Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 25.
Published in final edited form as: Nat Cancer. 2024 Jun 27;5(6):844–865. doi: 10.1038/s43018-024-00777-2

Pharmacologic Targeting of the Cancer Epigenome

Nathaniel W Mabe 1,2, Jennifer A Perry 1, Clare F Malone 1,2, Kimberly Stegmaier 1,2,3
PMCID: PMC11936478  NIHMSID: NIHMS2067715  PMID: 38937652

Abstract

Epigenetic dysregulation is increasingly appreciated as a hallmark of cancer, including disease initiation, maintenance, and therapy resistance. As a result, there have been advances in the development and evaluation of epigenetic therapies for cancer, revealing significant promise, but also challenges. Three epigenetic inhibitor classes are approved in the United States, and many more are currently undergoing clinical investigation. In this Review, we discuss recent developments for each epigenetic drug class and their implications for therapy, as well as highlight new insights into the role of epigenetics in cancer.

Epigenetics of Cancer

Epigenetics is broadly defined by the non-genetic biological processes underlying the regulation of chromatin architecture with no changes to the primary DNA sequence. Epigenetic regulation is paramount to establishing the gene expression patterns that define cell identity, differentiation, and response to environmental stimuli1. Dysregulation of epigenetic processes has long been appreciated as a core feature of cancer, with mutations in genes encoding epigenetic regulatory proteins among the most frequently observed2. In recent years, manifestations of epigenetic dysregulation, such as phenotype plasticity and non-genetic epigenetic reprogramming, have also been incorporated as fundamental hallmarks of cancers3.

The process of interpreting DNA-encoded information to guide cellular phenotypes involves a complex network of DNA elements, transcription factors, epigenome modifiers, and biophysical interactions. DNA is packaged tightly in nucleosomes consisting of 8 histone protein subunits that can be modified to facilitate regions of high transcription (euchromatin) or low transcription (heterochromatin)4. During transcription, nucleosome positioning is adjusted through ATP-dependent SWI/SNF chromatin remodeling complexes, allowing access to cis-regulatory elements on DNA, such as promoters and enhancers, to which trans-regulatory transcription factors can bind in a sequence-dependent manner to modulate transcription through RNA polymerase II activity and its associated complexes (e.g., CDK9/CCNT1 (P-TEFb), Mediator, etc.)5,6. Among the most studied epigenetic mechanisms regulating transcription are post-translational modifications (PTMs), either on cytosines of DNA or on amino acids of histone tails of nucleosomes, that facilitate the recruitment of chromatin modifiers, transcription factors, or compact the DNA7. Historically, epigenetic regulation has been defined by families of enzymatic writers and erasers that maintain the balance of deposition and removal of histone tail PTMs and the protein readers that recognize these PTMs8. Other epigenetic regulators, including scaffolding proteins that facilitate histone regulation, such as Menin, or chromatin remodelers (e.g., the BAF complex), have emerged as therapeutic targets in cancer, but do not fit cleanly into the reader, writer, and eraser categories.

Therapeutic Targeting of Epigenetic Proteins

Epigenetic proteins that regulate gene expression are frequently mutated in cancer, and as their mechanisms are increasingly being elucidated, therapeutic agents that alter their function are being developed. Here, we survey the history and recent advances in targeting epigenetic proteins in cancer, describing Food and Drug Administration (FDA)-approved small molecules and those in clinical trials. Currently, epigenetic inhibitors representing three distinct classes are approved by the FDA in the United States: DNA methyltransferase (DNMT), Histone deacetylase (HDAC), and Enhancer of Zeste Homolog-2 (EZH2) inhibitors. However, at least ten other classes of epigenetic inhibitors are currently being investigated in clinical trials (Figure 1). The development of these epigenetic chemical modulators has significantly expanded our understanding of how cancer cells utilize epigenetic and transcriptional machinery to promote their growth and survival.

Figure 1: Epigenetic Targets in Cancer.

Figure 1:

Major pharmacologic drug classes that target epigenetic processes and are currently in clinical testing or approved by government regulatory agencies. * indicates FDA approval.

Targeting Common Essential Chromatin Regulators

Some epigenetic regulators are common essential genes and their inhibition mimics cytotoxic chemotherapy while other epigenetic regulators may be selective dependencies or synthetic lethal in specific cancer subsets. While these differences had to be empirically determined in the past, genome-scale loss-of-function screening efforts in large panels of cancer cell lines have generated data sets, such as the Cancer Dependency Map (DepMap)9, in which genes can be broadly classified as common essential, selectively enriched, or non-essential across over 1000 cancer cell line models. Using these data, we can predict whether response to knockout of a particular epigenetic gene and its small molecule therapies by proxy are more likely to perform like a cytotoxic chemotherapy (e.g., TOP1 inhibitors) versus a targeted therapy (e.g., EGFR inhibitor)10. Common essential genes, including DNA methyltransferases (e.g., DNMT1), histone deacetylases (e.g., HDAC3) and bromodomain and extra-terminal domain (BET) proteins (e.g., BRD4), are among current epigenetic targets of interest for clinical development.

DNA Methyltransferase Inhibitors and Histone Deacetylase Inhibitors

DNMT inhibitors and HDAC inhibitors belong to the first developed classes of small molecules targeting epigenetic regulators and have been extensively studied and reviewed elsewhere11,12. The clinical impact of these drugs has been notable, but almost completely limited to hematologic malignancies. DNMT inhibitors 5’-azacytidine and decitabine are FDA approved for myelodysplastic syndrome, 5’-azacytidine for juvenile myelomonocytic leukemia, and 5’-azacytidine and decitabine are commonly used off-label for treatment of acute myeloid leukemia (AML) and are FDA approved in combination with venetoclax in older adults diagnosed with AML. HDAC inhibitors are FDA approved for cutaneous T-cell lymphomas (vorinostat, romidepsin)13, peripheral T-cell lymphoma (belinostat)14, and multiple myeloma (panobinostat),15 and many other clinical candidate HDAC inhibitors are being studied in solid and hematologic malignancies (Figure 1 and Table 1).

Table 1:

Summary of ongoing and completed clinical trials for epigenetic drugs.

Drug Disease State In combination with Study Number Phase Status Reason for withdrawal/termination Reference articles (PMID)
DNMT inhibitors
5’-azacytidine * MDS, juvenile myelomonocytic leukemia
Decitabine * MDS
guadecitabine** (SGI-110) AML Standard of care NCT02920008 Phase 3 Completed
MDS, Chronic myelomonocytic leukemia NCT02907359 Phase 3 Completed
AML NCT02348489 Phase 3 Completed 37276510
HDAC inhibitors
romidepsin * Cutaneous T-cell lymphoma FDA approved
vorinostat * Cutaneous T-cell lymphoma FDA approved
belinostat * Peripheral T-cell lymphoma FDA approved
panobinostat * MM FDA approved
tucidinostat (chidamide) ** Peripheral T-cell lymphoma azacitidine and CHOP NCT05075460 Phase 3 Not yet recruiting
DLBCL R-CHOP NCT04231448 Phase 3 Active, not recruiting
ER+ Breast cancer exemestane NCT02482753 Phase 3 Completed 31036468
Periperal T cell lymphoma NCT04668690 Phase 3 Not yet recruiting
ER+ Breast cancer exemestane NCT05253066 Phase 3 Not yet recruiting
Ph-like ALL dasatinib NCT03564470 Phase 3 Recruiting
DLBCL or T cell lymphoma cladribine, gemcitabine and busulfan or BCNU, etoposide, cytarabine, and melphalan. NCT05466318 Phase 3 Recruiting
ER+/HER2- advanced Breast cancer after CDK4/6 failure NCT05335473 Phase 3 Recruiting
Advanced CRC tucidinostat + sintilimab + bev vs. second-line FOLFIRI + Bev NCT05768503 Phase 3 Recruiting
T-cell lymphoma Compared versus SHR2554 NCT06122389 Phase 3 Recruiting
Stage III/IV extranodal NK/T-cell lymphoma tucidinostat + anti-PD1 + pegaspargase NCT06255795 Phase 3 Not yet recruiting
entinostat (MS-275) ** ER+/HER2- breast cancer exemestane NCT03538171 Phase 3 Active, not recruiting
ER+/HER2- breast cancer exemestane NCT02115282 Phase 3 Active, not recruiting
Citarinostat (ACY-241) Advanced solid malignancies paclitaxel NCT02551185 Phase 1 Completed 35070991
NSCLC nivolumab NCT02635061 Phase 1 Active, not recruiting
MM pomalidomide and dexamethasone NCT02400242 Phase 1 Active, not recruiting
Melanoma ipilimumab and nivolumab NCT02935790 Phase 1 Completed
Smoldering MM PVX-410 + citarinostat +/− lenalidomide NCT02886065 Phase 1 Active, not recruiting
KA2507 Biliary tract cancer NCT04186156 Phase 2 Withdrawn Study not progressing
Advanced solid malignancies NCT03008018 Phase 1 Completed 33947698
abexinostat (PCI-24781) DLBCL, Mantle Cell lymphoma ibrutinib NCT03939182 Phase 1/2 Active, not recruiting
Follicular lymphoma NCT03934567 Phase 2 Recruiting
Follicular lymphoma NCT03600441 Phase 2 Active, not recruiting
DLBCL NCT03936153 Phase 2 Recruiting
Breast and gynecologic cancers palbociclib and fulvestrant NCT04498520 Phase 1 Withdrawn Funding
Renal cell carcinoma pazopanib NCT03592472 Phase 3 Recruiting
Advanced solid malignancies pembrolizumab NCT03590054 Phase 1 Active, not recruiting
Non-Hodgkins lymphoma NCT04024696 Phase1/2 Active, not recruiting
Sarcoma doxorubicin NCT01027910 Phase 1/2 Completed 25536954
Non-Hodgkins lymphoma, MM, leukemia NCT01149668 Phase 1 Completed
Advanced solid malignancies pazopanib NCT01543763 Phase 1 Active, not recruiting
Non-Hodgkins lymphoma, Mantle cell lymphoma NCT00724984 Phase 1/2 Completed 26482040
Advanced solid malignancies NCT00473577 Phase 1 Completed
Recurrent glioma Temodar + abexinostat NCT05698524 Phase 1 Recruiting
pracinostat (SB939) MDS azacitidine NCT03151304 Phase 2 Terminated Sponsor decision
myelofibrosis ruxolitinib NCT02267278 Phase 2 Completed 30632841
AML azacitadine NCT01912274 Phase 2 Completed 30760466
MDS azacitadine or decitabine NCT01993641 Phase 2 Completed
MDS azacitidine NCT01873703 Phase 2 Completed 28094841
AML gemtuzumab NCT03848754 Phase 1 Completed 36401944
AML azacitadine NCT03151408 Phase 3 Terminated Lack of Efficacy
translocation-associated sarcomas NCT01112384 Phase 2 Completed 25632070
Advanced solid and hematologic malignancies NCT00741234 Phase 1 Completed 28841236
CRPC NCT01075308 Phase 2 Completed 25983041
Myelofibrosis NCT01200498 Phase 2 Completed
Pediatric solid and hematologic tumors NCT01184274 Phase 1 Completed
Advanced solid malignancies NCT00504296 Phase 1 Completed
fimepinostat (CUDC-907) DIPG, high-grade glioma, and medulloblastoma NCT03893487 Phase 1 Active, not recruiting
DLBCL, HGBL NCT01742988 Phase 1 Completed 28860342
Metastatic and locally advanced thyroid cancer NCT03002623 Phase 2 Terminated
Advanced Cancers NCT02307240 Phase 1 Completed
RR DLBCL NCT02674750 Phase 2 Completed
Children and young adults with RR solid tumors and lymphoma NCT02909777 Phase 1 Active, not recruiting
quisinostat (JNJ-26481585) epithelial ovarian cancer paclitaxel + carboplatin NCT02948075 Phase 2 Completed
T cell cutaneous lymphoma NCT01486277 Phase 2 Completed
SCLC and ovarian cancer standard chemotherapy NCT02728492 Phase 1 Completed
Leukemias and MDS NCT00676728 Phase 1 Terminated Sponsor decision
Advanced solid malignancies or lymphoma NCT00677105 Phase 1 Completed
MM bortezomib and dexamethasone NCT01464112 Phase 1 Completed
ricolinostat (ACY-1215) MM pomalidomide and dexamethasone NCT01997840 Phase 1/2 Active, not recruiting
Lymphoid malignancies NCT02091063 Phase 1/2 Completed 33458921
Breast cancer nab-paclitaxel NCT02632071 Phase 1 Completed 36585452
MM bortezomib and dexamethasone NCT01323751 Phase 1/2 Completed
MM pomalidomide and dexamethasone NCT02189343 Phase 1 Completed
Cholangiocarcinoma NCT02856568 Phase 1 Withdrawn Site dropped study
Gynecologic cancers paclitaxel and bevacizumab NCT02661815 Phase 1 Terminated Sponsor decision
Chronic lymphocytic leukemia ibrutinib or idelalisib NCT02787369 Phase 1 Active, not recruiting
MM lenalidomide and dexamethasone NCT01583283 Phase 1 Completed 27646843
nanatinostat (VRx-3996) EBV-positive lymphomas valganciclovir NCT05011058 Phase 2 Recruiting
EBV-positive solid tumors valganciclovir and pembrolizumab NCT05166577 Phase 1/2 Recruiting
EBV-positive lymphomas valganciclovir NCT03397706 Phase 1/2 Recruiting
Advanced Cancers NCT06302140 Phase 1 Active, not recruiting
ivaltinostat (CG200745) pancreatic adenocarcinoma capecitabine NCT05249101 Phase 1/2 Recruiting
Advanced pancreatic cancer Gemcitabine + erlotinib NCT02737228 Phase1/2 Unknown
Bocodepsin (OKI-179) Advanced Solid Tumors NCT03931681 Phase 1 Completed
MEK inhibition NCT05340621 Phase 2 Active, not recruiting
CUDC-101 Advanced solid malignancies NCT01702285 Phase 1/2 Terminated Unknown
Advanced solid malignancies NCT00728793 Phase 1 Completed
advanced head and neck, gastric, breast, liver, and NSCLC tumors NCT01171924 Phase 1 Completed
Head and neck cancers radiotherapy and cisplatin NCT01384799 Phase 1 Completed 25573383
MPT0E028 Advanced solid malignancies NCT02350868 Phase 1 Completed
BET inhibitors
Trotabresib (CC-90010) Astrocytoma, GBM NCT04047303 Phase 1 Active, not recruiting
non-hodgkins lymphoma, advanced solid tumors NCT03220347 Phase 1 Active, not recruiting
GBM temozolomide +/− radiotherapy NCT04324840 Phase 1 Active, not recruiting
pediatric solid tumors (Neuroblastoma, medulloblastoma, sarcomas) and lymphomas NCT03936465 Phase 1 Recruiting
HER2+ metastatic breast cancer Vinorelbine + radiation NCT06137651 Phase 1/2 Not yet recruiting
BMS-986158 pediatric solid tumors (Neuroblastoma, medulloblastoma, sarcomas) and lymphomas NCT03936465 Phase 1 Recruiting
Myelofibrosis alone or with ruxolitinib or fedratinib NCT04817007 Phase 1/2 Active, not recruiting
Advanced cancers +/− nivolumab NCT02419417 Phase 1/2 Completed 36077617
MM CC-92480 NCT05372354 Phase 1/2 Recruiting
PLX51107 AML, MDS azacitidine NCT04022785 Phase 1 Completed
Advanced cancers; lymphoma, AML; MDS NCT02683395 Phase 1 Terminated Sponsor Decision
AZD5153 Malignant solid tumors (Ovarian, Breast, Pancreatic, Prostate) and Lymphoma +/−olaparib NCT03205176 Phase 1 Completed
Non hodgkins Lymphoma, DLBCL acalabrutinib NCT03527147 Phase 1 Completed
AML venetoclax NCT03013998 Phase 1/2 Recruiting
SF1126 Hepatocellular carcinoma nivolumab NCT03059147 Phase 1 Terminated Lack of recruitment
Neuroblastoma NCT02337309 Phase 1 Terminated Low accrual
Metastatic Squamous Neck Cancer With Occult Primary Squamous Cell Carcinoma NCT02644122 Phase 2 Terminated Slow enrollment
Advanced Cancers NCT00907205 Phase 1 Completed
Mivebresib (ABBV-075) Breast cancer, NSCLC, AML, MM, prostate cancer, SCLC, Non-Hodgkins Lymphoma NCT02391480 Phase 1 Completed 33934351
Myelofibrosis +/− navitoclax or ruxolitinib NCT04480086 Phase 1 Terminated
Molibresib (GSK525762) AML, Non-Hodgkins lymphoma, MM NCT01943851 Phase 1 Completed 36350312
Ras-mutated cancers (SCLC, NSCLC, colorectal cancer, pancreatic cancer) trametinib NCT03266159 Phase 2 Withdrawn Unknown
ER+/HER2- breast cancer fulvestrant NCT02964507 Phase 1 Terminated
NMC, SCLC, NSCLC, colorectal cancer, neuroblastoma, CRPC, breast cancer, MYCN-driven solid tumors NCT01587703 Phase 1 Completed 34724226
CRPC androgen deprivation NCT03150056 Phase 1 Terminated protocol-defined futility
Solid tumors and lymphoma entinostat NCT03925428 Phase 1 Withdrawn Unknown
NMC chemotherapy NCT04116359 Phase 1/2 Withdrawn Unknown
birabresib (MK-8628/OTX015) NMC, Breast Cancer, NSCLC, CRPC NCT02698176 Phase 1 Terminated Limited efficacy
NMC, Breast Cancer, NSCLC, CRPC, Pancreatic NCT02259114 Phase 1 Completed 29733771
AML, DLBCL NCT02698189 Phase 1 Terminated
AML, ALL, DLBCL, MM NCT01713582 Phase 1 Completed 27063978
GBM NCT02296476 Phase 2 Terminated Limited efficacy
Newly diagnosed AML not candidate for standard chemotherapy azacitidine NCT02303782 Phase 1/2 Withdrawn
BI 894999 Advanced solid tumors or DLBCL NCT02516553 Phase 1 Completed 37556913
BAY 1238097 Solid tumors NCT02369029 Phase 1 Terminated Toxicity 30711772
pelabresib (CPI-0610) MPNST NCT02986919 Phase 2 Terminated Low enrollment
MM NCT02157636 Phase 1 Completed
Lymphoma NCT01949883 Phase 1 Completed 36923307
Advanced malignancies NCT05391022 Phase 1 Completed
Myelofibrosis +/− ruxolitinib NCT02158858 Phase 2 Active, not recruiting
Myelofibrosis +/− ruxolitinib NCT04603495 Phase 3 Active, Not recruiting
INCB054329 Advanced malignancies NCT02431260 Phase1/2 Terminated PK variability 31527168
INCB057643 Myelofibrosis +/− ruxolitinib NCT04279847 Phase 1 Recruiting
Advanced solid malignancies select agents NCT02711137 Phase 1/2 Terminated Safety concerns 31527168
Advanced solid malignancies pembrolizumab and epacadostat NCT02959437 Phase 1/2 Terminated Sponsor Decision 37087488
FT-1101 AML, MDS, NHL +/− azacitidine NCT02543879 Phase 1 Completed
ODM-207 Advanced solid malignancies NCT03035591 Phase 1/2 Completed 32989226
ZEN-3694 Advanced solid malignancies chemotherapy + capecitabine NCT05803382 Phase 1 Recruiting
Metastatic CRPC enzalutamide, pembrolizumab NCT04471974 Phase 2 Recruiting
Advanced solid malignancies Talazoparib NCT05327010 Phase 2 Recruiting
Recurrent ovarian and endometrial cancers M1774 NCT05950464 Phase 1b Recruiting
TNBC Pembrolizumab, standard chemotherapy NCT05422794 Phase 1b Recruiting
Advanced solid tumors with RAS alterations and TNBC Binimetinib NCT05111561 Phase 1 Recruiting
NMC Etoposide + Cisplatin NCT05019716 Phase1/2 Recruiting
NMC, other solid tumors abemaciclib NCT05372640 Phase 1 Recruiting
Advanced solid tumors and lymphomas Entinostat NCT05053971 Phase1/2 Recruiting
TNBC Talazoparib NCT03901469 Phase 2 Active. Not recruiting
Solid tumors Nivolumab +/− ipilimumab NCT04840589 Phase1/1b Recruiting
CRPC Enzalutamide NCT04986423 Phase 2b Recruiting
CRPC NCT02705469 Phase 1 Completed
CRPC Enzalutamide NCT02711956 Phase 1b/2a Completed 32694156
Squamous cell lung cancer NCT05607108 Phase 2 Recruiting
Ovarian cancer, Peritoneal cancer, fallopian tube cancer talazoparib NCT05071937 Phase 2 Recruiting
CRC in addition to chemo + cetuximab + encorafenib NCT06102902 Phase 1 Recruiting
Advanced solid malignancies Plus Niraparib NCT06161493 Phase 1 Recruiting
RO6870810 MM +/− daratumumab NCT03068351 Phase 1b Completed
Advanced ovarian or TNBC atezolizumab NCT03292172 Phase 1b Terminated Portfolio prioritization
AML, MDS NCT02308761 Phase 1 Completed 33586590
Advanced solid tumors NCT01987362 Phase 1 Completed 33311588
DLBCL, high-grade B-cell lymphoma Venetoclax +/− rituximab NCT03255096 Phase 1b Completed 34581757
EZH2 inhibitors
Tazemetostat * epithelioid sarcoma and EZH2 mutant follicular lymphoma FDA approved
ARID1A mutant cancers NCT05023655 Phase 2 Recruiting
malignant mesothelioma NCT02860286 Phase 2 Completed 35588752
MPNST NCT04917042 Phase 2 Recruiting
INI1-negative solid tumors or synovial sarcoma NCT02601950 Phase 2 Active, not recruiting
INI1-negative solid tumors or synovial sarcoma NCT02601937 Phase 1 Completed
B-cell NHL with EZH2 mutations NCT03456726 Phase 2 Completed 34159682
Gynecologic cancers NCT03348631 Phase 2 Active, not recruiting
GSK2816126 DLBCL, Follicular lymphoma, MM NCT02082977 Phase 1 Terminated No efficacy at MTD 31471312
PF-06821497 SCLC, Follicular lymphoma, CRPC NCT03460977 Phase 1 Recruiting
SHR2554 Relapsed/Refractory mature lymphoid malignancies NCT03603951 Phase 1 Recruiting
CRPC SHR3680 NCT03741712 Phase 1/2 Terminated Sponsor decision
Advanced solid tumors and B-cell lymphomas SHR1701 NCT04407741 Phase 1/2 Recruiting
Relapsed/Refractory NHL SHR1701 NCT05896046 Phase 1/2 Recruiting
Relapsed/Refractory PTCL Umbrella study NCT05559008 Phase 1/2 Recruiting
Advanced Breast Cancer Umbrella study NCT04355858 Phase 2 Recruiting
TNBC Umbrella study NCT03805399 Phase 1/2 Recruiting
T-cell lymphoma Compared versus chidamide NCT06122389 Phase 3 Recruiting
Peripheral T-cell lymphoma (treatment-naïve) With cheomtherapy NCT06173999 Phase 1b/2 Not yet recruiting
CPI-1205 B-cell Lymphomas NCT02395601 Phase 1 Completed
Advanced Solid tumors NCT03525795 Phase 1/2 Completed
CRPC NCT03480646 Phase 1/2 Unknown
Valemetostat (DS-3201b) Sinonasal cancer, squamous NSCLC, Squamous Cell Carcinoma, Head and Neck Cancers Pembrolizumab NCT05879484 Phase 1/2 Not yet recruiting
Relapsed/Refractory PTCL, adult T cell leukemia/lymphoma NCT04703192 Phase 2 Active, not recruiting
B-cell lymphoma NCT04842877 Phase 2 Active, not recruiting
T-cell leukemia/lymphoma NCT04102150 Phase 2 Active, not recruiting
prostate, urothelial, renal cell carcinomas ipilimumab NCT04388852 Phase 1 Recruiting
FL Rituximab, Lenalidomide NCT05683171 Phase 1/2 Recruiting
HER2-low breast cancer Trastuzumab NCT05633979 Phase 1 Recruiting
Hepatocellular carcinoma With atezolizumab and bevacizumab NCT06294548 Phase 1b/2 Not yet recruiting
Advanced solid tumors With DXd ADC NCT06244485 Phase 1 Not yet recruiting
Relapsed or refractor NHL With CC-99282 NCT03930953 Phase 1/2 Recruiting
HH2853 NHL, advanced solid tumors NCT04390737 Phase 1/2 Recruiting
EED inhibitor
MAK683 DLBCL and solid tumors NCT02900651 Phase 1/2 Active, not recruiting
APG-5918 Advanced Solid Tumors and Lymphomas including SMARCB1 mutated sarcomas and EZH2 mutant B cell lymphomas NCT05415098 Phase 1 Recruiting
ORIC-944 Metastatic Prostate Cancer NCT05413421 Phase 1 Recruiting
SMARCA2/4 inhibitor
FHD-286 Metastatic Uveal Melanoma NCT04879017 Phase 1 Active, not recruiting
AML, MDS NCT04891757 Phase 1 Recruiting
SMARCA2 degrader
PRT3789 Advanced solid tumors with SMARCA4 mutation Monotherapy and in como with docetaxel NCT05639761 Phase 1 Recruiting
BRD9 degrader
FHD-609 Synovial Sarcoma NCT04965753 Phase 1 Active, not recruiting
CFT8634 SMARCB1-perturbed cancers NCT05355753 Phase1/2 Terminated No efficacy in highly treated patients
DOT1L inhibitor
pinometostat (EPZ-5676) MLL-r leukemias Azacitidine NCT03701295 Phase 1 completed
MLL-r AML Daunorubicin, Cytarabine NCT03724084 Phase 1/2 Terminated Unknown
MLL-r leukemias NCT02141828 Phase 1 Completed
MLL-r leukemias NCT01684150 Phase 1 Completed 29724899
Menin inhibitors
BMF-219 AML, ALL With KMT2A/MLL1r, NPM1 NCT05153330 Phase 1 Recruiting
KRAS mutant CRC, NSCLC, PDAC NCT05631574 Phase 1 Recruiting
Revumenib (SNDX-5613) Acute Leukemia NCT05406817 Phase 1 Recruiting
Acute Leukemia Chemotherapy NCT05326516 Phase 1 Active, not recruiting
AML with MLL Rearrangement or NPM1 Mutation Cobicistat NCT04065399 Phase 1/2 Recruiting
AML Venetoclax, ASTX727 NCT05360160 Phase 1/2 Recruiting
Untreated AML - genome screening master study Many drugs of which SNDX-5613 is one NCT03013998 Phase 1/2 Recruiting
AML Combination with 7+3+midostaurin NCT06313437 Phase 1 Not yet recruiting
Leukemias associated with HOX cluster genes NCT06229912 Phase 2 Not yet recruiting
Pediatric and young adult AML NCT06177067 Phase 1 Not yet recruiting
AML In combination with BCL2 inhibitor NCT06284486 Phase 2 Not yet recruiting
CRC and other solid tumors NCT05731947 Phase 1/2 Recruiting
AML NCT06226571 Phase 1 Not yet recruiting
AML Standard chemotherapy for newly diagnosed patients with NPM1 or MLL alterations NCT05886049 Phase 1b Recruiting
AML And giltertinib in FLT3 mutated AML with concurrent MLL or NPM1 alterations NCT06222580 Phase 1 Not yet recruiting
AML NCT05761171 Phase 2 Active, not recruiting
Ziftomenib (KO-539) relapsed or refractory AML NCT04067336 Phase 1/2 Recruiting
AML Combination with venetoclax/azacitidine, venetoclax, or 7+3 NCT05735184 Phase 1 Recruiting
AML Various combinations NCT06001788 Phase 1 Recruiting
JNJ-75276617 Acute Leukemia NCT04811560 Phase 1 Recruiting
AML with MLL Rearrangement or NPM1 Mutation Venetoclax, Azacitidine NCT05453903 Phase 1/2 Recruiting
AML with MLL Rearrangement, NPM1 Mutation, Nucleoporin Mutation Chemotherapy NCT05521087 Phase 1 Not yet recruiting
DSP-5336 Acute Leukemia NCT04988555 Phase 1/2 Recruiting
DS-1594b Acute Leukemia Azacitidine, Venetoclax, mini-HCVD NCT04752163 Phase 1/2 Completed
P300/CBP inhibitors
FT-7051 CRPC NCT04575766 Phase 1 Terminated Sponsor decision
Inobrodib (CCS1477) NHL, MM, AML, MDS alone or with cancer-specific regimens NCT04068597 Phase 1/2a Recruiting
CRPC, NSCLC, Breast cancer, advanced solid tumors alone or in combination with select cancer-specific drug regimens NCT03568656 Phase 1/2a Recruiting
EP31670/NEO2734 CRPC, NMC NCT05488548 Phase 1 Recruiting
KAT6A inhibitor
PF-07248144 ER+ / HER2- breast cancer, NSCLC, CRPC Fulvestrant, Letrozole, Palbociclib NCT04606446 Phase 1 Recruiting
LSD1 inhibitors
Iadademstat (ORY-1001) FLT3 mutant AML Gilteritinib NCT05546580 Phase 1 Recruiting
Relapsed SCLC In combination with paclitaxel NCT05420636 Phase 2 Recruiting
SCLC In combination with atezolizumab or durvalumab NCT06287775 Phase 1/2 Not yet recruiting
GSK2879552 MDS Azacitidine NCT02929498 Phase 1 Terminated The risk benefit in MDS does not favor continuation of the study
SCLC NCT02034123 Phase 1 Terminated The risk benefit in SCLC does not favor continuation of the study 31260835
AML All-Trans Retinoic Acid (ATRA) NCT02177812 Phase 1 Terminated The risk benefit in relapsed refractory AML does not favor continuation of the study
Bomedemstat (IMG-7289 / MK-3543) Myelofibrosis NCT05223920 Phase 2 Active. Not recruiting
Myelofibrosis Ruxolitinib NCT05569538 Phase 2 Recruiting
SCLC Atezolizumab NCT05191797 Phase 1/2 Recruiting
AML Venetoclax NCT05597306 Phase 1 Recruiting
Advanced myeloid malignancies With or without ATRA NCT02842827 Phase 1/2 Completed
INCB059872 Ewing Sarcoma NCT03514407 Phase 1 Terminated Strategic Business Decision
Solid Tumors, Hematologic Malignancies ATRA, Azacitidine, Nivolumab NCT02712905 Phase 1/2 Terminated Strategic Business Decision
Solid Tumors Pembrolizumab, Epacadostat NCT02959437 Phase 1/2 Terminated Study terminated by Sponsor
Seclidemstat (SP-2577) Solid Tumors NCT03895684 Phase 1 Completed
Gynecologic Cancers with SWI/SNF pathway mutations Pembrolizumab NCT04611139 Phase 1 Withdrawn Salarius discontinued support
Ewing or Ewing-related Sarcomas Cyclophosphamide, Topotecan NCT03600649 Phase 1 Active. Not recruiting
Ewing or Ewing-related Sarcomas NCT05266196 Phase 1/2 Enrolling by invitation
MDS, Chronic Myelomonocytic Leukemia Azacitidine NCT04734990 Phase 1/2 Recruiting
CC-90011 Prostate Cancer Abiraterone, Prednisone NCT04628988 Phase 1 Completed
SCLC, Squamous NSCLC Nivolumab NCT04350463 Phase 2 Completed
SCLC Cisplatin, Etoposide, Carboplatin, Nivolumab NCT03850067 Phase 1 Active, not recruiting
AML Venetoclax, Azacitidine NCT04748848 Phase 1 Terminated Business objectives have changed
Solid Tumors, Non-Hodgkin’s Lymphomas Rifampicin, Itraconazole NCT02875223 Phase 1 Active, not recruiting
NSD2 inhibitors
KTX-1001 t(4;14) and NSD2 mutant multiple myeloma NCT05651932 Phase 1 Recruiting
Acronym Disease
DLBCL Diffuse large B-cell lymphoma
AML Acute Myeloid Leukemia
MDS Myelodysplastic syndromes
MM Multiple myeloma
NSCLC Non-Small Cell Lung Cancer
SCLC Small Cell Lung Cancer
CRPC Castration-resistant prostate cancer
NMC NUT midline carcinoma
ALL Acute lymphocytic leukemia
GBM Glioblastoma Multiforme
MPNST Malignant Peripheral Nerve Sheath Tumor
DIPG Diffuse intrinsic pontine glioma
HGBL High-grade B-cell lymphoma
PTCL Peripheral T-cell cutaneous lymphoma
*=

FDA-approved drugs, Only approved indications are shown

**=

investigational drugs for which there are ≥ 20 ongoing clinical trials. Only phase 3 trials shown in this table for these drugs.

The limited utility of DNMT and HDAC inhibitors for treating solid tumors highlights the double-edged sword of targeting common essential genes, which have a narrow therapeutic window in part because target proteins are crucial for the survival of both healthy cells and cancer cells. Minimizing toxicity of single-agent DNMT inhibitors may be difficult due the essentiality of DNMT1. However, selective targeting of HDACs may be achievable due to the large number of redundancies between HDACs enzymes. The therapeutic efficacy of HDAC inhibitors is likely mediated through inhibition of Class I HDACs (HDACs1-3,8), although the specificity for individual HDAC members varies between small molecules and off-target effects may partially mediate additional therapeutic effects16,17. Only HDAC3 is predicted to be common essential and plays a crucial role in chromatin remodeling and gene regulation in most cells/tissues. In contrast HDAC1 and HDAC2 are paralogs that induce cell death only when knocked out (or inhibited) together18. Therefore, selective inhibition of either of the paralogs HDAC1 or HDAC2 could provide therapeutic benefit while minimizing toxicity, as some disease lineages have shown enriched dependency on a single paralog10,19,. Further, recent compounds, such as TNG260, could inhibit HDACs in certain chromatin-modifying complexes (e.g., CoREST), which may overcome some toxicity by enabling an additional level of specificity.

Bromodomain and Extra-terminal Domain Protein Inhibitors

The BET protein family consists of members BRD2, BRD3, BRD4, and BRDT, although BRD4 is most investigated as a cancer target. BRD4 is a common essential gene, suggesting that pan-BET inhibitors would result in significant off-tumor toxicity20. BRD4 binds acetylated lysine residues, recruits the transcriptional elongation complex P-TEFb, and enables contact with the Mediator complex to facilitate transcription through RNA polymerase II21. Oncoproteins, such as c-Myc or N-Myc, have been shown to co-localize with BRD4 to drive cancer cell proliferation. Inhibition of BET proteins via the blocking of their bromodomains blocks the expression of MYC22 or MYCN23 mRNA itself, and collapses oncogenic transcriptional networks, suggesting a therapeutic window for BET inhibition. BET inhibition also downregulates transcriptional networks associated with other oncogenic transcription factors such as EWS-FLI124, BRD3/4-NUT25, and MLL9-AF926, suggesting broad utility for cancers dependent on transcription factor-driven proliferation.

The evaluation of BET inhibitors has demonstrated a requirement of BRD4 to maintain oncogenic transcription, and activity is observed in numerous preclinical models of cancer25,27. Several highly potent and selective BET inhibitors have been developed for study in clinical trials (Figure 1 and Table 1). BET inhibitors have a clear genetic indication for their use in BRD3/4-NUT fusions in midline NUT carcinomas, and clinical trials have demonstrated clinical benefit for a subset of these patients28,29. Responses, however, have generally been transient, and BET inhibitors have faced the challenge of dose-limiting hematological toxicity, particularly thrombocytopenia, limiting their activity as single agents in most other malignancies30,31. Therefore, caution is required in the choice of combination therapies with BET inhibitors, particularly those with overlapping hematologic toxicities.

Targeting Cancer-Selective Dependencies and Synthetic Lethalities Involving Chromatin Regulation

The gold standard for therapeutic targets is cancer-selective or synthetic lethal dependencies for which reliance can be predicated based on cancer context. Somatic mutations resulting in gain-of-function (GOF) of epigenetic modifiers are among the most enriched groups of mutated proteins, and specific mutations have been well covered in other reviews32. The high frequency makes these activating mutations attractive therapeutic targets, and indeed there is an FDA approval in this case (EZH2 inhibitors in EZH2 mutant follicular lymphoma)33. Synthetic lethal-like dependencies provide another target class. The loss of a protein’s function, either through loss-of-function (LOF) mutations or epigenetic silencing, can lead to reliance on the function of another protein, often a protein paralog or a downstream signaling mechanism, to maintain viability. For example, epigenetic targets, such as P300, are of interest in cancers harboring mutations in its histone acetyltransferase paralog CREBBP34. Further, recent data suggest DNMT3A mutations or TET2 mutations in AML cells predispose to response to 5-azacytidine35,36.

Chromatin complexes often maintain epigenetic control by balancing two opposing activities. The inactivation of one epigenetic complex can create a reliance on the unopposed activity of another chromatin complex network. In the sections below, we will discuss evidence of such “epigenetic antagonism” where LOF of one epigenetic protein leads to dependence on an alternative epigenetic complex (Figure 2A). For example, EZH2 inhibitors are approved for such an instance (e.g., SMARCB1-deficient epithelioid sarcomas)37. In sum, these cancer-selective and synthetic lethal relationships exploit a vulnerability that is unique to the cancer and might mitigate off-tumor toxicities.

Figure 2: Therapeutic Targeting of the Epigenome.

Figure 2:

A) Wild-type (WT) BAF complex evicts PRC1/PRC2 from chromatin to establish open chromatin regions, while loss of BAF activity (e.g., through loss-of-function (LOF) mutations) results in unopposed PRC2 signaling. B) Epigenetic proteins participate in oncogenic signaling by associating with oncogenic transcription factors.

Polycomb Repressive Complex Inhibitors

With recent advances in our understanding of the connection between genetic drivers of cancer and epigenetic regulation, the Polycomb Repressive Complex 2 (PRC2) has emerged as an important therapeutic target. PRC2, comprised of the core protein subunits EED, SUZ12, and EZH2, is responsible for the mono-, di-, and tri-methylation of histone 3 lysine 27 (H3K27)38,39. Polycomb-dependent gene repression is a critical determinant of lineage commitment, differentiation, and cell identity.

The functional role of PRC2 in oncogenic signaling is complex, with both oncogenic and tumor suppressive activity observed depending on the cell of origin. GOF mutations in the EZH2 subunit are observed in DLBCL, follicular lymphomas, and in rare subsets of hepatosplenic T-cell lymphomas, melanomas, and Ewing sarcomas4044. EZH2 overexpression also occurs in numerous cancers and is associated with poorer prognosis45,46. As such, EZH2 knockout has been shown to induce differentiation and reduce cancer cell viability in many pre-clinical models45,4749. PRC2 is opposed by trithorax protein complexes, including the chromatin remodeling BRG1/BRM-associated factor (BAF) complex. The BAF complex quickly evicts PRC2 that is reversible with loss of BAF activity (Figure 2A)50. LOF mutations in certain members of the BAF complex, including SMARCB1 and ARID1A mutations lead to unopposed PRC2 activity that results in dependence on PRC24952. These are all examples where inhibition of EZH2 by small molecules are shown to be beneficial. In contrast, deletions or LOF mutations in EED, EZH2, or SUZ12 have been observed in approximately 25% of T-cell acute lymphoblastic leukemias53, almost half of early T-cell precursor acute lymphoblastic leukemias54, in some myeloid malignancies55,56, and in the majority of malignant peripheral nerve sheath tumors57. Lysine-to-methionine mutations in histone 3 (H3K27M) in pediatric diffuse midline gliomas also alter EZH2 activity58. The distinctive roles of PRC2 in different tumor lineages suggest tissue-specific functions of PRC2.

Small molecule discovery programs to identify inhibitors of PRC2 have been extensive. The SET domain of EZH2 utilizes the cofactor S-adenosyl-methionine (SAM) to facilitate H3K27 methylation and EZH2 inhibitors compete with SAM to inhibit PRC2 activity in both EZH2 wild-type and mutant cell lines47. The EZH2 inhibitor tazemetostat has been approved in the United States for EZH2 mutant follicular lymphoma and epithelioid sarcomas with LOF SMARCB1 mutations based on positive clinical studies33,37. While LOF EZH2 mutations are observed in T cell lymphoblastic leukemias and myeloid malignancies,53,59 there has been only one case reported of a secondary T-cell malignancy during treatment with an EZH2 inhibitor. Nevertheless, as these inhibitors become more commonly used it will be important to report any identified cases of secondary malignancy. Other EZH2 inhibitors are also being evaluated in clinical studies (Figure 1 and Table 1). Interestingly, valemetostat inhibits both EZH1 and EZH2 and is suggested to induce greater antitumor effects than EZH2 alone60. In addition to EZH2 inhibitors, first-in-class EED inhibitors bind and block the H3K27me3 recognition pocket of EED to disrupt protein-protein interactions between PRC2 subunits, including both EHZ1 and EZH2 (Figure 1 and Table 1). MAK683 is currently being evaluated in DLBCL and nasopharyngeal carcinoma, with early reports suggesting signs of activity in DLBCL61. ORIC-944 is currently being studied in metastatic prostate cancer, and APG-5918 is being evaluated across numerous advanced solid tumors, including SMARCB1-mutant sarcomas and EZH2-mutant B-cell lymphomas.

BAF Complex Inhibitors

The ATP-dependent chromatin remodeling BAF complex (SWI/SNF complex in yeast) is crucial for the regulation of gene expression and differentiation and antagonizes the repressive PRC2 complex.50 The BAF family consists of three large, multimeric protein complexes: canonical BAF (cBAF), polybromo-associated BAF (pBAF), and non-canonical BAF (ncBAF)62. Each complex includes one of two DNA-stimulated ATPase catalytic subunits (SMARCA4 or SMARCA2), which are responsible for nucleosome displacement and transcriptional activation. A deficiency in either SMARCA2 or SMARCA4 results in a synthetic lethal paralog related dependency on the other subunit63,64. Mutations in BAF are abundant in cancer; many subunits of the BAF complex can be altered, including BRD9, ARID1A/B, PBRM1, ARID2, SMARCC1, SMARCC2, SS18, and SMARCB1. Mutations and alterations in BAF complex family members result in unique residual complex compositions that alter chromatin accessibility and dynamics, which likely underlies their oncogenic potential65. Alternatively, these alterations can incur synthetic lethal-like dependencies in various cancers.

A small molecule allosteric inhibitor of the SMARCA2/4 ATPases, FHD-286 (patent US 2023/0138480), is currently in clinical evaluation for AML and myelodysplastic syndrome (NCT04891757) and metastatic uveal melanoma (NCT04879017) (Figure 1 and Table 1). The clinical trial evaluating patients with AML was halted after a patient receiving FHD-286 showed significant toxicity from differentiation syndrome, likely due to on-target inhibition of SMARCA2/4, suggesting SMARCA2/4 may hold promise as a therapeutic target in AML. However, because the inhibitor targets both SMARCA2 and SMARCA4 paralogs, it raises questions as to whether prolonged inhibition will be cytotoxic to all cells, including healthy tissue. Ideally, an inhibitor would selectively target either SMARCA2 or SMARCA4 to best leverage the synthetic lethal relationship, such compounds are now in active development (Figure 1 and Table 1)66,67. Further, BAF subunits, including SMARCA2 and SMARCA4, are mutated in as many as 20% of cancers and are considered tumor suppressors in many contexts; thus more work is warranted with regards to evaluating the risk of secondary malignancy with SMARCA2/4 inhibitors68.

While targeting the obligate ATPase activity of the BAF complex is predicted to be lethal to all cells, targeting selective subunits that are dependencies in certain contexts seems promising. Recent work has demonstrated that perturbation of the cBAF complex can lead to dependency on the ncBAF complex and more specifically on the ncBAF complex subunit, BRD9.69,70 In this case, biallelic inactivation of SMARCB1, a subunit of the cBAF complex, portends a dependency on BRD9 in malignant rhabdoid tumors69,70. In an alternate mechanism, synovial sarcoma is defined by a rearrangement that creates the SS18-SSX fusion protein, which displaces and degrades SMARCB1, mimicking LOF SMARCB171. In both cases, SMARCB1 LOF is compensated for by the ncBAF complex, thereby resulting in a synthetic lethality on the ncBAF complex subunit BRD969,72. The clinical candidate PROTACs CFT863473 and FHD-609 (patent US 2023/0142883) degrade BRD9 (Figure 1) and are currently being studied in SS18-SSX fusion synovial sarcoma and other tumors with SMARCB1-loss (Table 1). Notably, enrollment was recently paused for FHD-609 due to reported QTc prolongation, a risk factor for cardiac arrythmia, which was observed at higher doses of the drug.

Targeting of Oncoprotein Complexes

The most direct and effective approach to alter oncogenic gene expression programs is to directly target the activity of the mutant/altered transcription factors that drive cancer. However, drugging these proteins remains challenging. One method to circumvent this problem is to target the critical co-factors and chromatin complexes that are necessary for their oncogenic function (Figure 2B). This is the case in a subset of acute leukemias with KMT2A (MLL1) rearrangements. While KMT2A itself is a histone methyltransferase, the fusion oncoprotein excludes the SET domain of KMT2A, activating leukemic gene expression independent of histone methylation74, typically due to rearrangements with mediators of transcriptional elongation such as AF4, AF9, ENL, and AF1075 To date, several indirect targeting strategies have been developed to block the KMT2A-fusion protein through targeting of its cofactors. These include small molecules targeting Menin76 and DOT1L,77 which reverse leukemic gene expression and exhibit therapeutic effects in pre-clinical leukemia models. While the concept of targeting leukemic gene expression through inhibition of chromatin complexes is well developed in KMT2A-r AML and ALL, it is becoming increasingly clear that similar mechanisms are at play in other leukemias, including NPM1 mutant and NUP98-rearranged AML78,79.

DOT1L Inhibitors

The histone methyltransferase DOT1L complexes with numerous proteins and is responsible for the mono-, di-, and trimethylation of H3K7980,81. DOT1L-dependent H3K79 methylation is thought to be required for transcriptional elongation at active gene loci80. DOT1L is a non-SET domain enzyme that utilizes SAM as a cofactor to perform its catalytic function82. A subset of acute leukemias harboring KMT2A-r or NPM1 mutations is defined by the overexpression of transcription factors in the HOXA/B cluster and their co-factor MEIS1. These transcription factors reprogram the epigenome to promote leukemogenesis83. The most prominent known role for DOT1L is in KMT2A-r leukemias where aberrant H3K79me2 at KMT2A-r-bound sites is a central feature. The small molecule pinometostat potently inhibits DOT1L activity through occupation of the SAM binding site, and induced differentiation of KMT2A-r leukemia and reduced KMT2A-r leukemia in preclinical models84. Despite the multiple pre-clinical DOT1L inhibitors studied, only pinometostat has been tested in the clinic (Figure 1 and Table 1). An early phase clinical trial demonstrated proof-of-concept clinical remissions as a single agent and general tolerability85. However, response rates have been modest and the continuous 28-day infusion of pinometostat is burdensome. Thus, if DOT1L inhibition is to be pursued as a pharmacologic strategy moving forward, pharmacologic inhibitors with more robust pharmacokinetic properties will be required.

Menin Inhibitors

Menin, initially reported as a scaffolding protein and recently implicated as a reader of H3K79me286, is encoded by the Multiple Endocrine Neoplasia 1 (MEN1) gene and is ubiquitously expressed across tissues. Menin binds KMT2A/MLL1 via its N-terminus Menin binding domain and is an essential subunit of the MLL1/2 COMPASS-like complex, a H3K4 histone methyltransferase. The MLL1/2 COMPASS-like complex is required for the maintenance of MEIS1 and the HOX gene expression in AML, and inhibition of Menin significantly reduced tumor burden and induced differentiation in pre-clinical models79,87. Numerous studies have shown that subsets of genetically defined acute leukemias, namely those with KMT2A-rearrangements, NPM1 mutations or NUP98-rearrangements, strongly depend on Menin to promote MEIS1/HOXA transcription78,79,87.

The strong support for Menin inhibitors in hematological malignancies has rapidly expanded the number of clinical studies evaluating their safety and efficacy (Table 1). Numerous small molecule inhibitors, which bind the Menin binding pocket of KMT2A, thereby blocking the physical interaction between Menin and KMT2A have been developed (Figure 1A)88,89. In acute leukemias, this disruption evicts Menin/KMT2A from chromatin and typically downregulates transcription of HOXA genes and MEIS1 in acute leukemias that are dependent on Menin in both the KMT2A-r and KMT2A wild-type settings79,88. Early clinical data for revumenib showed 59% overall response rates in KMT2A-r leukemias and 36% in NPM1 mutant leukemias90. Early clinical reports for ziftomenib showed an overall response rate of 42% and 75% in patients that exhibit differentiation syndrome91. Menin inhibitors are orally bioavailable and reported as generally well tolerated. However, serious adverse events have been identified, including asymptomatic prolongation of the QTc interval as a dose-limiting toxicity specifically with revumenib90. Differentiation syndrome, managed by corticosteroids and hydroxyurea, was also reported for both revumenib and ziftomenib, and is consistent with on-target activity of agents that induce AML differentiation90. Menin inhibition has been shown to have activity in some pre-clinical solid tumor contexts9294, but clinical trials in these disease contexts have not yet been reported.

Recent evidence from the AUGMENT-101 (NCT04065399) study in patients treated with revumenib suggests that resistance to Menin inhibition occurs after approximately two cycles of treatment95. Patients harboring MEN1 mutations at residues M327, G331, T349, and S160 demonstrated reduced drug affinity across multiple inhibitors in vitro without affecting the Menin-KMT2A interaction. Interestingly, all these mutations observed in patients were predicted both using unbiased base-editing CRISPR-Cas9 based screening of the MEN1 gene in vitro and in PDX samples treated in vivo. This is the first epigenetic therapy to our knowledge whereby clinical resistance in patients has been demonstrated to be mediated through somatic mutations in the drug target. These data provide gold standard evidence for the on-target mechanism of drug action and evidence for strong dependency of these disease subsets on Menin. As with resistance to tyrosine kinase inhibitors, overcoming resistance to first-generation Menin inhibitors by increasing affinity for the altered binding pocket with second-generation drugs may be possible.

Targeting Non-Essential Genes Involved in Transcription

The epigenetic cancer dependencies described above have fueled the development of highly selective and potent inhibitors targeting epigenetic complexes. Other epigenetic targets, such as the histone acetylase P300 and histone demethylase LSD1, demonstrate indiscriminate efficacy across numerous cancer lineages, although do not quite cross the threshold of being considered pan-essential, and thus may demonstrate fewer dose-limiting toxicities than cytotoxic chemotherapies.

Histone Acetyltransferase Inhibitors

The acetylation of histones is a dynamic process that is tightly regulated by the activity of histone acetyltransferases (HATs) and HDACs. Acetylation of lysines, including H3K9 and H3K18 at gene promoters and H3K27 at gene enhancers and promoters, is associated with gene transcription. Enzymatic HAT subunits are part of larger multimeric complexes, and the other members of the complex can impact HAT specificity and localization96. There are three main HAT complex groups that regulate the acetylation of core histones: MYST (KAT5, KAT6A/B, KAT7, KAT8), GCN5/PCAF (KAT2A/KAT2B), and CBP/EP300 (CREBBP/EP300). While HATs were originally discovered for their histone acetyltransferase activity, this has become a misnomer as a growing number of non-histone targets of these acetyltransferases have been discovered97.

Targeting HATs is a relatively new strategy compared to HDACs, but nonetheless there is growing evidence these targets could have a role in cancer therapy. The CBP/P300 family acetylates H3K18 and H3K27 and has also received interest in cancers dependent on super-enhancer driven transcriptional programs. CBP/P300 function as coactivators of transcription factors, such as androgen receptor in prostate cancer98. EP300 knockout alone has broad activity across numerous cancer lineages, but is not considered pan-essential, perhaps due to a partial compensation from CBP activity as CREBBP mutations are associated with stronger EP300 dependency34. Confirming the synthetic lethal relationship, CREBBP dependency is pronounced in DLBCL harboring damaging mutations in EP30034,99. However, while both proteins acetylate histones, they likely also have some non-redundant roles, as genetic knockout of EP300 is notably less tolerated than knockout of CREBBP100. There are two pharmacologic classes of CBP/P300 inhibitors, enzymatic HAT inhibitors and small molecule bromodomain inhibitors that block CBP/P300 activity101,102. Head-to-head comparisons of HAT versus bromodomain inhibitors have yet to be performed. It is likely that off-target effects specific to each class of compounds could enable greater activity in specific subsets of disease. For example, CBP/P300 bromodomain inhibitors may inhibit other bromodomain-containing proteins (e.g., BRD4) and could be more effective in BET-inhibitor response contexts such as midline NUT carcinoma103. Despite showing pre-clinical efficacy, HAT domain inhibitors have not yet been successfully bridged to clinical trials. In contrast, two CBP/P300 bromodomain inhibitors are being tested in clinical trials. CCS1477 (inobrodib) is currently in Phase I clinical trials for both solid and hematologic malignancies and FT-7051 is being studied for metastatic castration-resistant prostate cancer (Figure 1 and Table 1).

P300-selective, CBP-selective, and pan-targeting PROTACs, resulting in proteolysis of the target protein, have also been developed100,104,105. As compared to the catalytic inhibitor, a P300-selective PROTAC demonstrated rapid apoptotic cell death rather than cell cycle arrest, suggesting that P300 has catalytic-independent functions that can only be targeted through degradation100. Thus, deploying new pharmacologic modalities (i.e., degradation) may provide unique functions as compared to their catalytic inhibitor counterparts and would more closely resemble phenotypes observed with CRISPR knockouts (e.g., DepMap). However, PROTAC structures are much larger than typical small molecule catalytic inhibitors and thus may suffer from challenges related to solubility, permeability, and pharmacokinetics.

While the therapeutic utility of CBP/P300 is effective across numerous cancers due to the requirement for their activity to maintain cell viability, other HATs, such as the KAT6A/KAT6B enzymes in the MYST family, have an enriched dependency based on cancer lineage and/or genetics. Recurrent translocations involving KAT6A and KAT6B occur in AML,106 and KAT6A has been shown to be a critical driver of the stem cell-like properties in AML107. Amplifications of KAT6A and/or KAT6B also occur in solid tumors, including breast, colon, lung, endometrial, and ovarian cancers and medulloblastoma108111. KAT6A/KAT6B are required for suppression of senescence through locus-specific acetylation of H3K9 and H3K23, although the mechanism by which this occurs and whether the KAT6A/B inhibition phenotype is mediated through histone acetylation is still unclear112,113. Selective catalytic inhibitors targeting KAT6A/B have now been reported112,114, and the KAT6A/B inhibitor PF-07248144 is being investigated in ER+ breast cancer (Figure 1 and Table 1).

The remaining class of HATs include KAT2A/KAT2B and are commonly associated with acetylation of H3K9 and H3K14115,116. While inhibitors targeting KAT2A/KAT2B have not yet entered clinical trials, there is pre-clinical evidence that KAT2A HAT activity is a key activator of MYC transcription in some MYC-driven cancers117,118 and we speculate it will likely be an activator in some MYCN-driven cancers. In addition to catalytic and bromodomain inhibitors, more recently, drug discovery efforts against KAT2A/B HATs have expanded to include PROTACs, which have now been investigated pre-clinically119.

Histone Demethylase Inhibitors

Histone demethylases can broadly be divided into two families: flavin adenine dinucleotide (FAD)-dependent (KDM1) and 2-OG-dependent JmjC domain-containing histone demethylases120. Of the 20+ histone demethylases, only chemistry efforts targeting the FAD-dependent family member LSD1/KDM1A have successfully transitioned into clinical trials. LSD1 complexes with the NuRD and CoREST complexes to demethylate H3K4me1/me2121,122 and with nuclear hormone receptors to demethylate H3K9me1/me2123, resulting in inactivation or activation of transcription, respectively. Inhibition of LSD1 is efficacious in pre-clinical models of AML and small cell lung cancer (SCLC),124126 and DepMap predicts the greatest dependency on LSD1 in AML across the over 1000 cancer cell line models screened9. A subset of SCLCs that are defined by neuroendocrine-like lineage and highly express ASCL1 are strongly responsive to LSD1 inhibitors by decreasing neuroendocrine expression patterns and increasing NOTCH signaling in preclinical studies125. Similarly, LSD1 promotes stemness features and lineage plasticity in prostate cancer127, suggesting that histone demethylase inhibitors may have a broad role in preventing mechanisms of neuroendocrine plasticity across numerous cancer types.

Pre-clinical success of LSD1 inhibitors in SCLC and AML advanced the testing of potent LSD1 inhibitors in clinical studies (Table 1). Encouraging data was seen in relapsed/refractory AML with Iadademstat128. Clinical trials evaluating Iadademstat in tumors with neuroendocrine plasticity, such as prostate cancer and SCLC, are set to begin shortly. Bomedemstat is being evaluated for efficacy in combination with anti-PD1 immunotherapy as upfront therapy in SCLC and in combination with venetoclax in AML. Not all clinically evaluated LSD1 inhibitors have shown promising signs of efficacy, however. Clinical trials evaluating GSK2879552 were terminated due to significant adverse events, encephalopathy in the SCLC trial, and limited efficacy in SCLC and AML129. Encephalopathy has not been reported with any other LSD1 inhibitors so it may represent a compound-specific, off-target effect.

Emerging Pre-clinical Epigenetic Targets

While this review provides detailed pharmacologic and clinical data supporting the use of epigenetic inhibitors being tested in the clinic, there are several epigenetic targets emerging as promising candidates in pre-clinical studies.

The Histone Ubiquitylase Complex PRC1

The Polycomb Repressive Complex 1 (PRC1) complex, which is responsible for the ubiquitylation of lysine 119 on histone 2A (H2AK119Ub), commonly co-localizes with the PRC2 complex to facilitate gene repression130. PRC1 contains one of two E3 ubiquitin ligase RING1 paralogues, RING1A or RING1B, and is subdivided into either canonical or non-canonical complexes depending on whether the PRC1 complex contains a chromobox (CBX) protein or one of two paralogs, RYBP or YAF2, respectively (Figure 3). RING1A/B interacts with one or more PCGF proteins to enhance its ubiquitin ligase activity131,132. Canonical PRC1 commonly complexes with PCGF2/4, while each of four non-canonical complexes PRC1.1, PRC1.3/PRC1.5, or PRC1.6, are defined by containing PCGF1, PCGF3/5, or PCGF6, respectively133.

Figure 3: New Epigenetic Targets in Cancer.

Figure 3:

Schematic showing select complexes (PRC1, ENL within the super elongation complex, and ASH1L) that are of interest for the development of new therapeutics.

PRC1 activity has been increasingly recognized as important in cancer132, although the contribution of individual PRC1 complexes is not well understood. Genomic alterations in PRC1 are relatively rare, except for alterations in BCOR and its paralog BCORL1, subunits of the PRC1.1 complex. Loss-of-function BCOR/BCORL1 mutations are observed in myeloid malignancies134, T-cell lymphomas135, and cancers of the central nervous system136. BCOR genomic rearrangements are particularly enriched in pediatric solid tumors. Most clear cell sarcomas of the kidney and primitive mesenchymal myxoid tumors of infancy harbor BCOR with internal tandem duplications136. BCOR-CCNB3, BCOR-MAML3, ZC3H7B-BCOR, BCOR-RARE, BCOR-CREBBP and EP300-BCOR fusions are also observed in various undifferentiated Ewing-like sarcomas, leukemias, and malignancies of the central nervous system136139. Notably these fusions are very rare and not well documented, so the function of BCOR in these disease states is unknown. Most fusions retain BCL6 and PCGF1 binding domains, suggesting that these domains are likely still important for their function; however careful dissection of these fusions is needed to further understand their mechanism.

Due the complexity of the PRC1 family, selective and potent inhibitors of canonical and variant PRC1 are needed to discern their individual contributions in cancer. Inhibitors of RING1 show response rates in leukemia models in vitro140. Currently, there is no strong clinical rationale for targeting RING1 in leukemia, although it has been suggested that RING1 regulates self-renewal potential of both normal and leukemia cells141. Therefore, it will be important to study whether the combined loss of RING1A and RING1B is tolerated in healthy tissues due to loss of compensatory paralog function. RING1A/B integrate into every PRC1 variant complex, and therefore RING1 inhibitors cannot differentiate functions of the different complexes. USP7 inhibitors, largely reported to be active in TP53 wildtype malignancies by destabilizing MDM2, have also been described as destabilizing the PRC1.1 complex through disrupting PCGF1/RING1 interactions142. Numerous inhibitors targeting CBX proteins, including CBX2143, CBX4/7144, CBX6145, and CBX8146,147 have also been identified. DepMap does not show strong single gene dependency on any of the CBX proteins, perhaps due to redundancies between CBX paralogs. Multiplexed CRISPR-Cas9 studies evaluating the combinatorial effect of double knockout of CBX proteins would of interest to evaluate the utility of non-selective CBX inhibitors in diverse cancer lineages.

Chromatin reader ENL

ENL/MLLT1, a histone lysine acetylation reader, promotes transcription through interactions with the super elongation complex and regulation of chromatin remodeling and gene expression via binding to modified histones (Figure 3). Two groups simultaneously identified a role for the ENL_YEATS domain in the control of leukemogenic gene expression in KMT2A-r AML148,149. Chromosomal translocations of KMT2A-ENL are an initiator of leukemogenesis and the fusion protein has been shown to be a viable therapeutic target150. Likewise, recurrent hotspot mutations in the YEATS domain of ENL have been described in Wilms tumor, a pediatric kidney cancer, and have been demonstrated to drive hyperactivation of transcription, supporting oncogenesis151,152. Since being identified as a potential therapeutic target in leukemia, multiple peptidomimetics and small molecule inhibitors of ENL, which displace ENL from chromatin by blocking its YEATS domain interaction with acetylated histones, have been developed153160. ENL degraders have also been developed155,161. Interestingly, both disruption of the interaction between the ENL YEATS domain and acetylated histones and depletion of the protein significantly suppress leukemia progression with minimal and reversable effects on normal hematopoiesis157,158. It is important to note that these drugs target the YEATS domain more generally, inhibiting both ENL and its paralog AF9, which is required for normal hematopoiesis162. Interestingly, AF9 is not a dependency in KMT2A-r leukemias and most KMT2A-AF9 proteins lack the AF9 YEATS domain148150. Thus, ENL inhibitors likely are not effective by selective inhibition of AF9 or the KMT2A-AF9 fusion, but rather wild-type ENL may be operating through a Menin-like mechanism whereby ENL inhibitors impair wild-type ENL function and subsequent transcriptional elongation. A therapeutic window might be enhanced if the ENL-containing oncogenic signaling complex that specifically regulates KMT2A-r-dependent genes is distinct from the endogenous ENL-containing complex, similar to recent observations for DOT1L163. Although preclinical data and probe molecules developed to date support the translation of ENL inhibitors for the treatment of KMT2A-r leukemia, DepMap data does not show as strong of a dependency as for Menin. Thus, these compounds may not translate to as strong of a clinical response. It will be interesting to test ENL inhibitors in combination with Menin inhibitors to evaluate whether they exhibit synergism.

Histone Methyltransferase ASH1L

Another emerging target is ASH1L, a histone methyltransferase that positively regulates gene expression via methylation of H3K36 and recruitment of KMT2A to chromatin (Figure 3). Based on these molecular functions, ASH1L was found to be required for the initiation and maintenance of KMT2A-r leukemia164,165. ASH1L also has been implicated in the pathogenesis of other AMLs with high HOX gene expression, and it is overexpressed in thyroid, breast, and liver cancers166168, making ASH1L an attractive target for drug discovery. The first-in-class small molecule inhibitor of ASH1L, AS-99, has been reported to induce cell death and differentiation, suppress KMT2A fusion driven gene expression, and reduce leukemia burden in vivo169. Like ENL, ASH1L does not show as strong of a dependency in DepMap for hematologic malignancies as compared to Menin. However, it does show a strong enrichment for some solid malignancies including ovarian and endometrial cancers, suggesting it might be translated in other diseases. It will be exciting to watch this class of drugs develop into clinical molecules for the treatment of cancer; however, caution should be taken as LOF mutations and haploinsufficiency of ASH1L are linked to many developmental disorders, including autism170.

Therapeutic considerations

Non-genetic Reprogramming and Plasticity

While genetic drivers are critical to both oncogenesis and drug resistance, non-genetic mechanisms are being increasingly recognized as drivers of tumor relapse171174. Much like genetic heterogeneity, cancers often are composed of heterogenous populations of cells that exist in unique transcriptional states. Although it is controversial whether cancer cell states that are associated with therapy resistance pre-exist or transition when exposed to therapy, it is established that tumors can quickly and reversibly alter their cell state by epigenetic and transcriptional reprogramming (Figure 4A)175,176. Tumor cells that persist in the presence of therapeutic pressure can arise from cells derived from different transcriptional states177,178. This intrinsic ability of cancer cells to present a different lineage state can promote metastasis and chemoresistance175,179. The ability of tumor cells to quickly transit between epigenetic states may be partially explained by the presence of bivalent promoters, that is the paradoxical presence of both active H3K4me3 and repressive H3K27me3 marks at promoters that are transcriptionally repressed but poised for transcriptional activation180. Treatment-refractory breast cancer cells have been shown to utilize bivalent promoters to withstand therapeutic stress, and bivalent promoters are enriched in relapsed cancers181183. Similarly, enhancer rewiring has been demonstrated in preclinical studies to render resistance to chromatin modifying compounds such as BET inhibitors184. While signaling cues that promote epigenetic plasticity are not well known, it is likely that tumor cell extrinsic and intrinsic factors can promote epigenetic reprogramming184. For example, extrinsic factors in the tumor microenvironment promote residual cell survival and clonal expansion185,186. It will be important to use advanced lineage tracing, single-cell sequencing platforms, and cell barcoding technologies to increase clarity as to how cells pass through cell states to adapt to cell intrinsic and extrinsic pressures.

Figure 4: Epigenetic/Lineage Plasticity as a mechanism of therapeutic resistance.

Figure 4:

A) Drug-tolerant tumor cells harbor the intrinsic ability to reversibly alter their gene expression programs to overcome drug treatments. B) Cancer cells can co-opt lineage-associated differentiation states to overcome therapeutic or environmental stressors. C) Inhibition of an epigenetic protein can select for the upregulated activity of an alternative epigenetic pathway.

Despite inter-tumoral variability within cancers arising from a specific tissue, cancers of the same lineage mostly cluster together in expression profiles, likely by retaining transcriptional features of their tissue of origin187,188. However, a feature of some cancers is the intrinsic ability to leverage developmentally linked cell states to promote tumor growth, metastasis, and evasion of therapeutic interventions (Figure 4B)189. It has long been observed that epithelial cancers can undergo an epithelial-to-mesenchymal transition, a state more resistant to cytotoxic, targeted, and immunotherapies190,191. A similar adrenergic-to-mesenchymal transition has been observed in neuroblastoma that can also mediate resistance to cytotoxic and immune-therapies192195. Lineage plasticity is also observed in the neuroendocrine trans-differentiation of a subset of prostate cancers196, Ewing sarcoma197, conversion of non-small cell lung cancers to small cell lung cancers198,199, and phenotype switching between “proliferative” and “invasive” phenotypes in melanoma200. Similarly, pediatric ALLs have been shown to “switch” to a myeloid lineage when treated with CD19 CAR T cells201. Some interventions have been shown to alter the differentiation trajectories of cancers into therapy-sensitive states, such as methotrexate in melanoma202, EZH2 inhibitors in prostate cancer194,203, and KDM5A/B inhibitors in lung and breast cancer172,204,205. Our ability to target drug-tolerant phenotypic states could be paramount to the eradication of residual tumor cells.

Activation of Alternate Epigenetic Pathways Following Treatment with Epigenetic Inhibitors

LOF mutations that occur naturally in epigenetic complexes can result in changes in the activity of alternate epigenetic pathways due to compensation or a release of regulation. It is likely that treatment with epigenetic inhibitors will produce similar changes in the activity of epigenetic pathways. For example, a recent study suggests that malignant rhabdoid tumors overcome EZH2 inhibition via loss of activity of NSD1, a H3K36me3 methyltransferase206. Knockout of the H3K36me3 demethylase, KDM2B, restored EZH2 sensitivity. In another study, it was found that Menin inhibition releases the KMT2A complex from active genes, inducing a re-localization of the KMT2A complex to bivalent chromatin (Figure 4C)207. The combination of EZH2 and Menin inhibitors reduced cell viability more than either inhibitor alone207. Thus, combinatorial epigenetic therapies that prevent changes in compensatory epigenetic pathways may be more efficacious in clinical trials than single agents. We must also be aware that epigenetic inhibition may in some contexts induce cell states that are more aggressive or that promote secondary malignancies208,209. Further, LOF in alternate pathways may also mediate resistance. For example, NCOR1 and HDAC3 knockout was found to confer resistance to P300/CBP inhibitors; thus, altered histone acetylation dynamics may affect the response to clinical P300/CBP inhibitors210. In sum, these data provide a strong rationale for a better understanding of how cancer cells rewire epigenetic pathways following epigenetic inhibition and suggests that combinatorial epigenetic inhibition may be necessary to achieve durable responses.

Utilizing Epigenetic Inhibitors as Therapy “Boosters”

While sustained therapeutic doses of some single agent epigenetic therapies may be difficult to achieve in humans, emerging evidence suggests that low-dose or short interval delivery of epigenetic therapies may be useful as an adjunctive therapy to enhance the activity of chemotherapy, radiotherapy, and immunotherapy. For example, tolerated doses of vorinostat in combination with the radiotherapeutic 131-metaiodobenzylguanidine (MIBG) in patients with neuroblastoma were associated with increased response rates as compared to either therapy alone211. The biology of this synergism remains unknown. It is possible that vorinostat prevents the downregulation of the transporter that is required for MIBG uptake, promotes further DNA damage, or alters the cell state to one that is more amenable to radiation-related killing. EZH2 inhibitors have also been found to sensitize lung cancer to standard chemotherapy212 or promote increased expression of tumor cell antigens such as GD2194,213,214 or MHC expression207,215.

Epigenetics and phase transition

Emerging data suggest that the physicochemical properties generated by organization of proteins into membrane-less compartments also affect the structure and function of chromatin216218. Chromatin modifiers, transcription factors219, and histones220 are highly enriched for intrinsically disordered regions that can promote liquid-liquid phase separation. We are just beginning to understand how condensate biology affects partitioning of small molecules, such as epigenetic inhibitors, into specific epigenetic compartments and how these biophysical properties regulate epigenetic mechanisms221,222. For example, the BET inhibitor JQ1 preferentially concentrates in BRD4 condensates, suggesting localization within the cell may be important for pharmacodynamic response222. Further, fusion oncoproteins can harbor intrinsically disordered regions (IDRs) and are prone to generate phase-separated compartments that can modulate transcriptional activity and promote leukemogenesis223. A recent study experimentally validated that 58% of fusion oncoproteins, particularly those localized to the nucleus, can form condensates224. Using machine learning, the authors predict that as many as 67% of fusion oncoproteins may be able to form condensates. It has been shown, for example, that NUP98 fusions form condensates that drive leukemogenic transcriptional programs223. Further, driving concentrations of fusion oncoproteins through loss of regulators also disrupted oncogenic signaling activity by altering condensate properties225,226. This suggests a “Goldilocks” principle by which fusion oncoprotein levels must be fine-tuned and either low or high expression disrupts condensate formation and oncogenic activity. Thus, potential avenues of targeting “undruggable” fusion oncoproteins may be through targeting IDRs to modulate condensate properties.

Summary and Future Directions

New insights into the biology of cancer epigenetics have created a strong foundation for the development of small molecule inhibitors targeting at least thirteen unique classes of epigenetic targets (Figure 1). Excitingly, advances in medicinal chemistry have expanded the toolkit to target previously non-targetable proteins. The slate of new small molecule inhibitors includes molecular protein degraders (e.g., BRD9), allosteric inhibitors of catalytic function (e.g., EED, SMARCA2/4) and disruptors of protein-protein interactions (e.g., Menin-KMT2A). Selective degradation of protein targets via PROTACs may be particularly useful, as such degradation may enable distinction between catalytic-dependent versus scaffolding functions of epigenetic complexes. However, caution is warranted in interpreting the phenotype of degraders in the absence of orthogonal models (e.g., CRISPR knockout). A pre-print suggests that degradation of BRD4 also forms neo-amino-terminal peptides that neutralize Inhibitor of Apoptosis proteins, subsequently inducing cell death227. This may explain why cell death was observed with P300/CBP degraders and not the HAT or bromodomain CBP/P300 inhibitors100. The discovery of natural glue-like degraders, such as lenalidomide and related derivatives, that degrade transcription factors such as IKZF1 and IKZF3, offers the promise of degrading other transcription factors, including transcription factor fusions. Beyond targeting tumor-intrinsic epigenetic regulation described in this review, epigenetic inhibitors may also hold promise in promoting response to immunotherapy such as by preventing T cell exhaustion, modulating immune cell populations in the tumor microenvironment, or promoting antigen presentation228230.

As described, the development of new classes of compounds, such as ENL, PRC1 (RING1), and ASH1L inhibitors shows promise in AML and could be applied to other disease states. Additionally, a first-in-class NSD2 inhibitor, KTX-1001, will soon go into clinical trial for testing in t(4;14) and NSD2 gain-of-function (E1099K) mutant multiple myeloma (Figure 1 and Table 1). While we used a narrow perspective to define drugs that target epigenetic processes in this review to only those inhibitors that perturb the function of proteins that regulate chromatin/transcription factor function, there are other epigenetic-adjacent inhibitors also currently in clinical trials. These include those targeting the m6A methyltransferase METTL3, the kinases CDK7 and CDK9, PRMT5, and IDH1/2.

Despite much promise for epigenetic therapies in cancer, response rates in humans remain poor for solid tumors. In these patients, this may be due to dose-limiting toxicities that prevent therapeutic dosing (e.g., HDAC inhibitors) or inadequate drug concentrations within the tumor. Thus, optimization of pharmacokinetics and limiting off-tumor toxicity will be paramount to establish durable and tolerated therapy responses in patients. Therapeutic boosting with low-dose epigenetic inhibitors is another implementation strategy for epigenetic inhibitors in the clinic. Further, in vitro studies with solid tumor models suggest that epigenetic heterogeneity and adaptation may contribute to rapid drug resistance in the absence of clear genetic drivers of disease, providing another barrier to effective treatment of solid tumors. The implementation of single-cell spatial transcriptomics and epigenomics will hopefully shed light on whether cellular interactions or location can influence the response to epigenetic inhibitors in solid tumors. As we accumulate more information on encoded epigenetic, genetic, and drug sensitivity properties of cancers, we can integrate these data to design rationally guided strategies to treat and provide curative options for patients.

Acknowledgements

This work was funded by the National Institutes of Health (P01 CA217959 (K.S.); R35 CA283977 (K.S); CA261035 (N.W.M); CA279915 (N.W.M); CA243266 (C.F.M)), the Leukemia and Lymphoma Society, the Rally Foundation, and a Pediatric Stand Up to Cancer Catalyst Grant supported by Bristol-Myers Squibb (SU2C#6143). K.S. is also funded by the St. Jude Children’s Research Hospital Collaborative Research Program.

Disclosures

K.S. receives grant funding from the DFCI/Novartis Drug Discovery Program and from KronosBio, is a member of the SAB and has stock options with Auron Therapeutics and has consulted for AstraZeneca. All other authors have no disclosures.

References

  • 1.Allis CD & Jenuwein T The molecular hallmarks of epigenetic control. Nat Rev Genet 17, 487–500 (2016). 10.1038/nrg.2016.59 [DOI] [PubMed] [Google Scholar]
  • 2.Mashtalir N et al. A Structural Model of the Endogenous Human BAF Complex Informs Disease Mechanisms. Cell 183, 802–817 e824 (2020). 10.1016/j.cell.2020.09.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hanahan D Hallmarks of Cancer: New Dimensions. Cancer Discov 12, 31–46 (2022). 10.1158/2159-8290.CD-21-1059 [DOI] [PubMed] [Google Scholar]
  • 4.Lai WKM & Pugh BF Understanding nucleosome dynamics and their links to gene expression and DNA replication. Nat Rev Mol Cell Biol 18, 548–562 (2017). 10.1038/nrm.2017.47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mittal P & Roberts CWM The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat Rev Clin Oncol 17, 435–448 (2020). 10.1038/s41571-020-0357-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cramer P Organization and regulation of gene transcription. Nature 573, 45–54 (2019). 10.1038/s41586-019-1517-4 [DOI] [PubMed] [Google Scholar]
  • 7.Chi P, Allis CD & Wang GG Covalent histone modifications--miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer 10, 457–469 (2010). 10.1038/nrc2876 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Conery AR, Rocnik JL & Trojer P Small molecule targeting of chromatin writers in cancer. Nat Chem Biol 18, 124–133 (2022). 10.1038/s41589-021-00920-5 [DOI] [PubMed] [Google Scholar]
  • 9.Tsherniak A et al. Defining a Cancer Dependency Map. Cell 170, 564–576 e516 (2017). 10.1016/j.cell.2017.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dharia NV et al. A first-generation pediatric cancer dependency map. Nat Genet 53, 529–538 (2021). 10.1038/s41588-021-00819-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lyko F The DNA methyltransferase family: a versatile toolkit for epigenetic regulation. Nat Rev Genet 19, 81–92 (2018). 10.1038/nrg.2017.80 [DOI] [PubMed] [Google Scholar]
  • 12.Baylin SB & Jones PA Epigenetic Determinants of Cancer. Cold Spring Harb Perspect Biol 8 (2016). 10.1101/cshperspect.a019505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Whittaker SJ et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 28, 4485–4491 (2010). 10.1200/JCO.2010.28.9066 [DOI] [PubMed] [Google Scholar]
  • 14.O’Connor OA et al. Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results of the Pivotal Phase II BELIEF (CLN-19) Study. J Clin Oncol 33, 2492–2499 (2015). 10.1200/JCO.2014.59.2782 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.San-Miguel JF et al. Overall survival of patients with relapsed multiple myeloma treated with panobinostat or placebo plus bortezomib and dexamethasone (the PANORAMA 1 trial): a randomised, placebo-controlled, phase 3 trial. Lancet Haematol 3, e506–e515 (2016). 10.1016/S2352-3026(16)30147-8 [DOI] [PubMed] [Google Scholar]
  • 16.Eckschlager T, Plch J, Stiborova M & Hrabeta J Histone Deacetylase Inhibitors as Anticancer Drugs. Int J Mol Sci 18 (2017). 10.3390/ijms18071414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lechner S et al. Target deconvolution of HDAC pharmacopoeia reveals MBLAC2 as common off-target. Nat Chem Biol 18, 812–820 (2022). 10.1038/s41589-022-01015-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Najm FJ et al. Chromatin complex dependencies reveal targeting opportunities in leukemia. Nat Commun 14, 448 (2023). 10.1038/s41467-023-36150-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zhang Y et al. Collateral lethality between HDAC1 and HDAC2 exploits cancer-specific NuRD complex vulnerabilities. Nat Struct Mol Biol 30, 1160–1171 (2023). 10.1038/s41594-023-01041-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chang L, Ruiz P, Ito T & Sellers WR Targeting pan-essential genes in cancer: Challenges and opportunities. Cancer Cell 39, 466–479 (2021). 10.1016/j.ccell.2020.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yang Z et al. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol Cell 19, 535–545 (2005). 10.1016/j.molcel.2005.06.029 [DOI] [PubMed] [Google Scholar]
  • 22.Delmore JE et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011). 10.1016/j.cell.2011.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Puissant A et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov 3, 308–323 (2013). 10.1158/2159-8290.CD-12-0418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hensel T et al. Targeting the EWS-ETS transcriptional program by BET bromodomain inhibition in Ewing sarcoma. Oncotarget 7, 1451–1463 (2016). 10.18632/oncotarget.6385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Filippakopoulos P et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010). 10.1038/nature09504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dawson MA et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533 (2011). 10.1038/nature10509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Doroshow DB, Eder JP & LoRusso PM BET inhibitors: a novel epigenetic approach. Ann Oncol 28, 1776–1787 (2017). 10.1093/annonc/mdx157 [DOI] [PubMed] [Google Scholar]
  • 28.Piha-Paul SA et al. Phase 1 Study of Molibresib (GSK525762), a Bromodomain and Extra-Terminal Domain Protein Inhibitor, in NUT Carcinoma and Other Solid Tumors. JNCI Cancer Spectr 4, pkz093 (2020). 10.1093/jncics/pkz093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Stathis A et al. Clinical Response of Carcinomas Harboring the BRD4-NUT Oncoprotein to the Targeted Bromodomain Inhibitor OTX015/MK-8628. Cancer Discov 6, 492–500 (2016). 10.1158/2159-8290.CD-15-1335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shorstova T, Foulkes WD & Witcher M Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 124, 1478–1490 (2021). 10.1038/s41416-021-01321-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sun Y et al. Safety and Efficacy of Bromodomain and Extra-Terminal Inhibitors for the Treatment of Hematological Malignancies and Solid Tumors: A Systematic Study of Clinical Trials. Front Pharmacol 11, 621093 (2020). 10.3389/fphar.2020.621093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Garraway LA & Lander ES Lessons from the cancer genome. Cell 153, 17–37 (2013). 10.1016/j.cell.2013.03.002 [DOI] [PubMed] [Google Scholar]
  • 33.Morschhauser F et al. Tazemetostat for patients with relapsed or refractory follicular lymphoma: an open-label, single-arm, multicentre, phase 2 trial. Lancet Oncol 21, 1433–1442 (2020). 10.1016/S1470-2045(20)30441-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ogiwara H et al. Targeting p300 Addiction in CBP-Deficient Cancers Causes Synthetic Lethality by Apoptotic Cell Death due to Abrogation of MYC Expression. Cancer Discov 6, 430–445 (2016). 10.1158/2159-8290.CD-15-0754 [DOI] [PubMed] [Google Scholar]
  • 35.Scheller M et al. Hotspot DNMT3A mutations in clonal hematopoiesis and acute myeloid leukemia sensitize cells to azacytidine via viral mimicry response. Nat Cancer 2, 527–544 (2021). 10.1038/s43018-021-00213-9 [DOI] [PubMed] [Google Scholar]
  • 36.Bejar R et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood 124, 2705–2712 (2014). 10.1182/blood-2014-06-582809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gounder M et al. Tazemetostat in advanced epithelioid sarcoma with loss of INI1/SMARCB1: an international, open-label, phase 2 basket study. Lancet Oncol 21, 1423–1432 (2020). 10.1016/S1470-2045(20)30451-4 [DOI] [PubMed] [Google Scholar]
  • 38.Czermin B et al. Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell 111, 185–196 (2002). 10.1016/s0092-8674(02)00975-3 [DOI] [PubMed] [Google Scholar]
  • 39.Muller J et al. Histone methyltransferase activity of a Drosophila Polycomb group repressor complex. Cell 111, 197–208 (2002). 10.1016/s0092-8674(02)00976-5 [DOI] [PubMed] [Google Scholar]
  • 40.Hodis E et al. A landscape of driver mutations in melanoma. Cell 150, 251–263 (2012). 10.1016/j.cell.2012.06.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.McKinney M et al. The Genetic Basis of Hepatosplenic T-cell Lymphoma. Cancer Discov 7, 369–379 (2017). 10.1158/2159-8290.CD-16-0330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Morin RD et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298–303 (2011). 10.1038/nature10351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Pikman Y et al. Targeting EZH2 for the treatment of hepatosplenic T-cell lymphoma. Blood Adv 4, 1265–1269 (2020). 10.1182/bloodadvances.2019001256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tirode F et al. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov 4, 1342–1353 (2014). 10.1158/2159-8290.CD-14-0622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Chen L et al. CRISPR-Cas9 screen reveals a MYCN-amplified neuroblastoma dependency on EZH2. J Clin Invest 128, 446–462 (2018). 10.1172/JCI90793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Duan R, Du W & Guo W EZH2: a novel target for cancer treatment. J Hematol Oncol 13, 104 (2020). 10.1186/s13045-020-00937-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Qi W et al. Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation. Proc Natl Acad Sci U S A 109, 21360–21365 (2012). 10.1073/pnas.1210371110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Richter GH et al. EZH2 is a mediator of EWS/FLI1 driven tumor growth and metastasis blocking endothelial and neuro-ectodermal differentiation. Proc Natl Acad Sci U S A 106, 5324–5329 (2009). 10.1073/pnas.0810759106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wilson BG et al. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18, 316–328 (2010). 10.1016/j.ccr.2010.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kadoch C et al. Dynamics of BAF-Polycomb complex opposition on heterochromatin in normal and oncogenic states. Nat Genet 49, 213–222 (2017). 10.1038/ng.3734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bitler BG et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med 21, 231–238 (2015). 10.1038/nm.3799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Knutson SK et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A 110, 7922–7927 (2013). 10.1073/pnas.1303800110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ntziachristos P et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nat Med 18, 298–301 (2012). 10.1038/nm.2651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang J et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 481, 157–163 (2012). 10.1038/nature10725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ernst T et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 42, 722–726 (2010). 10.1038/ng.621 [DOI] [PubMed] [Google Scholar]
  • 56.Nikoloski G et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet 42, 665–667 (2010). 10.1038/ng.620 [DOI] [PubMed] [Google Scholar]
  • 57.Lee W et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat Genet 46, 1227–1232 (2014). 10.1038/ng.3095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lewis PW et al. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340, 857–861 (2013). 10.1126/science.1232245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chi S et al. Abstract A175: Phase 1 study of the EZH2 inhibitor, tazemetostat, in children with relapsed or refractory INI1-negative tumors including rhabdoid tumors, epithelioid sarcoma, chordoma, and synovial sarcoma. Molecular Cancer Therapeutics 17, A175–A175 (2018). 10.1158/1535-7163.Targ-17-a175 [DOI] [Google Scholar]
  • 60.Honma D et al. Novel orally bioavailable EZH1/2 dual inhibitors with greater antitumor efficacy than an EZH2 selective inhibitor. Cancer Sci 108, 2069–2078 (2017). 10.1111/cas.13326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Phase I/II Study of MAK683 in Patients with Advanced Malignancies, Including Diffuse Large B-Cell Lymphoma.
  • 62.Centore RC, Sandoval GJ, Soares LMM, Kadoch C & Chan HM Mammalian SWI/SNF Chromatin Remodeling Complexes: Emerging Mechanisms and Therapeutic Strategies. Trends Genet 36, 936–950 (2020). 10.1016/j.tig.2020.07.011 [DOI] [PubMed] [Google Scholar]
  • 63.Ehrenhofer-Wolfer K et al. SMARCA2-deficiency confers sensitivity to targeted inhibition of SMARCA4 in esophageal squamous cell carcinoma cell lines. Sci Rep 9, 11661 (2019). 10.1038/s41598-019-48152-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hoffman GR et al. Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers. Proc Natl Acad Sci U S A 111, 3128–3133 (2014). 10.1073/pnas.1316793111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Schick S et al. Systematic characterization of BAF mutations provides insights into intracomplex synthetic lethalities in human cancers. Nat Genet 51, 1399–1410 (2019). 10.1038/s41588-019-0477-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kofink C et al. A selective and orally bioavailable VHL-recruiting PROTAC achieves SMARCA2 degradation in vivo. Nat Commun 13, 5969 (2022). 10.1038/s41467-022-33430-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cantley J et al. Selective PROTAC-mediated degradation of SMARCA2 is efficacious in SMARCA4 mutant cancers. Nat Commun 13, 6814 (2022). 10.1038/s41467-022-34562-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hodges C, Kirkland JG & Crabtree GR The Many Roles of BAF (mSWI/SNF) and PBAF Complexes in Cancer. Cold Spring Harb Perspect Med 6 (2016). 10.1101/cshperspect.a026930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Michel BC et al. A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation. Nat Cell Biol 20, 1410–1420 (2018). 10.1038/s41556-018-0221-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang X et al. SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation. Nat Genet 49, 289–295 (2017). 10.1038/ng.3746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li J et al. A Role for SMARCB1 in Synovial Sarcomagenesis Reveals That SS18-SSX Induces Canonical BAF Destruction. Cancer Discov 11, 2620–2637 (2021). 10.1158/2159-8290.CD-20-1219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Brien GL et al. Targeted degradation of BRD9 reverses oncogenic gene expression in synovial sarcoma. Elife 7 (2018). 10.7554/eLife.41305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jackson KL et al. Abstract ND09: The discovery and characterization of CFT8634: A potent and selective degrader of BRD9 for the treatment of SMARCB1-perturbed cancers. Cancer Research 82, ND09–ND09 (2022). 10.1158/1538-7445.Am2022-nd09 [DOI] [Google Scholar]
  • 74.Milne TA et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol Cell 10, 1107–1117 (2002). 10.1016/s1097-2765(02)00741-4 [DOI] [PubMed] [Google Scholar]
  • 75.Mohan M, Lin C, Guest E & Shilatifard A Licensed to elongate: a molecular mechanism for MLL-based leukaemogenesis. Nat Rev Cancer 10, 721–728 (2010). 10.1038/nrc2915 [DOI] [PubMed] [Google Scholar]
  • 76.Grembecka J et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat Chem Biol 8, 277–284 (2012). 10.1038/nchembio.773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Daigle SR et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011). 10.1016/j.ccr.2011.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Heikamp EB et al. The menin-MLL1 interaction is a molecular dependency in NUP98-rearranged AML. Blood 139, 894–906 (2022). 10.1182/blood.2021012806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Uckelmann HJ et al. Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science 367, 586–590 (2020). 10.1126/science.aax5863 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Schubeler D et al. The histone modification pattern of active genes revealed through genome-wide chromatin analysis of a higher eukaryote. Genes Dev 18, 1263–1271 (2004). 10.1101/gad.1198204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Mohan M et al. Linking H3K79 trimethylation to Wnt signaling through a novel Dot1-containing complex (DotCom). Genes Dev 24, 574–589 (2010). 10.1101/gad.1898410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Min J, Feng Q, Li Z, Zhang Y & Xu RM Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase. Cell 112, 711–723 (2003). 10.1016/s0092-8674(03)00114-4 [DOI] [PubMed] [Google Scholar]
  • 83.Sun Y et al. HOXA9 Reprograms the Enhancer Landscape to Promote Leukemogenesis. Cancer Cell 34, 643–658 e645 (2018). 10.1016/j.ccell.2018.08.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Daigle SR et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122, 1017–1025 (2013). 10.1182/blood-2013-04-497644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Stein EM et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 131, 2661–2669 (2018). 10.1182/blood-2017-12-818948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lin J et al. Menin “reads” H3K79me2 mark in a nucleosomal context. Science 379, 717–723 (2023). 10.1126/science.adc9318 [DOI] [PubMed] [Google Scholar]
  • 87.Yokoyama A et al. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 123, 207–218 (2005). 10.1016/j.cell.2005.09.025 [DOI] [PubMed] [Google Scholar]
  • 88.Krivtsov AV et al. A Menin-MLL Inhibitor Induces Specific Chromatin Changes and Eradicates Disease in Models of MLL-Rearranged Leukemia. Cancer Cell 36, 660–673 e611 (2019). 10.1016/j.ccell.2019.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Shi A et al. Structural insights into inhibition of the bivalent menin-MLL interaction by small molecules in leukemia. Blood 120, 4461–4469 (2012). 10.1182/blood-2012-05-429274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Issa GC et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature (2023). 10.1038/s41586-023-05812-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Erba HP et al. Update on a Phase 1/2 First-in-Human Study of the Menin-KMT2A (MLL) Inhibitor Ziftomenib (KO-539) in Patients with Relapsed or Refractory Acute Myeloid Leukemia. Blood 140, 153–156 (2022). 10.1182/blood-2022-167412 [DOI] [Google Scholar]
  • 92.Malik R et al. Targeting the MLL complex in castration-resistant prostate cancer. Nat Med 21, 344–352 (2015). 10.1038/nm.3830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Svoboda LK et al. Tumorigenicity of Ewing sarcoma is critically dependent on the trithorax proteins MLL1 and menin. Oncotarget 8, 458–471 (2017). 10.18632/oncotarget.13444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hemming ML et al. MOZ and Menin-MLL Complexes Are Complementary Regulators of Chromatin Association and Transcriptional Output in Gastrointestinal Stromal Tumor. Cancer Discov 12, 1804–1823 (2022). 10.1158/2159-8290.CD-21-0646 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Perner F et al. MEN1 mutations mediate clinical resistance to menin inhibition. Nature 615, 913–919 (2023). 10.1038/s41586-023-05755-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lee KK & Workman JL Histone acetyltransferase complexes: one size doesn’t fit all. Nat Rev Mol Cell Biol 8, 284–295 (2007). 10.1038/nrm2145 [DOI] [PubMed] [Google Scholar]
  • 97.Yang XJ & Seto E Lysine acetylation: codified crosstalk with other posttranslational modifications. Mol Cell 31, 449–461 (2008). 10.1016/j.molcel.2008.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Welti J et al. Targeting the p300/CBP Axis in Lethal Prostate Cancer. Cancer Discov 11, 1118–1137 (2021). 10.1158/2159-8290.CD-20-0751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Nie M et al. Genome-wide CRISPR screens reveal synthetic lethal interaction between CREBBP and EP300 in diffuse large B-cell lymphoma. Cell Death Dis 12, 419 (2021). 10.1038/s41419-021-03695-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Durbin AD et al. EP300 Selectively Controls the Enhancer Landscape of MYCN-Amplified Neuroblastoma. Cancer Discov 12, 730–751 (2022). 10.1158/2159-8290.CD-21-0385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Hay DA et al. Discovery and optimization of small-molecule ligands for the CBP/p300 bromodomains. J Am Chem Soc 136, 9308–9319 (2014). 10.1021/ja412434f [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lasko LM et al. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 550, 128–132 (2017). 10.1038/nature24028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Morrison-Smith CD et al. Combined Targeting of the BRD4-NUT-p300 Axis in NUT Midline Carcinoma by Dual Selective Bromodomain Inhibitor, NEO2734. Mol Cancer Ther 19, 1406–1414 (2020). 10.1158/1535-7163.MCT-20-0087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Thomas JE 2nd, et al. Discovery of Exceptionally Potent, Selective, and Efficacious PROTAC Degraders of CBP and p300 Proteins. J Med Chem 66, 8178–8199 (2023). 10.1021/acs.jmedchem.3c00492 [DOI] [PubMed] [Google Scholar]
  • 105.Vannam R et al. Targeted degradation of the enhancer lysine acetyltransferases CBP and p300. Cell Chem Biol 28, 503–514 e512 (2021). 10.1016/j.chembiol.2020.12.004 [DOI] [PubMed] [Google Scholar]
  • 106.Borrow J et al. The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein. Nat Genet 14, 33–41 (1996). 10.1038/ng0996-33 [DOI] [PubMed] [Google Scholar]
  • 107.Yan F et al. KAT6A and ENL Form an Epigenetic Transcriptional Control Module to Drive Critical Leukemogenic Gene-Expression Programs. Cancer Discov 12, 792–811 (2022). 10.1158/2159-8290.CD-20-1459 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Northcott PA et al. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet 41, 465–472 (2009). 10.1038/ng.336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zack TI et al. Pan-cancer patterns of somatic copy number alteration. Nat Genet 45, 1134–1140 (2013). 10.1038/ng.2760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Turner-Ivey B et al. KAT6A, a chromatin modifier from the 8p11-p12 amplicon is a candidate oncogene in luminal breast cancer. Neoplasia 16, 644–655 (2014). 10.1016/j.neo.2014.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Saglam O, Tang Z, Tang G, Medeiros LJ & Toruner GA KAT6A amplifications are associated with shorter progression-free survival and overall survival in patients with endometrial serous carcinoma. PLoS One 15, e0238477 (2020). 10.1371/journal.pone.0238477 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Baell JB et al. Inhibitors of histone acetyltransferases KAT6A/B induce senescence and arrest tumour growth. Nature 560, 253–257 (2018). 10.1038/s41586-018-0387-5 [DOI] [PubMed] [Google Scholar]
  • 113.Lv D et al. Histone Acetyltransferase KAT6A Upregulates PI3K/AKT Signaling through TRIM24 Binding. Cancer Res 77, 6190–6201 (2017). 10.1158/0008-5472.CAN-17-1388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sharma S et al. Abstract 1130: First-in-class KAT6A/KAT6B inhibitor CTx-648 (PF-9363) demonstrates potent anti-tumor activity in ER+ breast cancer with KAT6A dysregulation. Cancer Research 81, 1130–1130 (2021). 10.1158/1538-7445.Am2021-1130 [DOI] [Google Scholar]
  • 115.Grant PA et al. Expanded lysine acetylation specificity of Gcn5 in native complexes. J Biol Chem 274, 5895–5900 (1999). 10.1074/jbc.274.9.5895 [DOI] [PubMed] [Google Scholar]
  • 116.Jin Q et al. Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/p300-mediated H3K18/27ac in nuclear receptor transactivation. EMBO J 30, 249–262 (2011). 10.1038/emboj.2010.318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Farria AT et al. Transcriptional Activation of MYC-Induced Genes by GCN5 Promotes B-cell Lymphomagenesis. Cancer Res 80, 5543–5553 (2020). 10.1158/0008-5472.CAN-20-2379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Mustachio LM, Roszik J, Farria A & Dent SYR Targeting the SAGA and ATAC Transcriptional Coactivator Complexes in MYC-Driven Cancers. Cancer Res 80, 1905–1911 (2020). 10.1158/0008-5472.CAN-19-3652 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bassi ZI et al. Modulating PCAF/GCN5 Immune Cell Function through a PROTAC Approach. ACS Chem Biol 13, 2862–2867 (2018). 10.1021/acschembio.8b00705 [DOI] [PubMed] [Google Scholar]
  • 120.Fang Y, Liao G & Yu B LSD1/KDM1A inhibitors in clinical trials: advances and prospects. J Hematol Oncol 12, 129 (2019). 10.1186/s13045-019-0811-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wang Y et al. LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell 138, 660–672 (2009). 10.1016/j.cell.2009.05.050 [DOI] [PubMed] [Google Scholar]
  • 122.Kim SA, Zhu J, Yennawar N, Eek P & Tan S Crystal Structure of the LSD1/CoREST Histone Demethylase Bound to Its Nucleosome Substrate. Mol Cell 78, 903–914 e904 (2020). 10.1016/j.molcel.2020.04.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Metzger E et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 437, 436–439 (2005). 10.1038/nature04020 [DOI] [PubMed] [Google Scholar]
  • 124.Maes T et al. ORY-1001, a Potent and Selective Covalent KDM1A Inhibitor, for the Treatment of Acute Leukemia. Cancer Cell 33, 495–511 e412 (2018). 10.1016/j.ccell.2018.02.002 [DOI] [PubMed] [Google Scholar]
  • 125.Augert A et al. Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci Signal 12 (2019). 10.1126/scisignal.aau2922 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Mohammad HP et al. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell 28, 57–69 (2015). 10.1016/j.ccell.2015.06.002 [DOI] [PubMed] [Google Scholar]
  • 127.Sehrawat A et al. LSD1 activates a lethal prostate cancer gene network independently of its demethylase function. Proc Natl Acad Sci U S A 115, E4179–E4188 (2018). 10.1073/pnas.1719168115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Salamero O et al. First-in-Human Phase I Study of Iadademstat (ORY-1001): A First-in-Class Lysine-Specific Histone Demethylase 1A Inhibitor, in Relapsed or Refractory Acute Myeloid Leukemia. J Clin Oncol 38, 4260–4273 (2020). 10.1200/JCO.19.03250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bauer TM et al. Phase I, Open-Label, Dose-Escalation Study of the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy of GSK2879552 in Relapsed/Refractory SCLC. J Thorac Oncol 14, 1828–1838 (2019). 10.1016/j.jtho.2019.06.021 [DOI] [PubMed] [Google Scholar]
  • 130.McGinty RK, Henrici RC & Tan S Crystal structure of the PRC1 ubiquitylation module bound to the nucleosome. Nature 514, 591–596 (2014). 10.1038/nature13890 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Fursova NA et al. Synergy between Variant PRC1 Complexes Defines Polycomb-Mediated Gene Repression. Mol Cell 74, 1020–1036 e1028 (2019). 10.1016/j.molcel.2019.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Parreno V, Martinez AM & Cavalli G Mechanisms of Polycomb group protein function in cancer. Cell Res 32, 231–253 (2022). 10.1038/s41422-021-00606-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Scelfo A et al. Functional Landscape of PCGF Proteins Reveals Both RING1A/B-Dependent-and RING1A/B-Independent-Specific Activities. Mol Cell 74, 1037–1052 e1037 (2019). 10.1016/j.molcel.2019.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Schaefer EJ et al. BCOR and BCORL1 Mutations Drive Epigenetic Reprogramming and Oncogenic Signaling by Unlinking PRC1.1 from Target Genes. Blood Cancer Discov 3, 116–135 (2022). 10.1158/2643-3230.BCD-21-0115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kang JH et al. The mutation of BCOR is highly recurrent and oncogenic in mature T-cell lymphoma. BMC Cancer 21, 82 (2021). 10.1186/s12885-021-07806-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Astolfi A et al. BCOR involvement in cancer. Epigenomics 11, 835–855 (2019). 10.2217/epi-2018-0195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Pisapia DJ et al. Fusions involving BCOR and CREBBP are rare events in infiltrating glioma. Acta Neuropathol Commun 8, 80 (2020). 10.1186/s40478-020-00951-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Yamamoto Y et al. BCOR as a novel fusion partner of retinoic acid receptor alpha in a t(X;17)(p11;q12) variant of acute promyelocytic leukemia. Blood 116, 4274–4283 (2010). 10.1182/blood-2010-01-264432 [DOI] [PubMed] [Google Scholar]
  • 139.Tauziede-Espariat A et al. The EP300:BCOR fusion extends the genetic alteration spectrum defining the new tumoral entity of “CNS tumors with BCOR internal tandem duplication”. Acta Neuropathol Commun 8, 178 (2020). 10.1186/s40478-020-01064-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Shukla S et al. Small-molecule inhibitors targeting Polycomb repressive complex 1 RING domain. Nat Chem Biol 17, 784–793 (2021). 10.1038/s41589-021-00815-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Rizo A et al. Repression of BMI1 in normal and leukemic human CD34(+) cells impairs self-renewal and induces apoptosis. Blood 114, 1498–1505 (2009). 10.1182/blood-2009-03-209734 [DOI] [PubMed] [Google Scholar]
  • 142.Maat H et al. The USP7-TRIM27 axis mediates non-canonical PRC1.1 function and is a druggable target in leukemia. iScience 24, 102435 (2021). 10.1016/j.isci.2021.102435 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wang S et al. A Potent, Selective CBX2 Chromodomain Ligand and Its Cellular Activity During Prostate Cancer Neuroendocrine Differentiation. Chembiochem 22, 2335–2344 (2021). 10.1002/cbic.202100118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Stuckey JI et al. A cellular chemical probe targeting the chromodomains of Polycomb repressive complex 1. Nat Chem Biol 12, 180–187 (2016). 10.1038/nchembio.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Milosevich N et al. Selective Inhibition of CBX6: A Methyllysine Reader Protein in the Polycomb Family. ACS Med Chem Lett 7, 139–144 (2016). 10.1021/acsmedchemlett.5b00378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Milosevich N et al. Polycomb Paralog Chromodomain Inhibitors Active against Both CBX6 and CBX8*. ChemMedChem 16, 3027–3034 (2021). 10.1002/cmdc.202100262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wang S et al. Optimization of Ligands Using Focused DNA-Encoded Libraries To Develop a Selective, Cell-Permeable CBX8 Chromodomain Inhibitor. ACS Chem Biol 15, 112–131 (2020). 10.1021/acschembio.9b00654 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Erb MA et al. Transcription control by the ENL YEATS domain in acute leukaemia. Nature 543, 270–274 (2017). 10.1038/nature21688 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wan L et al. ENL links histone acetylation to oncogenic gene expression in acute myeloid leukaemia. Nature 543, 265–269 (2017). 10.1038/nature21687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Hu H et al. The ENL YEATS epigenetic reader domain critically links MLL-ENL to leukemic stem cell frequency in t(11;19) Leukemia. Leukemia 37, 190–201 (2023). 10.1038/s41375-022-01765-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Perlman EJ et al. MLLT1 YEATS domain mutations in clinically distinctive Favourable Histology Wilms tumours. Nat Commun 6, 10013 (2015). 10.1038/ncomms10013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Wan L et al. Impaired cell fate through gain-of-function mutations in a chromatin reader. Nature 577, 121–126 (2020). 10.1038/s41586-019-1842-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Asiaban JN et al. Cell-Based Ligand Discovery for the ENL YEATS Domain. ACS Chem Biol 15, 895–903 (2020). 10.1021/acschembio.0c00124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Christott T et al. Discovery of a Selective Inhibitor for the YEATS Domains of ENL/AF9. SLAS Discov 24, 133–141 (2019). 10.1177/2472555218809904 [DOI] [PubMed] [Google Scholar]
  • 155.Garnar-Wortzel L et al. Chemical Inhibition of ENL/AF9 YEATS Domains in Acute Leukemia. ACS Cent Sci 7, 815–830 (2021). 10.1021/acscentsci.0c01550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Jiang Y et al. Selective Targeting of AF9 YEATS Domain by Cyclopeptide Inhibitors with Preorganized Conformation. J Am Chem Soc 142, 21450–21459 (2020). 10.1021/jacs.0c10324 [DOI] [PubMed] [Google Scholar]
  • 157.Li X et al. Structure-guided development of YEATS domain inhibitors by targeting pi-pi-pi stacking. Nat Chem Biol 14, 1140–1149 (2018). 10.1038/s41589-018-0144-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Liu Y et al. Small-Molecule Inhibition of the Acyl-Lysine Reader ENL as a Strategy against Acute Myeloid Leukemia. Cancer Discov 12, 2684–2709 (2022). 10.1158/2159-8290.CD-21-1307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Ma XR et al. Discovery of Selective Small-Molecule Inhibitors for the ENL YEATS Domain. J Med Chem 64, 10997–11013 (2021). 10.1021/acs.jmedchem.1c00367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Moustakim M et al. Discovery of an MLLT1/3 YEATS Domain Chemical Probe. Angew Chem Int Ed Engl 57, 16302–16307 (2018). 10.1002/anie.201810617 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Li X, Yao Y, Wu F & Song Y A proteolysis-targeting chimera molecule selectively degrades ENL and inhibits malignant gene expression and tumor growth. J Hematol Oncol 15, 41 (2022). 10.1186/s13045-022-01258-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Pina C, May G, Soneji S, Hong D & Enver T MLLT3 regulates early human erythroid and megakaryocytic cell fate. Cell Stem Cell 2, 264–273 (2008). 10.1016/j.stem.2008.01.013 [DOI] [PubMed] [Google Scholar]
  • 163.Gilan O et al. CRISPR-ChIP reveals selective regulation of H3K79me2 by Menin in MLL leukemia. Nat Struct Mol Biol (2023). 10.1038/s41594-023-01087-4 [DOI] [PubMed] [Google Scholar]
  • 164.Zhu L et al. ASH1L Links Histone H3 Lysine 36 Dimethylation to MLL Leukemia. Cancer Discov 6, 770–783 (2016). 10.1158/2159-8290.CD-16-0058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Aljazi MB, Gao Y, Wu Y, Mias GI & He J Histone H3K36me2-Specific Methyltransferase ASH1L Promotes MLL-AF9-Induced Leukemogenesis. Front Oncol 11, 754093 (2021). 10.3389/fonc.2021.754093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Fujimoto A et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat Genet 48, 500–509 (2016). 10.1038/ng.3547 [DOI] [PubMed] [Google Scholar]
  • 167.Liu L, Kimball S, Liu H, Holowatyj A & Yang ZQ Genetic alterations of histone lysine methyltransferases and their significance in breast cancer. Oncotarget 6, 2466–2482 (2015). 10.18632/oncotarget.2967 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Xu B et al. Novel role of ASH1L histone methyltransferase in anaplastic thyroid carcinoma. J Biol Chem 295, 8834–8845 (2020). 10.1074/jbc.RA120.013530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Rogawski DS et al. Discovery of first-in-class inhibitors of ASH1L histone methyltransferase with anti-leukemic activity. Nat Commun 12, 2792 (2021). 10.1038/s41467-021-23152-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Zhang C, Xu L, Zheng X, Liu S & Che F Role of Ash1l in Tourette syndrome and other neurodevelopmental disorders. Dev Neurobiol 81, 79–91 (2021). 10.1002/dneu.22795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Walens A et al. Adaptation and selection shape clonal evolution of tumors during residual disease and recurrence. Nat Commun 11, 5017 (2020). 10.1038/s41467-020-18730-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Sharma SV et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010). 10.1016/j.cell.2010.02.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Gupta PB et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146, 633–644 (2011). 10.1016/j.cell.2011.07.026 [DOI] [PubMed] [Google Scholar]
  • 174.Shaffer SM et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 546, 431–435 (2017). 10.1038/nature22794 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Yang D et al. Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution. Cell 185, 1905–1923 e1925 (2022). 10.1016/j.cell.2022.04.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Neftel C et al. An Integrative Model of Cellular States, Plasticity, and Genetics for Glioblastoma. Cell 178, 835–849 e821 (2019). 10.1016/j.cell.2019.06.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Goyal Y et al. Diverse clonal fates emerge upon drug treatment of homogeneous cancer cells. Nature 620, 651–659 (2023). 10.1038/s41586-023-06342-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Oren Y et al. Cycling cancer persister cells arise from lineages with distinct programs. Nature 596, 576–582 (2021). 10.1038/s41586-021-03796-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Alvarez-Varela A et al. Mex3a marks drug-tolerant persister colorectal cancer cells that mediate relapse after chemotherapy. Nat Cancer (2022). 10.1038/s43018-022-00402-0 [DOI] [PubMed] [Google Scholar]
  • 180.Voigt P, Tee WW & Reinberg D A double take on bivalent promoters. Genes Dev 27, 1318–1338 (2013). 10.1101/gad.219626.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Marsolier J et al. H3K27me3 conditions chemotolerance in triple-negative breast cancer. Nat Genet 54, 459–468 (2022). 10.1038/s41588-022-01047-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Mabe NW et al. Epigenetic silencing of tumor suppressor Par-4 promotes chemoresistance in recurrent breast cancer. J Clin Invest 128, 4413–4428 (2018). 10.1172/JCI99481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Chaffer CL et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 154, 61–74 (2013). 10.1016/j.cell.2013.06.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Iniguez AB et al. Resistance to Epigenetic-Targeted Therapy Engenders Tumor Cell Vulnerabilities Associated with Enhancer Remodeling. Cancer Cell 34, 922–938 e927 (2018). 10.1016/j.ccell.2018.11.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Ohta Y et al. Cell-matrix interface regulates dormancy in human colon cancer stem cells. Nature (2022). 10.1038/s41586-022-05043-y [DOI] [PubMed] [Google Scholar]
  • 186.Fane ME et al. Stromal changes in the aged lung induce an emergence from melanoma dormancy. Nature 606, 396–405 (2022). 10.1038/s41586-022-04774-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Warren A et al. Global computational alignment of tumor and cell line transcriptional profiles. Nat Commun 12, 22 (2021). 10.1038/s41467-020-20294-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Kadur Lakshminarasimha Murthy P et al. Epigenetic basis of oncogenic-Kras-mediated epithelial-cellular proliferation and plasticity. Dev Cell 57, 310–328 e319 (2022). 10.1016/j.devcel.2022.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Filbin MG et al. Developmental and oncogenic programs in H3K27M gliomas dissected by single-cell RNA-seq. Science 360, 331–335 (2018). 10.1126/science.aao4750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Nieto MA, Huang RY, Jackson RA & Thiery JP Emt: 2016. Cell 166, 21–45 (2016). 10.1016/j.cell.2016.06.028 [DOI] [PubMed] [Google Scholar]
  • 191.Yang J et al. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 21, 341–352 (2020). 10.1038/s41580-020-0237-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.van Groningen T et al. Neuroblastoma is composed of two super-enhancer-associated differentiation states. Nat Genet 49, 1261–1266 (2017). 10.1038/ng.3899 [DOI] [PubMed] [Google Scholar]
  • 193.Boeva V et al. Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries. Nat Genet 49, 1408–1413 (2017). 10.1038/ng.3921 [DOI] [PubMed] [Google Scholar]
  • 194.Mabe NW et al. Transition to a mesenchymal state in neuroblastoma confers resistance to anti-GD2 antibody via reduced expression of ST8SIA1. Nat Cancer (2022). 10.1038/s43018-022-00405-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Gartlgruber M et al. Super enhancers define regulatory subtypes and cell identity in neuroblastoma. Nature Cancer 2, 114–128 (2020). 10.1038/s43018-020-00145-w [DOI] [PubMed] [Google Scholar]
  • 196.Linder S et al. Drug-induced epigenomic plasticity reprograms circadian rhythm regulation to drive prostate cancer towards androgen-independence. Cancer Discov (2022). 10.1158/2159-8290.CD-21-0576 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Apfelbaum AA et al. EWS::FLI1 and HOXD13 Control Tumor Cell Plasticity in Ewing Sarcoma. Clin Cancer Res 28, 4466–4478 (2022). 10.1158/1078-0432.CCR-22-0384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Rubin MA, Bristow RG, Thienger PD, Dive C & Imielinski M Impact of Lineage Plasticity to and from a Neuroendocrine Phenotype on Progression and Response in Prostate and Lung Cancers. Mol Cell 80, 562–577 (2020). 10.1016/j.molcel.2020.10.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Oser MG, Niederst MJ, Sequist LV & Engelman JA Transformation from non-small-cell lung cancer to small-cell lung cancer: molecular drivers and cells of origin. Lancet Oncol 16, e165–172 (2015). 10.1016/S1470-2045(14)71180-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Hoek KS et al. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res 68, 650–656 (2008). 10.1158/0008-5472.CAN-07-2491 [DOI] [PubMed] [Google Scholar]
  • 201.Gardner R et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 127, 2406–2410 (2016). 10.1182/blood-2015-08-665547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Saez-Ayala M et al. Directed phenotype switching as an effective antimelanoma strategy. Cancer Cell 24, 105–119 (2013). 10.1016/j.ccr.2013.05.009 [DOI] [PubMed] [Google Scholar]
  • 203.Davies A et al. An androgen receptor switch underlies lineage infidelity in treatment-resistant prostate cancer. Nat Cell Biol 23, 1023–1034 (2021). 10.1038/s41556-021-00743-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Yamamoto S et al. JARID1B is a luminal lineage-driving oncogene in breast cancer. Cancer Cell 25, 762–777 (2014). 10.1016/j.ccr.2014.04.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Hinohara K et al. KDM5 Histone Demethylase Activity Links Cellular Transcriptomic Heterogeneity to Therapeutic Resistance. Cancer Cell 34, 939–953 e939 (2018). 10.1016/j.ccell.2018.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Drosos Y et al. NSD1 mediates antagonism between SWI/SNF and polycomb complexes and is required for transcriptional activation upon EZH2 inhibition. Mol Cell 82, 2472–2489 e2478 (2022). 10.1016/j.molcel.2022.04.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Sparbier CE et al. Targeting Menin disrupts the KMT2A/B and polycomb balance to paradoxically activate bivalent genes. Nat Cell Biol (2023). 10.1038/s41556-022-01056-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Zhang Y et al. Genome-wide CRISPR screen identifies PRC2 and KMT2D-COMPASS as regulators of distinct EMT trajectories that contribute differentially to metastasis. Nat Cell Biol 24, 554–564 (2022). 10.1038/s41556-022-00877-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Avgustinova A et al. Loss of G9a preserves mutation patterns but increases chromatin accessibility, genomic instability and aggressiveness in skin tumours. Nat Cell Biol 20, 1400–1409 (2018). 10.1038/s41556-018-0233-x [DOI] [PubMed] [Google Scholar]
  • 210.Hogg SJ et al. Targeting histone acetylation dynamics and oncogenic transcription by catalytic P300/CBP inhibition. Mol Cell 81, 2183–2200 e2113 (2021). 10.1016/j.molcel.2021.04.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.DuBois SG et al. Randomized Phase II Trial of MIBG Versus MIBG, Vincristine, and Irinotecan Versus MIBG and Vorinostat for Patients With Relapsed or Refractory Neuroblastoma: A Report From NANT Consortium. J Clin Oncol 39, 3506–3514 (2021). 10.1200/JCO.21.00703 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Gardner EE et al. Chemosensitive Relapse in Small Cell Lung Cancer Proceeds through an EZH2-SLFN11 Axis. Cancer Cell 31, 286–299 (2017). 10.1016/j.ccell.2017.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Kailayangiri S et al. EZH2 Inhibition in Ewing Sarcoma Upregulates GD2 Expression for Targeting with Gene-Modified T Cells. Mol Ther 27, 933–946 (2019). 10.1016/j.ymthe.2019.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Reppel L et al. Targeting disialoganglioside GD2 with chimeric antigen receptor-redirected T cells in lung cancer. J Immunother Cancer 10 (2022). 10.1136/jitc-2021-003897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Ennishi D et al. Molecular and Genetic Characterization of MHC Deficiency Identifies EZH2 as Therapeutic Target for Enhancing Immune Recognition. Cancer Discov 9, 546–563 (2019). 10.1158/2159-8290.CD-18-1090 [DOI] [PubMed] [Google Scholar]
  • 216.Palikyras S & Papantonis A Modes of phase separation affecting chromatin regulation. Open Biol 9, 190167 (2019). 10.1098/rsob.190167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Lyon AS, Peeples WB & Rosen MK A framework for understanding the functions of biomolecular condensates across scales. Nat Rev Mol Cell Biol 22, 215–235 (2021). 10.1038/s41580-020-00303-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Shin Y & Brangwynne CP Liquid phase condensation in cell physiology and disease. Science 357 (2017). 10.1126/science.aaf4382 [DOI] [PubMed] [Google Scholar]
  • 219.van der Lee R et al. Classification of intrinsically disordered regions and proteins. Chem Rev 114, 6589–6631 (2014). 10.1021/cr400525m [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Hansen JC, Lu X, Ross ED & Woody RW Intrinsic protein disorder, amino acid composition, and histone terminal domains. J Biol Chem 281, 1853–1856 (2006). 10.1074/jbc.R500022200 [DOI] [PubMed] [Google Scholar]
  • 221.Kilgore HR & Young RA Learning the chemical grammar of biomolecular condensates. Nat Chem Biol (2022). 10.1038/s41589-022-01046-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Klein IA et al. Partitioning of cancer therapeutics in nuclear condensates. Science 368, 1386–1392 (2020). 10.1126/science.aaz4427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Chandra B et al. Phase Separation Mediates NUP98 Fusion Oncoprotein Leukemic Transformation. Cancer Discov 12, 1152–1169 (2022). 10.1158/2159-8290.CD-21-0674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Tripathi S et al. Defining the condensate landscape of fusion oncoproteins. Nature Communications 14 (2023). 10.1038/s41467-023-41655-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Chong S et al. Tuning levels of low-complexity domain interactions to modulate endogenous oncogenic transcription. Mol Cell 82, 2084–2097 e2085 (2022). 10.1016/j.molcel.2022.04.007 [DOI] [PubMed] [Google Scholar]
  • 226.Seong BKA et al. TRIM8 modulates the EWS/FLI oncoprotein to promote survival in Ewing sarcoma. Cancer Cell 39, 1262–1278 e1267 (2021). 10.1016/j.ccell.2021.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Chen S-H et al. Constitutive protein degradation induces acute cell death via proteolysis products. Biorxiv (2023). 10.1101/2023.02.06.527237 [DOI] [Google Scholar]
  • 228.Belk JA, Daniel B & Satpathy AT Epigenetic regulation of T cell exhaustion. Nat Immunol 23, 848–860 (2022). 10.1038/s41590-022-01224-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Villanueva L, Alvarez-Errico D & Esteller M The Contribution of Epigenetics to Cancer Immunotherapy. Trends Immunol 41, 676–691 (2020). 10.1016/j.it.2020.06.002 [DOI] [PubMed] [Google Scholar]
  • 230.Hiatt JB et al. Inhibition of LSD1 with Bomedemstat Sensitizes Small Cell Lung Cancer to Immune Checkpoint Blockade and T-Cell Killing. Clin Cancer Res 28, 4551–4564 (2022). 10.1158/1078-0432.CCR-22-1128 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES