+ T lymphocytes [130], highlighting the multi-faceted opportunity to re-sensitize tumors to androgen targeted therapies by inhibiting EZH2. Currently, three EZH2 inhibitors have progressed to clinical testing in Metastatic Castration-Resistant Prostate Cancer for combination with enzalutamide or other androgen deprivation therapies (NCT03480646, NCT04179864, NCT03460977) [131–133].

KAT6A and KAT6B are members of the MYST family of histone acetyltransferases. Dysregulation of MYST family HATs has been implicated in the activation of oncogenic gene expression programs in human cancers [134]. KAT6A was found to be amplified and/or overexpressed in 10–15% of breast cancers and shown to be a significant dependency in breast cancer cell lines overexpressing KAT6A [135]. KAT6A shRNA-mediated knockdown reduced ERα levels in ER+ breast cancer cells, highlighting a role for KAT6A in regulating ERα-mediated growth of breast cancer cells [135]. These findings supported further investigation of the clinical utility of targeting KAT6A in ER+ breast cancers. A first-in-class KAT6A/B inhibitor PF-07248144 is being tested as a single agent and in combination with either fulvestrant or letrozole + palbociclib in advanced solid tumors (NCT04606446) [136].

2.3.2. Epigenetic therapy in combination with immune checkpoint therapies (ICTs)

There is an increasing amount of the literature supporting the idea of combining epigenetic therapies with ICTs to enhance their antitumor effects. There are many biological rationales proposed, including a) increase in neoantigen expression; b) increase in antigen presentation; c) rejuvenation of exhausted T-cells; d) increase in T cell differentiation and memory; e) induction of other immune cells including dendritic cells and NK cells; f) activation of interferons [96,137–139]. A number of clinical studies have been initiated to explore the benefit of combining epigenetic drugs with ICTs (see Table 3). In this section, we review a few selected examples to demonstrate the key principles and learnings.

Table 3.

Epigenetic inhibitors tested in the clinic in combination with immunotherapies (active trials).

Epi- therapy Immunotherapy
Indication Clinical trial Number
Vaccine Checkpoint therapy    
Demethylation agents (DNMTi)
Azacytidine, Decitabine
Allogeneic colon cancer cell vaccine (GVAX) and cyclophosphamide   Metastatic colon cancer NCT01966289
DEC-205/NY-ESO-1 Fusion Protein CDX-1401   MDS/AML NCT03358719
  Pembrolizumab Metastatic melanoma, NSCLC NCT02816021, NCT02546986
  Nivolumab and/or Ipilimumab MDS NCT02530463
HDACi
ACY-241, Entinostat, ZEN-3694
  Nivolumab and Ipilimumab Metastatic unresectable HER2-negative breast cancer, NSCLC, Solids NCT02453620
NCT02635061
NCT04840589
  Pembrolizumab Metastatic CRPC NCT04471974
EZH2i
CPI-1205, DS-3201
  Pembrolizumab Advanced urothelial cancer, NSCLC, Head & Neck NCT03854474
NCT05879484
  Ipilimumab Metastatis Prostate, Urothelial and Renal cell cancers NCT04388852
  Atezolizumab and Bevacizumab Hepatocellular Carcinoma NCT06294548
  Rituximab Follicular Lymphoma NCT05683171
  SHR1701 R/R cHL,
Advanced solids
R/R BC NHL
NCT05896046
NCT04407741
BETi
INCB057643, BMS986158
  Pembrolizumab + Epacadostat (IDO1 inhibitor) Advanced solid cancers NCT02959437
  Ruxolitinib or Fedratinib Myelofibrosis NCT04817007
LSD1i
ORY-1001,IMG-7289
  Atezolizumab SCLC NCT06287775
NCT05191797

Seminal work from Baylin and colleagues demonstrated the role of DNA methylation inhibitors, such as azacytidine, in increasing antigen presentation and major histocompatibility complex I (MHC-I) expression, thereby augmenting antitumor immune responses [140–143]. DNMTi induces double-stranded RNA (dsRNA) production through reactivation of transposable elements like endogenous retroviruses (ERVs), long interspersed retrotransposable elements (LINEs) and short interspersed retrotransposable elements (SINEs) and inverted-repeat Alu elements. Elevated levels of dsRNA are sensed by toll-like receptor 3 (TLR3), RNA helicases retinoic acid-inducible gene protein I (RIG-I) or melanoma differentiation-associated protein 5 (MDA5) mediated pathway to activate Type I interferon response [144]. DNMTis were also shown to reverse immune cell exhaustion and restore anti-tumor responses [145–147]. These preclinical studies have led to the excitement to combine DNMTi with ICTs in clinical trials. When Hodgkin Lymphoma patients were treated with programmed cell death protein 1 (PD-1) antibody camrelizumab in combination with decitabine (NCT02961101), there was a meaningful improvement in both complete response (CR) rates (71%) and progression-free survival (PFS) (100%) when compared to PD-1 monotherapy (32% and 76% respectively) [148]. It will be interesting to see if newer DNMTi will bring broader impacts to enhance ICTs in different cancer types.

Lysine-specific demethylase 1 (LSD1), a histone demethylase, was reported to enhance the efficacy of immunotherapy by acting on both the tumor intrinsic and immune related pathways [149]. In tumor cells, LSD1 inhibition can lead to an increased antigen presentation [150]. More importantly, LSD1 is also involved in the development and regulation of T cells, macrophages, cancer-associated fibroblasts (CAFs) and NK cells [151–154]. With LSD1 inhibitors already in the clinic, these preclinical findings have led to the initiation of several clinical trials that combine LSD1 inhibitors with immune checkpoint inhibitors [155–157].

Functional genomic screens continue to reveal new epigenetic targets that can enhance immune-mediated anti-tumor response. An epigenetic-focused CRISPR/Cas9 screen in chronically stimulated mouse CD8+ T cells revealed that SWI/SNF chromatin remodeling complex components, including ARID1A, DPF2, SMARCC1, and SMARCA4 were among the top scoring hits depleted in the PD1+TIM3+ T cells, highlighting a role of SWI/SNF in regulating T cell exhaustion [158]. Confirmatory studies using single gRNA mediated knockdown of SMARCA4, SMARCC1, and DPF2 validated findings from the primary screen. Pharmacological inhibition of SWI/SNF using SMARCA4/2 ATPase inhibitor or PROTAC degrader resulted in similar decrease in PD1+TIM3+ population and decrease in terminal T cell exhaustion marker CD39+ in multiple donors while increasing the effector T cell pool. Transcriptomics and epigenomic analysis showed that key hallmark genes of exhaustion including TOX, ENTPD1, ITGA2, and TIGIT were directly bound and regulated by the SWI/SNF complex. In an OVA antigen expressing B16 melanoma model, the pretreatment of CD8+ OT-1 T cells with the SMARCA4/2 ATPase inhibitor enhanced their in vivo anti-tumor activity. In another example, genome-wide CRISPR/Cas9 study identified EZH2 as an important player in mediating transcriptional silencing of MHC-I antigen processing pathway to promote immune evasion of T-cell mediated immunity [159]. EZH2 inhibitor can reverse epigenetic silencing of MHC-I antigen processing pathway to re-establish T cell-mediated immunity. Kim et al. has comprehensively reviewed the multi-faceted role of EZH2 in overcoming resistance to immune evasion [160].

In addition to the above examples, there is abundant literature demonstrating that targeting epigenetic regulators, including HDACs, G9a, SETDB1, ADAR1 can enhance ICTs. Here, we also caution that current preclinical models carry major challenges and limitations and might not adequately inform the complex immune-mediated anti-tumor mechanisms nor the complex tumor microenvironment in patients. Therefore, findings from preclinical models might not translate in clinical trials. For example, the HDAC inhibitor Entinostat was shown to have strong anti-tumor effects when combined with ICTs in syngeneic breast and pancreatic cancer mouse models [161]. However, in a trial (NCT02708680) testing atezolizumab in combination with entinostat, no significant benefit was observed [162]. Entinostat also did not show meaningful clinical benefit in the ENCORE 601 trial for non-small cell lung cancer (NSCLC) in combination with PD-1 inhibitor pembrolizumab [163]. To drive the success of rational combination therapies, a thorough mechanistic understanding of each drug’s biological impact on immune and tumor cells is needed.

Reverse translation from clinical experience could be a better path forward to inform biological hypotheses with epigenetic regulation and immune-mediated anti-tumor mechanisms. For example, Goswami et al. [164] observed that ipilimumab increases EZH2 expression in human T cells across various tumor indications and the increased EZH2 expression in T cells inversely correlates with clinical outcome. Following the initial clinical observation, preclinical models were developed and validated the hypothesis that pharmacological inhibition of EZH2 in combination with ipilimumab enhances effector-like T cell responses, providing a strong rationale for testing the combination of EZH2 inhibitor with ipilimumab in the clinic. This led to the initiation of a clinical trial combining EZH1/2 inhibitor (DS3201) with ipilimumab (NCT04388852). In these ongoing clinical trials, comprehensive translational biomarker data needs to be collected to inform how EZH2 inhibition impacts immune cells, tumor cells, and stromal cells. Findings from patient tumors can then be used to develop new hypotheses and assessed in the preclinical setting.

2.3.3. Epigenetics and therapy resistance

The development of drug resistance is a major challenge to the long-term effectiveness of cancer therapies. In addition to on-target genetic mutation causing drug resistance, studies with acquired resistance models also revealed significant contribution of epigenetic reprogramming to the development and maintenance of resistance states, especially in models that lack clear genetic driver mechanisms [165,166]. Drug resistance can be mediated by a subpopulation of cells that persist after treatment, also known as “Drug Tolerant Persisters” (DTPs) [167]. DTPs have stem cell-like features with a high degree of epigenetic plasticity. For instance, EGFR inhibitor-induced DTPs were found to highly express KDM5A. Inhibition of KDM5A or treatment with HDACi led to reduced survival of DTPs upon EGFRi treatment [168]. Profiling of osimertinib DTPs using RNA-seq and ATAC-seq identified consistent activation of the TEAD pathway [169]. These osimertinib DTPs were shown to be sensitive to BRD4 or TEAD inactivation. There is emerging evidence that DTPs adapt to therapeutic intervention through epigenomic modifications, transcriptomic regulation, metabolic remodeling, and altered interactions with the tumor microenvironment [170,171].

de Miguel et al. profiled chromatin accessibility in five pairs of parental and osimertinib resistant lung cancer models and showed that osimertinib-resistant cells harbor a differentiated chromatin accessibility pattern compared to parental cells [172]. Chromatin accessibility gains are enriched in promoters and enhancer regions for target genes associated with upregulation of EMT and receptor tyrosine kinase (RTK) signaling pathways, and downregulation of epithelial cell differentiation and cell–cell adhesion. A diverse set of resistance-associated transcriptional programs were observed, underscoring the heterogeneity of the resistant state. In search of chromatin associated factors that govern these changes, SWI/SNF chromatin remodeling complex component SMARCA4 and SWI/SNF associated transcription factors including TP53, SOX2 were identified. The contribution of SMARCA4 in driving the resistance states was further substantiated by concordant change in SWI/SNF chromatin occupancy in the enhancer regions of the differentially expressed genes. Osimertinib resistance can be reversed in a subset of models by SMARCA4 inhibition. Moreover, SMARCA4 perturbation can deepen the response of patient-derived xenograft models to Osimertinib.

Epigenetic alterations can drive pro-survival signaling, DNA repair, expression of drug efflux transporters, promote EMT, and reprogram metabolic pathways to cause drug resistance [173]. In such cases, utilizing a mechanism-based combinatorial approach including epigenetic therapies to overcome drug resistance is an attractive approach. The concept of epigenetic “priming” to re-sensitize resistant tumors to conventional chemotherapy targeted therapies or immunotherapies has been gaining great attention. One of the mechanisms by which pretreatment or “epigenetic-priming” increases the efficacy of the combination partner is through the reactivation of tumor-suppressor genes. For example, DLBCL patients treated with low-dose azacytidine showed reactivation of tumor suppressor SMAD1 and were re-sensitized to doxorubicin treatment leading to a higher remission rate [174].

While there are ample preclinical examples supporting the rationale of combining with epigenetic therapy to combat or prevent resistance, we are not aware of any successes in clinical trials. The main challenge is that unlike genetic-based resistance mechanisms, it is still not clear if epigenetic mechanisms of resistance such as DTPs occur in human cancer. With the advance of single-cell sequencing and improved ability to perform biomarker analysis using clinical samples, it will be important to clearly demonstrate the specific “epigenetic alterations” driving therapy resistance in cancer patients.

2.4. Novel modalities for targeting epigenetic factors

In the last decade, targeted protein degradation including PROTAC and Molecular Glue (MG) degraders has gained interest as novel drugging modalities [175,176]. These molecules are able to induce target protein degradation mimicking the effects of genetic knockdown and have demonstrated differential and in some cases superior activity compared to traditional catalytic inhibitors. These approaches are particularly relevant to epigenetic drug discovery as many epigenetic regulators have protein complex scaffolding functions beyond enzymatic activities.

There is increasing evidence from the studies of Menin/MLL inhibitors and EED/EZH2 inhibitors that protein degraders can further improve therapeutic efficacy [177–179]. Since 2021, multiple protein degraders have entered clinical trials, including SMARCA2 (PRT3789 [NCT05639751] and PRT7732 [NCT06560645]) degraders. In the case of selective SMARCA2 PROTACs, a nonselective SMARCA2/4 dual binder against the bromodomain was linked to an E3 ubiquitin ligase warhead. Selectivity toward SMARCA2 was engineered by optimizing the ternary structure between target protein and E3 ligase. Utilizing the PROTAC modality to improve selectivity and druggability is an exciting avenue for the development of more effective epigenetic therapeutics.

Another novel targeting modality that holds great potential is molecular glue approach. Molecular glues can promote protein–protein interaction, and in many cases molecular glues are designed to enhance the interaction between a target protein and an E3 ligase to promote target protein degradation. In 2019, Blake et al. reported the development of a BRD4 monovalent degrader GNE-0011, which recruited DCAF16 as the E3 ligase to mediate its degradation activity [180,181]. By applying a transposable covalent handle to existing binders, Toriki et al. reported HDAC1/3 and SMARCA2/4 molecular glues that recruit RNF126 to mediate the degradation effects. While no molecular glue targeting an epigenetic driver has progressed into clinical testing yet, we expect breakthroughs in this field can lead to more effective epigenetic drugs with improved potency and selectivity in the near future.

3. Conclusions

Cancer epigenetic drug discovery is a rapidly evolving field that holds great promise for the development of more effective and targeted therapies. Advances in cancer epigenetic research have already led to the discovery of epigenetic biomarkers that can be used for early cancer detection, risk assessment, and monitoring of treatment response. With the advancement of novel drugging approaches, diverse types of small-molecule inhibitors, including catalytic inhibitors, protein–protein interaction inhibitors, and protein degraders, can be designed for specific targets. The FDA approval of DNMT, HDAC, EZH2, and IDH1/2 inhibitors has demonstrated that targeting epigenetic mechanisms can be effective in cancer therapy. We are particularly encouraged by the increased pace and interest in the clinical development of many epigenetic agents (Tables 1–3). Due to space limitations, we have focused the discussion on epigenetics targeted agents under active clinical investigation, but we expect that the next wave of epigenetic agents to enter the clinic will have broad impacts in many if not all cancers. Epigenetic drugs when used in combination with other treatment modalities, such as chemotherapy, radiation therapy, or immunotherapy, could enhance treatment response and overcome drug resistance. The exciting field of cancer epigenetics holds tremendous potential for revolutionizing the cancer treatment landscape in the coming years.

4. Future perspectives

The future of cancer epigenetic drug discovery is poised to benefit significantly from advancements in drug discovery and epigenomic sequencing technologies. First, the development of new drugging modalities, such as PROTACs, molecular glue and covalent drugging strategies, holds great promise to develop more effective drugs targeting epigenetic regulators. These new capabilities have the potential to target previously “undruggable” epigenetic regulators, expanding the repertoire of targets. Second, by mapping the epigenomic landscape of different cancer types, we can now more precisely define different cancer subtypes by combining genetic and epigenomic annotations. The integration of single-cell epigenomics and multi-omics approaches will further enhance our understanding of the dynamic nature of the cancer cell states and reveal additional novel cancer-specific aberrations and vulnerabilities. This, together with combinatorial CRISPR screens in preclinical models, will help uncover the next wave of epigenetic targets for cancer therapies. In addition, cancer-specific metabolic states should be considered during drug design and optimization to maximize cancer targeting while sparing non-cancerous tissues. Last but not the least, reverse translation using clinical observations to inform preclinical research will play a pivotal role in developing mechanism-based combination strategies with epigenetic drugs. Here, the ability to apply new epigenomic sequencing technologies, including single-cell sequencing, to patient tumor samples will transform our understanding of tumor epigenetic state in “real-time.” This deeper insight into epigenomic dysregulation in cancer patients will provide more precision in guiding mechanism-based combination strategies.

Acknowledgments

We thank Steven Criscione, James Brownell, Ming Tang and John Reicha (all employees of AstraZeneca) for proofreading and providing valuable comments. We would also like to acknowledge that we were unable to cite many references due to space constraints.

Funding Statement

This paper was not funded.

Article highlights

Cancer epigenetic drug discovery

  • Dysregulated epigenetic landscape is a hallmark of all cancers and can be therapeutically targeted.

  • FDA approval of inhibitors against DNMTs, HDACs, and EZH2 showed initial success in several cancer indications.

Precision targeted epigenetic therapeutics

  • Precision-targeted epigenetic therapeutics will have broader impact to all cancers.

  • Genetic mutations of epigenetic regulators are common in all cancers and offer new opportunities to develop epigenetic drugs targeting these newly defined cancer segments.

  • New generation of targeted epigenetic drugs based on epigenetic oncogene drivers and synthetic lethality, such as inhibitors against PRMT5 and Menin, is showing great promise in clinical studies.

Combination Therapy

  • Mechanism-based combination strategies will realize the full potential of epigenetic drugs.

  • Epigenetic therapy can enhance and deepen the therapeutic benefits of immune checkpoint therapies.

  • Epigenetic therapy can be used to combat therapy resistance.

Disclosure statement

All authors are employees of AstraZeneca. Alex Rossi is now an employee of Flare therapeutics.

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Author contributions

Rajita Vatapalli, Ho Man Chan, Alex P. Rossi, Jingwen Zhang wrote the main manuscript text. Rajita Vatapalli and Alex P. Rossi made the tables and figures.

References

Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.

  • 1.Russo VEA, Martienssen RA, Riggs AD.. Epigenetic mechanisms of gene regulation. In: Russo VEA, editor. Cold Spring harbor monograph series. Vol. 32. Plainview, N.Y: Cold Spring Harbor Laboratory Press; 1996. p. 692. [Google Scholar]
  • 2.Bird A. Perceptions of epigenetics. Nature. 2007;447(7143):396–398. doi: 10.1038/nature05913 [DOI] [PubMed] [Google Scholar]
  • 3.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293(5532):1074–1080. doi: 10.1126/science.1063127 [DOI] [PubMed] [Google Scholar]
  • 4.Gasser SM, Li E. Epigenetics and disease: pharmaceutical opportunities. Preface Prog Drug Res. 2011;67:v–viii. [PubMed] [Google Scholar]
  • 5.Arrowsmith CH, Bountra C, Pv F, et al. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11(5):384–400. doi: 10.1038/nrd3674 [DOI] [PubMed] [Google Scholar]
  • 6.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683–692. doi: 10.1016/j.cell.2007.01.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Baylin SB, Jones PA. A decade of exploring the cancer epigenome - biological and translational implications. Nat Rev Cancer. 2011;11(10):726–734. doi: 10.1038/nrc3130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ma X, Yu L, Liu Y, et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature. 2018;555(7696):371–376. doi: 10.1038/nature25795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pradat Y, Viot J, Yurchenko AA, et al. Integrative pan-cancer genomic and transcriptomic analyses of refractory metastatic cancer. Cancer Discov. 2023;13(5):1116–1143. doi: 10.1158/2159-8290.CD-22-0966 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Priestley P, Baber J, Lolkema MP, et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature. 2019;575(7781):210–216. doi: 10.1038/s41586-019-1689-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dawson MA. The cancer epigenome: concepts, challenges, and therapeutic opportunities. Science. 2017;355(6330):1147–1152. doi: 10.1126/science.aam7304 [DOI] [PubMed] [Google Scholar]
  • 12.Bennett RL, Licht JD. Targeting Epigenetics in cancer. Annu Rev Pharmacol Toxicol. 2018;58:187–207. doi: 10.1146/annurev-pharmtox-010716-105106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sorm F, Piskala A, Čihák A, et al. 5-azacytidine, a new, highly effective cancerostatic. Experientia. 1964;20(4):202–203. doi: 10.1007/BF02135399 [DOI] [PubMed] [Google Scholar]
  • 14.Gnyszka A, Jastrzebski Z, Flis S. DNA methyltransferase inhibitors and their emerging role in epigenetic therapy of cancer. Anticancer Res. 2013;33(8):2989–2996. [PubMed] [Google Scholar]
  • 15.Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008;123(1):8–13. doi: 10.1002/ijc.23607 [DOI] [PubMed] [Google Scholar]
  • 16.Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell. 1980;20(1):85–93. doi: 10.1016/0092-8674(80)90237-8 [DOI] [PubMed] [Google Scholar]
  • 17.Pappalardi MB, Keenan K, Cockerill M, et al. 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. doi: 10.1038/s43018-021-00249-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol. 2014;6(4):a018713. doi: 10.1101/cshperspect.a018713 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Atadja PW. HDAC inhibitors and cancer therapy. Prog Drug Res. 2011;67:175–195. [DOI] [PubMed] [Google Scholar]
  • 20.Yang XJ, Seto E. HATs and HDACs: from structure, function and regulation to novel strategies for therapy and prevention. Oncogene. 2007;26(37):5310–5318. doi: 10.1038/sj.onc.1210599 [DOI] [PubMed] [Google Scholar]
  • 21.Furumai R, Komatsu Y, Nishino N, et al. Potent histone deacetylase inhibitors built from trichostatin a and cyclic tetrapeptide antibiotics including trapoxin. Proc Natl Acad Sci USA. 2001;98(1):87–92. doi: 10.1073/pnas.98.1.87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kinashi H, Someno K, Sakaguchi K. Isolation and characterization of concanamycins A, B and C. J Antibiot (Tokyo). 1984;37(11):1333–1343. doi: 10.7164/antibiotics.37.1333 [DOI] [PubMed] [Google Scholar]
  • 23.Yoshimitsu M, Ando K, Ishida T, et al. Oral histone deacetylase inhibitor HBI-8000 (tucidinostat) in Japanese patients with relapsed or refractory non-Hodgkin’s lymphoma: phase I safety and efficacy. Jpn J Clin Oncol. 2022;52(9):1014–1020. doi: 10.1093/jjco/hyac086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Suo J, Zhu K, Zhuang C, et al. Efficacy and safety of tucidinostat in patients with advanced hormone receptor-positive human epidermal growth factor receptor 2-negative breast cancer: real-world insights. Ann Transl Med. 2023;11(12):409. doi: 10.21037/atm-23-1913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sun Y, Hong JH, Ning Z, et al. Therapeutic potential of tucidinostat, a subtype-selective HDAC inhibitor, in cancer treatment. Front Pharmacol. 2022;13:932914. doi: 10.3389/fphar.2022.932914 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Marchion D, Munster P. Development of histone deacetylase inhibitors for cancer treatment. Expert Rev Anticancer Ther. 2007;7(4):583–598. doi: 10.1586/14737140.7.4.583 [DOI] [PubMed] [Google Scholar]
  • 27.Li Y, Seto E. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harb Perspect Med. 2016;6(10). doi: 10.1101/cshperspect.a026831 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bradner JE, West N, Grachan ML, et al. Chemical phylogenetics of histone deacetylases. Nat Chem Biol. 2010;6(3):238–243. doi: 10.1038/nchembio.313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cheshmazar N, Hamzeh-Mivehroud M, Charoudeh HN, et al. Current trends in development of HDAC-based chemotherapeutics. Life Sci. 2022;308:120946. doi: 10.1016/j.lfs.2022.120946 [DOI] [PubMed] [Google Scholar]
  • 30.Zhang Z, Wang G, Li Y, et al. Recent progress in DNA methyltransferase inhibitors as anticancer agents. Front Pharmacol. 2022;13:1072651. doi: 10.3389/fphar.2022.1072651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bondarev AD, Attwood MM, Jonsson J, et al. Recent developments of HDAC inhibitors: emerging indications and novel molecules. Br J Clin Pharmacol. 2021;87(12):4577–4597. doi: 10.1111/bcp.14889 [DOI] [PubMed] [Google Scholar]
  • 32.Shanmugam G, Rakshit S, Sarkar K. HDAC inhibitors: targets for tumor therapy, immune modulation and lung diseases. Transl Oncol. 2022;16:101312. doi: 10.1016/j.tranon.2021.101312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stegmeier F, Warmuth M, Sellers WR, et al. Targeted cancer therapies in the twenty-first century: lessons from imatinib. Clin Pharmacol Ther. 2010;87(5):543–552. doi: 10.1038/clpt.2009.297 [DOI] [PubMed] [Google Scholar]
  • 34.Stuart DD, Guzman-Perez A, Brooijmans N, et al. Precision oncology comes of age: designing best-in-class small molecules by integrating two decades of advances in chemistry. Target Biol Data Sci Cancer Discov. 2023;13(10):2131–2149. doi: 10.1158/2159-8290.CD-23-0280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46. doi: 10.1158/2159-8290.CD-21-1059 [DOI] [PubMed] [Google Scholar]
  • 36.Chan HM, Arrowsmith C, editors. Epigenetic Drug Discovery. Vol. 83. Royal Society of Chemistry; 2024. [Google Scholar]; •• This book is an excellent resource for an introduction to epigenetic drug discovery.
  • 37.Yap DB, Chu J, Berg T, et al. 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. doi: 10.1182/blood-2010-11-321208 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• First report of oncogenic driver mutations in EZH2.
  • 38.Milne TA, Kim J, Wang GG, et al. Multiple interactions recruit MLL1 and MLL1 fusion proteins to the HOXA9 locus in leukemogenesis. Mol Cell. 2010;38(6):853–863. doi: 10.1016/j.molcel.2010.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chesi M, Nardini E, Lim RS, et al. 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]
  • 40.Jaffe JD, Wang Y, Chan HM, et al. Global chromatin profiling reveals NSD2 mutations in pediatric acute lymphoblastic leukemia. Nat Genet. 2013;45(11):1386–1391. doi: 10.1038/ng.2777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.French CA, Ramirez CL, Kolmakova J, et al. BRD-NUT oncoproteins: a family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells. Oncogene. 2008;27(15):2237–2242. doi: 10.1038/sj.onc.1210852 [DOI] [PubMed] [Google Scholar]
  • 42.Crew AJ, Clark J, Fisher C, et al. Fusion of SYT to two genes, SSX1 and SSX2, encoding proteins with homology to the Kruppel-associated box in human synovial sarcoma. Embo J. 1995;14(10):2333–2340. doi: 10.1002/j.1460-2075.1995.tb07228.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Molenaar RJ, Maciejewski JP, Wilmink JW, et al. Wild-type and mutated IDH1/2 enzymes and therapy responses. Oncogene. 2018;37(15):1949–1960. doi: 10.1038/s41388-017-0077-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807–1812. doi: 10.1126/science.1164382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765–773. doi: 10.1056/NEJMoa0808710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Qi W, Chan H, Teng L, et al. 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. doi: 10.1073/pnas.1210371110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Amatori S, Tavolaro S, Gambardella S, et al. The dark side of histones: genomic organization and role of oncohistones in cancer. Clin Epigenetics. 2021;13(1):71. doi: 10.1186/s13148-021-01057-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Yu JR, LeRoy G, Bready D, et al. The H3K36me2 writer-reader dependency in H3K27M-DIPG. Sci Adv. 2021;7(29). doi: 10.1126/sciadv.abg7444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhang Y, Fang D. The incorporation loci of H3.3K36M determine its preferential prevalence in chondroblastomas. Cell Death Dis. 2021;12(4):311. doi: 10.1038/s41419-021-03597-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Maze I, Noh KM, Soshnev AA, et al. Every amino acid matters: essential contributions of histone variants to mammalian development and disease. Nat Rev Genet. 2014;15(4):259–271. doi: 10.1038/nrg3673 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Soshnev AA, Cd A, Cesarman E, et al. Histone H1 mutations in lymphoma: a Link(er) between chromatin organization, developmental reprogramming, and cancer. Cancer Res. 2021;81(24):6061–6070. doi: 10.1158/0008-5472.CAN-21-2619 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hoy SM. Tazemetostat: First Approval. Drugs. 2020;80(5):513–521. doi: 10.1007/s40265-020-01288-x [DOI] [PubMed] [Google Scholar]
  • 53.Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647(1–2):21–29. doi: 10.1016/j.mrfmmm.2008.07.010 [DOI] [PubMed] [Google Scholar]
  • 54.Morin RD, Johnson NA, Severson TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet. 2010;42(2):181–185. doi: 10.1038/ng.518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.McCabe MT, Graves AP, Ganji G, et al. Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27). Proc Natl Acad Sci USA. 2012;109(8):2989–2994. doi: 10.1073/pnas.1116418109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lohr JG, Stojanov P, Lawrence MS, et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc Natl Acad Sci USA. 2012;109(10):3879–3884. doi: 10.1073/pnas.1121343109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bodor C, O’Riain C, Wrench D, et al. EZH2 Y641 mutations in follicular lymphoma. Leukemia. 2011;25(4):726–729. doi: 10.1038/leu.2010.311 [DOI] [PubMed] [Google Scholar]
  • 58.Knutson SK, Wigle TJ, Warholic NM, et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat Chem Biol. 2012;8(11):890–896. doi: 10.1038/nchembio.1084 [DOI] [PubMed] [Google Scholar]
  • 59.McCabe MT, Ott HM, Ganji G, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012;492(7427):108–112. doi: 10.1038/nature11606 [DOI] [PubMed] [Google Scholar]
  • 60.Qi W, Zhao K, Gu J, et al. An allosteric PRC2 inhibitor targeting the H3K27me3 binding pocket of EED. Nat Chem Biol. 2017;13(4):381–388. doi: 10.1038/nchembio.2304 [DOI] [PubMed] [Google Scholar]
  • 61.Morschhauser F, Tilly H, Chaidos A, et al. 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. doi: 10.1016/S1470-2045(20)30441-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bernt KM, Armstrong SA. Targeting epigenetic programs in mll-rearranged leukemias. Hematology Am Soc Hematol Educ Program. 2011;2011:354–360. [DOI] [PubMed] [Google Scholar]
  • 63.Winters AC, Bernt KM. MLL-Rearranged leukemias-an update on science and clinical approaches. Front Pediatr. 2017;5:4. doi: 10.3389/fped.2017.00004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Feng Q, Wang H, Hh N, et al. Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain. Curr Biol. 2002;12(12):1052–1058. doi: 10.1016/S0960-9822(02)00901-6 [DOI] [PubMed] [Google Scholar]
  • 65.Daigle SR, Olhava EJ, Therkelsen CA, et al. Potent inhibition of DOT1L as treatment of mll-fusion leukemia. Blood. 2013;122(6):1017–1025. doi: 10.1182/blood-2013-04-497644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Vatapalli R, Sagar V, Rodriguez Y, et al. Histone methyltransferase DOT1L coordinates AR and MYC stability in prostate cancer. Nat Commun. 2020;11(1):4153. doi: 10.1038/s41467-020-18013-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Daigle SR, Olhava EJ, Therkelsen CA, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20(1):53–65. doi: 10.1016/j.ccr.2011.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kurani H, Razavipour SF, Harikumar KB, et al. DOT1L is a novel cancer stem cell target for triple-negative breast cancer. Clin Cancer Res. 2022;28(9):1948–1965. doi: 10.1158/1078-0432.CCR-21-1299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Salvati A, Gigantino V, Nassa G, et al. The histone methyltransferase DOT1L is a functional component of estrogen receptor alpha signaling in ovarian cancer cells. Cancers (Basel). 2019;11(11). doi: 10.3390/cancers11111720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Yi Y, Ge S. Targeting the histone H3 lysine 79 methyltransferase DOT1L in mll-rearranged leukemias. J Hematol Oncol. 2022;15(1):35. doi: 10.1186/s13045-022-01251-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Karatas H, Townsend EC, Cao F, et al. High-affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction. J Am Chem Soc. 2013;135(2):669–682. doi: 10.1021/ja306028q [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Li DD, Chen WL, Xl X, et al. Structure-based design and synthesis of small molecular inhibitors disturbing the interaction of MLL1-WDR5. Eur J Med Chem. 2016;118:1–8. doi: 10.1016/j.ejmech.2016.04.032 [DOI] [PubMed] [Google Scholar]
  • 73.Krivtsov AV, Evans K, Gadrey JY, et al. 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. doi: 10.1016/j.ccell.2019.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bai H, Zhang SQ, Lei H, et al. Menin-mll protein-protein interaction inhibitors: a patent review (2014-2021). Expert Opin Ther Pat. 2022;32(5):507–522. [DOI] [PubMed] [Google Scholar]
  • 75.Kuhn MW, Song E, Feng Z, et al. Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 2016;6(10):1166–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kuhn MW, Armstrong SA. Designed to kill: novel menin-mll inhibitors target mll-rearranged leukemia. Cancer Cell. 2015;27(4):431–433. [DOI] [PubMed] [Google Scholar]
  • 77.Perner F, Armstrong SA. Targeting chromatin complexes in myeloid malignancies and beyond: from basic mechanisms to clinical innovation. Cells. 2020;9(12). doi: 10.3390/cells9122721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Candoni A, Coppola G. A 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. doi: 10.3390/hematolrep16020024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kuo AJ, Cheung P, Chen K, et al. NSD2 links dimethylation of histone H3 at lysine 36 to oncogenic programming. Mol Cell. 2011;44(4):609–620. doi: 10.1016/j.molcel.2011.08.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.O’Neil NJ, Bailey ML, Hieter P. Synthetic lethality and cancer. Nat Rev Genet. 2017;18(10):613–623. doi: 10.1038/nrg.2017.47 [DOI] [PubMed] [Google Scholar]
  • 81.Ryan CJ, Lp D, Sj P, et al. Complex synthetic lethality in cancer. Nat Genet. 2023;55(12):2039–2048. doi: 10.1038/s41588-023-01557-x [DOI] [PubMed] [Google Scholar]
  • 82.McDonald ER 3rd, De WA, Mr S, et al. Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell. 2017;170(3):577–592 e10. doi: 10.1016/j.cell.2017.07.005 [DOI] [PubMed] [Google Scholar]
  • 83.Tsherniak A, Vazquez F, Pg M, et al. Defining a cancer dependency map. Cell. 2017;170(3):564–576 e16. doi: 10.1016/j.cell.2017.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This is an extensive resource for defining new synthetic lethal relationships and identifying new targets.
  • 84.Nawy T. A pan-cancer atlas. Nat Methods. 2018;15(6):407. doi: 10.1038/s41592-018-0020-4 [DOI] [PubMed] [Google Scholar]
  • 85.Bailey MH, Tokheim C, Porta-Pardo E, et al. Comprehensive characterization of cancer Driver genes and mutations. Cell. 2018;173(2):371–385 e18. doi: 10.1016/j.cell.2018.02.060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Shen H, Laird PW. Interplay between the cancer genome and epigenome. Cell. 2013;153(1):38–55. doi: 10.1016/j.cell.2013.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505(7484):495–501. doi: 10.1038/nature12912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Han E, Kim J, Jung MJ, et al. Malignant rhabdoid tumor of the kidney in an adult with loss of INI1 expression and mutation in the SMARCB1 gene. J Pathol Transl Med. 2021;55(2):145–153. doi: 10.4132/jptm.2021.01.26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wilson BG, Wang X, Shen X, et al. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell. 2010;18(4):316–328. doi: 10.1016/j.ccr.2010.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This study elaborates on synthetic lethality between EZH2 and SMARCB1 leading to the first approval of Tazemetostat in Epithelioid sarcoma.
  • 90.Buscarlet M, Provost S, Zada YF, et al. DNMT3A and TET2 dominate clonal hematopoiesis and demonstrate benign phenotypes and different genetic predispositions. Blood. 2017;130(6):753–762. doi: 10.1182/blood-2017-04-777029 [DOI] [PubMed] [Google Scholar]
  • 91.Langemeijer SM, Kuiper RP, Berends M, et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet. 2009;41(7):838–842. [DOI] [PubMed] [Google Scholar]
  • 92.Delhommeau F, Dupont S, Vd V, et al. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360(22):2289–2301. doi: 10.1056/NEJMoa0810069 [DOI] [PubMed] [Google Scholar]
  • 93.Jankowska AM, Szpurka H, Rv T, et al. Loss of heterozygosity 4q24 and TET2 mutations associated with myelodysplastic/myeloproliferative neoplasms. Blood. 2009;113(25):6403–6410. doi: 10.1182/blood-2009-02-205690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lin Y, Lin Z, Cheng K, et al. Prognostic role of TET2 deficiency in myelodysplastic syndromes: a meta-analysis. Oncotarget. 2017;8(26):43295–43305. doi: 10.18632/oncotarget.17177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Im AP, Ar S, Mp C, et al. DNMT3A and IDH mutations in acute myeloid leukemia and other myeloid malignancies: associations with prognosis and potential treatment strategies. Leukemia. 2014;28(9):1774–1783. doi: 10.1038/leu.2014.124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Huang A, LA G, Ashworth A, et al. Synthetic lethality as an engine for cancer drug target discovery. Nat Rev Drug Discov. 2020;19(1):23–38. doi: 10.1038/s41573-019-0046-z [DOI] [PubMed] [Google Scholar]
  • 97.Knutson SK, Nm W, Tj W, et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci USA. 2013;110(19):7922–7927. doi: 10.1073/pnas.1303800110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kadoch C, Dc H, Hodges C, et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet. 2013;45(6):592–601. doi: 10.1038/ng.2628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kadoch C, Crabtree GR. Mammalian SWI/SNF chromatin remodeling complexes and cancer: mechanistic insights gained from human genomics. Sci Adv. 2015;1(5):e1500447. doi: 10.1126/sciadv.1500447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Parker NA, Al-Obaidi A, Lalich D, et al. SMARCB1/INI1-deficient extrarenal rhabdoid tumor: a case report of a rare and aggressive soft tissue Sarcoma. Cureus. 2020;12(5):e8273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kryukov GV, Fh W, Jr R, et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science. 2016;351(6278):1214–1218. doi: 10.1126/science.aad5214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mavrakis KJ, Er M, Mr S, et al. Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5. Science. 2016;351(6278):1208–1213. [DOI] [PubMed] [Google Scholar]; •• This study was the first to demonstrate synthetic lethality in MTAP deleted cancers and highlights how effectively MTA inhibits PRMT5 enzyme leading to development of MTA cooperative PRMT5 inhibitors.
  • 103.Sasaki M, Ogiwara H. Synthetic lethal therapy based on targeting the vulnerability of SWI/SNF chromatin remodeling complex-deficient cancers. Cancer Sci. 2020;111(3):774–782. doi: 10.1111/cas.14311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Centore RC, Gj S, Lm S, et al. Mammalian SWI/SNF chromatin remodeling complexes: emerging mechanisms and therapeutic strategies. Trends Genet. 2020;36(12):936–950. doi: 10.1016/j.tig.2020.07.011 [DOI] [PubMed] [Google Scholar]
  • 105.Vangamudi B, Ta P, Pk S, et al. 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. doi: 10.1158/0008-5472.CAN-14-3798 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Oike T, Ogiwara H, Tominaga Y, et al. 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. doi: 10.1158/0008-5472.CAN-12-4593 [DOI] [PubMed] [Google Scholar]
  • 107.Hoffman GR, Rahal R, Buxton F, et al. Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers. Proc Natl Acad Sci USA. 2014;111(8):3128–3133. doi: 10.1073/pnas.1316793111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Fernando TM, Piskol R, Bainer R, et al. Functional characterization of SMARCA4 variants identified by targeted exome-sequencing of 131,668 cancer patients. Nat Commun. 2020;11(1):5551. doi: 10.1038/s41467-020-19402-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Yang L, Tu W, Huang L, et al. 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. doi: 10.1021/acs.jmedchem.3c00953 [DOI] [PubMed] [Google Scholar]
  • 110.Kofink C, Trainor N, Mair B, et al. A selective and orally bioavailable vhl-recruiting PROTAC achieves SMARCA2 degradation in vivo. Nat Commun. 2022;13(1):5969. doi: 10.1038/s41467-022-33430-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Cantley J, Ye X, Rousseau E, et al. Selective protac-mediated degradation of SMARCA2 is efficacious in SMARCA4 mutant cancers. Nat Commun. 2022;13(1):6814. doi: 10.1038/s41467-022-34562-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Reichl KD, Lee ECY, Gopalsamy A. Synthetic lethality: targeting SMARCA2 ATPase in SMARCA4-deficient tumors - a review of patent literature from 2019-30 June 2023. Expert Opin Ther Pat. 2024;34(3):159–169. [DOI] [PubMed] [Google Scholar]
  • 113.Lee ECY, Reichl KD, Gopalsamy A. Synthetic lethality: targeting the SMARCA2 bromodomain for degradation in SMARCA4-deficient tumors - a review of patent literature from 2019-June 2023. Expert Opin Ther Pat. 2024;34:1–19. [DOI] [PubMed] [Google Scholar]
  • 114.PRT3789 monotherapy and in combo w/Docetaxel in participants w/Advanced or metastatic solid tumors w/SMARCA4 mutation. Available from: https://www.clinicaltrials.gov/study/NCT05639751?term=SMARCA4%20Gene%20Mutation&rank=1
  • 115.Koferle A, Schlattl A, Hörmann A, et al. Interrogation of cancer gene dependencies reveals paralog interactions of autosome and sex chromosome-encoded genes. Cell Rep. 2022;39(2):110636. doi: 10.1016/j.celrep.2022.110636 [DOI] [PubMed] [Google Scholar]
  • 116.Helming KC, Wang X, Bg W, et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat Med. 2014;20(3):251–254. doi: 10.1038/nm.3480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ogiwara H, Sasaki M, Mitachi T, et al. 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. doi: 10.1158/2159-8290.CD-15-0754 [DOI] [PubMed] [Google Scholar]
  • 118.Bitler BG, Aird KM, Zhang R. Epigenetic synthetic lethality in ovarian clear cell carcinoma: EZH2 and ARID1A mutations. Mol Cell Oncol. 2016;3(1):e1032476. doi: 10.1080/23723556.2015.1032476 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.LaFave LM, Béguelin W, Koche R, et al. Loss of BAP1 function leads to EZH2-dependent transformation. Nat Med. 2015;21(11):1344–1349. doi: 10.1038/nm.3947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kawano S, Ar G, Tsuda M, et al. Preclinical evidence of anti-tumor activity induced by EZH2 inhibition in human models of synovial sarcoma. PLOS ONE. 2016;11(7):e0158888. doi: 10.1371/journal.pone.0158888 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Alimova I, Dk B, Ps H, et al. Inhibition of EZH2 suppresses self-renewal and induces radiation sensitivity in atypical rhabdoid teratoid tumor cells. Neuro Oncol. 2013;15(2):149–160. doi: 10.1093/neuonc/nos285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yamamichi N, Yamamichi-Nishina M, Mizutani T, et al. The brm gene suppressed at the post-transcriptional level in various human cell lines is inducible by transient HDAC inhibitor treatment, which exhibits antioncogenic potential. Oncogene. 2005;24(35):5471–5481. doi: 10.1038/sj.onc.1208716 [DOI] [PubMed] [Google Scholar]
  • 123.Marjon K, Mj C, Quang P, et al. MTAP deletions in cancer create vulnerability to targeting of the MAT2A/PRMT5/RIOK1 Axis. Cell Rep. 2016;15(3):574–587. doi: 10.1016/j.celrep.2016.03.043 [DOI] [PubMed] [Google Scholar]
  • 124.DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019;133(1):7–17. doi: 10.1182/blood-2018-08-868752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Ku SY, Rosario S, Wang Y, et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science. 2017;355(6320):78–83. doi: 10.1126/science.aah4199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Varambally S, Dhanasekaran SM, Zhou M, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419(6907):624–629. doi: 10.1038/nature01075 [DOI] [PubMed] [Google Scholar]
  • 127.Bai Y, Zhang Z, Cheng L, et al. Inhibition of enhancer of zeste homolog 2 (EZH2) overcomes enzalutamide resistance in castration-resistant prostate cancer. J Biol Chem. 2019;294(25):9911–9923. doi: 10.1074/jbc.RA119.008152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Xu K, Wu ZJ, Groner AC, et al. EZH2 oncogenic activity in castration-resistant prostate cancer cells is polycomb-independent. Science. 2012;338(6113):1465–1469. doi: 10.1126/science.1227604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kim J, Lee Y, Lu X, et al. Polycomb- and methylation-independent roles of EZH2 as a transcription activator. Cell Rep. 2018;25(10):2808–2820 e4. doi: 10.1016/j.celrep.2018.11.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Fischetti I, Botti L, Sulsenti R, et al. Combined therapy targeting AR and EZH2 curbs castration-resistant prostate cancer enhancing anti-tumor T-cell response. Epigenomics. 2024;16(9):653–670. doi: 10.2217/epi-2023-0374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.PF-06821497 treatment of Relapsed/Refractory SCLC, castration resistant prostate cancer, and follicular lymphoma. Available from: https://www.clinicaltrials.gov/study/NCT03460977
  • 132.A study of tazemetostat with enzalutamide or Abiraterone/Prednisone in participants with advanced prostate cancer (CELLO-1). Available from: https://clinicaltrials.gov/study/NCT04179864
  • 133.ProSTAR: a study evaluating CPI-1205 in patients with metastatic castration resistant prostate cancer. Available from: https://clinicaltrials.gov/study/NCT03480646
  • 134.Avvakumov N, Cote J. The MYST family of histone acetyltransferases and their intimate links to cancer. Oncogene. 2007;26(37):5395–5407. doi: 10.1038/sj.onc.1210608 [DOI] [PubMed] [Google Scholar]
  • 135.Yu L, Liang Y, Cao X, et al. Identification of MYST3 as a novel epigenetic activator of ERalpha frequently amplified in breast cancer. Oncogene. 2017;36(20):2910–2918. doi: 10.1038/onc.2016.433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Study of PF-07248144 in advanced or metastatic solid tumors (KAT6). Available from: https://clinicaltrials.gov/study/NCT04606446
  • 137.Liu Z, Ren Y, Weng S, et al. A new trend in cancer treatment: the combination of epigenetics and immunotherapy. Front Immunol. 2022;13:809761. doi: 10.3389/fimmu.2022.809761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Cao J, Yan Q. Cancer Epigenetics, tumor immunity, and immunotherapy. Trends Cancer. 2020;6(7):580–592. doi: 10.1016/j.trecan.2020.02.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Gomez S, Tabernacki T, Kobyra J, et al. Combining epigenetic and immune therapy to overcome cancer resistance. Semin Cancer Biol. 2020;65:99–113. doi: 10.1016/j.semcancer.2019.12.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Topper MJ, Vaz M, Chiappinelli KB, et al. Epigenetic therapy ties MYC depletion to reversing immune evasion and treating lung cancer. Cell. 2017;171(6):1284–1300 e21. doi: 10.1016/j.cell.2017.10.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Li H, Chiappinelli KB, Guzzetta AA, et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget. 2014;5(3):587–598. doi: 10.18632/oncotarget.1782 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Kanholm T, Rentia U, Hadley M, et al. Oncogenic transformation drives DNA methylation loss and transcriptional activation at transposable element loci. Cancer Res. 2023;83(15):2584–2599. doi: 10.1158/0008-5472.CAN-22-3485 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Chiappinelli KB, Zahnow CA, Ahuja N, et al. Combining epigenetic and immunotherapy to combat cancer. Cancer Res. 2016;76(7):1683–1689. doi: 10.1158/0008-5472.CAN-15-2125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015;162(5):974–986. doi: 10.1016/j.cell.2015.07.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ghoneim HE, Fan Y, Moustaki A, et al. De Novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell. 2017;170(1):142–157 e19. doi: 10.1016/j.cell.2017.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Prinzing B, Zebley CC, Petersen CT, et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity. Sci Transl Med. 2021;13(620):eabh0272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Jones PA, Ohtani H, Chakravarthy A, et al. Epigenetic therapy in immune-oncology. Nat Rev Cancer. 2019;19(3):151–161. [DOI] [PubMed] [Google Scholar]; •• This is a great review that highlights the role of DNA methylation inhibitors in inducing a viral mimicry state in cells which forms the basis for combinations of epigenetic therapy with immune therapy.
  • 148.Nie J, Wang C, Liu Y, et al. Addition of low-dose decitabine to anti-PD-1 antibody camrelizumab in Relapsed/Refractory classical Hodgkin Lymphoma. J Clin Oncol. 2019;37(17):1479–1489. doi: 10.1200/JCO.18.02151 [DOI] [PubMed] [Google Scholar]
  • 149.Mamun MA, Zhang Y, Zhao JY, et al. LSD1: an emerging face in altering the tumor microenvironment and enhancing immune checkpoint therapy. J Biomed Sci. 2023;30(1):60. doi: 10.1186/s12929-023-00952-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Nguyen EM, Taniguchi H, Chan JM, et al. Targeting lysine-specific demethylase 1 rescues Major histocompatibility complex class I antigen presentation and overcomes programmed death-ligand 1 blockade resistance in SCLC. J Thorac Oncol. 2022;17(8):1014–1031. doi: 10.1016/j.jtho.2022.05.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Liu C, Liu L, Chen X, et al. LSD1 stimulates cancer-associated fibroblasts to drive Notch3-dependent self-renewal of liver cancer stem-like cells. Cancer Res. 2018;78(4):938–949. doi: 10.1158/0008-5472.CAN-17-1236 [DOI] [PubMed] [Google Scholar]
  • 152.Yang RF, Zhao GW, Liang ST, et al. Lysine-specific demethylase 1 represses THP-1 monocyte-to-macrophage differentiation. Chin Med Sci J. 2013;28(2):82–87. doi: 10.1016/S1001-9294(13)60027-9 [DOI] [PubMed] [Google Scholar]
  • 153.Qiu F, Jiang P, Zhang G, et al. Priming with LSD1 inhibitors promotes the persistence and antitumor effect of adoptively transferred T cells. Nat Commun. 2024;15(1):4327. doi: 10.1038/s41467-024-48607-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Xueqing X, Yongcan P, Wei L, et al. Regulation of T cells in the tumor microenvironment by histone methylation: LSD1 inhibition-a new direction for enhancing immunotherapy. Heliyon. 2024;10(2):e24457. doi: 10.1016/j.heliyon.2024.e24457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Azacitidine combined with pembrolizumab and epacadostat in subjects with advanced solid tumors (ECHO-206). Available from: https://www.clinicaltrials.gov/study/NCT02959437?term=lsd1&viewType=Table&page=3&rank=30
  • 156.A safety and efficacy study of CC-90011 in combination with nivolumab in subjects with advanced cancers. Available from: https://www.clinicaltrials.gov/study/NCT04350463?term=lsd1&viewType=Table&page=4&rank=34
  • 157.Bomedemstat and maintenance immunotherapy for treatment of newly diagnosed extensive stage small cell lung cancer. Available from: https://www.clinicaltrials.gov/study/NCT05191797?term=lsd1&viewType=Table&page=4&rank=36
  • 158.Battistello E, Hixon KA, Comstock DE, et al. Stepwise activities of mSWI/SNF family chromatin remodeling complexes direct T cell activation and exhaustion. Mol Cell. 2023;83(8):1216–1236 e12. doi: 10.1016/j.molcel.2023.02.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Burr ML, Sparbier CE, Chan KL, et al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell. 2019;36(4):385–401 e8. doi: 10.1016/j.ccell.2019.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Kim HJ, Cantor H, Cosmopoulos K. Overcoming immune checkpoint blockade resistance via EZH2 inhibition. Trends Immunol. 2020;41(10):948–963. doi: 10.1016/j.it.2020.08.010 [DOI] [PubMed] [Google Scholar]
  • 161.Christmas BJ, Ci R, Hopkins AC, et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol Res. 2018;6(12):1561–1577. doi: 10.1158/2326-6066.CIR-18-0070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.O’Shaughnessy J, Moroose RL, Babu K, et al. Results of ENCORE 602 (TRIO025), a phase II, randomized, placebo-controlled, double-blinded, multicenter study of atezolizumab with or without entinostat in patients with advanced triple-negative breast cancer (aTNBC). Journal of Clinical Oncology. 2020;38(15_suppl):1014–1014. doi: 10.1200/JCO.2020.38.15_suppl.1014 [DOI] [Google Scholar]
  • 163.Hellmann MD, Janne PA, Opyrchal M, et al. Entinostat plus pembrolizumab in patients with metastatic NSCLC Previously treated with anti-pd-(L)1 therapy. Clin Cancer Res. 2021;27(4):1019–1028. doi: 10.1158/1078-0432.CCR-20-3305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Goswami S, Apostolou I, Zhang J, et al. Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy. J Clin Invest. 2018;128(9):3813–3818. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This report demonstrates the first example of reverse translation from clinic. The authors observed EZH2 upregulation in T cells in patients which leads to resistance to anti-CTLA4 therapy which in turn led to clinical assessment of combinations with EZH2 inhibitors.
  • 165.Brown R, Curry E, Magnani L, et al. Poised epigenetic states and acquired drug resistance in cancer. Nat Rev Cancer. 2014;14(11):747–753. doi: 10.1038/nrc3819 [DOI] [PubMed] [Google Scholar]
  • 166.Marine JC, Dawson SJ, Dawson MA. Non-genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer. 2020;20(12):743–756. doi: 10.1038/s41568-020-00302-4 [DOI] [PubMed] [Google Scholar]
  • 167.Leonce C, Saintigny P, Ortiz-Cuaran S. Cell-intrinsic mechanisms of drug tolerance to systemic therapies in cancer. Mol Cancer Res. 2022;20(1):11–29. doi: 10.1158/1541-7786.MCR-21-0038 [DOI] [PubMed] [Google Scholar]
  • 168.Sharma SV, Dy L, Li B, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141(1):69–80. doi: 10.1016/j.cell.2010.02.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Criscione SW, Martin MJ, Oien DB, et al. The landscape of therapeutic vulnerabilities in EGFR inhibitor osimertinib drug tolerant persister cells. NPJ Precis Oncol. 2022;6(1):95. doi: 10.1038/s41698-022-00337-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Shen H. Subclonal mutations in epigenetic regulators bring fitness in times of stress. Cancer Cell. 2023;41(1):7–9. doi: 10.1016/j.ccell.2022.11.004 [DOI] [PubMed] [Google Scholar]
  • 171.Loukas I, Simeoni F, Milan M, et al. Selective advantage of epigenetically disrupted cancer cells via phenotypic inertia. Cancer Cell. 2023;41(1):70–87 e14. doi: 10.1016/j.ccell.2022.10.002 [DOI] [PubMed] [Google Scholar]
  • 172.de Miguel FJ, Gentile C, Feng WW, et al. Mammalian SWI/SNF chromatin remodeling complexes promote tyrosine kinase inhibitor resistance in egfr-mutant lung cancer. Cancer Cell. 2023;41(8):1516–1534 e9. doi: 10.1016/j.ccell.2023.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Adhikari S, Bhattacharya A, Adhikary S, et al. The paradigm of drug resistance in cancer: an epigenetic perspective. Biosci Rep. 2022;42(4). doi: 10.1042/BSR20211812 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Clozel T, Yang S, Elstrom RL, et al. Mechanism-based epigenetic chemosensitization therapy of diffuse large B-cell lymphoma. Cancer Discov. 2013;3(9):1002–1019. doi: 10.1158/2159-8290.CD-13-0117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Webb T, Craigon C, Ciulli A. Targeting epigenetic modulators using PROTAC degraders: current status and future perspective. Bioorg Med Chem Lett. 2022;63:128653. doi: 10.1016/j.bmcl.2022.128653 [DOI] [PubMed] [Google Scholar]
  • 176.Peng X, Hu Z, Zeng L, et al. Overview of epigenetic degraders based on PROTAC, molecular glue, and hydrophobic tagging technologies. Acta Pharm Sin B. 2024;14(2):533–578. doi: 10.1016/j.apsb.2023.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hsu JH, Rasmusson T, Robinson J, et al. EED-Targeted PROTACs degrade EED, EZH2, and SUZ12 in the PRC2 complex. Cell Chem Biol. 2020;27(1):41–46 e17. [DOI] [PubMed] [Google Scholar]
  • 178.Wang X, Cao W, Zhang J, et al. A covalently bound inhibitor triggers EZH2 degradation through chip-mediated ubiquitination. Embo J. 2017;36(9):1243–1260. doi: 10.15252/embj.201694058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Wu Y, Doepner M, Hojnacki T, et al. Disruption of the menin-mll interaction triggers menin protein degradation via ubiquitin-proteasome pathway. Am J Cancer Res. 2019;9(8):1682–1694. [PMC free article] [PubMed] [Google Scholar]
  • 180.Toriki ES, Papatzimas JW, Nishikawa K, et al. Rational chemical design of molecular glue degraders. ACS Cent Sci. 2023;9(5):915–926. doi: 10.1021/acscentsci.2c01317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Blake RA. Abstract 4452: GNE-0011, a novel monovalent BRD4 degrader. Cancer Research. 2019;79(13_Supplement):4452–4452. doi: 10.1158/1538-7445.Am2019-4452 [DOI] [Google Scholar]

Articles from Epigenomics are provided here courtesy of Taylor & Francis

RESOURCES